Abstract
Purpose:
Treatment for patients with recurrent endometrioid endometrial cancer (EEC) are limited as paclitaxel is the only second line chemotherapy with a response rate >13%. Targeting PIK3/mTOR in combination with hormonal therapy has shown promise. The addition of metformin may enhance this response. We conducted a phase II study evaluating everolimus, letrozole, and metformin in advanced/recurrent ECC.
Experimental design:
A Simon two-stage design was employed. Women with ≤2 prior chemotherapy regimens for recurrence were eligible. Pre-treatment biopsy was required, followed by everolimus 10mg PO, letrozole 2.5mg PO, and metformin 500mg PO BID on a 4 week cycle. The primary endpoint was clinical benefit (CB), defined as complete response (CR), partial response (PR), or stable disease (SD) confirmed at 16 weeks. Patients were treated until progression or toxicity.
Results:
Sixty-two patients were enrolled. Median age was 62 years (40 – 77) with 401 cycles completed, median of 6 cycles (1 – 31). Fifty-four patients were evaluable for response with a CB rate of 50% (27/54). Best overall response (OR) was PR 28% (15/54) and SD 22% (12/54). Thirteen patients received > 12 cycles. Median follow-up was 17.9 months (2 – 47). Median PFS was 5.7 (95% CI 3.0 to 8.1) and OS was 19.6 months (95% CI 14.2 to 26.3). Positive progesterone receptor expression was associated with CB (89.5% versus 27.3%, p=0.001).
Conclusions:
Everolimus, letrozole and metformin resulted in 50% CB and 28% OR in women with recurrent EC. Progesterone receptor positive tumors may have better response; validation studies are needed.
Introduction
Endometrial cancer is the most common gynecologic malignancy in the United States with an estimated 61,880 new diagnoses and 12,160 expected deaths this year. Both the incidence and mortality associated with endometrial cancer have continued to increase; in part because of the limited treatment options for women with advanced or recurrent disease. A number of studies done through the Gynecologic Oncology Group in women with recurrent endometrial cancer have shown poor responses to salvage chemotherapy. Paclitaxel was the only active single agent chemotherapeutic with a response rate (RR) of 27.3% in women with advanced or recurrent endometrial cancer. These data however, were in women without prior treatment with taxane-based chemotherapy 1. Currently, the combination of paclitaxel and carboplatin is often used at first line treatment in advanced or recurrent endometrial cancer 2. As a result, the focus of second line therapy has been on non-taxane treatment. Other agents studied including etoposide, liposomal doxorubicin and topotecan have shown less than promising results with RR between 4 – 13% 3, 4. Bevacizumab was approved in the recurrent setting based on 6-month progression free survival (PFS) of 40% and overall RR of 13.5% 5. As a result, the major focus of clinical research studies has included alternative treatment strategies.
The Cancer Genome Atlas and other molecular studies have identified that aberrations in the PI3K/AKT/mTOR pathway are common in endometrioid endometrial cancer (EEC), with loss of PTEN found in up to 80% of tumors 6. In addition, mutations in PTEN and PIK3CA have been identified in up to 50% of tumors7. A number of studies have evaluated the role of single agent mTOR inhibition in recurrent endometrial cancer as both primary and second-line therapy 8–10. While objective RR ranged from 0–24%, stable disease (SD) rates have been as high as 90% resulting in further study.
A phase II study evaluating the combination of everolimus and letrozole in women with advanced and recurrent endometrial cancer showed a clinical benefit (CB) rate of 40% with an objective RR of 32% including nine complete and two partial responses 11. In this trial, nine patients were on metformin either prior to entry or started on metformin due to elevated glucose, a common side effect of everolimus. Among the small number of patients on metformin, the CB rate was 78% with an objective RR of 56% compared to CB rate of 38% and an objective RR of 23% in patients not taking metformin11. Previously, we demonstrated that a short course of oral metformin prior to endometrial cancer surgery resulted in down-regulation of the AKT/mTOR pathway at the tissue level 12. In addition, preclinical studies using a xenograft mouse model of endometrial cancer showed that metformin inhibited cell proliferation, induced apoptosis and decreased tumor growth with the greatest response seen in cells harboring activating KRAS mutations. Metformin displaced constitutively active KRAS from the cell membrane causing uncoupling of the MAPK signaling pathway. These findings provided rationale for combining metformin and a PI3K-targeted agent, particularly in KRAS mutation positive endometrial cancer 13.
The objective of the current study was to estimate the CB rate of the combination of everolimus, letrozole and metformin in women with advanced or recurrent EEC. In addition, we performed an exploratory analysis to determine if specific molecular features, including presence of a KRAS mutation were associated with response to therapy.
Patients and Methods
This was a phase II, open-label trial performed within the MD Anderson Cancer Research Network. Patients enrolled at MD Anderson Cancer Center (main campus and Houston area locations), Lyndon B. Johnson Hospital (Houston, Texas), and Cooper Health System (Camden, New Jersey). The primary objective of the study was to determine the CB of everolimus, letrozole, and metformin in patients with recurrent or advanced, non-curable EEC. Secondary objectives included treatment toxicity, progression free survival (PFS), time to disease progression and overall survival (OS) in this patient population. Exploratory objective included molecular analysis on tumor tissue obtained at study entry to determine if these findings informed response to therapy. The study was registered on clinicaltrials.gov as study number NCT01797523. The Institutional Review Board at each site approved the study in compliance with the Declaration of Helsinki.
Patient Population
Eligible patients had advanced or recurrent histologically confirmed endometrioid or mixed-endometrioid endometrial adenocarcinoma, which was refractory to curative therapy. At least 18 patients had to have a KRAS mutation to be able to determine if presence of this mutation was associated with response. Patients were excluded if they had a carcinosarcoma, sarcoma, pure clear cell carcinoma or any component of serous carcinoma. Patients were allowed up to two prior chemotherapeutic regimens for recurrent disease. Chemotherapy administered in conjunction with primary radiation as a radio-sensitizer was not counted as prior therapy. Prior hormonal therapy, including letrozole was allowed. Patients with known diabetes, including those already on metformin were also eligible. All patients were required to have RECIST version 1.1 measurable disease and ECOG performance status of 0 to 2. 14. Pretreatment hematologic, renal, and hepatic function tests were required to be grade 0 or 1 according to Common Terminology Criteria for Adverse Events (CTCAE, version 4.0).
The study schema is shown in Figure 1. After informed consent was obtained, patients were enrolled on the trial and underwent a pre-treatment biopsy for molecular analysis. If a patient had a biopsy for recurrence within 3 months of enrollment, this tissue was requested and used for molecular testing. If inadequate tissue was obtained during the research biopsy, archival tissue from either the primary tumor or a recurrence biopsy were requested for molecular testing. After completion of the biopsy, patients underwent a 7–10 day lead-in with metformin only. This included 500 mg PO daily for at least 3 days, then increase to 500 mg BID until day 1, cycle 1. If patients were already on metformin, their baseline dose was titrated up to the study dose of 500mg PO BID. If their dose was ≥ 1000mg daily, they continued their usual dose.
The starting dose was everolimus 10mg PO daily, letrozole 2.5mg PO daily, and metformin 500mg BID (unless they were on a higher dose of metformin prior to enrollment). Each cycle consisted of 4 weeks of therapy. Patients continued treatment until disease progression, dose-limiting toxicity or withdrawal of consent. The primary endpoint was CB rate defined as prolonged SD ( ≥ 16 weeks), partial response or complete response by RECIST 1.1 criteria. Imaging was performed every 2 cycles until cycle 6 and then every 3 cycles, unless there was a new symptom suggestive of progression or obvious progression on physical exam. Toxicity was assessed using the National Cancer Institute CTCAE (v4.0).
Correlative studies
Paraffin-embedded sections of endometrial tumor tissue were cut at 4μm thickness and used for immunohistochemical (IHC) analysis following antigen retrieval with citrate buffer (PH 6.0). Sections were incubated in primary antibody against Phospho-S6 Ribosomal Protein (Ser235/236) (pS6p, 1:75), Progesterone receptor (PgR, 1:500) (Cell Signaling, MA), Phosphatase and tensin homolog (PTEN, 1:100), and Estrogen receptor (ER, 1:35) (DAKO Incorporation), followed by the incubation with biotinylated anti rabbit or anti mouse IgG and streptavidin-HRP (Dako Incorporation). Diaminobenzidine solution was applied to visualize the complex. The sections were counterstained with Mayer’s hematoxylin. To evaluate differential expression levels of the markers, the following scoring systems were used: PTEN was scored as positive (>90% of tumor cells show positive cytoplasmic and nuclear staining), negative (<1% of tumor cells show positive staining in cytoplasmic and nuclear), and heterogeneous (tumors show positive and negative staining foci) 15. For PgR and ER, the proportion and intensity of staining was used to determine H score -- % of tumor cells stained with certain intensity multiplied by intensity score (0: none; 1: weak; 2: intermediate; 3: strong) was calculated, with the score ranging from 0 to 300. Tumor tissue staining with H score ≥1 was reported as positive, and negative staining was reported as H score<1 16. For pS6P: H score ranging from 0 to 300 was used to report tumor cells staining.
Molecular evaluation of mutational status was performed using a next-generation sequencing panel of hotspots from 50 genes (see supplemental data) in a clinical molecular diagnostics lab 17.
Statistical analysis
A Simon two-stage design was employed with 25 patients evaluable for efficacy in the first phase18. If there were at least 12 patients with CB measured after 2 cycles of therapy (at 8 weeks), the additional 29 patients enrolled in the second stage for 54 evaluable patients. This study design had a 91% power with a 10% significance level to reject a CB of 40% in favor of a CB rate of 60% (the hypothesized rate). This design had a 0.73 probability of stopping after the first stage if the CB rate was actually 40% and a 0.08 probability of stopping after the first stage if the CB rate was actually 60%.
Descriptive statistics were used to summarize the demographic and clinical characteristics of patients, as well as tumor response. The CB rate (CR + PR + SD) measured after 8 weeks (2 cycles) of therapy and confirmed after 16 weeks (4 cycles) of therapy, was estimated with an exact 95% binomial confidence interval. Duration of CB for those patients with at least SD after 8 weeks (2 cycles) and confirmed after 16 weeks (4 cycles), was calculated from the end of cycle 2 to the date of disease progression or date of last contact. PFS was calculated from day 1, cycle 1 until date of progression or last contact. OS was calculated from day 1, cycle 1 until date of death or last contact. Patients, who came off trial due to toxicity or in the absence of progression, were censored at last known follow up. Patients on active treatment were censored at last tumor assessment if they were alive and without progression. Kaplan-Meier method was used to analyze PFS and OS, and Fisher’s exact test was used to compare CB rate in patients with and without molecular alterations. Study data were collected and managed using REDCap 19. All statistical analyses were performed using SAS 9.4 for Windows (Copyright 2002–2012 by SAS Institute Inc, Cary, NC).
Results
Sixty-two patients enrolled in the study between October 2013 and May 2016. Eight patients were consented but were not evaluable for response due to withdrawal of consent (2), bowel obstruction/rapid progression prior to completion of cycle 1 (2), diagnosis of thyroid cancer prior to initiation of therapy (1), change in treatment plan to radiation (1), no measurable disease (1) and death due to a car accident during cycle 1 (1). The demographic characteristics of the 54 evaluable patients are shown in table 1.
Table 1.
Characteristic | No. of Patients (N=54) | % |
---|---|---|
Age (median, years) | 62 (40 – 77) | |
Body mass index (BMI, kg/m2) | 33.3 (16.9 – 65.5) | |
Stage of Disease | ||
I | 13 | 24 |
II | 4 | 7 |
III | 10 | 19 |
IV | 21 | 39 |
unknown | 6 | 11 |
Histology | ||
Endometrioid | 50 | 93 |
Mixed endometrioid | 4 | 7 |
Grade | ||
1 | 10 | 18 |
2 | 22 | 41 |
3 | 20 | 37 |
unknown | 2 | 4 |
Number of prior chemotherapy agents* | ||
0 | 8 | 15 |
1 | 22 | 41 |
2 | 24 | 44 |
Previous radiation therapy | ||
Yes | 35 | 65 |
No | 19 | 35 |
Does not include radio-sensitizing chemotherapy
Median age was 62 years. Median body mass index (BMI) was 33.3 kg/m2. Ninety-three percent had pure endometrioid histology; 18% grade 1, 41% grade 2, 37% grade 3, 4% unknown grade. A majority of patients (85%) had received at least one prior chemotherapy agent for recurrence and 65% had received prior radiation therapy. Five patients (9%) were on metformin prior to enrollment in the study.
Among the 54 evaluable patients, median number of cycles was 6 (range 1 – 31) with a total of 410 cycles completed. Clinical outcomes are shown in table 2. The overall CB rate was 50% (95%CI 36.1 to 63.9); with 28% PR and 22% SD confirmed after16 weeks of therapy. Figure 2 is a swimmer plot depicting both clinical outcome and duration of response. The median duration of response for those with a confirmed CB after 16 weeks was 7.1 months (range 2.9 to 26.6). The six month PFS was 0.41 (95% CI 0.27 – 0.54) and 12 month PFS 0.25 (95% CI 0.14 – 0.37). A majority of patients (90%) came off study due to progression of disease. One patient remains on active treatment. She has received 31 cycles and had a PR. The median follow-up time was 17.9 months (range 2 – 47 months). The median PFS was 5.7 months (95% CI 3. to 8.2) and median OS was 19.6 months (95% CI 14.2 to 26.3). At the time of this analysis, 67% of patients were deceased.
Table 2.
Outcome (N=54) | % | |
---|---|---|
Median number of cycles | 6 (1–31) | |
Clinical benefit* | ||
No | 27 | 50 |
Yes | 27 | 50 |
Best response | ||
Partial response | 15 | 28 |
Stable disease | 12 | 22 |
Progressive disease | 27 | 50 |
Reason off study (n = 53) | ||
Progressive disease | 48 | 90 |
Withdrew consent | 3 | 6 |
Non- compliance | 1 | 2 |
Toxicity | 1 | 2 |
Current status | ||
Alive with disease | 21 | |
Deceased | 33 | |
Median follow up | 17.9 (2 – 47) | |
Median progression free survival | 5.7 months | |
Median overall survival | 19.6 months | |
Confirmed after 4 cycles of therapy (16 weeks)
Toxicity was evaluated in the 59 patients who had at least one dose of treatment and had a follow up evaluation for side effects. The majority of adverse events were grade 1 and 2 and manageable with supportive care. The grade2, 3 and 4 adverse events are listed in Table 3. There were no grade 5 events. Anemia was the most common adverse event with 85% of patients having at least grade two. Grade 3 anemia was found in 24%, followed by hypertriglyceridemia (15%), hyperglycemia (9%), hyponatremia (7%), fatigue (6%), and thrombocytopenia (6%). Four patients required a dose reduction to everolimus 5 mg daily due to pneumonitis after 4 cycles (2), elevated creatinine after 9 cycles (1) and persistent grade 3 thrombocytopenia after 4 cycles. All patients remained on study at the reduced dose. One patient had to discontinue metformin due to persistent grade 2 diarrhea that was not tolerable for the patient. Only one patient was taken off study due to toxicity; grade 3 liver function tests after cycle 1, which did not resolve within 28 days. Of note, she had a PR on follow up imaging after only receiving one cycle of therapy.
Table 3.
Adverse Event (N=59) | Grade 2 | Grade 3 | Grade 4 | Total Grade 3 and 4 |
---|---|---|---|---|
Anemia | 36 | 14 | 0 | 14(24%) |
Hypertriglyceridemia | 14 | 7 | 1 | 8(15%) |
Hyperglycemia | 15 | 5 | 0 | 5 (9%) |
Hyponatremia | 0 | 4 | 0 | 4 (7%) |
Fatigue | 17 | 3 | 0 | 3 (6%) |
Thrombocytopenia | 4 | 3 | 0 | 3 (6%) |
Abdominal pain | 11 | 2 | 0 | 2 (4%) |
Mucositis oral | 13 | 2 | 0 | 2 (4%) |
Infection | 3 | 2 | 0 | 2 (4%) |
Elevated liver function test | 4 | 2 | 0 | 2 (4%) |
Pain | 4 | 2 | 0 | 2 (4%) |
Forty-seven patients had tissue available for molecular analysis (Table 4). A majority were biopsies done at the time of recurrence (51% on study and 24% archived tissue). For 19%, the primary tumor tissue was evaluated.
Table 4.
Pathologic Findings | N | % |
---|---|---|
Tissue evaluated | ||
Pre-treatment biopsy on study | 24 | 51 |
Archived primary tumor | 9 | 19 |
Archived recurrence biopsy | 11 | 24 |
Unknown | 3 | 6 |
Immunohistochemistry | ||
Estrogen positive | 29/32 | 91 |
Estrogen negative | 3/32 | 9 |
Progesterone positive | 19/30 | 63 |
Progesterone negative | 11/30 | 37 |
Mutation Detected by CMS50 Panel (N=47) | ||
Yes | 47 | 100 |
No | 0 | 0 |
Median number of mutations (range) | 2 (1–4) | |
Most common mutations | ||
PTEN | 28 | 60 |
PIK3CA | 22 | 47 |
KRAS | 18 | 38 |
TP53 | 12 | 26 |
CTNNB1 | 11 | 23 |
AKT1 | 2 | 11 |
The majority of tumors were ER and PgR positive, 91% and 63% respectively. Of the 15 samples that had data on ER status at both primary diagnosis and recurrence, 100% matched (95% CI: 78% – 100%). Of the 12 samples that had data on PgR status at both primary and recurrence, 75% matched (95% CI: 43%−95%). There was no difference in CB rate based on ER status (ER neg 33% (95% CI: 0.8% – 91%) versus ER pos 69% (49% - 85%), p=0.27). However, there was a significant difference in CB rate based on PgR status (PgR neg 27% (6% – 61%)) versus PgR pos 90% (67% – 99%), p=0.001). The difference in overall response (PgR neg 9% (0.2% – 41%) versus pos 45% (23% – 68%), p=0.06) was present but not statistically significant.
Next generation sequencing was performed on 47 (87%) tissue samples. The median number of mutations detected was 2 (range 1–4). The most common mutations were PTEN (60%), PIK3CA (47%), KRAS (38%), TP53 (26%) and CTNNB1 (23%). There was no difference in CB rate based on PTEN (p=0.78) or KRAS mutation status (p=0.99). There were 9 patients who had mutational analysis on their primary tumor in addition to the research biopsy performed as part of the study. Among these, the presence of a mutation in both samples matched 100% for PTEN (7 patients, 95% CI 59–100%), 100% AKT (2 patients, 95% CI 16–100%), 100% for PIK3CA (5 patients, 95% CI 48–100%), 100% for KRAS (5 patients, 95% CI 48–100%), 60% for CTNNB1 (5 patients, 95% CI 15–95%), and 50% for p53 (2 patients, 95%CI 1–99%).
Discussion
The combination of everolimus, letrozole and metformin resulted in CB for 50% of women with advanced or recurrent endometrial cancer, with an overall RR of 28%. The median duration of response was 7 months, with a six month PFS of 41%. These results compare favorably to currently approved therapies for recurrent endometrial cancer. The combination was well tolerated, with a manageable toxicity profile. PgR status was associated with response, with a 90% CB rate and 45% overall RR among women who had PgR expression in their tumor. This is among the first phase 2 studies in recurrent endometrial cancer to find a candidate biomarker associated with clinical benefit. Further validation studies are needed.
A number of single agent studies targeting mTOR have shown RR between 0–24% with SD rates as high as 90% 8–10, 20. The high CB rate has led to additional studies combining targets, including hormonal therapy. The concept of combining hormonal therapy with mTOR inhibition stems from preclinical data that suggest that blocking the PI3K/AKT/mTOR pathway may overcome resistance to hormonal therapy 21. Co-targeting the mTOR pathway and estrogen receptor has shown benefit for patients with hormone receptor positive (HR+) breast cancer in clinical trials 22. In a phase 3 randomized trial BOLERO-2, the addition of everolimus to endocrine therapy had significant improvement in progression free but not overall survival 23, 24.
The combination of mTOR inhibition and hormonal therapy in endometrial cancer has also been reported, however, with less consistent results. Fleming et al. conducted a randomized phase II trial of intravenous temsirolimus +/− megesterol alternating with tamoxifen (considered the most effective hormonal regimen) in women with recurrent endometrial cancer. There was no added benefit seen with the addition of hormones to temsirolimus. The study was closed early, however, due to toxicity with an increase in venous thrombosis seen in women on the hormonal arm 20. At our institution, a single arm phase II study of everolimus and letrozole by Slomovitz et al. reported a CB rate of 40% and an overall RR of 32% 11. The promising results from this study laid the foundation for the current study.
Elevated serum glucose is a common side effect of many mTOR inhibitors. As a result, oral hypoglycemic agents will be initiated to manage this on-target event. During the phase II trial by Slomovitz et al. evaluating everolimus and letrozole, patients appeared to benefit from the addition of metformin 11. In a posthoc analysis, the RR among patients on metformin was higher than those not receiving metformin. Specifically, there were 9 patients on metformin either prior to study enrollment or started on metformin due to elevated glucose. Among this subgroup, the CB rate was 78% with an objective RR of 56% compared to CB rate of 38% and an objective RR of 23% in patients not taking metformin.
Metformin is thought to have both a direct and indirect effect on cell growth and metabolism 25, 26. In the direct model, metformin activates AMPK, which results in phosphorylation of tuberous sclerosis 2 protein. This inhibits mTOR signaling which ultimately inhibits cell growth. Metformin also acts indirectly by increasing insulin sensitivity, increasing uptake of glucose in the cell, and ultimately decreasing circulating levels of insulin. Both insulin and IGF-1 are known growth factors that promote cell growth, so decreasing insulin would have a negative effect on cell proliferation.
Based on preclinical data using metformin in endometrial cancer cell lines, we anticipated that tumors with a KRAS mutation, would be more likely to respond and this study was powered to detect this difference 13. In this study, patients were required to have a baseline biopsy for molecular analysis to determine if molecular features at the time of recurrence could predict response to therapy. Interestingly, 100% of patients had at ≥1 mutation detected by next generation sequencing. PTEN was the most common, followed by PIK3CA, KRAS, TP53 and CTNNB1. Although available for comparison in only 9 patients, there was a high concordance between mutational analysis in the primary tumor compared to the research biopsy done at recurrence. Both ER and PgR positivity were high, 91% and 63%. Like many other studies in endometrial cancer, presence of PTEN or PIK3CA mutation was not associated with response to mTOR inhibition. Although we hypothesized that KRAS mutation would be associated with response to therapy, there was no association between this biomarker or any of the detected mutations and response to therapy. PgR status measured by IHC, however, correlated with response to therapy. Interestingly, there was a high correlation between ER and PgR status in the primary tumor and the recurrent tumor among those who had testing at both time points. This is important, as it is a readily available marker when determining the best treatment for patients with EEC. Prospective integrated trials are warranted to validate this association.
Although mechanism was not evaluated in this study, we suggest that PgR status in advanced/recurrent endometrial cancer could provide a readout of coupled, functional ER/PgR signaling. As a prototypical estrogen-regulated gene, expression of PgR suggests that downstream estrogen-regulated signaling is active, and thus, potentially more sensitive to hormonal agents. The response to anti-estrogen activity of letrozole is expected to be highest in this setting. ER status alone may not indicate the functional status of downstream ER signaling. Further, metformin has been shown to influence both ER/PgR signaling and PI3K/AKT/mTOR signaling providing further support to the relevance of PgR in predicting response to this regimen 27–29. Previous studies have shown crosstalk between ER/PgR and PI3K/AKT/mTOR signaling 30. Alternatively, the differential response in PgR+ cases could reflect increased benefit in cases that are ER+ and PgR+. A larger cohort of ER- and PgR- cases are required to evaluate this relationship. Further studies will be necessary both for validation and to understand the mechanism underlying PgR status and response to letrozole, metformin, and everolimus.
Future studies focus on novel combination therapies, which may add benefit to mTOR inhibition and/or hormonal therapy, as well as studies evaluating molecular diagnostics to identify subgroups most likely to respond to these combinations. CDK4/6 inhibition has shown benefit in combination with endocrine therapy in several HR+ breast cancer populations 31–33. The combination of everolimus, exemestane and ribociclib (CDK4/6 inhibitor) is ongoing in HR+ breast cancer (clinicaltrials.gov, NCT01857193). Similarly, we are enrolling in a phase II randomized study combining everolimus, letrozole +/− ribociclib to determine if the addition of a CDK4/6 inhibitor will add benefit for women with recurrent EEC (clinicaltrials.gov, NCT03008408). These trials should continue to explore the relationship between molecular features in the tumor and benefit from targeted therapies, to identify the best treatment for individuals with EEC.
Everolimus, letrozole and metformin resulted in a CBR of 50% and an overall RR of 28% in all patients. Response may be enhanced in women with PgR positive tumors. These findings, as well as the preliminary report from GOG-3007, suggest that the backbone of everolimus and letrozole are effective in treating women with recurrent EEC and may be a reasonable choice in second-line therapy34. This is the second trial to suggest there is benefit from the addition of metformin, without an increase in toxicity for patients. This study confirmed that the combination of everolimus, letrozole and metformin showed activity in the treatment of recurrent endometrioid endometrial cancer.
Supplementary Material
Translational relevance.
In this phase 2 study, the combination of everolimus, letrozole and metformin was well tolerated and 50% of women had clinical benefit, with a 28% objective response rate and a 6 month PFS of 41%. These results compare favorably to currently approved therapies for advanced and recurrent endometrial cancer. In our biomarker exploratory analysis, we found that objective response rates (45%) and the clinical benefit (90%) were highest among women with progesterone receptor positive tumors. Further validation studies are need to correlate this candidate biomarker with clinical outcome in recurrent endometrial cancer
Acknowledgments
This work was presented as an oral abstract at the American Society of Clinical Oncology Annual Meeting in 2016 in Chicago, Illinois, USA.
Funding: This work was supported in part by Cancer Center Support Grant (NCI Grant P30 CA016672), Andrew Sabin Family Fellowship, NCI SPORE in Uterine Cancer (2P50 CA098258–06), and Novartis
Appendix 1. Genes in CMS50 Next Generation Sequencing PlatformAKT1
BRAF
EZH2
FGFR1
GNAS
GNAQ
GNA11
IDH1
IDH2
FGFR2
KRAS
NRAS
PIK3CA
MET
RET
EGFR
JAK2
MPL
PDGFRA
PTEN
TP53
FGFR3
FLT3
KIT
ERBB2
ABL1
HNF1A
HRAS
ATM
RB1
CDH1
SMAD4
STK11
ALK
SRC
SMARCB1
VHL
MLH1
CTNNB1
KDR
FBXW7
APC
CSF1R
NPM1
SMO
ERBB4
CDKN2A
NOTCH1
PTPN11
SMAD4
Footnotes
Disclosures by author
Soliman – Funded research: Novartis, Advisory board: Clovis, Janssen
Westin – Funded research: ArQule, Astrazeneca, Novartis, Biomarin, Critical Outcome Technologies, Bayer, Tesaro, Cotinga Pharmaceuticals, Clovis Oncology, Roche/Genentech. Scientific advisory: Roche, Astrazeneca, Ovation Sciences, Medivation, Genentech, Vermillion, Casdin Capital, Medscape, Clovis Oncology, Watermark Research Partners, Gerson Lehrman Group, Vaniam Group, Bioascent, Tesaro, Merck, Pfizer
Iglesias – none
Fellman – none
Yuan - none
Zhang – none
Yates – none
Broaddus – none
Slomovitz – Scientific advisory: Advaxis, AstraZeneca, Clovis, Tesaro, Janssen
Lu – none
Coleman - Funded Research: Genentech/Roche, AstraZeneca, Clovis, Janssen, Merck, Novartis
Scientific Advisory: Genentech/Roche, AstraZeneca, Clovis, Janssen, Merck, Novartis, Genmab, Tesaro, Gamamab, Immunogen, Oncomed, Pfizer, Agenus, Oncolytics, Novocure
References
- 1.Lincoln S, Blessing JA, Lee RB, Rocereto TF. Activity of paclitaxel as second-line chemotherapy in endometrial carcinoma: a Gynecologic Oncology Group study. Gynecol Oncol. 2003;88: 277–281. [DOI] [PubMed] [Google Scholar]
- 2.Miller D FV, Fleming G, Mannel R, Cohn D, Matsumoto T, Tewari K, DiSilvestro P, Pearl M, Zaino R. Randomized phase III noninferiority trial of first line chemotherapy for metastatic or recurrent endometrial carcinoma: a Gynecologic Oncology Group study. Gynecol Oncol 125: 771–3, 2012. (Abstract). Gynecol Oncol Case Rep. 2012;125: 771–773. [Google Scholar]
- 3.Muggia FM, Blessing JA, Sorosky J, Reid GC. Phase II trial of the pegylated liposomal doxorubicin in previously treated metastatic endometrial cancer: a Gynecologic Oncology Group study. J Clin Oncol. 2002;20: 2360–2364. [DOI] [PubMed] [Google Scholar]
- 4.Miller DS, Blessing JA, Lentz SS, Waggoner SE. A phase II trial of topotecan in patients with advanced, persistent, or recurrent endometrial carcinoma: a gynecologic oncology group study. Gynecol Oncol. 2002;87: 247–251. [DOI] [PubMed] [Google Scholar]
- 5.Aghajanian C, Sill MW, Darcy KM, et al. Phase II trial of bevacizumab in recurrent or persistent endometrial cancer: a Gynecologic Oncology Group study. J Clin Oncol. 2011;29: 2259–2265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Mutter GL, Lin MC, Fitzgerald JT, et al. Altered PTEN expression as a diagnostic marker for the earliest endometrial precancers. J Natl Cancer Inst. 2000;92: 924–930. [DOI] [PubMed] [Google Scholar]
- 7.Oda K, Stokoe D, Taketani Y, McCormick F. High frequency of coexistent mutations of PIK3CA and PTEN genes in endometrial carcinoma. Cancer Res. 2005;65: 10669–10673. [DOI] [PubMed] [Google Scholar]
- 8.Slomovitz BM, Lu KH, Johnston T, et al. A phase 2 study of the oral mammalian target of rapamycin inhibitor, everolimus, in patients with recurrent endometrial carcinoma. Cancer. 2010;116: 5415–5419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Colombo N, McMeekin DS, Schwartz PE, et al. Ridaforolimus as a single agent in advanced endometrial cancer: results of a single-arm, phase 2 trial. Br J Cancer. 2013;108: 1021–1026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Oza AM, Elit L, Tsao MS, et al. Phase II study of temsirolimus in women with recurrent or metastatic endometrial cancer: a trial of the NCIC Clinical Trials Group. J Clin Oncol. 2011;29: 3278–3285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Slomovitz BM, Jiang Y, Yates MS, et al. Phase II study of everolimus and letrozole in patients with recurrent endometrial carcinoma. J Clin Oncol. 2015;33: 930–936. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Soliman PT, Zhang Q, Broaddus RR, et al. Prospective evaluation of the molecular effects of metformin on the endometrium in women with newly diagnosed endometrial cancer: A window of opportunity study. Gynecol Oncol. 2016;143: 466–471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Iglesias DA, Yates MS, van der Hoeven D, et al. Another surprise from Metformin: novel mechanism of action via K-Ras influences endometrial cancer response to therapy. Mol Cancer Ther. 2013;12: 2847–2856. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45: 228–247. [DOI] [PubMed] [Google Scholar]
- 15.Djordjevic B, Hennessy BT, Li J, et al. Clinical assessment of PTEN loss in endometrial carcinoma: immunohistochemistry outperforms gene sequencing. Mod Pathol. 2012;25: 699–708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Kraus JA, Dabbs DJ, Beriwal S, Bhargava R. Semi-quantitative immunohistochemical assay versus oncotype DX((R)) qRT-PCR assay for estrogen and progesterone receptors: an independent quality assurance study. Mod Pathol. 2012;25: 869–876. [DOI] [PubMed] [Google Scholar]
- 17.Singh RR, Patel KP, Routbort MJ, et al. Clinical validation of a next-generation sequencing screen for mutational hotspots in 46 cancer-related genes. J Mol Diagn. 2013;15: 607–622. [DOI] [PubMed] [Google Scholar]
- 18.Simon R Optimal two-stage designs for phase II clinical trials. Control Clin Trials. 1989;10: 1–10. [DOI] [PubMed] [Google Scholar]
- 19.Harris PA, Taylor R, Thielke R, Payne J, Gonzalez N, Conde JG. Research electronic data capture (REDCap)--a metadata-driven methodology and workflow process for providing translational research informatics support. J Biomed Inform. 2009;42: 377–381. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Fleming GF, Filiaci VL, Marzullo B, et al. Temsirolimus with or without megestrol acetate and tamoxifen for endometrial cancer: a gynecologic oncology group study. Gynecol Oncol. 2014;132: 585–592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Gu C, Zhang Z, Yu Y, et al. Inhibiting the PI3K/Akt pathway reversed progestin resistance in endometrial cancer. Cancer Sci. 2011;102: 557–564. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Royce M, Bachelot T, Villanueva C, et al. Everolimus Plus Endocrine Therapy for Postmenopausal Women With Estrogen Receptor-Positive, Human Epidermal Growth Factor Receptor 2-Negative Advanced Breast Cancer: A Clinical Trial. JAMA Oncol. 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Baselga J, Campone M, Piccart M, et al. Everolimus in postmenopausal hormone-receptor-positive advanced breast cancer. N Engl J Med. 2012;366: 520–529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Piccart M, Hortobagyi GN, Campone M, et al. Everolimus plus exemestane for hormone-receptor-positive, human epidermal growth factor receptor-2-negative advanced breast cancer: overall survival results from BOLERO-2dagger. Ann Oncol. 2014;25: 2357–2362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Del Barco S, Vazquez-Martin A, Cufi S, et al. Metformin: multi-faceted protection against cancer. Oncotarget. 2011;2: 896–917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Shackelford DB, Shaw RJ. The LKB1-AMPK pathway: metabolism and growth control in tumour suppression. Nat Rev Cancer. 2009;9: 563–575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Xiong F, Xiao J, Bai Y, Zhang Y, Li Q, Lishuang X. Metformin inhibits estradiol and progesterone-induced decidualization of endometrial stromal cells by regulating expression of progesterone receptor, cytokines and matrix metalloproteinases. Biomed Pharmacother. 2019;109: 1578–1585. [DOI] [PubMed] [Google Scholar]
- 28.Collins G, Mesiano S, DiFeo A. Effects of Metformin on Cellular Proliferation and Steroid Hormone Receptors in Patient-Derived, Low-Grade Endometrial Cancer Cell Lines. Reprod Sci. 2019;26: 609–618. [DOI] [PubMed] [Google Scholar]
- 29.Hu M, Zhang Y, Feng J, et al. Uterine progesterone signaling is a target for metformin therapy in PCOS-like rats. J Endocrinol. 2018;237: 123–137. [DOI] [PubMed] [Google Scholar]
- 30.Simoncini T, Hafezi-Moghadam A, Brazil DP, Ley K, Chin WW, Liao JK. Interaction of oestrogen receptor with the regulatory subunit of phosphatidylinositol-3-OH kinase. Nature. 2000;407: 538–541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Sonke GS, Hart LL, Campone M, et al. Ribociclib with letrozole vs letrozole alone in elderly patients with hormone receptor-positive, HER2-negative breast cancer in the randomized MONALEESA-2 trial. Breast Cancer Res Treat. 2018;167: 659–669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Slamon DJ, Neven P, Chia S, et al. Phase III Randomized Study of Ribociclib and Fulvestrant in Hormone Receptor-Positive, Human Epidermal Growth Factor Receptor 2-Negative Advanced Breast Cancer: MONALEESA-3. J Clin Oncol. 2018: JCO2018789909. [DOI] [PubMed] [Google Scholar]
- 33.O’Shaughnessy J, Petrakova K, Sonke GS, et al. Ribociclib plus letrozole versus letrozole alone in patients with de novo HR+, HER2- advanced breast cancer in the randomized MONALEESA-2 trial. Breast Cancer Res Treat. 2018;168: 127–134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Slomovitz BM F VL, Coleman RL, Walker JL, Fleury AC, Holman LL, Miller DS. GOG 3007, a randomized phase II(RP2) trial of everolimus and letrozole (EL) or hormonal therapy (medroxyprogesterone acetate/tamoxifen, PT) in women with advanced, persistent or recurrent endometrial cancinoma (EC): A GOG FOundation Study. Gynecol Oncol. 2018;149: 2. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.