Tumor board presentation of a woman with metastatic, hormone receptor-positive, mismatch repair-deficient endometrial cancer

IF 503.1 1区 医学 Q1 ONCOLOGY CA: A Cancer Journal for Clinicians Pub Date : 2022-01-25 DOI:10.3322/caac.21715
Jennifer A. Bennett MD, Gini F. Fleming MD, Katherine C. Kurnit MD, Kathryn A. Mills MD, Willem Jan vanWeelden MD
{"title":"Tumor board presentation of a woman with metastatic, hormone receptor-positive, mismatch repair-deficient endometrial cancer","authors":"Jennifer A. Bennett MD,&nbsp;Gini F. Fleming MD,&nbsp;Katherine C. Kurnit MD,&nbsp;Kathryn A. Mills MD,&nbsp;Willem Jan vanWeelden MD","doi":"10.3322/caac.21715","DOIUrl":null,"url":null,"abstract":"<p>A 65-year-old woman presented to an outside hospital with vaginal bleeding in 2009. Dilatation and curettage showed endometrial endometrioid carcinoma, International Federation of Gynecology and Obstetrics (FIGO) grade 2. She underwent exploratory laparotomy with pelvic washings, total abdominal hysterectomy with bilateral salpingo-oophorectomy, bilateral pelvic and para-aortic lymphadenectomy, and an omental biopsy. Pathology was read as uterine carcinosarcoma, homologous type, invading 3.5 cm into a 4-cm-thick myometrium, with involvement of the endocervical epithelium (no stromal involvement) and lymphovascular invasion. All 14 of the sampled lymph nodes (6 left pelvic, 1 left common iliac, 1 left aortic, 3 right pelvic, 2 right common iliac, and 1 right aortic) were negative for carcinoma. The tumor-node-metastasis (TNM) pathologic stage using current staging (2018 FIGO Cancer Report<span><sup>1</sup></span>) was pT1b N0 <i>American Joint Committee on Cancer [AJCC] Cancer Staging Manual</i>, eighth edition), FIGO stage IB. Postoperative positron emission tomography/computed tomography (CT) imaging studies demonstrated no evidence of disease.</p><p>The patient was otherwise healthy. Her father had lung cancer, but there was no other history of cancer in the family. She received pelvic radiotherapy and 4 cycles of paclitaxel 135 mg/m<sup>2</sup> plus ifosfamide 1600 mg/m<sup>2</sup>, with mesna and granulocyte-colony–stimulating factor support.</p><p>Five years later (at age 71 years), she developed abdominal pain and vomiting and was managed conservatively for a partial small bowel obstruction. CT scans of the abdomen and pelvis showed a large, heterogeneously enhancing mass in the left suprarenal space, measuring 8.9 × 8.6 cm, likely originating from the left adrenal gland. A lobular mass in the posterior left lung base measuring 3.2 × 1.6 cm was also noted. A CT scan of the chest revealed multiple, bilateral pulmonary nodules, the largest measuring 2.4 × 2.2 cm, some with central cavitation, most consistent with metastatic disease. A fine-needle aspirate of the suprarenal mass showed poorly differentiated carcinoma with extensive necrosis.</p><p>At that time, the patient presented to a university hospital. Review of the original pathology reclassified the tumor as endometrial endometrioid carcinoma, FIGO grade 3 (Fig. 1A), with the suprarenal mass morphologically consistent with the patient's primary. Estrogen receptor (ER) and progesterone receptor (PR) were strongly and diffusely positive (Fig. 1B,C).</p><p>Subtyping of high-grade endometrial carcinomas is diagnostically challenging for pathologists, with even expert gynecologic pathologists reaching consensus in only 63% to 72% of tumors.<span><sup>2, 3</sup></span> The most common disputes include endometrioid versus serous carcinoma, serous versus clear cell carcinoma, and endometrioid/clear cell/undifferentiated carcinoma versus carcinosarcoma.<span><sup>2</sup></span> Morphologically, high-grade endometrial carcinoma can show extensive solid growth, whereas low-grade endometrioid carcinoma may have a spindled morphology,<span><sup>4</sup></span> both of which may mimic carcinosarcoma with a homologous sarcomatous component. Although immunohistochemistry can be useful in subtyping endometrioid, serous, and clear cell carcinomas, it is generally unhelpful for distinguishing carcinosarcomas from pure endometrial carcinomas. Because carcinosarcomas are considered a subtype of endometrial carcinoma<span><sup>5</sup></span> rather than a mixed epithelial and mesenchymal tumor, as previously hypothesized, it is not surprising that the sarcoma (as well as the carcinoma) portions of the tumor may show cytokeratin and PAX8 expression, eliminating the utility of these stains in differentiating between carcinosarcoma and pure endometrial carcinoma.<span><sup>6, 7</sup></span></p><p>The College of American Pathologists recommends performing ER immunohistochemistry on stage III, IV, and recurrent endometrial carcinomas to predict response to endocrine therapy.<span><sup>8</sup></span> Unlike in breast carcinoma, there is not a uniform reporting system for hormone receptors in endometrial carcinoma; however, it is recommended that the number of positive cells and staining intensity be documented.<span><sup>9</sup></span></p><p>The patient was presented with the option of chemotherapy with carboplatin plus paclitaxel. She declined chemotherapy. She was also presented with the option of participation in a clinical trial, Gynecologic Oncology Group (GOG) 3007 (a randomized phase II trial of everolimus and letrozole or hormonal therapy [medroxyprogesterone acetate/tamoxifen], in women with advanced, persistent, or recurrent endometrial cancer; Clinicaltrials.gov identifier NCT02228681). She elected to participate in GOG 3007 and was randomized to therapy with medroxyprogesterone acetate/tamoxifen. She was treated with oral tamoxifen 20 mg twice daily (continuously), with the addition of medroxyprogesterone acetate 200 mg daily on alternating weeks (days 8-14 and 22-28 of a 28-day cycle). She had a partial response to therapy (Fig. 2) and remained on study for 19 cycles (about 1.5 years), at which time she had mild disease progression in the lung. She was switched to single-agent letrozole without further response.</p><p>Of note, had her tumor not been reclassified as an endometrioid endometrial carcinoma, she would have been ineligible for GOG 3007 because that trial, as well as many current ongoing trials for women with endometrial cancer (including those testing immunotherapy), excludes carcinosarcomas.</p><p>Whereas, historically, progestins were front-line therapy for metastatic endometrial cancer, currently, they are most commonly used in the second or later lines, although it is not clear that their activity is fully maintained in this setting. Single-agent progestins (most frequently oral medroxyprogesterone acetate or megestrol acetate),<span><sup>10, 11</sup></span> aromatase inhibitors (letrozole, anastrozole),<span><sup>12, 13</sup></span> selective ER modulators (tamoxifen, arzoxifene),<span><sup>14, 15</sup></span> fulvestrant,<span><sup>16, 17</sup></span> or combination therapies, such as progestin/tamoxifen<span><sup>18, 19</sup></span> or everolimus (an mTOR inhibitor)/letrozole,<span><sup>20, 21</sup></span> have all been investigated and have shown some efficacy. A 2017 meta-analysis evaluated 16 studies that included second-line endocrine therapy and reported an objective response rate of 18.5%.<span><sup>22</sup></span> When patients with stable disease were also included, the clinical benefit rate was 36%. By comparison, a randomized trial of tamoxifen versus anastrazole for first-line therapy of metastatic breast cancer (almost one-half of patients had hormone receptor-positive tumors and, in slightly greater than one-half, the tumor hormone receptor status was unknown) demonstrated an objective response rate of approximately 33% in both arms.<span><sup>23</sup></span> The current National Comprehensive Cancer Network guidelines include multiple endocrine therapy options for endometrial cancer, but they note that these therapies are typically reserved for patients who have low-grade endometrioid tumors with more indolent behavior.<span><sup>24</sup></span></p><p>More recently, the combination of everolimus plus letrozole and the combination of tamoxifen plus medroxyprogesterone acetate were evaluated in GOG 3007, the study in which our patient enrolled. Although this was a randomized, phase 2 study of women with advanced or recurrent endometrial cancer and included both first-line and second-line settings, it is important to note that the 2 regimens were not directly compared. Preliminary results were presented at the 2018 Society for Gynecologic Oncology Annual Meeting. The progression-free survival was 6.3 months for the everolimus/letrozole arm and 3.8 months for the medroxyprogesterone acetate/tamoxifen arm.<span><sup>25</sup></span> However, in patients who had received prior chemotherapy, progression-free survival was 3.3 months and 3.2 months, respectively. Overall survival for the medroxyprogesterone acetate/tamoxifen group was 16.6 months but was not yet calculable for the everolimus/letrozole group. In addition, a phase 2 trial of letrozole plus ribociclib (a cyclin-dependent kinase inhibitor) was recently published. Of the 20 patients with endometrial cancer enrolled on the trial, 55% were still on treatment at 12 weeks.<span><sup>26</sup></span> Similar to the studies discussed above, those patients who had low-grade endometrioid tumors seemed to derive the most benefit.</p><p>Discussion regarding the use of endocrine therapy before chemotherapy in the metastatic setting is ongoing. A 2017 meta-analysis evaluated results from 23 studies of hormonal therapy in the first-line setting and found that the overall response rate (ORR) was highest in patients who had hormone receptor-positive tumors, at 32.5%,<span><sup>22</sup></span> and that low-grade tumors had higher response rates than high-grade tumors. The ORR for ER-negative tumors was only 9.2%. The authors also reported that single-agent aromatase inhibitors produced the lowest response rates of the endocrine therapy classes, with a mean ORR of only 8.6%. For patients treated on GOG 3007 in the first-line setting, the objective response rate was 53% for everolimus/letrozole compared with 43% for tamoxifen/medroxyprogesterone acetate, and progression-free survival was 21.6 months in the everolimus/letrozole arm but only 6.6 months in the medroxyprogesterone acetate/tamoxifen arm.<span><sup>25</sup></span> Given the limitations of cross-trial comparisons, it is difficult to determine whether an endocrine regimen may be similar to front-line chemotherapy in patients with endocrine-sensitive tumors. A head-to-head trial of front-line chemotherapy versus endocrine therapy seems unlikely at this point, but we believe that there is a subset of patients with hormone receptor-positive endometrioid tumors for whom first-line endocrine therapy may be appropriate and may be as effective as and better tolerated than chemotherapy.</p><p>Most ongoing trials are looking at novel combinations that include hormonal therapy rather than new single-agent hormonal therapies. Several studies are evaluating the use of cyclin-dependent kinase inhibitors (palbociclib, ribociclib) in combination with letrozole (Clinicaltrials.gov identifiers NCT03675893, NCT02730429, NCT03008408, and NCT04049227) or fulvestrant (Clinicaltrials.gov identifier NCT03643510). A recent window-of-opportunity trial evaluating the combination of medroxyprogesterone acetate with entinostat (a histone deacetylase inhibitor) showed no difference in PR expression from before to after treatment, although some decrease in Ki-67 expression was seen, suggesting that this combination could be of interest in the future.<span><sup>27</sup></span></p><p>As in breast and prostate cancer, hormones play an important role in endometrial cancer development and treatment. During the menstrual cycle, proliferative effects of estrogens on the endometrium are counteracted by progestins in a strictly regulated balance. In case of excessive estrogen secretion, proliferative effects may not be sufficiently controlled by progesterone, and endometrial cancer can develop; unopposed estrogen exposure in the postmenopausal setting also increases the risk for endometrial cancer.<span><sup>28</sup></span> In 1961, Kelley and Baker first described the use of synthetic progestins to treat 21 patients with recurrent endometrial cancer.<span><sup>29</sup></span> Later, other hormonal drugs, including tamoxifen and aromatase inhibitors, were shown to exhibit antitumor effects in endometrial cancer. The introduction of endocrine treatment also initiated efforts to identify predictive biomarkers for the selection of patients who would benefit most from such therapy, which is generally less toxic than chemotherapy.</p><p>At the time of progression on letrozole, the patient remained asymptomatic and still did not desire chemotherapy. Further testing on the tumor was performed at this time because mismatch repair (MMR) testing was not standard at the time of her original presentation. The endometrial carcinoma showed loss of expression for MLH1 (Fig. 1D) and PMS2, retained expression for MSH2 and MSH6, and exhibited <i>MLH1</i> promoter hypermethylation. Next-generation sequencing (NGS) showed mutations in <i>MAP2K1</i> (<i>MEK1</i>), <i>PIK3CA</i>, <i>PTEN</i>, and <i>MED12</i>. She was switched to single-agent pembrolizumab. She did not experience any toxicity and had further disease shrinkage (Fig. 3); after 2 years of therapy, there is no evidence of progression.</p><p>The National Comprehensive Cancer Network currently recommends universal MMR protein/microsatellite instability (MSI) testing for all endometrial carcinomas,<span><sup>44</sup></span> regardless of patient age, personal/family history, or tumor subtype. Approximately 30% of endometrial carcinomas are MMR-deficient, thus identifying this subset helps screen for Lynch syndrome (2%-6% of MMR-deficient tumors),<span><sup>45, 46</sup></span> guide management based on molecular classification,<span><sup>47</sup></span> and recognize patients who are eligible for targeted therapy with immune checkpoint inhibitors.<span><sup>48</sup></span> The 2 techniques for detecting MMR deficiency include immunohistochemistry for MMR proteins (MLH1, PMS2, MSH2, and MSH6) and molecular testing for MSI. Because these 2 methods are highly concordant (94%; <i>P</i> &lt; .001),<span><sup>49</sup></span> immunohistochemistry is generally the preferred screening test because of its cost effectiveness and accessibility.</p><p>MMR proteins repair DNA base-base mismatches and small insertion-deletions (including those that alter microsatellite length) to prevent propagation of these mutations. They form heterodimers, pairing MLH1 with PMS2 and pairing MSH2 with MSH6. PMS2 and MSH6 must bind to their respective partners; however, MLH1 and MSH2 can form heterodimers with other proteins.<span><sup>50</sup></span> In normal cells as well as MMR-proficient carcinomas, all 4 MMR proteins show diffuse (typically strong) nuclear expression, whereas defects in one or more proteins result in loss of nuclear staining for that protein(s). Four abnormal staining patterns may be observed: 1) loss of MLH1 and PMS2, 2) loss of PMS2, 3) loss of MSH2 and MSH6, and 4) loss of MSH6. All patterns may occur in either the germline or somatic setting, but combined MLH1 and PMS2 loss is generally a result of sporadic <i>MLH1</i> promoter hypermethylation, which can be confirmed by methylation-specific polymerase chain reaction (PCR)/multiplex ligation-dependent probe amplification.<span><sup>51</sup></span> If <i>MLH1</i> promoter hypermethylation is absent or another abnormal staining pattern is present, genetics counseling with germline testing is recommended.</p><p>Alternatively, MSI testing directly detects unstable microsatellites (ie, microsatellites with variable length between tumor cells) and is either PCR-based or NGS-based.<span><sup>52</sup></span> The PCR-based assay requires matched tumor and normal tissue, tests several (from 5 to 10) microsatellite loci, and classifies each locus as <i>stable</i> or <i>unstable</i>. The tumor is then assigned an overall MSI status (MSI-high, MSI-low, microsatellite-stable) based on the percentage of unstable loci. However, because this test was originally designed for colorectal carcinoma screening, it is not as sensitive for endometrial carcinoma,<span><sup>53</sup></span> Furthermore, PCR-based testing is less sensitive for detecting MSH6 deficiency, which is more common in endometrial carcinomas than in colorectal carcinomas.<span><sup>54, 55</sup></span> In contrast, NGS-based tests can be performed on tumor without matched normal tissue, evaluate hundreds of microsatellite loci, and result in a quantitative score, which then can be grouped into 3 categories: MSI-H, MSI-indeterminate, and microsatellite-stable.<span><sup>52</sup></span> Although NGS-based MSI testing circumvents many of the shortcomings of PCR-based assays, NGS is often costly and has a longer turnaround time than the other modalities.</p><p>The Cancer Genome Atlas categorized endometrial cancers into 4 distinct molecular subtypes: <i>POLE</i>/ultramutated, MSI-H, copy number low, and copy number high.<span><sup>56</sup></span> These subtypes have been correlated with the risk of recurrence. Up to 30% of primary and recurrent endometrial tumors are MSI-H (MMR-deficient).<span><sup>57, 58</sup></span> Cancers in the MSI-H category, as discussed above, have a deficiency in their DNA MMR proteins, which leads to a high DNA mutational burden. It is believed that this increases the tumor immunogenicity by creating more neoantigens that can be identified and targeted by T cells and is responsible for the efficacy of immune checkpoint inhibitors for the treatment of these tumors.<span><sup>59</sup></span> Immune checkpoint inhibitors are also likely to be effective in the treatment of ultramutated <i>POLE</i>-mutant tumors, although these are less commonly found in the metastatic setting.<span><sup>60</sup></span></p><p>Pembrolizumab, the first site-agnostic therapy approved by the US Food and Drug Administration (FDA), was approved in 2017 for the treatment MSI-H/MMR-deficient solid tumors<span><sup>61</sup></span> regardless of tumor origin. It targets the PD-1 (programmed death-1) receptor on the surface of T cells, thereby blocking the immune-suppressing ligands PD-L1 and PD-L2 (programmed death ligand-1 and programmed death ligand-2, respectively) on the cancer cells from interacting with PD-1 and inactivating the T cells. The KEYNOTE 158 trial (Clinicaltrials.gov identifier NCT02628067), which enrolled patients with pretreated MSI-H/MMR-deficient tumors of multiple histologies, included 49 patients with endometrial cancer. Of these, 8 patients had a complete response, and 20 had a partial response, yielding an overall response rate of 57.1%. Response duration at the time of reporting was from 2.9 to ≥27 months.<span><sup>62</sup></span> A second, small, prospective trial of pembrolizumab only in patients with MSI-H/MMR-deficient, recurrent endometrial cancers (n = 25) who had received at least one prior line of chemotherapy found an overall response rate of 58%; interestingly, the response rate was 100% in the 6 patients with somatic loss of MMR proteins and only 44% in patients with <i>MLH1</i> promotor methylation.<span><sup>63</sup></span> Subsequently, dostarlimab (an anti–PD-1 monoclonal antibody) has also been approved, both specifically for second-line treatment of MSI-H/MMR-deficient endometrial cancer and for MMR-deficient solid tumors of any histology that have progressed on or after prior therapy and for which there is no satisfactory alternative treatment option. Although immune checkpoint inhibitors can produce severe or fatal autoimmune toxicity, for many patients, such as ours, they are well tolerated.<span><sup>64, 65</sup></span></p><p>The current FDA approval for pembrolizumab in patients with MSI-H/MMR-deficient tumors, including endometrial cancer, is for those whose cancer has progressed after prior treatment and who have no satisfactory alternative treatment options. An obvious direction of research is the use of immune checkpoint inhibitors in patients with endometrial cancer who have not received prior cytotoxic therapy. On June 29, 2020, the FDA approved pembrolizumab for first-line use in the treatment of patients with MSI-H/MMR-deficient <i>colorectal cancer</i> based on results of the KEYNOTE-177 trial (Clinicaltrials.gov identifier NCT02563002), which randomized 307 patients with MSI-H/MMR-deficient colorectal cancer to receive either pembrolizumab 200 mg intravenously every 3 weeks or investigator's choice of 5-fluorouracil-based therapy with or without bevacizumab or cetuximab. Pembrolizumab was given to patients on the chemotherapy arm at the time of progression. The median progression-free survival was 16.5 months for those receiving pembrolizumab versus 8.2 months for those receiving chemotherapy (hazard ratio, 0.60; <i>P</i> = .0002). Among responders, 83% of patients who received pembrolizumab versus 35% of those who received chemotherapy had ongoing responses at 24 months.<span><sup>66</sup></span> Toxicity was lower in the pembrolizumab arm, with 56% versus 78% of patients having grade 3 adverse events, respectively. Fourteen percent of patients in the pembrolizumab group and 12% of those in the chemotherapy group discontinued therapy because of adverse events. Health-related quality of life showed a clinically meaningful improvement from baseline to week 18 in scores on the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire Core 30 global health status/quality of life for pembrolizumab versus chemotherapy (<i>P</i> = .0002), and the median time to deterioration for global health status/quality of life was longer with pembrolizumab than with chemotherapy (hazard ratio, 0.61; <i>P</i> = .019).<span><sup>67</sup></span> This paradigm (chemotherapy vs single-agent immunotherapy up front in MSI-H/MMR-deficient tumors) is also being explored in patients with endometrial cancer. The GOG-3064/ENGOT-en15 trial should open shortly: this is a phase 3 randomized trial of pembrolizumab versus platinum doublet chemotherapy limited to participants with MMR-deficient advanced or recurrent endometrial carcinoma in the first-line setting.</p><p>Although single-agent immune checkpoint inhibitors produce a low response rate in patients with advanced endometrial cancer that is <i>not</i> MSI-H/MMR-deficient, a combination therapy, pembrolizumab plus lenvatinib (a tyrosine kinase inhibitor targeting vascular endothelial growth factor receptor as well as other receptor tyrosine kinases) has recently been FDA-approved in this setting. A response rate of 36.2% was reported with the combination for microsatellite-stable tumors.<span><sup>68</sup></span> The addition of pembrolizumab to standard front-line chemotherapy (paclitaxel and carboplatin) is also being investigated (Clinicaltrials.gov identifier NCT03914612) in a randomized trial as is the addition of dostarlimab (Clinicaltrials.gov identifier NCT03981796)<span><sup>69, 70</sup></span> and durvalumab (Clinicaltrials.gov identifier NCT04269200); all of these trials are open to patients regardless of MSI tumor status. It is hoped that the results of these trials will help improve outcomes for women with advanced endometrial cancer.</p><p>Gini F. Fleming's institution receives funding for institutional trials from Tesaro/GSK, Compugen, Incyte, AbbVie, Eisai, Celldex, AstraZeneca, Corcept, Plexxicon, and Astellas for which she serves as principal investigator; and honoraria from UpToDate outside the submitted work. Katherine C. Kurnit participate on a Data Safety Monitoring Board or Advisory Board at LEAP Therapeutics through the Gynecologic Oncology Group Foundation outside the submitted work. Jennifer A. Bennett, Kathryn A. Mills, and Willem Jan vanWeelden made no disclosures.</p>","PeriodicalId":137,"journal":{"name":"CA: A Cancer Journal for Clinicians","volume":"72 2","pages":"102-111"},"PeriodicalIF":503.1000,"publicationDate":"2022-01-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.3322/caac.21715","citationCount":"2","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"CA: A Cancer Journal for Clinicians","FirstCategoryId":"3","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.3322/caac.21715","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"ONCOLOGY","Score":null,"Total":0}
引用次数: 2

Abstract

A 65-year-old woman presented to an outside hospital with vaginal bleeding in 2009. Dilatation and curettage showed endometrial endometrioid carcinoma, International Federation of Gynecology and Obstetrics (FIGO) grade 2. She underwent exploratory laparotomy with pelvic washings, total abdominal hysterectomy with bilateral salpingo-oophorectomy, bilateral pelvic and para-aortic lymphadenectomy, and an omental biopsy. Pathology was read as uterine carcinosarcoma, homologous type, invading 3.5 cm into a 4-cm-thick myometrium, with involvement of the endocervical epithelium (no stromal involvement) and lymphovascular invasion. All 14 of the sampled lymph nodes (6 left pelvic, 1 left common iliac, 1 left aortic, 3 right pelvic, 2 right common iliac, and 1 right aortic) were negative for carcinoma. The tumor-node-metastasis (TNM) pathologic stage using current staging (2018 FIGO Cancer Report1) was pT1b N0 American Joint Committee on Cancer [AJCC] Cancer Staging Manual, eighth edition), FIGO stage IB. Postoperative positron emission tomography/computed tomography (CT) imaging studies demonstrated no evidence of disease.

The patient was otherwise healthy. Her father had lung cancer, but there was no other history of cancer in the family. She received pelvic radiotherapy and 4 cycles of paclitaxel 135 mg/m2 plus ifosfamide 1600 mg/m2, with mesna and granulocyte-colony–stimulating factor support.

Five years later (at age 71 years), she developed abdominal pain and vomiting and was managed conservatively for a partial small bowel obstruction. CT scans of the abdomen and pelvis showed a large, heterogeneously enhancing mass in the left suprarenal space, measuring 8.9 × 8.6 cm, likely originating from the left adrenal gland. A lobular mass in the posterior left lung base measuring 3.2 × 1.6 cm was also noted. A CT scan of the chest revealed multiple, bilateral pulmonary nodules, the largest measuring 2.4 × 2.2 cm, some with central cavitation, most consistent with metastatic disease. A fine-needle aspirate of the suprarenal mass showed poorly differentiated carcinoma with extensive necrosis.

At that time, the patient presented to a university hospital. Review of the original pathology reclassified the tumor as endometrial endometrioid carcinoma, FIGO grade 3 (Fig. 1A), with the suprarenal mass morphologically consistent with the patient's primary. Estrogen receptor (ER) and progesterone receptor (PR) were strongly and diffusely positive (Fig. 1B,C).

Subtyping of high-grade endometrial carcinomas is diagnostically challenging for pathologists, with even expert gynecologic pathologists reaching consensus in only 63% to 72% of tumors.2, 3 The most common disputes include endometrioid versus serous carcinoma, serous versus clear cell carcinoma, and endometrioid/clear cell/undifferentiated carcinoma versus carcinosarcoma.2 Morphologically, high-grade endometrial carcinoma can show extensive solid growth, whereas low-grade endometrioid carcinoma may have a spindled morphology,4 both of which may mimic carcinosarcoma with a homologous sarcomatous component. Although immunohistochemistry can be useful in subtyping endometrioid, serous, and clear cell carcinomas, it is generally unhelpful for distinguishing carcinosarcomas from pure endometrial carcinomas. Because carcinosarcomas are considered a subtype of endometrial carcinoma5 rather than a mixed epithelial and mesenchymal tumor, as previously hypothesized, it is not surprising that the sarcoma (as well as the carcinoma) portions of the tumor may show cytokeratin and PAX8 expression, eliminating the utility of these stains in differentiating between carcinosarcoma and pure endometrial carcinoma.6, 7

The College of American Pathologists recommends performing ER immunohistochemistry on stage III, IV, and recurrent endometrial carcinomas to predict response to endocrine therapy.8 Unlike in breast carcinoma, there is not a uniform reporting system for hormone receptors in endometrial carcinoma; however, it is recommended that the number of positive cells and staining intensity be documented.9

The patient was presented with the option of chemotherapy with carboplatin plus paclitaxel. She declined chemotherapy. She was also presented with the option of participation in a clinical trial, Gynecologic Oncology Group (GOG) 3007 (a randomized phase II trial of everolimus and letrozole or hormonal therapy [medroxyprogesterone acetate/tamoxifen], in women with advanced, persistent, or recurrent endometrial cancer; Clinicaltrials.gov identifier NCT02228681). She elected to participate in GOG 3007 and was randomized to therapy with medroxyprogesterone acetate/tamoxifen. She was treated with oral tamoxifen 20 mg twice daily (continuously), with the addition of medroxyprogesterone acetate 200 mg daily on alternating weeks (days 8-14 and 22-28 of a 28-day cycle). She had a partial response to therapy (Fig. 2) and remained on study for 19 cycles (about 1.5 years), at which time she had mild disease progression in the lung. She was switched to single-agent letrozole without further response.

Of note, had her tumor not been reclassified as an endometrioid endometrial carcinoma, she would have been ineligible for GOG 3007 because that trial, as well as many current ongoing trials for women with endometrial cancer (including those testing immunotherapy), excludes carcinosarcomas.

Whereas, historically, progestins were front-line therapy for metastatic endometrial cancer, currently, they are most commonly used in the second or later lines, although it is not clear that their activity is fully maintained in this setting. Single-agent progestins (most frequently oral medroxyprogesterone acetate or megestrol acetate),10, 11 aromatase inhibitors (letrozole, anastrozole),12, 13 selective ER modulators (tamoxifen, arzoxifene),14, 15 fulvestrant,16, 17 or combination therapies, such as progestin/tamoxifen18, 19 or everolimus (an mTOR inhibitor)/letrozole,20, 21 have all been investigated and have shown some efficacy. A 2017 meta-analysis evaluated 16 studies that included second-line endocrine therapy and reported an objective response rate of 18.5%.22 When patients with stable disease were also included, the clinical benefit rate was 36%. By comparison, a randomized trial of tamoxifen versus anastrazole for first-line therapy of metastatic breast cancer (almost one-half of patients had hormone receptor-positive tumors and, in slightly greater than one-half, the tumor hormone receptor status was unknown) demonstrated an objective response rate of approximately 33% in both arms.23 The current National Comprehensive Cancer Network guidelines include multiple endocrine therapy options for endometrial cancer, but they note that these therapies are typically reserved for patients who have low-grade endometrioid tumors with more indolent behavior.24

More recently, the combination of everolimus plus letrozole and the combination of tamoxifen plus medroxyprogesterone acetate were evaluated in GOG 3007, the study in which our patient enrolled. Although this was a randomized, phase 2 study of women with advanced or recurrent endometrial cancer and included both first-line and second-line settings, it is important to note that the 2 regimens were not directly compared. Preliminary results were presented at the 2018 Society for Gynecologic Oncology Annual Meeting. The progression-free survival was 6.3 months for the everolimus/letrozole arm and 3.8 months for the medroxyprogesterone acetate/tamoxifen arm.25 However, in patients who had received prior chemotherapy, progression-free survival was 3.3 months and 3.2 months, respectively. Overall survival for the medroxyprogesterone acetate/tamoxifen group was 16.6 months but was not yet calculable for the everolimus/letrozole group. In addition, a phase 2 trial of letrozole plus ribociclib (a cyclin-dependent kinase inhibitor) was recently published. Of the 20 patients with endometrial cancer enrolled on the trial, 55% were still on treatment at 12 weeks.26 Similar to the studies discussed above, those patients who had low-grade endometrioid tumors seemed to derive the most benefit.

Discussion regarding the use of endocrine therapy before chemotherapy in the metastatic setting is ongoing. A 2017 meta-analysis evaluated results from 23 studies of hormonal therapy in the first-line setting and found that the overall response rate (ORR) was highest in patients who had hormone receptor-positive tumors, at 32.5%,22 and that low-grade tumors had higher response rates than high-grade tumors. The ORR for ER-negative tumors was only 9.2%. The authors also reported that single-agent aromatase inhibitors produced the lowest response rates of the endocrine therapy classes, with a mean ORR of only 8.6%. For patients treated on GOG 3007 in the first-line setting, the objective response rate was 53% for everolimus/letrozole compared with 43% for tamoxifen/medroxyprogesterone acetate, and progression-free survival was 21.6 months in the everolimus/letrozole arm but only 6.6 months in the medroxyprogesterone acetate/tamoxifen arm.25 Given the limitations of cross-trial comparisons, it is difficult to determine whether an endocrine regimen may be similar to front-line chemotherapy in patients with endocrine-sensitive tumors. A head-to-head trial of front-line chemotherapy versus endocrine therapy seems unlikely at this point, but we believe that there is a subset of patients with hormone receptor-positive endometrioid tumors for whom first-line endocrine therapy may be appropriate and may be as effective as and better tolerated than chemotherapy.

Most ongoing trials are looking at novel combinations that include hormonal therapy rather than new single-agent hormonal therapies. Several studies are evaluating the use of cyclin-dependent kinase inhibitors (palbociclib, ribociclib) in combination with letrozole (Clinicaltrials.gov identifiers NCT03675893, NCT02730429, NCT03008408, and NCT04049227) or fulvestrant (Clinicaltrials.gov identifier NCT03643510). A recent window-of-opportunity trial evaluating the combination of medroxyprogesterone acetate with entinostat (a histone deacetylase inhibitor) showed no difference in PR expression from before to after treatment, although some decrease in Ki-67 expression was seen, suggesting that this combination could be of interest in the future.27

As in breast and prostate cancer, hormones play an important role in endometrial cancer development and treatment. During the menstrual cycle, proliferative effects of estrogens on the endometrium are counteracted by progestins in a strictly regulated balance. In case of excessive estrogen secretion, proliferative effects may not be sufficiently controlled by progesterone, and endometrial cancer can develop; unopposed estrogen exposure in the postmenopausal setting also increases the risk for endometrial cancer.28 In 1961, Kelley and Baker first described the use of synthetic progestins to treat 21 patients with recurrent endometrial cancer.29 Later, other hormonal drugs, including tamoxifen and aromatase inhibitors, were shown to exhibit antitumor effects in endometrial cancer. The introduction of endocrine treatment also initiated efforts to identify predictive biomarkers for the selection of patients who would benefit most from such therapy, which is generally less toxic than chemotherapy.

At the time of progression on letrozole, the patient remained asymptomatic and still did not desire chemotherapy. Further testing on the tumor was performed at this time because mismatch repair (MMR) testing was not standard at the time of her original presentation. The endometrial carcinoma showed loss of expression for MLH1 (Fig. 1D) and PMS2, retained expression for MSH2 and MSH6, and exhibited MLH1 promoter hypermethylation. Next-generation sequencing (NGS) showed mutations in MAP2K1 (MEK1), PIK3CA, PTEN, and MED12. She was switched to single-agent pembrolizumab. She did not experience any toxicity and had further disease shrinkage (Fig. 3); after 2 years of therapy, there is no evidence of progression.

The National Comprehensive Cancer Network currently recommends universal MMR protein/microsatellite instability (MSI) testing for all endometrial carcinomas,44 regardless of patient age, personal/family history, or tumor subtype. Approximately 30% of endometrial carcinomas are MMR-deficient, thus identifying this subset helps screen for Lynch syndrome (2%-6% of MMR-deficient tumors),45, 46 guide management based on molecular classification,47 and recognize patients who are eligible for targeted therapy with immune checkpoint inhibitors.48 The 2 techniques for detecting MMR deficiency include immunohistochemistry for MMR proteins (MLH1, PMS2, MSH2, and MSH6) and molecular testing for MSI. Because these 2 methods are highly concordant (94%; P < .001),49 immunohistochemistry is generally the preferred screening test because of its cost effectiveness and accessibility.

MMR proteins repair DNA base-base mismatches and small insertion-deletions (including those that alter microsatellite length) to prevent propagation of these mutations. They form heterodimers, pairing MLH1 with PMS2 and pairing MSH2 with MSH6. PMS2 and MSH6 must bind to their respective partners; however, MLH1 and MSH2 can form heterodimers with other proteins.50 In normal cells as well as MMR-proficient carcinomas, all 4 MMR proteins show diffuse (typically strong) nuclear expression, whereas defects in one or more proteins result in loss of nuclear staining for that protein(s). Four abnormal staining patterns may be observed: 1) loss of MLH1 and PMS2, 2) loss of PMS2, 3) loss of MSH2 and MSH6, and 4) loss of MSH6. All patterns may occur in either the germline or somatic setting, but combined MLH1 and PMS2 loss is generally a result of sporadic MLH1 promoter hypermethylation, which can be confirmed by methylation-specific polymerase chain reaction (PCR)/multiplex ligation-dependent probe amplification.51 If MLH1 promoter hypermethylation is absent or another abnormal staining pattern is present, genetics counseling with germline testing is recommended.

Alternatively, MSI testing directly detects unstable microsatellites (ie, microsatellites with variable length between tumor cells) and is either PCR-based or NGS-based.52 The PCR-based assay requires matched tumor and normal tissue, tests several (from 5 to 10) microsatellite loci, and classifies each locus as stable or unstable. The tumor is then assigned an overall MSI status (MSI-high, MSI-low, microsatellite-stable) based on the percentage of unstable loci. However, because this test was originally designed for colorectal carcinoma screening, it is not as sensitive for endometrial carcinoma,53 Furthermore, PCR-based testing is less sensitive for detecting MSH6 deficiency, which is more common in endometrial carcinomas than in colorectal carcinomas.54, 55 In contrast, NGS-based tests can be performed on tumor without matched normal tissue, evaluate hundreds of microsatellite loci, and result in a quantitative score, which then can be grouped into 3 categories: MSI-H, MSI-indeterminate, and microsatellite-stable.52 Although NGS-based MSI testing circumvents many of the shortcomings of PCR-based assays, NGS is often costly and has a longer turnaround time than the other modalities.

The Cancer Genome Atlas categorized endometrial cancers into 4 distinct molecular subtypes: POLE/ultramutated, MSI-H, copy number low, and copy number high.56 These subtypes have been correlated with the risk of recurrence. Up to 30% of primary and recurrent endometrial tumors are MSI-H (MMR-deficient).57, 58 Cancers in the MSI-H category, as discussed above, have a deficiency in their DNA MMR proteins, which leads to a high DNA mutational burden. It is believed that this increases the tumor immunogenicity by creating more neoantigens that can be identified and targeted by T cells and is responsible for the efficacy of immune checkpoint inhibitors for the treatment of these tumors.59 Immune checkpoint inhibitors are also likely to be effective in the treatment of ultramutated POLE-mutant tumors, although these are less commonly found in the metastatic setting.60

Pembrolizumab, the first site-agnostic therapy approved by the US Food and Drug Administration (FDA), was approved in 2017 for the treatment MSI-H/MMR-deficient solid tumors61 regardless of tumor origin. It targets the PD-1 (programmed death-1) receptor on the surface of T cells, thereby blocking the immune-suppressing ligands PD-L1 and PD-L2 (programmed death ligand-1 and programmed death ligand-2, respectively) on the cancer cells from interacting with PD-1 and inactivating the T cells. The KEYNOTE 158 trial (Clinicaltrials.gov identifier NCT02628067), which enrolled patients with pretreated MSI-H/MMR-deficient tumors of multiple histologies, included 49 patients with endometrial cancer. Of these, 8 patients had a complete response, and 20 had a partial response, yielding an overall response rate of 57.1%. Response duration at the time of reporting was from 2.9 to ≥27 months.62 A second, small, prospective trial of pembrolizumab only in patients with MSI-H/MMR-deficient, recurrent endometrial cancers (n = 25) who had received at least one prior line of chemotherapy found an overall response rate of 58%; interestingly, the response rate was 100% in the 6 patients with somatic loss of MMR proteins and only 44% in patients with MLH1 promotor methylation.63 Subsequently, dostarlimab (an anti–PD-1 monoclonal antibody) has also been approved, both specifically for second-line treatment of MSI-H/MMR-deficient endometrial cancer and for MMR-deficient solid tumors of any histology that have progressed on or after prior therapy and for which there is no satisfactory alternative treatment option. Although immune checkpoint inhibitors can produce severe or fatal autoimmune toxicity, for many patients, such as ours, they are well tolerated.64, 65

The current FDA approval for pembrolizumab in patients with MSI-H/MMR-deficient tumors, including endometrial cancer, is for those whose cancer has progressed after prior treatment and who have no satisfactory alternative treatment options. An obvious direction of research is the use of immune checkpoint inhibitors in patients with endometrial cancer who have not received prior cytotoxic therapy. On June 29, 2020, the FDA approved pembrolizumab for first-line use in the treatment of patients with MSI-H/MMR-deficient colorectal cancer based on results of the KEYNOTE-177 trial (Clinicaltrials.gov identifier NCT02563002), which randomized 307 patients with MSI-H/MMR-deficient colorectal cancer to receive either pembrolizumab 200 mg intravenously every 3 weeks or investigator's choice of 5-fluorouracil-based therapy with or without bevacizumab or cetuximab. Pembrolizumab was given to patients on the chemotherapy arm at the time of progression. The median progression-free survival was 16.5 months for those receiving pembrolizumab versus 8.2 months for those receiving chemotherapy (hazard ratio, 0.60; P = .0002). Among responders, 83% of patients who received pembrolizumab versus 35% of those who received chemotherapy had ongoing responses at 24 months.66 Toxicity was lower in the pembrolizumab arm, with 56% versus 78% of patients having grade 3 adverse events, respectively. Fourteen percent of patients in the pembrolizumab group and 12% of those in the chemotherapy group discontinued therapy because of adverse events. Health-related quality of life showed a clinically meaningful improvement from baseline to week 18 in scores on the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire Core 30 global health status/quality of life for pembrolizumab versus chemotherapy (P = .0002), and the median time to deterioration for global health status/quality of life was longer with pembrolizumab than with chemotherapy (hazard ratio, 0.61; P = .019).67 This paradigm (chemotherapy vs single-agent immunotherapy up front in MSI-H/MMR-deficient tumors) is also being explored in patients with endometrial cancer. The GOG-3064/ENGOT-en15 trial should open shortly: this is a phase 3 randomized trial of pembrolizumab versus platinum doublet chemotherapy limited to participants with MMR-deficient advanced or recurrent endometrial carcinoma in the first-line setting.

Although single-agent immune checkpoint inhibitors produce a low response rate in patients with advanced endometrial cancer that is not MSI-H/MMR-deficient, a combination therapy, pembrolizumab plus lenvatinib (a tyrosine kinase inhibitor targeting vascular endothelial growth factor receptor as well as other receptor tyrosine kinases) has recently been FDA-approved in this setting. A response rate of 36.2% was reported with the combination for microsatellite-stable tumors.68 The addition of pembrolizumab to standard front-line chemotherapy (paclitaxel and carboplatin) is also being investigated (Clinicaltrials.gov identifier NCT03914612) in a randomized trial as is the addition of dostarlimab (Clinicaltrials.gov identifier NCT03981796)69, 70 and durvalumab (Clinicaltrials.gov identifier NCT04269200); all of these trials are open to patients regardless of MSI tumor status. It is hoped that the results of these trials will help improve outcomes for women with advanced endometrial cancer.

Gini F. Fleming's institution receives funding for institutional trials from Tesaro/GSK, Compugen, Incyte, AbbVie, Eisai, Celldex, AstraZeneca, Corcept, Plexxicon, and Astellas for which she serves as principal investigator; and honoraria from UpToDate outside the submitted work. Katherine C. Kurnit participate on a Data Safety Monitoring Board or Advisory Board at LEAP Therapeutics through the Gynecologic Oncology Group Foundation outside the submitted work. Jennifer A. Bennett, Kathryn A. Mills, and Willem Jan vanWeelden made no disclosures.

查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
肿瘤委员会报告1例转移性、激素受体阳性、错配修复缺陷的子宫内膜癌
一名65岁妇女于2009年因阴道出血到外院就诊。子宫内膜扩张刮除显示子宫内膜样癌,国际妇产科学联合会(FIGO) 2级。她接受了探查性剖腹手术和盆腔清洗,全腹子宫切除术和双侧输卵管卵巢切除术,双侧盆腔和主动脉旁淋巴结切除术,以及大网膜活检。病理:子宫癌肉瘤,同源型,浸润3.5 cm至4 cm厚肌层,累及宫颈内上皮(未累及间质),淋巴血管浸润。所有14个淋巴结(6个左盆腔淋巴结,1个左髂总淋巴结,1个左主动脉淋巴结,3个右盆腔淋巴结,2个右髂总淋巴结,1个右主动脉淋巴结)均阴性。采用当前分期(2018年FIGO癌症报告1)的肿瘤-淋巴结-转移(TNM)病理分期为pT1b N0美国癌症联合委员会[AJCC]癌症分期手册,第八版),FIGO分期为IB期。术后正电子发射断层扫描/计算机断层扫描(CT)成像研究未发现疾病证据。病人在其他方面都很健康。她的父亲患有肺癌,但家族中没有其他癌症病史。患者接受盆腔放疗,紫杉醇135 mg/m2 +异环磷酰胺1600 mg/m2治疗4个周期,网膜和粒细胞集落刺激因子支持。5年后(71岁),患者出现腹痛和呕吐,因部分性小肠梗阻接受保守治疗。腹部和骨盆CT扫描显示左侧肾上间隙有一个巨大的非均匀强化肿块,大小为8.9 × 8.6 cm,可能起源于左侧肾上腺。左后肺底小叶肿块,尺寸为3.2 × 1.6 cm。胸部CT扫描显示多发双侧肺结节,最大的为2.4 × 2.2 cm,部分伴中央空化,最符合转移性疾病。细针穿刺肾上肿块显示低分化癌伴广泛坏死。当时,病人被送到了一所大学医院。对原始病理的回顾将肿瘤重新分类为子宫内膜子宫内膜样癌,FIGO 3级(图1A),肾上肿块在形态上与患者原发灶一致。雌激素受体(ER)和孕激素受体(PR)呈强烈且弥漫性阳性(图1B,C)。对于病理学家来说,高级别子宫内膜癌的亚型诊断具有挑战性,即使是专家妇科病理学家也只有63%至72%的肿瘤达成共识。最常见的争议包括子宫内膜样癌与浆液性癌、浆液性癌与透明细胞癌、子宫内膜样癌/透明细胞癌/未分化癌与癌肉瘤形态学上,高级别子宫内膜癌可表现为广泛的实体生长,而低级别子宫内膜样癌可表现为纺锤形形态,4这两种形态都可能类似于具有同源肉瘤成分的癌肉瘤。虽然免疫组织化学可用于子宫内膜样癌、浆液性癌和透明细胞癌的分型,但它通常无助于区分癌肉瘤和纯子宫内膜癌。由于癌肉瘤被认为是子宫内膜癌的一种亚型,而不是先前假设的上皮和间质混合肿瘤,因此肿瘤的肉瘤(以及癌)部分可能显示细胞角蛋白和PAX8表达并不奇怪,消除了这些染色在鉴别癌肉瘤和纯子宫内膜癌中的作用。美国病理学家协会推荐对III期、IV期和复发性子宫内膜癌进行ER免疫组化,以预测对内分泌治疗的反应与乳腺癌不同,子宫内膜癌的激素受体没有统一的报告系统;但是,建议记录阳性细胞的数量和染色强度。患者可选择卡铂加紫杉醇化疗。她拒绝化疗。她还可以选择参加临床试验,妇科肿瘤组(GOG) 3007(依维莫司和来曲唑或激素治疗[醋酸甲孕酮/他莫昔芬]的随机II期试验),用于晚期、持续性或复发性子宫内膜癌的妇女;Clinicaltrials.gov识别码NCT02228681)。她选择参加GOG 3007,并随机接受醋酸甲孕酮/他莫昔芬治疗。患者口服他莫昔芬20 mg,每日2次(连续),同时加用醋酸甲孕酮200 mg,每隔一周(8-14天和22-28天,每28天一个周期)。她对治疗有部分反应(图2),并继续研究了19个周期(约1个周期)。 5年),当时她的肺部有轻微的疾病进展。她转而使用单药来曲唑,没有进一步的反应。值得注意的是,如果她的肿瘤没有被重新分类为子宫内膜样子宫内膜癌,她将没有资格参加GOG 3007试验,因为该试验以及许多目前正在进行的针对子宫内膜癌女性的试验(包括那些测试免疫疗法的试验)不包括癌肉瘤。然而,从历史上看,孕激素是转移性子宫内膜癌的一线治疗方法,目前,它们最常用于二线或二线以上,尽管尚不清楚它们的活性在这种情况下是否完全维持。单药孕激素(最常见的是口服醋酸甲羟孕酮或醋酸甲地孕酮),10,11芳香化酶抑制剂(来曲唑,阿那曲唑),12,13选择性内质酶调节剂(他莫昔芬,阿唑昔芬),14,15氟维司汀,16,17或联合治疗,如黄体激素/他莫昔芬18,19或依维莫司(一种mTOR抑制剂)/来曲唑,20,21都已被研究并显示出一定的疗效。2017年的一项荟萃分析评估了16项纳入二线内分泌治疗的研究,结果显示客观缓解率为18.5%当病情稳定的患者也纳入时,临床获益率为36%。通过比较,他莫昔芬和阿纳唑用于转移性乳腺癌一线治疗的随机试验(几乎一半的患者患有激素受体阳性肿瘤,略多于一半的患者肿瘤激素受体状态未知)显示,两组的客观反应率约为33% 23目前的国家综合癌症网络指南包括子宫内膜癌的多种内分泌治疗方案,但他们指出,这些治疗通常是为低级别子宫内膜样肿瘤患者保留的,这些患者的行为更懒惰。最近,依维莫司联合来曲唑和他莫昔芬联合醋酸甲孕酮在GOG 3007中进行了评估,该研究纳入了本患者。尽管这是一项针对晚期或复发子宫内膜癌女性的随机二期研究,包括一线和二线治疗,但需要注意的是,这两种治疗方案并没有直接进行比较。初步结果在2018年妇科肿瘤学会年会上公布。依维莫司/来曲唑组无进展生存期为6.3个月,醋酸甲孕酮/他莫昔芬组为3.8个月然而,在接受过化疗的患者中,无进展生存期分别为3.3个月和3.2个月。醋酸甲孕酮/他莫昔芬组的总生存期为16.6个月,但依维莫司/来曲唑组的总生存期尚未计算出来。此外,最近发表了一项来曲唑加核糖环尼(一种细胞周期蛋白依赖性激酶抑制剂)的2期试验。在参加试验的20名子宫内膜癌患者中,55%的患者在12周时仍在接受治疗与上述研究类似,低级别子宫内膜样肿瘤患者似乎获益最多。关于转移性肿瘤化疗前使用内分泌治疗的讨论正在进行中。2017年的一项荟萃分析评估了一线环境中23项激素治疗研究的结果,发现激素受体阳性肿瘤患者的总缓解率(ORR)最高,为32.5%,22并且低级别肿瘤的缓解率高于高级别肿瘤。er阴性肿瘤的ORR仅为9.2%。作者还报道了单药芳香化酶抑制剂在内分泌治疗类别中产生最低的应答率,平均ORR仅为8.6%。在一线接受GOG 3007治疗的患者中,依维莫司/来曲唑的客观缓解率为53%,而他莫昔芬/醋酸羟孕酮的客观缓解率为43%,依维莫司/来曲唑组的无进展生存期为21.6个月,醋酸羟孕酮/他莫昔芬组的无进展生存期仅为6.6个月鉴于交叉试验比较的局限性,很难确定内分泌方案是否与内分泌敏感肿瘤患者的一线化疗相似。在这一点上,一线化疗与内分泌治疗的正面试验似乎不太可能,但我们相信有一部分激素受体阳性子宫内膜样肿瘤患者,一线内分泌治疗可能是合适的,可能与化疗一样有效,而且耐受性更好。大多数正在进行的试验都在寻找包括激素治疗在内的新组合,而不是新的单药激素治疗。一些研究正在评估周期蛋白依赖性激酶抑制剂(palbociclib, ribociclib)与来曲唑联合使用的情况(临床试验)。 NCT03675893、NCT02730429、NCT03008408和NCT04049227)或fulvestrant (Clinicaltrials.gov标识符NCT03643510)。最近一项评估醋酸甲羟孕酮与恩替诺他(一种组蛋白去乙酰化酶抑制剂)联合使用的机会之窗试验显示,治疗前后PR表达没有差异,尽管Ki-67表达有所下降,这表明这种联合治疗在未来可能会引起人们的兴趣。与乳腺癌和前列腺癌一样,激素在子宫内膜癌的发展和治疗中也起着重要作用。在月经周期中,雌激素对子宫内膜的增殖作用被黄体酮在严格调节的平衡中抵消。雌激素分泌过多时,孕激素不能充分控制增殖作用,可发生子宫内膜癌;绝经后无对抗性雌激素暴露也会增加患子宫内膜癌的风险1961年,Kelley和Baker首次描述了使用合成孕激素治疗21例复发性子宫内膜癌患者后来,其他激素药物,包括他莫昔芬和芳香酶抑制剂,在子宫内膜癌中显示出抗肿瘤作用。内分泌治疗的引入也开启了识别预测性生物标志物的努力,以选择从这种治疗中获益最多的患者,这种治疗通常比化疗毒性更小。在来曲唑治疗进展时,患者仍无症状,仍不愿化疗。由于错配修复(MMR)检测在她最初出现时并不标准,因此此时对肿瘤进行了进一步的检测。子宫内膜癌显示MLH1(图1D)和PMS2的表达缺失,MSH2和MSH6的表达保留,MLH1启动子超甲基化。下一代测序(NGS)显示MAP2K1 (MEK1)、PIK3CA、PTEN和MED12突变。她转而使用单药派姆单抗。患者未出现任何毒性反应,并进一步缩小(图3);治疗2年后,无进展迹象。国家综合癌症网络目前建议对所有子宫内膜癌进行通用MMR蛋白/微卫星不稳定性(MSI)检测,44与患者年龄、个人/家族史或肿瘤亚型无关。大约30%的子宫内膜癌存在mmr缺陷,因此确定这一亚群有助于筛查Lynch综合征(占mmr缺陷肿瘤的2%-6%),44,46指导基于分子分类的管理,47并识别有资格接受免疫检查点抑制剂靶向治疗的患者48检测MMR缺乏症的两种技术包括MMR蛋白(MLH1、PMS2、MSH2和MSH6)的免疫组化和MSI的分子检测。因为这两种方法的一致性很高(94%;P & lt;.001),49由于其成本效益和可及性,免疫组织化学通常是首选的筛查试验。MMR蛋白修复DNA碱基错配和小的插入缺失(包括那些改变微卫星长度的插入缺失),以防止这些突变的传播。它们形成异源二聚体,将MLH1与PMS2配对,将MSH2与MSH6配对。PMS2和MSH6必须与各自的伙伴绑定;然而,MLH1和MSH2可以与其他蛋白形成异源二聚体在正常细胞和精通MMR的癌细胞中,所有4种MMR蛋白都表现出弥漫性(通常是强的)核表达,而一种或多种蛋白的缺陷导致该蛋白的核染色丧失。可观察到四种异常染色模式:1)MLH1和PMS2缺失,2)PMS2缺失,3)MSH2和MSH6缺失,4)MSH6缺失。所有模式都可能发生在种系或体细胞环境中,但MLH1和PMS2的联合缺失通常是MLH1启动子散发性超甲基化的结果,这可以通过甲基化特异性聚合酶链反应(PCR)/多重连接依赖探针扩增来证实如果MLH1启动子超甲基化不存在或存在其他异常染色模式,建议进行生殖系检测的遗传学咨询。另外,MSI检测直接检测不稳定的微卫星(即肿瘤细胞之间长度可变的微卫星),基于pcr或基于ngs。52基于pcr的分析需要匹配肿瘤和正常组织,测试几个(从5到10)微卫星位点,并将每个位点分类为稳定或不稳定。然后根据不稳定位点的百分比为肿瘤分配总体MSI状态(MSI高、MSI低、微卫星稳定)。 然而,由于该检测最初是为结直肠癌筛查而设计的,因此对子宫内膜癌的敏感性不高。53此外,基于pcr的检测对检测MSH6缺乏症的敏感性较低,而MSH6缺乏症在子宫内膜癌中比在结直肠癌中更常见。54,55相比之下,基于ngs的检测可以在没有匹配的正常组织的肿瘤上进行,评估数百个微卫星位点,并得出定量评分,然后可以将其分为3类:MSI-H、msi -不确定和微卫星稳定52尽管基于NGS的MSI检测规避了基于pcr的分析的许多缺点,但NGS通常是昂贵的,并且比其他方式有更长的周转时间。癌症基因组图谱将子宫内膜癌分为4种不同的分子亚型:极/超突变、MSI-H、低拷贝数和高拷贝数这些亚型与复发风险相关。高达30%的原发性和复发性子宫内膜肿瘤是MSI-H (mmr缺陷)。57,58如上所述,MSI-H类癌症的DNA MMR蛋白缺乏,这导致了高DNA突变负担。据信,这增加了肿瘤的免疫原性,产生了更多的新抗原,可以被T细胞识别和靶向,这是免疫检查点抑制剂治疗这些肿瘤的有效性的原因免疫检查点抑制剂也可能对治疗超突变的极突变肿瘤有效,尽管这些在转移性肿瘤中不太常见。pembrolizumab是美国食品和药物管理局(FDA)批准的首个位点不确定疗法,于2017年被批准用于治疗MSI-H/ mmr缺陷实体肿瘤61,无论肿瘤来源如何。它靶向T细胞表面的PD-1(程序性死亡-1)受体,从而阻断癌细胞上的免疫抑制配体PD-L1和PD-L2(分别为程序性死亡配体-1和程序性死亡配体-2)与PD-1的相互作用,使T细胞失活。KEYNOTE 158试验(Clinicaltrials.gov标识符NCT02628067)纳入了49例子宫内膜癌患者,这些患者均为经预处理的多种组织学的MSI-H/ mmr缺陷肿瘤。其中,8例患者完全缓解,20例患者部分缓解,总缓解率为57.1%。报告时的反应持续时间为2.9至≥27个月另一项小型前瞻性试验发现,派姆单抗仅用于MSI-H/ mmr缺陷、复发性子宫内膜癌(n = 25)患者,这些患者之前至少接受过一次化疗,总有效率为58%;有趣的是,6例MMR蛋白体细胞缺失患者的缓解率为100%,而MLH1启动子甲基化患者的缓解率仅为44%随后,dostarlimab(一种抗pd -1单克隆抗体)也被批准,专门用于二线治疗MSI-H/ mmr缺陷子宫内膜癌和任何组织学上的mmr缺陷实体瘤,这些实体瘤在先前治疗中或之后已经进展,并且没有令人满意的替代治疗方案。尽管免疫检查点抑制剂可以产生严重或致命的自身免疫毒性,但对于许多患者,例如我们的患者,它们的耐受性良好。64,65目前FDA批准pembrolizumab用于MSI-H/ mmr缺陷肿瘤(包括子宫内膜癌)患者,用于那些在先前治疗后癌症进展且没有令人满意的替代治疗方案的患者。一个明显的研究方向是在未接受过细胞毒性治疗的子宫内膜癌患者中使用免疫检查点抑制剂。2020年6月29日,FDA根据KEYNOTE-177试验(Clinicaltrials.gov identifier NCT02563002)的结果批准派姆单抗用于一线治疗MSI-H/ mmr缺陷型结直肠癌患者,该试验随机选择307例MSI-H/ mmr缺陷型结直肠癌患者接受派姆单抗200 mg静脉注射,每3周一次,或研究者选择基于5-氟嘧啶的治疗,或不使用贝伐单抗或西妥昔单抗。Pembrolizumab在进展时给予化疗组的患者。接受派姆单抗的患者中位无进展生存期为16.5个月,而接受化疗的患者中位无进展生存期为8.2个月(风险比,0.60;P = 0.0002)。在应答者中,接受派姆单抗的患者中有83%在24个月时持续应答,而接受化疗的患者中有35%派姆单抗组的毒性较低,分别为56%和78%的患者发生3级不良事件。派姆单抗组中14%的患者和化疗组中12%的患者因不良事件而停止治疗。 欧洲癌症研究和治疗组织生活质量问卷Core 30总体健康状况/生活质量评分显示,与化疗相比,与健康相关的生活质量从基线到第18周有临床意义的改善(P = 0.0002),并且与化疗相比,派姆单抗组总体健康状况/生活质量恶化的中位时间更长(风险比,0.61;P = .019)这种模式(化疗vs单药免疫治疗预先治疗MSI-H/ mmr缺陷肿瘤)也正在子宫内膜癌患者中进行探索。GOG-3064/ENGOT-en15试验将很快开放:这是一项派姆单抗与铂双药化疗的3期随机试验,仅限于一线mmr缺陷晚期或复发子宫内膜癌患者。尽管单药免疫检查点抑制剂在非MSI-H/ mmr缺陷的晚期子宫内膜癌患者中产生低反应率,但一种联合治疗,派姆单抗加lenvatinib(一种靶向血管内皮生长因子受体和其他受体酪氨酸激酶的酪氨酸激酶抑制剂)最近已被fda批准用于这种情况。联合治疗微卫星稳定型肿瘤有效率为36.2%pembrolizumab加入标准一线化疗(紫杉醇和卡铂)也正在一项随机试验中进行研究(Clinicaltrials.gov标识符NCT03914612), dostarlimab (Clinicaltrials.gov标识符NCT03981796)69、70和durvalumab (Clinicaltrials.gov标识符NCT04269200);所有这些试验都对患者开放,无论MSI肿瘤状态如何。希望这些试验的结果将有助于改善晚期子宫内膜癌妇女的预后。Gini F. Fleming的机构接受来自Tesaro/GSK、Compugen、Incyte、AbbVie、Eisai、Celldex、AstraZeneca、concept、Plexxicon和Astellas的机构试验资金,她担任首席研究员;以及UpToDate在提交作品之外的酬金。Katherine C. Kurnit在提交的工作之外通过妇科肿瘤小组基金会参与LEAP Therapeutics的数据安全监测委员会或咨询委员会。珍妮弗·a·贝内特、凯瑟琳·a·米尔斯和威廉·扬·范·韦尔登没有透露任何信息。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 去求助
来源期刊
CiteScore
873.20
自引率
0.10%
发文量
51
审稿时长
1 months
期刊介绍: CA: A Cancer Journal for Clinicians" has been published by the American Cancer Society since 1950, making it one of the oldest peer-reviewed journals in oncology. It maintains the highest impact factor among all ISI-ranked journals. The journal effectively reaches a broad and diverse audience of health professionals, offering a unique platform to disseminate information on cancer prevention, early detection, various treatment modalities, palliative care, advocacy matters, quality-of-life topics, and more. As the premier journal of the American Cancer Society, it publishes mission-driven content that significantly influences patient care.
期刊最新文献
Preparation and Characterization of Low-Voltage Responsive Nanocomposite Shape-Changing Hydrogels/Carbon Nanofibers With Enhanced Mechanical Properties Introducing Antifouling Properties Onto Janus-Like Decellularized Corneas via Graft-From Zwitterionic Polymers Comparison of Swollen vs. Compounded Cross-linked High-Cis-1,4-Polybutadiene/n-Tetracosane Shape Memory Polymers Synthesis of Ferrocene-Containing Schiff Bases Based on 3-Aminopropyltriethoxysilane Oligomers for Covalent Modification of Glass Surfaces and Creation of Soft Magnetic Materials Fabrication of Hydroxyl Modified Hollow Glass Microsphere Composite Isocyanate-Based Polyimide Foam and Optimization Strategy Based on Different Bonding Mechanisms
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1