J Gynecol Oncol. 2024;35:e78. Forthcoming. English.
Published online Mar 22, 2024.
© 2024. Asian Society of Gynecologic Oncology, Korean Society of Gynecologic Oncology, and Japan Society of Gynecologic Oncology
Original Article

Abnormal p53 expression is associated with poor outcomes in grade I or II, stage I, endometrioid carcinoma: a retrospective single-institute study

Yu-Wei Chang,1 Hsiao-Li Kuo,2 Tzu-Chien Chen,3 Jessica Chen,4 Ling Lim,4 Kung-Liahng Wang,1,5 and Jen-Ruei Chen1,5,6
    • 1Department of Obstetrics & Gynecology, MacKay Memorial Hospital, Taipei, Taiwan.
    • 2Department of Nursing, MacKay Memorial Hospital, Taipei, Taiwan.
    • 3Department of Obstetrics & Gynecology, MacKay Memorial Hospital, Hsinchu Branch, Hsinchu, Taiwan.
    • 4Department of Obstetrics & Gynecology, MacKay Memorial Hospital, Tamsui Branch, New Taipei City, Taiwan.
    • 5Department of General Education, MacKay Junior College of Medicine, Nursing and Management, New Taipei City, Taiwan.
    • 6Department of Medicine, MacKay Medical College, New Taipei City, Taiwan.
Received November 06, 2023; Revised February 04, 2024; Accepted February 25, 2024.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (https://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Objective

The Cancer Genome Atlas study revealed an association between copy-number high (p53 abnormal) genetic mutation and poor prognosis in endometrial cancer in 2013. This retrospective study investigated outcomes in patients with abnormal p53 expression and stage I, low-grade endometrial endometrioid carcinoma (EEC).

Methods

We enrolled women with stage I, grade 1 or 2 EEC who received comprehensive staging and adjuvant therapy between January 2019 and December 2022 at MacKay Memorial Hospital, Taipei, Taiwan. Pathologists interpreted immunohistochemistry stains of cancerous tissues to detect p53 mutation. We compared recurrence, survival, progression-free survival, and overall survival between p53 abnormal and p53 normal groups.

Results

Of the 115 patients included, 26 had pathologically confirmed abnormal p53 expression. Of these 26 patients, five (19.2%) experienced recurrence, and two died due to disease progression. By contrast, no patients in the normal p53 group experienced disease recurrence or died due to disease progression. Significant intergroup differences were discovered in recurrent disease status (19.4% vs. 0%, p<0.001), mortality (7.7% vs. 0%, p<0.001), and progression-free survival (p<0.001). The overall survival (p=0.055) also showed powerful worse trend.

Conclusion

For patients with stage I, low-grade EEC, abnormal p53 expression may be used as an indicator of poor prognosis. Therefore, we suggest considering aggressive adjuvant therapies for these patients.

Synopsis

Abnormal expression of p53 is rare in low grade endometrial endometrioid carcinoma. Abnormal expression of p53 was associated with aggressive behavior in disease recurrence, metastases and survival according to the cancer genome atlas study. This retrospective study tried to figure out the role of p53 molecular factor in such non-aggressive pathological type endometrial cancer.

Graphical Abstract

Keywords
Endometrial Cancer; Endometrioid Carcinoma; Low Grade; Molecular Classification; p53 IHC

INTRODUCTION

Endometrial cancer (EC) is the most common type of gynecological cancer. In 2020, approximately 417,000 new cases of EC were identified worldwide [1, 2]. The most common type of EC is endometrioid carcinoma (EEC), which constitutes approximately 80% of cases of EC. EEC is often diagnosed in perimenopausal women and has a better prognosis [3]. Surgical pathological examination is the primary method for staging EC, and surgery is the primary method for treating EC. Due to noticeable clinical symptoms, EC is usually identified at an early stage, resulting in a 5-year survival rate of more than 90% for stage I EC [4].

Adjuvant therapy after surgical treatment of stage I EEC depends on various patient factors, such as age, tumor grade, myometrium invasion, and lymphovascular space invasion [5, 6]. These clinicopathologic factors may also influence the choice of adjuvant therapy [7]. Traditional pathological immunohistochemical (IHC) stain have been used to identify genetic alterations in EC, but the Cancer Genome Atlas has also been employed for this purpose and has provided insights into a new framework for subclassifying EC on the basis of four molecular subtypes: ultramutated (POLE-mutant), hypermutated (mismatch repair deficient [MMRd]), copy-number low (no specific molecular profile), and copy-number high (p53-abnormal) [8, 9]. The copy-number high subtype is mainly associated with type-II EC and is uncommon in type-I EEC (only 2%–15% of cases) [10]. Specifically, p53-mutated EEC is extremely rare among grade 1 and 2 tumors, and the effect of p53 mutation on prognosis is controversial.

We conducted a retrospective study to investigate the effect of p53 mutation on patient outcomes in clinicopathologic stage I, grade 1 or 2 EEC.

MATERIALS AND METHODS

1. Data collection

This retrospective, chart review study was conducted after obtaining approval from the Institutional Review Board of MacKay Memorial Hospital, Taipei, Taiwan (23MMHIS200e). Data from the period January 2019 to December 2022 were collected. Patients were enrolled if they had FIGO stage I, grade 1–2 EEC, if p53 IHC data were available for them, and if they received comprehensive staging and adjuvant therapy at MacKay Memorial Hospital. Patients were excluded if they did not have pure EEC (mixed cell types), if they had type-II EC or grade 3 EEC, if they had stage II or higher double ovarian cancer or EC, if they did not have p53 IHC data or comprehensive staging, or if they did not complete adjuvant treatment. We retrospectively reviewed electronic medical records of clinic notes, operation records, and pathologic reports. The FIGO staging system (version 2009) was used for staging. Morphological risk factors—including age, tumor grade, myometrial invasion, and lymphovascular space invasion—were recorded. Molecular alterations, such as MMRd/MMRp and TP53 mutation status, were also recorded, as was adjuvant therapy.

2. IHC staining for p53 protein

Cancerous tissue was processed into formalin-fixed, paraffin-embedded sections in accordance with the standard protocol. IHC staining was performed to detect localization of p53 protein. The DO7 clone is widely used as a p53 antibody for nuclear staining. A normal staining pattern indicated wild-type p53. IHC staining revealed a variable proportion of tumor cell nuclei that were positive and had varying intensity, ranging from 1% to 80% depending on the cellular differentiation and proliferation index. Abnormal p53 IHC staining patterns were classified as overexpression, complete absence, cytoplasmic, or heterogenous patterns. The former three pattern types were considered to be aberrant and indicative of mutations. Overexpression was characterized by strong nuclear positivity involving more than 75% of the tumor cells. Complete absence was characterized by the complete loss of nuclear staining but still the presence of internal control staining (fibroblasts, endothelial cells, or lymphocytes). The cytoplasmic pattern was characterized by positive cytoplasmic staining with variable nuclear staining. The final pattern was termed the subclonal p53 IHC staining pattern and was characterized by at least 10% of the total area having mutation-type staining patterns—the overexpression, complete absence, and cytoplasmic patterns—with other areas exhibiting wild-type p53 IHC staining. If cases were referred from local medical doctors or other hospitals, IHC data from outside medical records were also included in the analysis.

3. Statistical analysis

Our primary outcomes were recurrence, survival, progression-free survival (PFS), and overall survival (OS). PFS was defined as the time from surgery to the date of confirmation of recurrence or last follow-up before April 1, 2023, if no evidence of recurrence had been obtained. OS was defined as the time from operation to death, when evidence of recurrence was obtained, or the last follow-up before April 1, 2023, if no evidence of recurrence had been obtained. Recurrence was confirmed using imaging or biopsy. Data were analyzed using R (R Foundation for Statistical Computing, Vienna, Austria, https://www.R-project.org/) [11]. Continuous variables were compared using the nonpaired t test. Categorical variables were evaluated using chi-square or Fisher’s exact tests. Survival analysis was performed using Kaplan–Meier analysis and log-rank tests. Survival curves were calculated using the survminer package of R [12]. Statistical significance was indicated by p<0.05.

RESULTS

From January 2019 to December 2022, 537 patients with EC were registered in the cancer registry of our institute. We reviewed these patients’ charts to identify eligible patients. In total, 115 patients were eligible for analysis (Fig. 1). Of these, 89 were categorized into the normal_p53 group and 26 were categorized into the abnormal_p53 group. Patient characteristics are shown in Table 1 and did not differ between the groups. No significant differences in deep myometrium invasion, surgical approach, lymphovascular space invasion, MMR status, or adjuvant therapy were discovered. The normal_p53 group was significantly younger than the abnormal_p53 group (54 vs. 63 years, p=0.047).

Fig. 1
Participant recruitment process.
EEC, endometrioid carcinoma; MMH, MacKay Memorial Hospital.

In the abnormal_p53 group, five patients (19.2%) had recurrent disease. Specifically, two patients had local regional recurrence, and three patients had distant recurrence. Of these five patients with recurrent disease, two died due to disease progression. By contrast, no patients in the normal_p53 group experienced recurrent disease or died due to disease progression. Significant intergroup differences were found in recurrent disease status (19.4% vs. 0%, p<0.001) and mortality (7.7% vs. 0%, p<0.001). Additionally, significant difference was determined in PFS (p=0.0003; Fig. 2) and powerful worse trend was revealed in OS (p=0.055; Fig. 3), as revealed by survival analysis performed using Kaplan–Meier analysis and the log-rank test.

Fig. 2
Progression-free survival.
Abnormal = abnormal_p53 group; wild-type = normal_p53 group.

Fig. 3
Overall survival.
Abnormal = abnormal_p53 group; wild-type = normal_p53 group.

Cox regression analysis and proportional-hazards models were used to assess recurrence and mortality and to identify potential clinical contributors, such as FIGO stage, grade, surgical approach, lymphovascular space invasion, and MMR status. However, hazards ratios could not be calculated because no recurrent disease or mortality occurred in the normal_p53 group.

Recurrence was encountered in 5 (19.2%) patients in p53 abnormal expression group. The detail information of these patients is shown in Table 2. All patients were absence of lymphatic vascular space invasion. Two of them had abnormal IHC of MMR protein(s). Two patients had over half myometrial invasion and received adjuvant vaginal brachytherapy only according to current treatment guideline. However, they both developed lung metastases and one died because of failure of salvage systemic therapies. The remaining 3 cases were observed without adjuvant therapy due to their low pathological risks. One of them developed lung metastasis and died even aggressive radiation and chemotherapy. The remaining two cases developed pelvic recurrence. After surgical debulking and salvage systemic therapies, currently, they are still alive with disease.

Table 2
Patient characteristics in recurrent cases (n=5)

DISCUSSION

EC is the most common gynecological malignancy in developed countries. Poor diet, obesity, diabetes mellitus, and genetic factors, such as MMRd status, are potential risk factors for EC [13, 14, 15]. Surgical pathological examination is the primary method for staging EC and determining the appropriate adjuvant treatment. A new staging system was announced by FIGO in 2023; however, most current guidelines follow the 2009 recommendations and apply adjuvant management on the basis of morphological factors [7, 16]. EC is typically classified on the basis of the molecular characteristics of the cancerous tissue. A flowchart was developed for this purpose by the Cancer Genome Atlas study in 2013 [10]. According to the flowchart, EC is classified as POLE mutation, MMRd, copy-number low (MMRp), or copy-number high (abnormal p53) by using next generation gene sequencing for POLE and IHC staining for MMR proteins (MLH-1, MSH-2, MSH-6, and PMS-2) and p53 expression. However, these molecular classifications are not currently taken into account when determining the choice of adjuvant management despite the fact that they may be important prognostic factors.

Most patients with low-grade EEC have normal pattern or wild-type p53 expression. By contrast, patients with type-II EC or high-grade EEC commonly have aberrant or mutated p53 expression, namely the overexpression, complete absence, or cytoplasmic pattern [17]. According to Stelloo et al., p53 mutations may be indicators of a poor prognosis in EEC. They analyzed data from the PORTEC 1 and 2 studies, using both univariate and multivariate regression models, and discovered associations between p53 mutations and locoregional, distant recurrence and OS. They recommended escalating adjuvant therapy if IHC staining revealed abnormal p53 in addition to considering traditional morphological risk factors [18].

Abnormal mutant-type p53 staining is rare in low-grade EEC. This presents a paradoxical scenario in which a morphologically low-grade pattern in EEC exhibits molecular alterations associated with aggressive clinical behavior, such as p53 mutation [9]. This situation is estimated to occur in 10% to 15% of instances. Currently, adjuvant therapies for these cases are the same as those for cases involving wild-type p53 because of the practical guidelines. This is because p53 status is not considered a decisive factor in the treatment of EC [19]. However, Kurnit et al. showed that p53 mutation status could be integrated into risk stratification systems for early-stage, low-grade EEC because TP53 mutation is associated with significantly poorer PFS (hazard ratio [HR]=2.49; 95% confidence interval [CI]=1.05–5.90; p=0.04) [20]. Yano et al. [21] suggested that low-grade EEC with TP53-aberrant expression should be reclassified as high-grade (i.e., grade 3 disease) because of poor PFS (HR=2.91; p<0.001) and OS (HR=3.62; p<0.001) compared with low-grade EEC with TP53-normal expression. In addition, subgroup analysis of both early-stage (FIGO stage I/II) and advanced-stage (FIGO stage III/IV) EEC obtained similar results [21]. These studies have provided strong evidence of the possibility of p53 mutations being associated with poor prognosis in EEC. Our study specifically focused on patients with abnormal p53 and stage I, low-grade EEC to indicate their poor prognosis and to highlight the need for escalation of adjuvant therapies beyond what is recommended by current guidelines.

The TP53 gene is a type of tumor suppressor gene located on the short arm of chromosome 17p13.1 [22]. The normal p53 protein acts as a transcription factor and plays a crucial role in regulating various cellular processes, such as the cell cycle, apoptosis, genomic stability, and carcinogenesis [23, 24]. Mutations in the TP53 gene are frequently observed in high-grade serous ovarian cancer, fallopian tube cancer, peritoneal cancer, and EC [25]. In clinical practice, p53 IHC is used to predict TP53 mutation status due to its accessibility and affordability [17]. However, interpreting p53 IHC results can be technically challenging. This can be due to factors such as delayed fixation, inadequate presence of normal cells, or stronger staining of tumor cells [26]. Proper training in interpretation techniques and adherence to laboratory protocols are essential for obtaining reliable results from p53 IHC analysis.

Another challenging aspect is the heterogenous expression of p53 and subclonal TP53 mutations. Currently, this group is associated with certain mutator phenotypes, such as POLE ultramutation and MMRd/MMRp IHC, but cannot be classified into a single molecular subtype [27]. A universally accepted definition for subclonal abnormal p53 expression has not been established. We conducted subgroup analysis on heterogenous p53 expression; however, the results did not reveal better outcomes compared with those of the homogenous p53 abnormal group (not shown in the results and tables), inconsistent with the findings of several studies. This discrepancy may be attributable to statistical power limitations due to an insufficient number of cases in our study. Multicenter collaborative trials with more cases are warranted.

Our study has several limitations. First, this was a retrospective study with a limited number of cases from a single institute. Although we initially identified more than 500 patients, we had to omit most of them due to a lack of p53 IHC data. In our institute, we began routine p53 IHC staining for EC after mid-2021. Additional funding is required to obtain the missing p53 IHC data in these excluded cases. Second, most of the p53 IHC data were incomplete or ambiguous due to differences in p53 IHC nomenclature between nongynecological specialist pathologists in our department. Third, ultramutated (POLE-mutant) EEC could not be identified in our study due to a lack of facilities.

Finally, this study simply indicated an association between abnormal p53 and a poorer prognosis in low-grade EEC. On the basis of our findings, we recommend that patients receive aggressive therapies beyond what is recommended by current guidelines. This is because the risks of recurrence, mortality, and poor outcomes may be higher than current guidelines estimate. In the future, we may compare outcomes between aggressive adjuvant therapies and traditional adjuvant modalities recommended by current guidelines in cases of low-grade EEC with abnormal p53 IHC.

Notes

Conflict of Interest:No potential conflict of interest relevant to this article was reported.

Author Contributions:

  • Conceptualization: C.

  • Data curation: K.

  • Resources: C.

  • Software: C.

  • Writing - original draft: C.

  • Writing - review & editing: C.

References

    1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2021;71:209–249.
    1. Lu KH, Broaddus RR. Endometrial cancer. N Engl J Med 2020;383:2053–2064.
    1. Bokhman JV. Two pathogenetic types of endometrial carcinoma. Gynecol Oncol 1983;15:10–17.
    1. Crosbie EJ, Kitson SJ, McAlpine JN, Mukhopadhyay A, Powell ME, Singh N. Endometrial cancer. Lancet 2022;399:1412–1428.
    1. Keys HM, Roberts JA, Brunetto VL, Zaino RJ, Spirtos NM, Bloss JD, et al. A phase III trial of surgery with or without adjunctive external pelvic radiation therapy in intermediate risk endometrial adenocarcinoma: a Gynecologic Oncology Group study. Gynecol Oncol 2004;92:744–751.
    1. Creutzberg CL, van Putten WL, Koper PC, Lybeert ML, Jobsen JJ, Wárlám-Rodenhuis CC, et al. Surgery and postoperative radiotherapy versus surgery alone for patients with stage-1 endometrial carcinoma: multicentre randomised trial. PORTEC Study Group. Post Operative Radiation Therapy in Endometrial Carcinoma. Lancet 2000;355:1404–1411.
    1. Abu-Rustum N, Yashar C, Arend R, Barber E, Bradley K, Brooks R, et al. Uterine neoplasms, version 1.2023, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw 2023;21:181–209.
    1. Kandoth C, Schultz N, Cherniack AD, Akbani R, Liu Y, Shen H, et al. Integrated genomic characterization of endometrial carcinoma. Nature 2013;497:67–73.
    1. Vermij L, Smit V, Nout R, Bosse T. Incorporation of molecular characteristics into endometrial cancer management. Histopathology 2020;76:52–63.
    1. Murali R, Delair DF, Bean SM, Abu-Rustum NR, Soslow RA. Evolving roles of histologic evaluation and molecular/genomic profiling in the management of endometrial cancer. J Natl Compr Canc Netw 2018;16:201–209.
    1. R Core Team. R Foundation for Statistical Computing. R: a language and environment for statistical computing [Internet]. Vienna: R Foundation for Statistical Computing; 2024 [cited 2023 Jul 31].
    1. Kassambara A, Kosinski M, Biecek P. Survminer: Drawing Survival Curves using ‘ggplot2.’ R package version 0.4.9 (2021) [Internet]. Vienna: Institute for Statistics and Mathematics; 2021 [cited 2023 Jul 31].
    1. Lauby-Secretan B, Scoccianti C, Loomis D, Grosse Y, Bianchini F, Straif K, et al. Body fatness and cancer--viewpoint of the IARC Working Group. N Engl J Med 2016;375:794–798.
    1. Saed L, Varse F, Baradaran HR, Moradi Y, Khateri S, Friberg E, et al. The effect of diabetes on the risk of endometrial Cancer: an updated a systematic review and meta-analysis. BMC Cancer 2019;19:527.
    1. Moroney MR, Davies KD, Wilberger AC, Sheeder J, Post MD, Berning AA, et al. Molecular markers in recurrent stage I, grade 1 endometrioid endometrial cancers. Gynecol Oncol 2019;153:517–520.
    1. Oaknin A, Bosse TJ, Creutzberg CL, Giornelli G, Harter P, Joly F, et al. Endometrial cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol 2022;33:860–877.
    1. Singh N, Piskorz AM, Bosse T, Jimenez-Linan M, Rous B, Brenton JD, et al. p53 immunohistochemistry is an accurate surrogate for TP53 mutational analysis in endometrial carcinoma biopsies. J Pathol 2020;250:336–345.
    1. Stelloo E, Nout RA, Osse EM, Jürgenliemk-Schulz IJ, Jobsen JJ, Lutgens LC, et al. Improved risk assessment by integrating molecular and clinicopathological factors in early-stage endometrial cancer-combined analysis of the PORTEC cohorts. Clin Cancer Res 2016;22:4215–4224.
    1. Bosse T, Nout RA, McAlpine JN, McConechy MK, Britton H, Hussein YR, et al. Molecular classification of grade 3 endometrioid endometrial cancers identifies distinct prognostic subgroups. Am J Surg Pathol 2018;42:561–568.
    1. Kurnit KC, Kim GN, Fellman BM, Urbauer DL, Mills GB, Zhang W, et al. CTNNB1 (beta-catenin) mutation identifies low grade, early stage endometrial cancer patients at increased risk of recurrence. Mod Pathol 2017;30:1032–1041.
    1. Yano M, Ito K, Yabuno A, Ogane N, Katoh T, Miyazawa M, et al. Impact of TP53 immunohistochemistry on the histological grading system for endometrial endometrioid carcinoma. Mod Pathol 2019;32:1023–1031.
    1. Tanaka T, Watanabe M, Yamashita K. Potential therapeutic targets of TP53 gene in the context of its classically canonical functions and its latest non-canonical functions in human cancer. Oncotarget 2018;9:16234–16247.
    1. Wang H, Guo M, Wei H, Chen Y. Targeting p53 pathways: mechanisms, structures, and advances in therapy. Signal Transduct Target Ther 2023;8:92.
    1. Bode AM, Dong Z. Post-translational modification of p53 in tumorigenesis. Nat Rev Cancer 2004;4:793–805.
    1. Cole AJ, Dwight T, Gill AJ, Dickson KA, Zhu Y, Clarkson A, et al. Assessing mutant p53 in primary high-grade serous ovarian cancer using immunohistochemistry and massively parallel sequencing. Sci Rep 2016;6:26191.
    1. Köbel M, Ronnett BM, Singh N, Soslow RA, Gilks CB, McCluggage WG. Interpretation of P53 immunohistochemistry in endometrial carcinomas: toward increased reproducibility. Int J Gynecol Pathol 2019;38 Suppl 1:S123–S131.
    1. León-Castillo A, Gilvazquez E, Nout R, Smit VT, McAlpine JN, McConechy M, et al. Clinicopathological and molecular characterisation of ‘multiple-classifier’ endometrial carcinomas. J Pathol 2020;250:312–322.

Metrics
Share
Figures

1 / 3

Tables

1 / 2

PERMALINK