Next Article in Journal
Preclinical Evaluation of Artesunate as an Antineoplastic Agent in Ovarian Cancer Treatment
Next Article in Special Issue
Distinct Somatic Alteration Features Identified by Gene Panel Sequencing in Korean Triple-Negative Breast Cancer with High Ki67 Expression
Previous Article in Journal
A Comprehensive Review of Computer-Aided Diagnosis of Major Mental and Neurological Disorders and Suicide: A Biostatistical Perspective on Data Mining
Previous Article in Special Issue
Suitability of Bronchoscopic Biopsy Tissue Samples for Next-Generation Sequencing
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

PD-L1 Expression Is an Independent Marker for Lymph Node Metastasis in Middle Eastern Endometrial Cancer

by
Abdul K. Siraj
1,†,
Sandeep Kumar Parvathareddy
1,†,
Padmanaban Annaiyappanaidu
1,
Nabil Siraj
1,
Maha Al-Rasheed
1,
Ismail A. Al-Badawi
2,
Fouad Al-Dayel
3 and
Khawla S. Al-Kuraya
1,*
1
Human Cancer Genomic Research, Research Center, King Faisal Specialist Hospital and Research Center, P.O. Box 3354, Riyadh 11211, Saudi Arabia
2
Department of Obstetrics-Gynecology, King Faisal Specialist Hospital and Research Center, P.O. Box 3354, Riyadh 11211, Saudi Arabia
3
Department of Pathology, King Faisal Specialist Hospital and Research Centre, P.O. Box 3354, Riyadh 11211, Saudi Arabia
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Diagnostics 2021, 11(3), 394; https://doi.org/10.3390/diagnostics11030394
Submission received: 17 January 2021 / Revised: 20 February 2021 / Accepted: 23 February 2021 / Published: 25 February 2021

Abstract

:
Programmed death ligand 1 (PD-L1) expression in endometrial cancer (EC) tumor cells have been reported in several studies with inconsistent results. Furthermore, there is scarcity of data on the prevalence and prognostic significance of PD-L1 expression in EC from Middle Eastern ethnicity. We aimed to assess PD-L1 expression in a large cohort of Middle Eastern EC and to correlate this with clinico-pathological factors, as well as mismatch repair (MMR) protein status and patients’ outcome. PD-L1 expression was investigated using immunohistochemistry on tissue microarray in an unselected cohort of 440 EC. Kaplan–Meier and logistic regression analysis were used to compare the outcome and prognostic factors. PD-L1 expression in tumor tissue was detected in 18.9% (83/440) EC cases with no impact on survival. When stratified for MMR protein status, PD-L1 expression was similar for both MMR deficient and MMR proficient ECs. However, the expression of PD-L1 in tumor cells was significantly associated with type II (non-endometrioid) histology (p = 0.0005) and lymph node metastasis (p = 0.0172). Multivariate analysis showed PD-L1 expression to be an independent risk factor for lymph node metastasis (odds ratio: 2.94; 95% CI: 1.26–6.84; p = 0.0123). In conclusion, PD-L1 was strongly associated with non-endometrioid EC and was an independent prognostic marker of lymph node metastasis.

1. Introduction

Endometrial cancer (EC) is the most common gynecological malignancy in the Western countries [1,2]. Similarly, in Saudi Arabia, EC is the most common gynecological cancer and the fourth most common cancer among Saudi women [3]. The incidence is increasing in Saudi Arabia, which could be partially attributed to the high prevalence of obesity in this population [3,4,5]. Although most cases present at early stage and have a good prognosis, about 15–20% of patient present with advanced stage and aggressive morphology [6,7]. Recurrence and metastasis remain a challenge despite the available therapeutic options of surgery, chemotherapy, radiotherapy, and hormonal therapy.
Recently, immune checkpoint inhibitor has emerged as a promising therapeutic option in oncology and has significantly altered the standard treatment for many advanced cancers [8]. The most common mechanisms underlying immunotherapy are programmed cell death protein-1 (PD-1) and programmed death ligand-1 (PD-L1) which serve as immune checkpoints in tumor micro environment [9,10]. The PD-1/PD-L1 axis is a critical immune modulatory pathway that inhibits the immune reaction to cancer cells by negatively regulating T-cell functions [10,11].
After the Food and Drug Administration (FDA) approval of PD-L1 inhibitors for treatment of microsatellite instable and metastatic cancers in several types of malignancies including EC [12,13,14,15,16], this promising therapeutic modality has been the encouraging force behind the exploration of PD-L1 expression in EC in several recent reports [17,18,19,20,21,22]. However, results from these studies were not consistent. The discrepancies between these reports could be attributed to differences in cohort size, patient’s ethnicity, antibody used, and cut-off values.
At present, the prevalence of PD-L1 expression in Middle Eastern EC has not been explored in detail. Therefore, we conducted this study to determine the expression of PD-L1 in a large cohort of EC and to evaluate the utility of PD-L1 as prognostic biomarker of EC in this ethnicity.

2. Materials and Methods

2.1. Sample Selection

Archival samples from 440 EC patients diagnosed between 1990 and 2016 at the King Faisal Specialist Hospital and Research Center (Riyadh, Saudi Arabia) were included in the study. Detailed clinico-pathological data were noted from case records and have been summarized in Table 1. Endometrioid endometrial carcinomas were categorized as Type I and all other histologic subtypes were classified as Type II EC. The International Federation of Obstetrics and Gynecology (FIGO) system was used for staging and grading of tumors. Depth of myometrial invasion ≥50% was considered high myometrial invasion, whereas <50% was considered low myometrial invasion. All patients had surgery as their primary treatment. A total of 29.1% (128/440) of patients received radiotherapy alone following surgery. Moreover, 19.1% (84/440) of patients received adjuvant chemotherapy, of which 69 patients received carboplatin and paclitaxel combination whereas 15 patients received carboplatin alone. Overall survival was defined as the length of time from the date of diagnosis that patients diagnosed with the disease are still alive. Recurrence-free survival was defined as the length of time after primary treatment for a cancer ends that the patient survives without any signs or symptoms of that cancer. Disease-free survival was defined as the length of time after primary treatment for a cancer ends that the patient survives without any signs or symptoms of that cancer or death. All samples were obtained from patients with approval from the Institutional Review Board of the hospital. For the study, waiver of consent was obtained for archived paraffin tissue blocks from the Research Advisory Council (RAC) under project RAC# 2180 001 (approved on 7 March 2018).

2.2. Tissue Microarray Construction and Immunohistochemistry

All samples were analyzed in a tissue microarray (TMA) format. TMA construction was performed as described earlier [23]. Briefly, tissue cylinders with a diameter of 0.6 mm were punched from representative tumor regions of each donor tissue block and brought into recipient paraffin block using a modified semiautomatic robotic precision instrument (Beecher Instruments, Woodland, WI, USA). Two cores of EC were arrayed from each case.
Tissue microarray slides were processed and stained manually as described previously [24]. Primary antibody against PD-L1 (E1L3N, 1:50 dilution, pH 9.0, Cell Signaling Technology, Danvers, MA, USA) was used. A membranous and/or cytoplasmic staining was observed. Only the membrane staining was considered for scoring. PD-L1 was scored as described previously [25]. Briefly, the proportion of positively stained cells was calculated as a percentage for each core and the scores were averaged across two tissue cores from the same tumor to yield a single percent staining score representing each cancer patient. For the purpose of statistical analysis, the scores were dichotomized. Cases showing expression level of ≥5% were classified as positive and those with less than 5% as negative. Only staining of tumor cells was considered for percentage calculation.
MMR protein staining and evaluation was done as described previously [26]. Briefly, MMR protein expression was evaluated using MSH2, MSH6, MLH1, and PMS2 proteins. Tumor was classified as deficient MMR if any of the four proteins showed loss of staining in cancer with concurrent positive staining in the nuclei of normal epithelial cells. Otherwise, they were classified as proficient MMR.
IHC scoring was done by two pathologists, blinded to the clinico-pathological characteristics. Discordant scores were reviewed together to achieve agreement.

2.3. POLE Mutation Analysis

POLE mutation data were available from our previous study [27].

2.4. Statistical Analysis

The associations between clinico-pathological variables and protein expression were performed using contingency table analysis and Chi square tests. Survival curves were generated using the Kaplan–Meier method. Multivariate logistic regression analysis was performed to assess the correlation of lymph node metastasis and clinico-pathological variables. Two-sided tests were used for statistical analyses with a limit of significance defined as p-value of <0.05. Data analyses was performed using the JMP11.0 (SAS Institute, Inc., Cary, NC, USA) software package.

3. Results

3.1. Patient Characteristics

Median age of the study population was 59.3 years (range: 26–94 years). The majority of tumors were of Type I (endometrioid adenocarcinoma) histology, accounting for 88.0% (387/440) of ECs, with an almost equal distribution among the three grades. A nearly equal number of cases showed high and low myometrial invasion (50.2% vs. 49.8%). The majority of the cases were Stage I tumors (64.8%; 285/440). MMR deficient tumors accounted for 12.1% (53/440) of ECs in our cohort (Table 1).

3.2. PD-L1 Protein Expression in Endometrial Cancer and Its Clinico-Pathological Associations

PD-L1 protein expression was analyzed immunohistochemically in 440 EC samples. PD-L1 expression was noted in 18.9% (83/440) of ECs (Figure 1) and found to be significantly associated with Type II (non-endometrioid) histology (p = 0.0005) and lymph node metastasis (p = 0.0172). However, no association was found between PD-L1 expression and mismatch repair (MMR) immunohistochemistry (p = 0.4435) (Table 2). We next analyzed the association between PD-L1 expression and DNA polymerase epsilon (POLE) mutation. POLE mutation was detected in 0.5% (2/431) of cases in our cohort and was not associated with PD-L1 expression. Both the cases showing POLE mutation were negative for PD-L1 (Table 2).

3.3. Prognostic Impact of PD-L1 Expression in Endometrial Cancer

We evaluated the effect of PD-L1 expression on overall survival (OS), recurrence-free survival (RFS), and disease-free survival (DFS). However, PD-L1 expression was not associated with OS (p = 0.6963), RFS (p = 0.5351), or DFS (p = 0.7336) (Figure 2A–C). In multivariate logistic regression analysis, PD-L1 expression was an independent risk factor for lymph node metastasis in EC (odds ratio: 2.94; 95% CI: 1.26–6.84; p = 0.0123) (Table 3).

4. Discussion

Immunotherapies have gained much attention in current oncology practice. In recent years, PD-L1 has shown impressive clinical results in many solid tumors [15,16,28,29,30,31]. Therefore, identification of PD-L1 expression in tumors can offer important value in guiding clinical decisions to use immunotherapy. Different studies have investigated the role of PD-L1 in EC [17,18,19,20,21,22]. However, the results did not reach consensus. Moreover, data about PD-L1 expression in EC from Middle Eastern ethnicity are limited [32,33]. Therefore, in this study, we determined the expression of PD-L1 by IHC in a large cohort of EC and explored its correlation with clinico-pathological features.
In the present study, the incidence of PD-L1 expression was 18.9% in EC patients. This is consistent with previous reports where the identified PD-L1 expression in EC ranges from 14% to 56% [20,32,34,35]. We found a significant association between PD-L1 expression and Type II EC, which is in agreement with previous reports [34,35].
Interestingly, PD-L1 expression was significantly associated with lymph node metastasis and multivariate analysis demonstrated that high PD-L1 expression is an independent marker for lymph node metastasis. In concordance with our results, previously, few studies have reported the correlation between PD-L1 expression and risk of lymph node metastasis [36,37,38]. This finding is of important clinical implication, since PD-L1 expression could be considered as a potential biomarker for lymph node metastasis in EC. Identifying predictive factors for immunotherapeutic agents’ response is of great importance in helping oncologists to select patients who might benefit from immunotherapy.
MMR deficient status has been found to be a good predictive factor for immunotherapy response [39,40]. Furthermore, previous studies have shown that PD-L1 overexpression is significantly associated with MMR deficient tumors [20,21,41]. Expression of PD-L1 was similar in MMR deficient and proficient tumors in our cohort with no significant differences found in PD-L1 expression in both groups. This finding suggests that PD-L1 has no predictive advantage in MMR deficient EC subgroup and possibly PD-L1 expression status alone should be applied in selecting patients for immunotherapy regardless of their MMR status. A recent study has also shown lack of association between PD-L1 expression and MMR status in a large cohort of Western EC [34].
EC patients harboring POLE mutation have been shown to offer better immunotherapy response [42]. Therefore, we took advantage of availability of POLE mutation data on 431 patients [27] and analyzed the association with PD-L1 expression. Our results confirmed a lack of correlation between PD-L1 expression and POLE mutation in this cohort. Our result is contradicting previously reported higher frequency of PD-L1 expression in POLE mutant and MMR deficient tumors [43].
There are conflicting results in the literature, not only on PD-L1 expression incidence in EC and their clinico-molecular associations, but also their prognostic value and impact on patient survival. With regards to patient outcome, our results demonstrated that PD-L1 expression is not associated with patients’ overall survival, disease-free survival, or recurrence-free survival. This is in line with a previous study [32] where patients’ survival was not affected by PD-L1 expression in either tumor cells or immune infiltrates. Another large study on 689 EC samples evaluated the expression of PD-L1 on tumor cells and found no association with disease-specific survival, even after stratifying by histologic subtype [34]. Furthermore, a recent large metanalysis with more than 1600 patients suggested that PD-L1 expression is not associated with poor prognosis in endometrial cancer patients. However, they conclude that PD-L1 expression is positively correlated with poor differentiation and advanced stage in endometrial cancer [44]. These findings are in concordance with our study results, where we show that PD-L1 expression is strongly associated with lymph node metastasis. Since lymph node metastasis is an indicator of advanced disease, our study could provide further evidence that PD-L1 might not correlate with overall patient’s prognosis but can be used as a marker for advanced disease in endometrial cancer.

5. Conclusions

We detected PD-L1 expression in 18.9% of EC cases. We also demonstrated that PD-L1 was strongly associated with non-endometroid EC. This study also could not reveal any association between PD-L1 expression and patient survival. More importantly, our results show PD-L1 expression to be an independent prognostic marker of lymph node metastasis. Lymph node metastasis is an indicator of advanced disease and our study provides evidence that PD-L1 could be used as a marker of advanced disease in endometrial cancer. More studies are needed to identify patients who might benefit from immunotherapy in EC from this ethnicity, and to determine the role of immunotherapy in EC patients with lymph node metastasis.

Author Contributions

Conceptualization, K.S.A.-K. and A.K.S.; methodology, S.K.P. and A.K.S.; software, S.K.P.; validation, A.K.S., P.A., and N.S.; formal analysis, A.K.S. and S.K.P.; investigation, A.K.S., S.K.P., P.A., N.S., and M.A.-R.; resources, I.A.A.-B. and F.A.-D.; data curation, S.K.P.; writing—original draft preparation, K.S.A.-K.; writing—review and editing, A.K.S. and S.K.P.; visualization, A.K.S. and S.K.P.; supervision, K.S.A.-K. and A.K.S.; project administration, K.S.A.-K. and A.K.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki, and approved by the Institutional Review Board of King Faisal Specialist Hospital and Research Centre (protocol code 2180 001; 07 March 2018).

Informed Consent Statement

Patient consent was waived since only retrospective patient data and archival tissue samples were used for the study.

Data Availability Statement

The data presented in this study are available in the article.

Acknowledgments

The authors would like to thank Felisa DeVera for her technical assistance.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Howlader, N.; Krapcho, M.; Miller, D.; Bishop, K.; Kosary, C.; Yu, M.; Cronin, K.A. (Eds.) SEER Cancer Statistics Review, 1975–2014; National Cancer Institute: Bethesda, MD, USA, 2017. [Google Scholar]
  2. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2020. CA A Cancer J. Clin. 2020, 70, 7–30. [Google Scholar] [CrossRef]
  3. Alrawaji, A.; Alshahrani, Z.; Alzahrani, W.; Alomran, F.; Almadouj, A.; Alshehri, S.; Alzahrani, A.; Bazarbashi, S.; Alhashmi, H.; Almutlaq, H.; et al. Cancer Incidence Report Saudi Arabia 2015. In Saudi Cancer Registry; Saudi Health Council 2015: Riyadh, Saudi Arabia, 2018. [Google Scholar]
  4. Althubiti, M.A.; Eldein, M.M.N. Trends in the incidence and mortality of cancer in Saudi Arabia. Saudi Med. J. 2018, 39, 1259. [Google Scholar] [CrossRef]
  5. World Health Organization; Global Health Observatory (GHO) Data. Overweight and Obesity, Adults Aged 18+. World Health Organization [updated 16 June 2015]. Available online: http://www.who.int/gho/ncd/risk_factors/overweight_text/en (accessed on 22 December 2020).
  6. Colombo, N.; Creutzberg, C.; Amant, F.; Bosse, T.; González-Martín, A.; Ledermann, J.; Marth, C.; Nout, R.; Querleu, D.; Mirza, M.R. ESMO-ESGO-ESTRO consensus conference on endometrial cancer: Diagnosis, treatment and follow-up. Int. J. Gynecol. Cancer 2016, 26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Remmerie, M.; Janssens, V. Targeted therapies in type II endometrial cancers: Too little, but not too late. Int. J. Mol. Sci. 2018, 19, 2380. [Google Scholar] [CrossRef] [Green Version]
  8. Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Zou, W.; Wolchok, J.D.; Chen, L. PD-L1 (B7-H1) and PD-1 pathway blockade for cancer therapy: Mechanisms, response biomarkers, and combinations. Sci. Transl. Med. 2016, 8, rv324–rv328. [Google Scholar] [CrossRef] [Green Version]
  10. Chen, J.; Jiang, C.; Jin, L.; Zhang, X. Regulation of PD-L1: A novel role of pro-survival signalling in cancer. Ann. Oncol. 2016, 27, 409–416. [Google Scholar] [CrossRef]
  11. Topalian, S.L.; Taube, J.M.; Anders, R.A.; Pardoll, D.M. Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat. Rev. Cancer 2016, 16, 275–287. [Google Scholar] [CrossRef]
  12. Sheng, X.; Yan, X.; Chi, Z.; Si, L.; Cui, C.; Tang, B.; Li, S.; Mao, L.; Lian, B.; Wang, X. Axitinib in combination with toripalimab, a humanized immunoglobulin G4 monoclonal antibody against programmed cell death-1, in patients with metastatic mucosal melanoma: An open-label phase IB trial. J. Clin. Oncol. 2019, 37, 2987–2999. [Google Scholar] [CrossRef] [PubMed]
  13. Dang, T.O.; Ogunniyi, A.; Barbee, M.S.; Drilon, A. Pembrolizumab for the treatment of PD-L1 positive advanced or metastatic non-small cell lung cancer. Expert Rev. Anticancer Ther. 2016, 16, 13–20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Antill, Y.C.; Kok, P.S.; Robledo, K.; Barnes, E.; Friedlander, M.; Baron-Hay, S.E.; Shannon, C.M.; Coward, J.; Beale, P.J.; Goss, G. Activity of durvalumab in advanced endometrial cancer (AEC) according to mismatch repair (MMR) status: The phase II PHAEDRA trial (ANZGOG1601). J. Clin. Oncol. 2019, 37, 5501. [Google Scholar] [CrossRef]
  15. Balar, A.V.; Galsky, M.D.; Rosenberg, J.E.; Powles, T.; Petrylak, D.P.; Bellmunt, J.; Loriot, Y.; Necchi, A.; Hoffman-Censits, J.; Perez-Gracia, J.L. Atezolizumab as first-line treatment in cisplatin-ineligible patients with locally advanced and metastatic urothelial carcinoma: A single-arm, multicentre, phase 2 trial. Lancet 2017, 389, 67–76. [Google Scholar] [CrossRef] [Green Version]
  16. McDermott, D.F.; Sosman, J.A.; Sznol, M.; Massard, C.; Gordon, M.S.; Hamid, O.; Powderly, J.D.; Infante, J.R.; Fassò, M.; Wang, Y.V. Atezolizumab, an anti-programmed death-ligand 1 antibody, in metastatic renal cell carcinoma: Long-term safety, clinical activity, and immune correlates from a phase Ia study. J. Clin. Oncol. 2016, 34, 833–842. [Google Scholar] [CrossRef] [PubMed]
  17. Pasanen, A.; Ahvenainen, T.; Pellinen, T.; Vahteristo, P.; Loukovaara, M.; Bützow, R. PD-L1 expression in endometrial carcinoma cells and intratumoral immune cells: Differences across histologic and TCGA-based molecular subgroups. Am. J. Surg. Pathol. 2020, 44, 174–181. [Google Scholar] [CrossRef]
  18. Asaka, S.; Yen, T.-T.; Wang, T.-L.; Shih, I.-M.; Gaillard, S. T cell-inflamed phenotype and increased Foxp3 expression in infiltrating T-cells of mismatch-repair deficient endometrial cancers. Mod. Pathol. 2019, 32, 576–584. [Google Scholar] [CrossRef] [PubMed]
  19. Kim, J.; Kim, S.; Lee, H.S.; Yang, W.; Cho, H.; Chay, D.B.; Cho, S.J.; Hong, S.; Kim, J.-H. Prognostic implication of programmed cell death 1 protein and its ligand expressions in endometrial cancer. Gynecol. Oncol. 2018, 149, 381–387. [Google Scholar] [CrossRef] [PubMed]
  20. Li, Z.; Joehlin-Price, A.S.; Rhoades, J.; Ayoola-Adeola, M.; Miller, K.; Parwani, A.V.; Backes, F.J.; Felix, A.S.; Suarez, A.A. Programmed death ligand 1 expression among 700 consecutive endometrial cancers: Strong association with mismatch repair protein deficiency. Int. J. Gynecol. Cancer 2018, 28, 59–68. [Google Scholar] [CrossRef]
  21. Bregar, A.; Deshpande, A.; Grange, C.; Zi, T.; Stall, J.; Hirsch, H.; Reeves, J.; Sathyanarayanan, S.; Growdon, W.B.; Rueda, B.R. Characterization of immune regulatory molecules B7-H4 and PD-L1 in low and high grade endometrial tumors. Gynecol. Oncol. 2017, 145, 446–452. [Google Scholar] [CrossRef] [Green Version]
  22. Inaguma, S.; Wang, Z.; Lasota, J.; Sarlomo-Rikala, M.; McCue, P.A.; Ikeda, H.; Miettinen, M. Comprehensive immunohistochemical study of programmed cell death ligand 1 (PD-L1). Analysis in 5536 cases revealed consistent expression in trophoblastic tumors. Am. J. Surg. Pathol. 2016, 40, 1133. [Google Scholar] [CrossRef] [Green Version]
  23. Siraj, A.; Bavi, P.; Abubaker, J.; Jehan, Z.; Sultana, M.; Al-Dayel, F.; Al-Nuaim, A.; Alzahrani, A.; Ahmed, M.; Al-Sanea, O. Genome-wide expression analysis of Middle Eastern papillary thyroid cancer reveals c-MET as a novel target for cancer therapy. J. Pathol. 2007, 213, 190–199. [Google Scholar] [CrossRef]
  24. Bavi, P.; Jehan, Z.; Atizado, V.; Al-Dossari, H.; Al-Dayel, F.; Tulbah, A.; Amr, S.S.; Sheikh, S.S.; Ezzat, A.; El-Solh, H. Prevalence of fragile histidine triad expression in tumors from Saudi Arabia: A tissue microarray analysis. Cancer Epidemiol. Prev. Biomark. 2006, 15, 1708–1718. [Google Scholar] [CrossRef] [Green Version]
  25. Mesnage, S.; Auguste, A.; Genestie, C.; Dunant, A.; Pain, E.; Drusch, F.; Gouy, S.; Morice, P.; Bentivegna, E.; Lhomme, C. Neoadjuvant chemotherapy (NACT) increases immune infiltration and programmed death-ligand 1 (PD-L1) expression in epithelial ovarian cancer (EOC). Ann. Oncol. 2017, 28, 651–657. [Google Scholar] [CrossRef]
  26. Siraj, A.K.; Prabhakaran, S.; Bavi, P.; Bu, R.; Beg, S.; Hazmi, M.A.; Al-Rasheed, M.; Al-Assiri, M.; Sairafi, R.; Al-Dayel, F. Prevalence of Lynch syndrome in a Middle Eastern population with colorectal cancer. Cancer 2015, 121, 1762–1771. [Google Scholar] [CrossRef]
  27. Siraj, A.K.; Parvathareddy, S.K.; Bu, R.; Iqbal, K.; Siraj, S.; Masoodi, T.; Concepcion, R.M.; Ghazwani, L.O.; AlBadawi, I.; Al-Dayel, F. Germline POLE and POLD1 proofreading domain mutations in endometrial carcinoma from Middle Eastern region. Cancer Cell Int. 2019, 19, 334. [Google Scholar] [CrossRef]
  28. Fehrenbacher, L.; von Pawel, J.; Park, K.; Rittmeyer, A.; Gandara, D.R.; Aix, S.P.; Han, J.-Y.; Gadgeel, S.M.; Hida, T.; Cortinovis, D.L. Updated efficacy analysis including secondary population results for OAK: A randomized phase III study of atezolizumab versus docetaxel in patients with previously treated advanced non–small cell lung cancer. J. Thorac. Oncol. 2018, 13, 1156–1170. [Google Scholar] [CrossRef] [Green Version]
  29. Emens, L.A.; Cruz, C.; Eder, J.P.; Braiteh, F.; Chung, C.; Tolaney, S.M.; Kuter, I.; Nanda, R.; Cassier, P.A.; Delord, J.-P. Long-term clinical outcomes and biomarker analyses of atezolizumab therapy for patients with metastatic triple-negative breast cancer: A phase 1 study. JAMA Oncol. 2019, 5, 74–82. [Google Scholar] [CrossRef] [PubMed]
  30. Eng, C.; Kim, T.W.; Bendell, J.; Argilés, G.; Tebbutt, N.C.; Di Bartolomeo, M.; Falcone, A.; Fakih, M.; Kozloff, M.; Segal, N.H. Atezolizumab with or without cobimetinib versus regorafenib in previously treated metastatic colorectal cancer (IMblaze370): A multicentre, open-label, phase 3, randomised, controlled trial. Lancet Oncol. 2019, 20, 849–861. [Google Scholar] [CrossRef]
  31. Liu, J.F.; Gordon, M.; Veneris, J.; Braiteh, F.; Balmanoukian, A.; Eder, J.P.; Oaknin, A.; Hamilton, E.; Wang, Y.; Sarkar, I. Safety, clinical activity and biomarker assessments of atezolizumab from a Phase I study in advanced/recurrent ovarian and uterine cancers. Gynecol. Oncol. 2019, 154, 314–322. [Google Scholar] [CrossRef] [PubMed]
  32. Sungu, N.; Yildirim, M.; Desdicioglu, R.; Başaran Aydoğdu, Ö.; Kiliçarslan, A.; Tatli Doğan, H.; Kiliç Yazgan, A.; Akyol, M.; Erdoğan, F. Expression of immunomodulatory molecules PD-1, PD-L1, and PD-L2, and their relationship with clinicopathologic characteristics in endometrial cancer. Int. J. Gynecol. Pathol. 2019, 38, 404–413. [Google Scholar] [CrossRef] [PubMed]
  33. Al-Hussaini, M.; Lataifeh, I.; Jaradat, I.; Abdeen, G.; Otay, L.; Badran, O.; Abu Sheikha, A.; Dayyat, A.; El Khaldi, M.; Ashi Al-Loh, S. Undifferentiated endometrial carcinoma, an immunohistochemical study including PD-L1 testing of a series of cases from a single cancer center. Int. J. Gynecol. Pathol. 2018, 37, 564–574. [Google Scholar] [CrossRef] [PubMed]
  34. Engerud, H.; Berg, H.F.; Myrvold, M.; Halle, M.K.; Bjorge, L.; Haldorsen, I.S.; Hoivik, E.A.; Trovik, J.; Krakstad, C. High degree of heterogeneity of PD-L1 and PD-1 from primary to metastatic endometrial cancer. Gynecol. Oncol. 2020, 157, 260–267. [Google Scholar] [CrossRef]
  35. Mo, Z.; Liu, J.; Zhang, Q.; Chen, Z.; Mei, J.; Liu, L.; Yang, S.; Li, H.; Zhou, L.; You, Z. Expression of PD-1, PD-L1 and PD-L2 is associated with differentiation status and histological type of endometrial cancer. Oncol. Lett. 2016, 12, 944–950. [Google Scholar] [CrossRef] [Green Version]
  36. Kamiya, S.; Kato, J.; Kamiya, T.; Yamashita, T.; Sumikawa, Y.; Hida, T.; Horimoto, K.; Sato, S.; Takahashi, H.; Sawada, M. Association between PD-L1 expression and lymph node metastasis in cutaneous squamous cell carcinoma. Asia-Pac. J. Clin. Oncol. 2020, 16, e108–e112. [Google Scholar] [CrossRef]
  37. Slater, N.A.; Googe, P.B. PD-L1 expression in cutaneous squamous cell carcinoma correlates with risk of metastasis. J. Cutan. Pathol. 2016, 43, 663–670. [Google Scholar] [CrossRef]
  38. García-Pedrero, J.M.; Martínez-Camblor, P.; Diaz-Coto, S.; Munguia-Calzada, P.; Vallina-Alvarez, A.; Vazquez-Lopez, F.; Rodrigo, J.P.; Santos-Juanes, J. Tumor programmed cell death ligand 1 expression correlates with nodal metastasis in patients with cutaneous squamous cell carcinoma of the head and neck. J. Am. Acad. Dermatol. 2017, 77, 527–533. [Google Scholar] [CrossRef]
  39. Le, D.T.; Uram, J.N.; Wang, H.; Bartlett, B.R.; Kemberling, H.; Eyring, A.D.; Skora, A.D.; Luber, B.S.; Azad, N.S.; Laheru, D. PD-1 blockade in tumors with mismatch-repair deficiency. N. Engl. J. Med. 2015, 372, 2509–2520. [Google Scholar] [CrossRef] [Green Version]
  40. Le, D.T.; Durham, J.N.; Smith, K.N.; Wang, H.; Bartlett, B.R.; Aulakh, L.K.; Lu, S.; Kemberling, H.; Wilt, C.; Luber, B.S. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017, 357, 409–413. [Google Scholar] [CrossRef] [Green Version]
  41. Sloan, E.A.; Ring, K.L.; Willis, B.C.; Modesitt, S.C.; Mills, A.M. PD-L1 expression in mismatch repair-deficient endometrial carcinomas, including lynch syndrome-associated and MLH1 promoter hypermethylated tumors. Am. J. Surg. Pathol. 2017, 41, 326–333. [Google Scholar] [CrossRef] [PubMed]
  42. Bhangoo, M.S.; Boasberg, P.; Mehta, P.; Elvin, J.A.; Ali, S.M.; Wu, W.; Klempner, S.J. Tumor mutational burden guides therapy in a treatment refractory POLE-mutant uterine carcinosarcoma. Oncologist 2018, 23, 518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Howitt, B.E.; Shukla, S.A.; Sholl, L.M.; Ritterhouse, L.L.; Watkins, J.C.; Rodig, S.; Stover, E.; Strickland, K.C.; D’Andrea, A.D.; Wu, C.J. Association of polymerase e–mutated and microsatellite-instable endometrial cancers with neoantigen load, number of tumor-infiltrating lymphocytes, and expression of PD-1 and PD-L1. JAMA Oncol. 2015, 1, 1319–1323. [Google Scholar] [CrossRef] [PubMed]
  44. Lu, L.; Li, Y.; Luo, R.; Xu, J.; Feng, J.; Wang, M. Prognostic and clinicopathological role of PD-L1 in endometrial cancer: A meta-analysis. Front. Oncol. 2020, 10, 632. [Google Scholar] [CrossRef] [PubMed]
Figure 1. PD-L1 immunohistochemical staining in endometrial cancer tissue microarray (TMA). Representative examples of tumors showing (A) high expression and (B) low expression (right panel) of PD-L1. (20×/0.70 objective on an Olympus BX 51 microscope. (Olympus America Inc, Center Valley, PA, USA) with the inset showing a 40× 0.85 aperture magnified view of the same TMA spot).
Figure 1. PD-L1 immunohistochemical staining in endometrial cancer tissue microarray (TMA). Representative examples of tumors showing (A) high expression and (B) low expression (right panel) of PD-L1. (20×/0.70 objective on an Olympus BX 51 microscope. (Olympus America Inc, Center Valley, PA, USA) with the inset showing a 40× 0.85 aperture magnified view of the same TMA spot).
Diagnostics 11 00394 g001
Figure 2. Survival Analysis of PD-L1 protein expression. Kaplan–Meier survival plot showing no statistically significant difference between PD-L1 positive and negative tumors for (A) overall survival, (B) recurrence-free survival and (C) disease-free survival.
Figure 2. Survival Analysis of PD-L1 protein expression. Kaplan–Meier survival plot showing no statistically significant difference between PD-L1 positive and negative tumors for (A) overall survival, (B) recurrence-free survival and (C) disease-free survival.
Diagnostics 11 00394 g002
Table 1. Clinico-pathological variables for the patient cohort (n = 440).
Table 1. Clinico-pathological variables for the patient cohort (n = 440).
Clinico-Pathological Parametern (%)
Age
Median59.3
Range(IQR) ^53.0–66.2
Histologic subtype
Type I387 (88.0)
Type II53 (12.0)
Myometrial invasion
High221 (50.2)
Low219 (49.8)
Histological grade
Well differentiated148 (33.6)
Moderately differentiated147 (33.4)
Poorly differentiated130 (29.6)
Unknown15 (3.4)
pT
T1308 (70.0)
T255 (12.5)
T358 (13.2)
T419 (4.3)
pN
N0410 (93.2)
N1-230 (6.8)
pM
M0417 (94.8)
M123 (5.2)
Tumor Stage
I285 (64.8)
II48 (10.9)
III70 (15.9)
IV37 (8.4)
Primary treatment
Surgery440 (100.0)
Adjuvant chemotherapy 84 (19.1)
Paclitaxel and Carboplatin69 (15.7)
Carboplatin only 15 (3.4)
Adjuvant Radiotherapy175 (39.8)
^ IQR—inter quartile range; pT—pathologic tumor size; pN—pathologic lymph node metastasis; pM—pathologic distant metastasis.
Table 2. Association of PD-L1 protein expression with clinico-pathological characteristics in endometrial cancer.
Table 2. Association of PD-L1 protein expression with clinico-pathological characteristics in endometrial cancer.
TotalPD-L1 PositivePD-L1 Negativep Value
No.%No.%No.%
No. of patients440 8318.935781.1
Age (years)
≤6023653.64217.819482.20.5387
>6020446.44120.116379.9
Histologic subtype
Type I38787.96316.332483.70.0005 *
Type II5312.12037.73362.3
Myometrial invasion
High22150.24922.217277.80.0741
Low21949.83415.518584.5
Grade
Grade 114834.82214.912685.10.0893
Grade 214734.62416.312383.7
Grade 313030.63224.69875.4
pT
T130870.05317.225582.80.0570
T25512.51425.54174.5
T35813.2813.85086.2
T4194.3842.11157.9
pN
N041093.27217.633882.40.0172 *
N1-N2306.81136.71963.3
pM
M041794.87718.534081.50.3822
M1235.2626.11773.9
Tumor Stage
I28564.84716.523883.50.2443
II4810.91122.93777.1
III7015.91420.05680.0
IV378.41129.72670.3
MMR IHC
dMMR5312.1815.14584.90.4435
pMMR38787.97519.431280.6
POLE mutation
Present20.500.02100.00.3576
Absent42999.58219.134780.9
5 year overall survival 90.9 87.00.6963
5 year recurrence-free survival 85.7 83.60.5351
5 year disease-free survival 78.9 76.80.7336
* significant p-value; IHC—immunohistochemistry; dMMR—deficient mismatch repair; pMMR—proficient mismatch repair; POLE—DNA polymerase epsilon.
Table 3. Multivariate analysis to assess relationship between lymph node metastasis and clinico-pathological characteristics.
Table 3. Multivariate analysis to assess relationship between lymph node metastasis and clinico-pathological characteristics.
Clinico-Pathological VariablesOdds Ratio95% CIp-Value
Age
Age>60 years (vs. ≤60 years)0.650.29–1.470.3020
Histologic grade
Grade 3 (vs. Grade 1–2)2.140.92–4.960.0755
Myometrial invasion
High (vs. Low)2.841.01–7.940.0473 *
pT
T3-4 (vs. T1-2)2.420.90–6.490.0801
pM
M1 (VS. M0)0.630.03–11.740.7548
Tumor Stage
IV (vs I–III)0.280.03–2.750.2766
MMR statusd
MMR (vs. pMMR)1.200.41–3.580.7376
PD-L1 expression
Positive (vs. Negative)2.941.26–6.840.0123 *
* significant p-value.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Siraj, A.K.; Parvathareddy, S.K.; Annaiyappanaidu, P.; Siraj, N.; Al-Rasheed, M.; Al-Badawi, I.A.; Al-Dayel, F.; Al-Kuraya, K.S. PD-L1 Expression Is an Independent Marker for Lymph Node Metastasis in Middle Eastern Endometrial Cancer. Diagnostics 2021, 11, 394. https://doi.org/10.3390/diagnostics11030394

AMA Style

Siraj AK, Parvathareddy SK, Annaiyappanaidu P, Siraj N, Al-Rasheed M, Al-Badawi IA, Al-Dayel F, Al-Kuraya KS. PD-L1 Expression Is an Independent Marker for Lymph Node Metastasis in Middle Eastern Endometrial Cancer. Diagnostics. 2021; 11(3):394. https://doi.org/10.3390/diagnostics11030394

Chicago/Turabian Style

Siraj, Abdul K., Sandeep Kumar Parvathareddy, Padmanaban Annaiyappanaidu, Nabil Siraj, Maha Al-Rasheed, Ismail A. Al-Badawi, Fouad Al-Dayel, and Khawla S. Al-Kuraya. 2021. "PD-L1 Expression Is an Independent Marker for Lymph Node Metastasis in Middle Eastern Endometrial Cancer" Diagnostics 11, no. 3: 394. https://doi.org/10.3390/diagnostics11030394

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

Article Metrics

Back to TopTop