Clinical and prognostic significance of sPD-1 and sPD-L1 in ovarian cancer

Abstract

Introduction. Ovarian cancer is considered one of the most aggressive gynecological malignancies. Unfortunately, the methods that are currently considered the gold standard for the treatment of ovarian cancer, in most cases, lead to frequent relapses and progression of the disease after a limited period of time. Since the prognosis of this disease is often associated with the infiltration of the tumor by immune cells, immune checkpoint inhibitors can be used to activate the antitumor immune response in the treatment of tumors of this type. However, to date there is insufficient data to effectively select patients who can fully respond to this therapy.

The aim of the study is to study the level of soluble forms of PD-1 and PD-L1 (sPD-1 and sPD-L1) in ovarian tumors, their clinical and prognostic significance.

Material and methods. The study included 136 patients and 35 healthy women who were treated at the N.N. Blokhin NMRC of Oncology, MOH of Russia. The clinical diagnosis in all patients was confirmed by morphological examination of the tumor according to the International Histological Classification of Ovarian Tumors, adopted by WHO in 2014. The study included patients with epithelial ovarian cancer of three histological types: serous (77), endometrioid (13) and mucinous (8); benign neoplasms (22) and borderline tumors (10) of the ovaries, as well as non-epithelial ovarian tumors: dysgerminoma (3), sertolioma (1) and granulosa cell tumor (2). The levels of sPD-L1 and sPD-1 proteins was determined in blood plasma obtained according to standard methods before the start of specific treatment using an enzyme-linked immunosorbent assay. Statistical analysis of the obtained results was carried out using GraphPad Prizm v. 10. When comparing indicators and analyzing their relationships, nonparametric Mann–Whitney and Kruskal–Wallis tests and Spearman’s rank correlation coefficient were used. Overall survival analysis was performed by constructing survival curves using the Kaplan–Meier method. Differences were considered statistically significant at p < 0.05.

Results. The study showed that the median level of sPD-1 in the blood plasma of healthy women in the group of comparison was 43.8 (33.6–54.6) pg/ml, and in the group of patients with ovarian cancer – 47.1 (35.1–61. 6) pg/ml. The median level of sPD-L1 in blood plasma in the group of comparison was 60.8 (26.2–91.7) pg/ml and was not significantly higher than in patients with ovarian cancer 40.5 (15.7–78) pg/ml (p = 0.075). The level of sPD-L1 in blood plasma was significantly reduced in benign and non-epithelial malignant ovarian tumors (p = 0.022 and p = 0.006, respectively) compared to the group of comparison. It should be noted that the level of sPD-L1 in patients with nonepithelial ovarian tumors was significantly lower compared to epithelial tumors (p = 0.02). For sPD-L1 level, there is a significant association with tumor progression factors. Thus, the level of sPD-L1 is significantly higher in the later stages of the disease, in the case of larger tumors, in the presence of ascites and regional metastases. The analysis of prognostic significance showed that a high level of sPD-L1 protein in the plasma of patients with malignant ovarian tumors is a significant unfavorable prognostic factor (HR = 2.28; p = 0.023).

Conclusion. The study showed that the level of sPD-L1 in the blood plasma of patients with ovarian tumors reflects the extent of the tumor process and can potentially be an effective tool for monitoring the course of this disease. Also, our results and literature data indicate the prospects of using sPD-L1 level as a marker of the effectiveness of OC therapy, and therefore the prognosis of the disease.

Keywords:ovarian cancer; sPD-1; sPD-L1; prognosis

For citation: Kushlinskii N.E., Kovaleva O.V., Grachev A.N., Tsekatunov D.A., Kushlinskiy D.N., Alferov A.A., Kuzmin Yu.B., Shvedova A.D., Klimanov I.A., Stilidi I.S. Clinical and prognostic significance of sPD-1/sPD-L1 in ovarian cancer. Immunology. 2024; 45 (2): 183–92. DOI: https://doi.org/10.33029/1816-2134-2024-45-2-183-192

Funding. The study had no sponsor support.

Conflict of interests. The authors declare no conflict of interests.

Authors’ contributions. Concept of the study – Kushlinskii N.E., Stilidi I.S.; material collection and processing – Kushlinskiy D.N., Alferov A.A., Shvedova A.D.; histological examination of tumors – Tsekatunov D.A.; statistical processing, text writing – Kovaleva O.V., Grachev A.N., Klimanov I.A.

References

1. Dumitru A., Dobrica E.C., Croitoru A., Cretoiu S.M., Gaspar B.S. Focus on PD-1/PD-L1 as a Therapeutic Target in Ovarian Cancer. Int. J. Mol. Sci. 2022; 23 (20): 12067. DOI: https://doi.org/10.3390/ijms232012067

2. Hamanishi J., Mandai M., Ikeda T., Minami M., Kawaguchi A., Murayama T., Kanai M., Mori Y., Matsumoto S., Chikuma S., et al. Safety and Antitumor Activity of Anti-PD-1 Antibody, Nivolumab, in Patients With Platinum-Resistant Ovarian Cancer. J. Clin. Oncol. 2015; 33 (34): 4015-22. DOI: https://doi.org/10.1200/JCO.2015.62.3397

3. Hamanishi J., Mandai M., Iwasaki M., Okazaki T., Tanaka Y., Yamaguchi K., Higuchi T., Yagi H., Takakura K., Minato N., et al. Programmed cell death 1 ligand 1 and tumor-infiltrating CD8+ T lymphocytes are prognostic factors of human ovarian cancer. Proc. Natl. Acad. Sci. U S A. 2007; 104 (9): 3360-5. DOI: https://doi.org/10.1073/pnas.0611533104

4. Webb J.R., Milne K., Kroeger D.R., Nelson B.H. PD-L1 expression is associated with tumor-infiltrating T cells and favorable prognosis in high-grade serous ovarian cancer. Gynecol Oncol. 2016; 141 (2): 293-302. DOI: https://doi.org/10.1016/j.ygyno.2016.03.008

5. Zhao B., Zhao H., Zhao J. Efficacy of PD-1/PD-L1 blockade monotherapy in clinical trials. Ther. Adv. Med. Oncol. 2020; 12: 1758835920937612. DOI: https://doi.org/10.1177/1758835920937612

6. Chen X.J., Yuan S.Q., Duan J.L., Chen Y.M., Chen S., Wang Y., Li Y.F. The Value of PD-L1 Expression in Predicting the Efficacy of Anti-PD-1 or Anti-PD-L1 Therapy in Patients with Cancer: A Systematic Review and Meta-Analysis. Dis Markers. 2020; 2020: 6717912. DOI: https://doi.org/10.1155/2020/6717912

7. Swiderska J., Kozlowski M., Nowak K., Rychlicka M., Branecka-Wozniak D., Kwiatkowski S., Pius-Sadowska E., Machalinski B., Cymbaluk-Ploska A. Clinical Relevance of Soluble Forms of Immune Checkpoint Molecules sPD-1, sPD-L1, and sCTLA-4 in the Diagnosis and Prognosis of Ovarian Cancer. Diagnostics (Basel). 2022; 12 (1): 189. DOI: https://doi.org/10.3390/diagnostics12010189

8. Niu M., Liu Y., Yi M., Jiao D., Wu K. Biological Characteristics and Clinical Significance of Soluble PD-1/PD-L1 and Exosomal PD-L1 in Cancer. Front Immunol. 2022; 13: 827921. DOI: https://doi.org/10.3389/fimmu.2022.827921

9. Oh S.Y., Kim S., Keam B., Kim T.M., Kim D.W., Heo D.S. Soluble PD-L1 is a predictive and prognostic biomarker in advanced cancer patients who receive immune checkpoint blockade treatment. Sci. Rep. 2021; 11 (1): 19712. DOI: https://doi.org/10.1038/s41598-021-99311-y

10. Chiarucci C., Cannito S., Daffina M.G., Amato G., Giacobini G., Cutaia O., Lofiego M.F., Fazio C., Giannarelli D., Danielli R., et al. Circulating Levels of PD-L1 in Mesothelioma Patients from the NIBIT-MESO-1 Study: Correlation with Survival. Cancers (Basel). 2020; 12 (2): 361. DOI: https://doi.org/10.3390/cancers12020361

11. Ding X.C., Wang L.L., Zhu Y.F., Li Y.D., Nie S.L., Yang J., Liang H., Weichselbaum R.R., Yu J.M., Hu M. The Change of Soluble Programmed Cell Death-Ligand 1 in Glioma Patients Receiving Radiotherapy and Its Impact on Clinical Outcomes. Front Immunol. 2020; 11: 580335. DOI: https://doi.org/10.3389/fimmu.2020.580335

12. Finkelmeier F., Canli O., Tal A., Pleli T., Trojan J., Schmidt M., Kronenberger B., Zeuzem S., Piiper A., Greten F.R., Waidmann O. High levels of the soluble programmed death-ligand (sPD-L1) identify hepatocellular carcinoma patients with a poor prognosis. Eur. J. Cancer. 2016; 59: 152-9. DOI: https://doi.org/10.1016/j.ejca.2016.03.002

13. Bailly C., Thuru X., Quesnel B. Soluble Programmed Death Ligand-1 (sPD-L1): A Pool of Circulating Proteins Implicated in Health and Diseases. Cancers (Basel). 2021; 13 (12): 3034. DOI: https://doi.org/10.3390/cancers13123034

14. Mortensen J.B., Monrad I., Enemark M.B., Ludvigsen M., Kamper P., Bjerre M., d’Amore F. Soluble programmed cell death protein 1 (sPD-1) and the soluble programmed cell death ligands 1 and 2 (sPD-L1 and sPD-L2) in lymphoid malignancies. Eur. J. Haematol. 2021; 107 (1): 81-91. DOI: https://doi.org/10.1111/ejh.13621

15. Fanale D., Brando C., Corsini L.R., Cutaia S., Di Donna M.C., Randazzo U., Filorizzo C., Lisanti C., Magrin L., Gurrera V., et al. Low plasma PD-L1 levels, early tumor onset and absence of peritoneal carcinomatosis improve prognosis of women with advanced high-grade serous ovarian cancer. BMC cancer. 2023; 23 (1): 437. DOI: https://doi.org/10.1186/s12885-023-10911-5

16. Buderath P., Schwich E., Jensen C., Horn P.A., Kimmig R., Kasimir-Bauer S., Rebmann V. Soluble Programmed Death Receptor Ligands sPD-L1 and sPD-L2 as Liquid Biopsy Markers for Prognosis and Platinum Response in Epithelial Ovarian Cancer. Front. Oncol. 2019; 9: 1015. DOI: https://doi.org/10.3389/fonc.2019.01015

17. Larrinaga G., Solano-Iturri J.D., Errarte P., Unda M., Loizaga-Iriarte A., Perez-Fernandez A., Echevarria E., Asumendi A., Manini C., Angulo J.C., Lopez J.I. Soluble PD-L1 Is an Independent Prognostic Factor in Clear Cell Renal Cell Carcinoma. Cancers (Basel). 2021; 13 (4): 667. DOI: https://doi.org/10.3390/cancers13040667

18. Gershtein E.S., Mochalova A.S., Korotkova E.A., Samoilova E.V., Vashketova O.I., Kuzmin Yu.B., Sokolov N.Yu., Kushlinskii N.E. Dynamics of soluble forms of immune checkpoint components PD-1/PD-L1/B7-H3, CD314/ULBP1 and HLA-G in the peripheral blood of melanoma patients treated with blockers of the programmed cell death protein PD-1. Bull. Expert. Biol. Med. 2023; 175 (4): 482-7. DOI: https://doi.org/10.1007/s10517-023-05891-2 (in Russian)

19. Barron C.C., Stefanova I., Cha Y., Elsolh K., Zereshkian A., Gaafour N., McWhirter E. Chronic immune-related adverse events in patients with cancer receiving immune checkpoint inhibitors: a systematic review. J. Immunother. Cancer. 2023; 11 (8): e006500. DOI: https://doi.org/10.1136/jitc-2022-006500

20. Quandt Z., Young A., Anderson M. Immune checkpoint inhibitor diabetes mellitus: a novel form of autoimmune diabetes. Clin. Exp. Immunol. 2020; 200 (2): 131-140. DOI: https://doi.org/10.1111/cei.13424

21. Borunova A.A., Shoua E.K., Zakharova E.N., Chertkova A.I., Kadagidze Z.G., Shoua I.B., Davydova T.V., Kushlinskii N.E., Babkina I .V., Osipova N.G., Ternavsky A.P., Doroshev I.A., Kononets P.V., Stilidi I.S., Zabotina T.N. Features of cellular immunity of medical workers in the first wave of the SARS-COV2 infection pandemic. Immunology. 2023; 44 (6): 776-87. DOI: https://doi.org/10.33029/0206-4952-2023-44-6-776-787 (in Russian)

22. Pacholczak-Madej R., Kosałka-Węgiel J., Kuszmiersz P., Mituś J.W., Püsküllüoğlu M., Grela-Wojewoda A., Korkosz M., BazanSocha S. Immune Checkpoint Inhibitor Related Rheumatological Complications: Cooperation between Rheumatologists and Oncologists. Int. J. Environ Res. Public Health. 2023; 20 (6): 4926. DOI: https://doi.org/10.3390/ijerph20064926

23. Pichugin A.V., Podsvirova S.A., Ushakova E.I., Spirin D.M., Lebedeva E.S., Ataullakhanov R.I. Reprogramming of myeloid cells of the tumor microenvironment increases the effi ciency of CTLA-4 and PD-1 blockade in experimental malignant melanoma immunotherapy. Immunologiya. 2022; 43 (6): 673–90. DOI: https://doi.org/10.33029/0206-4952-2022-43-6-673-690 (in Russian)

All articles in our journal are distributed under the Creative Commons Attribution 4.0 International License (CC BY 4.0 license)


JOURNALS of «GEOTAR-Media»