Peripheral blood CD8+-lymphocytes populations and its value for direct and outlying treatment results in triple negative breast cancer patients

Abstract

Introduction. Triple negative breast cancer (TN BC) is characterized by an aggressive course and a high likelihood of the disease recurrence. An important role in the regulation of the growth of malignant neoplasms, as well as in the effectiveness of various types of antitumor therapy, is played by the immune system. Establishing the relationship of some CD8-positive lymphocyte populations with the effectiveness of treating patients with breast cancer may help establish reliable immunological predictive and prognostic factors and increase the effectiveness of treatment for patients.

The aim of the study was to determine the value of the original number of CD8+ peripheral blood lymphocytes (CD8+, CD3+CD8+, CD8+CD28+, CD8+CD28, CD3CD8+) cells for immediate (degree of therapeutic tumor pathomorphosis) and distant (progression of disease and survival) of treatment results of patients with three-year negative breast cancer.

Material and methods. In patients with II–III stages of TN BC (n = 72, age median 50 years) before the start of neoadjuvant chemotherapy (NACT) (cisplatin + paclitaxel), the percentage of peripheral blood CD8+, CD3+CD8+, CD8+CD28+, CD8+CD28, CD3CD8+ lymphocytes was determined using a multi-parameter cytometric analysis. The relationship of the initial number of these cell populations with the degree of therapeutic pathomorphosis (TP), disease progression, and patient survival was studied. The observation period was 60 months.

Results. In patients with TP grade 4, in patients without disease progression and surviving patients, the percentage of CD8+CD28+-lymphocytes and the ratio of CD8+CD28/CD8+CD28 were statistically significantly higher before treatment than in patients with TP grade 1–3, recurrent patients and deceased patients. An increase of the initial number of CD8+CD28+-lymphocytes and the СD8+CD28/CD8+CD28 ratio was associated with an increase in relapse-free (RFS) and overall survival (OS) of patients compared with patients in whom these showings were reduced. In patients who died during the observation period, the number of CD3CD8+-lymphocytes was increased prior to chemotherapy. An increase in the number of these cells was also associated with a decrease in OS.

Conclusion. Populations of CD8+CD28+ peripheral blood lymphocytes and the ratio of CD8+CD28/CD8+CD28 cells could be possible predictive factors for patients with II–III stages of TNBC receiving NACT (cisplatin and paclitaxel).

Keywords:TN breast cancer; CD8+-lymphocyte populations; neoadjuvant chemotherapy; degree of therapeutic pathomorphosis; relapse-free survival; overall survival

For citation: Zabotina T.N., Chertkova A.I., Kadagidze Z.G., Borunova A.A., Shoua E.K., Gordeeva O.O., Meshche-ryakov А.А. Peripheral blood CD8+-lymphocytes populations and its value for direct and outlying treatment results in triple negative breast cancer patients. Immunologiya. 2020; 41 (4): 326-36. DOI: https://doi.org/10.33029/0206-4952-2020-41-3-326-336 (in Russian)

Funding. The study had no sponsor support.

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

References

1. Reis-Filho J.S., Tutt A.N.J. Triple negative tumours: a critical review. Histopathology. 2008; 52: 108–18.

2. Kumar P., Aggarwal R. An overview of triple-negative breast cancer. Arch. Gynecol. Obstet. 2016; 293 (2): 247–69.

3. Bianchini G., Balko J.M., Mayer I.A., Sanders M.E., Gianni L. Triple-negative breast cancer: challenges and opportunities of a heterogeneous disease. Nat. Rev. Clin. Oncol. 2016; 13 (11): 674–90.

4. Rivenbark A.G., O’Connor S.M., Coleman W.B. Molecular and cellular heterogeneity in breast cancer: challenges for personalized medicine. Am. J. Pathol. 2013; 183 (4): 1113–24.

5. Ali A.M., Ansari J.A.K., El-Aziz N.M.A., Abozeed W.N., Warith A.M.A., Alsaleh K. Triple negative breast cancer: a tale of two decades anticancer agents. Med. Chem. 2017; 17 (4): 491–9.

6. Voena C., Chiarle R. Advances in cancer immunology and cancer immunotherapy. Discov. Med. 2016; 21 (114): 125–33.

7. Wu J., Waxman D.J. Immunogenic chemotherapy: dose and schedule dependence and combination with immunotherapy. Cancer Lett. 2018; 419: 210–21.

8. Emens L.A., Middleton G. The interplay of immunotherapy and chemotherapy: harnessing potential synergies. Cancer Immunol. Res. 2015; 3 (5): 436–43.

9. Ye J.C., Formenti S.C. Integration of radiation and immunotherapy in breast cancer - treatment implications. Breast. 2018; 38: 66–74.

10. Zitvogel L., Kepp O., Kroemer G. Immune parameters affecting the efficacy of chemotherapeutic regimens. Nat. Rev. Clin. Oncol. 2011; 8 (3): 151–60.

11. Behren A., Thompson E.W., Anderson R.L., Ferrao P.T. Editorial: cancer plasticity and the microenvironment: implications for immunity and therapy response. Front. Oncol. 2019; 9: 276.

12. Denkert C., von Minckwitz G., Darb-Esfahani S., Lederer B., Heppner B.I., Weber K.E., et al. Tumour-infiltrating lymphocytes and prognosis in different subtypes of breast cancer: a pooled analysis of 3771 patients treated with neoadjuvant therapy. Lancet Oncol. 2018; 19 (1): 40–50.

13. Stovgaard E.S., Nielsen D., Hogdall E., Balslev E. Triple negative breast cancer – prognostic role of immune-related factors: a systematic review. Acta Oncol, 2018; 57 (1): 74–82.

14. Vihervuori H., Autere T.A., Repo H., Kurki S., Kallio L., Lintunen M.M. Tumor-infiltrating lymphocytes and CD8+ T cells predict survival of triple-negative breast cancer. J. Cancer Res. Clin. Oncol. 2019; 145 (12): 3105–14.

15. Baxevanis C.N., Sofopoulos M., Fortis S.P., Perez S.A. The role of immune infiltrates as prognostic biomarkers in patients with breast cancer. Cancer Immunol. Immunother. 2019; 68: 1671–80.

16. McAllister S.S., Weinberg R.A. The tumour-induced systemic environment as a critical regulator of cancer progression and metastasis. Nat. Cell. Biol. 2014; 16 (8): 717–27.

17. Spitzer M.H., Carmi Y., Reticker-Flynn N.E., Kwek S.S., Madhireddy D., Martins M.M., et al. Systemic immunity is required for effective cancer immunotherapy. Cell. 2017; 168 (3): 487–502.

18. Wang L., Simons D.L., Lu X., Tu T.Y., Avalos C., Chang A.Y., et al. Breast cancer induces systemic immune changes on cytokine signaling in peripheral blood monocytes and lymphocytes. EBioMedicine. 2020; 52: 102631.

19. He Q., Li G., Ji X., Ma L., Wang X., Li Y., et al. Impact of the immune cell population in peripheral blood on response and survival in patients receiving neoadjuvant chemotherapy for advanced gastric cancer. Tumour Biol. 2017; 39 (5): 1–8.

20. Lin K.R., Pang D.M., Jin Y.B., Hu Q., Pan Y.M., Cui J.H., et al. Circulating CD8+ T-cell repertoires reveal the biological characteristics of tumors and clinical responses to chemotherapy in breast cancer patients. Cancer Immunol. Immunother. 2018; 67 (11): 1743–52.

21. Zenan H., Zixiong L., Zhicheng Y., Mei H., Xiongbin Y., Tiantian W., et al. Clinical prognostic evaluation of immunocytes in different molecular subtypes of breast cancer. J. Cell. Physiol. 2019; 234 (11): 20 584–602.

22. Péguillet I., Milder M., Louis D., Vincent-Salomon A., Dorval T., Piperno-Neumann S., et al. High numbers of differentiated effector CD4 T cells are found in patients with cancer and correlate with clinical response after neoadjuvant therapy of breast cancer. Cancer Res. 2014; 74 (8): 2204–16.

23. Disis M.L., Stanton S.E. Immunotherapy in breast cancer: an introduction. Breast. 2018; 37: 196–9.

24. Huff W.X., Kwon J.H., Henriquez M., Fetcko K., Dey M. The evolving role of CD8+CD28- immunosenescent T cells in cancer immunology. Int. J. Mol. Sci. 2019; 20 (11): E2810.

25. Tabakov D.V., Zabotina T.N., Zakharova E.N., Borunova A.A., Korotkova O.V., Chertkova A.I., et al. The subpopulation balance of effector cells in the peripheral blood of cancer patients. Immunologiya. 2019; 40 (3): 20–7. DOI: https://doi.org/10.24411/0206-4952-2019-13002 (in Russian)

26. Wang K., Xu J., Zhang T., Xue D. Tumor-infiltrating lymphocytes in breast cancer predict the response to chemotherapy and survival outcome: a meta-analysis. Oncotarget. 2016; 7 (28): 44288–98.

27. He J. Folate-modified chitosan nanoparticles containing the ip-10 gene enhance melanoma-specific cytotoxic CD8(+)CD28(+) T lymphocyte responses. Theranostics. 2016; 6 (5): 752–61.

28. Fiorentini S., Licenziati S. Alessandri G., Castelli .F, Caligaris S., Bonafede M., et al. CD11b expression identifies CD8+CD28+ T lymphocytes with phenotype and function of both naive/memory and effector cells. J. Immunol. 2001; 166 (2): 900–7.

29. Song G., Wang X., Jia J., Yuan Y., Wan F., Zhou X., et al. Elevated level of peripheral CD8(+)CD28(-) T lymphocytes are an independent predictor of progression-free survival in patients with metastatic breast cancer during the course of chemotherapy. Cancer Immunol. Immunother. 2013; 62 (6): 1123–30.

30. Wang S., Chen L. T lymphocyte co-signaling pathways of the B7-CD28 family. Cell. Mol. Immunol. 2004; 1 (1): 37–42.

31. Chen X., Liu Q., Xiang A.P. CD8+CD28 T cells: Not only age-related cells but a subset of regulatory T cells. Cell. Mol. Immunol. 2018; 15: 734–6.

32. Song Q., Ren J., Zhou X., Wang X., Song G., Hobeika A., et al. Circulating CD8+CD28- suppressor T cells tied to poorer prognosis among metastatic breast cancer patients receiving adoptive T-cell therapy: a cohort study. Cytotherapy. 2018; 20 (1): 126–33.

33. Crouse J., Xu H.C., Lang P.A., Oxenius A. NK cells regulating T cell responses: mechanisms and outcome. Trends Immunol. 2015; 36 (1): 49–58.

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»