Skip to main content

Advertisement

Log in

The upregulation of immune checkpoints after photodynamic therapy reducing immune effect for treating breast cancer

  • Original Article
  • Published:
Lasers in Medical Science Aims and scope Submit manuscript

Abstract

The immune effect induced by photodynamic therapy (PDT) has a limited effect on breast tumor. This study hypothesized that suppressive immune checkpoints on T cells might upregulate after PDT, which may reduce the antitumor effect of PDT for treating breast tumor. This study explored the alteration of immune checkpoint for the first time. A bilateral subcutaneous transplanted breast tumor mice model was established, and right tumors imitated primary tumors, and left tumors imitated distant tumors. Primary tumors were treated with PDT mediated by hematoporphyrin derivatives (HpD-PDT). Costimulatory molecules (ICOS, OX40, and 4-1BB) and immune checkpoints (PD1, LAG-3, CTLA-4, TIM-3, TIGIT) on tumor infiltrating T cells after HpD-PDT were analyzed by flow cytometry. Antitumor and immune effects were also assessed after HpD-PDT combined with anti-PD1 and LAG-3 antibodies. Primary tumors were suppressed, but distant tumors could not be inhibited after HpD-PDT. The number of T cells was increased, but function did not enhance after HpD-PDT. Additionally, costimulatory molecules (ICOS, OX40, and 4-1BB) were not elevated, but the suppressive immune checkpoints on tumor infiltrating T cells were upregulated after HpD-PDT. Notably, PD1+ LAG-3+ CD4+ T and PD1+ LAG-3+ CD8+ T cells were significantly increased. When PD1 and LAG-3 blockade combined with HpD-PDT, both primary and distant tumors were significantly suppressed, and antitumor immune effects were significantly enhanced. HpD-PDT could upregulate the PD1+ LAG-3+ CD4+ T and PD1+ LAG-3+ CD8+ T cells. Dual blockade of PD1 and LAG-3 immune checkpoints can enhance the antitumor effect of HpD-PDT.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

Data Availability

All data about this article are availability for this journal.

References

  1. Siegel RL, Miller KD, Fuchs HE, Jemal A (2022) Cancer statistics, 2022. CA: Cancer J Clin 72:7–33. https://doi.org/10.3322/caac.21708.

  2. Cao M, Chen W (2021) Interpretation on the global cancer statistics of GLOBOCAN 2020. Chinese Med Frontier J 13:63–69

    Google Scholar 

  3. Yang X, Zhang Y (2019) Advances in the treatment of breast cancer with cutaneous metastasis. Chin J Clin Oncol 46:1237–1240

    Google Scholar 

  4. Ostanska E, Aebisher D, Bartusik-Aebisher D (2021) The potential of photodynamic therapy in current breast cancer treatment methodologies. Biomed Pharmacot Biomed Pharmacot 137:111302. https://doi.org/10.1016/j.biopha.2021.111302

    Article  CAS  Google Scholar 

  5. Li B, Xie S, Wilson BC (2009) Advances in dosimetry of photodynamic therapy. Progress in Biochem Biophysics 36:676–683

    Article  Google Scholar 

  6. Castano AP, Mroz P, Hamblin MR (2006) Photodynamic therapy and anti-tumour immunity. Nat Rev Cancer 6:535–545. https://doi.org/10.1038/nrc1894

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Donohoe C, Senge MO, Arnaut LG, Gomes-da-Silva LC (2019) Cell death in photodynamic therapy: from oxidative stress to anti-tumor immunity. Biochim Biophys Acta 1872:188308. https://doi.org/10.1016/j.bbcan.2019.07.003

    Article  CAS  Google Scholar 

  8. Garg AD, Vandenberk L, Koks C, Verschuere T, Boon L, Van Gool SW, Agostinis P (2016) Dendritic cell vaccines based on immunogenic cell death elicit danger signals and T cell-driven rejection of high-grade glioma. Sci Translational Med 8:328ra327. https://doi.org/10.1126/scitranslmed.aae0105

    Article  CAS  Google Scholar 

  9. Kessel D (2019) Apoptosis, paraptosis and autophagy: death and survival pathways associated with photodynamic therapy. Photochem Photobiol 95:119–125. https://doi.org/10.1111/php.12952

    Article  CAS  PubMed  Google Scholar 

  10. Kessel D, Oleinick NL (2010) Photodynamic therapy and cell death pathways. Methods Molecul Biol (Clifton, NJ) 635:35–46. https://doi.org/10.1007/978-1-60761-697-9_3

    Article  CAS  Google Scholar 

  11. Anzengruber F, Avci P, de Freitas LF, Hamblin MR (2015) T-cell mediated anti-tumor immunity after photodynamic therapy: why does it not always work and how can we improve it? Photochem Photobiol Sci: Official J Europ Photochem Assoc Europ Soc Photobiol 14:1492–1509. https://doi.org/10.1039/c4pp00455h

    Article  CAS  Google Scholar 

  12. Wang Y, Gu Y (2017) Advances in clinical application and study on tumor-targeted photodynamic therapy. Chin J Laser Med Surg 26:279–287

    CAS  Google Scholar 

  13. Long S, Zhao YB, Xu YY, Li H, Zhao HY, Chen DF, Zeng J, Qiu HX, Li XS, Gu Y (2022) Immune response induced by hematoporphyrin derivatives mediated photodynamic therapy: immunogenic cell death and elevated costimulatory molecules. J Innov Opt Health Sci 15:12. https://doi.org/10.1142/s1793545822400028

    Article  CAS  Google Scholar 

  14. Choi Y, Shi Y, Haymaker CL, Naing A, Ciliberto G, Hajjar J (2020) T-cell agonists in cancer immunotherapy. J Immunother Cancer 8:e000966. https://doi.org/10.1136/jitc-2020-000966

    Article  PubMed  PubMed Central  Google Scholar 

  15. Smt A, Vsn A, Jd A, Eea B (2020) Immune checkpoints in the tumor microenvironment. Semin Cancer Biol 65:1–12. https://doi.org/10.1016/j.semcancer.2019.06.021

    Article  CAS  Google Scholar 

  16. Henderson BW, Busch TM, Snyder JW (2006) Fluence rate as a modulator of PDT mechanisms. Lasers Surg Med 38:489–493. https://doi.org/10.1002/lsm.20327

    Article  PubMed  Google Scholar 

  17. Coutier S, Bezdetnaya LN, Foster TH, Parache RM, Guillemin F (2002) Effect of irradiation fluence rate on the efficacy of photodynamic therapy and tumor oxygenation in meta-tetra (hydroxyphenyl) chlorin (mTHPC)-sensitized Ht29 xenografts in nude mice. Radiat Res 158:339–345. https://doi.org/10.1667/0033-7587(2002)158[0339:eoifro]2.0.co;2

    Article  CAS  PubMed  Google Scholar 

  18. Nath S, Obaid G, Hasan T (2019) The course of immune stimulation by photodynamic therapy: bridging fundamentals of photochemically induced immunogenic cell death to the enrichment of T-Cell repertoire. Photochem Photobiol 95:1288–1305. https://doi.org/10.1111/php.13173

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Dong C, Juedes AE, Temann UA, Shresta S, Allison JP, Ruddle NH, Flavell RA (2001) ICOS co-stimulatory receptor is essential for T-cell activation and function. Nature 409:97–101. https://doi.org/10.1038/35051100

    Article  CAS  PubMed  Google Scholar 

  20. Croft M (2009) The role of TNF superfamily members in T-cell function and diseases. Nat Rev Immunol 9:271–285. https://doi.org/10.1038/nri2526

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Sasidharan Nair V, El Salhat H, Taha RZ, John A, Ali BR, Elkord E (2018) DNA methylation and repressive H3k9 and H3k27 trimethylation in the promoter regions of Pd-1, Ctla-4, Tim-3, Lag-3, Tigit, and Pd-L1 genes in human primary breast cancer. Clin Epigenetics 10:1–12. https://doi.org/10.1186/s13148-018-0512-1

    Article  CAS  Google Scholar 

  22. Cramer GM, Moon EK, Cengel KA, Busch TM (2020) Photodynamic therapy and immune checkpoint blockade. Photochem Photobiol 96:954–961. https://doi.org/10.1111/php.13300

    Article  CAS  PubMed  Google Scholar 

  23. Fourcade J, Kudela P, Sun Z, Shen H, Land SR, Lenzner D, Guillaume P, Luescher IF, Sander C, Ferrone S, Kirkwood JM, Zarour HM (2009) Pd-1 is a regulator of Ny-Eso-1-Specific Cd8+ T Cell expansion in melanoma patients. J Immunol 182:5240–5249. https://doi.org/10.4049/jimmunol.0803245

    Article  CAS  PubMed  Google Scholar 

  24. Zahm CD, Moseman JE, Delmastro LE, Mcneel DG (2021) Pd-1 and Lag-3 blockade improve anti-tumor vaccine efficacy. Oncoimmunology 10:1912892. https://doi.org/10.1080/2162402X.2021.1912892

    Article  PubMed  PubMed Central  Google Scholar 

  25. Zhao Y, Liu X, Liu X, Yu J, Bai X, Wu X, Guo X, Liu Z, Liu X (2022) Combination of phototherapy with immune checkpoint blockade: theory and practice in cancer. Front Immunol 13:955920. https://doi.org/10.3389/fimmu.2022.955920

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. He X, Xu C (2020) Immune checkpoint signaling and cancer immunotherapy. Cell Res 30:660–669. https://doi.org/10.1038/s41422-020-0343-4

    Article  PubMed  PubMed Central  Google Scholar 

  27. Du H, Yi Z, Wang L, Li Z, Niu B, Ren G (2020) The co-expression characteristics of LAG3 and PD-1 on the T cells of patients with breast cancer reveal a new therapeutic strategy. Int Immunopharmacol 78:106113. https://doi.org/10.1016/j.intimp.2019.106113

    Article  CAS  PubMed  Google Scholar 

  28. Sharma P, Hu-Lieskovan S, Wargo JA, Ribas A (2017) Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell 168:707–723. https://doi.org/10.1016/j.cell.2017.01.017

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Spranger S, Gajewski TF (2018) Impact of oncogenic pathways on evasion of antitumour immune responses. Nat Rev Cancer 18:139–147. https://doi.org/10.1038/nrc.2017.117

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Mimura K, Kua LF, Xiao JF, Asuncion BR, Nakayama Y, Syn N, Fazreen Z, Soong R, Kono K, Yong WP (2021) Combined inhibition of Pd-1/Pd-L1, Lag-3, and Tim-3 axes augments antitumor immunity in gastric cancer-T cell coculture models. gastric cancer : official. J Int Gastric Cancer Ass Japanese Gastric Cancer Ass 24:611–623. https://doi.org/10.1007/s10120-020-01151-8

    Article  CAS  Google Scholar 

Download references

Funding

This work was supported by the National Natural Science Foundation of China (61975239), the Beijing Natural Science Foundation (7222174), the Beijing Science and Technology New Star Program (Z211100002121168), and the Medical and Health Technology Innovation Project of the Chinese Academy of Medical Sciences (2019-I2M-5–061).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: Shan Long, Xiaosong Li, and Ying Gu; data curation: Haixia Qiu, Hongyou Zhao, Jing Zeng, and Defu Chen; investigation: Shan Long, Yibing Zhao, Yuanyuan Xu, Bo Wang, and Hui Li; writing—original draft: Shan Long and Jiakang Shao; writing—review and editing: Xiaosong Li and Ying Gu; revising—manuscript: Bo Wang and Yingshu Cui.

Corresponding authors

Correspondence to Xiaosong Li or Ying Gu.

Ethics declarations

Ethical approval

Animal experiment was performed according to the procedures previously approved by the Animal Ethics Committee of Chinese People’s Liberation Army General Hospital (approval 2019-X15-68).

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Below is the link to the electronic supplementary material.

Supplementary file1 (DOCX 604 KB)

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Long, S., Wang, B., Cui, Y. et al. The upregulation of immune checkpoints after photodynamic therapy reducing immune effect for treating breast cancer. Lasers Med Sci 38, 243 (2023). https://doi.org/10.1007/s10103-023-03894-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s10103-023-03894-z

Keywords

Navigation