Skip to main content

Advertisement

Log in

Induction of ferroptosis by photodynamic therapy and enhancement of antitumor effect with ferroptosis inducers

  • Original Article—Alimentary Tract
  • Published:
Journal of Gastroenterology Aims and scope Submit manuscript

Abstract

Background

Photodynamic therapy (PDT) is an effective tumor treatment that involves the administration of a photosensitizer to generate cytotoxic 1O2 [reactive oxygen species (ROS)] from molecular oxygen that is produced from energy absorption following tumor irradiation at specific wavelengths. Ferroptosis is induced by the disruption of the glutathione peroxidase 4 (GPX4) antioxidant system, leading to lipid peroxidation. We hypothesized that talaporfin sodium-photodynamic therapy (TS-PDT)-generated ROS would lead to ferroptosis via accumulation of lipid peroxidation.

Methods

Cell viability assay in TS-PDT-treated cells in combination with a ferroptosis inhibitor (ferrostatin-1: Fer-1) or ferroptosis inducers (imidazole ketone erastin: IKE, Ras-selective lethal 3: RSL3) was performed. Accumulation of lipid peroxidation, GPX4 antioxidant system and cystine/glutamate antiporter (system xc) activity in TS-PDT-treated cells was investigated. In xenograft mice, the antitumor effect of TS-PDT in combination with ferroptosis inducers (IKE or sorafenib) was examined.

Results

TS-PDT-induced cell death was partly suppressed by Fer-1 and accompanied by lipid peroxidation. TS-PDT combined with IKE or RSL3 enhanced the induction of cell death. TS-PDT inhibited cystine uptake activity via system xc. In vivo, the combination of TS-PDT and ferroptosis inducers (IKE or sorafenib) reduced tumor volume.

Conclusion

This study found that the mechanism underlying TS-PDT-induced ferroptosis constitutes direct lipid peroxidation by the generated ROS, and the inhibition of system xc, and that the combination of a ferroptosis inducer with TS-PDT enhances the antitumor effect of TS-PDT. Our findings suggest that ferroptosis-inducing therapies combined with PDT may benefit cancer patients.

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
Fig. 6

Similar content being viewed by others

Abbreviations

ATM:

Ataxia telangiectasia-mutated

FBS:

Fetal bovine serum

Fer-1:

Ferrostatin-1

FSP1:

Ferroptosis suppressor protein 1

GSH:

Reduced glutathione

GPX4:

Glutathione peroxidase 4

IKE:

Imidazole ketone erastin

MDA:

Malondialdehyde

NAC:

N-Acetylcysteine

NADPH:

Nicotinamide adenine dinucleotide phosphate hydrogen

Nec-1s:

Necrostatin-1s

PBS:

Phosphate-buffered saline

PDT:

Photodynamic therapy

PUFAs:

Polyunsaturated fatty acids

RIPK:

Receptor-interacting protein kinase

ROS:

Reactive oxygen species

RSL3:

Ras-selective lethal 3

SLC3A2:

Solute carrier family 3 member 2

SLC7A11:

Solute carrier family 7 member 11

TBARS:

Thiobarbituric acid-reactive substance

TS:

Talaporfin sodium

References

  1. Dolmans DE, Fukumura D, Jain RK. Photodynamic therapy for cancer. Nat Rev Cancer. 2003;3:380–7.

    Article  CAS  PubMed  Google Scholar 

  2. Agostinis P, Berg K, Cengel KA, et al. Photodynamic therapy of cancer: an update. CA A Cancer J Clin. 2011. https://doi.org/10.3322/caac.20114.

    Article  Google Scholar 

  3. Brown SB, Brown EA, Walker I. The present and future role of photodynamic therapy in cancer treatment. Lancet Oncol. 2004;5:497–508.

    Article  CAS  PubMed  Google Scholar 

  4. Yano T, Minamide T, Takashima K, et al. Clinical practice of photodynamic therapy using talaporfin sodium for esophageal cancer. J Clin Med. 2021. https://doi.org/10.3390/jcm10132785.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Yano T, Kasai H, Horimatsu T, et al. A multicenter phase II study of salvage photodynamic therapy using talaporfin sodium (ME2906) and a diode laser (PNL6405EPG) for local failure after chemoradiotherapy or radiotherapy for esophageal cancer. Oncotarget. 2017;8:22135–44.

    Article  PubMed  Google Scholar 

  6. Dang J, He H, Chen D, et al. Manipulating tumor hypoxia toward enhanced photodynamic therapy (PDT). Biomater Sci. 2017;5:1500–11.

    Article  CAS  PubMed  Google Scholar 

  7. Li RQ, Zhang C, Xie BR, et al. A two-photon excited O(2)-evolving nanocomposite for efficient photodynamic therapy against hypoxic tumor. Biomaterials. 2019;194:84–93.

    Article  PubMed  Google Scholar 

  8. Liu LH, Zhang YH, Qiu WX, et al. Dual-stage light amplified photodynamic therapy against hypoxic tumor based on an O(2) self-sufficient nanoplatform. Small. 2017. https://doi.org/10.1002/smll.201701621.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Liu WL, Liu T, Zou MZ, et al. Aggressive man-made red blood cells for hypoxia-resistant photodynamic therapy. Adv Mater (Deerfield Beach, Fla). 2018;30:e1802006.

    Article  Google Scholar 

  10. Dixon SJ, Lemberg KM, Lamprecht MR, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell. 2012;149:1060–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Martin-Sanchez D, Ruiz-Andres O, Poveda J, et al. Ferroptosis, but not necroptosis, is important in nephrotoxic folic acid-induced AKI. J Am Soc Nephrol. 2017;28:218–29.

    Article  CAS  PubMed  Google Scholar 

  12. Carlson BA, Tobe R, Yefremova E, et al. Glutathione peroxidase 4 and vitamin E cooperatively prevent hepatocellular degeneration. Redox Biol. 2016;9:22–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Guiney SJ, Adlard PA, Bush AI, et al. Ferroptosis and cell death mechanisms in Parkinson’s disease. Neurochem Int. 2017;104:34–48.

    Article  CAS  PubMed  Google Scholar 

  14. Wu X, Li Y, Zhang S, et al. Ferroptosis as a novel therapeutic target for cardiovascular disease. Theranostics. 2021;11:3052–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Carneiro BA, El-Deiry WS. Targeting apoptosis in cancer therapy. Nat Rev Clin Oncol. 2020;17:395–417.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Viswanathan VS, Ryan MJ, Dhruv HD, et al. Dependency of a therapy-resistant state of cancer cells on a lipid peroxidase pathway. Nature. 2017;547:453–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Hangauer MJ, Viswanathan VS, Ryan MJ, et al. Drug-tolerant persister cancer cells are vulnerable to GPX4 inhibition. Nature. 2017;551:247–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Jiang X, Stockwell BR, Conrad M. Ferroptosis: mechanisms, biology and role in disease. Nat Rev Mol Cell Biol. 2021;22:266–82.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Yang WS, Stockwell BR. Ferroptosis: death by lipid peroxidation. Trends Cell Biol. 2016;26:165–76.

    Article  CAS  PubMed  Google Scholar 

  20. Zhu T, Shi L, Yu C, et al. Ferroptosis promotes photodynamic therapy: supramolecular photosensitizer-inducer nanodrug for enhanced cancer treatment. Theranostics. 2019;9:3293–307.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Xu T, Ma Y, Yuan Q, et al. enhanced ferroptosis by oxygen-boosted phototherapy based on a 2-in-1 nanoplatform of ferrous hemoglobin for tumor synergistic therapy. ACS Nano. 2020;14:3414–25.

    Article  CAS  PubMed  Google Scholar 

  22. Miotto G, Rossetto M, Di Paolo ML, et al. Insight into the mechanism of ferroptosis inhibition by ferrostatin-1. Redox Biol. 2020;28:101328.

    Article  CAS  PubMed  Google Scholar 

  23. Karuppagounder SS, Alin L, Chen Y, et al. N-acetylcysteine targets 5 lipoxygenase-derived, toxic lipids and can synergize with prostaglandin E(2) to inhibit ferroptosis and improve outcomes following hemorrhagic stroke in mice. Ann Neurol. 2018;84:854–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Slee EA, Zhu H, Chow SC, et al. Benzyloxycarbonyl-Val-Ala-Asp (OMe) fluoromethylketone (Z-VAD.FMK) inhibits apoptosis by blocking the processing of CPP32. Biochem J. 1996. https://doi.org/10.1042/bj3150021.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Mikuš P, Pecher D, Rauová D, et al. Determination of novel highly effective necrostatin Nec-1s in rat plasma by high performance liquid chromatography hyphenated with quadrupole-time-of-flight mass spectrometry. Molecules (Basel, Switzerland). 2018. https://doi.org/10.3390/molecules23081946.

    Article  PubMed  Google Scholar 

  26. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods (San Diego, Calif). 2001;25:402–8.

    Article  CAS  PubMed  Google Scholar 

  27. Dixon SJ, Patel DN, Welsch M, et al. Pharmacological inhibition of cystine-glutamate exchange induces endoplasmic reticulum stress and ferroptosis. Elife. 2014. https://doi.org/10.7554/eLife.02523.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Sengupta A, Lichti UF, Carlson BA, et al. Targeted disruption of glutathione peroxidase 4 in mouse skin epithelial cells impairs postnatal hair follicle morphogenesis that is partially rescued through inhibition of COX-2. J Invest Dermatol. 2013;133:1731–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Shimomura T, Hirakawa N, Ohuchi Y, et al. Simple fluorescence assay for cystine uptake via the xCT in cells using selenocystine and a fluorescent probe. ACS Sensors. 2021;6:2125–8.

    Article  CAS  PubMed  Google Scholar 

  30. Koppula P, Zhuang L, Gan B. Cystine transporter SLC7A11/xCT in cancer: ferroptosis, nutrient dependency, and cancer therapy. Protein Cell. 2021;12:599–620.

    Article  CAS  PubMed  Google Scholar 

  31. Oleinick NL, Morris RL, Belichenko I. The role of apoptosis in response to photodynamic therapy: what, where, why, and how. Photochem Photobiol Sci Off J Eur Photochem Assoc Eur Soc Photobiol. 2002;1:1–21.

    CAS  Google Scholar 

  32. Aniogo EC, George BPA, Abrahamse H. Role of Bcl-2 family proteins in photodynamic therapy mediated cell survival and regulation. Molecules (Basel, Switzerland). 2020. https://doi.org/10.3390/molecules25225308.

    Article  PubMed  Google Scholar 

  33. Miki Y, Akimoto J, Moritake K, et al. Photodynamic therapy using talaporfin sodium induces concentration-dependent programmed necroptosis in human glioblastoma T98G cells. Lasers Med Sci. 2015;30:1739–45.

    Article  PubMed  Google Scholar 

  34. Song R, Li T, Ye J, et al. Acidity-activatable dynamic nanoparticles boosting ferroptotic cell death for immunotherapy of cancer. Adv Mater (Deerfield Beach, Fla). 2021;33:e2101155.

    Article  Google Scholar 

  35. Zhou Y, Chen K, Lin WK, et al. Photo-enhanced synergistic induction of ferroptosis for anti-cancer immunotherapy. Adv Healthc Mater. 2023. https://doi.org/10.1002/adhm.202300994.

    Article  PubMed  Google Scholar 

  36. Chen Q, Ma X, Xie L, et al. Iron-based nanoparticles for MR imaging-guided ferroptosis in combination with photodynamic therapy to enhance cancer treatment. Nanoscale. 2021;13:4855–70.

    Article  CAS  PubMed  Google Scholar 

  37. Plaetzer K, Kiesslich T, Krammer B, et al. Characterization of the cell death modes and the associated changes in cellular energy supply in response to AlPcS4-PDT. Photochem Photobiol Sci Off J Eur Photochem Assoc Eur Soc Photobiol. 2002;1:172–7.

    CAS  Google Scholar 

  38. Hassannia B, Vandenabeele P, Vanden BT. Targeting ferroptosis to iron out cancer. Cancer Cell. 2019;35:830–49.

    Article  CAS  PubMed  Google Scholar 

  39. Ye LF, Chaudhary KR, Zandkarimi F, et al. Radiation-induced lipid peroxidation triggers ferroptosis and synergizes with ferroptosis inducers. ACS Chem Biol. 2020;15:469–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Lei G, Zhang Y, Koppula P, et al. The role of ferroptosis in ionizing radiation-induced cell death and tumor suppression. Cell Res. 2020;30:146–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Shui S, Zhao Z, Wang H, et al. Non-enzymatic lipid peroxidation initiated by photodynamic therapy drives a distinct ferroptosis-like cell death pathway. Redox Biol. 2021;45:102056.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Turubanova VD, Balalaeva IV, Mishchenko TA, et al. Immunogenic cell death induced by a new photodynamic therapy based on photosens and photodithazine. J Immunother Cancer. 2019;7:350.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Lang X, Green MD, Wang W, et al. Radiotherapy and Immunotherapy promote tumoral lipid oxidation and ferroptosis via synergistic repression of SLC7A11. Cancer Discov. 2019;9:1673–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Tanaka M, Sasaki M, Suzuki T, et al. Combination of talaporfin photodynamic therapy and Poly (ADP-Ribose) polymerase (PARP) inhibitor in gastric cancer. Biochem Biophys Res Commun. 2021;539:1–7.

    Article  CAS  PubMed  Google Scholar 

  45. Seibt TM, Proneth B, Conrad M. Role of GPX4 in ferroptosis and its pharmacological implication. Free Radical Biol Med. 2019;133:144–52.

    Article  CAS  Google Scholar 

  46. Stockwell BR, Jiang X. The chemistry and biology of ferroptosis. Cell Chem Biol. 2020;27:365–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Sato H, Shiiya A, Kimata M, et al. Redox imbalance in cystine/glutamate transporter-deficient mice. J Biol Chem. 2005;280:37423–9.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We would like to thank Suzuka Asai and Yukimi Ito for their technical assistance. We are grateful for the assistance of the Research Equipment Sharing Center at the Nagoya City University.

Funding

This work was partially supported by the Japan Society for the Promotion of Science (JSPS) KAKENHI (2020–2022) (grant number 20K08361) (to M. Tanaka); JSPS KAKENHI (2023–2025) (grant number 23K07358) (to M. Tanaka); Toyoaki Scholarship Foundation (2022–2023) (grant number JOSE203179) (to M. Tanaka); Iketani Science and Technology Foundation (2022–2023) (grant number JOSE204007) (to M. Tanaka); JSPS KAKENHI (2022–2023) (grant number 22K20862) (to M. Sasaki); Grant from Bristol Myers Squibb (grant number JOSE202103) (to H. Kataoka) and JSPS KAKENHI (2023–2025) (grant number 23K07421) (to H. Kataoka); JSPS KAKENHI (2023–2024) (grant number 23K15019) (to M. Sasaki). The funders had no role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

All the authors contributed to the study conception and design. Study design was performed by YK and MT. Material preparation, data collection, and analysis were performed by YK, MS and MT. The first draft of the manuscript was written by YK. Funding was acquired by MS, MT and HK. All the authors commented on the final manuscript and approved the final manuscript.

Corresponding author

Correspondence to Mamoru Tanaka.

Ethics declarations

Conflict of interest

All the authors have no conflicts of interest to disclose.

Ethics approval

Our study was approved by the Animal Research Committee of the Nagoya City University (project number: 21-014).

Additional information

Publisher's Note

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

Supplementary Information

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

Kojima, Y., Tanaka, M., Sasaki, M. et al. Induction of ferroptosis by photodynamic therapy and enhancement of antitumor effect with ferroptosis inducers. J Gastroenterol 59, 81–94 (2024). https://doi.org/10.1007/s00535-023-02054-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00535-023-02054-y

Keywords

Navigation