Skip to main content

Advertisement

Log in

Chlorin e6-loaded goat milk-derived extracellular vesicles for Cerenkov luminescence-induced photodynamic therapy

  • Original Article
  • Published:
European Journal of Nuclear Medicine and Molecular Imaging Aims and scope Submit manuscript

An Editorial to this article was published on 11 November 2022

Abstract

Purpose

Photodynamic therapy (PDT) is a promising cancer treatment strategy with rapid progress in preclinical and clinical settings. However, the limitations in penetration of external light and precise delivery of photosensitizers hamper its clinical translation. As such, the internal light source such as Cerenkov luminescence (CL) from decaying radioisotopes offers new opportunities. Herein, we show that goat milk-derived extracellular vesicles (GEV) can act as a carrier to deliver photosensitizer Chlorin e6 (Ce6) and tumor-avid 18F-FDG can activate CL-induced PDT for precision cancer theranostics.

Methods

GEV was isolated via differential ultracentrifugation of commercial goat milk and photosensitizer Ce6 was loaded by co-incubation to obtain Ce6@GEV. Tumor uptake of Ce6@GEV was examined using confocal microscopy and flow cytometry. To demonstrate the ability of 18F-FDG to activate photodynamic effects against cancer cells, apoptosis rates were measured using flow cytometry, and the production of 1O2 was measured by reactive oxygen species (ROS) monitoring kit. Moreover, we used the IVIS device to detect Cherenkov radiation and Cerenkov radiation energy transfer (CRET). For animal experiments, a small-animal IVIS imaging system was used to visualize the accumulation of the GEV drug delivery system in tumors. PET/CT and CL images of the tumor site were performed at 0.5, 1, and 2 h. For in vivo antitumor therapy, changes of tumor volume, survival time, and body weight in six groups of tumor-bearing mice were monitored. Furthermore, the blood sample and organs of interest (heart, liver, spleen, lungs, kidneys, and tumor) were collected for hematological analysis, immunohistochemistry, and H&E staining.

Results

Confocal microscopy of 4T1 cells incubated with Ce6@GEV for 4 h revealed strong red fluorescence signals in the cytoplasm, which demonstrated that Ce6 loaded in GEV could be efficiently delivered into tumor cells. When Ce6@GEV and 18F-FDG co-existed incubated with 4T1 cells, the cell viability plummeted from more than 88.02 ± 1.30% to 23.79 ± 1.59%, indicating excellent CL-induced PDT effects. In vivo fluorescence images showed a peak tumor/liver ratio of 1.36 ± 0.09 at 24 h after Ce6@GEV injection. For in vivo antitumor therapy, Ce6@GEV + 18F-FDG group had the best tumor inhibition rate (58.02%) compared with the other groups, with the longest survival rate (35 days, 40%). During the whole treatment process, neither blood biochemical analysis nor histological observation revealed vital organ damage, suggesting the biosafety of this treatment strategy.

Conclusions

The simultaneous accumulation of 18F-FDG and Ce6 in tumor tissues is expected to overcome the deficiency of traditional PDT. This strategy has the potential to extend PDT to a variety of tumors, including metastases, using targeted radiotracers to provide internal excitation of light-responsive therapeutics. We expect that our method will play a critical role in precision treatment of deep solid tumors.

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.

Scheme 1
Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

Abbreviations

PDT :

photodynamic therapy

CL :

Cerenkov luminescence

UV :

ultraviolet

EVs :

extracellular vesicles

GEV :

goat milk-derived extracellular vesicles

PS :

photosensitizers

ROS :

reactive oxygen species

1 O 2 :

singlet oxygen

CRET :

Cerenkov radiation energy transfer

18 F-FDG :

18F-fluo-rodeoxyglucose

Ce6 :

Chlorin e6

FBS :

fetal bovine serum

PBS :

phosphate buffer saline

TEM :

transmission electron microscope

DLS :

dynamic light scattering

WB :

Western blot

IVIS :

In Vivo Imaging System

i.v. :

intravenous injection

p.i. :

post-injection

ALT :

alanine aminotransferase

AST :

aspartate aminotransferase

ALP :

alkaline phosphatase

BUN :

blood urea nitrogen

CRE :

creatinine

DDS :

drug delivery systems

EPR effect :

enhanced permeability and retention effect

References

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

    Article  CAS  Google Scholar 

  2. Liu S, Feng G, Tang BZ, Liu B. Recent advances of AIE light-up probes for photodynamic therapy. Chem Sci. 2021;12:6488–506. https://doi.org/10.1039/d1sc00045d.

    Article  CAS  Google Scholar 

  3. Chilakamarthi U, Giribabu L. Photodynamic therapy: past, present and future. Chem Rec. 2017;17:775–802. https://doi.org/10.1002/tcr.201600121.

    Article  CAS  Google Scholar 

  4. Grigalavicius M, Mastrangelopoulou M, Berg K, Arous D, Ménard M, Raabe-Henriksen T, et al. Proton-dynamic therapy following photosensitiser activation by accelerated protons demonstrated through fluorescence and singlet oxygen production. Nat Commun. 2019;10:3986. https://doi.org/10.1038/s41467-019-12042-7.

    Article  CAS  Google Scholar 

  5. Li X, Lovell JF, Yoon J, Chen X. Clinical development and potential of photothermal and photodynamic therapies for cancer. Nat Rev Clin Oncol. 2020;17:657–74. https://doi.org/10.1038/s41571-020-0410-2.

    Article  Google Scholar 

  6. Blum NT, Zhang Y, Qu J, Lin J, Huang P. Recent advances in self-exciting photodynamic therapy. Front Bioeng Biotechnol. 2020;8:594491. https://doi.org/10.3389/fbioe.2020.594491.

    Article  Google Scholar 

  7. Kavadiya S, Biswas P. Design of Cerenkov radiation-assisted photoactivation of TiO nanoparticles and reactive oxygen species generation for cancer treatment. J Nucl Med. 2019;60:702–9. https://doi.org/10.2967/jnumed.118.215608.

    Article  CAS  Google Scholar 

  8. Shaffer TM, Pratt EC, Grimm J. Utilizing the power of Cerenkov light with nanotechnology. Nat Nanotechnol. 2017;12:106–17. https://doi.org/10.1038/nnano.2016.301.

    Article  CAS  Google Scholar 

  9. Shaffer TM, Drain CM, Grimm J. Optical imaging of ionizing radiation from clinical sources. J Nucl Med. 2016;57:1661–6.

    Article  CAS  Google Scholar 

  10. Kotagiri N, Sudlow GP, Akers WJ, Achilefu S. Breaking the depth dependency of phototherapy with Cerenkov radiation and low-radiance-responsive nanophotosensitizers. Nat Nanotechnol. 2015;10:370–9. https://doi.org/10.1038/nnano.2015.17.

    Article  CAS  Google Scholar 

  11. Kamkaew A, Chen F, Zhan Y, Majewski RL, Cai W. Scintillating nanoparticles as energy mediators for enhanced photodynamic therapy. ACS Nano. 2016;10:3918–35. https://doi.org/10.1021/acsnano.6b01401.

    Article  CAS  Google Scholar 

  12. Ni D, Ferreira CA, Barnhart TE, Quach V, Yu B, Jiang D, et al. Magnetic targeting of nanotheranostics enhances Cerenkov radiation-induced photodynamic therapy. J Am Chem Soc. 2018;140:14971–9. https://doi.org/10.1021/jacs.8b09374.

    Article  CAS  Google Scholar 

  13. Lioret V, Bellaye P-S, Arnould C, Collin B, Decréau RA. Dual Cherenkov radiation-induced near-infrared luminescence imaging and photodynamic therapy toward tumor resection. J Med Chem. 2020;63:9446–56. https://doi.org/10.1021/acs.jmedchem.0c00625.

    Article  CAS  Google Scholar 

  14. Zhang Y, Hao Y, Chen S, Xu M. Photodynamic therapy of cancers with internal light sources: chemiluminescence, bioluminescence, and Cerenkov radiation. Front Chem. 2020;8:770. https://doi.org/10.3389/fchem.2020.00770.

    Article  CAS  Google Scholar 

  15. Wang R, Li X, Yoon J. Organelle-targeted photosensitizers for precision photodynamic therapy. ACS Appl Mater Interfaces. 2021;13:19543–71. https://doi.org/10.1021/acsami.1c02019.

    Article  CAS  Google Scholar 

  16. Ha D, Yang N, Nadithe V. Exosomes as therapeutic drug carriers and delivery vehicles across biological membranes: current perspectives and future challenges. Acta Pharm Sin B. 2016;6:287–96. https://doi.org/10.1016/j.apsb.2016.02.001.

    Article  Google Scholar 

  17. van Niel G, Carter DRF, Clayton A, Lambert DW, Raposo G, Vader P. Challenges and directions in studying cell-cell communication by extracellular vesicles. Nat Rev Mol Cell Biol. 2022. https://doi.org/10.1038/s41580-022-00460-3.

  18. Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ, Lötvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 2007;9:654–9.

    Article  CAS  Google Scholar 

  19. Wiklander OPB, Nordin JZ, O’Loughlin A, Gustafsson Y, Corso G, Mäger I, et al. Extracellular vesicle in vivo biodistribution is determined by cell source, route of administration and targeting. J Extracell Vesicles. 2015;4:26316. https://doi.org/10.3402/jev.v4.26316.

    Article  Google Scholar 

  20. Aqil F, Munagala R, Jeyabalan J, Agrawal AK, Kyakulaga A-H, Wilcher SA, et al. Milk exosomes - natural nanoparticles for siRNA delivery. Cancer Lett. 2019;449:186–95. https://doi.org/10.1016/j.canlet.2019.02.011.

    Article  CAS  Google Scholar 

  21. Mecocci S, Pietrucci D, Milanesi M, Pascucci L, Filippi S, Rosato V, et al. Transcriptomic Characterization of cow, donkey and goat milk extracellular vesicles reveals their anti-inflammatory and immunomodulatory potential. Int J Mol Sci. 2021;22. https://doi.org/10.3390/ijms222312759.

  22. Yuan A, Tang X, Qiu X, Jiang K, Wu J, Hu Y. Activatable photodynamic destruction of cancer cells by NIR dye/photosensitizer loaded liposomes. Chem Commun (Camb). 2015;51:3340-2. https://doi.org/10.1039/c4cc09689d.

  23. Yang G, Xu L, Chao Y, Xu J, Sun X, Wu Y, et al. Hollow MnO as a tumor-microenvironment-responsive biodegradable nano-platform for combination therapy favoring antitumor immune responses. Nat Commun. 2017;8:902. https://doi.org/10.1038/s41467-017-01050-0.

    Article  CAS  Google Scholar 

  24. Cheng L, Kamkaew A, Sun H, Jiang D, Valdovinos HF, Gong H, et al. Dual-modality positron emission tomography/optical image-guided photodynamic cancer therapy with chlorin e6-containing nanomicelles. ACS Nano. 2016;10:7721–30. https://doi.org/10.1021/acsnano.6b03074.

    Article  CAS  Google Scholar 

  25. Lee W, Jeon M, Choi J, Oh C, Kim G, Jung S, et al. Europium-diethylenetriaminepentaacetic acid loaded radioluminescence liposome nanoplatform for effective radioisotope-mediated photodynamic therapy. ACS Nano. 2020;14:13004–15. https://doi.org/10.1021/acsnano.0c04324.

    Article  CAS  Google Scholar 

  26. Kamkaew A, Cheng L, Goel S, Valdovinos HF, Barnhart TE, Liu Z, et al. Cerenkov radiation induced photodynamic therapy using chlorin e6-loaded hollow mesoporous silica nanoparticles. ACS Appl Mater Interfaces. 2016;8:26630–7.

    Article  CAS  Google Scholar 

  27. Sun D, Zhuang X, Xiang X, Liu Y, Zhang S, Liu C, et al. A novel nanoparticle drug delivery system: the anti-inflammatory activity of curcumin is enhanced when encapsulated in exosomes. Mol Ther. 2010;18:1606–14. https://doi.org/10.1038/mt.2010.105.

    Article  CAS  Google Scholar 

  28. Dothager RS, Goiffon RJ, Jackson E, Harpstrite S, Piwnica-Worms D. Cerenkov radiation energy transfer (CRET) imaging: a novel method for optical imaging of PET isotopes in biological systems. PLoS ONE. 2010;5:e13300. https://doi.org/10.1371/journal.pone.0013300.

    Article  CAS  Google Scholar 

  29. Robertson R, Germanos MS, Li C, Mitchell GS, Cherry SR, Silva MD. Optical imaging of Cerenkov light generation from positron-emitting radiotracers. Phys Med Biol. 2009;54:N355–N65. https://doi.org/10.1088/0031-9155/54/16/N01.

    Article  CAS  Google Scholar 

  30. Xu Y, Liu H, Cheng Z. Harnessing the power of radionuclides for optical imaging: Cerenkov luminescence imaging. J Nucl Med. 2011;52:2009–18. https://doi.org/10.2967/jnumed.111.092965.

    Article  Google Scholar 

  31. Cheng L, Jiang D, Kamkaew A, Valdovinos HF, Im H-J, Feng L, et al. Renal-clearable PEGylated porphyrin nanoparticles for image-guided photodynamic cancer therapy. Adv Funct Mater. 2017;27. https://doi.org/10.1002/adfm.201702928.

  32. Xia Q, Zhang Y, Li Z, Hou X, Feng N. Red blood cell membrane-camouflaged nanoparticles: a novel drug delivery system for antitumor application. Acta Pharm Sin B. 2019;9:675–89. https://doi.org/10.1016/j.apsb.2019.01.011.

    Article  Google Scholar 

  33. Large DE, Abdelmessih RG, Fink EA, Auguste DT. Liposome composition in drug delivery design, synthesis, characterization, and clinical application. Adv Drug Deliv Rev. 2021;176:113851. https://doi.org/10.1016/j.addr.2021.113851.

    Article  CAS  Google Scholar 

  34. Moghassemi S, Dadashzadeh A, Azevedo RB, Feron O, Amorim CA. Photodynamic cancer therapy using liposomes as an advanced vesicular photosensitizer delivery system. J Control Release. 2021;339:75–90. https://doi.org/10.1016/j.jconrel.2021.09.024.

    Article  CAS  Google Scholar 

  35. Milman N, Ginini L, Gil Z. Exosomes and their role in tumorigenesis and anticancer drug resistance. Drug Resist Updat. 2019;45. https://doi.org/10.1016/j.drup.2019.07.003.

  36. He C, Zheng S, Luo Y, Wang B. Exosome theranostics: biology and translational medicine. Theranostics. 2018;8:237–55. https://doi.org/10.7150/thno.21945.

    Article  CAS  Google Scholar 

  37. Santos-Coquillat A, González MI, Clemente-Moragón A, González-Arjona M, Albaladejo-García V, Peinado H, et al. Goat milk exosomes as natural nanoparticles for detecting inflammatory processes by optical imaging. Small. 2022;18:e2105421. https://doi.org/10.1002/smll.202105421.

    Article  CAS  Google Scholar 

  38. Munagala R, Aqil F, Jeyabalan J, Gupta RC. Bovine milk-derived exosomes for drug delivery. Cancer Lett. 2016;371:48–61. https://doi.org/10.1016/j.canlet.2015.10.020.

    Article  CAS  Google Scholar 

  39. Mulcahy LA, Pink RC, Carter DRF. Routes and mechanisms of extracellular vesicle uptake. J Extracell Vesicles. 2014;3. https://doi.org/10.3402/jev.v3.24641.

  40. Qian R, Jing B, Jiang D, Gai Y, Zhu Z, Huang X, et al. Multi-antitumor therapy and synchronous imaging monitoring based on exosome. Eur J Nucl Med Mol Imaging. 2022. https://doi.org/10.1007/s00259-022-05696-x.

  41. Jing B, Gai Y, Qian R, Liu Z, Zhu Z, Gao Y, et al. Hydrophobic insertion-based engineering of tumor cell-derived exosomes for SPECT/NIRF imaging of colon cancer. J Nanobiotechnology. 2021;19:7. https://doi.org/10.1186/s12951-020-00746-8.

    Article  CAS  Google Scholar 

  42. Gill RK, Mitchell GS, Cherry SR. Computed Cerenkov luminescence yields for radionuclides used in biology and medicine. Phys Med Biol. 2015;60:4263–80. https://doi.org/10.1088/0031-9155/60/11/4263.

    Article  CAS  Google Scholar 

  43. Zhang Y-N, Poon W, Tavares AJ, McGilvray ID, Chan WCW. Nanoparticle-liver interactions: cellular uptake and hepatobiliary elimination. J Control Release. 2016;240:332–48. https://doi.org/10.1016/j.jconrel.2016.01.020.

    Article  CAS  Google Scholar 

  44. Bonsergent E, Grisard E, Buchrieser J, Schwartz O, Théry C, Lavieu G. Quantitative characterization of extracellular vesicle uptake and content delivery within mammalian cells. Nat Commun. 2021;12:1864. https://doi.org/10.1038/s41467-021-22126-y.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank colleagues of the Key Laboratory of Molecular Imaging for discussions and comments on this manuscript.

Funding

This work was supported by the National Natural Science Foundation of China (No. 82102121, 82071966, and 81873904), and thanks to the Open Foundation of Hubei Provincial Key Laboratory of Molecular Imaging (No. 2021fzyx009).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, R. Guo, D. Jiang, and Y. Gai; methodology, R. Guo, R. Qian, Z. Zhu, and Y. Gao; formal analysis, B. Jing and B. Yang; investigation, X. Lan and R. An; writing–original draft, R. Guo and R. An; writing–review and editing, R. An, D. Jiang, and X. Lan; project administration, R. An, D. Jiang, and X. Lan; funding acquisition, Y. Gai, D. Jiang, R. An, and X. Lan.

Corresponding authors

Correspondence to Xiaoli Lan or Rui An.

Ethics declarations

Ethics approval

All animal experiment protocols were reviewed and approved by the Animal Care and Use Committee of Tongji Medical College, Huazhong University of Science and Technology. BALB/c mice (female, 4–5 weeks old, Beijing Vital River Laboratory Animal Technology Co., Ltd, China) fed in a pathogen-free environment.

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.

This article is part of the Topical Collection on Theragnostic

Supplementary information

ESM 1

(DOCX 3084 kb)

Rights and permissions

Springer Nature or its licensor 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

Guo, R., Jiang, D., Gai, Y. et al. Chlorin e6-loaded goat milk-derived extracellular vesicles for Cerenkov luminescence-induced photodynamic therapy. Eur J Nucl Med Mol Imaging 50, 508–524 (2023). https://doi.org/10.1007/s00259-022-05978-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00259-022-05978-4

Keywords

Navigation