Skip to main content

Advertisement

Log in

A novel 18F-labeled agonist for PET imaging of stimulator of interferon gene expression in tumor-bearing mice

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

Abstract

Purpose

Stimulator of interferon genes (STING) protein plays a vital role in the immune surveillance of tumor microenvironment. Monitoring STING expression in tumors benefits the relevant STING therapy. This study aimed to develop a novel 18F-labeled agonist, dimeric amidobenzimidazole (diABZI), and firstly evaluate the feasibility of noninvasive positron emission tomography (PET) imaging of STING expression in the tumor microenvironment.

Methods

An analog of the STING agonist NOTA-DABI was synthesized and labeled with 18F via Al18F-NOTA complexation (denoted as [18F]F-DABI). Physicochemical properties, STING protein-binding affinity, and specificity of [18F]F-DABI were evaluated using cell uptake and docking assays. In vivo small-animal PET imaging and biodistribution studies of [18F]F-DABI in tumor-bearing mice were performed to verify the pharmacokinetics and tumor targeting ability. The correlation between tumor uptake and STING expression was also analyzed.

Results

[18F]F-DABI was produced conveniently with high radiochemical yield (44 ± 15%), radiochemical purity (> 97%) and molar activity (15–30 GBq/μmol). In vitro binding assays demonstrated that [18F]F-DABI has a favorable affinity and specificity for STING with a KD of 12.98 ± 2.07 nM. In vivo studies demonstrated the specificity of [18F]F-DABI for PET imaging of STING expression with B16F10 tumor uptake of 10.93 ± 0.93%ID/g, which was significantly different from that of blocking groups (3.13 ± 0.88%ID/g, ***p < 0.0001). Furthermore, tumor uptake of [18F]F-DABI was well positively correlated with STING expression in different tumor types. Biodistribution results demonstrated that [18F]F-DABI was predominately uptaken in the liver and intestines, indicating its hepatobiliary elimination.

Conclusion

This proof-of-concept study demonstrated a STING-binding radioligand for PET imaging, which could be used as a potential companion diagnostic tool for related STING-agonist therapies.

Graphical abstract

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
Fig. 7

Similar content being viewed by others

Abbreviations

STING:

Stimulator of interferon genes

diABZI:

Dimeric amidobenzimidazole

PET:

Positron emission tomography

cGAS:

Cyclic guanosine monophosphate-adenosine monophosphate synthase

IFN-I:

Type I interferons

IHC:

Immunohistochemistry

HPLC:

High-performance liquid chromatography

NMR:

Nuclear magnetic resonance

DMXAA:

Vadimezan

FBS:

Fetal bovine serum

SDS-PAGE:

Sodium dodecyl-sulfate polyacrylamide gel electrophoresis

%ID/g:

Injected dose per gram of tissue

ROIs:

Regions of interest

p.i.:

Post-injection

i.t.:

Intratumorally injected

CDN:

Cyclic dinucleotide

References

  1. Sanmamed MF, Chen L. A paradigm shift in cancer immunotherapy: from enhancement to normalization. Cell. 2018;175:313–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Haslam A, Prasad V. Estimation of the percentage of US patients with cancer who are eligible for and respond to checkpoint inhibitor immunotherapy drugs. JAMA Netw Open. 2019;2:e192535.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Yarchoan M, Hopkins A, Jaffee EM. Tumor mutational burden and response rate to PD-1 inhibition. N Engl J Med. 2017;377:2500.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Motwani M, Pesiridis S, Fitzgerald KA. DNA sensing by the cGAS–STING pathway in health and disease. Nat Rev Genet. 2019;20:657–74.

    Article  CAS  PubMed  Google Scholar 

  5. Garland KM, Sheehy TL, Wilson JT. Chemical and biomolecular strategies for STING pathway activation in cancer immunotherapy. Chem Rev. 2022;122:5977–6039.

    Article  CAS  PubMed  Google Scholar 

  6. Yum S, Li M, Frankel AE, Chen ZJ. Roles of the cGAS-STING pathway in cancer immunosurveillance and immunotherapy. Annu Rev Cancer Biol. 2019;3:323–44.

    Article  Google Scholar 

  7. Gao P, Ascano M, Wu Y, Barchet W, Gaffney BL, Zillinger T, et al. Cyclic [G(2′,5′)pA(3′,5′)p] is the metazoan second messenger produced by DNA-activated cyclic GMP-AMP synthase. Cell. 2013;153:1094–107.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Corrales L, Glickman LH, McWhirter SM, Kanne DB, Sivick KE, Katibah GE, et al. Direct Activation of STING in the tumor microenvironment leads to potent and systemic tumor regression and immunity. Cell Rep. 2015;11:1018–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Fu J, Kanne DB, Leong M, Glickman LH, McWhirter SM, Lemmens E, et al. STING agonist formulated cancer vaccines can cure established tumors resistant to PD-1 blockade. Sci Transl Med. 2015;7(283):283ra52.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Challa SV, Zhou S, Sheri A, Padmanabhan S, Meher G, Gimi R, et al. Preclinical studies of SB 11285, a novel STING agonist for immuno-oncology. JCO. 2017;35:14616–14616.

    Article  Google Scholar 

  11. Kwon J, Bakhoum SF. The cytosolic DNA-sensing cGAS–STING pathway in cancer. Cancer Discov. 2020;10:26–39.

    Article  CAS  PubMed  Google Scholar 

  12. Pan B-S, Perera SA, Piesvaux JA, Presland JP, Schroeder GK, Cumming JN, et al. An orally available non-nucleotide STING agonist with antitumor activity. Science. 2020;369:eaba6098.

    Article  CAS  PubMed  Google Scholar 

  13. Chen Q, Boire A, Jin X, Valiente M, Er EE, Lopez-Soto A, et al. Carcinoma–astrocyte gap junctions promote brain metastasis by cGAMP transfer. Nature. 2016;533:493–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Lemos H, Mohamed E, Huang L, Ou R, Pacholczyk G, Arbab AS, et al. STING promotes the growth of tumors characterized by low antigenicity via IDO activation. Cancer Res. 2016;76:2076–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Liang D, Xiao-Feng H, Guan-Jun D, Er-Ling H, Sheng C, Ting-Ting W, et al. Activated STING enhances tregs infiltration in the HPV-related carcinogenesis of tongue squamous cells via the c-jun/CCL22 signal. Biochim Biophys Acta. 2015;1852:2494–503.

    Article  PubMed  Google Scholar 

  16. Lim S-O, Li C-W, Xia W, Cha J-H, Chan L-C, Wu Y, et al. Deubiquitination and Stabilization of PD-L1 by CSN5. Cancer Cell. 2016;30:925–39.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Sivick KE, Desbien AL, Glickman LH, Reiner GL, Corrales L, Surh NH, et al. Magnitude of therapeutic STING activation determines CD8+ T cell-mediated anti-tumor immunity. Cell Rep. 2018;25:3074–85.

    Article  CAS  PubMed  Google Scholar 

  18. Chon HJ, Kim H, Noh JH, Yang H, Lee WS, Kong SJ, et al. STING Signaling is a potential immunotherapeutic target in colorectal cancer. J Cancer. 2019;10:4932–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Raaby Gammelgaard K, Sandfeld-Paulsen B, Godsk SH, Demuth C, Meldgaard P, Sorensen BS, et al. cGAS-STING pathway expression as a prognostic tool in NSCLC. Transl Lung Cancer Res. 2021;10:340–54.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Woo S-R, Fuertes MB, Corrales L, Spranger S, Furdyna MJ, Leung MYK, et al. STING-dependent cytosolic DNA sensing mediates innate immune recognition of immunogenic tumors. Immunity. 2014;41:830–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Ishikawa H, Barber GN. STING is An Endoplasmic reticulum adaptor that facilitates innate immune signalling. Nature. 2008;455:674–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Flood BA, Higgs EF, Li S, Luke JJ, Gajewski TF. STING pathway agonism as a cancer therapeutic. Immunol Rev. 2019;290:24–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Hu Q, Wu J, Ren Y, Wu X, Gao L, Wang G, et al. Degree of STING activation is associated with disease outcomes. Gut. 2020;69:792.2-794.

    Article  Google Scholar 

  24. Jiang M, Chen P, Wang L, Li W, Chen B, Liu Y, et al. cGAS-STING, an important pathway in cancer immunotherapy. J Hematol Oncol. 2020;13:81.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Topalian SL, Taube JM, Anders RA, Pardoll DM. Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat Rev Cancer. 2016;16:275–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. van der Leun AM, Thommen DS, Schumacher TN. CD8+ T cell states in human cancer: insights from single-cell analysis. Nat Rev Cancer. 2020;20:218–32.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Bensch F, van der Veen EL, Lub-de Hooge MN, Jorritsma-Smit A, Boellaard R, Kok IC, et al. 89Zr-atezolizumab imaging as a non-invasive approach to assess clinical response to PD-L1 blockade in cancer. Nat Med. 2018;24:1852–8.

    Article  CAS  PubMed  Google Scholar 

  28. Xing Y, Chand G, Liu C, Cook GJR, O’Doherty J, Zhao L, et al. Early phase I study of a 99mTc-Labeled Anti-Programmed Death Ligand-1 (PD-L1) single-domain antibody in SPECT/CT assessment of PD-L1 expression in non–small cell lung cancer. J Nucl Med. 2019;60:1213–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Zhou X, Jiang J, Yang X, Liu T, Ding J, Nimmagadda S, et al. First-in-humans evaluation of a PD-L1–binding peptide PET radiotracer in non–small cell lung cancer patients. J Nucl Med. 2022;63:536–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Ramanjulu JM, Pesiridis GS, Yang J, Concha N, Singhaus R, Zhang S-Y, et al. Design of amidobenzimidazole STING receptor agonists with systemic activity. Nature. 2018;564:439–43.

    Article  CAS  PubMed  Google Scholar 

  31. Burdette DL, Monroe KM, Sotelo-Troha K, Iwig JS, Eckert B, Hyodo M, et al. STING is a direct innate immune sensor of cyclic di-GMP. Nature. 2011;478:515–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Meric-Bernstam F, Sandhu SK, Hamid O, Spreafico A, Kasper S, Dummer R, et al. Phase Ib study of MIW815 (ADU-S100) in combination with spartalizumab (PDR001) in patients (pts) with advanced/metastatic solid tumors or lymphomas. JCO. 2019;37:2507–2507.

    Article  Google Scholar 

Download references

Funding

This study was financially supported by the Major Research Plan of the National Natural Science Foundation of China (91959122), National Natural Science Foundation of China (81901805, 21976150, 21906135), Joint Fund of the National Natural Science Foundation of China—China National Nuclear Corporation for Nuclear Technology Innovation (U1967222), and Fundamental Research Funds for the Central Universities of China (20720210115).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: Xianzhong Zhang, Jianyang Fang, Zhide Guo; methodology: Jianyang Fang, Lixia Feng, Lingxin Meng, Lumei Huang, Huanhuan Liu, Jingchao Li and Rongqiang Zhuang formal analysis and investigation: Jianyang Fang, Xiaobo Wang, Deliang Zhang; writing (original draft preparation): Jianyang Fang; writing (review and editing): Xianzhong Zhang, Zhide Guo, Xiaobo Wang; funding acquisition: Xianzhong Zhang, Zhide Guo.

Corresponding authors

Correspondence to Zhide Guo or Xianzhong Zhang.

Ethics declarations

Ethics approval

All animal studies were performed according to the guidelines of the Animal Care Committee of Xiamen University.

Conflict of interest

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 Preclinical Imaging

Jianyang Fang, Lixia Feng, and Lingxin Meng are first authors who contributed equally to this work.

Supplementary Information

Below is the link to the electronic supplementary material.

Supplementary file1 (DOCX 5368 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

Fang, J., Feng, L., Meng, L. et al. A novel 18F-labeled agonist for PET imaging of stimulator of interferon gene expression in tumor-bearing mice. Eur J Nucl Med Mol Imaging 50, 27–37 (2022). https://doi.org/10.1007/s00259-022-05959-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00259-022-05959-7

Keywords

Navigation