Skip to main content

Advertisement

Log in

First-in-human CLDN18.2 functional diagnostic pet imaging of digestive system neoplasms enables whole-body target mapping and lesion detection

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

Abstract

Purpose

Claudin 18.2 (CLDN18.2) is a reliable target for lesion detection and could have clinical implications for epithelial tumors, especially digestive system neoplasms. However, there is no predictive technology for accurate whole-body mapping of CLDN18.2 expression in patients. This study assessed the safety of the 124I-18B10(10L) tracer and the feasibility of mapping whole-body CLDN18.2 expression using PET functional imaging.

Methods

The 124I-18B10(10L) probe was synthesized manually, and preclinical experiments including binding affinity and specific targeting ability were conducted after testing in vitro model cells. Patients with pathologically confirmed digestive system neoplasms were enrolled in an ongoing, open-label, single-arm, first-in-human (FiH) phase 0 trial (NCT04883970). 124I-18B10(10L) PET/CT or PET/MR and 18F-FDG PET were performed within one week.

Results

124I-18B10(10L) was successfully constructed with an over 95% radiochemical yield. The results of preclinical experiments showed that it had high stability in saline and high affinity in CLDN18.2 overexpressing cells (Kd = 4.11 nM). Seventeen patients, including 12 with gastric cancers, 4 with pancreatic cancers, and 1 with cholangiocarcinoma were enrolled. 124I-18B10(10L) displayed high uptake in the spleen and liver, and slight uptake in the bone marrow, lung, stomach and pancreas. The tracer uptake SUVmax in tumor lesions ranged from 0.4 to 19.5. Compared with that in lesions that had been treated with CLDN18.2-targeted therapy, 124I-18B10(10L) uptake was significantly higher in lesions that had not. Regional 124I-18B10(10L) PET/MR in two patients showed high tracer uptake in metastatic lymph nodes.

Conclusions

124I-18B10(10L) was successfully prepared and exhibited a high binding affinity and CLDN18.2 specificity in preclinical studies. As an FiH CLDN18.2 PET tracer, 124I-18B10(10L) was shown to be safe with acceptable dosimetry and to clearly reveal most lesions overexpressing CLDN18.2.

Trial registration

NCT04883970; URL: https://register.clinicaltrials.gov/. Registered 07 May 2021.

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

Data Availability

The datasets generated during and/or analyzed during the current study are available from the corresponding author on reasonable request. 

References

  1. Tsukita S, Furuse M, Itoh M. Multifunctional strands in tight junctions. Nat Rev Mol Cell Biol. 2001;2:285–93.

    Article  CAS  PubMed  Google Scholar 

  2. Gunzel D, Yu AS. Claudins and the modulation of tight junction permeability. Physiol Rev. 2013;93:525–69.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Zihni C, Mills C, Matter K, et al. Tight junctions: from simple barriers to multifunctional molecular gates. Nat Rev Mol Cell Biol. 2016;17:564–80.

    Article  CAS  PubMed  Google Scholar 

  4. Niimi T, Nagashima K, Ward JM, et al. claudin-18, a novel downstream target gene for the T/EBP/NKX2.1 homeodomain transcription factor, encodes lung- and stomach-specific isoforms through alternative splicing. Mol Cell Biol. 2001;21:7380–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Sahin U, Koslowski M, Dhaene K, et al. Claudin-18 splice variant 2 is a pan-cancer target suitable for therapeutic antibody development. Clin Cancer Res. 2008;14:7624–34.

    Article  CAS  PubMed  Google Scholar 

  6. Iwaya M, Hayashi H, Nakajima T, et al. Colitis-associated colorectal adenocarcinomas frequently express claudin 18 isoform 2: implications for claudin 18.2 monoclonal antibody therapy. Histopathology. 2021;79:227–37.

    Article  PubMed  Google Scholar 

  7. Kyuno D, Takasawa A, Takasawa K, et al. Claudin-18.2 as a therapeutic target in cancers: cumulative findings from basic research and clinical trials. Tissue Barriers. 2022;10:1967080.

  8. Klempner SJ, Ajani JA, Al-Batran S-E, et al. Phase II study of zolbetuximab plus pembrolizumab in claudin 18.2: Positive locally advanced or metastatic gastric or gastroesophageal junction adenocarcinoma (G/GEJ)—ILUSTRO Cohort 3. J Clin Oncol. 2021;39:TPS260–TPS.

  9. Yamaguchi K, Shitara K, Al-Batran SE, et al. 198TiP - SPOTLIGHT: Comparison of zolbetuximab or placebo + mFOLFOX6 as first-line treatment in patients with claudin18.2+/HER2– locally advanced unresectable or metastatic gastric or gastroesophageal junction adenocarcinoma (GEJ): A randomized phase III study. Annals Oncol. 2019;30:ix66–7.

    Article  Google Scholar 

  10. Xu RH, Ajani JA, Al-Batran SE, et al. 195TiP GLOW: Phase III study of first-line zolbetuximab + CAPOX versus placebo + CAPOX in Claudin18.2+/HER2- advanced/metastatic gastric or gastroesophageal junction adenocarcinoma (G/GEJ). Annals Oncol. 2020;31:S1315–6.

    Article  Google Scholar 

  11. Sahin U, Türeci Ö, Manikhas G, et al. FAST: A randomised phase II study of zolbetuximab (IMAB362) plus EOX vs EOX alone for first-line treatment of advanced CLDN18.2 positive gastric and gastro-oesophageal adenocarcinoma. Annals Oncol. 2021;32:609–19.

    Article  CAS  Google Scholar 

  12. Qi C, Gong J, Li J, et al. Claudin18.2-specific CAR T cells in gastrointestinal cancers: phase 1 trial interim results. Nat Med. 2022;28:1189–98.

  13. Kage H, Flodby P, Zhou B, et al. Dichotomous roles of claudins as tumor promoters or suppressors: lessons from knockout mice. Cell Mol Life Sci. 2019;76:4663–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Zhao C, Rong Z, Ding J, et al. Targeting Claudin 18.2 Using a Highly Specific Antibody Enables Cancer Diagnosis and Guided Surgery. Mol Pharm. 2022. Mar 28 [Epub ahead of print]: https://doi.org/10.1021/acs.molpharmaceut.1c00947.

  15. Zhu H, Ding J, Chong X, et al. Molecular imaging evaluation of a novel Claudin18.2. specific monoclonal antibody labeled with radionuclide. Annals Oncol. 2021;32:S1.

    Article  Google Scholar 

  16. Gong J, Li N, Gao W, et al. A phase I study of TST001, a high affinity humanized anti-CLDN18.2 monoclonal antibody, in combination with capecitabine and oxaliplatin (CAPOX) as a first-line treatment of advanced G/GEJ cancer. J Clin Oncol. 2022;40:4062.

    Article  Google Scholar 

  17. Divgi CR, Uzzo RG, Gatsonis C, et al. Positron Emission Tomography/Computed Tomography Identification of Clear Cell Renal Cell Carcinoma: Results From the REDECT Trial. J Clin Oncol. 2013;31:187–94.

    Article  PubMed  Google Scholar 

  18. Wang S, Zhu H, Ding J, et al. Positron emission tomography imaging of programmed death 1 expression in cancer patients using 124I-Labeled toripalimab: a pilot clinical translation study. Clin Nucl Med. 2021;46:382–8.

  19. Guo X, Zhou N, Chen Z, et al. Construction of 124I-trastuzumab for Noninvasive PET Imaging of HER2 Expression: From Patient-derived Xenograft Models to Gastric Cancer Patients. Gastric Cancer. 2020;23:614–26.

    Article  CAS  PubMed  Google Scholar 

  20. Jiang H, Shi Z, Wang P, et al. Claudin18.2-Specific Chimeric Antigen Receptor Engineered T Cells for the Treatment of Gastric Cancer. J Natl Cancer Inst. 2019;111:409–18.

    Article  PubMed  Google Scholar 

  21. Zhang J, Dong R, Shen L. Evaluation and reflection on claudin 18.2 targeting therapy in advanced gastric cancer. Chin J Cancer Res. 2020;32:263–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Gerwing M, Herrmann K, Helfen A, et al. The beginning of the end for conventional RECIST - novel therapies require novel imaging approaches. Nat Rev Clin Oncol. 2019;16:442–58.

    Article  CAS  PubMed  Google Scholar 

  23. Bensch F, van der Veen EL, Lub-de Hooge MN, 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.

  24. Niemeijer AN, Leung D, Huisman MC, et al. Whole body PD-1 and PD-L1 positron emission tomography in patients with non-small-cell lung cancer. Nat Commun. 2018;9:4664.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Kok IC, Hooiveld JS, van de Donk PP, et al. 89Zr-pembrolizumab imaging as a non-invasive approach to assess clinical response to PD-1 blockade in cancer. Ann Oncol. 2022;33:80–8.

  26. Türeci O, Sahin U, Schulze-Bergkamen H, et al. A multicentre, phase IIa study of zolbetuximab as a single agent in patients with recurrent or refractory advanced adenocarcinoma of the stomach or lower oesophagus: the MONO study. Ann Oncol. 2019;30:1487–95.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Sahin U, Schuler M, Richly H, et al. A phase I dose-escalation study of IMAB362 (Zolbetuximab) in patients with advanced gastric and gastro-oesophageal junction cancer. Eur J Cancer. 2018;100:17–26.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We acknowledge the staff of the Department of Nuclear Medicine, and the Department of Gastrointestinal Oncology, Peking University Cancer Hospital for their help.

Funding

This work was supported by the National Natural Science Foundation of China (82171980, 82171973, 82102091, 82072728), Beijing Hospitals Authority’ Ascent Plan (DFL20191102), Beijing Millions of Talent Projects A level funding (No. 2019A38), The Pilot Project (4 th Round) to Reform Public Development of Beijing Municipal Medical Research Institute (2021–1). The authors declare that there is no conflict of interest regarding the publication of this article. Intellectual property protection has been filed by Suzhou Transcenta Therapeutics Company and Peking University Cancer Hospital.

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to the study conception and design. Material preparation, data collection and analysis were performed by [Shujing Wang], [Changsong Qi] and [Jin Ding]. The first draft of the manuscript was written by [Shujing Wang], [Changsong Qi] and [Jin Ding] and all authors commented on previous versions of the manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Jing Gao, Zhi Yang, Cheng Zhang or Hua Zhu.

Ethics declarations

Ethical approval

All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki Declaration and its later amendments or comparable ethical standards. The clinical study was approved by the Ethics Committee of Peking University Cancer Hospital (2021KT57) and was registered at ClinicalTrial.gov (NCT04883970).

Informed consent

Informed consent was obtained from all individual participants included in the study.

Conflict of interest

None declared. Intellectual property protection has been filed by Suzhou Transcenta Therapeutics Company and Peking University Cancer Hospital.

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 Translational research.

Supplementary Information

Below is the link to the electronic supplementary material.

Supplementary file1 (DOCX 2.00 MB)

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

Wang, S., Qi, C., Ding, J. et al. First-in-human CLDN18.2 functional diagnostic pet imaging of digestive system neoplasms enables whole-body target mapping and lesion detection. Eur J Nucl Med Mol Imaging 50, 2802–2817 (2023). https://doi.org/10.1007/s00259-023-06234-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00259-023-06234-z

Keywords

Navigation