Skip to main content

Advertisement

Log in

New and effective EGFR-targeted fluorescence imaging technology for intraoperative rapid determination of lung cancer in freshly isolated tissue

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

Abstract

Purpose

During lung cancer surgery, it is very important to define tumor boundaries and determine the surgical margin distance. In previous research, systemically application of fluorescent probes can help medical professionals determine the boundaries of tumors and find small tumors and metastases, thereby improving the accuracy of surgical resection. However, there are very few safe and effective probes that can be applied to clinical trials up to now, which limits the clinical application of fluorescence imaging. Here we developed a new technology that can quickly identify the tumor area in the resected lung tissue during the operation and distinguish the tumor boundary and metastatic lymph nodes.

Experimental design

For animal studies, a PDX model of lung cancer was established. The tumors, lungs, and peritumoral muscle tissues of tumor-bearing mice were surgically removed and incubated with a probe targeting epidermal growth factor receptor (EGFR) for 20 min, and then imaged by a closed-field near-infrared two-zone (NIR-II) fluorescence imaging system. For clinical samples, ten surgically removed lung tissues and 60 lymph nodes from 10 lung cancer patients undergoing radical resection were incubated with the targeting probe immediately after intraoperative resection and imaged to identify the tumor area and distinguish the tumor boundary and metastatic lymph nodes. The accuracy of fluorescence imaging was confirmed by HE staining and immunohistochemistry.

Results

The ex vivo animal imaging experiments showed a fluorescence enhancement of tumor tissue. For clinical samples, our results showed that this new technology yielded more than 85.7% sensitivity and 100% specificity in identifying the tumor area in the resected lung tissue. The average fluorescence tumor-to-background ratio was 2.5 ± 1.3. Furthermore, we also used this technique to image metastatic lymph nodes intraoperatively and showed that metastatic lymph nodes have brighter fluorescence than normal lymph nodes, as the average fluorescence tumor-to-background signal ratio was 2.7 ± 1.1. Calculations on the results of the fluorescence signal in relation to the number of metastatic lymph nodes yielded values of 77.8% for sensitivity and 92.1% for specificity. We expect this new technology to be a useful diagnostic tool for rapid intraoperative pathological detection and margin determination.

Conclusions

By using fluorescently labeled anti-human EGFR recombinant antibody scFv fragment to incubate freshly isolated tissues during surgery, the probes can quickly accumulate in lung cancer tissues, which can be used to quickly identify tumor areas in the resected lung tissues and distinguish tumor boundaries and find metastases in lymph nodes. This technology is expected to be used for rapid intraoperative pathological detection and margin determination.

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
Scheme 1
Fig. 5
Fig. 6

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.

Data availability

The data generated or analyzed during this study are included in the manuscript and the supplementary information files.

References

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

    Article  Google Scholar 

  2. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71:209–49. https://doi.org/10.3322/caac.21660.

    Article  Google Scholar 

  3. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2021. CA Cancer J Clin. 2021;71:7–33. https://doi.org/10.3322/caac.21654.

    Article  Google Scholar 

  4. Hui Z, Men Y, Hu C, Kang J, Sun X, Bi N, et al. Effect of postoperative radiotherapy for patients with pIIIA-N2 non–small cell lung cancer after complete resection and adjuvant chemotherapy: the phase 3 PORT-C randomized clinical trial. JAMA Oncol. 2021;7:1178–85. https://doi.org/10.1001/jamaoncol.2021.1910.

    Article  Google Scholar 

  5. Chi C, Du Y, Ye J, Kou D, Qiu J, Wang J, et al. Intraoperative imaging-guided cancer surgery: from current fluorescence molecular imaging methods to future multi-modality imaging technology. Theranostics. 2014;4:1072–84. https://doi.org/10.7150/thno.9899.

    Article  Google Scholar 

  6. Okusanya OT, Hess NR, Luketich JD, Sarkaria IS. Infrared intraoperative fluorescence imaging using indocyanine green in thoracic surgery. Eur J Cardiothorac Surg. 2018;53:512–8.

    Article  Google Scholar 

  7. Kossatz S, Pirovano G, De Souza Franca PD, Strome AL, Sunny SP, Zanoni DK, et al. Validation of the use of a fluorescent PARP1 inhibitor for the detection of oral, oropharyngeal and oesophageal epithelial cancers. Nat Biomed Eng. 2020;4:272–85. https://doi.org/10.1038/s41551-020-0526-9.

    Article  CAS  Google Scholar 

  8. Hernot S, van Manen L, Debie P, Mieog JSD, Vahrmeijer AL. Latest developments in molecular tracers for fluorescence image-guided cancer surgery. Lancet Oncol. 2019;20:e354–67. https://doi.org/10.1016/s1470-2045(19)30317-1.

    Article  CAS  Google Scholar 

  9. Quan YH, Oh CH, Jung D, Lim J-Y, Choi BH, Rho J, et al. Evaluation of intraoperative near-infrared fluorescence visualization of the lung tumor margin with indocyanine green inhalation. JAMA Surg. 2020;155:732–40.

    Article  Google Scholar 

  10. Kennedy GT, Azari FS, Bernstein E, Marfatia I, Din A, Kucharczuk JC, et al. Targeted intraoperative molecular imaging for localizing nonpalpable tumors and quantifying resection margin distances. JAMA Surg. 2021;156:1043–50. https://doi.org/10.1001/jamasurg.2021.3757.

    Article  Google Scholar 

  11. Miyamoto H. Intraoperative pathology consultation during urological surgery: Impact on final margin status and pitfalls of frozen section diagnosis. Pathol Int. 2021;71:567–80. https://doi.org/10.1111/pin.13132.

    Article  Google Scholar 

  12. Shima T, Kinoshita T, Sasaki N, Uematsu M, Sugita Y, Shimizu R, et al. Feasibility of intraoperative diagnosis of lung adenocarcinoma in situ to avoid excessive resection. J Thorac Dis. 2021;13:1338–46. https://doi.org/10.21037/jtd-20-2710.

    Article  Google Scholar 

  13. Yeh Y-C, Nitadori J-I, Kadota K, Yoshizawa A, Rekhtman N, Moreira AL, et al. Using frozen section to identify histological patterns in stage I lung adenocarcinoma of ≤3 cm: accuracy and interobserver agreement. Histopathology. 2015;66:922–38. https://doi.org/10.1111/his.12468.

    Article  Google Scholar 

  14. Li F, Yang L, Zhao Y, Yuan L, Wang S, Mao Y. Intraoperative frozen section for identifying the invasion status of lung adenocarcinoma: a systematic review and meta-analysis. Int J Surg. 2019;72:175–84. https://doi.org/10.1016/j.ijsu.2019.10.047.

    Article  Google Scholar 

  15. Rosenthal EL, Warram JM, de Boer E, Chung TK, Korb ML, Brandwein-Gensler M, et al. Safety and tumor specificity of cetuximab-IRDye800 for surgical navigation in head and neck cancer. Clin Cancer Res. 2015;21:3658–66. https://doi.org/10.1158/1078-0432.CCR-14-3284.

    Article  CAS  Google Scholar 

  16. Lauwerends LJ, van Driel PB, de Jong RJB, Hardillo JA, Koljenovic S, Puppels G, et al. Real-time fluorescence imaging in intraoperative decision making for cancer surgery. Lancet Oncol. 2021;22:e186–95.

    Article  CAS  Google Scholar 

  17. van Keulen S, Nishio N, Birkeland A, Fakurnejad S, Martin B, Forouzanfar T, et al. The sentinel margin: intraoperative ex vivo specimen mapping using relative fluorescence intensity. Clin Cancer Res. 2019;25:4656–62. https://doi.org/10.1158/1078-0432.CCR-19-0319.

    Article  Google Scholar 

  18. Linssen MD, Ter Weele EJ, Allersma DP, Lub-de Hooge MN, van Dam GM, Jorritsma-Smit A, et al. Roadmap for the development and clinical translation of optical tracers cetuximab-800CW and trastuzumab-800CW. J Nucl Med. 2019;60:418–23. https://doi.org/10.2967/jnumed.118.216556.

    Article  CAS  Google Scholar 

  19. Rosenthal EL, Moore LS, Tipirneni K, de Boer E, Stevens TM, Hartman YE, et al. Sensitivity and specificity of cetuximab-IRDye800CW to identify regional metastatic disease in head and neck cancer. Clin Cancer Res. 2017;23:4744–52. https://doi.org/10.1158/1078-0432.CCR-16-2968.

    Article  CAS  Google Scholar 

  20. Gao RW, Teraphongphom N, de Boer E, van den Berg NS, Divi V, Kaplan MJ, et al. Safety of panitumumab-IRDye800CW and cetuximab-IRDye800CW for fluorescence-guided surgical navigation in head and neck cancers. Theranostics. 2018;8:2488–95. https://doi.org/10.7150/thno.24487.

    Article  CAS  Google Scholar 

  21. Day KE, Sweeny L, Kulbersh B, Zinn KR, Rosenthal EL. Preclinical comparison of near-infrared-labeled cetuximab and panitumumab for optical imaging of head and neck squamous cell carcinoma. Mol Imag Biol. 2013;15:722–9. https://doi.org/10.1007/s11307-013-0652-9.

    Article  Google Scholar 

  22. Vonk J, Voskuil FJ, de Wit JG, Heeman WT, Nagengast WB, van Dam GM, et al. Fluorescence grid analysis for the evaluation of piecemeal surgery in sinonasal inverted papilloma: a proof-of-concept study. Eur J Nucl Med Mol Imaging. 2022;49:1640–9. https://doi.org/10.1007/s00259-021-05567-x.

    Article  CAS  Google Scholar 

  23. Quan YH, Bao K, Kim K, Wang H, Yokomizo S, Park GK, et al. Abstract 5975: Dual-channel near-infrared fluorescence imaging for simultaneous identification of lung cancer and intersegmental plane. Cancer Res. 2022;82:5975. https://doi.org/10.1158/1538-7445.AM2022-5975.

    Article  Google Scholar 

  24. Yang J, He S, Hu Z, Zhang Z, Cao C, Cheng Z, et al. In vivo multifunctional fluorescence imaging using liposome-coated lanthanide nanoparticles in near-infrared-II/IIa/IIb windows. Nano Today. 2021;38:101120. https://doi.org/10.1016/j.nantod.2021.101120.

    Article  CAS  Google Scholar 

  25. Chang B, Li D, Ren Y, Qu C, Shi X, Liu R, et al. A phosphorescent probe for in vivo imaging in the second near-infrared window. Nat Biomed Eng. 2022;6:629–39. https://doi.org/10.1038/s41551-021-00773-2.

    Article  CAS  Google Scholar 

  26. Lei Z, Zhang F. Molecular engineering of NIR-II fluorophores for improved biomedical detection. Angew Chem Int Ed. 2021;60:16294–308. https://doi.org/10.1002/anie.202007040.

    Article  CAS  Google Scholar 

  27. Tao W, Farokhzad OC. Theranostic nanomedicine in the NIR-II window: classification, fabrication, and biomedical applications. Chem Rev. 2022;122:5405–7. https://doi.org/10.1021/acs.chemrev.2c00089.

    Article  CAS  Google Scholar 

  28. Hu Z, Fang C, Li B, Zhang Z, Cao C, Cai M, et al. First-in-human liver-tumour surgery guided by multispectral fluorescence imaging in the visible and near-infrared-I/II windows. Nat Biomed Eng. 2020;4:259–71. https://doi.org/10.1038/s41551-019-0494-0.

    Article  Google Scholar 

  29. Eatz TA, Eichberg DG, Lu VM, Di L, Komotar RJ, Ivan ME. Intraoperative 5-ALA fluorescence-guided resection of high-grade glioma leads to greater extent of resection with better outcomes: a systematic review. J Neurooncol. 2022;156:233–56. https://doi.org/10.1007/s11060-021-03901-9.

    Article  CAS  Google Scholar 

  30. Duan H, Pang Y, Zhao C, Zhou T, Sun C, Hou M, et al. A novel, minimally invasive technique to establish the animal model of spinal cord injury. Ann Transl Med. 2021;9:881. https://doi.org/10.21037/atm-21-2063.

    Article  Google Scholar 

  31. Patrunov Y, Sencha AN, Sencha E, Penyaeva E. Ultrasound of regional lymph nodes in breast cancer. In: Sukhikh GT, Sencha AN, editors. Multiparametric ultrasound diagnosis of breast diseases. Cham: Springer International Publishing; 2018. p. 265–84.

    Chapter  Google Scholar 

  32. Yinhao, Pan Ningbo, Chen Liangjian, Liu Chengbo, Liu Zhiqiang, Xu Jianhui, Zhang (2021) Recovery of photoacoustic images based on accurate ultrasound positioning. Visual Computing for Industry Biomedicine and Art 4(1):7. https://doi.org/10.1186/s42492-021-00072-2

  33. Lubner MG, Mankowski Gettle L, Kim DH, Ziemlewicz TJ, Dahiya N, Pickhardt P. Diagnostic and procedural intraoperative ultrasound: technique, tips and tricks for optimizing results. Br J Radiol. 2021;94:20201406. https://doi.org/10.1259/bjr.20201406.

    Article  Google Scholar 

  34. Fuentes AM, Ansari D, Burch TG, Mehta AI. Use of intraoperative MRI for resection of intracranial tumors: a nationwide analysis of short-term outcomes. J Clin Neurosci. 2022;99:152–7. https://doi.org/10.1016/j.jocn.2022.03.005.

    Article  Google Scholar 

  35. Golub D, Hyde J, Dogra S, Nicholson J, Kirkwood KA, Gohel P, et al. Intraoperative MRI versus 5-ALA in high-grade glioma resection: a network meta-analysis. J Neurosurg JNS. 2021;134:484–98. https://doi.org/10.3171/2019.12.JNS191203.

    Article  CAS  Google Scholar 

  36. Rogers CM, Jones PS, Weinberg JS. Intraoperative MRI for brain tumors. J Neurooncol. 2021;151:479–90. https://doi.org/10.1007/s11060-020-03667-6.

    Article  Google Scholar 

  37. Qiu Y, Zhang Y, Li M, Chen G, Fan C, Cui K, et al. Intraoperative detection and eradication of residual microtumors with gap-enhanced Raman tags. ACS Nano. 2018;12:7974–85. https://doi.org/10.1021/acsnano.8b02681.

    Article  CAS  Google Scholar 

  38. Cui K, Zhang Y, Chen G, Cui Y, Wu W, Zhao N, et al. Molecular regulation of polymeric Raman probes for ultrasensitive microtumor diagnosis and noninvasive microvessel imaging. Small. 2022;18:2106925. https://doi.org/10.1002/smll.202106925.

    Article  CAS  Google Scholar 

  39. Jiang C, Wang Y, Song W, Lu L. Delineating the tumor margin with intraoperative surface-enhanced Raman spectroscopy. Anal Bioanal Chem. 2019;411:3993–4006. https://doi.org/10.1007/s00216-019-01577-9.

    Article  CAS  Google Scholar 

  40. Jermyn M, Mok K, Mercier J, Desroches J, Pichette J, Saint-Arnaud K, et al. Intraoperative brain cancer detection with Raman spectroscopy in humans. Sci Translat Med. 2015;7:274ra19. https://doi.org/10.1126/scitranslmed.aaa2384.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported in part by the National Natural Science Foundation of China (grants no. 62027901), the Fundamental Research Funds for the Central Universities (grant no. JKF-YG-22-B005), the National Key Research and Development Program of China (under Grants 2017YFA0205200, 2016YFC0103803, and 2017YFA0700401), the National Natural Science Foundation of China (grants nos. 81227901, 81930053, 81827808, 81527805, 81971198, 81671851, 82003316, and 92059203), China Postdoctoral Science Foundation (grant nos. 2020M680301 and 2019TQ0018), Chinese Academy of Sciences Youth Innovation Promotion Association (grant no. 2018167), CAS Youth Interdisciplinary Team (JCTD-2021-08), the Strategic Priority Research Program of the Chinese Academy of Sciences (XDA16021200), Chinese Academy of Sciences Key Technology Talent Program, Project of High-Level Talents Team Introduction in Zhuhai City (Zhuhai HLHPTP201703), Beijing Natural Science Foundation (grant nos. 7214266, JQ19027), and Research and Development Fund of Peking University People’s Hospital (grant no. RS2020-05). The authors would like to acknowledge the instrumental technical support of the Multimodal Biomedical Imaging Experimental Platform, Institute of Automation, Chinese Academy of Sciences.

Author information

Authors and Affiliations

Authors

Contributions

Changjian Li synthesized the probe. Changjian Li and Jiahui Mi performed the experiments. Changjian Li and Yueqi Wang analyzed the data and wrote the manuscript. Jie Tian, Zhenhua Hu, and Jian Zhou supervised the designation of the experiments. Zeyu Zhang and Xiaoyong Guo assisted with data analysis.

Corresponding authors

Correspondence to Jian Zhou, Zhenhua Hu or Jie Tian.

Ethics declarations

Ethics approval and consent to participate

In human subjects, the study was approved by the Institutional Review Board at the Peking University People’s Hospital (Permit Number: 2021PHB315-001). In animal subjects, all experiments were performed according to the guidelines of the Institutional Animal Care and Use Committee of Beijing Municipal Science & Technology Commission (Permit Number: 2020–0049).

Consent for publication

All the co-authors approved the manuscript and agreed with submission to your esteemed journal.

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

Below is the link to the electronic supplementary material.

Supplementary file1 (DOCX 9770 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

Li, C., Mi, J., Wang, Y. et al. New and effective EGFR-targeted fluorescence imaging technology for intraoperative rapid determination of lung cancer in freshly isolated tissue. Eur J Nucl Med Mol Imaging 50, 494–507 (2023). https://doi.org/10.1007/s00259-022-05975-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00259-022-05975-7

Keywords

Navigation