Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Fundamentals and developments in fluorescence-guided cancer surgery

Abstract

Fluorescence-guided surgery using tumour-targeted imaging agents has emerged over the past decade as a promising and effective method of intraoperative cancer detection. An impressive number of fluorescently labelled antibodies, peptides, particles and other molecules related to cancer hallmarks have been developed for the illumination of target lesions. New approaches are being implemented to translate these imaging agents into the clinic, although only a few have made it past early-phase clinical trials. For this translational process to succeed, target selection, imaging agents and their related detection systems and clinical implementation have to operate in perfect harmony to enable real-time intraoperative visualization that can benefit patients. Herein, we review key aspects of this imaging cascade and focus on imaging approaches and methods that have helped to shed new light onto the field of intraoperative fluorescence-guided cancer surgery with the singular goal of improving patient outcomes.

Key points

  • Tumour-targeted fluorescence-guided surgery (FGS) has emerged as a promising technique for the intraoperative visualization of solid tumours.

  • Different approaches can be used to make imaging agents specifically bind to or interact with a target protein associated with a cancer hallmark.

  • Imaging systems for clinical tumour-targeted FGS are available and provide real-time qualitative images.

  • Phase III studies are currently ongoing to prove patient benefit from tumour-targeted FGS.

  • Future camera developments focus on the quantification of fluorescence imaging by correcting for an array of optical properties.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Important stages in the development and clinical translation of tumour-targeted imaging agents.
Fig. 2: Timeline of seminal discoveries leading to the development of tumour-targeted fluorescence-guided surgery.
Fig. 3: Overview of approaches to enable tumour-targeted imaging.
Fig. 4: Effect of photon and tissue properties in qualitative output in clinical fluorescence imaging.

Similar content being viewed by others

References

  1. Alam, I. S. et al. Emerging intraoperative imaging modalities to improve surgical precision. Mol. Imaging Biol. 20, 705–715 (2018).

    Article  PubMed  Google Scholar 

  2. Frangioni, J. V. Translating in vivo diagnostics into clinical reality. Nat. Biotechnol. 24, 909–913 (2006).

    Article  CAS  PubMed  Google Scholar 

  3. Vahrmeijer, A. L., Hutteman, M., van der Vorst, J. R., van de Velde, C. J. & Frangioni, J. V. Image-guided cancer surgery using near-infrared fluorescence. Nat. Rev. Clin. Oncol. 10, 507–518 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Guttmann, P., Ehrlich, P. Berliner Klinische Wochenschrift 953-956 (Springer, 1891).

  5. Moore, G. E. et al. The clinical use of fluorescein in neurosurgery; the localization of brain tumors. J. Neurosurg. 5, 392–398 (1948).

    Article  CAS  PubMed  Google Scholar 

  6. Fox, I. J. & Wood, E. H. Indocyanine green: physical and physiologic properties. Proc. Staff. Meet. Mayo Clin. 35, 732–744 (1960).

    CAS  PubMed  Google Scholar 

  7. Watts, G. Bernard Fisher. Lancet 394, 1900 (2019).

    Article  Google Scholar 

  8. Best, S. JOAN RIBAS GISPERT Coordination chemistry Wiley–VCH, 2008, 600 pp. (paperback) ISBN-10 3-527-31802-X ISBN-13 978-3-527-31802-5.

  9. Keereweer, S. et al. Optical image-guided cancer surgery: challenges and limitations. Clin. Cancer Res. 19, 3745–3754 (2013).

    Article  PubMed  Google Scholar 

  10. Hernot, S., van Manen, L., Debie, P., Mieog, J. S. D. & Vahrmeijer, A. L. Latest developments in molecular tracers for fluorescence image-guided cancer surgery. Lancet Oncol. 20, e354–e367 (2019).

    Article  CAS  PubMed  Google Scholar 

  11. de Geus, S. W. et al. Selecting tumor-specific molecular targets in pancreatic adenocarcinoma: paving the way for image-guided pancreatic surgery. Mol. Imaging Biol. 18, 807–819 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. van Oosten, M., Crane, L. M., Bart, J., van Leeuwen, F. W. & van Dam, G. M. Selecting potential targetable biomarkers for imaging purposes in colorectal cancer using target selection criteria (TASC): a novel target identification tool. Transl. Oncol. 4, 71–82 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Achterberg, F. B. et al. Clinical translation and implementation of optical imaging agents for precision image-guided cancer surgery. Eur. J. Nucl. Med. Mol. Imaging 48, 332–339 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Weissleder, R., Schwaiger, M. C., Gambhir, S. S. & Hricak, H. Imaging approaches to optimize molecular therapies. Sci. Transl. Med. 8, 355ps316 (2016).

    Article  CAS  Google Scholar 

  15. Hanahan, D. & Weinberg, R. A. Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011).

    Article  CAS  PubMed  Google Scholar 

  16. Baart, V. M. et al. EGFR and αvβ6 as promising targets for molecular imaging of cutaneous and mucosal squamous cell carcinoma of the head and neck region. Cancers 12, 1474 (2020).

    Article  CAS  PubMed Central  Google Scholar 

  17. de Gouw, D. et al. Identifying biomarkers in lymph node metastases of esophageal adenocarcinoma for tumor-targeted imaging. Mol. Diagn. Ther. 24, 191–200 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Boogerd, L. S. et al. Biomarker expression in rectal cancer tissue before and after neoadjuvant therapy. OncoTargets Ther. 11, 1655–1664 (2018).

    Article  Google Scholar 

  19. Vuijk, F. A. et al. Molecular targets for diagnostic and intraoperative imaging of pancreatic ductal adenocarcinoma after neoadjuvant FOLFIRINOX treatment. Sci. Rep. 10, 16211 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Tummers, W. S. et al. Selection of optimal molecular targets for tumor-specific imaging in pancreatic ductal adenocarcinoma. Oncotarget 8, 56816–56828 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Hartmans, E. et al. Potential red-flag identification of colorectal adenomas with wide-field fluorescence molecular endoscopy. Theranostics 8, 1458–1467 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Boogerd, L. S. F. et al. Safety and effectiveness of SGM-101, a fluorescent antibody targeting carcinoembryonic antigen, for intraoperative detection of colorectal cancer: a dose-escalation pilot study. Lancet Gastroenterol. Hepatol. 3, 181–191 (2018).

    Article  PubMed  Google Scholar 

  23. Achterberg, F. B. et al. Real-time surgical margin assessment using ICG-fluorescence during laparoscopic and robot-assisted resections of colorectal liver metastases. Ann. Transl. Med. 8, 1448 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Lamberts, L. E. et al. Tumor-specific uptake of fluorescent bevacizumab-IRDye800CW microdosing in patients with primary breast cancer: a phase I feasibility study. Clin. Cancer Res. 23, 2730–2741 (2017).

    Article  CAS  PubMed  Google Scholar 

  25. Nagengast, W. B. et al. Near-infrared fluorescence molecular endoscopy detects dysplastic oesophageal lesions using topical and systemic tracer of vascular endothelial growth factor A. Gut 68, 7–10 (2019).

    Article  CAS  PubMed  Google Scholar 

  26. Korb, M. L. et al. Use of monoclonal antibody-IRDye800CW bioconjugates in the resection of breast cancer. J. Surg. Res. 188, 119–128 (2014).

    Article  CAS  PubMed  Google Scholar 

  27. Marston, J. C. et al. Panitumumab-IRDye800CW for fluorescence-guided surgical resection of colorectal cancer. J. Surg. Res. 239, 44–51 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Harlaar, N. J. et al. Molecular fluorescence-guided surgery of peritoneal carcinomatosis of colorectal origin: a single-centre feasibility study. Lancet Gastroenterol. Hepatol. 1, 283–290 (2016).

    Article  PubMed  Google Scholar 

  29. Kanazawa, S. et al. VEGF, basic-FGF, and TGF-beta in Crohn’s disease and ulcerative colitis: a novel mechanism of chronic intestinal inflammation. Am. J. Gastroenterol. 96, 822–828 (2001).

    CAS  PubMed  Google Scholar 

  30. Derks, Y. H. W. et al. PSMA-targeting agents for radio- and fluorescence-guided prostate cancer surgery. Theranostics 9, 6824–6839 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. van der Fels, C. A. M. et al. Potential receptors for targeted imaging of lymph node metastases in penile cancer. Diagnostics 10, 694 (2020).

    Article  PubMed Central  CAS  Google Scholar 

  32. Boonstra, M. C. et al. Stromal targets for fluorescent-guided oncologic surgery. Front. Oncol. 5, 254 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Urano, Y. et al. Selective molecular imaging of viable cancer cells with pH-activatable fluorescence probes. Nat. Med. 15, 104–109 (2009).

    Article  CAS  PubMed  Google Scholar 

  34. Debie, P. & Hernot, S. Emerging fluorescent molecular tracers to guide intra-operative surgical decision-making. Front. Pharmacol. 10, 510 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. van Leeuwen, F. W. B. et al. Trending: radioactive and fluorescent bimodal/hybrid tracers as multiplexing solutions for surgical guidance. J. Nucl. Med. 61, 13–19 (2020).

    Article  PubMed  Google Scholar 

  36. Mayer, A. T. & Gambhir, S. S. The immunoimaging toolbox. J. Nucl. Med. 59, 1174–1182 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Choi, H. S. et al. Design considerations for tumour-targeted nanoparticles. Nat. Nanotechnol. 5, 42–47 (2010).

    Article  CAS  PubMed  Google Scholar 

  38. van Dam, G. M. et al. Intraoperative tumor-specific fluorescence imaging in ovarian cancer by folate receptor-α targeting: first in-human results. Nat. Med. 17, 1315–1319 (2011).

    Article  PubMed  CAS  Google Scholar 

  39. Warram, J. M. et al. Antibody-based imaging strategies for cancer. Cancer Metastasis Rev. 33, 809–822 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Gong, H., Kovar, J. L., Cheung, L., Rosenthal, E. L. & Olive, D. M. A comparative study of affibody, panitumumab, and EGF for near-infrared fluorescence imaging of EGFR- and EGFRvIII-expressing tumors. Cancer Biol. Ther. 15, 185–193 (2014).

    Article  CAS  PubMed  Google Scholar 

  41. Heath, C. H. et al. Use of panitumumab-IRDye800 to image cutaneous head and neck cancer in mice. Otolaryngol. Head Neck Surg. 148, 982–990 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Miller, S. E. et al. First-in-human intraoperative near-infrared fluorescence imaging of glioblastoma using cetuximab-IRDye800. J. Neurooncol. 139, 135–143 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Odenthal, J. et al. Targeting CD44v6 for fluorescence-guided surgery in head and neck squamous cell carcinoma. Sci. Rep. 8, 10467 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Rijpkema, M. et al. SPECT- and fluorescence image-guided surgery using a dual-labeled carcinoembryonic antigen-targeting antibody. J. Nucl. Med. 55, 1519–1524 (2014).

    Article  CAS  PubMed  Google Scholar 

  45. Debie, P. et al. Improved debulking of peritoneal tumor implants by near-infrared fluorescent nanobody image guidance in an experimental mouse model. Mol. Imaging Biol. 20, 361–367 (2018).

    Article  PubMed  Google Scholar 

  46. Gao, R. W. et al. Safety of panitumumab-IRDye800CW and cetuximab-IRDye800CW for fluorescence-guided surgical navigation in head and neck cancers. Theranostics 8, 2488–2495 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Hollandsworth, H. M. et al. Anti-carcinoembryonic antigen-related cell adhesion molecule antibody for fluorescence visualization of primary colon cancer and metastases in patient-derived orthotopic xenograft mouse models. Oncotarget 11, 429–439 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Koller, M. et al. Implementation and benchmarking of a novel analytical framework to clinically evaluate tumor-specific fluorescent tracers. Nat. Commun. 9, 3739 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Nishio, N. et al. Optimal dosing strategy for fluorescence-guided surgery with panitumumab-IRDye800CW in head and neck cancer. Mol. Imaging Biol. 22, 156–164 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Rosenthal, E. L. et al. Safety and tumor specificity of cetuximab-IRDye800 for surgical navigation in head and neck cancer. Clin. Cancer Res. 21, 3658–3666 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Tummers, W. S. et al. Detection of visually occult metastatic lymph nodes using molecularly targeted fluorescent imaging during surgical resection of pancreatic cancer. HPB 21, 883–890 (2019).

    Article  PubMed  Google Scholar 

  52. Tummers, W. S. et al. Intraoperative pancreatic cancer detection using tumor-specific multimodality molecular imaging. Ann. Surg. Oncol. 25, 1880–1888 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Tummers, W. S. et al. Regulatory aspects of optical methods and exogenous targets for cancer detection. Cancer Res. 77, 2197–2206 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Fakurnejad, S. et al. Intraoperative molecular imaging for ex vivo assessment of peripheral margins in oral squamous cell carcinoma. Front. Oncol. 9, 1476 (2019).

    Article  PubMed  Google Scholar 

  55. van Keulen, S. et al. The clinical application of fluorescence-guided surgery in head and neck cancer. J. Nucl. Med. 60, 758–763 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  56. de Valk, K. S. et al. Dose-finding study of a CEA-targeting agent, SGM-101, for intraoperative fluorescence imaging of colorectal cancer. Ann. Surg. Oncol. 28, 1832–1844 (2021).

    Article  PubMed  Google Scholar 

  57. Meijer, R. P. J. et al. Intraoperative detection of colorectal and pancreatic liver metastases using SGM-101, a fluorescent antibody targeting CEA. Eur. J. Surg. Oncol. 47, 667–673 (2021).

    Article  PubMed  Google Scholar 

  58. Schaap, D. P. et al. Carcinoembryonic antigen-specific, fluorescent image-guided cytoreductive surgery with hyperthermic intraperitoneal chemotherapy for metastatic colorectal cancer. Br. J. Surg. 107, 334–337 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Thurber, G. M., Schmidt, M. M. & Wittrup, K. D. Factors determining antibody distribution in tumors. Trends Pharmacol. Sci. 29, 57–61 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Lu, G. et al. Predicting therapeutic antibody delivery into human head and neck cancers. Clin. Cancer Res. 26, 2582–2594 (2020).

    Article  CAS  PubMed  Google Scholar 

  61. Moore, L. S. et al. Characterizing the utility and limitations of repurposing an open-field optical imaging device for fluorescence-guided surgery in head and neck cancer patients. J. Nucl. Med. 58, 246–251 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Debie, P. et al. Size and affinity kinetics of nanobodies influence targeting and penetration of solid tumours. J. Control. Rel. 317, 34–42 (2020).

    Article  CAS  Google Scholar 

  63. Zhang, M. et al. Near-infrared dye-labeled anti-prostate stem cell antigen minibody enables real-time fluorescence imaging and targeted surgery in translational mouse models. Clin. Cancer Res. 25, 188–200 (2019).

    Article  PubMed  Google Scholar 

  64. Tsai, W. K. et al. Dual-modality immunoPET/fluorescence imaging of prostate cancer with an anti-PSCA Cys-minibody. Theranostics 8, 5903–5914 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Boogerd, L. S. F. et al. Fluorescence-guided tumor detection with a novel anti-EpCAM targeted antibody fragment: preclinical validation. Surg. Oncol. 28, 1–8 (2019).

    Article  PubMed  Google Scholar 

  66. El-Sayed, A. et al. Evaluation of antibody fragment properties for near-infrared fluorescence imaging of HER3-positive cancer xenografts. Theranostics 8, 4856–4869 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Bernhard, W. et al. Near infrared imaging of epidermal growth factor receptor positive xenografts in mice with domain I/II specific antibody fragments. Theranostics 9, 974–985 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Bam, R. et al. Affibody-indocyanine green based contrast agent for photoacoustic and fluorescence molecular imaging of B7-H3 expression in breast cancer. Bioconjug. Chem. 30, 1677–1689 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Tummers, W. S. et al. Development and preclinical validation of a cysteine knottin peptide targeting integrin alphavbeta6 for near-infrared fluorescent-guided surgery in pancreatic cancer. Clin. Cancer Res. 24, 1667–1676 (2018).

    Article  CAS  PubMed  Google Scholar 

  70. Kintzing, J. R. & Cochran, J. R. Engineered knottin peptides as diagnostics, therapeutics, and drug delivery vehicles. Curr. Opin. Chem. Biol. 34, 143–150 (2016).

    Article  CAS  PubMed  Google Scholar 

  71. de Valk, K. S. et al. First-in-human assessment of cRGD-ZW800-1, a Zwitterionic, integrin-targeted, near-infrared fluorescent peptide in colon carcinoma. Clin. Cancer Res. 26, 3990–3998 (2020).

    Article  PubMed  Google Scholar 

  72. Leptin, M. The fibronectin receptor family. Nature 321, 728 (1986).

    Article  CAS  PubMed  Google Scholar 

  73. Kitagawa, T. et al. RGD-conjugated human ferritin nanoparticles for imaging vascular inflammation and angiogenesis in experimental carotid and aortic disease. Mol. Imaging Biol. 14, 315–324 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  74. Chen, H., Niu, G., Wu, H. & Chen, X. Clinical application of radiolabeled RGD peptides for PET imaging of integrin alphavbeta3. Theranostics 6, 78–92 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Iagaru, A. et al. 18F-FPPRGD2 PET/CT: pilot phase evaluation of breast cancer patients. Radiology 273, 549–559 (2014).

    Article  PubMed  Google Scholar 

  76. Lee, J. Y. K. et al. Review of clinical trials in intraoperative molecular imaging during cancer surgery. J. Biomed. Opt. 24, 1–8 (2019).

    Article  PubMed  Google Scholar 

  77. Olson, M. T., Ly, Q. P. & Mohs, A. M. Fluorescence guidance in surgical oncology: challenges, opportunities, and translation. Mol. Imaging Biol. 21, 200–218 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  78. Pogue, B. W., Rosenthal, E. L., Achilefu, S. & van Dam, G. M. Perspective review of what is needed for molecular-specific fluorescence-guided surgery. J. Biomed. Opt. 23, 1–9 (2018).

    PubMed  Google Scholar 

  79. Kelemen, L. E. The role of folate receptor α in cancer development, progression and treatment: cause, consequence or innocent bystander? Int. J. Cancer 119, 243–250 (2006).

    Article  CAS  PubMed  Google Scholar 

  80. Hartmann, L. C. et al. Folate receptor overexpression is associated with poor outcome in breast cancer. Int. J. Cancer 121, 938–942 (2007).

    Article  CAS  PubMed  Google Scholar 

  81. Kalli, K. R. et al. Folate receptor alpha as a tumor target in epithelial ovarian cancer. Gynecol. Oncol. 108, 619–626 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. O'Shannessy, D. J. et al. Folate receptor alpha expression in lung cancer: diagnostic and prognostic significance. Oncotarget 3, 414–425 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  83. Salazar, M. D. A. & Ratnam, M. The folate receptor: What does it promise in tissue-targeted therapeutics? Cancer Metastasis Rev. 26, 141–152 (2007).

    Article  CAS  PubMed  Google Scholar 

  84. Keating, J. J. et al. Intraoperative near-infrared fluorescence imaging targeting folate receptors identifies lung cancer in a large-animal model. Cancer 123, 1051–1060 (2016).

    Article  PubMed  CAS  Google Scholar 

  85. Mahalingam, S. M. et al. Evaluation of novel tumor-targeted near-infrared probe for fluorescence-guided surgery of cancer. J. Med. Chem. 61, 9637–9646 (2018).

    Article  CAS  PubMed  Google Scholar 

  86. Srinivasarao, M., Galliford, C. V. & Low, P. S. Principles in the design of ligand-targeted cancer therapeutics and imaging agents. Nat. Rev. Drug Discov. 14, 203–219 (2015).

    Article  CAS  PubMed  Google Scholar 

  87. Shum, C. F. et al. Novel use of folate-targeted intraoperative fluorescence, OTL38, in robot-assisted laparoscopic partial nephrectomy: report of the first three cases. J. Endourol. Case Rep. 2, 189–197 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  88. Hoogstins, C. E. S. et al. A novel tumor-specific agent for intraoperative near-infrared fluorescence imaging: a translational study in healthy volunteers and patients with ovarian cancer. Clin. Cancer Res. 22, 2929–2938 (2016).

    Article  CAS  PubMed  Google Scholar 

  89. Predina, J. D. et al. Intraoperative molecular imaging combined with positron emission tomography improves surgical management of peripheral malignant pulmonary nodules. Ann. Surg. 266, 479 (2017).

    Article  PubMed  Google Scholar 

  90. Predina, J. D. et al. A phase I clinical trial of targeted intraoperative molecular imaging for pulmonary adenocarcinomas. Ann. Thorac. Surg. 105, 901–908 (2018).

    Article  PubMed  Google Scholar 

  91. Boogerd, L. S. F. et al. Folate receptor-α targeted near-infrared fluorescence imaging in high-risk endometrial cancer patients: a tissue microarray and clinical feasibility study. Oncotarget 9, 791–801 (2018).

    Article  PubMed  Google Scholar 

  92. Randall, L. M., Wenham, R. M., Low, P. S., Dowdy, S. C. & Tanyi, J. L. A phase II, multicenter, open-label trial of OTL38 injection for the intra-operative imaging of folate receptor-alpha positive ovarian cancer. Gynecol. Oncol. 155, 63–68 (2019).

    Article  CAS  PubMed  Google Scholar 

  93. Kularatne, S. A. et al. Evaluation of novel prostate-specific membrane antigen-targeted near-infrared imaging agent for fluorescence-guided surgery of prostate cancer. Clin. Cancer Res. 25, 177–187 (2019).

    Article  CAS  PubMed  Google Scholar 

  94. Netherlands Trial Register. TrialRegister.nl https://www.trialregister.nl/trial/8552 (2020).

  95. Stummer, W. et al. Fluorescence-guided surgery with 5-aminolevulinic acid for resection of malignant glioma: a randomised controlled multicentre phase III trial. Lancet Oncol. 7, 392–401 (2006).

    Article  CAS  PubMed  Google Scholar 

  96. Maeda, H. Toward a full understanding of the EPR effect in primary and metastatic tumors as well as issues related to its heterogeneity. Adv. Drug Deliv. Rev. 91, 3–6 (2015).

    Article  CAS  PubMed  Google Scholar 

  97. Hare, J. I. et al. Challenges and strategies in anti-cancer nanomedicine development: An industry perspective. Adv. Drug Deliv. Rev. 108, 25–38 (2017).

    Article  CAS  PubMed  Google Scholar 

  98. Golombek, S. K. et al. Tumor targeting via EPR: strategies to enhance patient responses. Adv. Drug Deliv. Rev. 130, 17–38 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Sindhwani, S. et al. The entry of nanoparticles into solid tumours. Nat. Mater. 19, 566–575 (2020).

    Article  CAS  PubMed  Google Scholar 

  100. Wojtynek, N. E. & Mohs, A. M. Image-guided tumor surgery: the emerging role of nanotechnology. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 12, e1624 (2020).

    Article  PubMed  Google Scholar 

  101. Tanaka, N. et al. Whole-tissue biopsy phenotyping of three-dimensional tumours reveals patterns of cancer heterogeneity. Nat. Biomed. Eng. 1, 796–806 (2017).

    Article  CAS  PubMed  Google Scholar 

  102. Das, S. S. et al. Stimuli-responsive polymeric nanocarriers for drug delivery, imaging, and theragnosis. Polymers 12, 1397 (2020).

    Article  CAS  PubMed Central  Google Scholar 

  103. Hu, Z., Chen, W. H., Tian, J. & Cheng, Z. NIRF nanoprobes for cancer molecular imaging: approaching clinic. Trends Mol. Med. 26, 469–482 (2020).

    Article  CAS  PubMed  Google Scholar 

  104. Anselmo, A. C. & Mitragotri, S. A review of clinical translation of inorganic nanoparticles. AAPS J. 17, 1041–1054 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Anselmo, A. C. & Mitragotri, S. Nanoparticles in the clinic. Bioeng. Transl. Med. 1, 10–29 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  106. Wang, Y. et al. A nanoparticle-based strategy for the imaging of a broad range of tumours by nonlinear amplification of microenvironment signals. Nat. Mater. 13, 204–212 (2014).

    Article  CAS  PubMed  Google Scholar 

  107. Benezra, M. et al. Multimodal silica nanoparticles are effective cancer-targeted probes in a model of human melanoma. J. Clin. Invest. 121, 2768–2780 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Chen, F. et al. Cancer-targeting ultrasmall silica nanoparticles for clinical translation: physicochemical structure and biological property correlations. Chem. Mater. 29, 8766–8779 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Phillips, E. et al. Clinical translation of an ultrasmall inorganic optical-PET imaging nanoparticle probe. Sci. Transl. Med. 6, 260ra149 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  110. Zhao, T. et al. A transistor-like pH nanoprobe for tumour detection and image-guided surgery. Nat. Biomed. Eng. 1, 0006 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  111. Frangioni, J. V. The problem is background, not signal. Mol. Imaging 8, 303–304 (2009).

    Article  PubMed  Google Scholar 

  112. Hoogstins, C. et al. Setting standards for reporting and quantification in fluorescence-guided surgery. Mol. Imaging Biol. 21, 11–18 (2019).

    Article  PubMed  Google Scholar 

  113. Deken, M. M. et al. Near-infrared fluorescence imaging compared to standard sentinel lymph node detection with blue dye in patients with vulvar cancer — a randomized controlled trial. Gynecol. Oncol. 159, 672–680 (2020).

    Article  CAS  PubMed  Google Scholar 

  114. Koch, M., Symvoulidis, P. & Ntziachristos, V. Tackling standardization in fluorescence molecular imaging. Nat. Photonics 12, 505–515 (2018).

    Article  CAS  Google Scholar 

  115. Nishio, N. et al. Probe-based fluorescence dosimetry of an antibody-dye conjugate to identify head and neck cancer as a first step to fluorescence-guided tissue preselection for pathological assessment. Head Neck 42, 59–66 (2020).

    Article  PubMed  Google Scholar 

  116. Cahill, R. A. et al. Artificial intelligence indocyanine green (ICG) perfusion for colorectal cancer intra-operative tissue classification. Br. J. Surg. 108, 5–9 (2021).

    Article  CAS  PubMed  Google Scholar 

  117. Antaris, A. L. et al. A small-molecule dye for NIR-II imaging. Nat. Mater. 15, 235–242 (2016).

    Article  CAS  PubMed  Google Scholar 

  118. Wan, H. et al. A bright organic NIR-II nanofluorophore for three-dimensional imaging into biological tissues. Nat. Commun. 9, 1171–1179 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  119. Hu, Z. 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. 4, 259–271 (2020).

    Article  PubMed  Google Scholar 

  120. Kimura, R. H. et al. Evaluation of integrin alphavbeta6 cystine knot PET tracers to detect cancer and idiopathic pulmonary fibrosis. Nat. Commun. 10, 4673 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  121. Lu, G. et al. Co-administered antibody improves penetration of antibody-dye conjugate into human cancers with implications for antibody-drug conjugates. Nat. Commun. 11, 5667 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Garland, M., Yim, J. J. & Bogyo, M. A bright future for precision medicine: advances in fluorescent chemical probe design and their clinical application. Cell Chem. Biol. 23, 122–136 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Landau, M. J., Gould, D. J. & Patel, K. M. Advances in fluorescent-image guided surgery. Ann. Transl. Med. 4, 392 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  124. Staderini, M., Megia-Fernandez, A., Dhaliwal, K. & Bradley, M. Peptides for optical medical imaging and steps towards therapy. Bioorg. Med. Chem. 26, 2816–2826 (2018).

    Article  CAS  PubMed  Google Scholar 

  125. Yang, Z. et al. Macro-/micro-environment-sensitive chemosensing and biological imaging. Chem. Soc. Rev. 43, 4563–4601 (2014).

    Article  CAS  PubMed  Google Scholar 

  126. Mochida, A., Ogata, F., Nagaya, T., Choyke, P. L. & Kobayashi, H. Activatable fluorescent probes in fluorescence-guided surgery: Practical considerations. Bioorg. Med. Chem. 26, 925–930 (2018).

    Article  CAS  PubMed  Google Scholar 

  127. Whitley, M. J. et al. A phase I study of the safety and activation of a cathepsin-activatable fluorescent cancer-specific probe LUM015. J. Clin. Oncol. 32, TPS11135 (2014).

    Article  Google Scholar 

  128. Whitley, M. J. et al. A mouse-human phase 1 co-clinical trial of a protease-activated fluorescent probe for imaging cancer. Sci. Transl. Med. 8, 320ra324 (2016).

    Article  CAS  Google Scholar 

  129. Smith, B. L. et al. Real-time, intraoperative detection of residual breast cancer in lumpectomy cavity walls using a novel cathepsin-activated fluorescent imaging system. Breast Cancer Res. Treat. 171, 413–420 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  130. Esfahani, S. A. et al. Optical imaging with a novel cathepsin-activatable probe for enhanced detection of colorectal cancer. Am. J. Nucl. Med. Mol. Imaging 9, 230–242 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Smith, B. L. et al. Feasibility study of a novel protease-activated fluorescent imaging system for real-time, intraoperative detection of residual breast cancer in breast conserving surgery. Ann. Surg. Oncol. 27, 1854–1861 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  132. Liu, Y. et al. Molecular imaging and validation of margins in surgically excised nonmelanoma skin cancer specimens. J. Med. Imaging 6, 016001 (2019).

    Article  Google Scholar 

  133. Unkart, J. T. et al. Intraoperative tumor detection using a ratiometric activatable fluorescent peptide: a first-in-human phase 1 study. Ann. Surg. Oncol. 24, 3167–3173 (2017).

    Article  PubMed  Google Scholar 

  134. Kato, D. et al. Activity-based probes that target diverse cysteine protease families. Nat. Chem. Biol. 1, 33–38 (2005).

    Article  CAS  PubMed  Google Scholar 

  135. Blum, G., von Degenfeld, G., Merchant, M. J., Blau, H. M. & Bogyo, M. Noninvasive optical imaging of cysteine protease activity using fluorescently quenched activity-based probes. Nat. Chem. Biol. 3, 668–677 (2007).

    Article  CAS  PubMed  Google Scholar 

  136. Sexton, K. B., Witte, M. D., Blum, G. & Bogyo, M. Design of cell-permeable, fluorescent activity-based probes for the lysosomal cysteine protease asparaginyl endopeptidase (AEP)/legumain. Bioorg. Med. Chem. Lett. 17, 649–653 (2007).

    Article  CAS  PubMed  Google Scholar 

  137. Ofori, L. O. et al. Design of protease activated optical contrast agents that exploit a latent lysosomotropic effect for use in fluorescence-guided surgery. ACS Chem. Biol. 10, 1977–1988 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Yim, J. J., Tholen, M., Klaassen, A., Sorger, J. & Bogyo, M. Optimization of a protease activated probe for optical surgical navigation. Mol. Pharm. 15, 750–758 (2018).

    Article  CAS  PubMed  Google Scholar 

  139. Shrivastav, M. et al. Validation of near infrared fluorescence (NIRF) probes in vivo with dual laser NIRF endoscope. PLoS One 13, e0206568 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  140. Miampamba, M. et al. Sensitive in vivo visualization of breast cancer using ratiometric protease-activatable fluorescent imaging agent, AVB-620. Theranostics 7, 3369–3386 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Walker, E. et al. A protease-activated fluorescent probe allows rapid visualization of keratinocyte carcinoma during excision. Cancer Res. 80, 2045–2055 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Withana, N. P. et al. Labeling of active proteases in fresh-frozen tissues by topical application of quenched activity-based probes. Nat. Protoc. 11, 184–191 (2016).

    Article  CAS  PubMed  Google Scholar 

  143. Chyan, W. & Raines, R. T. Enzyme-activated fluorogenic probes for live-cell and in vivo imaging. ACS Chem. Biol. 13, 1810–1823 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Onoyama, H. et al. Rapid and sensitive detection of early esophageal squamous cell carcinoma with fluorescence probe targeting dipeptidylpeptidase IV. Sci. Rep. 6, 26399 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Mizushima, T. et al. Rapid detection of superficial head and neck squamous cell carcinoma by topically spraying fluorescent probe targeting dipeptidyl peptidase-IV. Head Neck 40, 1466–1475 (2018).

    Article  PubMed  Google Scholar 

  146. Yamamoto, K. et al. Detection of early adenocarcinoma of the esophagogastric junction by spraying an enzyme-activatable fluorescent probe targeting dipeptidyl peptidase-IV. BMC Cancer 20, 64–69 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Kuriki, Y. et al. Establishment of molecular design strategy to obtain activatable fluorescent probes for carboxypeptidases. J. Am. Chem. Soc. 140, 1767–1773 (2018).

    Article  CAS  PubMed  Google Scholar 

  148. Lozano-Torres, B. et al. An OFF–ON two-photon fluorescent probe for tracking cell senescence in vivo. J. Am. Chem. Soc. 139, 8808–8811 (2017).

    Article  CAS  PubMed  Google Scholar 

  149. Li, H. et al. Aminopeptidase N activatable fluorescent probe for tracking metastatic cancer and image-guided surgery via in situ spraying. J. Am. Chem. Soc. 142, 6381–6389 (2020).

    Article  CAS  PubMed  Google Scholar 

  150. Xu, K. et al. High selectivity imaging of nitroreductase using a near-infrared fluorescence probe in hypoxic tumor. Chem. Commun. 49, 2554–2556 (2013).

    Article  CAS  Google Scholar 

  151. Li, Y. et al. Ultrasensitive near-infrared fluorescence-enhanced probe for in vivo nitroreductase imaging. J. Am. Chem. Soc. 137, 6407–6416 (2015).

    Article  CAS  PubMed  Google Scholar 

  152. Jiao, J. et al. Quicker, deeper and stronger imaging: a review of tumor-targeted, near-infrared fluorescent dyes for fluorescence guided surgery in the preclinical and clinical stages. Eur. J. Pharm. Biopharm. 152, 123–143 (2020).

    Article  CAS  PubMed  Google Scholar 

  153. Singh, H. et al. A pH-responsive glycyrrhetinic-acid-modified small-molecule conjugate for NIR imaging of hepatocellular carcinoma (HCC). ChemBioChem 20, 614–620 (2018).

    Article  CAS  Google Scholar 

  154. Gioux, S., Choi, H. S. & Frangioni, J. V. Image-guided surgery using invisible near-infrared light: fundamentals of clinical translation. Mol. Imaging 9, 237–255 (2010).

    Article  CAS  PubMed  Google Scholar 

  155. Av, D. S., Lin, H., Henderson, E. R., Samkoe, K. S. & Pogue, B. W. Review of fluorescence guided surgery systems: identification of key performance capabilities beyond indocyanine green imaging. J. Biomed. Opt. 21, 80901 (2016).

    Article  Google Scholar 

  156. Gioux, S. et al. FluoSTIC: miniaturized fluorescence image-guided surgery system. J. Biomed. Opt. 17, 106014 (2012).

    Article  PubMed  Google Scholar 

  157. Sajedi, S., Sabet, H. & Choi, H. S. Intraoperative biophotonic imaging systems for image-guided interventions. Nanophotonics 8, 99–116 (2019).

    Article  PubMed  Google Scholar 

  158. Matz, G. et al. Chip-on-the-tip compact flexible endoscopic epifluorescence video-microscope for in-vivo imaging in medicine and biomedical research. Biomed. Opt. Express 8, 3329–3342 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Tummers, W. S. et al. Recommendations for reporting on emerging optical imaging agents to promote clinical approval. Theranostics 8, 5336–5347 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Singh, G., Gott, M. D., Pietzsch, H.-J. & Stephan, H. Nuclear and optical dual-labelled imaging agents. Design and challenges. Nuklearmedizin. Nucl. Med. 55, 41–50 (2016).

    Article  CAS  Google Scholar 

  161. Sibinga Mulder, B. G. et al. A dual-labeled cRGD-based PET/optical tracer for pre-operative staging and intraoperative treatment of colorectal cancer. Am. J. Nucl. Med. Mol. Imaging 8, 282–291 (2018).

    PubMed  PubMed Central  Google Scholar 

  162. Pogue, B. W. et al. Fluorescence-guided surgery and intervention — an AAPM emerging technology blue paper. Med. Phys. 45, 2681–2688 (2018).

    Article  PubMed  Google Scholar 

  163. Valdes, P. A., Angelo, J. P., Choi, H. S. & Gioux, S. qF-SSOP: real-time optical property corrected fluorescence imaging. Biomed. Opt. Express 8, 3597–3605 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. McCulloch, P. et al. No surgical innovation without evaluation: the IDEAL recommendations. Lancet 374, 1105–1112 (2009).

    Article  PubMed  Google Scholar 

  165. van Manen, L. et al. A practical guide for the use of indocyanine green and methylene blue in fluorescence-guided abdominal surgery. J. Surg. Oncol. 118, 283–300 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  166. Armstrong, G. et al. IntAct: intra-operative fluorescence angiography to prevent anastomotic leak in rectal cancer surgery: a randomized controlled trial. Colorectal Dis. 20, O226–O234 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Jafari, M. D. et al. The use of indocyanine green fluorescence to assess anastomotic perfusion during robotic assisted laparoscopic rectal surgery. Surg. Endosc. 27, 3003–3008 (2013).

    Article  PubMed  Google Scholar 

  168. Varela, R. et al. Outcomes of DIEP flap and fluorescent angiography: a randomized controlled clinical trial. Plast. Reconstr. Surg. 145, 1–10 (2020).

    Article  CAS  PubMed  Google Scholar 

  169. Mascagni, P. et al. New intraoperative imaging technologies: innovating the surgeon’s eye toward surgical precision. J. Surg. Oncol. 118, 265–282 (2018).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

The authors would like to thank Dr J.H. van Dierendonck for his artistic contribution to this article. The authors received support from the Dutch Cancer Society (KWF; young investigator grant 11289 to R.J.S.), the Dutch Organization for Scientific Research (NWO; Veni grant 016.196.059 to J.S.D.M.) and the European Commission H2020-MSCA-ITN-2019 (project number: 857894 – CAST to A.L.V.).

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to each stage of preparation of this manuscript for publication.

Corresponding author

Correspondence to Alexander L. Vahrmeijer.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Dedication

Many of the topics discussed in this Review were studied and described by Dr Sanjiv S. Gambhir, one of the founders of molecular imaging, who unfortunately passed away during the preparation of this manuscript in July 2020. We thank him for his exceptional mentorship and forward-looking vision.

Peer review information

Nature Reviews Clinical Oncology thanks M. Bouvet, J. Warram and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

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

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mieog, J.S.D., Achterberg, F.B., Zlitni, A. et al. Fundamentals and developments in fluorescence-guided cancer surgery. Nat Rev Clin Oncol 19, 9–22 (2022). https://doi.org/10.1038/s41571-021-00548-3

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41571-021-00548-3

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer