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.

  • Opinion
  • Published:

Fluorescence-guided surgery with live molecular navigation — a new cutting edge

Abstract

A glowing new era in cancer surgery may be dawning. Using fluorescently labelled markers, surgical molecular navigation means that tumours and nerves can be displayed in real time intra-operatively in contrasting pseudocolours, which allows more complete tumour resection while preserving important structures. These advances can potentially cause a paradigm shift in cancer surgery, improving patient outcome and decreasing overall health-care costs.

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

Figure 1: Live molecular detection of surgical margins.
Figure 2: Live molecular detection of nerves.

Similar content being viewed by others

References

  1. National Cancer Institute. Previous version: SEER cancer statistics review, 1975–2005 [online], (2008).

  2. Haque, R., Contreras, R., McNicoll, M. P., Eckberg, E. C. & Petitti, D. B. Surgical margins and survival after head and neck cancer surgery. BMC Ear Nose Throat Disord. 16, 2 (2006).

    Article  Google Scholar 

  3. Singletary, S. Surgical margins in patients with early-stage breast cancer treated with breast conservation therapy. Am. J. Surg. 184, 383–393 (2002).

    Article  PubMed  Google Scholar 

  4. Meric, F. et al. Positive surgical margins and ipsilateral breast tumor recurrence predict disease-specific survival after breast-conserving therapy. Cancer 97, 926–933 (2003).

    Article  PubMed  Google Scholar 

  5. Snijder, R., de la Riviere, A., Elbers, H. & van den Bosch, J. Survival in resected stage I lung cancer with residual tumor at the bronchial resection margin. Ann. Thorac. Surg. 65, 212–216 (1998).

    Article  CAS  PubMed  Google Scholar 

  6. Nagtegaal, I. D. & Quirke, P. What is the role for the circumferential margin in the modern treatment of rectal cancer? J. Clin. Oncol. 26, 303–312 (2008).

    Article  PubMed  Google Scholar 

  7. Dotan, Z. et al. Positive surgical margins in soft tissue following radical cystectomy for bladder cancer and cancer specific survival. J. Urol. 178, 2308–2312 (2007).

    Article  PubMed  Google Scholar 

  8. Wieder, J. A. & Soloway, M. S. Incidence, etiology, location, prevention and treatment of positive surgical margins after radical prostatectomy for prostate cancer. J. Urol. 160, 299–315 (1998).

    Article  CAS  PubMed  Google Scholar 

  9. Kumar, A., Puri, R., Gadgil, P. V. & Jatoi, I. Sentinel lymph node biopsy in primary breast cancer: window to management of the axilla. World J. Surg. 36, 1453–1459 (2012).

    Article  PubMed  Google Scholar 

  10. Wong, S. L. et al. Sentinel lymph node biopsy for melanoma: American Society of Clinical Oncology and Society of Surgical Oncology joint clinical practice guideline. J. Clin. Oncol. 30, 2912–2918 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Hutteman, M. et al. Randomized, double-blind comparison of indocyanine green with or without albumin premixing for near-infrared fluorescence imaging of sentinel lymph nodes in breast cancer patients. Breast Cancer Res. Treat. 127, 163–170 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. van der Vorst, J. R. et al. Randomized comparison of near-infrared fluorescence imaging using indocyanine green and 99m technetium with or without patent blue for the sentinel lymph node procedure in breast cancer patients. Ann. Surg. Oncol. 19, 4104–4111 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Emerson, D. K. et al. A receptor-targeted fluorescent radiopharmaceutical for multireporter sentinel lymph node imaging. Radiology 265, 186–193 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Joseph, M., Phillips, M., Farrell, T. M. & Rupp, C. C. Can residents safely and efficiently be taught single incision laparoscopic cholecystectomy? J. Surg. Educ. 69, 468–472 (2012).

    Article  PubMed  Google Scholar 

  15. Kandil, E. H., Noureldine, S. I., Yao, L. & Slakey, D. P. Robotic transaxillary thyroidectomy: an examination of the first one hundred cases. J. Am. Coll. Surg. 214, 558–564 (2012).

    Article  PubMed  Google Scholar 

  16. Lee, J. C., Jang, H. D. & Shin, B. J. Learning curve and clinical outcomes of minimally invasive transforaminal lumbar interbody fusion: our experience in 86 consecutive cases. Spine 37, 1548–1557 (2012).

    Article  PubMed  Google Scholar 

  17. Pappo, I. et al. Diagnostic performance of a novel device for real-time margin assessment in lumpectomy specimens. J. Surg. Res. 160, 277–281 (2010).

    Article  PubMed  Google Scholar 

  18. Rivera, R. J., Holmes, D. R. & Tafra, L. Analysis of the impact of intraoperative margin assessment with adjunctive use of MarginProbe versus standard of care on tissue volume removed. Int. J. Surg. Oncol. 2012, 868623 (2012).

    PubMed  PubMed Central  Google Scholar 

  19. Dotan, Z. A. et al. Detection of prostate cancer by radio-frequency near-field spectroscopy in radical prostatectomy ex vivo specimens. Prostate Cancer Prostatic Dis. 16, 73–78 (2012).

    Article  PubMed  Google Scholar 

  20. Pavlova, I. et al. Multiphoton microscopy and microspectroscopy for diagnostics of inflammatory and neoplastic lung. J. Biomed. Opt. 17, 036014 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Pierce, M. C. et al. Accuracy of in vivo multimodal optical imaging for detection of oral neoplasia. Cancer Prev. Res. 5, 801–809 (2012).

    Article  Google Scholar 

  22. Gurtner, G. C. et al. Intraoperative laser angiography using the SPY system: review of the literature and recommendations for use. Ann. Surg. Innov. Res. 7, 1 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Kuroda, K. et al. Intra-arterial injection fluorescein videoangiography in aneurysm surgery. Neurosurgery 72, 141–150 (2012).

    Google Scholar 

  24. Shin, D., Vigneswaran, N., Gillenwater, A. & Richards-Kortum, R. Advances in fluorescence imaging techniques to detect oral cancer and its precursors. Future Oncol. 6, 1143–1154 (2010).

    Article  PubMed  Google Scholar 

  25. Foersch, S. et al. Confocal laser endomicroscopy for diagnosis and histomorphologic imaging of brain tumors in vivo. PLoS ONE 7, e41760 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Vila, P. M. et al. Discrimination of benign and neoplastic mucosa with a High-Resolution Microendoscope (HRME) in head and neck cancer. Ann. Surg. Oncol. 19, 3534–3539 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Barker, F. G. et al. Survival and functional status after resection of recurrent glioblastoma multiforme. Neurosurgery 42, 709–720 (1998).

    Article  PubMed  Google Scholar 

  28. Swindle, P. et al. Do margins matter? The prognostic significance of positive surgical margins in radical prostatectomy specimens. J. Urol. 179, S47–S51 (2008).

    Article  PubMed  Google Scholar 

  29. Warner, J. N. et al. Impact of margin status at 37 months after robot assisted radical prostatectomy. Can. J. Urol. 18, 6043–6049 (2011).

    PubMed  Google Scholar 

  30. Chalfin, H. J. et al. Impact of surgical margin status on prostate-cancer-specific mortality. BJU Int. 110, 1684–1689 (2012).

    Article  PubMed  Google Scholar 

  31. Pleijhuis, R. G. et al. Obtaining adequate surgical margins in breast-conserving therapy for patients with early-stage breast cancer: current modalities and future directions. Ann. Surg. Oncol. 16, 2717–2730 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Atkins, J. et al. Positive margin rates following breast-conserving surgery for stage I-III breast cancer: palpable versus nonpalpable tumors. J. Surg. Res. 177, 109–115 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Cutter, J. L. et al. Topical application of activity-based probes for visualization of brain tumor tissue. PLoS ONE 7, e33060 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Verdoes, M. et al. A nonpeptidic cathepsin S activity-based probe for noninvasive optical imaging of tumor-associated macrophages. Chem. Biol. 19, 619–628 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Sheth, R. A. et al. Improved detection of ovarian cancer metastases by intraoperative quantitative fluorescence protease imaging in a pre-clinical model. Gynecol. Oncol. 112, 616–622 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Nguyen, Q. T. et al. Surgery with molecular fluorescence imaging using activatable cell-penetrating peptides decreases residual cancer and improves survival. Proc. Natl Acad. Sci. USA 107, 4317–4322 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Jiang, T. et al. Tumor imaging by means of proteolytic activation of cell-penetrating peptides. Proc. Natl Acad. Sci. USA 101, 17867–17872 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Olson, E. S. et al. In vivo characterization of activatable cell penetrating peptides for targeting protease activity in cancer. Integr. Biol. 1, 382–393 (2009).

    Article  CAS  Google Scholar 

  39. Aguilera, T. A., Olson, E. S., Timmers, M. M., Jiang, T. & Tsien, R. Y. Systemic in vivo distribution of activatable cell penetrating peptides is superior to that of cell penetrating peptides. Integr. Biol. 1, 371–381 (2009).

    Article  CAS  Google Scholar 

  40. Savariar, E. N. et al. Real-time in vivo molecular detection of primary tumors and metastases with ratiometric activatable cell-penetrating peptides. Cancer Res. 73, 855–864 (2013).

    Article  CAS  PubMed  Google Scholar 

  41. Nakajima, T. et al. Targeted, activatable, in vivo fluorescence imaging of prostate-specific membrane antigen (PSMA) positive tumors using the quenched humanized J591 antibody-indocyanine green (ICG) conjugate. Bioconjug. Chem. 22, 1700–1705 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Tran Cao, H. S. et al. Tumor-specific fluorescence antibody imaging enables accurate staging laparoscopy in an orthotopic model of pancreatic cancer. Hepatogastroenterology 59, 1994–1999 (2012).

    PubMed  Google Scholar 

  43. McElroy, M. et al. Imaging of primary and metastatic pancreatic cancer using a fluorophore-conjugated anti-CA19-9 antibody for surgical navigation. World J. Surg. 32, 1057–1066 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Hall, M. A. et al. Comparison of mAbs targeting epithelial cell adhesion molecule for the detection of prostate cancer lymph node metastases with multimodal contrast agents: quantitative small-animal PET/CT and NIRF. J. Nucl. Med. 53, 1427–1437 (2012).

    Article  CAS  PubMed  Google Scholar 

  45. Rosbach, K. J., Williams, M. D., Gillenwater, A. M. & Richards-Kortum, R. R. Optical molecular imaging of multiple biomarkers of epithelial neoplasia: epidermal growth factor receptor expression and metabolic activity in oral mucosa. Transl Oncol. 5, 160–171 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Urano, Y. et al. Rapid cancer detection by topically spraying a γ-glutamyltranspeptidase-activated fluorescent probe. Sci. Transl Med. 3, 110ra119 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  48. 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 

  49. Stummer, W. et al. Fluorescence-guided resection of glioblastoma multiforme by using 5-aminolevulinic acid-induced porphyrins: a prospective study in 52 consecutive patients. J. Neurosurg. 93, 1003–1013 (2000).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Schafer, C. et al. γ-glutamyl transferase expression in higher-grade astrocytic glioma. Acta Oncol. 40, 529–535 (2001).

    Article  CAS  PubMed  Google Scholar 

  52. Floeth, F. W. et al. Comparison of 18F-FET PET and 5-ALA fluorescence in cerebral gliomas. Eur. J. Nucl. Med. Mol. Imaging 38, 731–741 (2011).

    Article  CAS  PubMed  Google Scholar 

  53. Sun, C. et al. Tumor-targeted drug delivery and MRI contrast enhancement by chlorotoxin-conjugated iron oxide nanoparticles. Nanomed. 3, 495–505 (2008).

    Article  CAS  Google Scholar 

  54. Sun, C. et al. In vivo MRI detection of gliomas by chlorotoxin-conjugated superparamagnetic nanoprobes. Small 4, 372–379 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Olson, E. S. et al. Activatable cell penetrating peptides linked to nanoparticles as dual probes for in vivo fluorescence and MR imaging of proteases. Proc. Natl Acad. Sci. USA 107, 4311–4316 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Gibbs-Strauss, S. L. et al. Nerve-highlighting fluorescent contrast agents for image-guided surgery. Mol. Imaging 10, 91–101 (2011).

    Article  CAS  PubMed  Google Scholar 

  57. Whitney, M. et al. Fluorescent peptides highlight peripheral nerves during surgery in mice. Nature Biotech. 29, 352–356 (2011).

    Article  CAS  Google Scholar 

  58. Sturm, M. B. et al. Targeted imaging of esophageal neoplasia with a fluorescently labeled peptide: first-in-human results. Sci. Transl Med. 5, 184ra61 (2013).

    Article  PubMed  Google Scholar 

  59. 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. Nature Chem. Biol. 3, 668–677 (2007).

    Article  CAS  Google Scholar 

  60. Figueiredo, J. L., Alencar, H., Weissleder, R. & Mahmood, U. Near infrared thoracoscopy of tumoral protease activity for improved detection of peripheral lung cancer. Int. J. Cancer 118, 2672–2677 (2006).

    Article  CAS  PubMed  Google Scholar 

  61. Kelly, K., Alencar, H., Funovics, M., Mahmood, U. & Weissleder, R. Detection of invasive colon cancer using a novel, targeted, library-derived fluorescent peptide. Cancer Res. 64, 6247–6251 (2004).

    Article  CAS  PubMed  Google Scholar 

  62. Veiseh, M. et al. Tumor paint: a chlorotoxin:Cy5.5 bioconjugate for intraoperative visualization of cancer foci. Cancer Res. 67, 6882–6888 (2007).

    Article  CAS  PubMed  Google Scholar 

  63. Stroud, M. R., Hansen, S. J. & Olson, J. M. In vivo bio-imaging using chlorotoxin-based conjugates. Curr. Pharm. Des. 17, 4362–4371 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Ye, Y. et al. Near-infrared fluorescent divalent RGD ligand for integrin αvβ3-targeted optical imaging. Bioorg. Med. Chem. Lett. 22, 5405–5409 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Gao, J. et al. A novel clinically translatable fluorescent nanoparticle for targeted molecular imaging of tumors in living subjects. Nano Lett. 12, 281–286 (2012).

    Article  PubMed  Google Scholar 

  66. Zhu, L. et al. Dual-functional, receptor-targeted fluorogenic probe for in vivo imaging of extracellular protease expressions. Bioconjugate Chem. 22, 1001–1005 (2011).

    Article  CAS  Google Scholar 

  67. Wu, A. P. et al. Improved facial nerve identification with novel fluorescently labeled probe. Laryngoscope 121, 805–810 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  68. Cotero, V. E. et al. Intraoperative fluorescence imaging of peripheral and central nerves through a myelin-selective contrast agent. Mol. Imaging Biol. 14, 708–717 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  69. Tsien, R. Y. & Harootunian, A. T. Practical design criteria for a dynamic ratio imaging system. Cell Calcium 11, 93–109 (1990).

    Article  CAS  PubMed  Google Scholar 

  70. Orosco, R. K., Tsien, R. Y. & Nguyen, Q. T. Fluorescence imaging in surgery. IEEE Rev. Biomed. Eng. 6, 178–187 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  71. Taruttis, A. & Ntziachristos, V. Translational optical imaging. Am. J. Roentgenol. 199, 263–271 (2012).

    Article  Google Scholar 

  72. Nunn, A. D. The cost of developing imaging agents for routine clinical use. Invest. Radiol. 41, 206–212 (2006).

    Article  PubMed  Google Scholar 

  73. Mitsunaga, M. et al. Fluorescence endoscopic detection of murine colitis-associated colon cancer by topically applied enzymatically rapid-activatable probe. Gut 62, 1179–1186 (2013).

    Article  CAS  PubMed  Google Scholar 

  74. Ashitate, Y. et al. Simultaneous assessment of luminal integrity and vascular perfusion of the gastrointestinal tract using dual-channel near-infrared fluorescence. Mol. Imaging 11, 301–308 (2012).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors would like to thank S. Howell and L. Simon for helpful discussions on the regulatory aspects discussed in this manuscript, and M. Whitney for helpful scientific discussions.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Quyen T. Nguyen or Roger Y. Tsien.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

PowerPoint slides

Glossary

Approved

An approved drug is a drug that has been authorized for a therapeutic use by a ruling authority. In Europe, the European Medicines Agency (EMA) is the ruling authority. In the United States, the Food and Drug Administration (FDA), an agency within the Department of Health and Human Services, provides this approval. A list of FDA-approved drugs is updated regularly on the FDA Orange Book website (see Further information).

Autofluorescence

Fluorescence that is inherent to different tissues (such as muscle, fat or liver) in the absence of any exogenously applied compounds.

Chlorotoxin

A peptide that is found in the venom of the scorpion Leiurus quinquestriatus that blocks small-conductance chloride channels. Chlorotoxin binds preferentially to gliomas and tumours of neuroectodermal origin.

Indication

A drug is approved by the US Food and Drug Administration (FDA) or the European Medicines Agency (EMA) for certain uses or indications following approved Phase I, II and III clinical trials. These indications are supplied with the package inserts for the drugs.

Infrared reflectivity

The fraction of incident infrared light that is reflected, which can vary between different tissues.

Intratumour microheterogeneity

The idea that within a given neoplasm, different tumour cells may display differences in genetic mutations and/or protein expression that may confer advantages and disadvantages for metastatic potential or response to chemotherapy or radiotherapy.

Lymphoedema

A condition of localized fluid retention and tissue swelling caused by a compromised lymphatic system. Lymphoedema is a common consequence of lymph node removal (for example, during neck dissection or axillary lymph node dissection) in the surgical treatment of cancer.

Medical device

An instrument that is used to diagnose or treat disease or other medical conditions and that does not achieve its purpose through chemical action within or on the body. In the United States, the Food and Drug Administration (FDA) is responsible for medical device evaluation, through premarket evaluation (510(k)) to show similarity with a previously approved device or through premarket approval (PMA) applications. PMA approval for a new device is based on a determination by the FDA that the PMA contains sufficient valid scientific evidence to ensure that the device is safe and effective for its intended use or uses.

Micro-anatomical cyto-architecture

The shapes and sizes of cells, nuclei, organelles and extracellular structures, which are used to distinguish tumour from normal tissue at microscopic resolution.

Nonspecific hydrophobic stickiness

Nonspecific binding to tissue of hydrophobic molecules such as most near-infrared dyes (which emit at wavelengths >700nm), thus typically resulting in increased background fluorescence.

Off-label

The clinical practice of prescribing pharmaceuticals for an unapproved indication or in an unapproved age group, at an unapproved dose or through an unapproved form of administration. After a drug has been approved for one purpose, licensed physicians are able to prescribe it for other purposes that in their professional judgment are both safe and effective, and are not limited to the official, approved indications.

Open-field surgery

A type of surgery in which the surgeon's view of the patient's tissue is through direct visualization through a large skin incision. This is in contrast to minimally invasive surgery (that is, laparoscopic surgery, endoscopic surgery or robotic surgery) in which the surgeon views the patient's tissues through fibre-optic probes or cameras, which are inserted through natural orifices or small skin incisions and the images are displayed on a computer screen.

Quantum dots

Inorganic nanocrystals that typically have a diameter of 2–8 nm. Most are composed of an inner semiconductor core of CdSe, an outer shell of ZnS, and an organic coating to make the quantum dot biocompatible. Quantum dots can be engineered to emit fluorescent light in the ultraviolet to infrared spectrum by varying their size.

Raman scattering

A spectroscopic technique that differentiates different tissue types depending on the infrared spectra of different molecules (such as fat and collagen) within that tissue.

Ratiometric imaging

Measurement of the ratio of fluorescence intensities at two wavelengths. When the response of the fluorescent probe to the desired signal (for example, protease activity) is markedly different at two wavelengths, calculating the ratio of the fluorescence at those wavelengths isolates the desired signal from confounding factors such as the absolute concentration of the fluorescent probe, the thickness of tissue, the intensity of illumination and the sensitivity of detection, which all affect intensities at both wavelengths by equal proportions. Ratiometric activatable cell-penetrating peptides (RACPPs)40 use pairs of dyes such as Cy5 and Cy7 as donor and acceptor dyes for fluorescence resonance energy transfer, thus enabling emission ratiometric discrimination of protease activity.

Sentinel lymph node

(SLN). The first draining lymph node for a given anatomical site. For breast cancer, the SLNs are located in the axilla.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Nguyen, Q., Tsien, R. Fluorescence-guided surgery with live molecular navigation — a new cutting edge. Nat Rev Cancer 13, 653–662 (2013). https://doi.org/10.1038/nrc3566

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrc3566

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