1932

Abstract

Surgical success depends on the accuracy with which disease and vital tissue can be intraoperatively detected. However, the dominant visualization approach, i.e., human vision, does not see under the tissue surface and operates on low contrast between sites of disease, such as cancer, and the surrounding tissue. Intraoperative fluorescence imaging is emerging as a highly effective method to improve surgical vision and offers the potential to be intergrated seamlessly into the normal workflow of the operating room without causing disruption or undue delay. We review and compare two critical fluorescence imaging directions: one that uses nonspecific fluorescence dyes, addressing tissue perfusion and viability, and one that uses targeted agents, interrogating pathophysiological features of disease. These two approaches present detection sensitivity challenges that may differ by orders of magnitude and require different detection strategies. Nevertheless, fluorescence imaging provides the surgeon with previously unavailable real-time feedback that improves surgical precision and can become essential for interventional decision-making.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-med-051914-022043
2016-01-14
2024-04-18
Loading full text...

Full text loading...

/deliver/fulltext/med/67/1/annurev-med-051914-022043.html?itemId=/content/journals/10.1146/annurev-med-051914-022043&mimeType=html&fmt=ahah

Literature Cited

  1. Roberts DW, Hartov A, Kennedy FE. 1.  et al. 1998. Intraoperative brain shift and deformation: a quantitative analysis of cortical displacement in 28 cases. Neurosurgery 43:749–58 [Google Scholar]
  2. Labadie RF, Davis BM, Fitzpatrick JM. 2.  2005. Image-guided surgery: What is the accuracy?. Curr. Opin. Otolaryngol. Head Neck Surg. 13:27–31 [Google Scholar]
  3. Zanzonico P, Heller S. 3.  2000. The intraoperative gamma probe: basic principles and choices available. Proc. Semin. Nucl. Med. 30:33–48 [Google Scholar]
  4. Galloro G. 4.  2012. High technology imaging in digestive endoscopy. World J. Gastrointest. Endosc. 4:22–27 [Google Scholar]
  5. Freschi C, Ferrari V, Melfi F. 5.  et al. 2013. Technical review of the da Vinci surgical telemanipulator. Int. J. Med. Robot. Comput. Assist. Surg. 9:396–406 [Google Scholar]
  6. Moore G, Hunter S, Hubbard T. 6.  1949. Clinical and experimental studies of fluorescein dyes with special reference to their use for the diagnosis of central nervous system tumors. Ann. Surg. 130:637–42 [Google Scholar]
  7. Novotny H, Alvis D. 7.  1960. A method of photographing fluorescence in circulating blood of the human eye. Tech. Doc. Rep. SAM-TDR. USAF Sch. Aerosp. Med. 60:1–4 [Google Scholar]
  8. Leevy CM, Smith F, Longueville J. 8.  et al. 1967. Indocyanine green clearance as a test for hepatic function: evaluation by dichromatic ear densitometry. JAMA 200:236–40 [Google Scholar]
  9. Rowell LB, Marx HJ, Bruce RA. 9.  et al. 1966. Reductions in cardiac output, central blood volume, and stroke volume with thermal stress in normal men during exercise. J. Clin. Investig. 45:1801–16 [Google Scholar]
  10. Craandijk A, Van Beek C. 10.  1976. Indocyanine green fluorescence angiography of the choroid. Br. J. Ophthalmol. 60:377–85 [Google Scholar]
  11. Schaafsma BE, Mieog JSD, Hutteman M. 11.  et al. 2011. The clinical use of indocyanine green as a near-infrared fluorescent contrast agent for image-guided oncologic surgery. J. Surg. Oncol. 104:323–32 [Google Scholar]
  12. Polom K, Murawa D, Rho YS. 12.  et al. 2011. Current trends and emerging future of indocyanine green usage in surgery and oncology. Cancer 117:4812–22 [Google Scholar]
  13. Sekijima M, Tojimbara T, Sato S. 13.  et al. 2004. An intraoperative fluorescent imaging system in organ transplantation. Transplant. Proc. 36:2188–90 [Google Scholar]
  14. Holm C, Mayr M, Höfter E. 14.  et al. 2002. Intraoperative evaluation of skin-flap viability using laser-induced fluorescence of indocyanine green. Br. J. Plastic Surg. 55:635–44 [Google Scholar]
  15. Detter C, Russ D, Iffland A. 15.  et al. 2002. Near-infrared fluorescence coronary angiography: a new noninvasive technology for intraoperative graft patency control. Proc. Heart Surg. Forum 5:364–69 [Google Scholar]
  16. Ferroli P, Nakaji P, Acerbi F. 16.  et al. 2011. Indocyanine green (ICG) temporary clipping test to assess collateral circulation before venous sacrifice. World Neurosurg. 75:122–25 [Google Scholar]
  17. Allison AS, Bloor C, Faux W. 17.  et al. 2010. The angiographic anatomy of the small arteries and their collaterals in colorectal resections: some insights into anastomotic perfusion. Ann. Surg. 251:1092–97 [Google Scholar]
  18. Sherwinter DA. 18.  2012. Transanal near-infrared imaging of colorectal anastomotic perfusion. Surg. Laparosc. Endosc. Percutan. Tech. 22:433–36 [Google Scholar]
  19. Jonsson K, Jensen JA, Goodson W 3rd. 19.  et al. 1991. Tissue oxygenation, anemia, and perfusion in relation to wound healing in surgical patients. Ann. Surg. 214:605–13 [Google Scholar]
  20. Yamaguchi S, De Lorenzi F, Petit JY. 20.  et al. 2004. The “perfusion map” of the unipedicled TRAM flap to reduce postoperative partial necrosis. Ann. Plastic Surg. 53:205–9 [Google Scholar]
  21. Azuma R, Morimoto Y, Masumoto K. 21.  et al. 2008. Detection of skin perforators by indocyanine green fluorescence nearly infrared angiography. Plastic Reconstr. Surg. 122:1062–67 [Google Scholar]
  22. Liu DZ, Mathes DW, Zenn MR, Neligan PC. 22.  2011. The application of indocyanine green fluorescence angiography in plastic surgery. J. Reconstr. Microsurg. 27:355–64 [Google Scholar]
  23. Lyman GH, Temin S, Edge SB. 23.  et al. 2014. Sentinel lymph node biopsy for patients with early-stage breast cancer: American Society of Clinical Oncology clinical practice guideline update. J. Clin. Oncol. 32:1365–83 [Google Scholar]
  24. Yared M, Middleton L, Smith T. 24.  et al. 2002. Recommendations for sentinel lymph node processing in breast cancer. Am. J. Surg. Pathol. 26:377–82 [Google Scholar]
  25. Cox CE, Pendas S, Cox JM. 25.  et al. 1998. Guidelines for sentinel node biopsy and lymphatic mapping of patients with breast cancer. Ann. Surg. 227:645–53 [Google Scholar]
  26. Mieog JSD, Troyan SL, Hutteman M. 26.  et al. 2011. Toward optimization of imaging system and lymphatic tracer for near-infrared fluorescent sentinel lymph node mapping in breast cancer. Ann. Surg. Oncol. 18:2483–91 [Google Scholar]
  27. Wishart G, Loh S-W, Jones L, Benson J. 27.  2012. A feasibility study (ICG-10) of indocyanine green (ICG) fluorescence mapping for sentinel lymph node detection in early breast cancer. Eur. J. Surg. Oncol. (EJSO) 38:651–56 [Google Scholar]
  28. Verbeek FP, Troyan SL, Mieog JSD. 28.  et al. 2014. Near-infrared fluorescence sentinel lymph node mapping in breast cancer: a multicenter experience. Breast Cancer Res. Treat. 143:333–42 [Google Scholar]
  29. Fujiwara M, Mizukami T, Suzuki A, Fukamizu H. 29.  2009. Sentinel lymph node detection in skin cancer patients using real-time fluorescence navigation with indocyanine green: preliminary experience. J. Plastic Reconstr. Aesthetic Surg. 62:e373–e78 [Google Scholar]
  30. van den Berg NS, Brouwer OR, Klop WMC. 30.  et al. 2012. Concomitant radio- and fluorescence-guided sentinel lymph node biopsy in squamous cell carcinoma of the oral cavity using ICG-99mTc-nanocolloid. Eur. J. Nucl. Med. Mol. Imaging 39:1128–36 [Google Scholar]
  31. Crane L, Themelis G, Arts H. 31.  et al. 2011. Intraoperative near-infrared fluorescence imaging for sentinel lymph node detection in vulvar cancer: first clinical results. Gynecol. Oncol. 120:291–95 [Google Scholar]
  32. Crane LM, Themelis G, Pleijhuis RG. 32.  et al. 2011. Intraoperative multispectral fluorescence imaging for the detection of the sentinel lymph node in cervical cancer: a novel concept. Mol. Imaging Biol. 13:1043–49 [Google Scholar]
  33. Misiek M, Rzepka JK, Zalewski K. 33.  et al. 2014. Fluorescence-guided lymph node mapping with indocyanine green for endometrial cancer: a feasibility study. Clin. J. Oncol. (Meet. Abstr.) 32:e16549 [Google Scholar]
  34. Yamashita S-I, Tokuishi K, Miyawaki M. 34.  et al. 2012. Sentinel node navigation surgery by thoracoscopic fluorescence imaging system and molecular examination in non-small cell lung cancer. Ann. Surg. Oncol. 19:728–33 [Google Scholar]
  35. Rasmussen JC, Tan I-C, Marshall MV. 35.  et al. 2009. Lymphatic imaging in humans with near-infrared fluorescence. Curr. Opin. Biotechnol. 20:74–82 [Google Scholar]
  36. Abu-Rustum NR. 36.  2014. Sentinel lymph node mapping for endometrial cancer: a modern approach to surgical staging. J. Natl. Compr. Cancer Netw. 12:288–97 [Google Scholar]
  37. Kim CH, Soslow RA, Park KJ. 37.  et al. 2013. Pathologic ultrastaging improves micrometastasis detection in sentinel lymph nodes during endometrial cancer staging. Int. J. Gynecol. Cancer 23:964–70 [Google Scholar]
  38. Marano A, Priora F, Lenti LM. 38.  et al. 2013. Application of fluorescence in robotic general surgery: review of the literature and state of the art. World J. Surg. 37:2800–11 [Google Scholar]
  39. Rossi EC, Ivanova A, Boggess JF. 39.  2012. Robotically assisted fluorescence-guided lymph node mapping with ICG for gynecologic malignancies: a feasibility study. Gynecol. Oncol. 124:78–82 [Google Scholar]
  40. Jafari MD, Lee KH, Halabi WJ. 40.  et al. 2013. The use of indocyanine green fluorescence to assess anastomotic perfusion during robotic assisted laparoscopic rectal surgery. Surg. Endosc. 27:3003–8 [Google Scholar]
  41. Daskalaki D, Fernandes E, Wang X. 41.  et al. 2014. Indocyanine green (ICG) fluorescent cholangiography during robotic cholecystectomy: results of 184 consecutive cases in a single institution. Surg. Innov. 21:615–21 [Google Scholar]
  42. Frangioni JV. 42.  2003. In vivo near-infrared fluorescence imaging. Curr. Opin. Chem. Biol. 7:626–34 [Google Scholar]
  43. van Oosten M, Crane LM, Bart J. 43.  et al. 2011. 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 [Google Scholar]
  44. Moran MS, Schnitt SJ, Giuliano AE. 44.  et al. 2013. Society of Surgical Oncology–American Society for Radiation Oncology consensus guideline on margins for breast-conserving surgery with whole-breast irradiation in stages I and II invasive breast cancer. Int. J. Radiat. Oncol. Biol. Phys. 88:553–64 [Google Scholar]
  45. Fang W, Chen W, Chen G, Jiang Y. 45.  2005. Surgical management of thymic epithelial tumors: a retrospective review of 204 cases. Ann. Thorac. Surg. 80:2002–7 [Google Scholar]
  46. Mendenhall WM, Zlotecki RA, Hochwald SN. 46.  et al. 2005. Retroperitoneal soft tissue sarcoma. Cancer 104:669–75 [Google Scholar]
  47. McCahill LE, Single RM, Aiello Bowles EJ. 47.  et al. 2012. Variability in reexcision following breast conservation surgery. JAMA 307:467–75 [Google Scholar]
  48. Tewari A, Sooriakumaran P, Bloch DA. 48.  et al. 2012. Positive surgical margin and perioperative complication rates of primary surgical treatments for prostate cancer: a systematic review and meta-analysis comparing retropubic, laparoscopic, and robotic prostatectomy. Eur. Urol. 62:1–15 [Google Scholar]
  49. Anderson C, Uman G, Pigazzi A. 49.  2008. Oncologic outcomes of laparoscopic surgery for rectal cancer: a systematic review and meta-analysis of the literature. Eur. J. Surg. Oncol. 34:1135–42 [Google Scholar]
  50. van Dam GM, Themelis G, Crane LM. 50.  et al. 2011. Intraoperative tumor-specific fluorescence imaging in ovarian cancer by folate receptor-α targeting: first in-human results. Nat. Med. 17:1315–19 [Google Scholar]
  51. Hsiung P-L, Hardy J, Friedland S. 51.  et al. 2008. Detection of colonic dysplasia in vivo using a targeted heptapeptide and confocal microendoscopy. Nat. Med. 14:454–58 [Google Scholar]
  52. Atreya R, Neumann H, Neufert C. 52.  et al. 2014. In vivo imaging using fluorescent antibodies to tumor necrosis factor predicts therapeutic response in Crohn's disease. Nat. Med. 20:313–18 [Google Scholar]
  53. Sturm MB, Joshi BP, Lu S. 53.  et al. 2013. Targeted imaging of esophageal neoplasia with a fluorescently labeled peptide: first-in-human results. Sci. Transl. Med. 5:ra61 [Google Scholar]
  54. Scheuer W, van Dam GM, Dobosz M. 54.  et al. 2012. Drug-based optical agents: infiltrating clinics at lower risk. Sci. Transl. Med. 4:134ps11 [Google Scholar]
  55. Rosenthal EL, Warram JM, de Boer E. 55.  et al. 2015. Safety and tumor-specificity of cetuximab-IRDye800 for surgical navigation in head and neck cancer. Clin. Cancer Res. 213658–66
  56. Stummer W, Novotny A, Stepp H. 56.  et al. 2000. Fluorescence-guided resection of glioblastoma multiforme utilizing 5-ALA-induced porphyrins: a prospective study in 52 consecutive patients. J. Neurosurg. 93:1003–13 [Google Scholar]
  57. Stummer W, Pichlmeier U, Meinel T. 57.  et al. 2006. Fluorescence-guided surgery with 5-aminolevulinic acid for resection of malignant glioma: a randomised controlled multicentre phase III trial. Lancet Oncol. 7:392–401 [Google Scholar]
  58. Kamp MA, Grosser P, Felsberg J. 58.  et al. 2012. 5-aminolevulinic acid (5-ALA)-induced fluorescence in intracerebral metastases: a retrospective study. Acta Neurochir. 154:223–28 [Google Scholar]
  59. Kishi K, Fujiwara Y, Yano M. 59.  et al. 2012. Staging laparoscopy using ALA-mediated photodynamic diagnosis improves the detection of peritoneal metastases in advanced gastric cancer. J. Surg. Oncol. 106:294–98 [Google Scholar]
  60. Harada K, Harada Y, Beika M. 60.  et al. 2013. Detection of lymph node metastases in human colorectal cancer by using 5-aminolevulinic acid-induced protoporphyrin IX fluorescence with spectral unmixing. Int. J. Mol. Sci. 14:23140–52 [Google Scholar]
  61. Riegger C, Herrmann J, Nagarajah J. 61.  et al. 2012. Whole-body FDG PET/CT is more accurate than conventional imaging for staging primary breast cancer patients. Eur. J. Nucl. Med. Mol. Imaging 39:852–63 [Google Scholar]
  62. Poulsen MH, Bouchelouche K, Høilund-Carlsen PF. 62.  et al. 2012. [18F] fluoromethylcholine (FCH) positron emission tomography/computed tomography (PET/CT) for lymph node staging of prostate cancer: a prospective study of 210 patients. BJU Int. 110:1666–71 [Google Scholar]
  63. Tichauer KM, Samkoe KS, Gunn JR. 63.  et al. 2014. Microscopic lymph node tumor burden quantified by macroscopic dual-tracer molecular imaging. Nat. Med. 20:1348–53 [Google Scholar]
  64. Nguyen QT, Tsien RY. 64.  2013. Fluorescence-guided surgery with live molecular navigation—a new cutting edge. Nat. Rev. Cancer 13:653–62 [Google Scholar]
  65. Gibbs-Strauss SL, Nasr KA, Fish KM. 65.  et al. 2011. Nerve-highlighting fluorescent contrast agents for image-guided surgery. Mol. Imaging 10:91–101 [Google Scholar]
  66. Cotero VE, Siclovan T, Zhang R. 66.  et al. 2012. Intraoperative fluorescence imaging of peripheral and central nerves through a myelin-selective contrast agent. Mol. Imaging Biol. 14:708–17 [Google Scholar]
  67. Park MH, Hyun H, Ashitate Y. 67.  et al. 2014. Prototype nerve-specific near-infrared fluorophores. Theranostics 4:823–33 [Google Scholar]
  68. van Scheltinga AGT, van Dam GM, Nagengast WB. 68.  et al. 2011. Intraoperative near-infrared fluorescence tumor imaging with vascular endothelial growth factor and human epidermal growth factor receptor 2 targeting antibodies. J. Nucl. Med. 52:1778–85 [Google Scholar]
  69. Barbash GI, Glied SA. 69.  2010. New technology and health care costs—the case of robot-assisted surgery. N. Engl. J. Med. 363:701–4 [Google Scholar]
  70. Gambhir SS. 70.  2002. Molecular imaging of cancer with positron emission tomography. Nat. Rev. Cancer 2:683–93 [Google Scholar]
  71. Weber WA, Grosu AL, Czernin J. 71.  2008. Technology insight: advances in molecular imaging and an appraisal of PET/CT scanning. Nat. Clin. Pract. Oncol. 5:160–70 [Google Scholar]
  72. Takami T, Yamagata T, Naito K. 72.  et al. 2013. Intraoperative assessment of spinal vascular flow in the surgery of spinal intramedullary tumors using indocyanine green videoangiography. Surg. Neurol. Int. 4:135 [Google Scholar]
  73. Buchs NC, Hagen ME, Pugin F. 73.  et al. 2012. Intra-operative fluorescent cholangiography using indocyanin green during robotic single site cholecystectomy. Int. J. Med. Robot. Comput. Assist. Surg. 8:436–40 [Google Scholar]
/content/journals/10.1146/annurev-med-051914-022043
Loading
/content/journals/10.1146/annurev-med-051914-022043
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error