Intest Res.  2021 Jan;19(1):33-44. 10.5217/ir.2019.09175.

Endoscopic molecular imaging in inflammatory bowel disease

Affiliations
  • 1Department of Gastroenterology, Veterans Health Service Medical Center, Seoul, Korea
  • 2Department of Gastroenterology, Digestive Diseases Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea

Abstract

Molecular imaging is a technique for imaging the processes occurring in a living body at a molecular level in real-time, combining molecular cell biology with advanced imaging technologies using molecular probes and fluorescence. Gastrointestinal endoscopic molecular imaging shows great promise for improving the identification of neoplasms, providing characterization for patient stratification and assessing the response to molecular targeted therapy. In inflammatory bowel disease, endoscopic molecular imaging can be used to assess disease severity and predict therapeutic response and prognosis. Endoscopic molecular imaging is also able to visualize dysplasia in the presence of background inflammation. Several preclinical and clinical trials have evaluated endoscopic molecular imaging; however, this area is just beginning to evolve, and many issues have not been solved yet. In the future, it is expected that endoscopic molecular imaging will be of increasing interest among clinicians as a new technology for the identification and evaluation of colorectal neoplasm and colitis-associated cancer.

Keyword

Molecular imaging; Intestinal diseases; Inflammatory bowel disease; Colitis; Neoplasm

Figure

  • Fig. 1. Simultaneous white light (WL) and fluorescent light (FL) images of representative lesions of various morphological and histological subtypes found. (A-C) The lesions shown are clearly visible in WL and show clearly increased fluorescence. (D) A lesion that, although it was visible in WL, had enhanced visibility in FL. (E, F) Images representative of the 9 lesions that were only visible in FL. Polyps are indicated by the white arrows. (A) A 2-cm pedunculated (Paris 0-Ip) tubular adenoma. (B) A 4-cm subpedunculated (Paris 0-Isp) tubulovillous adenoma. (C) A 2-cm sessile (Paris 0-Is) serrated polyp. (D) A 5-mm flat elevated (Paris 0-IIa) tubular adenoma. (E, F) Flat (Paris 0-IIb) tubular adenomas, 5 mm and 4 mm in diameter, respectively. (G) Graph showing the relationship between the degree of fluorescence and the histological diagnosis. (H) Graph showing the relationship between c-Met expression and the degree of fluorescence. (I) Graph showing the relationship between c-Met expression and histological diagnosis. Error bars are means±standard error of the mean. aP<0.001, mixed-model analysis of variance. Adapted from Burggraaf J, et al. Nat Med 2015;21:955-961, with permission from Springer Nature [23].

  • Fig. 2. (A) Schematic diagram of sequential ex vivo staining with crystal violet (CV)-glutamic acid (Glu) and matrix metalloproteinases 14 (MMP14) antibody-quantum dot (Ab-QD) probes. (B) White light images (left panel) and in vivo imaging system images (right panel) of (from left) a normal colon tissue treated with the MMP14 Ab-QD probe, a tumor tissue treated with IgG-QD conjugates, and two other tumor colons treated with the MMP14 Ab–QD probe. Five regions were histopathologically analyzed (arrows 1, 2, 3, 4, and n). (C) Twophoton microscopy (TPM) images of mouse tumor colon stained by the MMP14 Ab-QD probe. Upper image: autofluorescence (Auto-FL) imaging, lower image: MMP14 Ab-QD probe signals (yellow pseudo-color). (D) Ratiometric signals after spraying all tissues with the CV-Glu probe at 5 and 30 minutes after treatment. (E) Overlay images of the CV-Glu probe radio frequency signal at 30 minutes (green pseudo-color) and MMP14 Ab-QD probe signal (red pseudo-color). (F) Time-dependent TPM images of the CV–Glu probe in the same area shown in panel (C). First row: CV–Glu; second row: CV; third row: overlay images; fourth row: ratiometric images. (G) TPM fluorescence images recorded moving down in the z-direction. First row: MMP14 Ab-QDs (yellow pseudo-color); second row: overlay images of CV-Glu probe. Scale bar: 50 µm. AOM, azoxymethane; DSS, dextran sodium sulfate. Adapted from Park Y, et al. Acs Nano 2014;8:8896-8910, with permission from John Wiley and Sons [24].

  • Fig. 3. Representative example of endoscopic submucosal dissection for colitis-associated dysplasia. (A) Large, non-ulcerated Paris type O-IIa dysplasia with a distinct border in the rectum. (B) Mucosal incision was performed after submucosal injection. (C) Mild but diffuse submucosal fibrosis and submucosal fat deposition. (D-F) The colonoscope was changed into a gastroscope to expose the submucosal layer more effectively, and en bloc resection was achieved. The final histology revealed low-grade dysplasia (42×40 mm in size, with clear lateral and vertical margins). Adapted from Yang DH, et al. Clin Endosc 2019;52:120-128 [29].

  • Fig. 4. Dysplastic lesion. (A) White light endoscopic view showing a polypoid lesion (Paris 0-Is) of the transverse colon. (B) After resection and coloration with the 100 μM VRPMPLQ peptide solution, confocal laser endomicroscopy shows active binding of the peptide to dysplastic colonocytes is observed. This along with passive accumulation of the peptide determines an increase in fluorescence. (C) Conventional histology (H&E, ×106) showing low-grade dysplasia. VRPMPLQ is a synthetic peptide conjugated with fluorescein, which shows more selective binding to dysplastic tissue than to normal mucosa. Adapted from De Palma GD, et al. PLoS One 2017;12:e0180509 [67].


Cited by  2 articles

Differentiating between Intestinal Tuberculosis and Crohn’s Disease May Be Complicated by Multidrug-resistant Mycobacterium tuberculosis
Seung Wook Hong, Sang Hyoung Park, Byong Duk Ye, Suk-Kyun Yang
Ewha Med J. 2021;44(3):93-96.    doi: 10.12771/emj.2021.44.3.93.

Is Multidrug-resistant Extrapulmonary Tuberculosis Important? If So, What Is Our Strategy?
Seong-Eun Kim
Ewha Med J. 2021;44(4):148-149.    doi: 10.12771/emj.2021.44.4.148.


Reference

1. Drossman DA. Challenges in the physician-patient relationship: feeling “drained”. Gastroenterology. 2001; 121:1037–1038.
Article
2. Matloff JL, Abidi W, Richards-Kortum R, Sauk J, Anandasabapathy S. High-resolution and optical molecular imaging for the early detection of colonic neoplasia. Gastrointest Endosc. 2011; 73:1263–1273.
Article
3. van Rijn JC, Reitsma JB, Stoker J, Bossuyt PM, van Deventer SJ, Dekker E. Polyp miss rate determined by tandem colonoscopy: a systematic review. Am J Gastroenterol. 2006; 101:343–350.
Article
4. Yalamarthi S, Witherspoon P, McCole D, Auld CD. Missed diagnoses in patients with upper gastrointestinal cancers. Endoscopy. 2004; 36:874–879.
Article
5. Dacosta RS, Wilson BC, Marcon NE. New optical technologies for earlier endoscopic diagnosis of premalignant gastrointestinal lesions. J Gastroenterol Hepatol. 2002; 17 Suppl:S85S104.
Article
6. Goetz M, Wang TD. Molecular imaging in gastrointestinal endoscopy. Gastroenterology. 2010; 138:828–833.
Article
7. Kim JW, Yoon SM, Myung SJ. Molecular imaging of gastrointestinal malignancies. Intest Res. 2011; 9:90–96.
Article
8. Kim SY, Myung SJ. Optical molecular imaging for diagnosing intestinal diseases. Clin Endosc. 2013; 46:620–626.
Article
9. Shahid MW, Buchner AM, Coron E, et al. Diagnostic accuracy of probe-based confocal laser endomicroscopy in detecting residual colorectal neoplasia after EMR: a prospective study. Gastrointest Endosc. 2012; 75:525–533.
Article
10. Shahid MW, Buchner AM, Heckman MG, et al. Diagnostic accuracy of probe-based confocal laser endomicroscopy and narrow band imaging for small colorectal polyps: a feasibility study. Am J Gastroenterol. 2012; 107:231–239.
Article
11. Shahid MW, Buchner AM, Raimondo M, Woodward TA, Krishna M, Wallace MB. Accuracy of real-time vs. blinded offline diagnosis of neoplastic colorectal polyps using probebased confocal laser endomicroscopy: a pilot study. Endoscopy. 2012; 44:343–348.
Article
12. Wallace MB, Meining A, Canto MI, et al. The safety of intravenous fluorescein for confocal laser endomicroscopy in the gastrointestinal tract. Aliment Pharmacol Ther. 2010; 31:548552.
Article
13. Yao J, Wang LV. Recent progress in photoacoustic molecular imaging. Curr Opin Chem Biol. 2018; 45:104–112.
Article
14. Yang JM, Favazza C, Chen R, et al. Simultaneous functional photoacoustic and ultrasonic endoscopy of internal organs in vivo. Nat Med. 2012; 18:1297–1302.
Article
15. Marten K, Bremer C, Khazaie K, et al. Detection of dysplastic intestinal adenomas using enzyme-sensing molecular beacons in mice. Gastroenterology. 2002; 122:406–414.
Article
16. Miller SJ, Joshi BP, Feng Y, Gaustad A, Fearon ER, Wang TD. In vivo fluorescence-based endoscopic detection of colon dysplasia in the mouse using a novel peptide probe. PLoS One. 2011; 6:e17384.
Article
17. Joshi BP, Liu Z, Elahi SF, Appelman HD, Wang TD. Near-infrared-labeled peptide multimer functions as phage mimic for high affinity, specific targeting of colonic adenomas in vivo (with videos). Gastrointest Endosc. 2012; 76:1197–1206.
Article
18. Zhou L, El-Deiry WS. Multispectral fluorescence imaging. J Nucl Med. 2009; 50:1563–1566.
Article
19. Miller SJ, Lee CM, Joshi BP, Gaustad A, Seibel EJ, Wang TD. Targeted detection of murine colonic dysplasia in vivo with flexible multispectral scanning fiber endoscopy. J Biomed Opt. 2012; 17:021103.
Article
20. Bae SM, Bae DJ, Do EJ, et al. Multi-spectral fluorescence imaging of colon dysplasia invivo using a multi-spectral endoscopy system. Transl Oncol. 2019; 12:226–235.
Article
21. Yoon SM, Myung SJ, Kim IW, et al. Application of near-infrared fluorescence imaging using a polymeric nanoparticlebased probe for the diagnosis and therapeutic monitoring of colon cancer. Dig Dis Sci. 2011; 56:3005–3013.
Article
22. Yoon SM, Myung SJ, Ye BD, et al. Near-infrared fluorescence imaging using a protease-specific probe for the detection of colon tumors. Gut Liver. 2010; 4:488–497.
Article
23. Burggraaf J, Kamerling IM, Gordon PB, et al. Detection of colorectal polyps in humans using an intravenously administered fluorescent peptide targeted against c-Met. Nat Med. 2015; 21:955–961.
Article
24. Park Y, Ryu YM, Jung Y, et al. Spraying quantum dot conjugates in the colon of live animals enabled rapid and multiplex cancer diagnosis using endoscopy. ACS Nano. 2014; 8:88968910.
Article
25. Park S, Lim SY, Bae SM, Kim SY, Myung SJ, Kim HJ. Indocyanine-based activatable fluorescence turn-on probe for γ-glutamyltranspeptidase and its application to the mouse model of colon cancer. ACS Sens. 2016; 1:579–583.
Article
26. Kim J, Do EJ, Moinova H, et al. Molecular imaging of colorectal tumors by targeting colon cancer secreted protein-2 (CCSP-2). Neoplasia. 2017; 19:805–816.
Article
27. Bae SM, Park SJ, Choi M, et al. PSP1, a phosphatidylserinerecognizing peptide, is useful for visualizing radiation-induced apoptosis in colorectal cancer in vitro and in vivo. Transl Oncol. 2018; 11:1044–1052.
Article
28. Park Y, Ryu YM, Wang T, et al. Colorectal cancer diagnosis using enzyme‐sensitive ratiometric fluorescence dye and antibody-quantum dot conjugates for multiplexed detection. Adv Funct Mater. 2018; 28:1703450.
Article
29. Yang DH, Rey I. Endoscopic submucosal dissection for colitis-associated dysplasia. Clin Endosc. 2019; 52:120–128.
Article
30. Jess T, Gamborg M, Matzen P, Munkholm P, Sørensen TI. Increased risk of intestinal cancer in Crohn’s disease: a metaanalysis of population-based cohort studies. Am J Gastroenterol. 2005; 100:2724–2729.
Article
31. Jess T, Rungoe C, Peyrin-Biroulet L. Risk of colorectal cancer in patients with ulcerative colitis: a meta-analysis of population-based cohort studies. Clin Gastroenterol Hepatol. 2012; 10:639–645.
Article
32. Kim BJ, Yang SK, Kim JS, et al. Trends of ulcerative colitis-associated colorectal cancer in Korea: a KASID study. J Gastroenterol Hepatol. 2009; 24:667–671.
Article
33. Mattar MC, Lough D, Pishvaian MJ, Charabaty A. Current management of inflammatory bowel disease and colorectal cancer. Gastrointest Cancer Res. 2011; 4:53–61.
34. Feagins LA, Souza RF, Spechler SJ. Carcinogenesis in IBD: potential targets for the prevention of colorectal cancer. Nat Rev Gastroenterol Hepatol. 2009; 6:297–305.
Article
35. Rubin DC, Shaker A, Levin MS. Chronic intestinal inflammation: inflammatory bowel disease and colitis-associated colon cancer. Front Immunol. 2012; 3:107.
Article
36. Saleh M, Trinchieri G. Innate immune mechanisms of colitis and colitis-associated colorectal cancer. Nat Rev Immunol. 2011; 11:9–20.
Article
37. Kawanishi S, Hiraku Y, Pinlaor S, Ma N. Oxidative and nitrative DNA damage in animals and patients with inflammatory diseases in relation to inflammation-related carcinogenesis. Biol Chem. 2006; 387:365–372.
Article
38. van Dieren JM, Wink JC, Vissers KJ, et al. Chromosomal and microsatellite instability of adenocarcinomas and dysplastic lesions (DALM) in ulcerative colitis. Diagn Mol Pathol. 2006; 15:216–222.
Article
39. Tahara T, Inoue N, Hisamatsu T, et al. Clinical significance of microsatellite instability in the inflamed mucosa for the prediction of colonic neoplasms in patients with ulcerative colitis. J Gastroenterol Hepatol. 2005; 20:710–715.
Article
40. Xie J, Itzkowitz SH. Cancer in inflammatory bowel disease. World J Gastroenterol. 2008; 14:378–389.
Article
41. Itzkowitz S. Colon carcinogenesis in inflammatory bowel disease: applying molecular genetics to clinical practice. J Clin Gastroenterol. 2003; 36(5 Suppl):S70–S74.
42. Itzkowitz SH, Harpaz N. Diagnosis and management of dysplasia in patients with inflammatory bowel diseases. Gastroenterology. 2004; 126:1634–1648.
Article
43. Murthy SK, Kiesslich R. Evolving endoscopic strategies for detection and treatment of neoplastic lesions in inflammatory bowel disease. Gastrointest Endosc. 2013; 77:351–359.
Article
44. Annese V, Beaugerie L, Egan L, et al. European evidencebased consensus: inflammatory bowel disease and malignancies. J Crohns Colitis. 2015; 9:945–965.
Article
45. Farraye FA, Odze RD, Eaden J, Itzkowitz SH. AGA technical review on the diagnosis and management of colorectal neoplasia in inflammatory bowel disease. Gastroenterology. 2010; 138:746–774.
Article
46. Cairns SR, Scholefield JH, Steele RJ, et al. Guidelines for colorectal cancer screening and surveillance in moderate and high risk groups (update from 2002). Gut. 2010; 59:666689.
Article
47. Kornbluth A, Sachar DB; Practice Parameters Committee of the American College of Gastroenterology. Ulcerative colitis practice guidelines in adults: American College of Gastroenterology, Practice Parameters Committee. Am J Gastroenterol. 2010; 105:501–523.
Article
48. Awais D, Siegel CA, Higgins PD. Modelling dysplasia detection in ulcerative colitis: clinical implications of surveillance intensity. Gut. 2009; 58:1498–1503.
Article
49. Subramanian V, Ramappa V, Telakis E, et al. Comparison of high definition with standard white light endoscopy for detection of dysplastic lesions during surveillance colonoscopy in patients with colonic inflammatory bowel disease. Inflamm Bowel Dis. 2013; 19:350–355.
Article
50. Kiesslich R, Neurath MF. Chromoendoscopy in inflammatory bowel disease. Gastroenterol Clin North Am. 2012; 41:291302.
Article
51. Brown SR, Baraza W, Din S, Riley S. Chromoscopy versus conventional endoscopy for the detection of polyps in the colon and rectum. Cochrane Database Syst Rev. 2016; 4–CD006439.
Article
52. Watanabe T, Ajioka Y, Mitsuyama K, et al. Comparison of targeted vs random biopsies for surveillance of ulcerative colitis-associated colorectal cancer. Gastroenterology. 2016; 151:1122–1130.
Article
53. Rubin DT, Rothe JA, Hetzel JT, Cohen RD, Hanauer SB. Are dysplasia and colorectal cancer endoscopically visible in patients with ulcerative colitis? Gastrointest Endosc. 2007; 65:998–1004.
Article
54. Rutter MD, Saunders BP, Wilkinson KH, Kamm MA, Williams CB, Forbes A. Most dysplasia in ulcerative colitis is visible at colonoscopy. Gastrointest Endosc. 2004; 60:334–339.
Article
55. American Society for Gastrointestinal Endoscopy Standards of Practice Committee, Shergill AK, Lightdale JR, et al. The role of endoscopy in inflammatory bowel disease. Gastrointest Endosc. 2015; 81:1101–1121.
Article
56. Laine L, Kaltenbach T, Barkun A, et al. SCENIC international consensus statement on surveillance and management of dysplasia in inflammatory bowel disease. Gastrointest Endosc. 2015; 81:489–501.
Article
57. Velayos F, Kathpalia P, Finlayson E. Changing paradigms in detection of dysplasia and management of patients with inflammatory bowel disease: is colectomy still necessary? Gastroenterology. 2017; 152:440–450.
Article
58. Annese V, Daperno M, Rutter MD, et al. European evidence based consensus for endoscopy in inflammatory bowel disease. J Crohns Colitis. 2013; 7:982–1018.
Article
59. Iacopini F, Saito Y, Yamada M, et al. Curative endoscopic submucosal dissection of large nonpolypoid superficial neoplasms in ulcerative colitis (with videos). Gastrointest Endosc. 2015; 82:734–738.
Article
60. Suzuki N, Toyonaga T, East JE. Endoscopic submucosal dissection of colitis-related dysplasia. Endoscopy. 2017; 49:12371242.
Article
61. Yang DH, Kim J, Song EM, et al. Outcomes of ulcerative colitis-associated dysplasia patients referred for potential endoscopic submucosal dissection. J Gastroenterol Hepatol. 2019; 34:1581–1589.
Article
62. Kinoshita S, Uraoka T, Nishizawa T, et al. The role of colorectal endoscopic submucosal dissection in patients with ulcerative colitis. Gastrointest Endosc. 2018; 87:1079–1084.
Article
63. Gounaris E, Martin J, Ishihara Y, et al. Fluorescence endoscopy of cathepsin activity discriminates dysplasia from colitis. Inflamm Bowel Dis. 2013; 19:1339–1345.
Article
64. Mason SD, Joyce JA. Proteolytic networks in cancer. Trends Cell Biol. 2011; 21:228–237.
Article
65. Mitsunaga M, Kosaka N, Choyke PL, et al. Fluorescence endoscopic detection of murine colitis-associated colon cancer by topically applied enzymatically rapid-activatable probe. Gut. 2013; 62:1179–1186.
Article
66. Pompella A, De Tata V, Paolicchi A, Zunino F. Expression of gamma-glutamyltransferase in cancer cells and its significance in drug resistance. Biochem Pharmacol. 2006; 71:231238.
67. De Palma GD, Colavita I, Zambrano G, et al. Detection of colonic dysplasia in patients with ulcerative colitis using a targeted fluorescent peptide and confocal laser endomicroscopy: a pilot study. PLoS One. 2017; 12:e0180509.
Article
68. Kiesslich R, Burg J, Vieth M, et al. Confocal laser endoscopy for diagnosing intraepithelial neoplasias and colorectal cancer in vivo. Gastroenterology. 2004; 127:706–713.
Article
69. Kiesslich R, Goetz M, Lammersdorf K, et al. Chromoscopyguided endomicroscopy increases the diagnostic yield of intraepithelial neoplasia in ulcerative colitis. Gastroenterology. 2007; 132:874–882.
Article
70. Caobelli F, Panarotto MB, Andreoli F, Ravelli A, De Agostini A, Giubbini R. Is 99mTc-HMPAO granulocyte scan an alternative to endoscopy in pediatric chronic inflammatory bowel disease (IBD)? Eur J Pediatr. 2011; 170:51–57.
Article
71. Treglia G, Quartuccio N, Sadeghi R, et al. Diagnostic performance of fluorine-18-fluorodeoxyglucose positron emission tomography in patients with chronic inflammatory bowel disease: a systematic review and a meta-analysis. J Crohns Colitis. 2013; 7:345–354.
Article
72. Caobelli F, Evangelista L, Quartuccio N, et al. Role of molecular imaging in the management of patients affected by inflammatory bowel disease: state-of-the-art. World J Radiol. 2016; 8:829–845.
Article
73. Wang H, Machtaler S, Bettinger T, et al. Molecular imaging of inflammation in inflammatory bowel disease with a clinically translatable dual-selectin-targeted US contrast agent: comparison with FDG PET/CT in a mouse model. Radiology. 2013; 267:818–829.
Article
74. Kiesslich R, Duckworth CA, Moussata D, et al. Local barrier dysfunction identified by confocal laser endomicroscopy predicts relapse in inflammatory bowel disease. Gut. 2012; 61:1146–1153.
Article
75. Atreya R, Neumann H, Neufert C, et al. In vivo imaging using fluorescent antibodies to tumor necrosis factor predicts therapeutic response in Crohn’s disease. Nat Med. 2014; 20:313–318.
Article
Full Text Links
  • IR
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr