Sei sulla pagina 1di 9

Digestive Endoscopy 2014; 26 (Suppl. 1): 43–51 doi: 10.1111/den.

12190

Review

Present and future perspectives of virtual


chromoendoscopy with i-scan and optical enhancement
technology
Helmut Neumann,1 Mitsuhiro Fujishiro,2 C. Mel Wilcox3 and Klaus Mönkemüller3
1
Department of Medicine 1, Interdisciplinary Endoscopy, University of Erlangen-Nuremberg, Erlangen, Germany;
2
Department of Endoscopy and Endoscopic Surgery, Graduate School of Medicine, The University of Tokyo,
Tokyo, Japan; and 3Basil Hirschowitz Endoscopic Center of Excellence, Division of Gastroenterology and
Hepatology, University of Alabama at Birmingham, Birmingham, USA

Despite being the current gold standard, white-light endoscopy polyps and inflammatory bowel diseases. Moreover, DLC tech-
may miss a significant proportion of lesions within the colorec- niques enable a more precise characterization of lesions through-
tum, thus leading to a misinterpretation of various disease find- out the whole luminal GI tract, thereby potentially enabling more
ings. Traditionally, dye-based chromoendoscopy is used to accurate endoscopic therapies. In the present review we focus
improve both detection and characterization of lesions during on the newly introduced dye-less chromoendoscopy technique
luminal gastrointestinal (GI) endoscopy. Recently introduced dye- i-scan and give an additional outlook on the recent development
less chromoendoscopy (DLC) techniques have overcome many of of optical enhancement technology.
the limitations of dye-based chromoendoscopy, thereby poten-
tially improving lesion recognition and characterization. In detail, Key words: chromoendoscopy, inflammatory bowel disease
DLC techniques allow for better detection of esophageal lesions, (IBD), i-scan, optical enhancement technology, polyp
gastric cancer and colorectal pathologies including colorectal

INTRODUCTION six were cancers. These data were recently confirmed by Pohl
and coworkers in a randomized two-center trial including
E NDOSCOPIC IMAGING OF the gastrointestinal (GI)
tract is routinely carried out by using white-light endos-
copy. However, standard white-light endoscopy may miss a
more than 1000 patients.6 DBC with methylene blue has also
been shown to allow a more accurate diagnosis of the extent
and severity of the inflammatory activity in inflammatory
significant amount of lesions, especially within the colorec-
bowel diseases compared with conventional colonoscopy.7 In
tum, and may also lead to a misinterpretation of findings,
addition, DBC allowed for the early detection of intraepithe-
thereby resulting in delayed or suboptimal therapies.1,2 In an
lial neoplasia and colitis-associated cancer in patients with
attempt to overcome the limitations of white-light endos-
long-standing ulcerative colitis.7
copy, dye-based chromoendoscopy (DBC) techniques were
Moreover, the impact of DBC for detection and charac-
introduced more than one decade ago.3,4 DBC has been
terization of esophageal lesions, including esophageal squa-
proven to be feasible for improved detection and character-
mous cell cancer, non-erosive reflux disease and Barrett’s
ization rates of various GI lesions. In this context, one early
esophagus, has been demonstrated.8–11
study evaluated the efficacy of indigocarmine dye spraying
In terms of stomach lesions, indigocarmine DBC has rou-
in 52 patients undergoing screening colonoscopy.5 Among
tinely been used after detection of gastric lesions by white-
48 patients with mucosa of normal appearance, 27 patients
light endoscopy for an extended period of time, especially in
showed 178 lesions after staining with a mean size of 3 mm.
Japan, although there was no clear evidence that DBC could
On histological investigation, 67 lesions were adenomas and
actually improve detection and characterization of gastric
lesions. In this context, Kawahara et al. have recently shown
that indigocarmine dye-spraying significantly improved
Corresponding: Helmut Neumann, Department of Medicine 1,
University of Erlangen-Nuremberg, Ulmenweg 18, Erlangen diagnostic accuracy of tumor extents and that a mixture of
91054, Germany. Email: helmut.neumann@uk-erlangen.de acetic acid and indigocarmine could lead to much higher
Received 17 June 2013; accepted 2 September 2013. accuracy in 108 already-known early gastric cancers.12

© 2013 The Authors 43


Digestive Endoscopy © 2013 Japan Gastroenterological Endoscopy Society
bs_bs_banner
44 H. Neumann et al. Digestive Endoscopy 2014; 26 (Suppl. 1): 43–51

Table 1 Summary of currently available dye-less chromoendoscopy (DLC) techniques


Technology Brand Company Specifications

Optical DLC NBI Olympus, Tokyo, Japan Evis Exera II & III
CBI Aohua Photoelectricity, Shanghai, China AQ-100
Virtual DLC i-scan Pentax, Tokyo, Japan EPKi, EPKi-5000, EPKi-7000
FICE Fujifilm, Tokyo, Japan EPX-4400

CBI, Compound Band Imaging; FICE, Fuji Intelligent Color Enhancement; NBI, narrow band imaging.

Additionally, intravital staining of the duodenum with Table 2 Potential applications and clinical advantages of virtual
indigocarmine was shown to be useful for detecting mucosal chromoendoscopy using i-scan
abnormalities, delineating their extent, and allowing targeted Esophagus
biopsies.13 Taken together, DBC may unmask mucosal • Early squamous cell cancer
lesions that are not easily identified by routine white-light • Gastric inlet patch
endoscopy. In addition, DBC facilitates visualization of the • Minimal change gastroesophageal reflux disease
margins of flat or depressed lesions and helps to predict • Barrett’s esophagus and early adenocarcinoma
lesion histology for an optimized endoscopic therapy. Nev- Stomach
ertheless, DBC has some potential limitations, as the dye • Atrophic gastritis
does not always coat the surface evenly, thereby often result- • Gastric antral vascular ectasia (GAVE)
ing in overstained and less stained mucosal areas, which, in • Early gastric cancer
turn, may affect the diagnosis and characterization of Duodenum
lesions. In addition, there are additional costs for dye spray- • Celiac disease
ing, including those for the dye and the spraying catheter. • Whipple’s disease
Furthermore, the procedure requires a learning phase and the
Colon
mucosa should be well prepared, as mucus or residual stool
• Polyps
may result in false-positive findings.
• Inflammatory bowel diseases
In an attempt to overcome the limitations of DBC, • Food allergies
dye-less chromoendoscopy (DLC) techniques have been
introduced (Table 1). These are divided into optical chromo-
endoscopy techniques, including narrow band imaging
(NBI; Olympus, Tokyo, Japan) and Compound Band time without any noticeable time delay. Currently, two
Imaging (CBI; Aohua Photoelectricity, Shanghai, China), virtual chromoendoscopy techniques are available, including
and virtual chromoendoscopy techniques including Fujifilm FICE and i-scan. While FICE has no standardized image
Intelligent Color Enhancement (FICE; Fujifilm, Tokyo, settings, a recent international consensus recommended
Japan) and i-scan (Pentax, Tokyo, Japan). DLC is activated uniform settings for i-scan, thereby allowing even the com-
simply by pushing a button on the handle of the endoscope, parison of different studies.14 As a standard feature, the
thereby enabling high-contrast imaging of the mucosal i-scan system is integrated into the EPKi high-definition
surface without any time delay or the need for special video processor units (Pentax, Tokyo, Japan). A detailed
equipment. description of the technology was recently presented by
In the present review, we will focus on present and future Kodashima and Fujishiro.15 In short, i-scan consists of three
perspectives of virtual chromoendoscopy using i-scan and different image algorithms: (i) surface enhancement (SE);
give an outlook on the advent of the recently introduced (ii) tone enhancement (TE); and (iii) contrast enhancement
optical enhancement technology (Table 2). (CE). Switching between different modes is done by pushing
a button on the handle of the endoscope. As explained by
Kodashima and Fujishiro, SE enhances light-dark contrast
TECHNIQUE OF VIRTUAL
by obtaining luminance intensity data for each pixel and
CHROMOENDOSCOPY
applying an algorithm that allows detailed observation of the

V IRTUAL CHROMOENDOSCOPY IS based on the


principle of digital post-processing. Accordingly, the
endoscopic image is reconstructed into virtual images in real
mucosal surface structure and lesion borders. In addition, CE
digitally adds blue color to relatively dark areas within the
endoscopic image by obtaining luminance intensity data for

© 2013 The Authors


Digestive Endoscopy © 2013 Japan Gastroenterological Endoscopy Society
Digestive Endoscopy 2014; 26 (Suppl. 1): 43–51 i-scan: Optical enhancement technology 45

each pixel and applying an algorithm that allows detailed visible changes and, afterwards, targeted biopsies were
observation of subtle irregularities around the surface and a carried out. It was found that Lugol’s solution significantly
detailed inspection of the mucosal vascular pattern morphol- improved the identification of patients with esophagitis and
ogy. Last, TE dissects and analyses the individual red-green- reduced misclassification, whereas i-scan and DBC helped
blue (RGB) components of a normal endoscopic image in to identify reflux-associated lesions. These data were also
real time and then alters the color frequencies of each com- confirmed by a recent prospective study from Korea includ-
ponent and recombines the components to a single, new ing over 500 patients.18 White-light endoscopy identified
color image without visible delay for the examiner. Thereby, reflux esophagitis in approximately 22% of patients,
TE enhances minute mucosal structures and subtle changes whereas i-scan identified reflux associated lesions in 30%
in color.15 of patients. The detection rate of minimal change GERD
Based on current consensus, three i-scan settings are rec- was significantly higher in the i-scan group as compared to
ommended as follows:14 (i) i-scan 1 for detection of lesions. white-light endoscopy, but was not different for erosive
This algorithm uses only SE to refine imaging of subtle reflux esophagitis grade A and grade B according to the
surface abnormalities without altering the brightness of the Los Angeles classification (LA). Additionally, i-scan
endoscopic picture. (ii) i-scan 2 mode was established for showed better interobserver agreement than white-light
characterization of lesions. This algorithm combines SE and endoscopy. These data were also confirmed by Kim and
TE, thereby enhancing minute mucosal changes and vessel coworkers.19
structures. (iii) Last, i-scan 3 adds CE to the endoscopic
image (in addition to SE and TE) and is recommended for
demarcation of lesions as it digitally adds blue color to PRESENT APPLICATIONS OF i-SCAN
darker edges within the endoscopic image. TECHNOLOGY IN THE STOMACH

PRESENT APPLICATIONS OF i-SCAN


D IAGNOSES OF ATROPHIC gastritis, vascular lesions
such as gastric antral vascular ectasia (GAVE), and
gastric neoplasms represent potential candidates for appli-
TECHNOLOGY IN THE ESOPHAGUS
cation of i-scan in the stomach (Figs 1,2). Currently, only

P ATIENTS WITH GASTROESOPHAGEAL reflux


disease (GERD) are divided into those with non-
erosive reflux disease (NERD), erosive reflux disease and
limited data are available on the use of i-scan for detection
of gastric lesions. However, in our experience, i-scan might
increase the detection of early gastric cancer in comparison
Barrett’s esophagus. Patients with NERD suffer from with white-light endoscopy.20 Additionally, our preliminary
typical reflux symptoms (e.g. heartburn) but show no results of i-scan revealed significantly higher accuracy in
visible mucosal lesions during white-light endoscopy.16 the diagnoses of lateral tumor extent in 21 gastric neo-
Hoffman and coworkers evaluated the efficacy of i-scan plasms in comparison with white-light endoscopy (95.2%
and chromoendoscopy with Lugol’s solution for differen- vs 66.7%).21 Taken together, i-scan could potentially be
tiation of reflux patients.17 In their study, the distal esopha- used for screening endoscopy to detect gastric lesions
gus of patients with heartburn was inspected with three and might also be useful for detailed inspection after detec-
imaging modalities. High-definition white-light endoscopy tion of suspicious lesions. Nevertheless, well-designed, pro-
was followed by i-scan and Lugol’s solution dye-spraying. spective studies on this topic are still missing and highly
The esophagus was evaluated for mucosal breaks and small anticipated.

A B

Figure 1 Intestinal-type intramucosal


gastric cancer at the lesser curvature of
the gastric body. (a) During white-light
endoscopy, correct identification of the
lesion is difficult. (b) i-scan endoscopy
enhances the lesion for better detec-
tion and characterization.

© 2013 The Authors


Digestive Endoscopy © 2013 Japan Gastroenterological Endoscopy Society
46 H. Neumann et al. Digestive Endoscopy 2014; 26 (Suppl. 1): 43–51

A B C

Figure 2 Intestinal-type intramucosal gastric cancer. (a) White-light endoscopy shows a small reddish depression in the lower gastric
body. (b) i-scan enhances the atrophic changes in the background as well as the tumor itself, which reveals that the tumor is located on
the atrophic border. (c) i-scan additionally reveals a clear demarcation line of the tumor to the surrounding tissue.

PRESENT APPLICATIONS OF i-SCAN instruments used for polypectomy, retrieval devices,


TECHNOLOGY FOR DUODENAL PATHOLOGIES physician and nurses fees, pathology assessments and
further surveillance. Therefore, the American Society for
O NLY LIMITED DATA are currently available on the
use of i-scan for the diagnosis of duodenal lesions.
One recent pilot study aimed to determine the diagnostic
Gastrointestinal Endoscopy (ASGE) recently introduced
a new concept for real-time endoscopic assessment of
the histology of diminutive polyps.25 The so-called PIVI
accuracy of i-scan for the evaluation of duodenal villous
(Preservation and Incorporation of Valuable endoscopic
patterns by using histology as the reference standard.22
Innovations) statement includes two new attractive para-
During endoscopy, duodenal villous patterns were evaluated
digms to reduce costs associated with diminutive colon
and classified as normal, partial villous atrophy, or marked
polyp resection. First, in order for colorectal polyps ≤5 mm
villous atrophy and the results were then compared with
in size to be resected and discarded, endoscopic technology
histology. For i-scan, an overall accuracy of 100% was dem-
should provide ≥90% agreement in assignment of post-
onstrated for the detection of marked villous atrophy pat-
polypectomy surveillance compared to decisions based on
terns, whereas the accuracy was 90% for determination of
pathological assessment of all identified polyps. Second, in
partial villous atrophy or normal villous patterns. Another
order for a technology used to guide the decision to leave
recent report highlighted the potential of i-scan for in vivo
suspected hyperplastic polyps ≤5 mm in size in place, the
diagnosis of Whipple’s disease.23 White-light endoscopy
technology should provide a negative predictive value
revealed pale yellow, shaggy mucosa with intermittent,
>90% for adenomatous polyp histology. One recent two-
superficial, erythematous eroded patches of the duodenum,
center trial examined whether i-scan was feasible to cor-
whereas i-scan additionally revealed edematous and
rectly identify hyperplastic and adenomatous colorectal
engorged duodenal villi filled with a white material repre-
lesions and also assessed the learning curve of this tech-
senting lymph. Moreover, concentric rings of the mucosa
nique.26 After a training set containing 20 images with
were visible, which have been identified to be specific for
known histology was reviewed to standardize image inter-
Whipple’s disease in previous studies using optical magni-
pretation, images from 110 colorectal lesions were ana-
fication endoscopy with 115-fold magnification.24
lyzed by using corresponding polypectomies as the
reference standard. Overall, four endoscopists with no prior
experience in i-scan image interpretation from two differ-
PRESENT APPLICATIONS OF i-SCAN
ent endoscopy centers evaluated the images. Overall accu-
TECHNOLOGY FOR COLORECTAL LESIONS
racy for the group was 74% for lesions 1 to 22, 80% for

C OLORECTAL CANCER IS the second most common


cause of cancer-related death in Europe and North
America and it is well accepted that screening colonoscopy
lesions 23 to 44, 84% for lesions 45 to 66, 88% for lesions
67 to 88, and 94% for lesions 89 to 110. Accuracy of i-scan
for prediction of polyp histology was not dependent on
reduces colorectal cancer mortality by early detection and polyp size (≤5 mm, 6–10 mm, >10 mm) and the ability to
endoscopic treatment (i.e. polypectomy). Nevertheless, the obtain high-quality images was stable over time and con-
unnecessary removal of non-adenomatous polyps results in stantly produced high-quality images. Whereas negative
enormous costs for the health-care system related to the predictive value was moderate in the first three sessions

© 2013 The Authors


Digestive Endoscopy © 2013 Japan Gastroenterological Endoscopy Society
Digestive Endoscopy 2014; 26 (Suppl. 1): 43–51 i-scan: Optical enhancement technology 47

(50–80%) it increased for the subsequent 22 lesions (71– significantly more patients with colorectal neoplasia (38%)
83%). In the last teaching sessions, two investigators compared with standard resolution endoscopy (13%). More-
reached the proposed negative predictive value ≥90% over, significantly more neoplastic (adenomatous and can-
according to the ASGE PIVI statement (80–100%). The cerous) lesions and more flat adenomas were detected by
authors concluded that accurate interpretation of i-scan using i-scan. Of interest, the final histological result could be
images for prediction of hyperplastic and adenomatous predicted with a nearly 99% accuracy. These data were also
colorectal lesions follows a learning curve but can be confirmed by Testoni et al. showing that i-scan during the
learned rapidly. withdrawal phase of colonoscopy significantly increased the
Another recent multicenter study including eight expert detection of colonic mucosal lesions, particularly the detec-
endoscopists assessed interobserver agreement in the visu- tion of small and non-protruding polyps.30 In contrast to
alization of the surfaces and margins of colorectal polyps these studies, one prospective, randomized, back-to-back
and in distinguishing neoplastic from non-neoplastic polyps trial was not able to show a significantly improved adenoma
by using i-scan.27 A total of 400 images were stored for detection rate when using i-scan.31 Patients were randomized
analysis. Distinction between neoplastic and non-neoplastic to the first withdrawal with either conventional high-
tissue was based on Kudo’s pit pattern classification, con- definition white-light or i-scan. In addition, all patients
sidering patterns I and II as non-neoplastic lesions and pat- underwent a second examination with high-definition white-
terns III, IV and V as neoplastic lesions (III and IV as light as the criterion standard. The adenoma detection rates
adenomatous and V as carcinomas). Overall, there was a during the first withdrawal of high-definition, i-scan 1 mode,
kappa agreement of 0.370 (P < 0.001) and 0.306 (P < 0.001) and i-scan 2 mode were 32%, 37%, and 33%, respectively
regarding pit pattern and margins, respectively. The kappa (P = 0.742), and the adenoma miss rates of each group were
agreement for the differentiation between neoplastic and 23%, 19%, and 16%, respectively (P = 0.513). Based on the
non-neoplastic lesions was 0.446 (P < 0.001). Accordingly, a multivariate analysis, the application of i-scan was not asso-
good interobserver agreement was observed for the evalua- ciated with an improvement in adenoma detection and the
tion of neoplastic and non-neoplastic lesions and a less good prevention of missed polyps. However, the prediction of
agreement for the evaluation of pit pattern and margins. The neoplastic and non-neoplastic colorectal lesions was more
authors concluded that adequate training is required in order precise in the i-scan group as compared with the high-
to interpret images acquired with the i-scan technique. definition white-light group.
One recent study compared the detection rate of mucosal The group from Frankfurt tested the efficacy of high-
lesions using i-scan and the withdrawal time of the instru- definition white-light endoscopy alone and in combination
ment among non-expert and expert endoscopists during with i-scan or methylene blue-aided chromoendoscopy in
screening colonoscopy for colorectal cancer.28 Overall, 542 screening for colorectal cancer.32 In 69 patients studied,
colonoscopies were carried out. Notably, it was found that i-scan augmented the identification of lesions from 176 to
i-scan enabled less experienced endoscopists to achieve 335 (P < 0.001) and chromoendoscopy to 646 (P < 0.001).
results comparable to those of experienced ones in detecting Additional detected lesions were mainly flat polyps and the
mucosal lesions (Fig. 3). number of neoplasias detected was not significantly different
Hoffman and coworkers prospectively compared high- between groups (high-definition: 5, i-scan: 11, chromoen-
definition colonoscopy with i-scan versus standard video doscopy: 11), but all could be correctly predicted using
colonoscopy.29 In their study, a total of 220 patients were i-scan or chromoendoscopy. Of note, i-scan was able
randomized in a 1:1 ratio. In this study, i-scan detected to predict neoplasia as precisely as chromoendoscopy.

A B

Figure 3 Non-polypoid lesion with a


mucus cap in the ascending colon visu-
alized by (a) high-definition white-light
endoscopy and (b) i-scan technology.
Surface characteristics and borders of
the lesion become more obvious by
using i-scan.

© 2013 The Authors


Digestive Endoscopy © 2013 Japan Gastroenterological Endoscopy Society
48 H. Neumann et al. Digestive Endoscopy 2014; 26 (Suppl. 1): 43–51

The potential of i-scan for real-time prediction of colorectal OPTICAL ENHANCEMENT TECHNOLOGY
polyp histology was also highlighted by a study from the
Modena group in Italy reporting on a sensitivity, specificity,
and accuracy of 95%, 82%, and 92%, respectively.33 Among
the subset of polyps with good or excellent quality images,
N EW IMAGE-ENHANCED ENDOSCOPIC technol-
ogy using band-limited light, optical enhancement
(OE), was developed by HOYA Co. (Tokyo, Japan) and is
sensitivity and accuracy of i-scan significantly improved to now equipped with the latest endoscopy system (Pentax
97% and 94%, respectively. Video Processor EPK-i7000; HOYA Co.). The new technol-
ogy combines digital signal processing in a similar way to
i-scan and optical filters that limit the spectral characteristics
of the illumination light.
PRESENT APPLICATIONS OF i-SCAN
Previous i-scan technology uses white light alone as an
TECHNOLOGY FOR INFLAMMATORY
illumination light and digital post-processing of the reflec-
BOWEL DISEASE
tion afterwards creates images yielding the virtual chromo-

A CCURATE ASSESSMENT OF disease activity and


extent in inflammatory bowel disease (IBD) appears to
be of paramount importance for optimized medical therapy.
endoscopic image. Although accumulating evidence has
shown the usefulness of i-scan in the clinical setting, emis-
sion of white light alone causes a potential limitation for the
Nevertheless, standard white-light endoscopy is likely an current i-scan technology to obtain higher contrast images of
insensitive test for diagnosis of IBD in the quiescent or even microvascular pattern on the mucosal surface in combination
mild phase of the disease.34,35 Therefore, multiple random with high magnification as shown by NBI with optical mag-
biopsies have to be taken because only histopathology is nification. The basic concept of OE is to overcome the dark-
currently able to predict the exact disease extent and severity ness of NBI which results in less usefulness for detectability
in IBD.36,37 in wide-range observation in the full extended gastrointesti-
One recent study evaluated whether i-scan has the poten- nal lumen. The new innovated optical filters achieve higher
tial to enhance assessment of disease severity and extent in overall transmittance by connecting the peaks of the hemo-
patients with mild or inactive IBD in comparison to high- globin absorption spectrum (415 nm, 540 nm and 570 nm)
definition white-light endoscopy (HD-WL).38 Consecutive creating a continuous wavelength spectrum. There are two
patients with IBD were randomly assigned in a 1:1 ratio to modes with different OE filters (Mode 1 and Mode 2)
undergo colonoscopy with HD-WL (Group 1) or i-scan (Fig. 4). In the Mode 1 optical filter, the main wavelengths of
(Group 2) and the mucosal vascular pattern and any mucosal spectral transmission correspond to the peaks of the hemo-
abnormalities were recorded. Average duration of the exami- globin absorption spectrum and the light between these
nation was 18 min in group A and 20.5 min in group B, peaks is continuously slightly emitted by raising baseline
which was not statistically significant. When comparing the transmittance. In the Mode 2 optical filter, the main wave-
endoscopic prediction of inflammatory extent and activity lengths of the short and mid-wavelength correspond to the
with the histological results, an overall agreement of 51% peaks of the hemoglobin absorption spectrum, and the main
and 56% (group A) and 92% and 90% (group B) was found, R wavelength of the RGB signal is added on the long-wave
respectively. These differences were statistically significant. side. The light between these peaks is also continuously
Therefore, the present study indicated that i-scan has the slightly emitted by raising baseline transmittance to achieve
potential to significantly improve the diagnosis of severity the maximum amount of illumination. Mode 1 is designed
and extent of mucosal inflammation in patients with IBD and mainly to improve visualization of microvessels with a suf-
may therefore open new implications for therapeutic inter- ficient amount of light, and Mode 2 is designed to improve
ventions in patients suffering from IBD. contrast of white-light observation by bringing the color tone
Moreover, one recent study reported on the potential of of the overall image closer to that of natural color (white
i-scan for prediction of mucosal changes in patients with color tone) with much more light than that of the Mode 1
suspected food allergy. Preliminary data suggested that filter. By using the optical filter (Mode 1 or Mode 2), the
patients with intestinal food allergy present with lymphoid microvascular patterns in addition to microsurface patterns
hyperplasia, slight mucosal edema and blurred mucosal vas- on the mucosal surface are clearly observed, which means
cular pattern. Based on these findings, i-scan could predict that the knowledge obtained by optical chromoendoscopy
food allergy with a sensitivity, specificity and accuracy of could potentially be brought into the i-scan technology
85%, 89%, and 86%, respectively. Positive and negative pre- (Figs 4–7). Representative cases of endoscopic images are
dictive value for i-scan to predict food allergy were 92% and shown in Figures 2–4. From our pilot study by using OE
80%, respectively.39 Mode 2 and Mode 1 we found that these methods may be

© 2013 The Authors


Digestive Endoscopy © 2013 Japan Gastroenterological Endoscopy Society
Digestive Endoscopy 2014; 26 (Suppl. 1): 43–51 i-scan: Optical enhancement technology 49

Figure 4 Characteristics of optical


filters and ex-vivo observation of
esophageal cancerous tissue.

A B C

Figure 5 Superficial esophageal cancer with submucosal invasion. (a) White-light image. (b) Mode 2 image. (c) Mode 1 with magnifica-
tion image.

useful for detection and characterization of gastrointestinal lesions. The new optical enhancement technology offers a
neoplasms, respectively.40 new potential for enhanced diagnosis of lesions throughout
the whole luminal gastrointestinal tract. This new technology
is promising to overcome higher miss rates and lower accu-
CONCLUSION racy of characterization. Accumulation of cases and well-
designed clinical trials in the near future are warranted and
A LTHOUGH RECENTLY INTRODUCED, various
large and prospective studies have already shown the
benefit of i-scan for patient management during ongoing
highly anticipated.

endoscopy. Indications for image-enhanced endoscopy with


CONFLICTS OF INTEREST
i-scan include diagnosis of minimal change esophagitis,
duodenal pathologies, colorectal adenomas and inflamma-
tory bowel disease. Future studies should now focus on the
detection of other esophageal pathologies including squa-
H ELMUT NEUMANN IS a recipient of the 2013 ASGE
Cook Medical Don Wilson Award. This work was
done during his awardee period at the Basil Hirschowitz
mous cell cancer and Barrett’s esophagus as well as gastric Endoscopic Center of Excellence, University of Alabama,

© 2013 The Authors


Digestive Endoscopy © 2013 Japan Gastroenterological Endoscopy Society
50 H. Neumann et al. Digestive Endoscopy 2014; 26 (Suppl. 1): 43–51

A B C

Figure 6 Intestinal-type intramucosal gastric cancer. (a) White-light image. (b) Mode 2 image. (c) Mode 1 with magnification image.

A B C

Figure 7 Laterally spreading-type intramucosal colon cancer. (a) White-light image. (b) Mode 2 image. (c) Mode 1 with magnification
image.

Birmingham, USA. Mitsuhiro Fujishiro has served as a lec- 5 Kiesslich R, von Bergh M, Hahn M. Chromoendoscopy
turer for Eisai Pharmaceuticals Co., Ltd and his department with indigocarmine improves the detection of adenomatous and
has obtained a donation from Dainihon Sumitomo Pharma- nonadenomatous lesions in the colon. Endoscopy 2001; 33:
ceuticals Co., Ltd. He is also a chief investigator in a coop- 1001–6.
6 Pohl J, Schneider A, Vogell H, Mayer G, Kaiser G, Ell C.
erative study between The University of Tokyo and HOYA
Pancolonic chromoendoscopy with indigo carmine versus stan-
Corporation regarding optical enhanced technology. The
dard colonoscopy for detection of neoplastic lesions: A ran-
other authors have no relevant conflicts of interest to disclose. domised two-centre trial. Gut 2011; 60: 485–90.
7 Kiesslich R, Fritsch J, Holtmann M et al. Methylene blue-aided
chromoendoscopy for the detection of intraepithelial neoplasia
REFERENCES
and colon cancer in ulcerative colitis. Gastroenterology 2003;
1 Menees SB, Kim HM, Elliott EE, Mickevicius JL, Graustein 124: 880–8.
BB, Schoenfeld PS. The impact of fair colonoscopy preparation 8 Inoue H, Rey JF, Lightdale C. Lugol chromoendoscopy for
on colonoscopy use and adenoma miss rates in patients under- esophageal squamous cell cancer. Endoscopy 2001; 33: 75–
going outpatient colonoscopy. Gastrointest. Endosc. 2013; 78: 9.
510–6. 9 Yoshikawa I, Yamasaki M, Yamasaki T, Kume K, Otsuki M.
2 Leufkens AM, van Oijen MG, Vleggaar FP, Siersema PD. Lugol chromoendoscopy as a diagnostic tool in so-called
Factors influencing the miss rate of polyps in a back-to-back endoscopy-negative GERD. Gastrointest. Endosc. 2005; 62:
colonoscopy study. Endoscopy 2012; 44: 470–5. 698–703.
3 Mönkemüller K, Fry LC, Zimmermann L, Mania A, Zabielski 10 Kiesslich R, Hahn M, Herrmann G, Jung M. Screening for
M, Jovanovic I. Advanced endoscopic imaging methods for specialized columnar epithelium with methylene blue: Chro-
colon neoplasia. Dig. Dis. 2010; 28: 629–40. moendoscopy in patients with Barrett’s esophagus and a
4 Neumann H, Neurath MF, Mudter J. New endoscopic normal control group. Gastrointest. Endosc. 2001; 53: 47–
approaches in IBD. World J. Gastroenterol. 2011; 17: 63–8. 52.

© 2013 The Authors


Digestive Endoscopy © 2013 Japan Gastroenterological Endoscopy Society
Digestive Endoscopy 2014; 26 (Suppl. 1): 43–51 i-scan: Optical enhancement technology 51

11 Horwhat JD, Maydonovitch CL, Ramos F et al. A randomized 27 Masci E, Mangiavillano B, Crosta C et al. Interobserver agree-
comparison of methylene blue-directed biopsy versus conven- ment among endoscopists on evaluation of polypoid colorectal
tional four-quadrant biopsy for the detection of intestinal meta- lesions visualized with the Pentax i-Scan technique. Dig. Liver
plasia and dysplasia in patients with long-segment Barrett’s Dis. 2013; 45: 207–10.
esophagus. Am. J. Gastroenterol. 2008; 103: 546–54. 28 Testoni PA, Notaristefano C, Di Leo M, Vailati C, Mazzoleni G,
12 Kawahara Y, Takenaka R, Okada H et al. Novel chromoendo- Viale E. High-definition with i-Scan gives comparable accuracy
scopic method using an acetic acid-indigocarmine mixture for for detecting colonic lesions by non-expert and expert endosco-
diagnostic accuracy in delineating the margin of early gastric pists. Dig. Liver Dis. 2013; 45: 481–6.
cancers. Dig. Endosc. 2009; 21: 14–9. 29 Hoffman A, Sar F, Goetz M et al. High definition colonoscopy
13 Kiesslich R, Mergener K, Naumann C et al. Value of chromo- combined with i-Scan is superior in the detection of colorectal
endoscopy and magnification endoscopy in the evaluation of neoplasias compared with standard video colonoscopy: A pro-
duodenal abnormalities: A prospective, randomized compari- spective randomized controlled trial. Endoscopy 2010; 42: 827–
son. Endoscopy 2003; 35: 559–63. 33.
14 Kiesslich R, Sanduleanu S (eds). I-Scan Mini-Atlas. Hamburg: 30 Testoni PA, Notaristefano C, Vailati C, Di Leo M, Viale E.
Pentax Europe, 2011. High-definition colonoscopy with i-Scan: Better diagnosis for
15 Kodashima S, Fujishiro M. Novel image-enhanced endoscopy small polyps and flat adenomas. World J. Gastroenterol. 2012;
with i-scan technology. World J. Gastroenterol. 2010; 16: 18: 5231–9.
1043–9. 31 Hong SN, Choe WH, Lee JH et al. Prospective, randomized,
16 Vieth M. Structural abnormalities of endoscopy-negative reflux back-to-back trial evaluating the usefulness of i-SCAN in
disease – real or perceived? Digestion 2008; 78: 24–30. screening colonoscopy. Gastrointest. Endosc. 2012; 75: 1011–
17 Hoffman A, Basting N, Goetz M et al. High-definition endos- 21.
copy with i-Scan and Lugol’s solution for more precise detec- 32 Hoffman A, Kagel C, Goetz M et al. Recognition and charac-
tion of mucosal breaks in patients with reflux symptoms. terization of small colonic neoplasia with high-definition colo-
Endoscopy 2009; 41: 107–12. noscopy using i-Scan is as precise as chromoendoscopy. Dig.
18 Kang HS, Hong SN, Kim YS et al. The efficacy of i-SCAN for Liver Dis. 2010; 42: 45–50.
detecting reflux esophagitis: A prospective randomized con- 33 Pigò F, Bertani H, Manno M et al. i-Scan high-definition white
trolled trial. Dis. Esophagus 2013; 26: 204–11. light endoscopy and colorectal polyps: Prediction of histology,
19 Kim MS, Choi SR, Roh MH et al. Efficacy of I-scan endoscopy interobserver and intraobserver agreement. Int. J. Colorectal
in the diagnosis of gastroesophageal reflux disease with Dis. 2013; 28: 399–406.
minimal change. Clin. Endosc. 2011; 44: 27–32. 34 Geboes K, Ectors N, D’Haens G, Rutgeerts P. Is ileoscopy with
20 Kodashima S, Fujishiro M, Koike K. Early detection of gastric biopsy worthwhile in patients presenting with symptoms of
cancer by i-scan and chromoendoscopy. Endosc. Dig. 2011; 23: inflammatory bowel disease? Am. J. Gastroenterol. 1998; 93:
733–9. 201–6.
21 Kodashima S, Goto O, Ono S et al. Usefulness of i-scan (TE-g) 35 Powell-Tuck J, Day DW, Buckell NA, Wadsworth J,
for diagnosis of lateral extent in gastric neoplasms. J. New. Rem. Lennard-Jones JE. Correlations between defined sigmoido-
Clin. 2009; 58: 1409–13. scopic appearances and other measures of disease activity in
22 Cammarota G, Ianiro G, Sparano L et al. Image-enhanced ulcerative colitis. Dig. Dis. Sci. 1982; 27: 533–7.
endoscopy with I-scan technology for the evaluation of duode- 36 Yantiss RK, Odze RD. Pitfalls in the interpretation of nonneo-
nal villous patterns. Dig. Dis. Sci. 2013; 58: 1287–92. plastic mucosal biopsies in inflammatory bowel disease. Am. J.
23 Neumann H, Neufert C, Vieth M, Siebler J, Mönkemüller K, Gastroenterol. 2007; 102: 890–904.
Neurath MF. High-definition endoscopy with i-scan enables 37 Neurath MF, Travis SP. Mucosal healing in inflammatory bowel
diagnosis of characteristic mucosal lesions in Whipple’s diseases: A systematic review. Gut 2012; 61: 1619–35.
disease. Endoscopy 2012; 44: 217–8. 38 Neumann H, Vieth M, Günther C et al. Virtual chromoendos-
24 Mönkemüller K, Fry LC, von Arnim U, Neumann H, Evert M, copy for prediction of severity and disease extent in patients
Malfertheiner P. Whipple’s disease: An endoscopic and histo- with inflammatory bowel disease: A randomized controlled
logic study. Digestion 2008; 77: 161–5. study. Inflamm. Bowel Dis. 2013; 19: 1935–42.
25 Rex DK, Kahi C, O’Brien M et al. The American Society for 39 Neumann H, Vieth M, Neurath MF, Zopf Y. High-definition
Gastrointestinal Endoscopy PIVI (Preservation and Incorpora- endoscopy with virtual chromoendoscopy for prediction of food
tion of Valuable Endoscopic Innovations) on real-time endo- allergy in real-time – an observational study. Gastrointest.
scopic assessment of the histology of diminutive colorectal Endosc. 2013; 77: 556–7.
polyps. Gastrointest. Endosc. 2011; 73: 419–22. 40 Kodashima S, Fujishiro M, Ono S et al. Evaluation of a new
26 Neumann H, Vieth M, Fry LC et al. Learning curve of virtual image-enhanced endoscopic technology employing band-
chromoendoscopy for the prediction of hyperplastic and adeno- limited light for detection of esophageal squamous cell carci-
matous colorectal lesions: A prospective 2-center study. Gas- noma. Dig. Endosc. 2013 29 Apr. doi: 10.1111/den.12108.
trointest. Endosc. 2013; 78: 115–20.

© 2013 The Authors


Digestive Endoscopy © 2013 Japan Gastroenterological Endoscopy Society

Potrebbero piacerti anche