AN UMET MEDICAL NEED: ADVANCES IN ENDOSCOPIC …



An unmet medical need: Advances in endoscopic imaging of colorectal neoplasia

1Andreas Stallmach, 1Carsten Schmidt, 3Alastair Watson, 2Ralf Kiesslich

1 Division of Gastroenterology, Hepatology and Infectious Diseases, Department of Internal Medicine II, Jena University Hospital

2 I. Medizinische Klinik, Universitätsklinikum, Johannes-Gutenberg Universität, Mainz

3 Norwich Medical School, University of East Anglia, Norwich

Corresponding author:

Andreas Stallmach

Division of Gastroenterology, Hepatology and Infectious Diseases, Department of Internal Medicine II, Friedrich Schiller University of Jena, Erlanger Allee 101, 07740 Jena, Germany.

Tel: +49-3641-9324221, Fax: +49-3641-9324222

E-mail-address: andreas.stallmach@med.uni-jena.de

Abstract

Gastrointestinal cancer is a major public health problem worldwide. Detection of early neoplastic lesions in gastrointestinal tract is essential for cure, because prognosis and survival are related to the size and stage of malignant lesions. Endoscopic screening and treatment of polyps could prevent approximately 80% of colorectal cancer (CRC). However, white light endoscopy is an imperfect technology since miss rates of up to 25% have been reported and polyps without malignant potential were treated without benefit but with additional costs and risks to the patient.  There are several known “human” predictors of an inadequate colonoscopy. These include patient characteristics such as poor bowel preparation, female gender, or inpatient status. Skills of the endoscopists are an important issue, as well. Therefore, a variety of advanced technologies has been attempted to overcome these issues. These new endoscopic imaging techniques allow a more precise classification of mucosal alterations with selection of patients for invasive therapy or surveillance. Further, molecular and functional imaging techniques could identify novel targets for therapies and new prospects to access response to therapies. However, at the “end of the day” a better endoscopic approach for CRC screening and surveillance depends on a good bowel preparation, a trained endoscopist spending sufficient time on a detailed examination together with an advanced endoscope.

“Healing is a matter of time, but it is sometimes also a matter of opportunity."— Hippocrates

Malignant disease is the second most frequently cause of death in Europe and the North America. Despite all previous prevention campaigns the Deutsche Krebshilfe (“German Cancer Aid”) forecasts that the annual incidence rate of cancer patients worldwide will increase to 15.7 million until 2020. It has been estimated that CRC accounted for 142.570 new cancer cases and 51.370 cancer deaths in the United States in 2010 [1]. The majority of CRC develops from normal tissue, followed by adenomatous polyps, which evolve from pre-invasive, to invasive and finally to metastatic carcinomas. In addition 10-15% of sporadic CRCs would have their origin in serrated polyps. These lesions include hyperplastic-type aberrant crypt foci, hyperplastic polyps, sessile serrated adenomas, admixed polyps and serrated adenomas, and constitute the so-called 'serrated pathway', which is characterized by early involvement of oncogenic BRAF mutations, excess CpG island methylation and subsequent low- or high-level DNA microsatellite instability (for review see [2]). Perhaps 30-50 % of the German population will develop adenomatous polyps during their life; about 7% of these people will develop an apparent carcinoma. Several data indicate that more than 2/3 of the persons could be retrieved through a consistent early diagnosis of CRC [3] [4] [5].

The central requirement for healing of tumour patients is an early diagnosis; only then are time frames for curative treatments opened. For gastrointestinal tumours, particularly the CRC, endoscopic examinations are established as diagnostic standard for early detection. The efficiency of the screening colonoscopy for early detection of CRC and consequently decreased CRC-associated mortality has been proven through a multiplicity of clinical studies. In addition to patients with a sporadic CRC other groups of patients, who have a higher risk for colon cancer, can be identified, i.e. patients with chronic inflammatory bowel disease (IBD) (ulcerative colitis and Crohn’s Disease), first-degree relatives of patients with colon carcinoma and a range of genetic determinate familial syndromes [6] [7]. In IBD, according to recent research invasive carcinoma develops also out of inflammatory diseases in line with a malignant transformation from intraepithelial neoplasias (IEN)/dysplasias [8] [9]. The early detection of an IEN/dysplasia identifies high risk patients and is therefore important for the prognosis of the patients.

The Problem

Endoscopic screening doubtlessly reduces CRC-related mortality [10]. Two previously - non-randomized - published trials showed that the incidence of CRC may be decreased by polypectomy by 66 - 90%. A randomized controlled trial in UK demonstrated that only one flexible sigmoidoscopy screening between 55 and 64 years of age resulted in a reduced incidence of colorectal cancer by 33% (0.67, 0.60-0.76) and mortality by 43% [11]. Therefore colonoscopy seems to be well accepted by the experts of the guideline conference from 2004 with high sensitivity and specificity rates for adenomas and carcinomas - especially for neoplasias localized in the proximal colon. However, white light endoscopy is an imperfect technology, since adenomas, the premalignant condition, can be missed during colonoscopy and polyps without malignant potential can be removed without benefit but additional costs and increased risk to patients [12].  Critical studies document, that up to 25% of the polyps in the colon will be missed as possible precancerous lesions by the classic surveillance endoscopy [13] [14] [15]. Interval carcinomas can also develop after an apparently “clear colonoscopy” in which significant neoplastic lesions were not recognized. Robertson et al. assumed that about 50% of the interval carcinomas were missed at the initial colonoscopy. Further, approximately 25% were classified as a relapse of an adenomatous polyp with incomplete polypectomy and about 25 % were judged as fast-growing de novo-carcinomas [16]. In patients with IBD this problem is even greater. In these diseases, which come along with a high risk of malignant transformation, the identification of inflammation associated neoplastic mucosal alterations during the colonoscopy is difficult, because these alterations can be found in macroscopic normal as well as in inflamed mucosa. In many cases non-neoplastic inflammatory alterations can be found, e.g. so-called pseudopolyps or inflammatory, which don’t have necessarily malignant potential. White light colonoscopy is not able to readily differentiate between chronic inflammation, regenerative changes or flat growing neoplasia.

In summary, it would be preferable pre-neoplastic lesions were detected with techniques with the highest sensitivity using a “red flag technology” and afterwards to classifying these lesions according to their malignant potential (“detect and characterize”).

The Patient

As the rate of colonoscopic examinations in respect of annually performed screening procedures has increased, so has the total number of inadequate or incomplete colonoscopy. Patient factors that have been identified to be associated with insufficient colonoscopy are female sex, increasing patient age, diverticular disease, and history of abdominal surgery. Additional factors that have been reported are a low body mass index, history of constipation, or laxative use [17] [18] [19]. Much of endoscopy training is aimed at ourvercoming these unfavourable feature so that all patients recieve and high quality, safe colonoscopy. Among these, poor bowel preparation is the most consistently reported factor predictive of an inadequate colonoscopy. Since diagnostic accuracy of colonoscopy strictly depends upon the quality of bowel cleansing, the ideal preparation for colonoscopy should reliably empty the large intestine from all faecal material allowing the optimal visualization of the mucosa without causing great patient's discomfort. However, bowel preparation is very often perceived as the most unpleasant part of the procedure in individuals undergoing screening colonoscopy. Data from a recent study showed a good correlation between colon cleanliness and detection rate of adenomas [20]. So a sufficiently cleaned colon using standard preparation techniques is the conditio sine qua non of an informative endoscopy. In a recently published study describing 8328 colonoscopies, 1243 (15%) were judged as be inadequate because of worst/absent intestinal toilette [21]. Therefore, there is an urgent need for increased patient education for adequate bowel preparation to permit a higher rate of sufficient colonoscopies. Currently, there are three different new medical devices available, which allow improved intraprocedural bowl cleansing [22] [23] [24]. It can be speculated that these devices will facilitate effective screening and surveillance colonoscopy.

The Endoscopist

Besides patient associated criteria, the skill of the colonoscopist has a major influence on specificity and sensitivity of the procedure to detect colorectal neoplasias. It is very important to detect all neoplasias, to recognize these as such and not to misinterpretate them. Already basic factors like colonoscopy withdrawal time influence the quality of the endoscopy. Toruner and co-workers demonstrated a significant correlation between examination time and flat dysplasia detection rate [25]. Higher rates of detecting adenomas were observed among endoscopists who had a longer colonoscopy withdrawal time (at least 6 minutes). Further, not only time but timing is relevant for high detections rates of colorectal neoplasia. Sanaka et al. described significantly higher adenoma detection rates in the morning (29.3%) compared with the afternoon (25.3%). There was a significant decline in adenoma detection rate with each subsequent hour of the day. Endoscopist fatigue is postulated as a factor leading to lower polyp detection rates in the afternoon compared with the morning [26].

Another important determinant of the success of the colonoscopy is the experience of the endoscopist (for review see [27]). Sonnenberg summarizes the problems of experience, education and visual cognition in his excellent short statement “We only see what we already know” [28]. Therefore, a good medical training and the learning how pathological findings could be detected are of great importance. Different education and sensitivity for small changes may result in the major differences between western and eastern endoscopists in how they interpret the shape of lesions of the colorectal mucosa. In Japan there will be detected clearly more preliminary stages and early stage carcinomas as in Europe or North America. Concerning this there were marked differences in determination of flat lesions, depressed lesions and those deemed "no polyp”.

The problem of failed detection of small or flat lesions can be minimized by using enhanced imaging technologies.

The Endoscope (enhanced imaging technologies)

A variety of new technologies have been attempted to overcome these issues based on advances in physics and biomedical engineering, these new technologies of endoscopic imaging include

• (virtual) chromoendoscopy

• autofluorescence

• light-scattered spectroscopy (LSS)

• Raman spectroscopy

• confocal laser endoscopy (cLE)

Since a recently published review [29] describe these technologies in detail, we will summarize only some aspects and discuss impacts on clinical strategies to improve quality of colonoscopic examination, both in diagnostic or surveillance settings.

(Virtual) chromoendoscopy

In chromoendoscopy, intravital dyes like indigo carmine or methylene blue are topically applied onto the mucosal surface to enhance superficial patterns and contrast of pathologic versus normal mucosa. Kudo et al. firstly described the value of chromoendoscopy combined with magnifying endoscopy, which unmask surface “pit-pattern” structure, which can be used to predict histology [30]. Chromoendoscopy is generally used in a targeted fashion to detect flat sporadic adenoma. However, in long lasting ulcerative colitis pan-chromoendoscopy is recommended [29].

There is convincing evidence from a multitude of studies that the detection of flat, circumscribed colitis-associated neoplasias is enhanced in patients with long-standing ulcerative colitis by a factor of 3 to 5 [24] [31]. The most recent ECCO guideline endorse chromoendoscopy as the better alternative as standard colonoscopy with random biopsies [32].

The success of chromoendoscopy has triggered research activities towards “virtual chromoendoscopy” by new light filters, such as narrow band imaging (NBI) or post processing techniques, such as i-scan or Fujinon intelligent chromoendoscopy which mimic chromoendoscopy.

NBI, FICE and i-scan could not show an enhanced adenoma detection rate compared to white light endoscopy [33] (see also figure 1). Probably high-definition per se contributed to high adenoma detection rates, but virtual chromoendoscopy may induce a learning effect even in experienced endoscopists as to the appreciation of flat adenomas in WLE [34]. However, virtual chromoendoscopy can be used to characterize colorectal lesions as precisely as chromoendoscopy [33].

Autofluorescence (AFI)

After exposure of tissues to light of a defined short wavelength, excitation of endogenous fluorophores results in the emission of light of a longer wavelength, which is termed autofluorescence. Due to changes of endogenous fluorophores in dysplastic tissue, the altered autofluorescence spectrum can be translated into a pseudo-coloured image in which normal tissue appears greenish in contrast to a purple rendering of dysplastic tissue.

AFI has been combined with high-resolution endoscopy and NBI in a single endoscope with 2 charge-coupled devices, a combination termed endoscopic trimodal imaging. AFI images are composed of total emitted autofluorescence after blue light excitation (390-470 nm) and green reflectance (540-560 nm). In screening colonoscopy, AFI was not able to significantly enhance the diagnostic yield in a randomized tandem colonoscopy trial on 100 patients [35]. The adenoma miss rates of AFI and high-definition endoscopy were 20% and 29%, respectively. In contrast, a study on 50 patients with ulcerative colitis from the same center found a significantly lower neoplasia miss rate for AFI than for WLE (0% vs 50%; P=.036), and every intraepithelial neoplasia was coloured purple on AFI (sensitivity 100%) [36]. However, despite promising results to date autofluorescence has not enhanced diagnostic yield and further research is required.

Light-scattered spectroscopy (LSS)

Light scattering spectroscopy is a type of reflectance spectroscopy that determines tissue structures by examining elastic scattering. Elastic light scattering is a process in which the illumination light does not undergo a shift in wavelength during the scattering event. Each wavelength of the illumination light is scattered to a different degree by dense structures in the tissue, such as nuclei. A portion of the light incident on the tissue is backscattered, or scattered directly in the backward direction, and can be collected by the fiberoptic probe that delivered the light to the tissue or a distinct collection optical fiber. Because no energy is lost during these scattering events and, therefore, no wavelength shift occurs, this type of spectroscopy is termed elastic. By analyzing the varying degree to which individual wavelengths are backscattered across a spectral range, it is possible to determine the size, number, and optical density of the dense structures within a tissue [29].

This light scattering information can be utilized to detect developing abnormalities in a tissue. For example, neoplastic tissue, which is characterized by enlarged nuclei, can be accurately detected at its earliest stages by elastic scattering spectroscopy. Even the field defect within the colon can be analyzed at any site within the colon if the colon harbours somewhere else cancer or advanced adenomas. The field defect is detected based on micro skeleton and capillary changes and was proven in animal and human studies [37].

Raman Spectroscopy

New molecular based techniques for the detection of protein profiles, such as Raman imaging spectroscopy have an enormous potential for the detection of molecular information from cells. Raman spectroscopy is a technique used to study vibrational and rotational conditions of molecules and relies on the inelastic scattering of monochromatic light (photons) on molecules. Light of a known frequency and polarization interacts with the sample and the scattered light is then analyzed for its frequency and polarization, which provides information on the characteristics of the sample [38]. The shifts in wavelengths are expressed as wave numbers and correspond to specific vibrations or rotations of particular molecular bonds. Tissues contain biomolecules that have distinct vibrational energies in their molecular bonds. Raman detects molecular components of tissue in a qualitative and quantitative way and subtle changes at a molecular level can be examined. In gastrointestinal oncology, Raman has been investigated as a diagnostic tool for characterizing cancer cells and early malignant changes. Raman spectroscopy has been used on single cells [39] and tissue [40] [41], and has been applied in vivo towards better diagnostics for dysplastic diseases in humans. Very recently, Michael A. Short and coworkers presented at the Digestive Disease Week 2011 in Chicago an Endoscopic Raman Spectroscopy System for the improving detection of early colonic neoplasias. They demonstrated that malignant ad benign tissues could be separated with 100% sensitivity and 89% specificity based on the high frequency region spectra. Even a simple comparison of ratios of lipid and protein peaks had a significant discrimination power [42]. Nevertheless future intensive research work and clinical studies are necessary before this promising technique could get clinical value.

Confocal laser endoscopy (CLE)

Confocal laser endomicroscopy (CLE) can be achieved with two different endoscopic models. First, a miniaturized confocal scanner has been integrated into the distal tip of a flexible endoscope [43]. Second is the use of confocal microscopy miniprobes. These probes can be fitted through the working channel of most endoscopes for clinical use [44]. Endomicroscopy with both techniques provides in vivo histology during ongoing endoscopy at subcellular resolution.

CLE mandates use of fluorescent agents. Most studies in humans have been performed with intravenous fluorescein sodium. Fluorescein quickly distributes within all compartments of the tissue, and CLE is possible within seconds after injection. It contrasts cellular and subcellular details, connective tissue, and vessel architecture at high resolution, but does not stain nuclei. Acriflavine as an alternative or additional contrast medium is applied topically to the colonic mucosa and predominantly stains nuclei and, to a lesser extent, cytoplasm [29].

Multiple trials have proven the high accuracy with which CLE can differentiate non-neoplastic from neoplastic tissue by using fluorescein aided endomicroscopy and simplified endomicroscopic criteria have been established [45] , [45] (see figure 2).

The combination of topical acriflavine and systemic fluorescein administration also allowed reliable differentiation of low-grade from high-grade intraepithelial neoplasia with high accuracy. Endomicroscopy is well suited for patients with high risk to develop colon cancer. In patients with long lasting ulcerative colitis, chromoendoscopy in combination with endomicroscopy enabled to significantly increase the diagnostic yield of intraepithelial neoplasias [24].

Molecular Imaging with Endomicroscopy

Currently, only a limited number of fluorescent staining protocols have been evaluated for CLE in humans, and new protocols are just being tested. Animal models do not face such constraints, and many of the staining protocols used in bench top confocal microscopy can be transferred to in vivo imaging. Even molecular imaging, that is, the fluorescent labeling of individual cells by their molecular signature, has been possible in vivo.

Molecular targeting of somatostatin receptor-positive tumor cells in vivo was demonstrated in a nude mouse xenograft model after injection of a carboxyfluorescein-labeled ligand [46]. More recently, differentiation of tumor cells based on their epithelial growth factor receptor and vascular epithelial growth factor expression patterns was achieved in vivo in a rodent model with human colorectal cancer xenografts [46]. In humans, the targeted labeling of neoplastic crypts has been possible during colonoscopy using a labeled heptapeptide derived from a phage library, showing a sensitivity and specificity on neoplastic lesions thereby coming close to in vivo immunohistochemistry [47].

A Short Summary and Perspectives

With the increasing demand for colonoscopy for CRC screening and surveillance of patients at risk, there as been an increasing interest in quality of colonoscopy. The “bottom line” for endoscopic detection of pre-neoplastic or neoplastic alterations depends on a good bowel preparation, and an experienced endoscopist spending sufficient time on a detailed examination. However, even under ideal conditions, conventional colonoscopy still fails to detect small neoplastic alterations, particularly in inflamed tissue. Several of novel technologies have the potential to improve detection rates of colorectal neoplasia in surveillance programs and in patients at increased risk. Image enhancement methods such as autofluorescent endoscopy or narrow-band-imaging have the potential to act as “red flat techniques” which enhance the recognition of mucosal alternations. The application of molecular specific stains such as fluorescent monoclonal antibodies or fluorophores should further increase the accuracy of these technologies. However, intensive research is necessary to evaluate sensitivity, specificity and most important safety of these new agents. These new technologies must be both effective and efficient in order to be incorporated on a widespread basis in an already economically stressed health system.

References

[1] A. Jemal, R. Siegel, J. Xu and E. Ward, CA Cancer J Clin 60, 277-300 (2011).

[2] M. J. Makinen, Histopathology 50, 131-150 (2007).

[3] S. J. Winawer, A. G. Zauber, M. N. Ho, M. J. O'Brien, L. S. Gottlieb, S. S. Sternberg, J. D. Waye, M. Schapiro, J. H. Bond, J. F. Panish and et al., N Engl J Med 329, 1977-1981 (1993).

[4] E. Thiis-Evensen, G. S. Hoff, J. Sauar, F. Langmark, B. M. Majak and M. H. Vatn, Scand J Gastroenterol 34, 414-420 (1999).

[5] F. Citarda, G. Tomaselli, R. Capocaccia, S. Barcherini and M. Crespi, Gut 48, 812-815 (2001).

[6] C. Schmidt, C. Bielecki, J. Felber and A. Stallmach, Minerva Gastroenterol Dietol 56, 189-201 (2010).

[7] A. Stallmach, C. Bielecki and C. Schmidt, Dig Dis 27, 584-590 (2009).

[8] H. E. Blum, Eur J Cancer 31A, 1369-1372 (1995).

[9] J. Xie and S. H. Itzkowitz, World J Gastroenterol 14, 378-389 (2008).

[10] B. Levin, D. A. Lieberman, B. McFarland, K. S. Andrews, D. Brooks, J. Bond, C. Dash, F. M. Giardiello, S. Glick, D. Johnson, C. D. Johnson, T. R. Levin, P. J. Pickhardt, D. K. Rex, R. A. Smith, A. Thorson and S. J. Winawer, Gastroenterology 134, 1570-1595 (2008).

[11] W. S. Atkin, R. Edwards, I. Kralj-Hans, K. Wooldrage, A. R. Hart, J. M. Northover, D. M. Parkin, J. Wardle, S. W. Duffy and J. Cuzick, Lancet 375, 1624-1633 (2011).

[12] L. Rabeneck, L. F. Paszat, R. J. Hilsden, R. Saskin, D. Leddin, E. Grunfeld, E. Wai, M. Goldwasser, R. Sutradhar and T. A. Stukel, Gastroenterology 135, 1899-1906, 1906 e1891 (2008).

[13] L. J. Hixson, M. B. Fennerty, R. E. Sampliner, D. McGee and H. Garewal, J Natl Cancer Inst 82, 1769-1772 (1990).

[14] D. K. Rex, C. S. Cutler, G. T. Lemmel, E. Y. Rahmani, D. W. Clark, D. J. Helper, G. A. Lehman and D. G. Mark, Gastroenterology 112, 24-28 (1997).

[15] J. C. van Rijn, J. B. Reitsma, J. Stoker, P. M. Bossuyt, S. J. van Deventer and E. Dekker, Am J Gastroenterol 101, 343-350 (2006).

[16] D. J. Robertson, E. R. Greenberg, M. Beach, R. S. Sandler, D. Ahnen, R. W. Haile, C. A. Burke, D. C. Snover, R. S. Bresalier, G. McKeown-Eyssen, J. S. Mandel, J. H. Bond, R. U. Van Stolk, R. W. Summers, R. Rothstein, T. R. Church, B. F. Cole, T. Byers, L. Mott and J. A. Baron, Gastroenterology 129, 34-41 (2005).

[17] G. Dafnis, F. Granath, L. Pahlman, A. Ekbom and P. Blomqvist, Dig Liver Dis 37, 113-118 (2005).

[18] J. C. Anderson, C. R. Messina, W. Cohn, E. Gottfried, S. Ingber, G. Bernstein, E. Coman and J. Polito, Gastrointest Endosc 54, 558-562 (2001).

[19] W. C. Cirocco and L. C. Rusin, Dis Colon Rectum 38, 964-968 (1995).

[20] S. Thomas-Gibson, P. Rogers, S. Cooper, R. Man, M. D. Rutter, N. Suzuki, D. Swain, A. Thuraisingam and W. Atkin, Endoscopy 38, 456-460 (2006).

[21] G. Geraci, F. Pisello, G. Modica, F. Li Volsi, T. Facella, G. Romeo, S. Maggio and C. Sciume, G Chir 28, 227-231 (2007).

[22] M. Moshkowitz, Y. Hirsch, I. Carmel, T. Duvdevany, I. Fabian, E. Willenz and J. Cohen, Endoscopy 42, 834-836 (2010).

[23] I. Juriene, J. Rigaux and J. M. Deviere, Gastrointestinal Endoscopy 71, AB157-AB157 (2010).

[24] R. Kiesslich, M. Goetz, K. Lammersdorf, C. Schneider, J. Burg, M. Stolte, M. Vieth, B. Nafe, P. R. Galle and M. F. Neurath, Gastroenterology 132, 874-882 (2007).

[25] M. Toruner, G. C. Harewood, E. V. Loftus, Jr., W. J. Sandborn, W. J. Tremaine, W. A. Faubion, K. W. Schroeder and L. J. Egan, Inflamm Bowel Dis 11, 428-434 (2005).

[26] M. R. Sanaka, N. Shah, K. D. Mullen, D. R. Ferguson, C. Thomas and A. J. McCullough, Am J Gastroenterol 101, 2726-2730 (2006).

[27] R. Lambert, M. Jeannerod and J. F. Rey, Endoscopy 36, 723-725 (2004).

[28] A. Sonnenberg, Gastroenterology 124 Suppl 1, A 353 (2003).

[29] M. Wallace, A. Meining, M. Canto, P. Fockens, S. Miehlke, T. Roesch, C. Lightdale, H. Pohl, D. CARR LOCKE and M. Löhr, Alimentary pharmacology & therapeutics 31, 548-552 (2010).

[30] S. E. Kudo, S. Tamura, T. Nakajima, H. Yamano, H. Kusaka and H. Watanabe, Gastrointestinal Endoscopy 44, 8-14 (1996).

[31] M. Rutter, B. Saunders, G. Schofield, A. Forbes, A. Price and I. Talbot, Gut 53, 256 (2004).

[32] G. Van Assche, A. Dignass, W. Reinisch, C. J. van der Woude, A. Sturm, M. De Vos, M. Guslandi, B. Oldenburg, I. Dotan and P. Marteau, Journal of Crohn's and Colitis 4, 63-101 (2010).

[33] J. Sauk, A. Hoffman, S. Anandasabapathy and R. Kiesslich, Gastroenterology Clinics of North America (2010).

[34] A. Adler, H. Pohl, I. S. Papanikolaou, H. Abou-Rebyeh, G. Schachschal, W. Veltzke-Schlieker, A. C. Khalifa, E. Setka, M. Koch, B. Wiedenmann and T. Rosch, Gut 57, 59-64 (2008).

[35] F. J. C. Van Den Broek, E. J. Van Soest, A. H. Naber, A. H. A. M. Van Oijen, R. C. Mallant-Hent, C. J. M. Böhmer, P. Scholten, P. C. F. Stokkers, W. A. Marsman and E. M. H. Mathus-Vliegen, The American journal of gastroenterology 104, 1498-1507 (2009).

[36] F. J. C. van den Broek, P. Fockens, S. van Eeden, J. B. Reitsma, J. C. H. Hardwick, P. C. F. Stokkers and E. Dekker, Gut 57, 1083 (2008).

[37] V. Backman and H. K. Roy, Gastroenterology 140, 35-41. e35 (2011).

[38] P. Matousek and N. Stone, Analyst 134, 1058-1066 (2009).

[39] K. Chen, Y. Qin, F. Zheng, M. Sun and D. Shi, Opt Lett 31, 2015-2017 (2006).

[40] E. Widjaja, W. Zheng and Z. Huang, Int J Oncol 32, 653-662 (2008).

[41] A. Beljebbar, O. Bouche, M. D. Diebold, P. J. Guillou, J. P. Palot, D. Eudes and M. Manfait, Crit Rev Oncol Hematol 72, 255-264 (2009).

[42] M. A. Short, S. F. Lam, D. A. Owen, C. MacAulay, I. T. Tai and H. Zeng, Development of an endoscopic raman spectroscopy system for theimproving detection of early colonic neoplasia, in Digestive Disease Week, Chicago (2011).

[43] R. Kiesslich, J. Burg, M. Vieth, J. Gnaendiger, M. Enders, P. Delaney, A. Polglase, W. McLaren, D. Janell and S. Thomas, Gastroenterology 127, 706-713 (2004).

[44] M. B. Wallace and P. Fockens, Respiratory Care Clinics of North America 136, 1509-1513 (2009).

[45] A. M. Buchner, M. W. Shahid, M. G. Heckman, M. Krishna, M. Ghabril, M. Hasan, J. E. Crook, V. Gomez, M. Raimondo and T. Woodward, Gastroenterology 138, 834-842 (2010).

[46] C. Fottner, E. Mettler, M. Goetz, E. Schirrmacher, M. Anlauf, D. Strand, R. Schirrmacher, G. Kloppel, P. Delaney and M. Schreckenberger, Endocrinology 151, 2179 (2010).

[47] P. L. Hsiung, J. Hardy, S. Friedland, R. Soetikno, C. B. Du, A. P. Wu, P. Sahbaie, J. M. Crawford, A. W. Lowe and C. H. Contag, Nature medicine 14, 454-458 (2008).

................
................

In order to avoid copyright disputes, this page is only a partial summary.

Google Online Preview   Download