51
|
Ma C, Li C, Jiang D, Gao X, Han J, Xu N, Wu Q, Nie G, Chen W, Lin F, Hou Y. Screening of a specific peptide binding to esophageal squamous carcinoma cells from phage displayed peptide library. Mol Cell Probes 2015; 29:182-9. [PMID: 25886968 DOI: 10.1016/j.mcp.2015.04.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 04/04/2015] [Accepted: 04/06/2015] [Indexed: 01/22/2023]
Abstract
To select a specifically binding peptide for imaging detection of human esophageal squamous cell carcinoma (ESCC), a phage-displayed 12-mer peptide library was used to screen the peptide that bind to ESCC cells specifically. After four rounds of bio-panning, the phage recovery rate gradually increased, and specific phage clones were effectively enriched. The 60 randomly selected phage clones were tested using cellular enzyme-linked immunosorbent assay (ELISA), and 41 phage clones were identified as positive clones with the over 2.10 ratio of absorbance higher than other clones, IRP and PBS controls. From the sequencing results of the positive clones, 14 peptide sequences were obtained and ESCP9 consensus sequence was identified as the peptide with best affinity to ESCC cells via competitive inhibition, fluorescence microscopy, and flow cytometry. The results indicate that the peptide ESCP9 can bind to ESCC cells specifically and sensitively, and it is a potential candidate to be developed as an useful molecule to the imaging detection and targeting therapy for ESCC.
Collapse
Affiliation(s)
- Caixia Ma
- Co-Innovation Center for Qinba Region's Sustainable Development, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Chunyan Li
- Co-Innovation Center for Qinba Region's Sustainable Development, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | | | - Xiaojie Gao
- College of Life and Geography Science, Qinghai Normal University, Xining, Qinghai 810008, China
| | - Juanjuan Han
- Co-Innovation Center for Qinba Region's Sustainable Development, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Nan Xu
- Co-Innovation Center for Qinba Region's Sustainable Development, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Qiong Wu
- Co-Innovation Center for Qinba Region's Sustainable Development, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Guochao Nie
- Center of Medical Nanomaterial, Yulin Normal College, Yulin, Guangxi 537000, China
| | - Wei Chen
- Department of Physics and the SAVANT Center, The University of Texas at Arlington, Arlington, TX 76019, USA
| | - Fenghuei Lin
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
| | - Yingchun Hou
- Co-Innovation Center for Qinba Region's Sustainable Development, Shaanxi Normal University, Xi'an, Shaanxi 710062, China.
| |
Collapse
|
52
|
Kang J, Chang JH, Wilson BC, Veilleux I, Bai Y, DaCosta R, Kim K, Ha S, Lee JG, Kim JS, Lee SG, Kim SM, Lee HJ, Ahn YB, Han S, Yoo Y, Song TK. A prototype hand-held tri-modal instrument for in vivo ultrasound, photoacoustic, and fluorescence imaging. THE REVIEW OF SCIENTIFIC INSTRUMENTS 2015; 86:034901. [PMID: 25832265 DOI: 10.1063/1.4915146] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Multi-modality imaging is beneficial for both preclinical and clinical applications as it enables complementary information from each modality to be obtained in a single procedure. In this paper, we report the design, fabrication, and testing of a novel tri-modal in vivo imaging system to exploit molecular/functional information from fluorescence (FL) and photoacoustic (PA) imaging as well as anatomical information from ultrasound (US) imaging. The same ultrasound transducer was used for both US and PA imaging, bringing the pulsed laser light into a compact probe by fiberoptic bundles. The FL subsystem is independent of the acoustic components but the front end that delivers and collects the light is physically integrated into the same probe. The tri-modal imaging system was implemented to provide each modality image in real time as well as co-registration of the images. The performance of the system was evaluated through phantom and in vivo animal experiments. The results demonstrate that combining the modalities does not significantly compromise the performance of each of the separate US, PA, and FL imaging techniques, while enabling multi-modality registration. The potential applications of this novel approach to multi-modality imaging range from preclinical research to clinical diagnosis, especially in detection/localization and surgical guidance of accessible solid tumors.
Collapse
Affiliation(s)
- Jeeun Kang
- Department of Electronic Engineering, Sogang University, Seoul 121-742, South Korea
| | - Jin Ho Chang
- Department of Electronic Engineering, Sogang University, Seoul 121-742, South Korea
| | - Brian C Wilson
- Princess Margaret Cancer Centre/University Health Network, Toronto, Ontario M5G 1L7, Canada
| | - Israel Veilleux
- Princess Margaret Cancer Centre/University Health Network, Toronto, Ontario M5G 1L7, Canada
| | - Yanhui Bai
- Princess Margaret Cancer Centre/University Health Network, Toronto, Ontario M5G 1L7, Canada
| | - Ralph DaCosta
- Princess Margaret Cancer Centre/University Health Network, Toronto, Ontario M5G 1L7, Canada
| | - Kang Kim
- Center for Ultrasound Molecular Imaging and Therapeutics, Department of Medicine, University of Pittsburgh School of Medicine and Heart and Vascular Institute, University of Pittsburgh Medical Center (UPMC), Pittsburgh, Pennsylvania 15213, USA
| | - Seunghan Ha
- Center for Ultrasound Molecular Imaging and Therapeutics, Department of Medicine, University of Pittsburgh School of Medicine and Heart and Vascular Institute, University of Pittsburgh Medical Center (UPMC), Pittsburgh, Pennsylvania 15213, USA
| | - Jong Gun Lee
- GE Ultrasound Korea, Seongnam 462-807, South Korea
| | | | | | - Sun Mi Kim
- Department of Radiology, Seoul National University of Bundang Hospital, Kyonggi-do, South Korea
| | - Hak Jong Lee
- Department of Radiology, Seoul National University of Bundang Hospital, Kyonggi-do, South Korea
| | - Young Bok Ahn
- Department of Electronic Engineering, Konkuk University, Seoul 143-701, South Korea
| | - Seunghee Han
- Sogang Institute of Advanced Technology, Sogang University, Seoul 121-742, South Korea
| | - Yangmo Yoo
- Department of Electronic Engineering, Sogang University, Seoul 121-742, South Korea
| | - Tai-Kyong Song
- Department of Electronic Engineering, Sogang University, Seoul 121-742, South Korea
| |
Collapse
|
53
|
Chang LC, Chiu HM, Shun CT, Liang JT, Lin JT, Chen CC, Lee YC, Wu MS. Mutational profiles of different macroscopic subtypes of colorectal adenoma reveal distinct pathogenetic roles for KRAS, BRAF and PIK3CA. BMC Gastroenterol 2014; 14:221. [PMID: 25551625 PMCID: PMC4296683 DOI: 10.1186/s12876-014-0221-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 12/12/2014] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Investigations of genetic alterations and correlations with histology or morphology could provide further insights into colorectal carcinogenesis. Nevertheless, such genetic changes were less investigated in adenoma stage and a comprehensive survey of oncogenic mutations in EGFR signaling pathway according to different morphologic subtypes has not been performed. METHODS A total of 94 neoplasms, including 34 polypoid adenoma, 16 lateral spreading tumors-granular (LST-G), 20 non-granular LST (LST-NG), and 24 depressed tumors, were subjected for mutational analysis of KRAS (exon 2), BRAF (exon 11 and 15), PIK3CA (exon 9 and 20), AKT (exon 4), EGFR (exon 18-24) and HER2 (exon18-24). RESULTS KRAS mutation was noted more frequently in LST (13/36, 36.1%) than polypoid neoplasms (5/34, 14.7%, p = 0.041). When comparing with LST-NG, LST-G had a significantly higher frequency of KRAS mutation. (9/16, 56.3% vs. 4/20, 20.0%, p = 0.024). BRAF mutation (V600E) was found in 2 of 36 (5.6%)LSTs and 1 of 34 (2.9%) polypoid lesions. The two LST lesions with BRAF mutation were pathologically proven to be serrated adenoma. PIK3CA mutation (exon 9 E545K) was identified only in LST (5/36, 13.9%). Mutations in KRAS, BRAF or PIK3CA occurred in a mutually exclusive manner. All mutations were absent in the specimens obtained from depressed type neoplasms. CONCLUSIONS Three different macroscopic subtypes of colorectal neoplasms display distinct carcinogenetic pathways in EGFR networking. Further molecular studies of CRCs should take macroscopic subtypes into consideration and highlight the importance of consensus and communication between endoscopic and pathologic diagnosis.
Collapse
|
54
|
Abstract
PURPOSE OF REVIEW The introduction of novel molecular imaging modalities that can not only define disease states on the basis of structural changes and morphology, but also allow in-vivo visualization and characterization of molecular and biochemical alterations on a cellular level add a new dimension to our current diagnostic possibilities. The advents of innovative endoscopic devices coupled with the introduction of novel targeting ligands contribute to the recent advances made in the field of molecular imaging. The purpose of this review is to present and discuss the concepts and the potential of novel endoscopic imaging modalities for immune cell monitoring in the intestine. RECENT FINDINGS Recent progress concerning molecular imaging studies in animals and human patients implicates that this approach can be used to improve detection of mucosal lesions in wide-field imaging and for in-vivo characterization of the mucosa with the ultimate goal of assessing the likelihood of response to targeted therapy with biological agents. In particular, molecular endomicroscopy for assessment of mucosal immune responses ('immunoendoscopy') emerges as a novel approach for optimized endoscopic diagnosis and individualized therapy. SUMMARY Molecular imaging modalities in the intestine have the immediate potential to have an impact on current clinical practice and could therefore open new frontiers for clinical endoscopy and give hope for improved diagnosis and targeted therapies.
Collapse
|
55
|
Hoetker MS, Goetz M. Molecular imaging in endoscopy. United European Gastroenterol J 2014; 1:84-92. [PMID: 24917945 DOI: 10.1177/2050640613483291] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Accepted: 02/18/2013] [Indexed: 02/06/2023] Open
Abstract
Molecular imaging focuses on the molecular signature of cells rather than morphological changes in the tissue. The need for this novel type of imaging arises from the often difficult detection and characterization especially of small and/or premalignant lesions. Molecular imaging specifically visualizes biological properties of a lesion and might thereby be able to close diagnostic gaps, e.g. when differentiating hyperplastic from neoplastic polyps or detecting the margins of intraepithelial neoplastic spread. Additionally, not only the detection and discrimination of lesions could be improved: based on the molecular features identified using molecular imaging, therapy regimens could be adjusted on the day of diagnosis to allow for personalized medicine and optimized care for each individual patient.
Collapse
Affiliation(s)
| | - Martin Goetz
- Universitätsklinikum Tübingen, Tübingen, Germany
| |
Collapse
|
56
|
Abstract
White light endoscopy has proven to be a very powerful tool in oncology. There is still, however, a need for better endoscopic techniques to overcome the current limitations of white light optics. New technologies that allow higher sensitivity, improved microanatomy and molecular characterization have been available for in vitro microscopy and are now being translated into in vivo endoscopy. Endoscopic molecular imaging is still in its infancy but holds the promise for enhancing sensitivity for early lesions, thus allowing earlier diagnosis and enabling early image-guided endoscopic intervention. A key feature of endoscopic molecular imaging is its increased sensitivity and specificity, which will be illustrated in this article, as well as describing perspectives on its future use in oncologic surgery.
Collapse
Affiliation(s)
- Towhid Ali
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892-1088, USA
| | | | | |
Collapse
|
57
|
Tsai TH, Fujimoto JG, Mashimo H. Endoscopic Optical Coherence Tomography for Clinical Gastroenterology. Diagnostics (Basel) 2014; 4:57-93. [PMID: 26852678 PMCID: PMC4665545 DOI: 10.3390/diagnostics4020057] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Revised: 04/18/2014] [Accepted: 04/22/2014] [Indexed: 12/12/2022] Open
Abstract
Optical coherence tomography (OCT) is a real-time optical imaging technique that is similar in principle to ultrasonography, but employs light instead of sound waves and allows depth-resolved images with near-microscopic resolution. Endoscopic OCT allows the evaluation of broad-field and subsurface areas and can be used ancillary to standard endoscopy, narrow band imaging, chromoendoscopy, magnification endoscopy, and confocal endomicroscopy. This review article will provide an overview of the clinical utility of endoscopic OCT in the gastrointestinal tract and of recent achievements using state-of-the-art endoscopic 3D-OCT imaging systems.
Collapse
Affiliation(s)
- Tsung-Han Tsai
- Department of Electrical Engineering and Computer Science and Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - James G Fujimoto
- Department of Electrical Engineering and Computer Science and Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - Hiroshi Mashimo
- Veterans Affairs Boston Healthcare System and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
58
|
Goda K, Kato T, Tajiri H. Endoscopic diagnosis of early Barrett's neoplasia: perspectives for advanced endoscopic technology. Dig Endosc 2014; 26:311-21. [PMID: 24754238 DOI: 10.1111/den.12294] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Accepted: 02/28/2014] [Indexed: 12/18/2022]
Abstract
Barrett's esophagus (BE) is a metaplastic condition that occurs secondary to gastroesophageal reflux disease. BE is also a precursor to esophageal adenocarcinoma, which, although still rare in Japan, is one of the most rapidly increasing cancers in Western countries. However, the prevalence of gastroesophageal reflux disease has increased significantly over the past few decades in Japan, possibly leading to an incremental rise in BE and the associated inherent risk of adenocarcinoma. Given the poor prognosis of advanced-stage Barrett's adenocarcinoma, endoscopic surveillance is recommended for subjects with BE to detect early neoplasias including dysplasia. However, endoscopic identification of dysplastic lesions is still not sufficiently reliable or subjective, making targeted therapy extremely difficult. Over the past few years, improvements in image resolution, image processing software, and optical filter technology have enabled identification of dysplasia and early cancer in BE patients. We retrieved as many studies on advanced endoscopic technologies in BE as possible from MEDLINE and PubMed. The present review focuses on the emergent clinically available technologies to provide an overview of the technologies, their practical applicability, current status, and future challenges.
Collapse
Affiliation(s)
- Kenichi Goda
- Department of Endoscopy, The Jikei University School of Medicine, Tokyo, Japan
| | | | | |
Collapse
|
59
|
Realdon S, Dassie E, Fassan M, Dall'Olmo L, Hatem G, Buda A, Arcidiacono D, Diamantis G, Zhang H, Greene MI, Sturniolo GC, Rugge M, Alberti A, Battaglia G. In vivo molecular imaging of HER2 expression in a rat model of Barrett's esophagus adenocarcinoma. Dis Esophagus 2014; 28:394-403. [PMID: 24708360 DOI: 10.1111/dote.12210] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Human epidermal growth factor receptor 2 (HER2) is involved in the malignant progression of several human cancers, including esophageal adenocarcinoma (EAC). The purpose of this study was to evaluate HER2 overexpression and to explore the feasibility of confocal laser endomicroscopy for in vivo molecular imaging of HER2 status in an animal model of Barrett's-related EAC. Rats underwent esophagojejunostomy with gastric preservation. At 30 weeks post-surgery, the esophagus of 46 rats was studied; endoscopic and histological findings were correlated with HER2 immunofluorescence on excised biopsies and gross specimens. At this age, 23/46 rats developed Barrett's esophagus (BE), and 6/46 had cancer (four EAC and two squamous cell carcinomas). A significant overexpression of HER2 was observed in esophageal adenocarcinoma compared with normal squamous esophagus (9.4-fold) and BE (6.0-fold). AKT and its phosphorylated form were also overexpressed in cancer areas. Molecular imaging was performed at 80 weeks post-surgery in four rats after tail injection of fluorescent-labeled anti-HER2 antibody. At this age, 3/4 rats developed advance adenocarcinoma and showed in vivo overexpression of HER2 by molecular confocal laser endomicroscopy with heterogeneous distribution within cancer; no HER2 signal was observed in normal or Barrett's tissues. Therefore, HER2 overexpression is a typical feature of the surgical induced model of EAC that can be easily quantified in vivo using an innovative mini-invasive approach including confocal endomicroscopy; this approach may avoid limits of histological evaluation of HER2 status on 'blinded' biopsies.
Collapse
Affiliation(s)
- S Realdon
- Endoscopy Unit, Istituto Oncologico Veneto - IOV-IRCCS, Padua, Italy; Venetian Institute of Molecular Medicine (VIMM), Padua, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
60
|
Choi KS, Jung HY. Confocal laser endomicroscopy and molecular imaging in barrett esophagus and stomach. Clin Endosc 2014; 47:23-30. [PMID: 24570880 PMCID: PMC3928487 DOI: 10.5946/ce.2014.47.1.23] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 12/10/2013] [Indexed: 12/20/2022] Open
Abstract
Detection of premalignant lesions in the upper gastrointestinal tract may facilitate endoscopic treatment and improve survival. Despite technological advances in white light endoscopy, its ability to detect premalignant lesions remains limited. Early detection could be improved by using advanced endoscopic imaging techniques, such as magnification endoscopy, narrow band imaging, i-scanning, flexible spectral imaging color enhancement, autofluorescence imaging, and confocal laser endomicroscopy (CLE), as these techniques may increase the rate of detection of mucosal abnormalities and allow optical diagnosis. The present review focuses on advanced endoscopic imaging techniques based on the use of CLE for diagnosing premalignant lesions in Barrett esophagus and stomach.
Collapse
Affiliation(s)
- Kwi-Sook Choi
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hwoon-Yong Jung
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
61
|
Yang C, Hou VW, Girard EJ, Nelson LY, Seibel EJ. Target-to-background enhancement in multispectral endoscopy with background autofluorescence mitigation for quantitative molecular imaging. JOURNAL OF BIOMEDICAL OPTICS 2014; 19:76014. [PMID: 25027002 PMCID: PMC4098034 DOI: 10.1117/1.jbo.19.7.076014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 06/25/2014] [Indexed: 05/05/2023]
Abstract
Fluorescence molecular imaging with exogenous probes improves specificity for the detection of diseased tissues by targeting unambiguous molecular signatures. Additionally, increased diagnostic sensitivity is expected with the application of multiple molecular probes. We developed a real-time multispectral fluorescence-reflectance scanning fiber endoscope (SFE) for wide-field molecular imaging of fluorescent dye-labeled molecular probes at nanomolar detection levels. Concurrent multichannel imaging with the wide-field SFE also allows for real-time mitigation of the background autofluorescence (AF) signal, especially when fluorescein, a U.S. Food and Drug Administration approved dye, is used as the target fluorophore. Quantitative tissue AF was measured for the ex vivo porcine esophagus and murine brain tissues across the visible and nearinfrared spectra. AF signals were then transferred to the unit of targeted fluorophore concentration to evaluate the SFE detection sensitivity for sodium fluorescein and cyanine. Next, we demonstrated a real-time AF mitigation algorithm on a tissue phantom, which featured molecular probe targeted cells of high-grade dysplasia on a substrate containing AF species. The target-to-background ratio was enhanced by more than one order of magnitude when applying the real-time AF mitigation algorithm. Furthermore, a quantitative estimate of the fluorescein photodegradation (photobleaching) rate was evaluated and shown to be insignificant under the illumination conditions of SFE. In summary, the multichannel laser-based flexible SFE has demonstrated the capability to provide sufficient detection sensitivity, image contrast, and quantitative target intensity information for detecting small precancerous lesions in vivo.
Collapse
Affiliation(s)
- Chenying Yang
- University of Washington, Department of Bioengineering, Seattle, Washington 98195, United States
| | - Vivian W. Hou
- University of Washington, Department of Biology, Seattle, Washington 98195, United States
| | - Emily J. Girard
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, Washington 98109, United States
| | - Leonard Y. Nelson
- University of Washington, Department of Mechanical Engineering, Seattle, Washington 98195, United States
| | - Eric J. Seibel
- University of Washington, Department of Mechanical Engineering, Seattle, Washington 98195, United States
- Address all correspondence to: Eric J. Seibel,
| |
Collapse
|
62
|
Abstract
Molecular imaging is a novel field in gastroenterology that uses fluorescently labelled probes to specifically highlight neoplastic lesions on the basis of their molecular signature. The development of molecular imaging has been driven by the need to improve endoscopic diagnosis and by progress in targeted therapies in gastrointestinal oncology to provide individualized treatment, which coincides with progress in endoscopy techniques and further miniaturization of detection devices. Different exogenous molecular probes for imaging include labelled antibodies, oligopeptides, affibodies(™) (Affibody AB, Bromma, Sweden), aptamers and activatable probes. Molecular imaging has been evaluated in two major indications: many trials have studied molecular imaging as a red flag technique to improve detection of lesions in wide-field imaging; on the other hand, microscopic analysis has been investigated for in vivo characterization of the molecular fingerprint of tumours with the ultimate goal of assessing the likelihood of response to targeted therapy. This Review focusses on the applications of molecular imaging that have immediate potential for translational science or imminent transition into clinical practice of gastrointestinal endoscopy.
Collapse
|
63
|
Urquhart P, DaCosta R, Marcon N. Endoscopic mucosal imaging of gastrointestinal neoplasia in 2013. Curr Gastroenterol Rep 2013; 15:330. [PMID: 23771504 DOI: 10.1007/s11894-013-0330-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The holy grail of gastrointestinal endoscopy consists of the detection, in vivo characterization, and endoscopic removal of early or premalignant mucosal lesions. While our ability to achieve this goal has improved substantially since the development of the modern video-endoscope, inadequate visual inspection, errors of interpretation, and lesion subtlety all contribute to the continued suboptimal detection and assessment of early neoplasia. A myriad of new technologies has thus emerged that may help resolve these shortcomings; high magnification endoscopes, as well as the techniques of dye-based and virtual chromoendoscopy, are now widely available, while confocal laser endomicroscopy and endocystoscopy, optical coherence tomography, and autofluorescence imaging are generally applicable only in a research setting. Such technologies can be broadly categorized according to whether they potentially afford endoscopists improved detection, or real-time characterization, of mucosal lesions. Enhanced detection of otherwise "invisible" lesions, such as a flat area of intramucosal adenocarcinoma within Barrett's esophagus, carries the potential of an endoscopic cure prior to the development into a more advanced or metastatic disease. The ability to characterize a lesion to achieve an in vivo diagnosis, such as a colonic polyp, potentially affords endoscopists the ability to decide which lesions require removal and which can be safely left behind or discarded without histological assessment. Furthermore targeted biopsies, such as in the surveillance of chronic colitis, may prove to be more accurate and efficacious than the current protocol of random biopsies. An important caveat in the discussion of developing technologies in early cancer detection is the fundamental importance of a health-care system that promotes screening programs to recruit at-risk individuals. The ideal tool to optimize the use of endoscopy in population screening would be a panel of reliable biomarkers (blood, stool, or urine) that could effectively select a high-risk group, thus reducing the indiscriminate use of an expensive technology. The following review summarizes the current endoscopic imaging techniques available, and in development, for the early identification of gastrointestinal neoplasia.
Collapse
Affiliation(s)
- P Urquhart
- St Michael's Hospital, Toronto, ON, Canada
| | | | | |
Collapse
|
64
|
Muguruma N, Miyamoto H, Okahisa T, Takayama T. Endoscopic molecular imaging: status and future perspective. Clin Endosc 2013; 46:603-10. [PMID: 24340252 PMCID: PMC3856260 DOI: 10.5946/ce.2013.46.6.603] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Revised: 09/30/2013] [Accepted: 09/30/2013] [Indexed: 12/13/2022] Open
Abstract
During the last decade, researchers have made great progress in the development of new image processing technologies for gastrointestinal endoscopy. However, diagnosis using conventional endoscopy with white light optical imaging is essentially limited, and ultimately, we still rely on the histopathological diagnosis from biopsy specimens. Molecular imaging represents the most novel imaging methods in medicine, and the future of endoscopic diagnosis is likely to be impacted by a combination of biomarkers and technology. Endoscopic molecular imaging can be defined as the visualization of molecular characteristics with endoscopy. These innovations will allow us not only to locate a tumor or dysplastic lesion but also to visualize its molecular characteristics and the activity of specific molecules and biological processes that affect tumor behavior and/or its response to therapy. In the near future, these promising technologies will play a central role in endoluminal oncology.
Collapse
Affiliation(s)
- Naoki Muguruma
- Department of Gastroenterology and Oncology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | | | | | | |
Collapse
|
65
|
Kim SY, Myung SJ. Optical molecular imaging for diagnosing intestinal diseases. Clin Endosc 2013; 46:620-6. [PMID: 24340254 PMCID: PMC3856262 DOI: 10.5946/ce.2013.46.6.620] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 10/12/2013] [Accepted: 10/17/2013] [Indexed: 12/20/2022] Open
Abstract
Real-time visualization of the molecular signature of cells can be achieved with advanced targeted imaging techniques using molecular probes and fluorescence endoscopy. This molecular optical imaging in gastrointestinal endoscopy is promising for improving the detection of neoplastic lesions, their characterization for patient stratification, and the assessment of their response to molecular targeted therapy and radiotherapy. In inflammatory bowel disease, this method can be used to detect dysplasia in the presence of background inflammation and to visualize inflammatory molecular targets for assessing disease severity and prognosis. Several preclinical and clinical trials have applied this method in endoscopy; however, this field has just started to evolve. Hence, many problems have yet to be solved to enable the clinical application of this novel method.
Collapse
Affiliation(s)
- Sang-Yeob Kim
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | | |
Collapse
|
66
|
Abstract
Gastrointestinal endoscopy had major technological improvements and novel technologies in recent years. High-definition endoscopy has permitted an increasingly detailed view of the mucosa during colonoscopy. Filter techniques that enhance analysis of vessel and surface structures. Autofluorescence imaging relies on functional imaging of tissue alterations. Endocytoscopy is an ultrahigh-contact microscopy procedure for cellular analysis of the epithelium. Endomicroscopy is an adaption of laser scanning microscopy for real-time intravital surface and subsurface microscopy during endoscopy. With these technologies, endoscopy has moved from prediction of histology based on morphologic patterns toward visualization of cellular and subcellular details, providing real-time histology.
Collapse
Affiliation(s)
- Martin Goetz
- Innere Medizin I, Universitätsklinikum Tübingen, Otfried-Müller-Street 10, Tübingen 72076, Germany.
| |
Collapse
|
67
|
Fluorescein derivatives in intravital fluorescence imaging. Cells 2013; 2:591-606. [PMID: 24709799 PMCID: PMC3972669 DOI: 10.3390/cells2030591] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 07/25/2013] [Accepted: 07/26/2013] [Indexed: 01/19/2023] Open
Abstract
Intravital fluorescence microscopy enables the direct imaging of fluorophores in vivo and advanced techniques such as fluorescence lifetime imaging (FLIM) enable the simultaneous detection of multiple fluorophores. Consequently, it is now possible to record distribution and metabolism of a chemical in vivo and to optimise the delivery of fluorophores in vivo. Recent clinical applications with fluorescein and other intravital fluorescent stains have occurred in neurosurgery, dermatology [including photodynamic therapy (PDT)] and endomicroscopy. Potential uses have been identified in periodontal disease, skin graft and cancer surgery. Animal studies have demonstrated that diseased tissue can be specifically stained with fluorophore conjugates. This review focuses on the fluorescein derived fluorophores in common clinical use and provides examples of novel applications from studies in tissue samples.
Collapse
|
68
|
Yang C, Hou V, Nelson LY, Seibel EJ. Mitigating fluorescence spectral overlap in wide-field endoscopic imaging. JOURNAL OF BIOMEDICAL OPTICS 2013; 18:86012. [PMID: 23966226 PMCID: PMC3767456 DOI: 10.1117/1.jbo.18.8.086012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
The number of molecular species suitable for multispectral fluorescence imaging is limited due to the overlap of the emission spectra of indicator fluorophores, e.g., dyes and nanoparticles. To remove fluorophore emission cross-talk in wide-field multispectral fluorescence molecular imaging, we evaluate three different solutions: (1) image stitching, (2) concurrent imaging with cross-talk ratio subtraction algorithm, and (3) frame-sequential imaging. A phantom with fluorophore emission cross-talk is fabricated, and a 1.2-mm ultrathin scanning fiber endoscope (SFE) is used to test and compare these approaches. Results show that fluorophore emission cross-talk could be successfully avoided or significantly reduced. Near term, the concurrent imaging method of wide-field multispectral fluorescence SFE is viable for early stage cancer detection and localization in vivo. Furthermore, a means to enhance exogenous fluorescence target-to-background ratio by the reduction of tissue autofluorescence background is demonstrated.
Collapse
Affiliation(s)
- Chenying Yang
- University of Washington, Department of Bioengineering, Seattle, Washington 98195, USA.
| | | | | | | |
Collapse
|
69
|
Liu Z, Miller SJ, Joshi BP, Wang TD. In vivo targeting of colonic dysplasia on fluorescence endoscopy with near-infrared octapeptide. Gut 2013; 62:395-403. [PMID: 22427239 PMCID: PMC3563943 DOI: 10.1136/gutjnl-2011-301913] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To demonstrate a near-infrared (NIR) peptide that is highly specific for colonic adenomas on fluorescence endoscopy in vivo. DESIGN A 3 mm diameter endoscope was adapted to deliver 671 nm illumination and collect NIR fluorescence (696-736 nm). Target (QPIHPNNM) and control (YTTNKH) peptides were labelled with Cy5.5, a NIR dye, and characterised by mass spectra. The peptides were topically administered separately (100 μM) through the endoscope's instrument channel into the distal colon of CPC;Apc mice, genetically engineered to spontaneously develop adenomas. After 5 min for incubation, the unbound peptides were rinsed off, and images were collected at a rate of 10 frames/s. Regions of interest were identified around the adenoma and adjacent normal-appearing mucosa on white light. Intensity measurements were made from these same regions on fluorescence, and the target-to-background ratio (TBR) was calculated. RESULTS An image resolution of 9.8 μm and field of view of 3.6 mm was achieved at a distance of 2.5 mm between the distal end of the instrument and the tissue surface. On mass spectra, the experimental mass-to-charge ratio for the Cy5.5-labelled target and control peptides agreed with expected values. The NIR fluorescence images of adenomas revealed individual dysplastic crypts with distorted morphology. By comparison, only amorphous surface features could be visualised from reflected NIR light. The average TBR for adenomas was found to be 3.42 ± 1.30 and 1.88 ± 0.38 for the target and control peptides, respectively, p=0.007. CONCLUSION A NIR peptide was shown to be highly specific for colonic adenomas on fluorescence endoscopy in vivo and to achieve sub-cellular resolution images.
Collapse
Affiliation(s)
- Zhongyao Liu
- Department of Medicine, Division of Gastroenterology, Ann Arbor, MI 48109
| | - Sharon J. Miller
- Department of Medicine, Division of Gastroenterology, Ann Arbor, MI 48109
| | - Bishnu P. Joshi
- Department of Medicine, Division of Gastroenterology, Ann Arbor, MI 48109
| | - Thomas D. Wang
- Department of Medicine, Division of Gastroenterology, Ann Arbor, MI 48109,Department of Biomedical Engineering, Ann Arbor, Michigan 48109
| |
Collapse
|
70
|
Yang C, Hou V, Nelson LY, Seibel EJ. Color-matched and fluorescence-labeled esophagus phantom and its applications. JOURNAL OF BIOMEDICAL OPTICS 2013; 18:26020. [PMID: 23403908 PMCID: PMC3569733 DOI: 10.1117/1.jbo.18.2.026020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
We developed a stable, reproducible three-dimensional optical phantom for the evaluation of a wide-field endoscopic molecular imaging system. This phantom mimicked a human esophagus structure with flexibility to demonstrate body movements. At the same time, realistic visual appearance and diffuse spectral reflectance properties of the tissue were simulated by a color matching methodology. A photostable dye-in-polymer technology was applied to represent biomarker probed "hot-spot" locations. Furthermore, fluorescent target quantification of the phantom was demonstrated using a 1.2 mm ultrathin scanning fiber endoscope with concurrent fluorescence-reflectance imaging.
Collapse
Affiliation(s)
- Chenying Yang
- University of Washington, Department of Bioengineering, 204 Fluke Hall, 4000 Mason Road, Seattle, WA 98195, USA.
| | | | | | | |
Collapse
|
71
|
Endomicroscopic Imaging of COX-2 Activity in Murine Sporadic and Colitis-Associated Colorectal Cancer. DIAGNOSTIC AND THERAPEUTIC ENDOSCOPY 2013; 2013:250641. [PMID: 23401648 PMCID: PMC3562574 DOI: 10.1155/2013/250641] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 12/26/2012] [Indexed: 12/21/2022]
Abstract
Although several studies propose a chemopreventive effect of aspirin for colorectal cancer (CRC) development, the general use of aspirin cannot be recommended due to its adverse side effects. As the protective effect of aspirin has been associated with an increased expression of COX-2, molecular imaging of COX-2, for instance, during confocal endomicroscopy could enable the identification of patients who would possibly benefit from aspirin treatment. In this pilot trial, we used a COX-2-specific fluorescent probe for detection of colitis-associated and sporadic CRC in mice using confocal microscopy. Following the injection of the COX-2 probe into tumor-bearing APCmin mice or mice exposed to the AOM + DSS model of colitis-associated cancer, the tumor-specific upregulation of COX-2 could be validated with in vivo fluorescence imaging. Subsequent confocal imaging of tumor tissue showed an increased number of COX-2 expressing cells when compared to the normal mucosa of healthy controls. COX-2-expression was detectable with subcellular resolution in tumor cells and infiltrating stroma cells. These findings pose a proof of concept and suggest the use of CLE for the detection of COX-2 expression during colorectal cancer surveillance endoscopy. This could improve early detection and stratification of chemoprevention in patients with CRC.
Collapse
|
72
|
Sevick-Muraca EM, Akers WJ, Joshi BP, Luker GD, Cutler CS, Marnett LJ, Contag CH, Wang TD, Azhdarinia A. Advancing the translation of optical imaging agents for clinical imaging. BIOMEDICAL OPTICS EXPRESS 2013; 4:160-170. [PMID: 23304655 PMCID: PMC3539189 DOI: 10.1364/boe.4.000160] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Revised: 11/13/2012] [Accepted: 11/23/2012] [Indexed: 05/29/2023]
Abstract
Despite the development of a large number of promising candidates, few contrast agents for established medical imaging modalities have successfully been translated over the past decade. The emergence of new imaging contrast agents that employ biomedical optics is further complicated by the relative infancy of the field and the lack of approved imaging devices compared to more established clinical modalities such as nuclear medicine. Herein, we propose a navigational approach (as opposed to a fixed "roadmap") for translation of optical imaging agents that is (i) proposed through consensus by four academic research programs that are part of the cooperative U54 NCI Network for Translational Research, (ii) developed through early experiences for translating optical imaging agents in order to meet distinctly varied needs in cancer diagnostics, and (iii) adaptable to the rapidly changing environment of academic medicine. We describe the pathways by which optical imaging agents are synthesized, qualified, and validated for preclinical testing, and ultimately translated for "first-in-humans" studies using investigational optical imaging devices. By identifying and adopting consensus approaches for seemingly disparate optical imaging modalities and clinical indications, we seek to establish a systematic method for navigating the ever-changing "roadmap" to most efficiently arrive at the destination of clinical adoption and improved outcome and survivorship for cancer patients.
Collapse
Affiliation(s)
- Eva M. Sevick-Muraca
- The University of Texas Health Science Center at Houston, Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, 1825 Pressler Street, Houston, TX 77030, USA
| | - Walter J. Akers
- Washington University School of Medicine, Department of Radiology, St. Louis, MO 63110, USA
| | - Bishnu P. Joshi
- The University of Michigan, School of Medicine, Department of Internal-Medicine-Division of Gastroenterology, Ann Arbor, MI 48109, USA
| | - Gary D. Luker
- The University of Michigan, School of Medicine, Department of Internal-Medicine-Division of Gastroenterology, Ann Arbor, MI 48109, USA
| | - Cathy S. Cutler
- University of Missouri Research Reactor Center (MURR), Radiopharmaceutical Sciences Institute, Nuclear Engineering and Sciences Institute, Nuclear Engineering, Columbia, MO 65211, USA
| | | | - Christopher H. Contag
- Stanford University, School of Medicine, Department of Pediatrics, Stanford, CA 94305, USA
| | - Thomas D. Wang
- The University of Michigan, School of Medicine, Department of Internal-Medicine-Division of Gastroenterology, Ann Arbor, MI 48109, USA
| | - Ali Azhdarinia
- The University of Texas Health Science Center at Houston, Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, 1825 Pressler Street, Houston, TX 77030, USA
| |
Collapse
|
73
|
Goetz M. Confocal Laser Endomicroscopy: Applications in Clinical and Translational Science—A Comprehensive Review. ACTA ACUST UNITED AC 2012. [DOI: 10.5402/2012/387145] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Confocal laser endomicroscopy (CLE) is a novel tool in the endoscopist’s armamentarium. It allows on-site histological information. The ability of gastroenterologists to interpret such microscopic information has been demonstrated in multiple studies from the upper and lower gastrointestinal tract. Recently, the field of application has expanded to provide hepatobiliary and intra-abdominal CLE imaging. CLE allows “smart,” targeted biopsies and is able to guide endoscopic interventions. But CLE is also translational in its approach and permits functional imaging that significantly impacts on our understanding of gastrointestinal diseases. Molecular imaging with CLE allows detection and characterization of lesions and may even be used for prediction of response to targeted therapy. This paper provides a comprehensive review over current applications of CLE in clinical applications and translational science.
Collapse
Affiliation(s)
- Martin Goetz
- Innere Medizin I, Universitätsklinikum Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
74
|
Matsunaga TO, Sheeran PS, Luois S, Streeter JE, Mullin LB, Banerjee B, Dayton PA. Phase-change nanoparticles using highly volatile perfluorocarbons: toward a platform for extravascular ultrasound imaging. Theranostics 2012; 2:1185-98. [PMID: 23382775 PMCID: PMC3563153 DOI: 10.7150/thno.4846] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Accepted: 11/02/2012] [Indexed: 12/20/2022] Open
Abstract
Recent efforts using perfluorocarbon (PFC) nanoparticles in conjunction with acoustic droplet vaporization has introduced the possibility of expanding the diagnostic and therapeutic capability of ultrasound contrast agents to beyond the vascular space. Our laboratories have developed phase-change nanoparticles (PCNs) from the highly volatile PFCs decafluorobutane (DFB, bp =-2 °C) and octafluoropropane (OFP, bp =-37 °C ) for acoustic droplet vaporization. Studies with commonly used clinical ultrasound scanners have demonstrated the ability to vaporize PCN emulsions with frequencies and mechanical indices that may significantly decrease tissue bioeffects. In addition, these contrast agents can be formulated to be stable at physiological temperatures and the perfluorocarbons can be mixed to modulate the balance between sensitivity to ultrasound and general stability. We herein discuss our recent efforts to develop finely-tuned diagnostic/molecular imaging agents for tissue interrogation. We discuss studies currently under investigation as well as potential diagnostic and therapeutic paradigms that may emerge as a result of formulating PCNs with low boiling point PFCs.
Collapse
|
75
|
In vivo molecular imaging of epidermal growth factor receptor in patients with colorectal neoplasia using confocal laser endomicroscopy. Cancer Lett 2012; 330:200-7. [PMID: 23220286 DOI: 10.1016/j.canlet.2012.11.044] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Revised: 11/25/2012] [Accepted: 11/27/2012] [Indexed: 01/12/2023]
Abstract
Epidermal growth factor receptor (EGFR) plays an important role in tumorigenesis of colorectal cancer (CRC), and its in vivo molecular imaging in rodent models has become the subject of an increased number of studies using novel imaging techniques for gastrointestinal endoscopy. Current study aimed to evaluate the use of confocal endomicroscopy (CLE) for in vivo molecular imaging of EGFR in patients with colorectal neoplasia. Molecular imaging of colorectal neoplasia in patients was performed by CLE after topical application of a fluorescent-labeled molecular probe against EGFR. Representative images of CLE were chosen to calculate EGFR-specific fluorescence intensity. Targeted biopsy specimens were taken from each examined site during in vivo imaging for histology and immunohistochemistry (IHC). During in vivo molecular imaging in 37 patients, an EGFR-specific fluorescence signal was present in 18/19 CRC, and 12/18 colorectal adenomas. No or only weak fluorescence signal was observed in vivo in 10 cases of normal mucosa. CLE is a novel tool that could be used in molecular imaging with specific targeting of EGFR in patients with colorectal neoplasia. This technique demonstrates a promising imaging approach for targeted therapies of colorectal neoplasia.
Collapse
|
76
|
Joshi BP, Liu Z, Elahi SF, Appelman H, 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; 76:1197-206.e1-5. [PMID: 23022051 PMCID: PMC3502727 DOI: 10.1016/j.gie.2012.07.017] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2012] [Accepted: 07/08/2012] [Indexed: 02/07/2023]
Abstract
BACKGROUND Fluorescent-labeled peptides are being developed to improve the endoscopic detection of colonic dysplasia. OBJECTIVE To demonstrate a near-infrared peptide multimer that functions as a phage mimic for in vivo detection of colonic adenomas. DESIGN A peptide multimer was synthesized by using trilysine as a dendritic wedge to mimic the presentation of peptides on phage, and all peptides, including the multimer, were fluorescent-labeled with Cy5.5. SETTING Small-animal imaging facility. ANIMAL SUBJECTS: Genetically engineered CPC;Apc mice that spontaneously develop colonic adenomas. INTERVENTION Near-infrared-labeled AKPGYLS peptide multimer was administered topically into the distal colons of the mice, and endoscopic images of adenomas were captured. Fluorescence intensities were quantified by target-to-background (T/B) ratios, and adenoma dimensions were measured with calipers after imaging. Validation of specific peptide binding was performed on cryosectioned specimens and cells by using confocal microscopy and flow cytometry. MAIN OUTCOME MEASUREMENTS Fluorescence T/B ratios from colonic adenomas and adjacent normal-appearing mucosa. RESULTS AKP-multimer, monomer, trilysine core, and Cy5.5 resulted in mean (± SD) T/B ratios of 3.85 ± 0.25, 2.21 ± 0.13, 1.56 ± 0.12, and 1.19 ± 0.11, respectively, P < .01 on in vivo imaging. Peptide multimer showed higher contrast and greater specificity for dysplastic crypts as compared with other probes. Peptide multimer demonstrated significantly greater binding to HT29 cells on flow cytometry and fluorescence microscopy in comparison to monomer and trilysine core. A binding affinity of 6.4 nm/L and time constant of 0.1136 minutes(-1) (8.8 minutes) was measured for multimer. LIMITATIONS Only distal colonic adenomas were imaged. CONCLUSION Peptide multimers combine strengths of multiple individual peptides to enhance binding interactions and demonstrate significantly higher specificity and affinity for tumor targets.
Collapse
Affiliation(s)
- Bishnu P. Joshi
- Department of Medicine, Division of Gastroenterology, Ann Arbor, MI 48109
| | - Zhongyao Liu
- Department of Medicine, Division of Gastroenterology, Ann Arbor, MI 48109
| | - Sakib F. Elahi
- Department of Biomedical Engineering, Ann Arbor, Michigan 48109
| | | | - Thomas D. Wang
- Department of Medicine, Division of Gastroenterology, Ann Arbor, MI 48109,Department of Biomedical Engineering, Ann Arbor, Michigan 48109
| |
Collapse
|
77
|
Abstract
Colorectal cancer represents one of the leading malignancies worldwide. Early endoscopic detection and removal of its precursor lesions, adenomas, and serrated hyperplastic polyps results in a decrease of colon cancer-related death. However, miss rates in adenoma detection up to 26% underline the need for high compliance to basic measures and further improvement in methodology and technology. Basic parameters affecting adenoma detection rates include sufficient training and awareness of the endoscopist, use of high-definition endoscopes, careful examination behind folds, cleansing the colon wall, accurate distention of the colon, and adequate withdrawal time. Advanced imaging techniques, introduced to further improve adenoma detection, have yielded mixed results. These include wide-angle colonoscopes, cap-assisted colonoscopy, and retroscopic methods which may add new obstacles to colonoscopy. Moreover, chromoendoscopy either 'virtual' or by topically applied dyes has been suggested to enhance the detection of colonic neoplasia. Yet, studies on patients with average cancer risk have failed to reproduce promising initial results. Similarly, although autofluorescence has not enhanced the diagnostic yield in screening a population at average risk, it may be useful in patients at increased cancer risk. Recently, technical feasibility of molecular imaging employing 'biomarkers' has been demonstrated, but needs further evaluation. The newest developments, employing light-scattering spectroscopy, suggest the existence of a 'field effect' of colonic carcinogenesis and may enable detection of the earliest neoplastic events and distant adenomas even when applied to normal-appearing mucosa. Upon confirmation, these technologies may result in a substantial change in patient management and risk stratification.
Collapse
Affiliation(s)
- Mario Anders
- Department of Interdisciplinary Endoscopy, University Hospital Hamburg Eppendorf, Hamburg, Germany.
| |
Collapse
|
78
|
Ding S, Blue RE, Chen Y, Scull B, Lund PK, Morgan D. Molecular Imaging of Gastric Neoplasia with Near-Infrared Fluorescent Activatable Probes. Mol Imaging 2012. [DOI: 10.2310/7290.2012.00014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
- Shengli Ding
- From the Department of Cell and Molecular Physiology and Division of Gastroenterology, University of North Carolina at Chapel Hill, Chapel Hill, NC; Department of Biological Sciences, Kent State University, Kent, OH; and Department of Gastroenterology, Vanderbilt University Medical Center, Nashville, TN
| | - Randall Eric Blue
- From the Department of Cell and Molecular Physiology and Division of Gastroenterology, University of North Carolina at Chapel Hill, Chapel Hill, NC; Department of Biological Sciences, Kent State University, Kent, OH; and Department of Gastroenterology, Vanderbilt University Medical Center, Nashville, TN
| | - Yijing Chen
- From the Department of Cell and Molecular Physiology and Division of Gastroenterology, University of North Carolina at Chapel Hill, Chapel Hill, NC; Department of Biological Sciences, Kent State University, Kent, OH; and Department of Gastroenterology, Vanderbilt University Medical Center, Nashville, TN
| | - Brooks Scull
- From the Department of Cell and Molecular Physiology and Division of Gastroenterology, University of North Carolina at Chapel Hill, Chapel Hill, NC; Department of Biological Sciences, Kent State University, Kent, OH; and Department of Gastroenterology, Vanderbilt University Medical Center, Nashville, TN
| | - Pauline Kay Lund
- From the Department of Cell and Molecular Physiology and Division of Gastroenterology, University of North Carolina at Chapel Hill, Chapel Hill, NC; Department of Biological Sciences, Kent State University, Kent, OH; and Department of Gastroenterology, Vanderbilt University Medical Center, Nashville, TN
| | - Douglas Morgan
- From the Department of Cell and Molecular Physiology and Division of Gastroenterology, University of North Carolina at Chapel Hill, Chapel Hill, NC; Department of Biological Sciences, Kent State University, Kent, OH; and Department of Gastroenterology, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
79
|
Ding S, Blue RE, Chen Y, Scull B, Lund PK, Morgan D. Molecular imaging of gastric neoplasia with near-infrared fluorescent activatable probes. Mol Imaging 2012; 11:507-515. [PMID: 23084251 PMCID: PMC3689298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023] Open
Abstract
Gastric cancer is the second leading cause of cancer mortality worldwide and is projected to rise to tenth in all-cause mortality in the near term. Early detection requires improved sensitivity and specificity of endoscopic imaging with novel methods. The objective of this study was to evaluate the utility of activatable molecular probes for the detection of gastric cancer both in vivo and ex vivo in a preclinical model. Smad4⁺/⁻ mice, which develop spontaneous gastric neoplasia, were compared to normal wild-type controls. Cathepsin-activatable and matrix metalloproteinase (MMP)-activatable molecular probes were injected 24 hours and 6 hours before imaging, respectively. In vivo imaging was performed using quantitative tomographic near-infrared fluorescence (NIRF) imaging. For validation, ex vivo imaging and histologic examination were performed. Molecular imaging in vivo of Smad4⁺/⁻ gastric cancer murine models revealed intense activation of both cathepsin B and MMP probes. Ex vivo imaging and histology confirmed that the detected neoplasms were adenocarcinomas and hyperplastic lesions. This study provides proof of principle that the cathepsin- and MMP-activatable molecular probes are activated in the Smad4⁺/⁻ murine model of spontaneous gastric adenocarcinoma and can be imaged by both in vivo and ex vivo NIRF methods. The cathepsin probe also detects hyperplastic lesions.
Collapse
Affiliation(s)
- Shengli Ding
- Department of Cell and Molecular Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Randall Eric Blue
- Department of Cell and Molecular Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Yijing Chen
- Department of Biological Sciences, Kent State University, Kent, OH
| | - Brooks Scull
- Department of Gastroenterology, Vanderbilt University Medical Center, Nashville, TN
| | - Pauline Kay Lund
- Department of Cell and Molecular Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Douglas Morgan
- Division of Gastroenterology, University of North Carolina, Chapel Hill, NC
| |
Collapse
|
80
|
Lyo V, Cattaruzza F, Kim TN, Walker AW, Paulick M, Cox D, Cloyd J, Buxbaum J, Ostroff J, Bogyo M, Grady EF, Bunnett NW, Kirkwood KS. Active cathepsins B, L, and S in murine and human pancreatitis. Am J Physiol Gastrointest Liver Physiol 2012; 303:G894-903. [PMID: 22899821 PMCID: PMC3469694 DOI: 10.1152/ajpgi.00073.2012] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Cathepsins regulate premature trypsinogen activation within acinar cells, a key initial step in pancreatitis. The identity, origin, and causative roles of activated cathepsins in pancreatic inflammation and pain are not defined. By using a near infrared-labeled activity-based probe (GB123) that covalently modifies active cathepsins, we localized and identified activated cathepsins in mice with cerulein-induced pancreatitis and in pancreatic juice from patients with chronic pancreatitis. We used inhibitors of activated cathepsins to define their causative role in pancreatic inflammation and pain. After GB123 administration to mice with pancreatitis, reflectance and confocal imaging showed significant accumulation of the probe in inflamed pancreas compared with controls, particularly in acinar cells and macrophages, and in spinal cord microglia and neurons. Biochemical analysis of pancreatic extracts identified them as cathepsins B, L, and S (Cat-B, Cat-L, and Cat-S, respectively). These active cathepsins were also identified in pancreatic juice from patients with chronic pancreatitis undergoing an endoscopic procedure for the treatment of pain, indicating cathepsin secretion. The cathepsin inhibitor K11777 suppressed cerulein-induced activation of Cat-B, Cat-L, and Cat-S in the pancreas and ameliorated pancreatic inflammation, nocifensive behavior, and activation of spinal nociceptive neurons. Thus pancreatitis is associated with an increase in the active forms of the proteases Cat-B, Cat-L, and Cat-S in pancreatic acinar cells and macrophages, and in spinal neurons and microglial cells. Inhibition of cathepsin activation ameliorated pancreatic inflammation and pain. Activity-based probes permit identification of proteases that are predictive biomarkers of disease progression and response to therapy and may be useful noninvasive tools for the detection of pancreatic inflammation.
Collapse
Affiliation(s)
- Victoria Lyo
- 1Department of Surgery, University of California, San Francisco, San Francisco, California;
| | - Fiore Cattaruzza
- 1Department of Surgery, University of California, San Francisco, San Francisco, California;
| | - Tyson N. Kim
- 1Department of Surgery, University of California, San Francisco, San Francisco, California;
| | - Austin W. Walker
- 1Department of Surgery, University of California, San Francisco, San Francisco, California;
| | - Margot Paulick
- 2Department of Pathology, Stanford University, Stanford, California;
| | - Daniel Cox
- 1Department of Surgery, University of California, San Francisco, San Francisco, California;
| | - Jordan Cloyd
- 1Department of Surgery, University of California, San Francisco, San Francisco, California;
| | - James Buxbaum
- 3Department of Gastroenterology, University of California, San Francisco, San Francisco, California; Departments of Pharmacology and Medicine, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia
| | - James Ostroff
- 3Department of Gastroenterology, University of California, San Francisco, San Francisco, California; Departments of Pharmacology and Medicine, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia
| | - Matthew Bogyo
- 2Department of Pathology, Stanford University, Stanford, California;
| | - Eileen F. Grady
- 1Department of Surgery, University of California, San Francisco, San Francisco, California;
| | - Nigel W. Bunnett
- 3Department of Gastroenterology, University of California, San Francisco, San Francisco, California; Departments of Pharmacology and Medicine, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia
| | - Kimberly S. Kirkwood
- 1Department of Surgery, University of California, San Francisco, San Francisco, California;
| |
Collapse
|
81
|
Murthy S, Goetz M, Hoffman A, Kiesslich R. Novel colonoscopic imaging. Clin Gastroenterol Hepatol 2012; 10:984-7. [PMID: 22835580 DOI: 10.1016/j.cgh.2012.07.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 07/18/2012] [Accepted: 07/19/2012] [Indexed: 02/06/2023]
Affiliation(s)
- Sanjay Murthy
- Department of Medicine, Mount Sinai Hospital and University of Toronto, Toronto, Canada
| | | | | | | |
Collapse
|
82
|
Lee MH, Buterbaugh K, Richards-Kortum R, Anandasabapathy S. Advanced endoscopic imaging for Barrett's Esophagus: current options and future directions. Curr Gastroenterol Rep 2012; 14:216-25. [PMID: 22453701 DOI: 10.1007/s11894-012-0259-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Barrett's esophagus is the precursor to esophageal adenocarcinoma, one of the most rapidly increasing cancers in the United States. Given the poor prognosis of late-stage adenocarcinoma, endoscopic surveillance is recommended for subjects with Barrett's esophagus to detect early neoplasia. Current guidelines recommend "random" four-quadrant biopsies taken every 1-2 cm throughout the Barrett's segment. However, this only samples a minority of epithelium and has been shown to miss areas of endoscopically- inapparent neoplasia (high grade dysplasia or cancer). Recent efforts have focused on developing novel diagnostic imaging technologies to detect the subtle epithelial changes associated with dysplasia and neoplasia in Barrett's esophagus. Some of these modalities serve as "red flag" technologies designed to detect areas of abnormality within large surface areas. Other technologies serve to characterize areas of visible abnormality, offering a higher spatial resolution to confirm/exclude the presence of neoplasia. This review summarizes several available and evolving imaging technologies used in the endoscopic diagnosis and surveillance of Barrett's associated neoplasia.
Collapse
Affiliation(s)
- Michelle H Lee
- Division of Gastroenterology, The Mount Sinai Medical Center, New York, NY 10029, USA
| | | | | | | |
Collapse
|
83
|
Aldrich MB, Marshall MV, Sevick-Muraca EM, Lanza G, Kotyk J, Culver J, Wang LV, Uddin J, Crews BC, Marnett LJ, Liao JC, Contag C, Crawford JM, Wang K, Reisdorph B, Appelman H, Turgeon DK, Meyer C, Wang T. Seeing it through: translational validation of new medical imaging modalities. BIOMEDICAL OPTICS EXPRESS 2012; 3:764-76. [PMID: 22574264 PMCID: PMC3345805 DOI: 10.1364/boe.3.000764] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Revised: 02/10/2012] [Accepted: 02/13/2012] [Indexed: 05/21/2023]
Abstract
Medical imaging is an invaluable tool for diagnosis, surgical guidance, and assessment of treatment efficacy. The Network for Translational Research (NTR) for Optical Imaging consists of four research groups working to "bridge the gap" between lab discovery and clinical use of fluorescence- and photoacoustic-based imaging devices used with imaging biomarkers. While the groups are using different modalities, all the groups face similar challenges when attempting to validate these systems for FDA approval and, ultimately, clinical use. Validation steps taken, as well as future needs, are described here. The group hopes to provide translational validation guidance for itself, as well as other researchers.
Collapse
Affiliation(s)
- Melissa B. Aldrich
- Center for Molecular Imaging, The Brown Foundation Institute for Molecular Medicine, The University of Texas Health Science Center-Houston, 1825 Pressler, 330-07, Houston, TX 77030, USA
| | - Milton V. Marshall
- Center for Molecular Imaging, The Brown Foundation Institute for Molecular Medicine, The University of Texas Health Science Center-Houston, 1825 Pressler, 330-07, Houston, TX 77030, USA
| | - Eva M. Sevick-Muraca
- Center for Molecular Imaging, The Brown Foundation Institute for Molecular Medicine, The University of Texas Health Science Center-Houston, 1825 Pressler, 330-07, Houston, TX 77030, USA
| | - Greg Lanza
- Department of Biomedical Engineering, Washington University in St. Louis, Campus Box 1097, One Brookings Drive, St. Louis, MO 63130, USA
| | - John Kotyk
- Department of Biomedical Engineering, Washington University in St. Louis, Campus Box 1097, One Brookings Drive, St. Louis, MO 63130, USA
| | - Joseph Culver
- Department of Biomedical Engineering, Washington University in St. Louis, Campus Box 1097, One Brookings Drive, St. Louis, MO 63130, USA
| | - Lihong V. Wang
- Department of Biomedical Engineering, Washington University in St. Louis, Campus Box 1097, One Brookings Drive, St. Louis, MO 63130, USA
| | - Jashim Uddin
- Department of Biochemistry, Vanderbilt University Medical Center, 850 Robinson Research Building, Nashville, TN 37232, USA
| | - Brenda C. Crews
- Department of Biochemistry, Vanderbilt University Medical Center, 850 Robinson Research Building, Nashville, TN 37232, USA
| | - Lawrence J. Marnett
- Department of Biochemistry, Vanderbilt University Medical Center, 850 Robinson Research Building, Nashville, TN 37232, USA
| | - Joseph C. Liao
- Department of Pediatrics, Clark Center, East Wing E150, 318 Campus Drive, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Chris Contag
- Department of Pediatrics, Clark Center, East Wing E150, 318 Campus Drive, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - James M. Crawford
- Department of Pathology, Hofstra North Shore-LIJ College of Medicine, 10 Nevada Drive, Lake Success, NY 11042, USA
| | - Ken Wang
- Gastroenterology and Hepatology, Mayo Clinic Rochester, 200 First Street SW, Rochester, MN 55905, USA
| | - Bill Reisdorph
- Michigan Institute for Clinical and Health Research, University of Michigan Medical School, 2800 Plymouth Road, NCRC Building 400, #4023, Ann Arbor, MI 48109, USA
| | - Henry Appelman
- Department of Pathology, University of Michigan Medical School, 1301 Catherine, Ann Arbor, MI 48109, USA
| | - D. Kim Turgeon
- Department of Internal Medicine, University of Michigan Medical School, 1500 East Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Charles Meyer
- Department of Radiology, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI 48109, USA
| | - Tom Wang
- Departments of Medicine and Biomedical Engineering, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI 48109, USA
| |
Collapse
|
84
|
Kwon YS, Cho YS, Yoon TJ, Kim HS, Choi MG. Recent advances in targeted endoscopic imaging: Early detection of gastrointestinal neoplasms. World J Gastrointest Endosc 2012; 4:57-64. [PMID: 22442742 PMCID: PMC3309894 DOI: 10.4253/wjge.v4.i3.57] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Revised: 01/22/2012] [Accepted: 03/02/2012] [Indexed: 02/05/2023] Open
Abstract
Molecular imaging has emerged as a new discipline in gastrointestinal endoscopy. This technology encompasses modalities that can visualize disease-specific morphological or functional tissue changes based on the molecular signature of individual cells. Molecular imaging has several advantages including minimal damage to tissues, repetitive visualization, and utility for conducting quantitative analyses. Advancements in basic science coupled with endoscopy have made early detection of gastrointestinal cancer possible. Molecular imaging during gastrointestinal endoscopy requires the development of safe biomarkers and exogenous probes to detect molecular changes in cells with high specificity anda high signal-to-background ratio. Additionally, a high-resolution endoscope with an accurate wide-field viewing capability must be developed. Targeted endoscopic imaging is expected to improve early diagnosis and individual therapy of gastrointestinal cancer.
Collapse
Affiliation(s)
- Yong-Soo Kwon
- Yong-Soo Kwon, Tae-Jong Yoon, Department of Applied Bioscience, CHA University, Seoul 135081, South Korea
| | | | | | | | | |
Collapse
|
85
|
Longitudinal molecular imaging with single cell resolution of disseminated ovarian cancer in mice with a LED-based confocal microendoscope. Mol Imaging Biol 2012; 13:1157-62. [PMID: 21136184 DOI: 10.1007/s11307-010-0455-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
PURPOSE We engineered a flexible fiber-optic microendoscope for longitudinal optical imaging studies in a mouse model of disseminated ovarian cancer. PROCEDURES The microendoscope delivers 470 nm excitation light from a light-emitting diode through a fiber-optic bundle with outer diameter of 680 μm. Optics were optimized to maximize power and lateral resolution. We used this instrument to repetitively monitor intraperitoneal growth of HeyA8 ovarian cancer cells stably transduced with green fluorescent protein over 4 weeks. RESULTS The microendoscope achieves 0.7 mW power and lateral resolution of 4 μm. Initial in vivo imaging studies visualized single cells and small clusters of malignant cells with subsequent studies showing tumor masses and vasculature. We also resolved single cells within intraperitoneal tumor masses. CONCLUSIONS These studies establish microendoscope technology with single cell resolution for minimally-invasive, longitudinal imaging in living animals. This technology will advance future molecular imaging studies of ovarian cancer and other diseases.
Collapse
|
86
|
Quantitative assessment of microbicide-induced injury in the ovine vaginal epithelium using confocal microendoscopy. BMC Infect Dis 2012; 12:48. [PMID: 22375797 PMCID: PMC3315435 DOI: 10.1186/1471-2334-12-48] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Accepted: 02/29/2012] [Indexed: 11/17/2022] Open
Abstract
Background The development of safe topical microbicides that can preserve the integrity of cervicovaginal tract epithelial barrier is of great interest as this may minimize the potential for increased susceptibility to STI infections. High resolution imaging to assess epithelial integrity in a noninvasive manner could be a valuable tool for preclinical testing of candidate topical agents. Methods A quantitative approach using confocal fluorescence microendoscopy (CFM) for assessment of microbicide-induced injury to the vaginal epithelium was developed. Sheep were treated intravaginally with one of five agents in solution (PBS; 0.02% benzalkonium chloride (BZK); 0.2% BZK) or gel formulation (hydroxyethyl cellulose (HEC); Gynol II nonoxynol-9 gel (N-9)). After 24 hours the vaginal tract was removed, labeled with propidium iodide (PI), imaged, then fixed for histology. An automated image scoring algorithm was developed for quantitative assessment of injury and applied to the data set. Image-based findings were validated with histological visual gradings that describe degree of injury and measurement of epithelial thickness. Results Distinct differences in PI staining were detected following BZK and N-9 treatment. Images from controls had uniformly distributed nuclei with defined borders, while those after BZK or N-9 showed heavily stained and disrupted nuclei, which increased in proportion to injury detected on histology. The confocal scoring system revealed statistically significant scores for each agent versus PBS controls with the exception of HEC and were consistent with histology scores of injury. Conclusions Confocal microendoscopy provides a sensitive, objective, and quantitative approach for non-invasive assessment of vaginal epithelial integrity and could serve as a tool for real-time safety evaluation of emerging intravaginal topical agents.
Collapse
|
87
|
Bézière N, Ntziachristos V. Optoacoustic imaging: an emerging modality for the gastrointestinal tract. Gastroenterology 2011; 141:1979-85. [PMID: 22015916 DOI: 10.1053/j.gastro.2011.10.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Nicolas Bézière
- Institute for Biological and Medical Imaging, Technische Universität München and Helmholtz Zentrum München, Munich, Germany
| | | |
Collapse
|
88
|
Kiesslich R, Goetz M, Hoffman A, Galle PR. New imaging techniques and opportunities in endoscopy. Nat Rev Gastroenterol Hepatol 2011; 8:547-53. [PMID: 21894196 DOI: 10.1038/nrgastro.2011.152] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Gastrointestinal endoscopy is undergoing major improvements, which are driven by new available technologies and substantial refinements of optical features. In this Review, we summarize available and evolving imaging technologies that could influence the clinical algorithm of endoscopic diagnosis. Detection, characterization and confirmation are essential steps required for proper endoscopic diagnosis. Optical and nonoptical methods can help to improve each step; these improvements are likely to increase the detection rate of neoplasias and reduce unnecessary endoscopic treatments. Furthermore, functional and molecular imaging are emerging as new diagnostic tools that could provide an opportunity for personalized medicine, in which endoscopy will define disease outcome or predict the response to targeted therapy.
Collapse
Affiliation(s)
- Ralf Kiesslich
- Department of Internal Medicine, Johannes Gutenberg University of Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany.
| | | | | | | |
Collapse
|
89
|
Goetz M, Watson A, Kiesslich R. Confocal laser endomicroscopy in gastrointestinal diseases. JOURNAL OF BIOPHOTONICS 2011; 4:498-508. [PMID: 21567975 DOI: 10.1002/jbio.201100022] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Revised: 04/19/2011] [Accepted: 04/25/2011] [Indexed: 05/30/2023]
Abstract
Confocal laser endomicroscopy (CLE) is a novel endoscopic technique permitting in vivo microscopy (optical biopsies) of the gastrointestinal mucosa. CLE has been studied in a multitude of diseases of the upper and lower gastrointestinal tract, including Barrett's esophagus, gastric inflammation and cancer, celiac disease, colorectal adenoma and carcinoma, and inflammatory bowel diseases. CLE has recently evolved and been studied for bile duct and liver imaging. CLE has shown overall high accuracy and enabled smart, targeted biopsies rather than untargeted sampling. Furthermore, the availability of real time microscopic information during endoscopy has immediate impact on therapeutic decisions and guides endoscopic interventions. CLE is also a unique tool for observation of (patho-)physiologic events in their natural environment (functional imaging) and has been linked to molecular imaging of gastrointestinal neoplasia in vivo, thereby broadening our understanding of mucosal pathology in clinical and basic science.
Collapse
Affiliation(s)
- Martin Goetz
- Medizinische Klinik und Poliklinik, Universitätsmedizin Mainz, Mainz, Germany
| | | | | |
Collapse
|
90
|
Elahi SF, Wang TD. Future and advances in endoscopy. JOURNAL OF BIOPHOTONICS 2011; 4:471-81. [PMID: 21751414 PMCID: PMC3517128 DOI: 10.1002/jbio.201100048] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2011] [Revised: 06/29/2011] [Accepted: 06/30/2011] [Indexed: 05/07/2023]
Abstract
The future of endoscopy will be dictated by rapid technological advances in the development of light sources, optical fibers, and miniature scanners that will allow for images to be collected in multiple spectral regimes, with greater tissue penetration, and in three dimensions. These engineering breakthroughs will be integrated with novel molecular probes that are highly specific for unique proteins to target diseased tissues. Applications include early cancer detection by imaging molecular changes that occur before gross morphological abnormalities, personalized medicine by visualizing molecular targets specific to individual patients, and image guided therapy by localizing tumor margins and monitoring for recurrence.
Collapse
Affiliation(s)
- Sakib F. Elahi
- Department of Biomedical Engineering, Ann Arbor, Michigan 48109
| | - Thomas D. Wang
- Department of Biomedical Engineering, Ann Arbor, Michigan 48109
- Department of Medicine, Division of Gastroenterology, Ann Arbor, Michigan 48109
| |
Collapse
|
91
|
Abstract
The future of endoscopy will be dictated by rapid technological advances in the development of light sources, optical fibers, and miniature scanners that will allow for images to be collected in multiple spectral regimes, with greater tissue penetration, and in three dimensions. These engineering breakthroughs will be integrated with novel molecular probes that are highly specific for unique proteins to target diseased tissues. Applications include early cancer detection by imaging molecular changes that occur before gross morphological abnormalities, personalized medicine by visualizing molecular targets specific to individual patients, and image guided therapy by localizing tumor margins and monitoring for recurrence.
Collapse
Affiliation(s)
- Sakib F Elahi
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | |
Collapse
|
92
|
Advances in bio-optical imaging for the diagnosis of early oral cancer. Pharmaceutics 2011; 3:354-78. [PMID: 24310585 PMCID: PMC3857071 DOI: 10.3390/pharmaceutics3030354] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Accepted: 06/27/2011] [Indexed: 12/20/2022] Open
Abstract
Oral cancer is among the most common malignancies worldwide, therefore early detection and treatment is imperative. The 5-year survival rate has remained at a dismal 50% for the past several decades. The main reason for the poor survival rate is the fact that most of the oral cancers, despite the general accessibility of the oral cavity, are not diagnosed until the advanced stage. Early detection of the oral tumors and its precursor lesions may be the most effective means to improve clinical outcome and cure most patients. One of the emerging technologies is the use of non-invasive in vivo tissue imaging to capture the molecular changes at high-resolution to improve the detection capability of early stage disease. This review will discuss the use of optical probes and highlight the role of optical imaging such as autofluorescence, fluorescence diagnosis (FD), laser confocal endomicroscopy (LCE), surface enhanced Raman spectroscopy (SERS), optical coherence tomography (OCT) and confocal reflectance microscopy (CRM) in early oral cancer detection. FD is a promising method to differentiate cancerous lesions from benign, thus helping in the determination of adequate resolution of surgical resection margin. LCE offers in vivo cellular imaging of tissue structures from surface to subsurface layers and has demonstrated the potential to be used as a minimally invasive optical biopsy technique for early diagnosis of oral cancer lesions. SERS was able to differentiate between normal and oral cancer patients based on the spectra acquired from saliva of patients. OCT has been used to visualize the detailed histological features of the oral lesions with an imaging depth down to 2–3 mm. CRM is an optical tool to noninvasively image tissue with near histological resolution. These comprehensive diagnostic modalities can also be used to define surgical margin and to provide a direct assessment of the therapeutic effectiveness.
Collapse
|
93
|
Wang RCC, Deen MJ, Armstrong D, Fang Q. Development of a catadioptric endoscope objective with forward and side views. JOURNAL OF BIOMEDICAL OPTICS 2011; 16:066015. [PMID: 21721816 DOI: 10.1117/1.3593148] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Autofluorescence endoscopy is a promising functional imaging technique to improve screening of pre-cancerous or early cancer lesions in the gastrointestinal (GI) tract. Tissue autofluorescence signal is weak compared to white light reflectance imaging. Conventional forward-viewing endoscopes are inefficient in the collection of light from objects of interest along on the GI luminal wall. A key component of a complete autofluorescence endoscope is the light collection module. In this paper, we report the design, optimization, prototype development, and testing of an endoscope objective that is capable of acquiring simultaneous forward and radial views. The radial-view optical design was optimized for a balance between image quality and light collection. Modulation transfer function (MTF), entrance pupil radius, manufacturability, and field-of-view were parameters used in the lens optimization. In comparison with the typical forward-viewing endoscopes, our nonsequential ray trace simulations suggest the proposed radial-view design is more practical in the light collection. To validate the proposed simulation methods, a 3:1 scaled-up prototype was fabricated. Contrast measurements were taken with the prototype, and then compared with the simulated MTF.
Collapse
Affiliation(s)
- Roy Chih Chung Wang
- McMaster University, Department of Engineering Physics, 1280 Main Street West, Hamilton, Ontario, L8S 4L7, Canada
| | | | | | | |
Collapse
|
94
|
Neves AA, Stöckmann H, Harmston RR, Pryor HJ, Alam IS, Ireland‐Zecchini H, Lewis DY, Lyons SK, Leeper FJ, Brindle KM. Imaging sialylated tumor cell glycans
in vivo. FASEB J 2011; 25:2528-37. [DOI: 10.1096/fj.10-178590] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- André A. Neves
- Cancer Research UKCambridge Research InstituteLi Ka Shing CentreCambridgeUK
| | - Henning Stöckmann
- Cancer Research UKCambridge Research InstituteLi Ka Shing CentreCambridgeUK
- Department of BiochemistryUniversity of CambridgeCambridgeUK
| | | | - Helen J. Pryor
- Cancer Research UKCambridge Research InstituteLi Ka Shing CentreCambridgeUK
| | - Israt S. Alam
- Cancer Research UKCambridge Research InstituteLi Ka Shing CentreCambridgeUK
| | | | - David Y. Lewis
- Cancer Research UKCambridge Research InstituteLi Ka Shing CentreCambridgeUK
| | - Scott K. Lyons
- Cancer Research UKCambridge Research InstituteLi Ka Shing CentreCambridgeUK
| | | | - Kevin M. Brindle
- Cancer Research UKCambridge Research InstituteLi Ka Shing CentreCambridgeUK
- Department of ChemistryUniversity of CambridgeCambridgeUK
| |
Collapse
|
95
|
Yentz S, Wang TD. Molecular imaging for guiding oncologic prognosis and therapy in esophageal adenocarcinoma. Hosp Pract (1995) 2011; 39:97-106. [PMID: 21576902 PMCID: PMC3227392 DOI: 10.3810/hp.2011.04.399] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In the past 30 years, the incidence of esophageal adenocarcinoma (ACA) has increased significantly. Sadly, advances in treatment have not followed the same trend, and the prognosis for patients with esophageal ACA remains poor, with a 5-year survival rate of only 15%. Like most cancers, early detection is the key to improving prognosis, but this outcome has proven difficult in the esophagus for several reasons: 1) patients present with advanced disease because "alarm symptoms," such as dysphagia, occur at a late stage, and 2) high-grade dysplasia (HGD) and early ACA are not visible on routine surveillance endoscopy. Currently, the recommended surveillance strategy involves collection of random biopsies, an imperfect technique that is limited by sampling error and is infrequently used because of the considerable time and cost it requires. Even in patients with biopsy-proven dysplasia, adequate guidance for clinical management decisions is still lacking. Dysplasia alone is not an entirely reliable biomarker for the risk of progression to ACA because the natural history of this condition is extremely variable. Clearly, there is a need for additional biomarkers that can better characterize this disease and thus improve our ability to treat patients on an individual basis. As we better understand the molecular changes that lead to the development of this cancer, new molecular biomarkers are needed to allow for more personalized diagnoses, surveillance, and treatment. Targeted agents against epidermal growth factor receptor (EGFR), human epidermal growth factor receptor 2 (HER2), and vascular endothelial growth factor (VEGF) are currently being evaluated for their role in combination chemotherapy for metastatic esophageal ACA. As these studies progress, a reliable approach for determining receptor status in individual patients is essential. Molecular imaging uses fluorescent probes that target specific cell-surface receptors, and has the potential to evaluate an individual patient's gene expression profile. By topically applying fluorescent probes to dysplastic epithelium during endoscopy, a variety of receptors can be visualized, and the response to treatment can be monitored in real time. This technique can mitigate the limitations of current surveillance protocols, allow for improved cancer detection, and be used for personalized treatment in the future.
Collapse
Affiliation(s)
- Sarah Yentz
- Division of Gastroenterology and Hepatology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | | |
Collapse
|
96
|
Gheonea DI, Cârţână T, Ciurea T, Popescu C, Bădărău A, Săftoiu A. Confocal laser endomicroscopy and immunoendoscopy for real-time assessment of vascularization in gastrointestinal malignancies. World J Gastroenterol 2011; 17:21-7. [PMID: 21218080 PMCID: PMC3016676 DOI: 10.3748/wjg.v17.i1.21] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2010] [Revised: 10/19/2010] [Accepted: 10/26/2010] [Indexed: 02/06/2023] Open
Abstract
Gastrointestinal cancers represent a major cause of morbidity and mortality, with incomplete response to chemotherapy in the advanced stages and poor prognosis. Angiogenesis plays a crucial part in tumor growth and metastasis, with most gastrointestinal cancers depending strictly on the development of a new and devoted capillary network. Confocal laser endomicroscopy is a new technology which allows in vivo microscopic analysis of the gastrointestinal mucosa and its microvascularization during ongoing endoscopy by using topically or systemically administered contrast agents. Targeting markers of angiogenesis in association with confocal laser endomicroscopic examination (immunoendoscopy), as a future challenge, will add functional analysis to the morphological aspect of the neoplastic process. This review describes previous experience in endomicroscopic examination of the upper and lower digestive tract with emphasis on vascularization, resulting in a broad spectrum of potential clinical applications, and also preclinical research that could be translated to human studies.
Collapse
|
97
|
Thekkek N, Anandasabapathy S, Richards-Kortum R. Optical molecular imaging for detection of Barrett’s-associated neoplasia. World J Gastroenterol 2011; 17:53-62. [PMID: 21218084 PMCID: PMC3016680 DOI: 10.3748/wjg.v17.i1.53] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2010] [Revised: 11/17/2010] [Accepted: 11/24/2010] [Indexed: 02/06/2023] Open
Abstract
Recent advancements in the endoscopic imaging of Barrett’s esophagus can be used to probe a wide range of optical properties that are altered with neoplastic progression. This review summarizes relevant changes in optical properties as well as imaging approaches that measures those changes. Wide-field imaging approaches include narrow-band imaging that measures changes in light scattering and absorption, and autofluorescence imaging that measure changes in endogenous fluorophores. High-resolution imaging approaches include optical coherence tomography, endocytoscopy, confocal microendoscopy, and high-resolution microendoscopy. These technologies, some coupled with an appropriate contrast agent, can measure differences in glandular morphology, nuclear morphology, or vascular alterations associated with neoplasia. Advances in targeted contrast agents are further discussed. Studies that have explored these technologies are highlighted; as are the advantages and limitations of each.
Collapse
|
98
|
Atreya R, Waldner MJ, Neurath MF. Molecular imaging: interaction between basic and clinical science. Gastroenterol Clin North Am 2010; 39:911-22. [PMID: 21093763 DOI: 10.1016/j.gtc.2010.08.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
One of the major proceedings in the field of gastrointestinal endoscopy has been the advent of molecular imaging, which possesses the potential to have a significant effect on the existing diagnostic and therapeutic paradigms. Molecular imaging encompasses different methods that enable the visualization of disease-specific morphologic or functional alterations of the mucosa based on the molecular signature of individual cells. This development has been made possible by advancements in basic science coupled with technological innovations in endoscopy, both facilitating the identification and characterization of mucosal lesions in vivo based on the lesions' molecular composition rather than their morphologic structure alone. Novel studies based on fluorescent antibody imaging pave the road toward clinical translation and give hope for improved diagnosis and targeted therapies in gastrointestinal diseases.
Collapse
Affiliation(s)
- Raja Atreya
- Medical Clinic I, University of Erlangen-Nuremberg, Ulmenweg 18, 91056 Erlangen, Germany
| | | | | |
Collapse
|
99
|
Li M, Anastassiades CP, Joshi B, Komarck CM, Piraka C, Elmunzer BJ, Turgeon DK, Johnson TD, Appelman H, Beer DG, Wang TD. Affinity peptide for targeted detection of dysplasia in Barrett's esophagus. Gastroenterology 2010; 139:1472-80. [PMID: 20637198 PMCID: PMC3319360 DOI: 10.1053/j.gastro.2010.07.007] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2010] [Revised: 06/30/2010] [Accepted: 07/07/2010] [Indexed: 12/23/2022]
Abstract
BACKGROUND & AIMS Dysplasia is a premalignant condition in Barrett's esophagus that is difficult to detect on endoscopy because of its flat architecture and patchy distribution. Peptides are promising for use as novel molecular probes that identify cell surface targets unique to disease and can be fluorescence-labeled for detection. We aim to select and validate an affinity peptide that binds to esophageal dysplasia for future clinical studies. METHODS Peptide selection was performed using phage display by removing nonspecific binders using Q-hTERT (intestinal metaplasia) cells and achieving specific binding against OE33 (esophageal adenocarcinoma) cells. Selective binding was confirmed on bound phage counts, enzyme-linked immunosorbent assay (ELISA), flow cytometry, competitive inhibition, and fluorescence microscopy. On stereomicroscopy, specific peptide binding to dysplasia on endoscopically resected specimens was assessed by rigorous registration of fluorescence intensity to histology in 1-mm intervals. RESULTS The peptide sequence SNFYMPL was selected and showed preferential binding to target cells. Reduced binding was observed on competition with unlabeled peptide in a dose-dependent manner, an affinity of K(d) = 164 nmol/L was measured, and peptide binding to the surface of OE33 cells was validated on fluorescence microscopy. On esophageal specimens (n = 12), the fluorescence intensity (mean ± SEM) in 1-mm intervals classified histologically as squamous (n = 145), intestinal metaplasia (n = 83), dysplasia (n = 61), and gastric mucosa (n = 69) was 46.5 ± 1.6, 62.3 ± 5.8, 100.0 ± 9.0, and 42.4 ± 3.0 arb units, respectively. CONCLUSIONS The peptide sequence SNFYMPL binds specifically to dysplasia in Barrett's esophagus and can be fluorescence labeled to target premalignant mucosa on imaging.
Collapse
Affiliation(s)
- Meng Li
- Department of Medicine, Division of Gastroenterology, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China,Department of Pharmacy, State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Costas P. Anastassiades
- Department of Medicine, Division of Gastroenterology, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Bishnu Joshi
- Department of Medicine, Division of Gastroenterology, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Chris M. Komarck
- Department of Medicine, Division of Gastroenterology, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Cyrus Piraka
- Department of Medicine, Division of Gastroenterology, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Badih J. Elmunzer
- Department of Medicine, Division of Gastroenterology, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Danielle K. Turgeon
- Department of Medicine, Division of Gastroenterology, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | | | - Henry Appelman
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109
| | - David G. Beer
- Department of Thoracic Surgery, University of Michigan, Ann Arbor, MI, 48109
| | - Thomas D. Wang
- Department of Medicine, Division of Gastroenterology, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China,Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109
| |
Collapse
|
100
|
Abstract
The risk of developing colon cancer is increased in colitis patients, particularly if the disease is extensive and its duration long-standing. Endoscopic guidelines have been developed with the goal of detecting early neoplastic changes prior to development of advanced malignancy. Unfortunately, the natural history of this superimposed neoplastic process in colitis appears to be very heterogeneous and poorly understood. Moreover, there are numerous confounding variables in colitis patients that limit accurate assessment of the surveillance effectiveness of colonoscopy and multi-site biopsy protocols. Although the clinical challenge posed to even the most experienced clinicians remains significant, evolving methods of endoscopic imaging may facilitate better evaluation of this highly select group of patients.
Collapse
|