1
|
Ren T, Yang MZ, Zhang WM, Qin LJ, Zhou SC, Cheng NN, Huang YJ, Sun J, Xu N, Sun HB, Zhang BB. A novel fluorescent dye selectively images and kills cancer stem cells by targeting mitochondria: Evidence from a cell line‑based zebrafish xenograft model. Oncol Lett 2023; 26:472. [PMID: 37809044 PMCID: PMC10551866 DOI: 10.3892/ol.2023.14058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 08/23/2023] [Indexed: 10/10/2023] Open
Abstract
Numerous agents such as near-infrared dyes that are characterized by specialized cancer imaging and cytotoxicity effects have key roles in cancer diagnosis and therapy via molecularly targeting special biological tissues, organelles and processes. In the present study, a novel fluorescent compound was demonstrated to inhibit cancer cell proliferation in a zebrafish model with slight in vivo toxicity. Further studies demonstrated selective staining of cancer cells and even putative cancer stem cells via accumulation of the dye in the mitochondria of cancer cells, compared with normal cells. Moreover, this compound was also used to image cancer cells in vivo using a zebrafish model. The compound displayed no apparent toxicity to the host animal. Overall, the data indicated that this compound was worthy of further evaluation due to its low toxicity and selective cancer cell imaging and killing effects. It could be a useful tool in cancer research.
Collapse
Affiliation(s)
- Tao Ren
- Department of Clinical Oncology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530000, P.R. China
| | - Meng-Zhe Yang
- Graduate School, Beijing TongRen Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Wei-Ming Zhang
- Department of Clinical Oncology, Wuming Hospital of Guangxi Medical University, Nanning, Guangxi 530199, P.R. China
| | - Liu-Jie Qin
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi 530000, P.R. China
| | - Shou-Chang Zhou
- Life Science Institute, Guangxi Medical University, Nanning, Guangxi 530000, P.R. China
| | - Nan-Nan Cheng
- Laboratory of Clinical Medicine, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| | - Yuan-Jiao Huang
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi 530000, P.R. China
- Life Science Institute, Guangxi Medical University, Nanning, Guangxi 530000, P.R. China
| | - Jing Sun
- School of Pharmacy, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Ning Xu
- Department of Clinical Oncology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530000, P.R. China
| | - Hua-Bing Sun
- School of Pharmacy, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Bei-Bei Zhang
- Institute of Biomedical Research, Yunnan University, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
2
|
Navolokin N, Lomova M, Bucharskaya A, Godage O, Polukonova N, Shirokov A, Grinev V, Maslyakova G. Antitumor Effects of Microencapsulated Gratiola officinalis Extract on Breast Carcinoma and Human Cervical Cancer Cells In Vitro. MATERIALS (BASEL, SWITZERLAND) 2023; 16:1470. [PMID: 36837099 PMCID: PMC9960207 DOI: 10.3390/ma16041470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/04/2023] [Accepted: 02/07/2023] [Indexed: 06/18/2023]
Abstract
Flavonoid-containing Gratiola officinalis extract has been studied in relation to breast carcinoma and human cervical cancer cells in encapsulated and native form. Encapsulation was realized in polymer shells, which were formed by the layer-by-layer method using sequential adsorption of poly(allylamine hydrochloride) and poly(sodium 4-styrenesulfonate) on the destructible cores. The extract was prepared by the author's method and characterized using high performance liquid chromatography. By means of optical and fluorescent microscopy, cell changes under the action of pure and encapsulated extracts were comprehensively studied, and statistical analysis was carried out. Cells were stained with propidium iodide, acridine orange, and Hoechst 33258. A fluorescence microscope with a digital video camera were used for cell imaging. The encapsulated extract caused 100% death of breast cancer SKBR-3 cells and 34% death of cervical cancer HeLa cells and prevented the formation of autophagosomes in both cultures. Analysis of the viability and morphological features of tumor cells under the action of microencapsulated extract allows us to consider microencapsulation as an effective strategy for delivering Gratiola officinalis extract to tumor cells and a promising way to overcome the protective autophagy.
Collapse
Affiliation(s)
- Nikita Navolokin
- Center for Collective Use of Experimental Oncology, Saratov State Medical University n.a. V.I. Razumovsky, Saratov 410012, Russia
- Science Medical Centre, Saratov State University, Saratov 410012, Russia
| | - Maria Lomova
- Science Medical Centre, Saratov State University, Saratov 410012, Russia
| | - Alla Bucharskaya
- Center for Collective Use of Experimental Oncology, Saratov State Medical University n.a. V.I. Razumovsky, Saratov 410012, Russia
- Science Medical Centre, Saratov State University, Saratov 410012, Russia
- Laser Molecular Imaging and Machine Learning Laboratory, Tomsk State University, Tomsk 634050, Russia
| | - Olga Godage
- Center for Collective Use of Experimental Oncology, Saratov State Medical University n.a. V.I. Razumovsky, Saratov 410012, Russia
| | - Natalya Polukonova
- Center for Collective Use of Experimental Oncology, Saratov State Medical University n.a. V.I. Razumovsky, Saratov 410012, Russia
| | - Alexander Shirokov
- Center for Collective Use of Experimental Oncology, Saratov State Medical University n.a. V.I. Razumovsky, Saratov 410012, Russia
- Science Medical Centre, Saratov State University, Saratov 410012, Russia
- Institute of Biochemistry and Physiology of Plants and Microorganisms, Russian Academy of Sciences (IBPPM RAS), Saratov 410028, Russia
| | - Vyacheslav Grinev
- Science Medical Centre, Saratov State University, Saratov 410012, Russia
- Institute of Biochemistry and Physiology of Plants and Microorganisms, Russian Academy of Sciences (IBPPM RAS), Saratov 410028, Russia
| | - Galina Maslyakova
- Center for Collective Use of Experimental Oncology, Saratov State Medical University n.a. V.I. Razumovsky, Saratov 410012, Russia
- Science Medical Centre, Saratov State University, Saratov 410012, Russia
| |
Collapse
|
3
|
Waldner MJ, Neurath MF. Molecular Endoscopy for the Diagnosis and Therapeutic Monitoring of Colorectal Cancer. Front Oncol 2022; 12:835256. [PMID: 35280747 PMCID: PMC8913894 DOI: 10.3389/fonc.2022.835256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 01/26/2022] [Indexed: 11/23/2022] Open
Abstract
Colorectal cancer (CRC) is one of the leading causes of cancer related death in the western world. Its successful treatment requires early detection and removal of precursor lesions as well as individualized treatment of advanced disease. During recent years, molecular imaging techniques have shown promising results to improve current clinical practice. For instance, molecular endoscopy resulted in higher detection rates of precursors in comparison to conventional endoscopy in preclinical and clinical studies. Molecular confocal endomicroscopy allowed a further classification of suspect lesions as well as the prediction and monitoring of the therapeutic response. In this review, we summarize recent achievements for molecular imaging of CRC in preclinical studies, initial clinical trials and the remaining challenges for future translation into clinical practice.
Collapse
Affiliation(s)
- Maximilian J Waldner
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Markus F Neurath
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
4
|
Zhu G, Wu Z, Lui S, Hu N, Wu M. Advances in Imaging Modalities and Contrast Agents for the Early Diagnosis of Colorectal Cancer. J Biomed Nanotechnol 2021; 17:558-581. [PMID: 35057884 DOI: 10.1166/jbn.2021.3064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Colorectal cancer is one of the most common gastrointestinal cancers worldwide. The mortality rate of colorectal cancer has declined by more than 20% due to the rapid development of early diagnostic techniques and effective treatment. At present, there are many diagnostic modalities
available for the evaluation of colorectal cancer, such as the carcinoembryonic antigen test, the fecal occult blood test, endoscopy, X-ray barium meal, computed tomography, magnetic resonance imaging, and radionuclide examination. Sensitive and specific imaging modalities have played an increasingly
important role in the diagnosis of colorectal cancer following the rapid development of novel contrast agents. This review discusses the applications and challenges of different imaging techniques and contrast agents applied to detect colorectal cancer, for the purpose of the early diagnosis
and treatment of patients with colorectal cancer.
Collapse
Affiliation(s)
- Guannan Zhu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zijun Wu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Su Lui
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Na Hu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Min Wu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
5
|
Ahmed S, Galle PR, Neumann H. Molecular endoscopic imaging: the future is bright. Ther Adv Gastrointest Endosc 2019; 12:2631774519867175. [PMID: 31517311 PMCID: PMC6724493 DOI: 10.1177/2631774519867175] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/10/2019] [Indexed: 12/24/2022] Open
Abstract
The prediction and final survival rate of gastrointestinal cancers are dependent on the stage of disease. The ideal would be to detect those gastrointestinal lesions at early stage or even premalignant forms which are difficult to detect by conventional endoscopy with white light optical imaging as they show minimum or no changes in morphological characteristics and are thus left untreated. The introduction of molecular imaging has greatly changed the pattern for detecting gastrointestinal lesions from purely macroscopic structural imaging to the molecular level. It allows microscopic examination of the gastrointestinal mucosa with endoscopy after the topical or systemic application of molecular probes. In recent years, major advancements in endoscopic instruments and specific molecular probes have been achieved. This review focuses on the current status of endoscopic imaging and highlights the application of molecular imaging in gastrointestinal and hepatic disease in the context of diagnosis and therapy based on recently published literature in this field. We also discuss the challenges of molecular endoscopic imaging, its future directions and potential that could have a tremendous impact on endoscopic research and clinical practice in future.
Collapse
Affiliation(s)
- Shakil Ahmed
- Department of Interdisciplinary Endoscopy, I. Medical Clinic and Polyclinic, University Hospital Mainz, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Peter R Galle
- Department of Interdisciplinary Endoscopy, I. Medical Clinic and Polyclinic, University Hospital Mainz, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Helmut Neumann
- Department of Interdisciplinary Endoscopy, I. Medical Clinic and Polyclinic, University Hospital Mainz, Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
6
|
Kopansky-Groisman E, Kogan-Zviagin I, Sella-Tavor O, Oron-Herman M, David A. Near-Infrared Fluorescent Activated Polymeric Probe for Imaging Intraluminal Colorectal Cancer Tumors. Biomacromolecules 2019; 20:3547-3556. [PMID: 31381303 DOI: 10.1021/acs.biomac.9b00806] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Detection and removal of preneoplastic tumors is crucial for successful colorectal cancer (CRC) therapy. Here we describe the design of a Cathepsin B (CB)-activated polymeric probe, P-(GGFLGK-IR783), for imaging CRC tumors established by intrarectal or subcutaneous (s.c.) implantation of human colon cancer cells (SW-480 and HT-29) in mice. Multiple copies of the near-infrared fluorescent (NIRF) dye IR783 were attached to a single HPMA copolymer backbone via a CB-cleavable linker (GFLG), and the influence of the dye loading on the fluorescence quenching and activation by CB was assessed in vitro, ex vivo, and in vivo. The optimal dose and dosing regimen of P-(GGFLGK-IR783) for colonic tumor detection was determined. Increasing the IR783 loading in the copolymer from 2.5 to 20 mol % resulted in quenching of the fluorescence signal that was activated in vitro by the action of CB from different origins. Following intravenous administration, P-(GGFLGK-IR783)7.5% preferentially accumulated in intrarectal and s.c. implanted tumors, allowing tumor visualization after 4 h and even 48 h postadministration. Activation of P-(GGFLGK-IR783)7.5% by CB was clearly detected in s.c. implanted tumors, revealing about a 4-fold increase in the fluorescence signal in tumors vs healthy colon tissue. The probe containing the CB-cleavable linker produced higher fluorescence signal intensity in tumors, relative to the noncleavable probe. These results indicate that P-(GGFLGK-IR783)7.5% may aid in detecting CRC tumors and can help to guide selective removal of polyps during colonoscopic procedures.
Collapse
Affiliation(s)
- Eva Kopansky-Groisman
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences , Ben-Gurion University of the Negev , Beer-Sheva 84105 , Israel
| | - Inga Kogan-Zviagin
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences , Ben-Gurion University of the Negev , Beer-Sheva 84105 , Israel
| | | | - Mor Oron-Herman
- Advanced Technology Center, Sheba Medical Center , Tel-Hashomer 52621 , Israel
| | - Ayelet David
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences , Ben-Gurion University of the Negev , Beer-Sheva 84105 , Israel
| |
Collapse
|
7
|
Bogdanov AA, Solovyev ID, Savitsky AP. Sensors for Proteolytic Activity Visualization and Their Application in Animal Models of Human Diseases. BIOCHEMISTRY (MOSCOW) 2019; 84:S1-S18. [PMID: 31213192 DOI: 10.1134/s0006297919140013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Various sensors designed for optical and photo(opto)acoustic imaging in living systems are becoming essential components of basic and applied biomedical research. Some of them including those developed for determining enzyme activity in vivo are becoming commercially available. These sensors can be used for various fluorescent signal detection methods: from whole body tomography to endoscopy with miniature cameras. Sensor molecules including enzyme-cleavable macromolecules carrying multiple quenched near-infrared fluorophores are able to deliver their payload in vivo and have long circulation time in bloodstream enabling detection of enzyme activity for extended periods of time at low doses of these sensors. In the future, more effective "activated" probes are expected to become available with optimized sensitivity to enzymatic activity, spectral characteristics suitable for intraoperative imaging of surgical field, biocompatibility and lack of immunogenicity and toxicity. New in vivo optical imaging methods such as the fluorescence lifetime and photo(opto)acoustic imaging will contribute to early diagnosis of human diseases. The use of sensors for in vivo optical imaging will include more extensive preclinical applications of experimental therapies. At the same time, the ongoing development and improvement of optical signal detectors as well as the availability of biologically inert and highly specific fluorescent probes will further contribute to the introduction of fluorescence imaging into the clinic.
Collapse
Affiliation(s)
- A A Bogdanov
- University of Massachusetts Medical School, Department of Radiology, Laboratory of Molecular Imaging Probes, Worcester, MA 01655, USA. .,A. N. Bach Institute of Biochemistry, Federal Research Center "Fundamentals of Biotechnology", Russian Academy of Sciences, Laboratory of Molecular Imaging, Moscow, 119071, Russia.,Lomonosov Moscow State University, Faculty of Bioengineering and Bioinformatics, Moscow, 119991, Russia
| | - I D Solovyev
- A. N. Bach Institute of Biochemistry, Federal Research Center "Fundamentals of Biotechnology", Russian Academy of Sciences, Laboratory of Molecular Imaging, Moscow, 119071, Russia.,A. N. Bach Institute of Biochemistry, Fundamentals of Biotechnology Federal Research Center, Russian Academy of Sciences, Laboratory of Physical Biochemistry, Moscow, 119071, Russia
| | - A P Savitsky
- A. N. Bach Institute of Biochemistry, Federal Research Center "Fundamentals of Biotechnology", Russian Academy of Sciences, Laboratory of Molecular Imaging, Moscow, 119071, Russia.,A. N. Bach Institute of Biochemistry, Fundamentals of Biotechnology Federal Research Center, Russian Academy of Sciences, Laboratory of Physical Biochemistry, Moscow, 119071, Russia
| |
Collapse
|
8
|
Ahmed S, Strand S, Weinmann-Menke J, Urbansky L, Galle PR, Neumann H. Molecular endoscopic imaging in cancer. Dig Endosc 2018; 30:719-729. [PMID: 29846982 DOI: 10.1111/den.13199] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 05/21/2018] [Indexed: 12/14/2022]
Abstract
Cancer is one of the major causes of death in both the USA and Europe. Molecular imaging is a novel field that is revolutionizing cancer management. It is based on the molecular signature of cells in order to study the human body both in normal and diseased conditions. The emergence of molecular imaging has been driven by the difficulties associated with cancer detection, particularly early-stage premalignant lesions which are often unnoticed as a result of minimal or no structural changes. Endoscopic surveillance is the standard method for early-stage cancer detection. In addition to recent major advancements in endoscopic instruments, significant progress has been achieved in the exploration of highly specific molecular probes and the combination of both will permit significant improvement of patient care. In this review, we provide an outline of the current status of endoscopic imaging and focus on recent applications of molecular imaging in gastrointestinal, hepatic and other cancers in the context of detection, targeted therapy and personalized medicine. As new imaging agents have the potential to broadly expand our cancer diagnostic capability, we will also present an overview of the main types of optical molecular probes with their pros and cons. We conclude by discussing the challenges and future prospects of the field.
Collapse
Affiliation(s)
- Shakil Ahmed
- Department of Interdisciplinary Endoscopy, I. Medical Clinic and Polyclinic, University Hospital Mainz, Mainz, Germany
| | - Susanne Strand
- Department of Interdisciplinary Endoscopy, I. Medical Clinic and Polyclinic, University Hospital Mainz, Mainz, Germany
| | - Julia Weinmann-Menke
- Department of Interdisciplinary Endoscopy, I. Medical Clinic and Polyclinic, University Hospital Mainz, Mainz, Germany
| | - Lana Urbansky
- Department of Interdisciplinary Endoscopy, I. Medical Clinic and Polyclinic, University Hospital Mainz, Mainz, Germany
| | - Peter R Galle
- Department of Interdisciplinary Endoscopy, I. Medical Clinic and Polyclinic, University Hospital Mainz, Mainz, Germany
| | - Helmut Neumann
- Department of Interdisciplinary Endoscopy, I. Medical Clinic and Polyclinic, University Hospital Mainz, Mainz, Germany
| |
Collapse
|
9
|
Ding S, Blue RE, Moorefield E, Yuan H, Lund PK. Ex Vivo and In Vivo Noninvasive Imaging of Epidermal Growth Factor Receptor Inhibition on Colon Tumorigenesis Using Activatable Near-Infrared Fluorescent Probes. Mol Imaging 2018; 16:1536012117729044. [PMID: 28884622 PMCID: PMC5595252 DOI: 10.1177/1536012117729044] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background: Near-infrared fluorescence (NIRF) imaging combined with enzyme-activatable NIRF probes has yielded promising results in cancer detection. Objective: To test whether 3-dimensional (3-D) noninvasive in vivo NIRF imaging can detect effects of epidermal growth factor receptor (EGFR) inhibitor on both polypoid and flat tumor load in azoxymethane (AOM)-induced colon tumors or tumors in ApcMin/+ mice. Methods: The AOM-injected KK-HIJ mice received EGFR inhibitor diet or chow diet. These and ApcMin/+ mice were given cathepsin-activatable probes (ProSense 680) before imaging. In vivo imaging was performed using quantitative tomographic NIRF imaging. Ex vivo imaging and histologic examination were performed. Dual imaging by micro computed tomography (CT) and 3D NIRF imaging was used to verify tumor location. Results: Tumor load reduction by EGFR inhibition was detected ex vivo using cathepsin B probes. In vivo imaging revealed intense activation of probes only in large tumors. Dual imaging with microCT and 3D NIRF imaging improved tumor detection in vivo. Conclusions: The 3-D NIRF imaging with ProSense 680 can detect and quantify drug effects on colon tumors ex vivo. The NIRF imaging with ProSense 680 probe has limitations as a valid nonendoscopic method for intestinal tumor detection. Combing with other imaging modalities will improve the specificity and sensitivity of intestinal tumor detection in vivo.
Collapse
Affiliation(s)
- Shengli Ding
- 1 Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Randall E Blue
- 1 Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Emily Moorefield
- 1 Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hong Yuan
- 2 Department of Radiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Pauline K Lund
- 1 Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
10
|
Waldner MJ, Rath T, Schürmann S, Bojarski C, Atreya R. Imaging of Mucosal Inflammation: Current Technological Developments, Clinical Implications, and Future Perspectives. Front Immunol 2017; 8:1256. [PMID: 29075256 PMCID: PMC5641553 DOI: 10.3389/fimmu.2017.01256] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 09/21/2017] [Indexed: 12/12/2022] Open
Abstract
In recent years, various technological developments markedly improved imaging of mucosal inflammation in patients with inflammatory bowel diseases. Although technological developments such as high-definition-, chromo-, and autofluorescence-endoscopy led to a more precise and detailed assessment of mucosal inflammation during wide-field endoscopy, probe-based and stationary confocal laser microscopy enabled in vivo real-time microscopic imaging of mucosal surfaces within the gastrointestinal tract. Through the use of fluorochromes with specificity against a defined molecular target combined with endoscopic techniques that allow ultrastructural resolution, molecular imaging enables in vivo visualization of single molecules or receptors during endoscopy. Molecular imaging has therefore greatly expanded the clinical utility and applications of modern innovative endoscopy, which include the diagnosis, surveillance, and treatment of disease as well as the prediction of the therapeutic response of individual patients. Furthermore, non-invasive imaging techniques such as computed tomography, magnetic resonance imaging, scintigraphy, and ultrasound provide helpful information as supplement to invasive endoscopic procedures. In this review, we provide an overview on the current status of advanced imaging technologies for the clinical non-invasive and endoscopic evaluation of mucosal inflammation. Furthermore, the value of novel methods such as multiphoton microscopy, optoacoustics, and optical coherence tomography and their possible future implementation into clinical diagnosis and evaluation of mucosal inflammation will be discussed.
Collapse
Affiliation(s)
- Maximilian J. Waldner
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Timo Rath
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Sebastian Schürmann
- Institute of Medical Biotechnology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christian Bojarski
- Department of Gastroenterology, Infectiology and Rheumatology, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Raja Atreya
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
11
|
Li K, Liu Y, Zhang S, Xu Y, Jiang J, Yin F, Hu Y, Han B, Ge S, Zhang L, Wang Y. Folate receptor-targeted ultrasonic PFOB nanoparticles: Synthesis, characterization and application in tumor-targeted imaging. Int J Mol Med 2017; 39:1505-1515. [PMID: 28487935 PMCID: PMC5428942 DOI: 10.3892/ijmm.2017.2975] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 04/24/2017] [Indexed: 12/22/2022] Open
Abstract
In this study, we aimed to determine an effective strategy for the synthesis of folate receptor (FR) targeted-nanoparticles (FRNPs). The nanoparticles used as ultrasound contrast agents (UCAs) were composed of a liquid core of perfluorooctyl bromide (PFOB) liposome and a targeted shell chemically conjugated with folic acid (FA) and polyethylene glycol (PEG). This was done in order to avoid recognition and clearance by the mononuclear phagocyte system [also known as the reticuloendothelial system (RES)] and enhance the targeting capability of the nanoparticles to tumors overexpressing folate receptor (FR). The FRNPs exhibited an average particle size of 301±10.8 nm and surface potential of 39.1±0.43 mV. Subsequently, in vitro, FRNPs labeled with FITC fluorescence dye were visibly uptaken into the cytoplasm of FR-overexpressing cancer cells (Bel7402 and SW620 cells), whereas the A549 cells expressing relatively low levels of FR just bound with few FRNPs. These results demonstrated that FRNPs have a high affinity to FR-overexpressing cancer cells. Additionally, in in vivo experiments, FRNPs achieved a greater enhancement of tumor ultrasound imaging and a longer enhancement time in FR-overexpressing tumors and the Cy7-labeled FRNPs exhibited a relatively high tumor-targeted distribution in FR-overexpressing tumors. Targeted ultrasound and fluorescence imaging revealed that FRNPs have the ability to target FR-overexpressing tumors and ex vivo fluorescence imaging was then used to further verify and confirm the presence of FRNPs in tumor tissues with histological analysis of the tumor slices. On the whole, our data demonstrate that the FRNPs may prove to be a promising candidate for the early diagnosis for FR-overexpressing tumors at the molecular and cellular levels.
Collapse
Affiliation(s)
- Keshi Li
- Department of Hepatobiliary Surgery, Ningbo First Hospital, Haishu, Ningbo, Zhejiang 315010, P.R. China
| | - Yahui Liu
- Department of Hepatobiliary Surgery, Ningbo First Hospital, Haishu, Ningbo, Zhejiang 315010, P.R. China
| | - Shengmin Zhang
- Department of Hepatobiliary Surgery, Ningbo First Hospital, Haishu, Ningbo, Zhejiang 315010, P.R. China
| | - Youfeng Xu
- Department of Hepatobiliary Surgery, Ningbo First Hospital, Haishu, Ningbo, Zhejiang 315010, P.R. China
| | - Jianshuai Jiang
- Department of Hepatobiliary Surgery, Ningbo First Hospital, Haishu, Ningbo, Zhejiang 315010, P.R. China
| | - Fengying Yin
- Department of Hepatobiliary Surgery, Ningbo First Hospital, Haishu, Ningbo, Zhejiang 315010, P.R. China
| | - Yue Hu
- Department of Hepatobiliary Surgery, Ningbo First Hospital, Haishu, Ningbo, Zhejiang 315010, P.R. China
| | - Baosan Han
- Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Yangpu, Shanghai 200092, P.R. China
| | - Shuxiong Ge
- Ningbo Medical School of Ningbo University, Jiangbei, Ningbo, Zhejiang 315211, P.R. China
| | - Li Zhang
- Ningbo Medical School of Ningbo University, Jiangbei, Ningbo, Zhejiang 315211, P.R. China
| | - Yong Wang
- Department of Hepatobiliary Surgery, Ningbo First Hospital, Haishu, Ningbo, Zhejiang 315010, P.R. China
| |
Collapse
|
12
|
Belykh E, Martirosyan NL, Yagmurlu K, Miller EJ, Eschbacher JM, Izadyyazdanabadi M, Bardonova LA, Byvaltsev VA, Nakaji P, Preul MC. Intraoperative Fluorescence Imaging for Personalized Brain Tumor Resection: Current State and Future Directions. Front Surg 2016; 3:55. [PMID: 27800481 PMCID: PMC5066076 DOI: 10.3389/fsurg.2016.00055] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 09/12/2016] [Indexed: 12/29/2022] Open
Abstract
INTRODUCTION Fluorescence-guided surgery is one of the rapidly emerging methods of surgical "theranostics." In this review, we summarize current fluorescence techniques used in neurosurgical practice for brain tumor patients as well as future applications of recent laboratory and translational studies. METHODS Review of the literature. RESULTS A wide spectrum of fluorophores that have been tested for brain surgery is reviewed. Beginning with a fluorescein sodium application in 1948 by Moore, fluorescence-guided brain tumor surgery is either routinely applied in some centers or is under active study in clinical trials. Besides the trinity of commonly used drugs (fluorescein sodium, 5-aminolevulinic acid, and indocyanine green), less studied fluorescent stains, such as tetracyclines, cancer-selective alkylphosphocholine analogs, cresyl violet, acridine orange, and acriflavine, can be used for rapid tumor detection and pathological tissue examination. Other emerging agents, such as activity-based probes and targeted molecular probes that can provide biomolecular specificity for surgical visualization and treatment, are reviewed. Furthermore, we review available engineering and optical solutions for fluorescent surgical visualization. Instruments for fluorescent-guided surgery are divided into wide-field imaging systems and hand-held probes. Recent advancements in quantitative fluorescence-guided surgery are discussed. CONCLUSION We are standing on the threshold of the era of marker-assisted tumor management. Innovations in the fields of surgical optics, computer image analysis, and molecular bioengineering are advancing fluorescence-guided tumor resection paradigms, leading to cell-level approaches to visualization and resection of brain tumors.
Collapse
Affiliation(s)
- Evgenii Belykh
- Department of Neurosurgery, St. Joseph’s Hospital and Medical Center, Barrow Neurological Institute, Phoenix, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
- Laboratory of Neurosurgery, Irkutsk Scientific Center of Surgery and Traumatology, Irkutsk, Russia
- Irkutsk State Medical University, Irkutsk, Russia
| | - Nikolay L. Martirosyan
- Department of Neurosurgery, St. Joseph’s Hospital and Medical Center, Barrow Neurological Institute, Phoenix, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Kaan Yagmurlu
- Department of Neurosurgery, St. Joseph’s Hospital and Medical Center, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Eric J. Miller
- University of Arizona College of Medicine – Phoenix, Phoenix, AZ, USA
| | - Jennifer M. Eschbacher
- Department of Neurosurgery, St. Joseph’s Hospital and Medical Center, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Mohammadhassan Izadyyazdanabadi
- Department of Neurosurgery, St. Joseph’s Hospital and Medical Center, Barrow Neurological Institute, Phoenix, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Liudmila A. Bardonova
- Department of Neurosurgery, St. Joseph’s Hospital and Medical Center, Barrow Neurological Institute, Phoenix, AZ, USA
- Laboratory of Neurosurgery, Irkutsk Scientific Center of Surgery and Traumatology, Irkutsk, Russia
- Irkutsk State Medical University, Irkutsk, Russia
| | - Vadim A. Byvaltsev
- Laboratory of Neurosurgery, Irkutsk Scientific Center of Surgery and Traumatology, Irkutsk, Russia
- Irkutsk State Medical University, Irkutsk, Russia
| | - Peter Nakaji
- Department of Neurosurgery, St. Joseph’s Hospital and Medical Center, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Mark C. Preul
- Department of Neurosurgery, St. Joseph’s Hospital and Medical Center, Barrow Neurological Institute, Phoenix, AZ, USA
| |
Collapse
|
13
|
Xie J, Yan C, Yan Y, Chen L, Song L, Zang F, An Y, Teng G, Gu N, Zhang Y. Multi-modal Mn-Zn ferrite nanocrystals for magnetically-induced cancer targeted hyperthermia: a comparison of passive and active targeting effects. NANOSCALE 2016; 8:16902-15. [PMID: 27427416 DOI: 10.1039/c6nr03916b] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
The high performance and increased tumor-targeting accumulation of magnetic nanocrystals (MNCs) are the most important considerations in cancer targeted magnetic hyperthermia (TMH). To achieve these goals, our study was firstly done using well-established fluorescence/magnetic Mn-Zn ferrite MNCs (core size: 14 nm) as multi-modal imaging contrast agents and highly-efficient "heat generators", which were coated with a biocompatible PEG-phospholipid (DSPE-PEG2000) and further modified by a cyclic tripeptide of arginine-glycine-aspartic acid (RGD). By using a mouse model bearing breast carcinoma (4T1), we then systematically compared PEGylated MNCs (MNCs@PEG)- and RGD-PEGylated MNCs (MNCs@RGD)-mediated tumor targeting abilities by intravenous administration. The MNCs@PEG-based passive targeting could successfully accumulate at the tumor due to the enhanced permeability and retention (EPR) effects, but the non-targeted localization might make the MNCs@PEG "leaking" from larger pores of tumor fenestrated vascular networks. Our designed MNCs@RGD, simultaneously functionalized with PEG and RGD ligands, might promote a synergistic effect including efficient tumor vasculature active targeting and EPR-mediated passive targeting, improving total MNC concentration and retention time in tumor tissues. By combining fluorescence/magnetic resonance (MR)/thermal multi-modal imaging-guided diagnostics and continuous TMH treatment under an alternating current magnetic field (ACMF, 2.58 kA m(-1), 390 kHz), the tumor surface could be heated to approximately 43-44 °C based on the MNC-mediated repeated injections. Sufficient temperature elevation induced the apoptosis of tumor cells, and inhibited the tumor angiogenesis. Compared with MNCs@PEG, the active MNCs@RGD-based tumor targeting MR image was significantly more efficient due to both the higher and long-lasting tumor accumulation, but its antitumor efficacy was not obviously improved in the TMH treatments. To achieve a singularly promising tumor TMH therapy, a greatly increased MNC content in tumor was needed. This insight indicated that not only the tumor vasculature targeting, but also the active tumor cells targeting of MNCs should receive considerable attention in future clinical TMH therapy application.
Collapse
Affiliation(s)
- Jun Xie
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Collaborative Innovation Center of Suzhou Nano Science and Technology, Southeast University, Nanjing 210096, P. R. China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Hodgson A, Wier EM, Fu K, Sun X, Wan F. Ultrasound imaging of splenomegaly as a proxy to monitor colon tumor development in Apc(min716/+) mice. Cancer Med 2016; 5:2469-76. [PMID: 27485505 PMCID: PMC5055147 DOI: 10.1002/cam4.842] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 07/02/2016] [Accepted: 07/04/2016] [Indexed: 01/13/2023] Open
Abstract
Animal models of colon cancer are widely used to understand the molecular mechanisms and pathogenesis of the disease. These animal models require a substantial investment of time and traditionally necessitate the killing of the animal to measure the tumor progression. Several in vivo imaging techniques are being used in both human clinics and preclinical studies, albeit at high cost and requiring particular expertise. Here, we report that the progression of splenomegaly coincides with and positively correlates to colon tumor development in Apcmin716/+ mice expressing a mutant gene encoding an adenomatous polyposis coli protein truncated at amino acid 716. Ultrasound image‐based spleen size measurement precisely mirrors splenomegaly development in vivo in the tumor‐laden Apcmin716/+ mice. Moreover, the spleen dimensions extracted from the ultrasound sonograms are positively correlated with normalized spleen weight and the number and area of colon tumors. Hence, we propose measuring the spleen size in vivo by ultrasound imaging as a novel approach to estimate splenomegaly development and to indirectly monitor colon tumor development in Apcmin716/+ mice. The widespread use of ultrasound machines in the laboratory setting, coupled with the fact that it is a noninvasive method, make it a straightforward and useful tool for monitoring the experimental progress of colon cancer in mice and determining end points without killing animals strictly for diagnostics purposes.
Collapse
Affiliation(s)
- Andrea Hodgson
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, 21025
| | - Eric M Wier
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, 21025
| | - Kai Fu
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, 21025
| | - Xin Sun
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, 21025
| | - Fengyi Wan
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, 21025. .,Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, 21287.
| |
Collapse
|
15
|
Tontini GE, Rath T, Neumann H. Advanced gastrointestinal endoscopic imaging for inflammatory bowel diseases. World J Gastroenterol 2016; 22:1246-1259. [PMID: 26811662 PMCID: PMC4716035 DOI: 10.3748/wjg.v22.i3.1246] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 10/15/2015] [Accepted: 11/09/2015] [Indexed: 02/06/2023] Open
Abstract
Gastrointestinal luminal endoscopy is of paramount importance for diagnosis, monitoring and dysplasia surveillance in patients with both, Crohn's disease and ulcerative colitis. Moreover, with the recent recognition that mucosal healing is directly linked to the clinical outcome of patients with inflammatory bowel disorders, a growing demand exists for the precise, timely and detailed endoscopic assessment of superficial mucosal layer. Further, the novel field of molecular imaging has tremendously expanded the clinical utility and applications of modern endoscopy, now encompassing not only diagnosis, surveillance, and treatment but also the prediction of individual therapeutic responses. Within this review, we describe how novel endoscopic approaches and advanced endoscopic imaging methods such as high definition and high magnification endoscopy, dye-based and dye-less chromoendoscopy, confocal laser endomicroscopy, endocytoscopy and molecular imaging now allow for the precise and ultrastructural assessment of mucosal inflammation and describe the potential of these techniques for dysplasia detection.
Collapse
|
16
|
Rath T, Tontini GE, Neurath MF, Neumann H. From the surface to the single cell: Novel endoscopic approaches in inflammatory bowel disease. World J Gastroenterol 2015; 21:11260-11272. [PMID: 26523101 PMCID: PMC4616203 DOI: 10.3748/wjg.v21.i40.11260] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 07/31/2015] [Accepted: 09/14/2015] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel diseases (IBD) comprise the two major entities Crohn's disease and ulcerative colitis and endoscopic imaging of the gastrointestinal tract has always been an integral and central part in the management of IBD patients. Within the recent years, mucosal healing emerged as a key treatment goal in IBD that substantially decides about the clinical outcome of IBD patients, thereby demanding for a precise, timely and detailed endoscopic assessment of the mucosal inflammation associated with IBD. Further, molecular imaging has tremendously expanded the clinical utility and applications of modern endoscopy, now encompassing not only diagnosis, surveillance, and treatment but also the prediction of individual therapy response. Within this review we describe novel endoscopic approaches and advanced endoscopic imaging methods for the diagnosis, treatment and surveillance of IBD patients. We begin by providing an overview over novel and advanced imaging techniques such as magnification endoscopy and dye-based and dye-less chromoendoscopy, endomicroscopy and endocytoscopy. We then describe how these techniques can be utilized for the precise and ultrastructural assessment of mucosal inflammation and dysplasia development associated with IBD and outline how they have enabled the endoscopist to gain insight onto the cellular level in real-time. Finally, we provide an outlook on how molecular imaging has rapidly evolved in the recent past and can be used to make individual predictions about the therapeutic response towards biological treatment.
Collapse
|
17
|
Korb ML, Warram JM, Grudzinski J, Weichert J, Jeffery J, Rosenthal EL. Breast cancer imaging using the near-infrared fluorescent agent, CLR1502. Mol Imaging 2015; 13. [PMID: 25743270 DOI: 10.2310/7290.2014.00040] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Positive margins after breast conservation surgery represent a significant problem in the treatment of breast cancer. The near-infrared fluorescence agent CLR1502 (Cellectar Biosciences, Madison, WI) was studied in a preclinical breast cancer model to determine imaging properties and ability to detect small islands of malignancy. Nude mice bearing human breast cancer flank xenografts were given a systemic injection of CLR1502, and imaging was performed using LUNA (Novadaq Technologies Inc., Richmond, BC) and Pearl Impulse (LI-COR Biosciences, Lincoln, NE) devices. Normal tissues were examined for fluorescence signal, and conventional and fluorescence histology was performed using the Odyssey scanner. Peak tumor to background ratio occurred 2 days after injection with CLR1502. The smallest amount of tumor that was imaged and detected using these devices was 1.9 mg, equivalent to 1.9 × 10⁶ cells. The highest fluorescence signal was seen in tumor and normal lymph node tissue, and the lowest fluorescence signal was seen in muscle and plasma. Human breast cancer tumors can be imaged in vivo with multiple optical imaging platforms using CLR1502. This pilot study supports further investigations of this fluorescent agent for improving surgical resection of malignancies, with the goal of eventual clinical translation.
Collapse
|
18
|
Lee SJ, Jeong YI, Park HK, Kang DH, Oh JS, Lee SG, Lee HC. Enzyme-responsive doxorubicin release from dendrimer nanoparticles for anticancer drug delivery. Int J Nanomedicine 2015; 10:5489-503. [PMID: 26357473 PMCID: PMC4559238 DOI: 10.2147/ijn.s87145] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Background Since cancer cells are normally over-expressed cathepsin B, we synthesized dendrimer-methoxy poly(ethylene glycol) (MPEG)-doxorubicin (DOX) conjugates using a cathepsin B-cleavable peptide for anticancer drug targeting. Methods Gly-Phe-Leu-Gly peptide was conjugated with the carboxylic acid end groups of a dendrimer, which was then conjugated with MPEG amine and doxorubicin by aid of carbodiimide chemistry (abbreviated as DendGDP). Dendrimer-MPEG-DOX conjugates without Gly-Phe-Leu-Gly peptide linkage was also synthesized for comparison (DendDP). Nanoparticles were then prepared using a dialysis procedure. Results The synthesized DendGDP was confirmed with 1H nuclear magnetic resonance spectroscopy. The DendDP and DendGDP nanoparticles had a small particle size of less than 200 nm and had a spherical morphology. DendGDP had cathepsin B-sensitive drug release properties while DendDP did not show cathepsin B sensitivity. Further, DendGDP had improved anticancer activity when compared with doxorubicin or DendDP in an in vivo CT26 tumor xenograft model, ie, the volume of the CT26 tumor xenograft was significantly inhibited when compared with xenografts treated with doxorubicin or DendDP nanoparticles. The DendGDP nanoparticles were found to be relatively concentrated in the tumor tissue and revealed stronger fluorescence intensity than at other body sites while doxorubicin and DendDP nanoparticles showed strong fluorescence intensity in the various organs, indicating that DendGDP has cathepsin B sensitivity. Conclusion DendGDP is sensitive to cathepsin B in tumor cells and can be used as a cathepsin B-responsive drug targeting strategy. We suggest that DendGDP is a promising vehicle for cancer cell targeting.
Collapse
Affiliation(s)
- Sang Joon Lee
- Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Young-Il Jeong
- Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Hyung-Kyu Park
- Department of Microbiology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Dae Hwan Kang
- Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea ; Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Gyeongnam, Republic of Korea
| | - Jong-Suk Oh
- Department of Microbiology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Sam-Gyu Lee
- Department of Physical and Rehabilitation Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Hyun Chul Lee
- Department of Microbiology, Chonnam National University Medical School, Gwangju, Republic of Korea
| |
Collapse
|
19
|
Abstract
Mounting evidence suggests that a more extensive surgical resection is associated with an improved life expectancy for both low-grade and high-grade glioma patients. However, radiographically complete resections are not often achieved in many cases because of the lack of sensitivity and specificity of current neurosurgical guidance techniques at the margins of diffuse infiltrative gliomas. Intraoperative fluorescence imaging offers the potential to improve the extent of resection and to investigate the possible benefits of resecting beyond the radiographic margins. Here, we provide a review of wide-field and high-resolution fluorescence-imaging strategies that are being developed for neurosurgical guidance, with a focus on emerging imaging technologies and clinically viable contrast agents. The strengths and weaknesses of these approaches will be discussed, as well as issues that are being addressed to translate these technologies into the standard of care.
Collapse
Affiliation(s)
- Jonathan T C Liu
- *Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York; ‡Barrow Brain Tumor Research Center, Division of Neurosurgical Oncology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona
| | | | | |
Collapse
|
20
|
Al Rawashdeh W, Arns S, Gremse F, Ehling J, Knüchel-Clarke R, Kray S, Spöler F, Kiessling F, Lederle W. Optical tomography of MMP activity allows a sensitive noninvasive characterization of the invasiveness and angiogenesis of SCC xenografts. Neoplasia 2015; 16:235-46, 246.e1. [PMID: 24784000 DOI: 10.1016/j.neo.2014.03.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 02/13/2014] [Accepted: 02/13/2014] [Indexed: 12/22/2022] Open
Abstract
For improved tumor staging and therapy control, imaging biomarkers are of great interest allowing a noninvasive characterization of invasiveness. In squamous epithelial skin and cervix lesions, transition to invasive stages is associated with enhanced matrix metalloproteinase (MMP) activity, increased angiogenesis, and worsened prognosis. Thus, we investigated MMP activity as imaging biomarker of invasiveness and the potential of optical tomography in characterizing the angiogenic and invasive behavior of skin squamous cell carcinoma (SCC) xenografts. MMP activity was measured in vivo in HaCaT-ras A-5RT3 tumors at different angiogenic and invasive stages (onset of angiogenesis, intermediate and highly angiogenic, invasive stage) and after 1 week of sunitinib treatment by fluorescence molecular tomography-microcomputed tomography imaging using an activatable probe. Treatment response was additionally assessed morphologically by optical coherence tomography (OCT). In vivo MMP activity significantly differed between the groups, revealing highest levels in the highly angiogenic, invasive tumors that were confirmed by immunohistochemistry. At the onset of angiogenesis with lowest MMP activity, fibroblasts were detected in the MMP-positive areas, whereas macrophages were absent. Accumulation of both cell types occurred in both invasive groups, again to a significantly higher degree at the most invasive and angiogenic stage. Sunitinib treatment significantly reduced the MMP activity and accumulation of fibroblasts and macrophages and blocked tumor invasion that was additionally visualized by OCT. Human cervical SCCs also showed high MMP activity and a similar stromal composition as the HaCaT xenografts, whereas normal tissue was negative. This study strongly suggests MMP activity as imaging biomarker and demonstrates the high sensitivity of optical tomography in determining tumor invasiveness that can morphologically be supported by OCT.
Collapse
Affiliation(s)
- Wa'el Al Rawashdeh
- Department of Experimental Molecular Imaging, Rheinisch-Westfaelische Technische Hochschule Aachen University, Aachen, Germany
| | - Susanne Arns
- Department of Experimental Molecular Imaging, Rheinisch-Westfaelische Technische Hochschule Aachen University, Aachen, Germany
| | - Felix Gremse
- Department of Experimental Molecular Imaging, Rheinisch-Westfaelische Technische Hochschule Aachen University, Aachen, Germany
| | - Josef Ehling
- Department of Experimental Molecular Imaging, Rheinisch-Westfaelische Technische Hochschule Aachen University, Aachen, Germany; Institute of Pathology, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Ruth Knüchel-Clarke
- Institute of Pathology, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Stefan Kray
- Institute for Semiconductor Electronics, RWTH Aachen University, Aachen, Germany
| | - Felix Spöler
- Institute for Semiconductor Electronics, RWTH Aachen University, Aachen, Germany
| | - Fabian Kiessling
- Department of Experimental Molecular Imaging, Rheinisch-Westfaelische Technische Hochschule Aachen University, Aachen, Germany
| | - Wiltrud Lederle
- Department of Experimental Molecular Imaging, Rheinisch-Westfaelische Technische Hochschule Aachen University, Aachen, Germany.
| |
Collapse
|
21
|
Near-Infrared Confocal Laser Endomicroscopy Detects Colorectal Cancer via an Integrin αvβ3 Optical Probe. Mol Imaging Biol 2015; 17:450-60. [DOI: 10.1007/s11307-015-0825-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
22
|
Lee EK, Kim CW, Kawanami H, Kishimura A, Niidome T, Mori T, Katayama Y. Utilization of a PNA-peptide conjugate to induce a cancer protease-responsive RNAi effect. RSC Adv 2015. [DOI: 10.1039/c5ra17737e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
We designed a new siRNA system which turns on RNAi responding to a cancer cell-specific protease by using a peptide nucleic acid (PNA)-peptide conjugate.
Collapse
Affiliation(s)
- Eun Kyung Lee
- Graduate School of Systems Life Sciences
- Kyushu University
- Fukuoka
- Japan
| | - Chan Woo Kim
- Department of Applied Chemistry
- Faculty of Engineering
- Kyushu University
- Fukuoka
- Japan
| | - Hiroyuki Kawanami
- Graduate School of Systems Life Sciences
- Kyushu University
- Fukuoka
- Japan
| | - Akihiro Kishimura
- Graduate School of Systems Life Sciences
- Kyushu University
- Fukuoka
- Japan
- Department of Applied Chemistry
| | - Takuro Niidome
- Department of Applied Chemistry
- Faculty of Engineering
- Kyushu University
- Fukuoka
- Japan
| | - Takeshi Mori
- Graduate School of Systems Life Sciences
- Kyushu University
- Fukuoka
- Japan
- Department of Applied Chemistry
| | - Yoshiki Katayama
- Graduate School of Systems Life Sciences
- Kyushu University
- Fukuoka
- Japan
- Department of Applied Chemistry
| |
Collapse
|
23
|
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
|
24
|
Folate/NIR 797-conjugated albumin magnetic nanospheres: synthesis, characterisation, and in vitro and in vivo targeting evaluation. PLoS One 2014; 9:e106483. [PMID: 25188308 PMCID: PMC4154716 DOI: 10.1371/journal.pone.0106483] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 07/30/2014] [Indexed: 12/24/2022] Open
Abstract
A practical and effective strategy for synthesis of Folate-NIR 797-conjugated Magnetic Albumin Nanospheres (FA-NIR 797-MAN) was developed. For this strategy, Magnetic Albumin Nanospheres (MAN), composed of superparamagnetic iron oxide nanoparticles (SPIONs) and bovine serum albumin (BSA), were covalently conjugated with folic acid (FA) ligands to enhance the targeting capability of the particles to folate receptor (FR) over-expressing tumours. Subsequently, a near-infrared (NIR) fluorescent dye NIR 797 was conjugated with FA-conjugated MAN for in vivo fluorescence imaging. The FA-NIR 797-MAN exhibited low toxicity to a human nasopharyngeal epidermal carcinoma cell line (KB cells). Additionally, in vitro and in vivo evaluation of the dynamic behaviour and targeting ability of FA-NIR 797-MAN to KB tumours validated the highly selective affinity of FA-NIR 797-MAN for FR-positive tumours. In summary, the FA-NIR 797-MAN prepared here exhibited great potential for tumour imaging, since the near-infrared fluorescence contrast agents target cells via FR-mediated endocytosis. The high fluorescence intensity together with the targeting effect makes FA-NIR 797-MAN a promising candidate for imaging, monitoring, and early diagnosis of cancer at the molecular and cellular levels.
Collapse
|
25
|
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: 1.7] [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
|
26
|
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
|
27
|
de Britto MAP, Soletti RC, Schanaider A, Madi K, de Souza HSP, Machado JC. Endoluminal ultrasound biomicroscopy as a reliable tool for in vivo assessment of colonic inflammation in rats. Int J Colorectal Dis 2013; 28:1613-1620. [PMID: 23925435 DOI: 10.1007/s00384-013-1755-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/23/2013] [Indexed: 02/04/2023]
Abstract
PURPOSE Endoscopic ultrasound (EUS) imaging of the colon is an important diagnostic tool for early neoplasia, although usually restricted to the rectum in inflammatory bowel disease (IBD). This study aimed to evaluate the ability of an endoluminal ultrasound biomicroscopic (eUBM) system to detect and characterize lesions simulating Crohn's disease in the colon of rats in vivo. METHODS Colitis was induced with trinitrobenzene sulfonic acid instillated in the distal colon. Eighteen Wistar rats were submitted to eUBM in three time points: week 1 group (18 animals examined on day 3 after colitis induction), week 2 group (12 animals on days 3 and 10), and week 3 group (7 animals on days 3, 10, and 17). This design yielded distinct inflammation intensities. Three untreated rats were used for acquisition of control images. Scores were used for comparison with histology. RESULTS Scores for eUBM and histology in the different moments of examination achieved a Spearman's rank correlation coefficient of 0.87 (p < 0.001). Findings of wall thickening presented positive predictive value (PPV) and sensitivity of 94 and of 100 %, respectively. Superficial and deep ulcers presented a PPV of 89 and 80 %, respectively, and negative predictive values of 100 and 85 %, respectively. CONCLUSION Accurate detection and analysis of the lesions was achieved. The model is essential for the clinical development of the technique and a reproducible method for the evaluation of experimental colitis. eUBM might be applicable in different segments of the gut, developing into a novel adjunct method for IBD evaluation.
Collapse
Affiliation(s)
- Marcelo Alexandre Pinto de Britto
- Post-Graduation Program in Surgical Sciences, Department of Surgery, School of Medicine, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | | | | | | | | |
Collapse
|
28
|
Mullins SR, Sameni M, Blum G, Bogyo M, Sloane BF, Moin K. Three-dimensional cultures modeling premalignant progression of human breast epithelial cells: role of cysteine cathepsins. Biol Chem 2013; 393:1405-16. [PMID: 23667900 DOI: 10.1515/hsz-2012-0252] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The expression of the cysteine protease cathepsin B is increased in early stages of human breast cancer.To assess the potential role of cathepsin B in premalignant progression of breast epithelial cells, we employed a 3D reconstituted basement membrane overlay culture model of MCF10A human breast epithelial cells and isogenic variants that replicate the in vivo phenotypes of hyper plasia(MCF10AneoT) and atypical hyperplasia (MCF10AT1). MCF10A cells developed into polarized acinar structures with central lumens. In contrast, MCF10AneoT and MCF10AT1 cells form larger structures in which the lumens are filled with cells. CA074Me, a cell-permeable inhibitor selective for the cysteine cathepsins B and L,reduced proliferation and increased apoptosis of MCF10A, MCF10AneoT and MCF10AT1 cells in 3D culture. We detected active cysteine cathepsins in the isogenic MCF10 variants in 3D culture with GB111, a cell-permeable activity based probe, and established differential inhibition of cathepsin B in our 3D cultures. We conclude that cathepsin B promotes proliferation and premalignant progression of breast epithelial cells. These findings are consistent with studies by others showing that deletion of cathepsin B in the transgenic MMTV-PyMT mice, a murine model that is predisposed to development of mammary cancer, reduces malignant progression.
Collapse
|
29
|
Zhang H, Uselman RR, Yee D. Exogenous near-infrared fluorophores and their applications in cancer diagnosis: biological and clinical perspectives. ACTA ACUST UNITED AC 2013; 5:241-51. [PMID: 21566703 DOI: 10.1517/17530059.2011.566858] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
INTRODUCTION Near-infrared fluorescent (NIRF) imaging is a rapidly growing research field which has the potential to be an important imaging modality in cancer diagnosis. Various exogenous NIR fluorophores have been developed for the technique, including small molecule fluorophores and nanoparticles. NIRF imaging has been used in animal models for the detection of cancer overthe last twenty years and has in recent years been used in human clinical trials. AREAS COVERED This article describes the types and characteristics of exogenous fluorophores available for in vivo fluorescent cancer imaging. The article also discusses the progression of NIRF cancer imaging over recent years and its future challenges, from both a biological and clinical perspective. in The review also looks at its application for lymph node mapping, tumor targeting and characterization, and tumor margin definition for surgical guidance. EXPERT OPINION NIRF imaging is not in routine clinical cancer practice; yet, the authors predict that techniques using NIR fluorophores for tumor margin definition and lymph node mapping will enter clinical practice in the near future. The authors also anticipate that NIRF imaging research will lead to the development of flurophores with 'high brightness' that will overcome the limited penetration of this modality and be better suited for non invasive tumor targeting.
Collapse
Affiliation(s)
- Hua Zhang
- Department of Medicine, Masonic Cancer Center, MMC 806, 420 Delaware St SE, Minneapolis, MN, 55455, USA,
| | | | | |
Collapse
|
30
|
Alves KZ, Soletti RC, de Britto MA, de Matos DG, Soldan M, Borges HL, Machado JC. In vivo endoluminal ultrasound biomicroscopic imaging in a mouse model of colorectal cancer. Acad Radiol 2013; 20:90-8. [PMID: 22959583 DOI: 10.1016/j.acra.2012.07.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 07/17/2012] [Accepted: 07/31/2012] [Indexed: 12/14/2022]
Abstract
RATIONALE AND OBJECTIVES The gold-standard tool for colorectal cancer detection is colonoscopy, but it provides only mucosal surface visualization. Ultrasound biomicroscopy allows a clear delineation of the epithelium and adjacent colonic layers. The aim of this study was to design a system to generate endoluminal ultrasound biomicroscopic images of the mouse colon, in vivo, in an animal model of inflammation-associated colon cancer. MATERIALS AND METHODS Thirteen mice (Mus musculus) were used. A 40-MHz miniprobe catheter was inserted into the accessory channel of a pediatric flexible bronchofiberscope. Control mice (n = 3) and mice treated with azoxymethane and dextran sulfate sodium (n = 10) were subjected to simultaneous endoluminal ultrasound biomicroscopy and white-light colonoscopy. The diagnosis obtained with endoluminal ultrasound biomicroscopy and colonoscopy was compared and confirmed by postmortem histopathology. RESULTS Endoluminal ultrasound biomicroscopic images showed all layers of the normal colon and revealed lesions such as lymphoid hyperplasias and colon tumors. Additionally, endoluminal ultrasound biomicroscopy was able to detect two cases of mucosa layer thickening, confirmed by histology. Compared to histologic results, the sensitivities of endoluminal ultrasound biomicroscopy and colonoscopy were 0.95 and 0.83, respectively, and both methods achieved specificities of 1.0. CONCLUSIONS Endoluminal ultrasound biomicroscopy can be used, in addition to colonoscopy, as a diagnostic method for colonic lesions. Moreover, experimental endoluminal ultrasound biomicroscopy in mouse models is feasible and might be used to further develop research on the differentiation between benign and malignant colonic diseases.
Collapse
|
31
|
Bogdanov AA, Mazzanti ML. Fluorescent macromolecular sensors of enzymatic activity for in vivo imaging. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 113:349-87. [PMID: 23244795 DOI: 10.1016/b978-0-12-386932-6.00009-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Macromolecular imaging probes (or sensors) of enzymatic activity have a unique place in the armamentarium of modern optical imaging techniques. Such probes were initially developed by attaching optically "silent" fluorophores via enzyme-sensitive linkers to large copolymers of biocompatible poly(ethylene glycol) and poly(amino acids). In diseased tissue, where the concentration of enzymes is high, the fluorophores are freed from the macromolecular carrier and regain their initial ability to fluoresce, thus allowing in vivo optical localization of the diseased tissue. This chapter describes the design and application of these probes and their alternatives in various areas of experimental medicine and gives an overview of currently available techniques that allow imaging of animals using visible and near-infrared light.
Collapse
Affiliation(s)
- Alexei A Bogdanov
- Laboratory of Molecular Imaging Probes, Department of Radiology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | | |
Collapse
|
32
|
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: 4.0] [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
|
33
|
|
34
|
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: 2.5] [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
|
35
|
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
|
36
|
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: 1.0] [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
|
37
|
Lam KWK, Lo SCL. Discovery of diagnostic serum biomarkers of gastric cancer using proteomics. Proteomics Clin Appl 2012; 2:219-28. [PMID: 21136826 DOI: 10.1002/prca.200780015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Gastric cancer has significant morbidity and mortality worldwide and locally. Good prognosis relies on an early diagnosis. However, this remains a challenge due to the lack of specific and sensitive serum biomarkers for early detection. Hence, there is a constant search for these biomarkers for screening purposes. Proteomic profiling enables a new approach to the discovery of biomarkers in disease. This review presents recent attempts in search of gastric cancer serum biomarker using proteomics. Different methodologies and different types of samples were employed by different groups of researchers. Major difficulties were encountered in the discovery processes, including interference from abundant proteins and continuous changing serum proteomes from different individuals.
Collapse
Affiliation(s)
- Katie Wing-Kei Lam
- The Proteomic Task Force, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China
| | | |
Collapse
|
38
|
Hoetker MS, Kiesslich R, Diken M, Moehler M, Galle PR, Li Y, Goetz M. Molecular in vivo imaging of gastric cancer in a human-murine xenograft model: targeting epidermal growth factor receptor. Gastrointest Endosc 2012; 76:612-20. [PMID: 22771099 DOI: 10.1016/j.gie.2012.05.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Accepted: 05/09/2012] [Indexed: 02/08/2023]
Abstract
BACKGROUND The prognosis of gastric cancer depends on early diagnosis. Targeted therapies against epidermal growth factor receptors (EGFRs) are currently emerging for the treatment of gastric cancer. OBJECTIVE To specifically visualize gastric cancer by using monoclonal antibodies targeting EGFR1 as molecular probes for in vivo molecular confocal laser endomicroscopy (mCLE) in a human-murine xenograft model. DESIGN Prospective in vivo animal study. SETTING Animal laboratory. INTERVENTIONS Human gastric carcinoma xenografts were examined in 26 nude mice by using mCLE after injection of fluorescently labeled antibodies. Nine mice received low-dose anti-EGFR1 antibodies, 7 mice cetuximab, and 7 control mice isotype antibodies. Three mice were screened for autofluorescence without injection. Macroscopic fluorescence was evaluated in 2 additional mice. MAIN OUTCOME MEASUREMENTS Molecular imaging of gastric cancer with confocal laser endomicroscopy. RESULTS Fluorescence intensity in the anti-EGFR1 (P = .0145) and cetuximab group (P = .0047) was significantly higher than in isotype control mice. The same protocol allowed macroscopic fluorescence detection of tumor xenografts. LIMITATIONS Animal model. CONCLUSIONS In vivo microscopic and macroscopic molecular imaging of gastric cancer is feasible in a human-murine xenograft model with both diagnostic and therapeutic antibodies targeting EGFR1. In perspective, mCLE could help diagnose and molecularly characterize gastric cancer during ongoing gastroscopy and may even assist in the prediction of response to therapy.
Collapse
Affiliation(s)
- Michael S Hoetker
- 1st Department of Medicine, University Medical Center of Johannes Gutenberg University Mainz, Mainz, Germany
| | | | | | | | | | | | | |
Collapse
|
39
|
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
|
40
|
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: 0.8] [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
|
41
|
Topical application of activity-based probes for visualization of brain tumor tissue. PLoS One 2012; 7:e33060. [PMID: 22427947 PMCID: PMC3302795 DOI: 10.1371/journal.pone.0033060] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Accepted: 02/06/2012] [Indexed: 11/21/2022] Open
Abstract
Several investigators have shown the utility of systemically delivered optical imaging probes to image tumors in small animal models of cancer. Here we demonstrate an innovative method for imaging tumors and tumor margins during surgery. Specifically, we show that optical imaging probes topically applied to tumors and surrounding normal tissue rapidly differentiate between tissues. In contrast to systemic delivery of optical imaging probes which label tumors uniformly over time, topical probe application results in rapid and robust probe activation that is detectable as early as 5 minutes following application. Importantly, labeling is primarily associated with peri-tumor spaces. This methodology provides a means for rapid visualization of tumor and potentially infiltrating tumor cells and has potential applications for directed surgical excision of tumor tissues. Furthermore, this technology could find use in surgical resections for any tumors having differential regulation of cysteine cathepsin activity.
Collapse
|
42
|
Patel V, Papineni RVL, Gupta S, Stoyanova R, Ahmed MM. A realistic utilization of nanotechnology in molecular imaging and targeted radiotherapy of solid tumors. Radiat Res 2012; 177:483-95. [PMID: 22404738 DOI: 10.1667/rr2597.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Precise dose delivery to malignant tissue in radiotherapy is of paramount importance for treatment efficacy while minimizing morbidity of surrounding normal tissues. Current conventional imaging techniques, such as magnetic resonance imaging (MRI) and computerized tomography (CT), are used to define the three-dimensional shape and volume of the tumor for radiation therapy. In many cases, these radiographic imaging (RI) techniques are ambiguous or provide limited information with regard to tumor margins and histopathology. Molecular imaging (MI) modalities, such as positron emission tomography (PET) and single photon-emission computed-tomography (SPECT) that can characterize tumor tissue, are rapidly becoming routine in radiation therapy. However, their inherent low spatial resolution impedes tumor delineation for the purposes of radiation treatment planning. This review will focus on applications of nanotechnology to synergize imaging modalities in order to accurately highlight, as well as subsequently target, tumor cells. Furthermore, using such nano-agents for imaging, simultaneous coupling of novel therapeutics including radiosensitizers can be delivered specifically to the tumor to maximize tumor cell killing while sparing normal tissue.
Collapse
Affiliation(s)
- Vivek Patel
- Department of Radiation Oncology, University of Miami, Miami, Florida 33136, USA
| | | | | | | | | |
Collapse
|
43
|
Miller SJ, Lee CM, Joshi BP, Gaustad A, Seibel EJ, Wang TD. Targeted detection of murine colonic dysplasia in vivo with flexible multispectral scanning fiber endoscopy. JOURNAL OF BIOMEDICAL OPTICS 2012; 17:021103. [PMID: 22463021 PMCID: PMC3380821 DOI: 10.1117/1.jbo.17.2.021103] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Revised: 09/17/2011] [Accepted: 09/21/2011] [Indexed: 05/20/2023]
Abstract
Gastrointestinal cancers are heterogeneous and can overexpress several protein targets that can be imaged simultaneously on endoscopy using multiple molecular probes. We aim to demonstrate a multispectral scanning fiber endoscope for wide-field fluorescence detection of colonic dysplasia. Excitation at 440, 532, and 635 nm is delivered into a single spiral scanning fiber, and fluorescence is collected by a ring of light-collecting optical fibers placed around the instrument periphery. Specific-binding peptides are selected with phage display technology using the CPC;Apc mouse model of spontaneous colonic dysplasia. Validation of peptide specificity is performed on flow cytometry and in vivo endoscopy. The peptides KCCFPAQ, AKPGYLS, and LTTHYKL are selected and labeled with 7-diethylaminocoumarin-3-carboxylic acid (DEAC), 5-carboxytetramethylrhodamine (TAMRA), and CF633, respectively. Separate droplets of KCCFPAQ-DEAC, AKPGYLS-TAMRA, and LTTHYKL-CF633 are distinguished at concentrations of 100 and 1 μM. Separate application of the fluorescent-labeled peptides demonstrate specific binding to colonic adenomas. The average target/background ratios are 1.71 ± 0.19 and 1.67 ± 0.12 for KCCFPAQ-DEAC and AKPGYLS-TAMRA, respectively. Administration of these two peptides together results in distinct binding patterns in the blue and green channels. Specific binding of two or more peptides can be distinguished in vivo using a novel multispectral endoscope to localize colonic dysplasia on real-time wide-field imaging.
Collapse
Affiliation(s)
- Sharon J. Miller
- University of Michigan, Department of Internal Medicine, Division of Gastroenterology, 109 Zina Pitcher Pl. BSRB 1522, Ann Arbor, Michigan 48109-2200
| | - Cameron M. Lee
- University of Washington, Department of Mechanical Engineering, Human Photonics Laboratory, Box 352600, Seattle, Washington 98195
| | - Bishnu P. Joshi
- University of Michigan, Department of Internal Medicine, Division of Gastroenterology, 109 Zina Pitcher Pl. BSRB 1522, Ann Arbor, Michigan 48109-2200
| | - Adam Gaustad
- University of Michigan, Department of Biomedical Engineering, Division of Gastroenterology, 109 Zina Pitcher Pl. BSRB 1522, Ann Arbor, Michigan 48109-2200
| | - Eric J. Seibel
- University of Washington, Department of Mechanical Engineering, Human Photonics Laboratory, Box 352600, Seattle, Washington 98195
| | - Thomas D. Wang
- University of Michigan, Department of Internal Medicine, Division of Gastroenterology, 109 Zina Pitcher Pl. BSRB 1522, Ann Arbor, Michigan 48109-2200
- University of Michigan, Department of Biomedical Engineering, Division of Gastroenterology, 109 Zina Pitcher Pl. BSRB 1522, Ann Arbor, Michigan 48109-2200
- Address all correspondence to: Thomas D. Wang, University of Michigan, Department of Biomedical Engineering, Division of Gastroenterology, 109 Zina Pitcher Pl. BSRB 1522, Ann Arbor, Michigan 48109-2200. Tel: +734 936 1228; Fax: +734 647 7950; E-mail:
| |
Collapse
|
44
|
Alves KZ, Borges HL, Soletti RC, Viana ALP, Petrella LI, Soldan M, Chagas VL, Schanaider A, Machado JC. Features of in vitro ultrasound biomicroscopic imaging and colonoscopy for detection of colon tumor in mice. ULTRASOUND IN MEDICINE & BIOLOGY 2011; 37:2086-2095. [PMID: 22033129 DOI: 10.1016/j.ultrasmedbio.2011.09.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2011] [Revised: 07/13/2011] [Accepted: 09/06/2011] [Indexed: 05/31/2023]
Abstract
The present work tested the capability of ultrasound biomicroscopy (UBM), at 45 MHz, to provide cross-sectional images with appropriate resolution and contrast to detect tumors and determine their penetration depths on the colon of mice, Mus musculus (Linnaeus 1758), treated with carcinogen for colon tumor induction. B-mode images were obtained, in vitro, from each animal (13 treated and 4 untreated) colon opened longitudinally and immersed in saline solution at room temperature. Prior to UBM inspection, all animals were also examined by colonoscopy. The layers of normal colon identified by UBM are: mucosa (hyperechoic), muscularis mucosae (hypoechoic), submucosa (hyperechoic) and muscularis externa (hypoechoic). UBM images of colon lesions presented structures corresponding to tumors (hyperechoic), lymphoid hyperplasia (hypoechoic) and polypoid tumors (hyperechoic). Additionally, tumoral lesion invasion through the colon was also identified. When compared with histopathologic analysis, all colon lesions detected by UBM were confirmed, while colonoscopic findings had two false negatives.
Collapse
Affiliation(s)
- Kelly Z Alves
- Biomedical Engineering Program, COPPE, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
In vivo diagnosis of murine pancreatic intraepithelial neoplasia and early-stage pancreatic cancer by molecular imaging. Proc Natl Acad Sci U S A 2011; 108:9945-50. [PMID: 21628592 DOI: 10.1073/pnas.1100890108] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a fatal disease with poor patient outcome often resulting from late diagnosis in advanced stages. To date methods to diagnose early-stage PDAC are limited and in vivo detection of pancreatic intraepithelial neoplasia (PanIN), a preinvasive precursor of PDAC, is impossible. Using a cathepsin-activatable near-infrared probe in combination with flexible confocal fluorescence lasermicroscopy (CFL) in a genetically defined mouse model of PDAC we were able to detect and grade murine PanIN lesions in real time in vivo. Our diagnostic approach is highly sensitive and specific and proved superior to clinically established fluorescein-enhanced imaging. Translation of this endoscopic technique into the clinic should tremendously improve detection of pancreatic neoplasia, thus reforming management of patients at risk for PDAC.
Collapse
|
46
|
van Oosten M, Crane LM, Bart J, van Leeuwen FW, van Dam GM. Selecting Potential Targetable Biomarkers for Imaging Purposes in Colorectal Cancer Using TArget Selection Criteria (TASC): A Novel Target Identification Tool. Transl Oncol 2011; 4:71-82. [PMID: 21461170 PMCID: PMC3069650 DOI: 10.1593/tlo.10220] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Revised: 10/23/2010] [Accepted: 11/01/2010] [Indexed: 12/19/2022] Open
Abstract
Peritoneal carcinomatosis (PC) of colorectal origin is associated with a poor prognosis. However, cytoreductive surgery combined with hyperthermic intraperitoneal chemotherapy is available for a selected group of PC patients, which significantly increases overall survival rates up to 30%. As a consequence, there is substantial room for improvement. Tumor targeting is expected to improve the treatment efficacy of colorectal cancer (CRC) further through 1) more sensitive preoperative tumor detection, thus reducing overtreatment; 2) better intraoperative detection and surgical elimination of residual disease using tumor-specific intraoperative imaging; and 3) tumor-specific targeted therapeutics. This review focuses, in particular, on the development of tumor-targeted imaging agents. A large number of biomarkers are known to be upregulated in CRC. However, to date, no validated criteria have been described for the selection of the most promising biomarkers for tumor targeting. Such a scoring system might improve the selection of the correct biomarker for imaging purposes. In this review, we present the TArget Selection Criteria (TASC) scoring system for selection of potential biomarkers for tumor-targeted imaging. By applying TASC to biomarkers for CRC, we identified seven biomarkers (carcinoembryonic antigen, CXC chemokine receptor 4, epidermal growth factor receptor, epithelial cell adhesion molecule, matrix metalloproteinases, mucin 1, and vascular endothelial growth factor A) that seem most suitable for tumor-targeted imaging applications in colorectal cancer. Further cross-validation studies in CRC and other tumor types are necessary to establish its definitive value.
Collapse
Affiliation(s)
- Marleen van Oosten
- Department of Surgery, Division of Surgical Oncology, Surgical Research Laboratory/BioOptical Imaging Center, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | | | | | | |
Collapse
|
47
|
In vivo fluorescence-based endoscopic detection of colon dysplasia in the mouse using a novel peptide probe. PLoS One 2011; 6:e17384. [PMID: 21408169 PMCID: PMC3050896 DOI: 10.1371/journal.pone.0017384] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2010] [Accepted: 02/01/2011] [Indexed: 12/22/2022] Open
Abstract
Colorectal cancer (CRC) is a major cause of cancer-related deaths in much of the
world. Most CRCs arise from pre-malignant (dysplastic) lesions, such as
adenomatous polyps, and current endoscopic screening approaches with white light
do not detect all dysplastic lesions. Thus, new strategies to identify such
lesions, including non-polypoid lesions, are needed. We aim to identify and
validate novel peptides that specifically target dysplastic colonic epithelium
in vivo. We used phage display to identify a novel peptide
that binds to dysplastic colonic mucosa in vivo in a
genetically engineered mouse model of colo-rectal tumorigenesis, based on
somatic Apc (adenomatous polyposis coli) gene
inactivation. Binding was confirmed using confocal microscopy on biopsied
adenomas and excised adenomas incubated with peptide ex vivo.
Studies of mice where a mutant Kras allele was somatically
activated in the colon to generate hyperplastic epithelium were also performed
for comparison. Several rounds of in vivo T7 library biopanning
isolated a peptide, QPIHPNNM.
The fluorescent-labeled peptide bound to dysplastic lesions on endoscopic
analysis. Quantitative assessment revealed the fluorescent-labeled peptide
(target/background: 2.17±0.61) binds ∼2-fold greater to the colonic
adenomas when compared to the control peptide (target/background:
1.14±0.15), p<0.01. The peptide did not bind to the non-dysplastic
(hyperplastic) epithelium of the Kras mice. This work is first
to image fluorescence-labeled peptide binding in vivo that is
specific towards colonic dysplasia on wide-area surveillance. This finding
highlights an innovative strategy for targeted detection to localize
pre-malignant lesions that can be generalized to the epithelium of hollow
organs.
Collapse
|
48
|
Razgulin A, Ma N, Rao J. Strategies for in vivo imaging of enzyme activity: an overview and recent advances. Chem Soc Rev 2011; 40:4186-216. [DOI: 10.1039/c1cs15035a] [Citation(s) in RCA: 226] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
49
|
Sandhu GS, Solorio L, Broome AM, Salem N, Kolthammer J, Shah T, Flask C, Duerk JL. Whole animal imaging. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2010; 2:398-421. [PMID: 20836038 DOI: 10.1002/wsbm.71] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Translational research plays a vital role in understanding the underlying pathophysiology of human diseases, and hence development of new diagnostic and therapeutic options for their management. After creating an animal disease model, pathophysiologic changes and effects of a therapeutic intervention on them are often evaluated on the animals using immunohistologic or imaging techniques. In contrast to the immunohistologic techniques, the imaging techniques are noninvasive and hence can be used to investigate the whole animal, oftentimes in a single exam which provides opportunities to perform longitudinal studies and dynamic imaging of the same subject, and hence minimizes the experimental variability, requirement for the number of animals, and the time to perform a given experiment. Whole animal imaging can be performed by a number of techniques including x-ray computed tomography, magnetic resonance imaging, ultrasound imaging, positron emission tomography, single photon emission computed tomography, fluorescence imaging, and bioluminescence imaging, among others. Individual imaging techniques provide different kinds of information regarding the structure, metabolism, and physiology of the animal. Each technique has its own strengths and weaknesses, and none serves every purpose of image acquisition from all regions of an animal. In this review, a broad overview of basic principles, available contrast mechanisms, applications, challenges, and future prospects of many imaging techniques employed for whole animal imaging is provided. Our main goal is to briefly describe the current state of art to researchers and advanced students with a strong background in the field of animal research.
Collapse
Affiliation(s)
- Gurpreet Singh Sandhu
- Department of Biomedical Engineering, Case Center of Imaging Research, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Luis Solorio
- Department of Biomedical Engineering, Case Center of Imaging Research, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Ann-Marie Broome
- Department of Biomedical Engineering, Case Center of Imaging Research, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Nicolas Salem
- Department of Biomedical Engineering, Case Center of Imaging Research, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Jeff Kolthammer
- Department of Biomedical Engineering, Case Center of Imaging Research, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Tejas Shah
- Department of Biomedical Engineering, Case Center of Imaging Research, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Chris Flask
- Department of Biomedical Engineering, Case Center of Imaging Research, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Jeffrey L Duerk
- Department of Biomedical Engineering, Case Center of Imaging Research, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
50
|
Yoon SM, Myung SJ, Ye BD, Kim IW, Lee NG, Ryu YM, Park K, Kim K, Kwon IC, Park YS, Park CS, Moon DH, Kim DH, Do MY, Byeon JS, Yang SK, Kim JH. Near-infrared fluorescence imaging using a protease-specific probe for the detection of colon tumors. Gut Liver 2010; 4:488-97. [PMID: 21253297 DOI: 10.5009/gnl.2010.4.4.488] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2010] [Accepted: 05/25/2010] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND/AIMS Early tumor detection is crucial for the prevention of colon cancer. Near-infrared fluorescence (NIRF) imaging using a target-activatable probe may permit earlier disease detection. Matrix metalloproteinases (MMPs) participate in tumorigenesis and tumor growth. The aim of this study was to determine whether NIRF imaging using an MMP-activatable probe can detect colon tumors at early stages. METHODS WE UTILIZED TWO MURINE COLON CANCER MODELS: a sporadic colon cancer model induced by azoxymethane (AOM), and a colitis-associated cancer model induced by a combination of AOM and dextran sodium sulfate (DSS). Colonic lesions were analyzed by histologic examination, Western blotting, immunohistochemical staining, and NIRF imaging using an MMP-activatable probe. RESULTS Multiple variable-sized tumors developed in both models and progressed from adenomas to adenocarcinomas over time. At the early stage of the AOM/DSS model, diffuse inflammation was observed within the tumors. MMP expression increased progressively through normal, inflammation, adenoma, and adenocarcionoma stages. NIRF signal intensities were strongly correlated with each tumor stage from adenoma to adenocarcinoma. NIRF imaging also distinguished tumors from inflamed mucosa. CONCLUSIONS NIRF imaging using a protease-activatable probe may be a useful tool for early tumor detection. This approach could translate to improve the endoscopic detection of colon tumors, especially in patients with inflammatory bowel disease.
Collapse
Affiliation(s)
- Soon Man Yoon
- Department of Internal Medicine, Chungbuk National University College of Medicine, Cheongju, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|