1
|
Georgieva TG, Darmoul D, Chen H, Cui H, Rice PFS, Barton JK, Besselsen DG, Ignatenko NA. Kallikrein-Related Peptidase 6 Contributes to Murine Intestinal Tumorigenesis Driven by a Mutant Adenomatous polyposis coli Gene. Cancers (Basel) 2024; 16:3842. [PMID: 39594797 PMCID: PMC11592602 DOI: 10.3390/cancers16223842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/06/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES The objective of this study was to assess the role of a secreted serine protease, kallikrein-related peptidase 6 (KLK6), during colorectal tumorigenesis driven by a mutant Adenomatous polyposis coli (APC) tumor suppressor gene. A first analysis of KLK6 expression in the intestinal tract of Apc-mutant multiple intestinal neoplasia (ApcMin/+) mice revealed up to four-fold induction of Klk6 mRNA levels in adenomas relative to its level in the adjacent mucosa. METHODS AND RESULTS The presence of KLK6 protein in the adenomatous areas was confirmed by immunohistochemistry and optical coherence tomography/laser-induced fluorescence (OCT/LIF) imaging. To assess the contribution of the KLK6 expression on the Apc-mutant intestinal and colon tumorigenesis, we engineered a mouse with floxed alleles of the Klk6 gene (Klk6lox/lox) and crossed it with a mouse expressing the truncated APC protein under control of the intestinal tract-specific human CDX2P9.5-NLS Cre transgene (CPC;Apcfl/fl;Klk6+/+). We found that CPC;Apcfl/fl mice with disrupted Klk6 gene expression (CPC;Apcfl/fl;Klk6fl/fl) had a significantly smaller average size of the small intestinal and colon crypts (p < 0.001 and p = 0.04, respectively) and developed a significantly fewer adenomas (p = 0.01). Moreover, a decrease in high-grade adenomas (p = 0.03) and adenomas with a diameter above 2 mm (p < 0.0001) was noted in CPC;Apcfl/fl;Klk6fl/fl mice. Further molecular analysis showed that Klk6 gene inactivation in the small intestine and colon tissues of CPC;Apcfl/fl;Klk6fl/fl mice resulted in a significant suppression of transforming growth factor β2 (TGF-β2) protein (p ≤ 0.02) and mitogen-activated protein kinase (MAPK) phosphorylation (p ≤ 0.01). CONCLUSIONS These findings demonstrate the oncogenic role of KLK6 in the mutant Apc-mediated intestinal tumorigenesis and suggest the utility of KLK6 for early diagnosis of colorectal tumors.
Collapse
Affiliation(s)
- Teodora G. Georgieva
- Genetically Engineered Mouse Models Core, The University of Arizona Bio5 Institute, Tucson, AZ 85721-0240, USA;
- Department of Cellular and Molecular Medicine, The University of Arizona Cancer Center, Tucson, AZ 85724-5024, USA; (H.C.); (H.C.)
| | - Dalila Darmoul
- Institut de Biologie Paris-Seine, Sorbonne Université, UMR CNRS 8256, INSERM ERL U1164, Biological Adaptation and Ageing, 75005 Paris, France;
| | - Hwudaurw Chen
- Department of Cellular and Molecular Medicine, The University of Arizona Cancer Center, Tucson, AZ 85724-5024, USA; (H.C.); (H.C.)
| | - Haiyan Cui
- Department of Cellular and Molecular Medicine, The University of Arizona Cancer Center, Tucson, AZ 85724-5024, USA; (H.C.); (H.C.)
| | - Photini F. S. Rice
- Department of Biomedical Engineering, The University of Arizona, Tucson, AZ 85721-0240, USA; (P.F.S.R.); (J.K.B.)
| | - Jennifer K. Barton
- Department of Biomedical Engineering, The University of Arizona, Tucson, AZ 85721-0240, USA; (P.F.S.R.); (J.K.B.)
| | - David G. Besselsen
- University Animal Care, The University of Arizona, Tucson, AZ 85721-0101, USA;
| | - Natalia A. Ignatenko
- Department of Cellular and Molecular Medicine, The University of Arizona Cancer Center, Tucson, AZ 85724-5024, USA; (H.C.); (H.C.)
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ 85724-5024, USA
| |
Collapse
|
2
|
Wartak A, Kelada AK, Leon Alarcon PA, Bablouzian AL, Ahsen OO, Gregg AL, Wei Y, Bollavaram K, Sheil CJ, Farewell E, VanTol S, Smith R, Grahmann P, Baillargeon AR, Gardecki JA, Tearney GJ. Dual-modality optical coherence tomography and fluorescence tethered capsule endomicroscopy. BIOMEDICAL OPTICS EXPRESS 2021; 12:4308-4323. [PMID: 34457416 PMCID: PMC8367220 DOI: 10.1364/boe.422453] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 06/02/2021] [Accepted: 06/02/2021] [Indexed: 06/13/2023]
Abstract
OCT tethered capsule endomicroscopy (TCE) is an emerging noninvasive diagnostic imaging technology for gastrointestinal (GI) tract disorders. OCT measures tissue reflectivity that provides morphologic image contrast, and thus is incapable of ascertaining molecular information that can be useful for improving diagnostic accuracy. Here, we introduce an extension to OCT TCE that includes a fluorescence (FL) imaging channel for attaining complementary, co-registered molecular contrast. We present the development of an OCT-FL TCE capsule and a portable, plug-and-play OCT-FL imaging system. The technology is validated in phantom experiments and feasibility is demonstrated in a methylene blue (MB)-stained swine esophageal injury model, ex vivo and in vivo.
Collapse
Affiliation(s)
- Andreas Wartak
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Dermatology, Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Alfred K. Kelada
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Paola A. Leon Alarcon
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Ara L. Bablouzian
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Osman O. Ahsen
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Dermatology, Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Abigail L. Gregg
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Yuxiao Wei
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA
| | - Keval Bollavaram
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Conor J. Sheil
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Dermatology, Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Edward Farewell
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Schuyler VanTol
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Rachel Smith
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Patricia Grahmann
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Aaron R. Baillargeon
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Joseph A. Gardecki
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Dermatology, Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Guillermo J. Tearney
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
3
|
Moody AS, Dayton PA, Zamboni WC. Imaging methods to evaluate tumor microenvironment factors affecting nanoparticle drug delivery and antitumor response. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2021; 4:382-413. [PMID: 34796317 PMCID: PMC8597952 DOI: 10.20517/cdr.2020.94] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/07/2021] [Accepted: 01/28/2021] [Indexed: 11/24/2022]
Abstract
Standard small molecule and nanoparticulate chemotherapies are used for cancer treatment; however, their effectiveness remains highly variable. One reason for this variable response is hypothesized to be due to nonspecific drug distribution and heterogeneity of the tumor microenvironment, which affect tumor delivery of the agents. Nanoparticle drugs have many theoretical advantages, but due to variability in tumor microenvironment (TME) factors, the overall drug delivery to tumors and associated antitumor response are low. The nanotechnology field would greatly benefit from a thorough analysis of the TME factors that create these physiological barriers to tumor delivery and treatment in preclinical models and in patients. Thus, there is a need to develop methods that can be used to reveal the content of the TME, determine how these TME factors affect drug delivery, and modulate TME factors to increase the tumor delivery and efficacy of nanoparticles. In this review, we will discuss TME factors involved in drug delivery, and how biomedical imaging tools can be used to evaluate tumor barriers and predict drug delivery to tumors and antitumor response.
Collapse
Affiliation(s)
- Amber S. Moody
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27599, USA
- Carolina Institute for Nanomedicine, Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, NC 27599, USA
| | - Paul A. Dayton
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, NC 27599, USA
| | - William C. Zamboni
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27599, USA
- Carolina Institute for Nanomedicine, Chapel Hill, NC 27599, USA
| |
Collapse
|
4
|
Candidate Biomarkers for Specific Intraoperative Near-Infrared Imaging of Soft Tissue Sarcomas: A Systematic Review. Cancers (Basel) 2021; 13:cancers13030557. [PMID: 33535618 PMCID: PMC7867119 DOI: 10.3390/cancers13030557] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/16/2021] [Accepted: 01/21/2021] [Indexed: 12/27/2022] Open
Abstract
Simple Summary Near-infrared imaging of tumors during surgery facilitates the oncologic surgeon to distinguish malignant from healthy tissue. The technique is based on fluorescent tracers binding to tumor biomarkers on malignant cells. Currently, there are no clinically available fluorescent tracers that specifically target soft tissue sarcomas. This review searched the literature to find candidate biomarkers for soft tissue sarcomas, based on clinically used therapeutic antibodies. The search revealed 7 biomarkers: TEM1, VEGFR-1, EGFR, VEGFR-2, IGF-1R, PDGFRα, and CD40. These biomarkers are abundantly present on soft tissue sarcoma tumor cells and are already being targeted with humanized monoclonal antibodies. The conjugation of these antibodies with a fluorescent dye will yield in specific tracers for image-guided surgery of soft tissue sarcomas to improve the success rates of tumor resections. Abstract Surgery is the mainstay of treatment for localized soft tissue sarcomas (STS). The curative treatment highly depends on complete tumor resection, as positive margins are associated with local recurrence (LR) and prognosis. However, determining the tumor margin during surgery is challenging. Real-time tumor-specific imaging can facilitate complete resection by visualizing tumor tissue during surgery. Unfortunately, STS specific tracers are presently not clinically available. In this review, STS-associated cell surface-expressed biomarkers, which are currently already clinically targeted with monoclonal antibodies for therapeutic purposes, are evaluated for their use in near-infrared fluorescence (NIRF) imaging of STS. Clinically targeted biomarkers in STS were extracted from clinical trial registers and a PubMed search was performed. Data on biomarker characteristics, sample size, percentage of biomarker-positive STS samples, pattern of biomarker expression, biomarker internalization features, and previous applications of the biomarker in imaging were extracted. The biomarkers were ranked utilizing a previously described scoring system. Eleven cell surface-expressed biomarkers were identified from which 7 were selected as potential biomarkers for NIRF imaging: TEM1, VEGFR-1, EGFR, VEGFR-2, IGF-1R, PDGFRα, and CD40. Promising biomarkers in common and aggressive STS subtypes are TEM1 for myxofibrosarcoma, TEM1, and PDGFRα for undifferentiated soft tissue sarcoma and EGFR for synovial sarcoma.
Collapse
|
5
|
Qiu L, Lin Q, Si Z, Tan H, Liu G, Zhou J, Wang T, Chen Y, Huang Y, Yu T, Jin M, Cheng D, Shi H. A Pretargeted Imaging Strategy for EGFR-Positive Colorectal Carcinoma via Modulation of Tz-Radioligand Pharmacokinetics. Mol Imaging Biol 2021; 23:38-51. [PMID: 32914391 DOI: 10.1007/s11307-020-01539-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/26/2020] [Accepted: 08/27/2020] [Indexed: 11/27/2022]
Abstract
PURPOSE Previously, we successfully developed a pretargeted imaging strategy (atezolizumab-TCO/[99mTc]HYNIC-PEG11-Tz) for evaluating programmed cell death ligand-1 (PD-L1) expression in xenograft mice. However, the surplus unclicked [99mTc]HYNIC-PEG11-Tz is cleared somewhat sluggishly through the intestines, which is not ideal for colorectal cancer (CRC) imaging. To shift the excretion of the Tz-radioligand to the renal system, we developed a novel Tz-radioligand by adding a polypeptide linker between HYNIC and PEG11. PROCEDURES Pretargeted molecular probes [99mTc]HYNIC-polypeptide-PEG11-Tz and cetuximab-TCO were synthesized. [99mTc]HYNIC-polypeptide-PEG11-Tz was evaluated for in vitro stability and in vivo blood pharmacokinetics. In vitro ligation reactivity of [99mTc]HYNIC-polypeptide-PEG11-Tz towards cetuximab-TCO was also tested. Biodistribution assay and imaging of [99mTc]HYNIC-polypeptide-PEG11-Tz were performed to observe its excretion pathway. Pretargeted biodistribution was measured at three different accumulation intervals to determine the optimal pretargeted interval time. Pretargeted (cetuximab-TCO 48 h/[99mTc]HYNIC-PEG11-Tz 6 h) and (cetuximab-TCO 48 h/[99mTc]HYNIC-Polypeptide-PEG11-Tz 6 h) imagings were compared to examine the effect of the excretion pathway on tumor imaging. RESULTS [99mTc]HYNIC-polypeptide-PEG11-Tz showed favorable in vitro stability and rapid blood clearance in mice. SEC-HPLC revealed almost complete reaction between cetuximab-TCO and [99mTc]HYNIC-polypeptide-PEG11-Tz in vitro, with the 8:1 Tz-to-mAb reaction providing a conversion yield of 87.83 ± 3.27 %. Biodistribution and imaging analyses showed that the Tz-radioligand was cleared through the kidneys. After 24, 48, and 72 h of accumulation in HCT116 tumor, the tumor-to-blood ratio of cetuximab-TCO was 0.83 ± 0.13, 1.40 ± 0.31, and 1.15 ± 0.21, respectively. Both pretargeted (cetuximab-TCO 48 h/[99mTc]HYNIC-PEG11-Tz 6 h) and (cetuximab-TCO 48 h/[99mTc]HYNIC-polypeptide-PEG11-Tz 6 h) clearly delineated HCT116 tumor. Pretargeted imaging strategy using cetuximab-TCO/[99mTc]HYNIC-polypeptide-PEG11-Tz could be used for diagnosing CRC, as the surplus unclicked [99mTc]HYNIC-polypeptide-PEG11-Tz was cleared through the urinary system, leading to low abdominal uptake background. CONCLUSION Our novel pretargeted imaging strategy (cetuximab-TCO/[99mTc]HYNIC-polypeptide-PEG11-Tz) was useful for imaging CRC, broadening the application scope of pretargeted imaging strategy. The pretargeted imaging strategy clearly delineated HCT116 tumor, showing that its use could be extended to selection of internalizing antibodies.
Collapse
Affiliation(s)
- Lin Qiu
- Department of Nuclear Medicine, Zhongshan Hospital Fudan University, No. 180, Fenglin Road, Xuhui District, Shanghai, 200032, China
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
| | - Qingyu Lin
- Department of Nuclear Medicine, Zhongshan Hospital Fudan University, No. 180, Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Zhan Si
- Department of Nuclear Medicine, Zhongshan Hospital Fudan University, No. 180, Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Hui Tan
- Department of Nuclear Medicine, Zhongshan Hospital Fudan University, No. 180, Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Guobing Liu
- Department of Nuclear Medicine, Zhongshan Hospital Fudan University, No. 180, Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Jun Zhou
- Department of Nuclear Medicine, Zhongshan Hospital Fudan University, No. 180, Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Tingting Wang
- Department of Nuclear Medicine, Zhongshan Hospital Fudan University, No. 180, Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Yue Chen
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
| | | | - Tao Yu
- WuXi AppTec, Shanghai, China
| | - Mingzhi Jin
- WuXi Biologics (Shanghai) Co., Ltd, Shanghai, China
| | - Dengfeng Cheng
- Department of Nuclear Medicine, Zhongshan Hospital Fudan University, No. 180, Fenglin Road, Xuhui District, Shanghai, 200032, China.
| | - Hongcheng Shi
- Department of Nuclear Medicine, Zhongshan Hospital Fudan University, No. 180, Fenglin Road, Xuhui District, Shanghai, 200032, China.
| |
Collapse
|
6
|
Glover B, Teare J, Patel N. The Status of Advanced Imaging Techniques for Optical Biopsy of Colonic Polyps. Clin Transl Gastroenterol 2020; 11:e00130. [PMID: 32352708 PMCID: PMC7145035 DOI: 10.14309/ctg.0000000000000130] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 01/03/2020] [Indexed: 12/19/2022] Open
Abstract
The progressive miniaturization of photonic components presents the opportunity to obtain unprecedented microscopic images of colonic polyps in real time during endoscopy. This information has the potential to act as "optical biopsy" to aid clinical decision-making, including the possibility of adopting new paradigms such as a "resect and discard" approach for low-risk lesions. The technologies discussed in this review include confocal laser endomicroscopy, optical coherence tomography, multiphoton microscopy, Raman spectroscopy, and hyperspectral imaging. These are in different stages of development and clinical readiness, but all show the potential to produce reliable in vivo discrimination of different tissue types. A structured literature search of the imaging techniques for colorectal polyps has been conducted. The significant developments in endoscopic imaging were identified for each modality, and the status of current development was discussed. Of the advanced imaging techniques discussed, confocal laser endomicroscopy is in clinical use and, under optimal conditions with an experienced operator, can provide accurate histological assessment of tissue. The remaining techniques show potential for incorporation into endoscopic equipment and practice, although further component development is needed, followed by robust prospective validation of accuracy. Optical coherence tomography illustrates tissue "texture" well and gives good assessment of mucosal thickness and layers. Multiphoton microscopy produces high-resolution images at a subcellular resolution. Raman spectroscopy and hyperspectral imaging are less developed endoscopically but provide a tissue "fingerprint" which can distinguish between tissue types. Molecular imaging may become a powerful adjunct to other techniques, with its ability to precisely label specific molecules within tissue and thereby enhance imaging.
Collapse
Affiliation(s)
- Ben Glover
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Julian Teare
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Nisha Patel
- Department of Surgery and Cancer, Imperial College London, London, UK
| |
Collapse
|
7
|
Li Y, Zhu Z, Chen JJ, Jing JC, Sun CH, Kim S, Chung PS, Chen Z. Multimodal endoscopy for colorectal cancer detection by optical coherence tomography and near-infrared fluorescence imaging. BIOMEDICAL OPTICS EXPRESS 2019; 10:2419-2429. [PMID: 31143497 PMCID: PMC6524571 DOI: 10.1364/boe.10.002419] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/20/2019] [Accepted: 04/01/2019] [Indexed: 05/07/2023]
Abstract
While colonoscopy is the gold standard for diagnosis and classification of colorectal cancer (CRC), its sensitivity and specificity are operator-dependent and are especially poor for small and flat lesions. Contemporary imaging modalities, such as optical coherence tomography (OCT) and near-infrared (NIR) fluorescence, have been investigated to visualize microvasculature and morphological changes for detecting early stage CRC in the gastrointestinal (GI) tract. In our study, we developed a multimodal endoscopic system with simultaneous co-registered OCT and NIR fluorescence imaging. By introducing a contrast agent into the vascular network, NIR fluorescence is able to highlight the cancer-suspected area based on significant change of tumor vascular density and morphology caused by angiogenesis. With the addition of co-registered OCT images to reveal subsurface tissue layer architecture, the suspected regions can be further investigated by the altered light scattering resulting from the morphological abnormality. Using this multimodal imaging system, an in vivo animal study was performed using a F344-ApcPircUwm rat, in which the layered architecture and microvasculature of the colorectal wall at different time points were demonstrated. The co-registered OCT and NIR fluorescence images allowed the identification and differentiation of normal colon, hyperplastic polyp, adenomatous polyp, and adenocarcinoma. This multimodal imaging strategy using a single imaging probe has demonstrated the enhanced capability of identification and classification of CRC compared to using any of these technologies alone, thus has the potential to provide a new clinical tool to advance gastroenterology practice.
Collapse
Affiliation(s)
- Yan Li
- Beckman Laser Institute, University of California, Irvine, Irvine, CA 92617, USA
- Department of Biomedical Engineering, University of California, Irvine, 5200 Engineering Hall, Irvine, CA 92697, USA
- Co-first authors with equal contribution
| | - Zhikai Zhu
- Beckman Laser Institute, University of California, Irvine, Irvine, CA 92617, USA
- Department of Biomedical Engineering, University of California, Irvine, 5200 Engineering Hall, Irvine, CA 92697, USA
- Co-first authors with equal contribution
| | - Jason J. Chen
- Beckman Laser Institute, University of California, Irvine, Irvine, CA 92617, USA
- Department of Biomedical Engineering, University of California, Irvine, 5200 Engineering Hall, Irvine, CA 92697, USA
| | - Joseph C. Jing
- Beckman Laser Institute, University of California, Irvine, Irvine, CA 92617, USA
- Department of Biomedical Engineering, University of California, Irvine, 5200 Engineering Hall, Irvine, CA 92697, USA
| | - Chung-Ho Sun
- Beckman Laser Institute, University of California, Irvine, Irvine, CA 92617, USA
| | - Sehwan Kim
- Department of Biomedical Engineering, College of Medicine, Dankook University, Cheonan 31116, South Korea
| | - Phil-Sang Chung
- Department of Otorhinolaryngology-Head & Neck Surgery, College of Medicine, Dankook University, Cheonan 31116, South Korea
| | - Zhongping Chen
- Beckman Laser Institute, University of California, Irvine, Irvine, CA 92617, USA
- Department of Biomedical Engineering, University of California, Irvine, 5200 Engineering Hall, Irvine, CA 92697, USA
| |
Collapse
|
8
|
LI YAN, JING JOSEPH, YU JUNXIAO, ZHANG BUYUN, HUO TIANCHENG, YANG QIANG, CHEN ZHONGPING. Multimodality endoscopic optical coherence tomography and fluorescence imaging technology for visualization of layered architecture and subsurface microvasculature. OPTICS LETTERS 2018; 43:2074-2077. [PMID: 29714749 PMCID: PMC6443372 DOI: 10.1364/ol.43.002074] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Endoscopic imaging technologies, such as endoscopic optical coherence tomography (OCT) and near-infrared fluorescence, have been used to investigate vascular and morphological changes as hallmarks of early cancer in the gastrointestinal tract. Here we developed a high-speed multimodality endoscopic OCT and fluorescence imaging system. Using this system, the architectural morphology and vasculature of the rectum wall were obtained simultaneously from a Sprague Dawley rat in vivo. This multimodality imaging strategy in a single imaging system permits the use of a single imaging probe, thereby improving prognosis by early detection and reducing costs.
Collapse
Affiliation(s)
- YAN LI
- Department of Biomedical Engineering and Beckman laser institute, University of California, Irvine, Irvine, California 92617, USA
- Department of Biomedical Engineering, University of California, 5200 Engineering Hall, Irvine, California 92697, USA
| | - JOSEPH JING
- Department of Biomedical Engineering and Beckman laser institute, University of California, Irvine, Irvine, California 92617, USA
- Department of Biomedical Engineering, University of California, 5200 Engineering Hall, Irvine, California 92697, USA
| | - JUNXIAO YU
- Department of Biomedical Engineering and Beckman laser institute, University of California, Irvine, Irvine, California 92617, USA
- Department of Biomedical Engineering, University of California, 5200 Engineering Hall, Irvine, California 92697, USA
| | - BUYUN ZHANG
- Department of Biomedical Engineering and Beckman laser institute, University of California, Irvine, Irvine, California 92617, USA
| | - TIANCHENG HUO
- Department of Biomedical Engineering and Beckman laser institute, University of California, Irvine, Irvine, California 92617, USA
| | - QIANG YANG
- Department of Biomedical Engineering and Beckman laser institute, University of California, Irvine, Irvine, California 92617, USA
| | - ZHONGPING CHEN
- Department of Biomedical Engineering and Beckman laser institute, University of California, Irvine, Irvine, California 92617, USA
- Department of Biomedical Engineering, University of California, 5200 Engineering Hall, Irvine, California 92697, USA
- Corresponding author:
| |
Collapse
|
9
|
Rice PFS, Ehrichs KG, Jones MS, Chen H, Hsu CH, Abril ER, Nagle RB, Besselsen DG, Barton JK, Ignatenko NA. Does Mutated K-RAS Oncogene Attenuate the Effect of Sulindac in Colon Cancer Chemoprevention? Cancer Prev Res (Phila) 2017; 11:16-26. [PMID: 29118162 DOI: 10.1158/1940-6207.capr-17-0230] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 09/15/2017] [Accepted: 10/23/2017] [Indexed: 11/16/2022]
Abstract
The NSAID sulindac has been successfully used alone or in combination with other agents to suppress colon tumorigenesis in patients with genetic predisposition and also showed its efficacy in prevention of sporadic colon adenomas. At the same time, some experimental and clinical reports suggest that a mutant K-RAS oncogene may negate sulindac antitumor efficacy. To directly assess sulindac activity at suppressing premalignant lesions carrying K-RAS mutation, we utilized a novel mouse model with an inducible colon-specific expression of the mutant K-ras oncogene (K-rasG12D ). Tumor development and treatment effects were monitored by minimally invasive endoscopic Optical coherence tomography. Expression of the mutant K-ras allele accelerated azoxymethane (AOM)-induced colon carcinogenesis in C57BL/6 mice, a strain otherwise resistant to this carcinogen. Sulindac completely prevented AOM-induced tumor formation in K-ras wild-type (K-ras wt) animals. In K-rasG12D -mutant mice, a 38% reduction in tumor number, an 83% reduction in tumor volume (P ≤ 0.01) and an increase in the number of adenoma-free mice (P = 0.04) were observed. The partial response of K-RasG12D animals to sulindac treatment was evident by the decrease in mucosal thickness (P < 0.01) and delay in progression of the precancerous aberrant crypt foci to adenomas. Molecular analyses showed significant induction in cyclooxygenase 2 (COX-2), cleaved caspase-3 (CC3), and Ki-67 expression by AOM, but not sulindac treatment, in all genotypes. Our data underscore the importance of screening for K-RAS mutations in individuals with colon polyps to provide more personalized interventions targeting mutant K-RAS signaling pathways. Cancer Prev Res; 11(1); 16-26. ©2017 AACR.
Collapse
Affiliation(s)
- Photini F S Rice
- Department of Biomedical Engineering, University of Arizona, Tucson, Arizona
| | - Kevin G Ehrichs
- Department of Biomedical Engineering, University of Arizona, Tucson, Arizona
| | - Mykella S Jones
- Department of Biomedical Engineering, University of Arizona, Tucson, Arizona
| | - Hwudarw Chen
- University of Arizona Cancer Center, University of Arizona, Tucson, Arizona
| | - Chiu-Hsieh Hsu
- Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, Arizona
| | - Edward R Abril
- University of Arizona Cancer Center, University of Arizona, Tucson, Arizona
| | - Raymond B Nagle
- Department of Pathology, University of Arizona, Tucson, Arizona
| | | | - Jennifer K Barton
- Department of Biomedical Engineering, University of Arizona, Tucson, Arizona.,University of Arizona Cancer Center, University of Arizona, Tucson, Arizona
| | - Natalia A Ignatenko
- University of Arizona Cancer Center, University of Arizona, Tucson, Arizona. .,Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona
| |
Collapse
|
10
|
Yan X, Song X, Wang Z. Construction of specific magnetic resonance imaging/optical dual-modality molecular probe used for imaging angiogenesis of gastric cancer. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2016; 45:399-403. [PMID: 27074993 DOI: 10.3109/21691401.2016.1167701] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The purpose of the study was to construct specific magnetic resonance imaging (MRI)/optical dual-modality molecular probe. Tumor-bearing animal models were established. MRI/optical dual-modality molecular probe was construed by coupling polyethylene glycol (PEG)-modified nano-Fe3O4 with specific targeted cyclopeptide GX1 and near-infrared fluorescent dyes Cy5.5. MRI/optical imaging effects of the probe were observed and the feasibility of in vivo double-modality imaging was discussed. It was found that, the double-modality probe was of high stability; tumor signal of the experimental group tended to be weak after injection of the probe, but rose to a level which was close to the previous level after 18 h (p > 0.05). We successively completed the construction of an ideal MRI/optical dual-modality molecular probe. MRI/optical dual-modality molecular probe which can selectively gather in gastric cancer is expected to be a novel probe used for diagnosing gastric cancer in the early stage.
Collapse
Affiliation(s)
- Xuejie Yan
- a Department of Spleen and Stomach Diseases , Hospital of Traditional Chinese Medicine , Yantai , Shandong , China
| | - Xiaoyan Song
- b Emergency Department , Hospital of Traditional Chinese Medicine , Yantai , Shandong , China
| | - Zhenbo Wang
- a Department of Spleen and Stomach Diseases , Hospital of Traditional Chinese Medicine , Yantai , Shandong , China
| |
Collapse
|
11
|
Bliley JM, Satish L, McLaughlin MM, Kling RE, Day JR, Grahovac TL, Kokai LE, Zhang W, Marra KG, Rubin JP. Imaging the Stromal Vascular Fraction during Soft-Tissue Reconstruction. Plast Reconstr Surg 2015; 136:1205-1215. [PMID: 26595017 DOI: 10.1097/prs.0000000000001815] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND Although fat grafting is an increasingly popular practice, suboptimal volume retention remains an obstacle. Graft enrichment with the stromal vascular fraction has gained attention as a method of increasing graft retention. However, few studies have assessed the fate and impact of transplanted stromal vascular fraction on fat grafts. In vivo imaging techniques can be used to help determine the influence stromal vascular fraction has on transplanted fat. METHODS Stromal vascular fraction was labeled with 1,1'-dioctadecyl-3,3,3',3'-tetramethylindotricarbocyanine iodide (DiR), a near-infrared dye, and tracked in vivo. Proliferation and differentiation of labeled cells were assessed to confirm that labeling did not adversely affect cellular function. Different doses of labeled stromal vascular fraction were tracked within fat grafts over time using the in vivo imaging system. RESULTS No significant differences in differentiation and proliferation were observed in labeled versus unlabeled cells (p > 0.05). A pilot study confirmed that stromal vascular fraction fluorescence was localized to fat grafts and different cell doses could be distinguished. A larger-scale in vivo study revealed that stromal vascular fraction fluorescence was statistically significant (p < 0.05) between different cell dose groups and this significance was maintained in higher doses (3 × 10(6) and 2 × 10(6) cells/ml of fat graft) for up to 41 days in vivo. CONCLUSIONS DiR labeling allowed the authors to differentiate between cell doses and confirm localization. This article supports the use of DiR labeling in conjunction with in vivo imaging as a tool for imaging stromal vascular fraction within fat grafts.
Collapse
Affiliation(s)
- Jacqueline M Bliley
- Pittsburgh, Pa. From the Departments of Plastic Surgery and Bioengineering, University of Pittsburgh; and the McGowan Institute for Regenerative Medicine
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Boonstra MC, Prakash J, Van De Velde CJH, Mesker WE, Kuppen PJK, Vahrmeijer AL, Sier CFM. Stromal Targets for Fluorescent-Guided Oncologic Surgery. Front Oncol 2015; 5:254. [PMID: 26636036 PMCID: PMC4653299 DOI: 10.3389/fonc.2015.00254] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 11/05/2015] [Indexed: 12/17/2022] Open
Abstract
Pre-operative imaging techniques are essential for tumor detection and diagnosis, but offer limited help during surgery. Recently, the applicability of imaging during oncologic surgery has been recognized, using near-infrared fluorescent dyes conjugated to targeting antibodies, peptides, or other vehicles. Image-guided oncologic surgery (IGOS) assists the surgeFon to distinguish tumor from normal tissue during operation, and can aid in recognizing vital structures. IGOS relies on an optimized combination of a dedicated fluorescent camera system and specific probes for targeting. IGOS probes for clinical use are not widely available yet, but numerous pre-clinical studies have been published and clinical trials are being established or prepared. Most of the investigated probes are based on antibodies or peptides against proteins on the membranes of malignant cells, whereas others are directed against stromal cells. Targeting stroma cells for IGOS has several advantages. Besides the high stromal content in more aggressive tumor types, the stroma is often primarily located at the periphery/invasive front of the tumor, which makes stromal targets particularly suited for imaging purposes. Moreover, because stroma up-regulation is a physiological reaction, most proteins to be targeted on these cells are “universal” and not derived from a specific genetic variation, as is the case with many upregulated proteins on malignant cancer cells.
Collapse
Affiliation(s)
- Martin C Boonstra
- Department of Surgery, Leiden University Medical Center , Leiden , Netherlands
| | - Jai Prakash
- Department of Biomaterial Science and Technology, Targeted Therapeutics, University of Twente , Enschede , Netherlands
| | | | - Wilma E Mesker
- Department of Surgery, Leiden University Medical Center , Leiden , Netherlands
| | - Peter J K Kuppen
- Department of Surgery, Leiden University Medical Center , Leiden , Netherlands
| | | | - Cornelis F M Sier
- Department of Surgery, Leiden University Medical Center , Leiden , Netherlands ; Antibodies for Research Applications BV , Gouda , Netherlands
| |
Collapse
|
13
|
LeGendre-McGhee S, Rice PS, Wall RA, Sprute KJ, Bommireddy R, Luttman AM, Nagle RB, Abril ER, Farrell K, Hsu CH, Roe DJ, Gerner EW, Ignatenko NA, Barton JK. Time-serial Assessment of Drug Combination Interventions in a Mouse Model of Colorectal Carcinogenesis Using Optical Coherence Tomography. CANCER GROWTH AND METASTASIS 2015; 8:63-80. [PMID: 26396545 PMCID: PMC4562605 DOI: 10.4137/cgm.s21216] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 07/05/2015] [Accepted: 07/07/2015] [Indexed: 02/07/2023]
Abstract
Optical coherence tomography (OCT) is a high-resolution, nondestructive imaging modality that enables time-serial assessment of adenoma development in the mouse model of colorectal cancer. In this study, OCT was utilized to evaluate the effectiveness of interventions with the experimental antitumor agent α-difluoromethylornithine (DFMO) and a nonsteroidal anti-inflammatory drug sulindac during early [chemoprevention (CP)] and late stages [chemotherapy (CT)] of colon tumorigenesis. Biological endpoints for drug interventions included OCT-generated tumor number and tumor burden. Immunochistochemistry was used to evaluate biochemical endpoints [Ki-67, cleaved caspase-3, cyclooxygenase (COX)-2, β-catenin]. K-Ras codon 12 mutations were studied with polymerase chain reaction-based technique. We demonstrated that OCT imaging significantly correlated with histological analysis of both tumor number and tumor burden for all experimental groups (P < 0.0001), but allows more accurate and full characterization of tumor number and burden growth rate because of its time-serial, nondestructive nature. DFMO alone or in combination with sulindac suppressed both the tumor number and tumor burden growth rate in the CP setting because of DFMO-mediated decrease in cell proliferation (Ki-67, P < 0.001) and K-RAS mutations frequency (P = 0.04). In the CT setting, sulindac alone and DFMO/sulindac combination were effective in reducing tumor number, but not tumor burden growth rate. A decrease in COX-2 staining in DFMO/sulindac CT groups (COX-2, P < 0.01) confirmed the treatment effect. Use of nondestructive OCT enabled repeated, quantitative evaluation of tumor number and burden, allowing changes in these parameters to be measured during CP and as a result of CT. In conclusion, OCT is a robust minimally invasive method for monitoring colorectal cancer disease and effectiveness of therapies in mouse models.
Collapse
Affiliation(s)
| | - Photini S Rice
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ, USA
| | - R Andrew Wall
- College of Optical Sciences, University of Arizona, Tucson, AZ, USA
| | - Kyle J Sprute
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ, USA
| | | | - Amber M Luttman
- College of Optical Sciences, University of Arizona, Tucson, AZ, USA
| | - Raymond B Nagle
- Department of Pathology, University of Arizona, Tucson, AZ, USA
| | - Edward R Abril
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, USA
| | - Katrina Farrell
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ, USA
| | - Chiu-Hsieh Hsu
- Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ, USA
| | - Denise J Roe
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, USA. ; Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ, USA
| | - Eugene W Gerner
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Natalia A Ignatenko
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, USA. ; Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Jennifer K Barton
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ, USA. ; College of Optical Sciences, University of Arizona, Tucson, AZ, USA. ; University of Arizona Cancer Center, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
14
|
Yoon Y, Jang WH, Xiao P, Kim B, Wang T, Li Q, Lee JY, Chung E, Kim KH. In vivo wide-field reflectance/fluorescence imaging and polarization-sensitive optical coherence tomography of human oral cavity with a forward-viewing probe. BIOMEDICAL OPTICS EXPRESS 2015; 6:524-35. [PMID: 25780742 PMCID: PMC4354576 DOI: 10.1364/boe.6.000524] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 12/31/2014] [Accepted: 01/07/2015] [Indexed: 05/18/2023]
Abstract
We report multimodal imaging of human oral cavity in vivo based on simultaneous wide-field reflectance/fluorescence imaging and polarization-sensitive optical coherence tomography (PS-OCT) with a forward-viewing imaging probe. Wide-field reflectance/fluorescence imaging and PS-OCT were to provide both morphological and fluorescence information on the surface, and structural and birefringent information below the surface respectively. The forward-viewing probe was designed to access the oral cavity through the mouth with dimensions of approximately 10 mm in diameter and 180 mm in length. The probe had field of view (FOV) of approximately 5.5 mm in diameter, and adjustable depth of field (DOF) from 2 mm to 10 mm by controlling numerical aperture (NA) in the detection path. This adjustable DOF was to accommodate both requirements for image-based guiding with high DOF and high-resolution, high-sensitivity imaging with low DOF. This multimodal imaging system was characterized by using a tissue phantom and a mouse model in vivo, and was applied to human oral cavity. Information of surface morphology and vasculature, and under-surface layered structure and birefringence of the oral cavity tissues was obtained. These results showed feasibility of this multimodal imaging system as a tool for studying oral cavity lesions in clinical applications.
Collapse
Affiliation(s)
- Yeoreum Yoon
- Department of Mechanical Engineering, Pohang University of Science and Technology, San 31, Hyoja-dong, Nam-gu, Pohang, Gyeongbuk 790-784,
South Korea
| | - Won Hyuk Jang
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, San 31, Hyoja-dong, Nam-gu, Pohang, Gyeongbuk 790-784,
South Korea
| | - Peng Xiao
- Department of Mechanical Engineering, Pohang University of Science and Technology, San 31, Hyoja-dong, Nam-gu, Pohang, Gyeongbuk 790-784,
South Korea
| | - Bumju Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology, San 31, Hyoja-dong, Nam-gu, Pohang, Gyeongbuk 790-784,
South Korea
| | - Taejun Wang
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, San 31, Hyoja-dong, Nam-gu, Pohang, Gyeongbuk 790-784,
South Korea
| | - Qingyun Li
- Department of Mechanical Engineering, Pohang University of Science and Technology, San 31, Hyoja-dong, Nam-gu, Pohang, Gyeongbuk 790-784,
South Korea
| | - Ji Youl Lee
- Department of Urology, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Seocho-gu, Seoul 137–040,
South Korea
| | - Euiheon Chung
- Department of Medical System Engineering and School of Mechatronics, Gwangju Institute of Science and Technology, 123 Cheomdan-gwagiro, Buk-gu, Gwangju 500-712,
South Korea
| | - Ki Hean Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology, San 31, Hyoja-dong, Nam-gu, Pohang, Gyeongbuk 790-784,
South Korea
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, San 31, Hyoja-dong, Nam-gu, Pohang, Gyeongbuk 790-784,
South Korea
| |
Collapse
|
15
|
Carbary-Ganz JL, Welge WA, Barton JK, Utzinger U. In vivo molecular imaging of colorectal cancer using quantum dots targeted to vascular endothelial growth factor receptor 2 and optical coherence tomography/laser-induced fluorescence dual-modality imaging. JOURNAL OF BIOMEDICAL OPTICS 2015; 20:096015. [PMID: 26397238 PMCID: PMC4963467 DOI: 10.1117/1.jbo.20.9.096015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 08/17/2015] [Indexed: 05/27/2023]
Abstract
Optical coherence tomography/laser induced fluorescence (OCT/LIF) dual-modality imaging allows for minimally invasive, nondestructive endoscopic visualization of colorectal cancer in mice. This technology enables simultaneous longitudinal tracking of morphological (OCT) and biochemical (fluorescence) changes as colorectal cancer develops, compared to current methods of colorectal cancer screening in humans that rely on morphological changes alone. We have shown that QDot655 targeted to vascular endothelial growth factor receptor 2 (QD655-VEGFR2) can be applied to the colon of carcinogen-treated mice and provides significantly increased contrast between the diseased and undiseased tissue with high sensitivity and specificity ex vivo. QD655-VEGFR2 was used in a longitudinal in vivo study to investigate the ability to correlate fluorescence signal to tumor development. QD655-VEGFR2 was applied to the colon of azoxymethane (AOM-) or saline-treated control mice in vivo via lavage. OCT/LIF images of the distal colon were taken at five consecutive time points every three weeks after the final AOM injection. Difficulties in fully flushing unbound contrast agent from the colon led to variable background signal; however, a spatial correlation was found between tumors identified in OCT images, and high fluorescence intensity of the QD655 signal, demonstrating the ability to detect VEGFR2 expressing tumors in vivo.
Collapse
Affiliation(s)
- Jordan L. Carbary-Ganz
- University of Arizona, Biomedical Engineering, Thomas W. Keating Bioresearch Building, 1657 E Helen Street, Tucson, Arizona 84721, United States
| | - Weston A. Welge
- University of Arizona, Biomedical Engineering, Thomas W. Keating Bioresearch Building, 1657 E Helen Street, Tucson, Arizona 84721, United States
| | - Jennifer K. Barton
- University of Arizona, Biomedical Engineering, Thomas W. Keating Bioresearch Building, 1657 E Helen Street, Tucson, Arizona 84721, United States
| | - Urs Utzinger
- University of Arizona, Biomedical Engineering, Thomas W. Keating Bioresearch Building, 1657 E Helen Street, Tucson, Arizona 84721, United States
| |
Collapse
|
16
|
Leung SJ, Rice PS, Barton JK. In vivo molecular mapping of the tumor microenvironment in an azoxymethane-treated mouse model of colon carcinogenesis. Lasers Surg Med 2014; 47:40-9. [PMID: 25487746 DOI: 10.1002/lsm.22309] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2014] [Indexed: 01/05/2023]
Abstract
BACKGROUND AND OBJECTIVE Development of miniaturized imaging systems with molecular probes enables examination of molecular changes leading to initiation and progression of colorectal cancer in an azoxymethane (AOM)-induced mouse model of the disease. Through improved and novel studies of animal disease models, more effective diagnostic and treatment strategies may be developed for clinical translation. We introduce use of a miniaturized multimodal endoscope with lavage-delivered fluorescent probes to examine dynamic microenvironment changes in an AOM-treated mouse model. STUDY DESIGN/MATERIALS AND METHODS The endoscope is equipped with optical coherence tomography (OCT) and laser induced fluorescence (LIF) imaging modalities. It is used with Cy5.5-conjugated antibodies to create time-resolved molecular maps of colon carcinogenesis. We monitored in vivo changes in molecular expression over a five month period for four biomarkers: epithelial growth factor receptor (EGFR), transferrin receptor (TfR), transforming growth factor beta 1 (TGFβ1), and chemokine (C-X-C motif) receptor 2 (CXCR2). In vivo OCT and LIF images were compared over multiple time points to correlate increases in biomarker expression with adenoma development. RESULTS This system is uniquely capable of tracking in vivo changes in molecular expression over time. Increased expression of the biomarker panel corresponded to sites of disease and offered predictive utility in highlighting sites of disease prior to detectable structural changes. Biomarker expression also tended to increase with higher tumor burden and growth rate in the colon. CONCLUSION We can use miniaturized dual modality endoscopes with fluorescent probes to study the tumor microenvironment in developmental animal models of cancer and supplement findings from biopsy and tissue harvesting.
Collapse
Affiliation(s)
- Sarah J Leung
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ
| | | | | |
Collapse
|
17
|
Pahlevaninezhad H, Lee AMD, Shaipanich T, Raizada R, Cahill L, Hohert G, Yang VXD, Lam S, MacAulay C, Lane P. A high-efficiency fiber-based imaging system for co-registered autofluorescence and optical coherence tomography. BIOMEDICAL OPTICS EXPRESS 2014; 5:2978-87. [PMID: 25401011 PMCID: PMC4230860 DOI: 10.1364/boe.5.002978] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 07/31/2014] [Accepted: 08/01/2014] [Indexed: 05/06/2023]
Abstract
We present a power-efficient fiber-based imaging system capable of co-registered autofluorescence imaging and optical coherence tomography (AF/OCT). The system employs a custom fiber optic rotary joint (FORJ) with an embedded dichroic mirror to efficiently combine the OCT and AF pathways. This three-port wavelength multiplexing FORJ setup has a throughput of more than 83% for collected AF emission, significantly more efficient compared to previously reported fiber-based methods. A custom 900 µm diameter catheter ‒ consisting of a rotating lens assembly, double-clad fiber (DCF), and torque cable in a stationary plastic tube ‒ was fabricated to allow AF/OCT imaging of small airways in vivo. We demonstrate the performance of this system ex vivo in resected porcine airway specimens and in vivo in human on fingers, in the oral cavity, and in peripheral airways.
Collapse
Affiliation(s)
- Hamid Pahlevaninezhad
- Integrative Oncology Department―Imaging Unit, BC Cancer Research Center, 675 West 10th Avenue, Vancouver, Canada
| | - Anthony M. D. Lee
- Integrative Oncology Department―Imaging Unit, BC Cancer Research Center, 675 West 10th Avenue, Vancouver, Canada
| | - Tawimas Shaipanich
- Integrative Oncology Department―Imaging Unit, BC Cancer Research Center, 675 West 10th Avenue, Vancouver, Canada
| | - Rashika Raizada
- Integrative Oncology Department―Imaging Unit, BC Cancer Research Center, 675 West 10th Avenue, Vancouver, Canada
| | - Lucas Cahill
- Integrative Oncology Department―Imaging Unit, BC Cancer Research Center, 675 West 10th Avenue, Vancouver, Canada
| | - Geoffrey Hohert
- Integrative Oncology Department―Imaging Unit, BC Cancer Research Center, 675 West 10th Avenue, Vancouver, Canada
| | - Victor X. D. Yang
- Biophotonics and Bioengineering Laboratory, Electrical and Computer Engineering, Ryerson University, Toronto, ON, Canada
| | - Stephen Lam
- Integrative Oncology Department―Imaging Unit, BC Cancer Research Center, 675 West 10th Avenue, Vancouver, Canada
| | - Calum MacAulay
- Integrative Oncology Department―Imaging Unit, BC Cancer Research Center, 675 West 10th Avenue, Vancouver, Canada
| | - Pierre Lane
- Integrative Oncology Department―Imaging Unit, BC Cancer Research Center, 675 West 10th Avenue, Vancouver, Canada
| |
Collapse
|
18
|
Carbary-Ganz JL, Barton JK, Utzinger U. Quantum dots targeted to vascular endothelial growth factor receptor 2 as a contrast agent for the detection of colorectal cancer. JOURNAL OF BIOMEDICAL OPTICS 2014; 19:086003. [PMID: 25104409 PMCID: PMC4124065 DOI: 10.1117/1.jbo.19.8.086003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 07/17/2014] [Accepted: 07/21/2014] [Indexed: 06/01/2023]
Abstract
We successfully labeled colorectal cancer in vivo using quantum dots targeted to vascular endothelial growth factor receptor 2 (VEGFR2). Quantum dots with emission centered at 655 nm were bioconjugated to anti-VEGFR2 antibodies through streptavidin/biotin linking. The resulting QD655-VEGFR2 contrast agent was applied in vivo to the colon of azoxymethane (AOM) treated mice via lavage and allowed to incubate. The colons were then excised, cut longitudinally, opened to expose the lumen, and imaged en face using a fluorescence stereoscope. The QD655-VEGFR2 contrast agent produced a significant increase in contrast between diseased and undiseased tissues, allowing for fluorescence-based visualization of the diseased areas of the colon. Specificity was assessed by observing insignificant contrast increase when labeling colons of AOM-treated mice with quantum dots bioconjugated to isotype control antibodies, and by labeling the colons of saline-treated control mice. This contrast agent has a great potential for in vivo imaging of the colon through endoscopy.
Collapse
Affiliation(s)
- Jordan L. Carbary-Ganz
- University of Arizona, Biomedical Engineering GIDP, Tucson, Arizona 85721, United States
| | - Jennifer K. Barton
- University of Arizona, Biomedical Engineering GIDP, Tucson, Arizona 85721, United States
| | - Urs Utzinger
- University of Arizona, Biomedical Engineering GIDP, Tucson, Arizona 85721, United States
| |
Collapse
|
19
|
Wang T, Li Q, Xiao P, Ahn J, Kim YE, Park Y, Kim M, Song M, Chung E, Chung WK, Ahn GO, Kim S, Kim P, Myung SJ, Kim KH. Gradient index lens based combined two-photon microscopy and optical coherence tomography. OPTICS EXPRESS 2014; 22:12962-70. [PMID: 24921493 DOI: 10.1364/oe.22.012962] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
We report a miniaturized probe-based combined two-photon microscopy (TPM) and optical coherence tomography (OCT) system. This system is to study the colorectal cancer in mouse models by visualizing both cellular and structural information of the colon in 3D with TPM and OCT respectively. The probe consisted of gradient index (GRIN) lenses and a 90° reflecting prism at its distal end for side-viewing, and it was added onto an objective lens-based TPM and OCT system. The probe was 2.2 mm in diameter and 60 mm in length. TPM imaging was performed by raster scanning of the excitation focus at the imaging speed of 15.4 frames/s. OCT imaging was performed by combining the linear sample translation and probe rotation along its axis. This miniaturized probe based dual-modal system was characterized with tissue phantoms containing fluorescent microspheres, and applied to image mouse colonic tissues ex vivo as a demonstration. As OCT and TPM provided structural and cellular information of the tissues respectively, this probe based multi-modal imaging system can be helpful for in vivo studies of preclinical animal models such as mouse colonic tumorigenesis.
Collapse
|
20
|
Pahlevaninezhad H, Lee AMD, Lam S, MacAulay C, Lane PM. Coregistered autofluorescence-optical coherence tomography imaging of human lung sections. JOURNAL OF BIOMEDICAL OPTICS 2014; 19:36022. [PMID: 24687614 DOI: 10.1117/1.jbo.19.3.036022] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 03/03/2014] [Indexed: 05/20/2023]
Abstract
Autofluorescence (AF) imaging can provide valuable information about the structural and metabolic state of tissue that can be useful for elucidating physiological and pathological processes. Optical coherence tomography (OCT) provides high resolution detailed information about tissue morphology. We present coregistered AF-OCT imaging of human lung sections. Adjacent hematoxylin and eosin stained histological sections are used to identify tissue structures observed in the OCT images. Segmentation of these structures in the OCT images allowed determination of relative AF intensities of human lung components. Since the AF imaging was performed on tissue sections perpendicular to the airway axis, the results show the AF signal originating from the airway wall components free from the effects of scattering and absorption by overlying layers as is the case during endoscopic imaging. Cartilage and dense connective tissue (DCT) are found to be the dominant fluorescing components with the average cartilage AF intensity about four times greater than that of DCT. The epithelium, lamina propria, and loose connective tissue near basement membrane generate an order of magnitude smaller AF signal than the cartilage fluorescence.
Collapse
|
21
|
Alex A, Noti M, Wojno EDT, Artis D, Zhou C. Characterization of eosinophilic esophagitis murine models using optical coherence tomography. BIOMEDICAL OPTICS EXPRESS 2014; 5:609-620. [PMID: 24575353 PMCID: PMC3920889 DOI: 10.1364/boe.5.000609] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 01/17/2014] [Accepted: 01/18/2014] [Indexed: 05/29/2023]
Abstract
Pre-clinical studies using murine models are critical for understanding the pathophysiological mechanisms underlying immune-mediated disorders such as Eosinophilic esophagitis (EoE). In this study, an optical coherence tomography (OCT) system capable of providing three-dimensional images with axial and transverse resolutions of 5 µm and 10 µm, respectively, was utilized to obtain esophageal images from a murine model of EoE-like disease ex vivo. Structural changes in the esophagus of wild-type (Tslpr(+/+) ) and mutant (Tslpr(-/-) ) mice with EoE-like disease were quantitatively evaluated and food impaction sites in the esophagus of diseased mice were monitored using OCT. Here, the capability of OCT as a label-free imaging tool devoid of tissue-processing artifacts to effectively characterize murine EoE-like disease models has been demonstrated.
Collapse
Affiliation(s)
- Aneesh Alex
- Department of Electrical and Computer Engineering, Lehigh University, Bethlehem, PA-18015, USA
- Center for Photonics and Nanoelectronics, Lehigh University, Bethlehem, PA-18015, USA
| | - Mario Noti
- Department of Microbiology University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Immunology, Perelman School of Medicine University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Elia D. Tait Wojno
- Department of Microbiology University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Immunology, Perelman School of Medicine University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David Artis
- Department of Microbiology University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Immunology, Perelman School of Medicine University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Chao Zhou
- Department of Electrical and Computer Engineering, Lehigh University, Bethlehem, PA-18015, USA
- Center for Photonics and Nanoelectronics, Lehigh University, Bethlehem, PA-18015, USA
- Bioengineering Program, Lehigh University, Bethlehem, PA-18015, USA
| |
Collapse
|
22
|
Narunsky L, Oren R, Bochner F, Neeman M. Imaging aspects of the tumor stroma with therapeutic implications. Pharmacol Ther 2013; 141:192-208. [PMID: 24134903 DOI: 10.1016/j.pharmthera.2013.10.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Accepted: 09/13/2013] [Indexed: 12/25/2022]
Abstract
Cancer cells rely on extensive support from the stroma in order to survive, proliferate and invade. The tumor stroma is thus an important potential target for anti-cancer therapy. Typical changes in the stroma include a shift from the quiescence promoting-antiangiogenic extracellular matrix to a provisional matrix that promotes invasion and angiogenesis. These changes in the extracellular matrix are induced by changes in the secretion of extracellular matrix proteins and glucose amino glycans, extravasation of plasma proteins from hyperpermeable vessels and release of matrix modifying enzymes resulting in cleavage and cross-linking of matrix macromolecules. These in turn alter the rigidity of the matrix and the exposure and release of cytokines. Changes in matrix rigidity and vessel permeability affect drug delivery and mediate resistance to cytotoxic therapy. These stroma changes are brought about not only by the cancer cells, but also through the action of many cell types that are recruited by tumors including immune cells, fibroblasts and endothelial cells. Within the tumor, these normal host cells are activated resulting in loss of inhibitory and induction of cancer promoting activities. Key to the development of stroma-targeted therapies, selective biomarkers were developed for specific imaging of key aspects of the tumor stroma.
Collapse
Affiliation(s)
- Lian Narunsky
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Roni Oren
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Filip Bochner
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Michal Neeman
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
23
|
Enhanced imaging in the GI tract: spectroscopy and optical coherence tomography. Gastrointest Endosc 2013; 78:568-73. [PMID: 24054739 DOI: 10.1016/j.gie.2013.07.024] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 07/11/2013] [Indexed: 02/07/2023]
|
24
|
Carns J, Keahey P, Quang T, Anandasabapathy S, Richards-Kortum R. Optical molecular imaging in the gastrointestinal tract. Gastrointest Endosc Clin N Am 2013; 23:707-23. [PMID: 23735112 PMCID: PMC3746803 DOI: 10.1016/j.giec.2013.03.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Recent developments in optical molecular imaging allow for real-time identification of morphologic and biochemical changes in tissue associated with gastrointestinal neoplasia. This review summarizes widefield and high-resolution imaging modalities in preclinical and clinical evaluation for the detection of colorectal cancer and esophageal cancer. Widefield techniques discussed include high-definition white light endoscopy, narrow band imaging, autofluoresence imaging, and chromoendoscopy; high-resolution techniques discussed include probe-based confocal laser endomicroscopy, high-resolution microendoscopy, and optical coherence tomography. New approaches to enhance image contrast using vital dyes and molecular-specific targeted contrast agents are evaluated.
Collapse
Affiliation(s)
- Jennifer Carns
- Department of Bioengineering, Rice University, Houston, TX 77005, United States,corresponding author for proofs
| | - Pelham Keahey
- Department of Bioengineering, Rice University, Houston, TX 77005, United States
| | - Timothy Quang
- Department of Bioengineering, Rice University, Houston, TX 77005, United States
| | | | - Rebecca Richards-Kortum
- Department of Bioengineering, Rice University, Houston, TX 77005, United States,corresponding author after publication
| |
Collapse
|
25
|
Yang JM, Chen R, Favazza C, Yao J, Li C, Hu Z, Zhou Q, Shung KK, Wang LV. A 2.5-mm diameter probe for photoacoustic and ultrasonic endoscopy. OPTICS EXPRESS 2012; 20:23944-23953. [PMID: 23188360 PMCID: PMC3601641 DOI: 10.1364/oe.20.023944] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Revised: 09/15/2012] [Accepted: 09/18/2012] [Indexed: 05/19/2023]
Abstract
We have created a 2.5-mm outer diameter integrated photo-acoustic and ultrasonic mini-probe which can be inserted into a standard video endoscope's instrument channel. A small-diameter focused ultrasonic transducer made of PMN-PT provides adequate signal sensitivity, and enables miniaturization of the probe. Additionally, this new endoscopic probe utilizes the same scanning mirror and micromotor-based built-in actuator described in our previous reports; however, the length of the rigid distal section of the new probe has been further reduced to ~35 mm. This paper describes the technical details of the mini-probe and presents experimental results that both quantify the imaging performance and demonstrate its in vivo imaging capability, which suggests that it could work as a mini-probe for certain clinical applications.
Collapse
Affiliation(s)
- Joon-Mo Yang
- Optical Imaging Laboratory, Department of Biomedical Engineering, Washington University in St. Louis, Campus Box 1097, One Brookings Drive, St. Louis, Missouri 63130,
USA
- These authors contributed equally to this work
| | - Ruimin Chen
- NIH Ultrasonic Transducer Resource Center, Department of Biomedical Engineering, University of Southern California, 1042 Downey Way, University Park, DRB 130, Los Angeles, California 90089,
USA
- These authors contributed equally to this work
| | - Christopher Favazza
- Optical Imaging Laboratory, Department of Biomedical Engineering, Washington University in St. Louis, Campus Box 1097, One Brookings Drive, St. Louis, Missouri 63130,
USA
| | - Junjie Yao
- Optical Imaging Laboratory, Department of Biomedical Engineering, Washington University in St. Louis, Campus Box 1097, One Brookings Drive, St. Louis, Missouri 63130,
USA
| | - Chiye Li
- Optical Imaging Laboratory, Department of Biomedical Engineering, Washington University in St. Louis, Campus Box 1097, One Brookings Drive, St. Louis, Missouri 63130,
USA
| | - Zhilin Hu
- Optical Imaging Laboratory, Department of Biomedical Engineering, Washington University in St. Louis, Campus Box 1097, One Brookings Drive, St. Louis, Missouri 63130,
USA
| | - Qifa Zhou
- NIH Ultrasonic Transducer Resource Center, Department of Biomedical Engineering, University of Southern California, 1042 Downey Way, University Park, DRB 130, Los Angeles, California 90089,
USA
| | - K. Kirk Shung
- NIH Ultrasonic Transducer Resource Center, Department of Biomedical Engineering, University of Southern California, 1042 Downey Way, University Park, DRB 130, Los Angeles, California 90089,
USA
| | - Lihong V. Wang
- Optical Imaging Laboratory, Department of Biomedical Engineering, Washington University in St. Louis, Campus Box 1097, One Brookings Drive, St. Louis, Missouri 63130,
USA
| |
Collapse
|
26
|
Lee MH, Buterbaugh K, Richards-Kortum R, Anandasabapathy S. Advanced endoscopic imaging for Barrett's Esophagus: current options and future directions. Curr Gastroenterol Rep 2012; 14:216-25. [PMID: 22453701 DOI: 10.1007/s11894-012-0259-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Barrett's esophagus is the precursor to esophageal adenocarcinoma, one of the most rapidly increasing cancers in the United States. Given the poor prognosis of late-stage adenocarcinoma, endoscopic surveillance is recommended for subjects with Barrett's esophagus to detect early neoplasia. Current guidelines recommend "random" four-quadrant biopsies taken every 1-2 cm throughout the Barrett's segment. However, this only samples a minority of epithelium and has been shown to miss areas of endoscopically- inapparent neoplasia (high grade dysplasia or cancer). Recent efforts have focused on developing novel diagnostic imaging technologies to detect the subtle epithelial changes associated with dysplasia and neoplasia in Barrett's esophagus. Some of these modalities serve as "red flag" technologies designed to detect areas of abnormality within large surface areas. Other technologies serve to characterize areas of visible abnormality, offering a higher spatial resolution to confirm/exclude the presence of neoplasia. This review summarizes several available and evolving imaging technologies used in the endoscopic diagnosis and surveillance of Barrett's associated neoplasia.
Collapse
Affiliation(s)
- Michelle H Lee
- Division of Gastroenterology, The Mount Sinai Medical Center, New York, NY 10029, USA
| | | | | | | |
Collapse
|
27
|
Vakoc BJ, Fukumura D, Jain RK, Bouma BE. Cancer imaging by optical coherence tomography: preclinical progress and clinical potential. Nat Rev Cancer 2012; 12:363-8. [PMID: 22475930 PMCID: PMC3560400 DOI: 10.1038/nrc3235] [Citation(s) in RCA: 180] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The past decade has seen dramatic technological advances in the field of optical coherence tomography (OCT) imaging. These advances have driven commercialization and clinical adoption in ophthalmology, cardiology and gastrointestinal cancer screening. Recently, an array of OCT-based imaging tools that have been developed for preclinical intravital cancer imaging applications has yielded exciting new capabilities to probe and to monitor cancer progression and response in vivo. Here, we review these results, forecast the future of OCT for preclinical cancer imaging and discuss its exciting potential to translate to the clinic as a tool for monitoring cancer therapy.
Collapse
Affiliation(s)
- Benjamin J Vakoc
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA.
| | | | | | | |
Collapse
|
28
|
Shukla R, Abidi WM, Richards-Kortum R, Anandasabapathy S. Endoscopic imaging: How far are we from real-time histology? World J Gastrointest Endosc 2011; 3:183-94. [PMID: 22013499 PMCID: PMC3196726 DOI: 10.4253/wjge.v3.i10.183] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2010] [Revised: 07/15/2011] [Accepted: 08/30/2011] [Indexed: 02/05/2023] Open
Abstract
Currently, in gastrointestinal endoscopy there is increasing interest in high resolution endoscopic technologies that can complement high-definition white light endoscopy by providing real-time subcellular imaging of the epithelial surface. These ‘optical biopsy’ technologies offer the potential to improve diagnostic accuracy and yield, while facilitating real-time decision-making. Although many endoscopic techniques have preliminarily shown high accuracy rates, these technologies are still evolving. This review will provide an overview of the most promising high-resolution imaging technologies, including high resolution microendoscopy, optical coherence tomography, endocytoscopy and confocal laser endoscopy. This review will also discuss the application and current limitations of these technologies for the early detection of neoplasia in Barrett’s esophagus, ulcerative colitis and colorectal cancer.
Collapse
Affiliation(s)
- Richa Shukla
- Richa Shukla, Wasif M Abidi, Sharmila Anandasabapathy, Department of Medicine, Mount Sinai School of Medicine, New York, NY 10029, United States
| | | | | | | |
Collapse
|
29
|
Matloff JL, Abidi W, Richards-Kortum R, Sauk J, Anandasabapathy S. High-resolution and optical molecular imaging for the early detection of colonic neoplasia. Gastrointest Endosc 2011; 73:1263-73. [PMID: 21628019 DOI: 10.1016/j.gie.2011.01.070] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Accepted: 01/29/2011] [Indexed: 02/08/2023]
Affiliation(s)
- Jeremy L Matloff
- Department of Medicine, Mount Sinai Medical Center, New York, New York 10029, USA
| | | | | | | | | |
Collapse
|