1
|
Chandy M, Wu JC. Molecular Imaging of Stem Cell Therapy in Ischemic Cardiomyopathy. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00065-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
2
|
Abstract
Regenerative medicine with the use of stem cells has appeared as a potential therapeutic alternative for many disease states. Despite initial enthusiasm, there has been relatively slow transition to clinical trials. In large part, numerous questions remain regarding the viability, biology and efficacy of transplanted stem cells in the living subject. The critical issues highlighted the importance of developing tools to assess these questions. Advances in molecular biology and imaging have allowed the successful non-invasive monitoring of transplanted stem cells in the living subject. Over the years these methodologies have been updated to assess not only the viability but also the biology of transplanted stem cells. In this review, different imaging strategies to study the viability and biology of transplanted stem cells are presented. Use of these strategies will be critical as the different regenerative therapies are being tested for clinical use.
Collapse
Affiliation(s)
- Fakhar Abbas
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Joseph C. Wu
- Molecular Imaging Program at Stanford, Stanford University, Stanford, CA, USA
- Department of Medicine (Cardiology), Stanford University, Stanford, CA, USA
| | - Sanjiv Sam Gambhir
- Molecular Imaging Program at Stanford, Stanford University, Stanford, CA, USA
- Department of Bio-Engineering, Stanford University, Stanford, CA, USA
| | | |
Collapse
|
3
|
Mészáros M, Porkoláb G, Kiss L, Pilbat AM, Kóta Z, Kupihár Z, Kéri A, Galbács G, Siklós L, Tóth A, Fülöp L, Csete M, Sipos Á, Hülper P, Sipos P, Páli T, Rákhely G, Szabó-Révész P, Deli MA, Veszelka S. Niosomes decorated with dual ligands targeting brain endothelial transporters increase cargo penetration across the blood-brain barrier. Eur J Pharm Sci 2018; 123:228-240. [PMID: 30031862 DOI: 10.1016/j.ejps.2018.07.042] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 07/13/2018] [Accepted: 07/19/2018] [Indexed: 12/14/2022]
Abstract
Nanoparticles targeting transporters of the blood-brain barrier (BBB) are promising candidates to increase the brain penetration of biopharmacons. Solute carriers (SLC) are expressed at high levels in brain endothelial cells and show a specific pattern at the BBB. The aim of our study was to test glutathione and ligands of SLC transporters as single or dual BBB targeting molecules for nanovesicles. High mRNA expression levels for hexose and neutral amino acid transporting SLCs were found in isolated rat brain microvessels and our rat primary cell based co-culture BBB model. Niosomes were derivatized with glutathione and SLC ligands glucopyranose and alanine. Serum albumin complexed with Evans blue (67 kDa), which has a very low BBB penetration, was selected as a cargo. The presence of targeting ligands on niosomes, especially dual labeling, increased the uptake of the cargo molecule in cultured brain endothelial cells. This cellular uptake was temperature dependent and could be decreased with a metabolic inhibitor and endocytosis blockers filipin and cytochalasin D. Making the negative surface charge of brain endothelial cells more positive with a cationic lipid or digesting the glycocalyx with neuraminidase elevated the uptake of the cargo after treatment with targeted nanocarriers. Treatment with niosomes increased plasma membrane fluidity, suggesting the fusion of nanovesicles with endothelial cell membranes. Targeting ligands elevated the permeability of the cargo across the BBB in the culture model and in mice, and dual-ligand decoration of niosomes was more effective than single ligand labeling. Our data indicate that dual labeling with ligands of multiple SLC transporters can potentially be exploited for BBB targeting of nanoparticles.
Collapse
Affiliation(s)
- Mária Mészáros
- Institute of Biophysics, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári krt. 62, H-6726 Szeged, Hungary; Doctoral School in Theoretical Medicine, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary
| | - Gergő Porkoláb
- Institute of Biophysics, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári krt. 62, H-6726 Szeged, Hungary; Foundation for the Future of Biomedical Sciences in Szeged, Pálfy u. 52/d, H-6725 Szeged, Hungary
| | - Lóránd Kiss
- Institute of Biophysics, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári krt. 62, H-6726 Szeged, Hungary
| | - Ana-Maria Pilbat
- Institute of Biochemistry, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári krt. 62, H-6726 Szeged, Hungary
| | - Zoltán Kóta
- Institute of Biophysics, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári krt. 62, H-6726 Szeged, Hungary
| | - Zoltán Kupihár
- Department of Medical Chemistry, Faculty of Medicine, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary
| | - Albert Kéri
- Department of Inorganic and Analytical Chemistry, Faculty of Science and Informatics, University of Szeged, Dóm tér 7, H-6720 Szeged, Hungary
| | - Gábor Galbács
- Department of Inorganic and Analytical Chemistry, Faculty of Science and Informatics, University of Szeged, Dóm tér 7, H-6720 Szeged, Hungary
| | - László Siklós
- Institute of Biophysics, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári krt. 62, H-6726 Szeged, Hungary
| | - András Tóth
- Institute of Biophysics, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári krt. 62, H-6726 Szeged, Hungary; Department of Biotechnology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary, Közép fasor 52, H-6726 Szeged, Hungary
| | - Lívia Fülöp
- Department of Medical Chemistry, Faculty of Medicine, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary
| | - Mária Csete
- Department of Optics and Quantum Electronics, Faculty of Science and Informatics, University of Szeged, Dóm tér 9, H-6720 Szeged, Hungary
| | - Áron Sipos
- Institute of Biophysics, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári krt. 62, H-6726 Szeged, Hungary
| | - Petra Hülper
- Department of Pediatrics I, University Medical Center Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - Péter Sipos
- Institute of Pharmaceutical Technology and Regulatory Affairs, Faculty of Pharmacy, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary
| | - Tibor Páli
- Institute of Biophysics, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári krt. 62, H-6726 Szeged, Hungary
| | - Gábor Rákhely
- Institute of Biophysics, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári krt. 62, H-6726 Szeged, Hungary; Department of Biotechnology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary, Közép fasor 52, H-6726 Szeged, Hungary
| | - Piroska Szabó-Révész
- Institute of Pharmaceutical Technology and Regulatory Affairs, Faculty of Pharmacy, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary
| | - Mária A Deli
- Institute of Biophysics, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári krt. 62, H-6726 Szeged, Hungary.
| | - Szilvia Veszelka
- Institute of Biophysics, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári krt. 62, H-6726 Szeged, Hungary.
| |
Collapse
|
4
|
Sun J, Miller JP, Hathi D, Zhou H, Achilefu S, Shokeen M, Akers WJ. Enhancing in vivo tumor boundary delineation with structured illumination fluorescence molecular imaging and spatial gradient mapping. JOURNAL OF BIOMEDICAL OPTICS 2016; 21:80502. [PMID: 27580364 PMCID: PMC5005463 DOI: 10.1117/1.jbo.21.8.080502] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 08/05/2016] [Indexed: 05/06/2023]
Abstract
Fluorescence imaging, in combination with tumor-avid near-infrared (NIR) fluorescent molecular probes, provides high specificity and sensitivity for cancer detection in preclinical animal models, and more recently, assistance during oncologic surgery. However, conventional camera-based fluorescence imaging techniques are heavily surface-weighted such that surface reflection from skin or other nontumor tissue and nonspecific fluorescence signals dominate, obscuring true cancer-specific signals and blurring tumor boundaries. To address this challenge, we applied structured illumination fluorescence molecular imaging (SIFMI) in live animals for automated subtraction of nonspecific surface signals to better delineate accumulation of an NIR fluorescent probe targeting α4β1 integrin in mice bearing subcutaneous plasma cell xenografts. SIFMI demonstrated a fivefold improvement in tumor-to-background contrast when compared with other full-field fluorescence imaging methods and required significantly reduced scanning time compared with diffuse optical spectroscopy imaging. Furthermore, the spatial gradient mapping enhanced highlighting of tumor boundaries. Through the relatively simple hardware and software modifications described, SIFMI can be integrated with clinical fluorescence imaging systems, enhancing intraoperative tumor boundary delineation from the uninvolved tissue.
Collapse
Affiliation(s)
- Jessica Sun
- Washington University School of Medicine, Department of Radiology, 4515 McKinley Avenue, St. Louis, Missouri 63110, United States
| | - Jessica P. Miller
- Washington University School of Medicine, Department of Radiology, 4515 McKinley Avenue, St. Louis, Missouri 63110, United States
- Washington University School of Medicine, Department of Biomedical Engineering, 4515 McKinley Avenue, St. Louis, Missouri 63110, United States
| | - Deep Hathi
- Washington University School of Medicine, Department of Radiology, 4515 McKinley Avenue, St. Louis, Missouri 63110, United States
- Washington University School of Medicine, Department of Biomedical Engineering, 4515 McKinley Avenue, St. Louis, Missouri 63110, United States
| | - Haiying Zhou
- Washington University School of Medicine, Department of Radiology, 4515 McKinley Avenue, St. Louis, Missouri 63110, United States
| | - Samuel Achilefu
- Washington University School of Medicine, Department of Radiology, 4515 McKinley Avenue, St. Louis, Missouri 63110, United States
- Washington University School of Medicine, Department of Biomedical Engineering, 4515 McKinley Avenue, St. Louis, Missouri 63110, United States
- Washington University School of Medicine, Department of Biochemistry and Molecular Biophysics, 4515 McKinley Avenue, St. Louis, Missouri 63110, United States
| | - Monica Shokeen
- Washington University School of Medicine, Department of Radiology, 4515 McKinley Avenue, St. Louis, Missouri 63110, United States
| | - Walter J. Akers
- Washington University School of Medicine, Department of Radiology, 4515 McKinley Avenue, St. Louis, Missouri 63110, United States
- Address all correspondence to: Walter J. Akers, E-mail:
| |
Collapse
|
5
|
Mu Y, Niedre M. Fast single photon avalanche photodiode-based time-resolved diffuse optical tomography scanner. BIOMEDICAL OPTICS EXPRESS 2015; 6:3596-3609. [PMID: 26417526 PMCID: PMC4574682 DOI: 10.1364/boe.6.003596] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 08/17/2015] [Accepted: 08/17/2015] [Indexed: 06/05/2023]
Abstract
Resolution in diffuse optical tomography (DOT) is a persistent problem and is primarily limited by high degree of light scatter in biological tissue. We showed previously that the reduction in photon scatter between a source and detector pair at early time points following a laser pulse in time-resolved DOT is highly dependent on the temporal response of the instrument. To this end, we developed a new single-photon avalanche photodiode (SPAD) based time-resolved DOT scanner. This instrument uses an array of fast SPADs, a femto-second Titanium Sapphire laser and single photon counting electronics. In combination, the overall instrument temporal impulse response function width was 59 ps. In this paper, we report the design of this instrument and validate its operation in symmetrical and irregularly shaped optical phantoms of approximately small animal size. We were able to accurately reconstruct the size and position of up to 4 absorbing inclusions, with increasing image quality at earlier time windows. We attribute these results primarily to the rapid response time of our instrument. These data illustrate the potential utility of fast SPAD detectors in time-resolved DOT.
Collapse
Affiliation(s)
- Ying Mu
- Department of Electrical and Computer Engineering, Dana Research Center, Northeastern University, Boston, MA, 02115, USA
| | - Mark Niedre
- Department of Electrical and Computer Engineering, Dana Research Center, Northeastern University, Boston, MA, 02115, USA
| |
Collapse
|
6
|
Jung HK, Wang K, Jung MK, Kim IS, Lee BH. In vivo near-infrared fluorescence imaging of apoptosis using histone H1-targeting peptide probe after anti-cancer treatment with cisplatin and cetuximab for early decision on tumor response. PLoS One 2014; 9:e100341. [PMID: 24949860 PMCID: PMC4065102 DOI: 10.1371/journal.pone.0100341] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 05/23/2014] [Indexed: 12/29/2022] Open
Abstract
Early decision on tumor response after anti-cancer treatment is still an unmet medical need. Here we investigated whether in vivo imaging of apoptosis using linear and cyclic (disulfide-bonded) form of ApoPep-1, a peptide that recognizes histone H1 exposed on apoptotic cells, at an early stage after treatment could predict tumor response to the treatment later. Treatment of stomach tumor cells with cistplatin or cetuximab alone induced apoptosis, while combination of cisplatin plus cetuximab more efficiently induced apoptosis, as detected by binding with linear and cyclic form of ApoPep-1. However, the differences between the single agent and combination treatment were more remarkable as detected with the cyclic form compared to the linear form. In tumor-bearing mice, apoptosis imaging was performed 1 week and 2 weeks after the initiation of treatment, while tumor volumes and weights were measured 3 weeks after the treatment. In vivo fluorescence imaging signals obtained by the uptake of ApoPep-1 to tumor was most remarkable in the group injected with cyclic form of ApoPep-1 at 1 week after combined treatment with cisplatin plus cetuximab. Correlation analysis revealed that imaging signals by cyclic ApoPep-1 at 1 week after treatment with cisplatin plus cetuximab in combination were most closely related with tumor volume changes (r2 = 0.934). These results demonstrate that in vivo apoptosis imaging using Apopep-1, especially cyclic ApoPep-1, is a sensitive and predictive tool for early decision on stomach tumor response after anti-cancer treatment.
Collapse
Affiliation(s)
- Hyun-Kyung Jung
- Department of Biochemistry and Cell Biology and School of Medicine, Kyungpook National University, Daegu, Korea
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu, Korea
| | - Kai Wang
- Department of Plastic Surgery, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Min Kyu Jung
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Korea
| | - In-San Kim
- Department of Biochemistry and Cell Biology and School of Medicine, Kyungpook National University, Daegu, Korea
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu, Korea
| | - Byung-Heon Lee
- Department of Biochemistry and Cell Biology and School of Medicine, Kyungpook National University, Daegu, Korea
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu, Korea
- * E-mail:
| |
Collapse
|
7
|
Ardeshirpour Y, Chernomordik V, Hassan M, Zielinski R, Capala J, Gandjbakhche A. In vivo fluorescence lifetime imaging for monitoring the efficacy of the cancer treatment. Clin Cancer Res 2014; 20:3531-9. [PMID: 24671949 DOI: 10.1158/1078-0432.ccr-13-1826] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
PURPOSE Advances in tumor biology created a foundation for targeted therapy aimed at inactivation of specific molecular mechanisms responsible for cell malignancy. In this paper, we used in vivo fluorescence lifetime imaging with HER2-targeted fluorescent probes as an alternative imaging method to investigate the efficacy of targeted therapy with 17-DMAG (an HSP90 inhibitor) on tumors with high expression of HER2 receptors. EXPERIMENTAL DESIGN HER2-specific Affibody, conjugated to Alexafluor 750, was injected into nude mice bearing HER2-positive tumor xenograft. The fluorescence lifetime was measured before treatment and monitored after the probe injections at 12 hours after the last treatment dose, when the response to the 17-DMAG therapy was the most pronounced as well as a week after the last treatment when the tumors grew back almost to their pretreatment size. RESULTS Imaging results showed significant difference between the fluorescence lifetimes at the tumor and the contralateral site (∼0.13 ns) in the control group (before treatment) and 7 days after the last treatment when the tumors grew back to their pretreatment dimensions. However, at the time frame that the treatment had its maximum effect (12 hours after the last treatment), the difference between the fluorescence lifetime at the tumor and contralateral site decreased to 0.03 ns. CONCLUSIONS The results showed a good correlation between fluorescence lifetime and the efficacy of the treatment. These findings show that in vivo fluorescence lifetime imaging can be used as a promising molecular imaging tool for monitoring the treatment outcome in preclinical models and potentially in patients.
Collapse
Affiliation(s)
- Yasaman Ardeshirpour
- Authors' Affiliations: NIH/National Institute of Child Health and Human Development, Bethesda
| | - Victor Chernomordik
- Authors' Affiliations: NIH/National Institute of Child Health and Human Development, Bethesda
| | - Moinuddin Hassan
- Authors' Affiliations: NIH/National Institute of Child Health and Human Development, Bethesda; Division of Physics, Office of Science and Engineering Laboratories, CDRH, FDA, Silver Spring
| | - Rafal Zielinski
- NIH/National Cancer Institute, Rockville, Maryland; and UT MD Anderson Cancer Center, Houston, Texas
| | - Jacek Capala
- NIH/National Cancer Institute, Rockville, Maryland; and
| | - Amir Gandjbakhche
- Authors' Affiliations: NIH/National Institute of Child Health and Human Development, Bethesda;
| |
Collapse
|
8
|
Abstract
Diffuse optical imaging is highly versatile and has a very broad range of applications in biology and medicine. It covers diffuse optical tomography, fluorescence diffuse optical tomography, bioluminescence, and a number of other new imaging methods. These methods of diffuse optical imaging have diversified instrument configurations but share the same core physical principle – light propagation in highly diffusive media, i.e., the biological tissue. In this review, the author summarizes the latest development in instrumentation and methodology available to diffuse optical imaging in terms of system architecture, light source, photo-detection, spectral separation, signal modulation, and lastly imaging contrast.
Collapse
|
9
|
Abstract
Advances in noninvasive imaging technologies that allow for in vivo dynamic monitoring of cells and cellular function in living research subjects have revealed new insights into cell biology in the context of intact organs and their native environment. In the field of hematopoiesis and stem cell research, studies of cell trafficking involved in injury repair and hematopoietic engraftment have made great progress using these new tools. Stem cells present unique challenges for imaging since after transplantation, they proliferate dramatically and differentiate. Therefore, the imaging modality used needs to have a large dynamic range, and the genetic regulatory elements used need to be stably expressed during differentiation. Multiple imaging technologies using different modalities are available, and each varies in sensitivity, ease of data acquisition, signal to noise ratios (SNR), substrate availability, and other parameters that affect utility for monitoring cell fates and function. For a given application, there may be several different approaches that can be used. For mouse models, clinically validated technologies such as magnetic resonance imaging (MRI) and positron emission tomography (PET) have been joined by optical imaging techniques such as in vivo bioluminescence imaging (BLI) and fluorescence imaging (FLI), and all have been used to monitor bone marrow and stem cells after transplantation into mice. Photoacoustic imaging that utilizes the sound created by the thermal expansion of absorbed light to generate an image best represents hybrid technologies. Each modality requires that the cells of interest be marked with a genetic reporter that acts as a label making them uniquely visible using that technology. For each modality, there are several labels to choose from. Multiple methods for applying these different labels are available. This chapter provides an overview of the imaging technologies and commonly used labels for each, as well as detailed protocols for gene delivery into hematopoietic cells for the purposes of applying these specific labels to cell trafficking. The goal of this chapter is to provide adequate background information to allow the design and implementation of an experimental system for in vivo imaging in mice.
Collapse
|
10
|
Liao WY, Li HJ, Chang MY, Tang ACL, Hoffman AS, Hsieh PCH. Comprehensive characterizations of nanoparticle biodistribution following systemic injection in mice. NANOSCALE 2013; 5:11079-11086. [PMID: 24072256 DOI: 10.1039/c3nr03954d] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Various nanoparticle (NP) properties such as shape and surface charge have been studied in an attempt to enhance the efficacy of NPs in biomedical applications. When trying to undermine the precise biodistribution of NPs within the target organs, the analytical method becomes the determining factor in measuring the precise quantity of distributed NPs. High performance liquid chromatography (HPLC) represents a more powerful tool in quantifying NP biodistribution compared to conventional analytical methods such as an in vivo imaging system (IVIS). This, in part, is due to better curve linearity offered by HPLC than IVIS. Furthermore, HPLC enables us to fully analyze each gram of NPs present in the organs without compromising the signals and the depth-related sensitivity as is the case in IVIS measurements. In addition, we found that changing physiological conditions improved large NP (200-500 nm) distribution in brain tissue. These results reveal the importance of selecting analytic tools and physiological environment when characterizing NP biodistribution for future nanoscale toxicology, therapeutics and diagnostics.
Collapse
Affiliation(s)
- Wei-Yin Liao
- Institute of Clinical Medicine, National Cheng Kung University & Hospital, Tainan 704, Taiwan, R.O.C..
| | | | | | | | | | | |
Collapse
|
11
|
Ma G. Background-free in vivo time domain optical molecular imaging using colloidal quantum dots. ACS APPLIED MATERIALS & INTERFACES 2013; 5:2835-2844. [PMID: 23448359 DOI: 10.1021/am3028519] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
The interest in optical molecular imaging of small animals in vivo has been steadily increased in the last two decades as it is being adopted by not only academic laboratories but also the biotechnical and pharmaceutical industries. In this Spotlight paper, the elements for in vivo optical molecular imaging are briefly reviewed, including contrast agents, i.e., various fluorescent reporters, and the most commonly used technologies to detect the reporters. The challenges particularly for in vivo fluorescence imaging are discussed and solutions to overcome the said-challenges are presented. An advanced imaging technique, in vivo fluorescence lifetime imaging, is introduced together with a few application examples. Taking advantage of the long fluorescence lifetime of quantum dots, a method to achieve background-free in vivo fluorescence small animal imaging is demonstrated.
Collapse
Affiliation(s)
- Guobin Ma
- ART Advanced Research Technologies Inc., 2300 Alfred-Nobel Boulevard, Montreal, Quebec, Canada H4S 2A4.
| |
Collapse
|
12
|
Cool SK, Breyne K, Meyer E, De Smedt SC, Sanders NN. Comparison of in vivo optical systems for bioluminescence and fluorescence imaging. J Fluoresc 2013; 23:909-20. [PMID: 23579930 DOI: 10.1007/s10895-013-1215-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Accepted: 04/04/2012] [Indexed: 01/26/2023]
Abstract
In vivo optical imaging has become a popular tool in animal laboratories. Currently, many in vivo optical imaging systems are available on the market, which often makes it difficult for research groups to decide which system fits their needs best. In this work we compared different commercially available systems, which can measure both bioluminescent and fluorescent light. The systems were tested for their bioluminescent and fluorescent sensitivity both in vitro and in vivo. The IVIS Lumina II was found to be most sensitive for bioluminescence imaging, with the Photon Imager a close second. Contrary, the Kodak system was, in vitro, the most sensitive system for fluorescence imaging. In vivo, the fluorescence sensitivity of the systems was similar. Finally, we examined the added value of spectral unmixing algorithms for in vivo optical imaging and demonstrated that spectral unmixing resulted in at least a doubling of the in vivo sensitivity. Additionally, spectral unmixing also enabled separate imaging of dyes with overlapping spectra which were, without spectral unmixing, not distinguishable.
Collapse
Affiliation(s)
- Steven K Cool
- Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | | | | | | | | |
Collapse
|
13
|
Ren S, Chen X, Wang H, Qu X, Wang G, Liang J, Tian J. Molecular Optical Simulation Environment (MOSE): a platform for the simulation of light propagation in turbid media. PLoS One 2013; 8:e61304. [PMID: 23577215 PMCID: PMC3620115 DOI: 10.1371/journal.pone.0061304] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 03/06/2013] [Indexed: 01/28/2023] Open
Abstract
The study of light propagation in turbid media has attracted extensive attention in the field of biomedical optical molecular imaging. In this paper, we present a software platform for the simulation of light propagation in turbid media named the “Molecular Optical Simulation Environment (MOSE)”. Based on the gold standard of the Monte Carlo method, MOSE simulates light propagation both in tissues with complicated structures and through free-space. In particular, MOSE synthesizes realistic data for bioluminescence tomography (BLT), fluorescence molecular tomography (FMT), and diffuse optical tomography (DOT). The user-friendly interface and powerful visualization tools facilitate data analysis and system evaluation. As a major measure for resource sharing and reproducible research, MOSE aims to provide freeware for research and educational institutions, which can be downloaded at http://www.mosetm.net.
Collapse
Affiliation(s)
- Shenghan Ren
- School of Life Sciences and Technology, Xidian University, Xi’an, Shaanxi, China
| | - Xueli Chen
- School of Life Sciences and Technology, Xidian University, Xi’an, Shaanxi, China
| | - Hailong Wang
- School of Life Sciences and Technology, Xidian University, Xi’an, Shaanxi, China
| | - Xiaochao Qu
- School of Life Sciences and Technology, Xidian University, Xi’an, Shaanxi, China
| | - Ge Wang
- Biomedical Imaging Center, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- * E-mail: (GW); (JL); (JT)
| | - Jimin Liang
- School of Life Sciences and Technology, Xidian University, Xi’an, Shaanxi, China
- * E-mail: (GW); (JL); (JT)
| | - Jie Tian
- School of Life Sciences and Technology, Xidian University, Xi’an, Shaanxi, China
- Institute of Automation, Chinese Academy of Sciences, Beijing, China
- * E-mail: (GW); (JL); (JT)
| |
Collapse
|
14
|
McCormack E, Silden E, West RM, Pavlin T, Micklem DR, Lorens JB, Haug BE, Cooper ME, Gjertsen BT. Nitroreductase, a near-infrared reporter platform for in vivo time-domain optical imaging of metastatic cancer. Cancer Res 2012; 73:1276-86. [PMID: 23233739 DOI: 10.1158/0008-5472.can-12-2649] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The ability to visualize reporter gene expression in vivo has revolutionized all facets of biologic investigation and none more so than imaging applications in oncology. Near-infrared reporter gene imaging may facilitate more accurate evaluation of chemotherapeutic response in preclinical models of orthotopic and metastatic cancers. We report the development of a cell permeable, quenched squarine probe (CytoCy5S), which is reduced by Escherichia coli nitroreductase (NTR), resulting in a near-infrared fluorescent product. Time-domain molecular imaging of NTR/CytoCy5S reporter platform permitted noninvasive monitoring of disease progression in orthotopic xenografts of disseminated leukemia, lung, and metastatic breast cancer. This methodology facilitated therapeutic evaluation of NTR gene-directed enzymatic prodrug therapy with conventional metronidazole antibiotics. These studies show NTR/CytoCy5S as a near-infrared gene reporter system with broad preclinical and prospective clinical applications within imaging, and gene therapy, of cancer.
Collapse
Affiliation(s)
- Emmet McCormack
- Institute of Medicine, Hematology Section; Department of Biomedicine, University of Bergen, Norway.
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Nakajima T, Sano K, Mitsunaga M, Choyke PL, Kobayashi H. Real-time monitoring of in vivo acute necrotic cancer cell death induced by near infrared photoimmunotherapy using fluorescence lifetime imaging. Cancer Res 2012; 72:4622-8. [PMID: 22800710 PMCID: PMC3445723 DOI: 10.1158/0008-5472.can-12-1298] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A new type of monoclonal antibody (mAb)-based, highly specific phototherapy (photoimmunotherapy; PIT) that uses a near infrared (NIR) phthalocyanine dye, IRDye700DX (IR700) conjugated with a mAb, has recently been described. NIR light exposure leads to immediate, target-selective necrotic cell death in vitro. Detecting immediate in vivo cell death is more difficult because it takes at least 3 days for the tumor to begin to shrink in size. In this study, fluorescence lifetime (FLT) was evaluated before and after PIT for monitoring the immediate cytotoxic effects of NIR mediated mAb-IR700 PIT. Anti-epidermal growth factor receptor (EGFR) panitumumab-IR700 was used for targeting EGFR-expressing A431 tumor cells. PIT with various doses of NIR light was conducted in cell pellets in vitro and in subcutaneously xenografted tumors in mice in vivo. FLT measurements were obtained before and 0, 6, 24, and 48 hours after PIT. In vitro, PIT at higher doses of NIR light immediately led to FLT shortening in A431 cells. In vivo PIT induced immediate shortening of FLT in treated tumors after a threshold NIR dose of 30 J/cm(2) or greater. In contrast, lower levels of NIR light (10 J/cm(2) or smaller) did not induce shortening of FLT. Prolongation of FLT in tissue surrounding the tumor site was noted 6 hours after PIT, likely reflecting phagocytosis by macrophages. In conclusion, FLT imaging can be used to monitor the acute cytotoxic effects of mAb-IR700-induced PIT even before morphological changes can be seen in the targeted tumors.
Collapse
Affiliation(s)
- Takahito Nakajima
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | |
Collapse
|
16
|
James ML, Gambhir SS. A molecular imaging primer: modalities, imaging agents, and applications. Physiol Rev 2012; 92:897-965. [PMID: 22535898 DOI: 10.1152/physrev.00049.2010] [Citation(s) in RCA: 729] [Impact Index Per Article: 56.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Molecular imaging is revolutionizing the way we study the inner workings of the human body, diagnose diseases, approach drug design, and assess therapies. The field as a whole is making possible the visualization of complex biochemical processes involved in normal physiology and disease states, in real time, in living cells, tissues, and intact subjects. In this review, we focus specifically on molecular imaging of intact living subjects. We provide a basic primer for those who are new to molecular imaging, and a resource for those involved in the field. We begin by describing classical molecular imaging techniques together with their key strengths and limitations, after which we introduce some of the latest emerging imaging modalities. We provide an overview of the main classes of molecular imaging agents (i.e., small molecules, peptides, aptamers, engineered proteins, and nanoparticles) and cite examples of how molecular imaging is being applied in oncology, neuroscience, cardiology, gene therapy, cell tracking, and theranostics (therapy combined with diagnostics). A step-by-step guide to answering biological and/or clinical questions using the tools of molecular imaging is also provided. We conclude by discussing the grand challenges of the field, its future directions, and enormous potential for further impacting how we approach research and medicine.
Collapse
Affiliation(s)
- Michelle L James
- Molecular Imaging Program, Department of Radiology, Stanford University, Palo Alto, CA 94305, USA
| | | |
Collapse
|
17
|
Filfil R, Paul-Roc B, Cantin C, Iqbal U, Tolkatchev D, Vinogradova A, Xu P, Ni F, O'Connor-McCourt MD, Lenferink AE. Molecular imaging of breast tumors using a near-infrared fluorescently labeled clusterin binding peptide. Int J Cancer 2012; 131:E681-92. [DOI: 10.1002/ijc.27368] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Accepted: 10/31/2011] [Indexed: 12/31/2022]
|
18
|
van de Ven SMWY, Elias SG, Chan CT, Miao Z, Cheng Z, De A, Gambhir SS. Optical imaging with her2-targeted affibody molecules can monitor hsp90 treatment response in a breast cancer xenograft mouse model. Clin Cancer Res 2012; 18:1073-81. [PMID: 22235098 DOI: 10.1158/1078-0432.ccr-10-3213] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
PURPOSE To determine whether optical imaging can be used for in vivo therapy response monitoring as an alternative to radionuclide techniques. For this, we evaluated the known Her2 response to 17-dimethylaminoethylamino-17-demethoxygeldanamycin hydrochloride (17-DMAG) treatment, an Hsp90 inhibitor. EXPERIMENTAL DESIGN After in vitro 17-DMAG treatment response evaluation of MCF7 parental cells and 2 HER2-transfected clones (clone A medium, B high Her2 expression), we established human breast cancer xenografts in nude mice (only parental and clone B) for in vivo evaluation. Mice received 120 mg/kg of 17-DMAG in 4 doses at 12-hour intervals intraperitonially (n = 14) or PBS as carrier control (n = 9). Optical images were obtained both pretreatment (day 0) and posttreatment (day 3, 6, and 9), always 5 hours postinjection of 500 pmol of anti-Her2 Affibody-AlexaFluor680 via tail vein (with preinjection background subtraction). Days 3 and 9 in vivo optical imaging signal was further correlated with ex vivo Her2 levels by Western blot after sacrifice. RESULTS Her2 expression decreased with 17-DMAG dose in vitro. In vivo optical imaging signal was reduced by 22.5% in clone B (P = 0.003) and by 9% in MCF7 parental tumors (P = 0.23) 3 days after 17-DMAG treatment; optical imaging signal recovered in both tumor types at days 6 to 9. In the carrier group, no signal reduction was observed. Pearson correlation of in vivo optical imaging signal with ex vivo Her2 levels ranged from 0.73 to 0.89. CONCLUSIONS Optical imaging with an affibody can be used to noninvasively monitor changes in Her2 expression in vivo as a response to treatment with an Hsp90 inhibitor, with results similar to response measurements in positron emission tomography imaging studies.
Collapse
|
19
|
Mathejczyk JE, Pauli J, Dullin C, Napp J, Tietze LF, Kessler H, Resch-Genger U, Alves F. Spectroscopically Well-Characterized RGD Optical Probe as a Prerequisite for Lifetime-Gated Tumor Imaging. Mol Imaging 2011. [DOI: 10.2310/7290.2011.00018] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Labeling of RGD peptides with near-infrared fluorophores yields optical probes for noninvasive imaging of tumors overexpressing αvβ3 integrins. An important prerequisite for optimum detection sensitivity in vivo is strongly absorbing and highly emissive probes with a known fluorescence lifetime. The RGD-Cy5.5 optical probe was derived by coupling Cy5.5 to a cyclic arginine–glycine–aspartic acid–d-phenylalanine–lysine (RGDfK) peptide via an aminohexanoic acid spacer. Spectroscopic properties of the probe were studied in different matrices in comparison to Cy5.5. For in vivo imaging, human glioblastoma cells were subcutaneously implanted into nude mice, and in vivo fluorescence intensity and lifetime were measured. The fluorescence quantum yield and lifetime of Cy5.5 were found to be barely affected on RGD conjugation but dramatically changed in the presence of proteins. By time domain fluorescence imaging, we demonstrated specific binding of RGD-Cy5.5 to glioblastoma xenografts in nude mice. Discrimination of unspecific fluorescence by lifetime-gated analysis further enhanced the detection sensitivity of RGD-Cy5.5-derived signals. We characterized RGD-Cy5.5 as a strongly emissive and stable probe adequate for selective targeting of αvβ3 integrins. The specificity and thus the overall detection sensitivity in vivo were optimized with lifetime gating, based on the previous determination of the probes fluorescence lifetime under application-relevant conditions.
Collapse
Affiliation(s)
- Julia Eva Mathejczyk
- From the Department of Molecular Biology of Neuronal Signals, Max-Planck-Institute for Experimental Medicine, Göttingen, Germany; Departments of Hematology and Oncology and Diagnostic Radiology, University Medical Center Göttingen, Göttingen, Germany; BAM Federal Institute for Materials Research and Testing, Berlin, Germany; Department of Organic and Biomolecular Chemistry, University of Göttingen, Göttingen, Germany; and Institute for Advanced Study and Center of Integrated Protein Science Munich,
| | - Jutta Pauli
- From the Department of Molecular Biology of Neuronal Signals, Max-Planck-Institute for Experimental Medicine, Göttingen, Germany; Departments of Hematology and Oncology and Diagnostic Radiology, University Medical Center Göttingen, Göttingen, Germany; BAM Federal Institute for Materials Research and Testing, Berlin, Germany; Department of Organic and Biomolecular Chemistry, University of Göttingen, Göttingen, Germany; and Institute for Advanced Study and Center of Integrated Protein Science Munich,
| | - Christian Dullin
- From the Department of Molecular Biology of Neuronal Signals, Max-Planck-Institute for Experimental Medicine, Göttingen, Germany; Departments of Hematology and Oncology and Diagnostic Radiology, University Medical Center Göttingen, Göttingen, Germany; BAM Federal Institute for Materials Research and Testing, Berlin, Germany; Department of Organic and Biomolecular Chemistry, University of Göttingen, Göttingen, Germany; and Institute for Advanced Study and Center of Integrated Protein Science Munich,
| | - Joanna Napp
- From the Department of Molecular Biology of Neuronal Signals, Max-Planck-Institute for Experimental Medicine, Göttingen, Germany; Departments of Hematology and Oncology and Diagnostic Radiology, University Medical Center Göttingen, Göttingen, Germany; BAM Federal Institute for Materials Research and Testing, Berlin, Germany; Department of Organic and Biomolecular Chemistry, University of Göttingen, Göttingen, Germany; and Institute for Advanced Study and Center of Integrated Protein Science Munich,
| | - Lutz-F. Tietze
- From the Department of Molecular Biology of Neuronal Signals, Max-Planck-Institute for Experimental Medicine, Göttingen, Germany; Departments of Hematology and Oncology and Diagnostic Radiology, University Medical Center Göttingen, Göttingen, Germany; BAM Federal Institute for Materials Research and Testing, Berlin, Germany; Department of Organic and Biomolecular Chemistry, University of Göttingen, Göttingen, Germany; and Institute for Advanced Study and Center of Integrated Protein Science Munich,
| | - Horst Kessler
- From the Department of Molecular Biology of Neuronal Signals, Max-Planck-Institute for Experimental Medicine, Göttingen, Germany; Departments of Hematology and Oncology and Diagnostic Radiology, University Medical Center Göttingen, Göttingen, Germany; BAM Federal Institute for Materials Research and Testing, Berlin, Germany; Department of Organic and Biomolecular Chemistry, University of Göttingen, Göttingen, Germany; and Institute for Advanced Study and Center of Integrated Protein Science Munich,
| | - Ute Resch-Genger
- From the Department of Molecular Biology of Neuronal Signals, Max-Planck-Institute for Experimental Medicine, Göttingen, Germany; Departments of Hematology and Oncology and Diagnostic Radiology, University Medical Center Göttingen, Göttingen, Germany; BAM Federal Institute for Materials Research and Testing, Berlin, Germany; Department of Organic and Biomolecular Chemistry, University of Göttingen, Göttingen, Germany; and Institute for Advanced Study and Center of Integrated Protein Science Munich,
| | - Frauke Alves
- From the Department of Molecular Biology of Neuronal Signals, Max-Planck-Institute for Experimental Medicine, Göttingen, Germany; Departments of Hematology and Oncology and Diagnostic Radiology, University Medical Center Göttingen, Göttingen, Germany; BAM Federal Institute for Materials Research and Testing, Berlin, Germany; Department of Organic and Biomolecular Chemistry, University of Göttingen, Göttingen, Germany; and Institute for Advanced Study and Center of Integrated Protein Science Munich,
| |
Collapse
|
20
|
Hülper P, Dullin C, Kugler W, Lakomek M, Erdlenbruch B. Monitoring proteins using in vivo near-infrared time-domain optical imaging after 2-O-hexyldiglycerol-mediated transfer to the brain. Mol Imaging Biol 2011; 13:275-83. [PMID: 20521132 PMCID: PMC3051066 DOI: 10.1007/s11307-010-0348-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Purpose The aim of the present study was to gain insight into the penetration, biodistribution, and fate of globulins in the brain after 2-O-hexyldiglycerol-induced blood–brain barrier opening. Procedures The spatial distribution of fluorescence probes was investigated after blood–brain barrier opening with intracarotid 2-O-hexyldiglycerol injection. Fluorescence intensity was visualized by microscopy (mice and rats) and by in vivo time-domain optical imaging. Results There was an increased 2-O-hexyldiglycerol-mediated transfer of fluorescence-labeled globulins into the ipsilateral hemisphere. Sequential in vivo measurements revealed that the increase in protein concentration lasted at least 96 h after administration. Ex vivo detection of tissue fluorescence confirmed the results obtained in vivo. Conclusion Globulins enter the healthy brain in conjunction with 2-O-hexyldiglycerol. Sequential in vivo near-infrared fluorescence measurements enable the visualization of the spatial distribution of antibodies in the brain of living small animals.
Collapse
Affiliation(s)
- Petra Hülper
- Kinderklinik der Universität Göttingen, Robert-Koch-Str. 40, 37099 Göttingen, Germany.
| | | | | | | | | |
Collapse
|
21
|
Napp J, Dullin C, Müller F, Uhland K, Petri JB, van de Locht A, Steinmetzer T, Alves F. Time-domain in vivo near infrared fluorescence imaging for evaluation of matriptase as a potential target for the development of novel, inhibitor-based tumor therapies. Int J Cancer 2010; 127:1958-74. [PMID: 20473895 DOI: 10.1002/ijc.25405] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Proteolytic enzymes expressed on the surface of tumor cells, and thus easily accessible to external interventions, represent useful targets for anticancer and antimetastatic therapies. In our study, we thoroughly evaluated matriptase, a trypsin-like transmembrane serine protease, as potential target for novel inhibitor-based tumor therapies. We applied time-domain near infrared fluorescence (NIRF) imaging to characterize expression and activity of matriptase in vivo in an orthotopic AsPC-1 pancreatic tumor model in nude mice. We show strong and tumor-specific binding of intravenously injected Cy5.5 labeled antimatriptase antibody (MT-Ab*Cy5.5) only to primary AsPC-1 tumors and their metastases over time within living mice, taking into account fluorescence intensities and fluorescence lifetimes of the applied probes. Specific binding of MT-Ab*Cy5.5 to tumor sites was confirmed by ex vivo NIRF imaging of tumor tissue, NIRF microscopy and by coregistration of the in vivo acquired NIRF intensity maps to anatomical structures visualized by flat-panel volume computed tomography (fpVCT) in living mice. Moreover, using an activatable synthetic substrate S*DY-681 we could clearly demonstrate that matriptase is proteolytically active in vitro as well as in vivo in tumor-bearing mice, and that application of synthetic active-site inhibitors having high affinity and selectivity toward matriptase can efficiently inhibit its proteolytic activity for at least 24 hr. We thus successfully applied NIRF imaging in combination with fpVCT to characterize matriptase as a promising molecular target for inhibitor-based cancer therapies.
Collapse
Affiliation(s)
- Joanna Napp
- Department of Haematology and Oncology, University Medical Center, Goettingen, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
22
|
de la Zerda A, Bodapati S, Teed R, Schipper ML, Keren S, Smith BR, Ng JST, Gambhir SS. A comparison between time domain and spectral imaging systems for imaging quantum dots in small living animals. Mol Imaging Biol 2009; 12:500-8. [PMID: 20012220 DOI: 10.1007/s11307-009-0290-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2009] [Revised: 07/06/2009] [Accepted: 07/29/2009] [Indexed: 10/20/2022]
Abstract
PURPOSE We quantified the performance of time-domain imaging (TDI) and spectral imaging (SI) for fluorescence imaging of quantum dots (QDs) in three distinct imaging instruments: eXplore Optix (TDI, Advanced Research Technologies Inc.), Maestro (SI, CRi Inc.), and IVIS-Spectrum (SI, Caliper Life Sciences Inc.). PROCEDURE The instruments were compared for their sensitivity in phantoms and living mice, multiplexing capabilities (ability to resolve the signal of one QD type in the presence of another), and the dependence of contrast and spatial resolution as a function of depth. RESULTS In phantoms, eXplore Optix had an order of magnitude better sensitivity compared to the SI systems, detecting QD concentrations of ~40 pM in vitro. Maestro was the best instrument for multiplexing QDs. Reduction of contrast and resolution as a function of depth was smallest with eXplore Optix for depth of 2-6 mm, while other depths gave comparable results in all systems. Sensitivity experiments in living mice showed that the eXplore Optix and Maestro systems outperformed the IVIS-Spectrum. CONCLUSION TDI was found to be an order of magnitude more sensitive than SI at the expense of speed and very limited multiplexing capabilities. For deep tissue QD imaging, TDI is most applicable for depths between 2 and 6 mm, as its contrast and resolution degrade the least at these depths.
Collapse
Affiliation(s)
- Adam de la Zerda
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), The Bio-X Program, Stanford, CA 94305, USA
| | | | | | | | | | | | | | | |
Collapse
|
23
|
May A, Bhaumik S, Gambhir SS, Zhan C, Yazdanfar S. Whole-body, real-time preclinical imaging of quantum dot fluorescence with time-gated detection. JOURNAL OF BIOMEDICAL OPTICS 2009; 14:060504. [PMID: 20059235 PMCID: PMC2801727 DOI: 10.1117/1.3269675] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
We describe a wide-field preclinical imaging system optimized for time-gated detection of quantum dot fluorescence emission. As compared to continuous wave measurements, image contrast was substantially improved by suppression of short-lifetime background autofluorescence. Real-time (8 frames/s) biological imaging of subcutaneous quantum dot injections is demonstrated simultaneously in multiple living mice.
Collapse
|
24
|
Kim SK, Kim JY, Choi YS, Lee MK, Lee BW, Lee HC, Lee EJ. A dual-reporter system for specific tracing of pancreatic ß-cell lines that non-invasively measures viable in vivo islet cells. Biotechnol Lett 2009; 32:53-7. [DOI: 10.1007/s10529-009-0113-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2009] [Revised: 08/14/2009] [Accepted: 08/14/2009] [Indexed: 11/28/2022]
|
25
|
Method of bioluminescence imaging for molecular imaging of physiological and pathological processes. Methods 2009; 48:139-45. [PMID: 19324090 DOI: 10.1016/j.ymeth.2009.03.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2009] [Accepted: 03/11/2009] [Indexed: 01/20/2023] Open
Abstract
Molecular imaging has emerged as a powerful tool in basic, pre-clinical and clinical research for monitoring a variety of molecular and cellular processes in living organisms. Optical imaging techniques, mainly bioluminescence imaging, have extensively been used to study biological processes because of their exquisite sensitivity and high signal-to noise ratio. However, current applications have mainly been limited to small animals due to attenuation and scattering of light by tissues but efforts are ongoing to overcome these hurdles. Here, we focus on bioluminescence imaging by giving a brief overview of recent advances in instrumentation, current available reporter gene-reporter probe systems and applications such as cell trafficking, protein-protein interactions and imaging endogenous processes.
Collapse
|
26
|
Marjono A, Yano A, Okawa S, Gao F, Yamada Y. Total light approach of time-domain fluorescence diffuse optical tomography. OPTICS EXPRESS 2008; 16:15268-85. [PMID: 18795065 DOI: 10.1364/oe.16.015268] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
In this study, time-domain fluorescence diffuse optical tomography in biological tissue is numerically investigated using a total light approach. Total light is a summation of excitation light and zero-lifetime emission light divided by quantum yield. The zero-lifetime emission light is an emitted fluorescence light calculated by assuming that the fluorescence lifetime is zero. The zero-lifetime emission light is calculated by deconvolving the actually measured emission light with a lifetime function, an exponential function for fluorescence decay. The object for numerical simulation is a 2-D 10 mm-radius circle with the optical properties simulating biological tissues for near infrared light, and contains regions with fluorophore. The inverse problem of fluorescence diffuse optical tomography is solved using time-resolved simulated measurement data of the excitation and total lights for reconstructing the bsorption coefficient and fluorophore concentration simultaneously. The mean time of flight is used as the featured data-type extracted from the time-resolved data. The reconstructed images of fluorophore concentration show good quantitativeness and spatial reproducibility. By use of the total light approach, computation is performed much faster than the conventional ones.
Collapse
Affiliation(s)
- Andhi Marjono
- Department of Mechanical Engineering and Intelligent Systems, University of Electro-Communications, Chofu, Tokyo, Japan.
| | | | | | | | | |
Collapse
|
27
|
DE LA ZERDA ADAM, ZAVALETA CRISTINA, KEREN SHAY, VAITHILINGAM SRIKANT, BODAPATI SUNIL, LIU ZHUANG, LEVI JELENA, SMITH BRYANR, MA TEJEN, ORALKAN OMER, CHENG ZHEN, CHEN XIAOYUAN, DAI HONGJIE, KHURI-YAKUB BUTRUST, GAMBHIR SANJIVS. Carbon nanotubes as photoacoustic molecular imaging agents in living mice. NATURE NANOTECHNOLOGY 2008; 3:557-62. [PMID: 18772918 PMCID: PMC2562547 DOI: 10.1038/nnano.2008.231] [Citation(s) in RCA: 788] [Impact Index Per Article: 46.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2007] [Accepted: 07/04/2008] [Indexed: 05/18/2023]
Abstract
Photoacoustic imaging of living subjects offers higher spatial resolution and allows deeper tissues to be imaged compared with most optical imaging techniques. As many diseases do not exhibit a natural photoacoustic contrast, especially in their early stages, it is necessary to administer a photoacoustic contrast agent. A number of contrast agents for photoacoustic imaging have been suggested previously, but most were not shown to target a diseased site in living subjects. Here we show that single-walled carbon nanotubes conjugated with cyclic Arg-Gly-Asp (RGD) peptides can be used as a contrast agent for photoacoustic imaging of tumours. Intravenous administration of these targeted nanotubes to mice bearing tumours showed eight times greater photoacoustic signal in the tumour than mice injected with non-targeted nanotubes. These results were verified ex vivo using Raman microscopy. Photoacoustic imaging of targeted single-walled carbon nanotubes may contribute to non-invasive cancer imaging and monitoring of nanotherapeutics in living subjects.
Collapse
Affiliation(s)
- ADAM DE LA ZERDA
- Molecular Imaging Program at Stanford, Department of Radiology and Bio-X Program, Stanford University, Palo Alto, California 94305, USA
- Department of Electrical Engineering, Stanford University, Palo Alto, California 94305, USA
| | - CRISTINA ZAVALETA
- Molecular Imaging Program at Stanford, Department of Radiology and Bio-X Program, Stanford University, Palo Alto, California 94305, USA
| | - SHAY KEREN
- Molecular Imaging Program at Stanford, Department of Radiology and Bio-X Program, Stanford University, Palo Alto, California 94305, USA
| | - SRIKANT VAITHILINGAM
- Department of Electrical Engineering, Stanford University, Palo Alto, California 94305, USA
| | - SUNIL BODAPATI
- Molecular Imaging Program at Stanford, Department of Radiology and Bio-X Program, Stanford University, Palo Alto, California 94305, USA
| | - ZHUANG LIU
- Department of Chemistry, Stanford University, Palo Alto, California 94305, USA
| | - JELENA LEVI
- Molecular Imaging Program at Stanford, Department of Radiology and Bio-X Program, Stanford University, Palo Alto, California 94305, USA
| | - BRYAN R. SMITH
- Molecular Imaging Program at Stanford, Department of Radiology and Bio-X Program, Stanford University, Palo Alto, California 94305, USA
| | - TE-JEN MA
- Department of Electrical Engineering, Stanford University, Palo Alto, California 94305, USA
| | - OMER ORALKAN
- Department of Electrical Engineering, Stanford University, Palo Alto, California 94305, USA
| | - ZHEN CHENG
- Molecular Imaging Program at Stanford, Department of Radiology and Bio-X Program, Stanford University, Palo Alto, California 94305, USA
| | - XIAOYUAN CHEN
- Molecular Imaging Program at Stanford, Department of Radiology and Bio-X Program, Stanford University, Palo Alto, California 94305, USA
| | - HONGJIE DAI
- Department of Chemistry, Stanford University, Palo Alto, California 94305, USA
| | - BUTRUS T. KHURI-YAKUB
- Department of Electrical Engineering, Stanford University, Palo Alto, California 94305, USA
| | - SANJIV S. GAMBHIR
- Molecular Imaging Program at Stanford, Department of Radiology and Bio-X Program, Stanford University, Palo Alto, California 94305, USA
- Department of Bioengineering, Stanford University, Palo Alto, California 94305, USA
- e-mail:
| |
Collapse
|