51
|
Guo H, He X, Liu M, Zhang Z, Hu Z, Tian J. Weight Multispectral Reconstruction Strategy for Enhanced Reconstruction Accuracy and Stability With Cerenkov Luminescence Tomography. IEEE TRANSACTIONS ON MEDICAL IMAGING 2017; 36:1337-1346. [PMID: 28182554 DOI: 10.1109/tmi.2017.2658661] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Cerenkov luminescence tomography (CLT) provides a novel technique for 3-D noninvasive detection of radiopharmaceuticals in living subjects. However, because of the severe scattering of Cerenkov light, the reconstruction accuracy and stability of CLT is still unsatisfied. In this paper, a modified weight multispectral CLT (wmCLT) reconstruction strategy was developed which split the Cerenkov radiation spectrum into several sub-spectral bands and weighted the sub-spectral results to obtain the final result. To better evaluate the property of the wmCLT reconstruction strategy in terms of accuracy, stability and practicability, several numerical simulation experiments and in vivo experiments were conducted and the results obtained were compared with the traditional multispectral CLT (mCLT) and hybrid-spectral CLT (hCLT) reconstruction strategies. The numerical simulation results indicated that wmCLT strategy significantly improved the accuracy of Cerenkov source localization and intensity quantitation and exhibited good stability in suppressing noise in numerical simulation experiments. And the comparison of the results achieved from different in vivo experiments further indicated significant improvement of the wmCLT strategy in terms of the shape recovery of the bladder and the spatial resolution of imaging xenograft tumors. Overall the strategy reported here will facilitate the development of nuclear and optical molecular tomography in theoretical study.
Collapse
|
52
|
Deán-Ben XL, Gottschalk S, Mc Larney B, Shoham S, Razansky D. Advanced optoacoustic methods for multiscale imaging of in vivo dynamics. Chem Soc Rev 2017; 46:2158-2198. [PMID: 28276544 PMCID: PMC5460636 DOI: 10.1039/c6cs00765a] [Citation(s) in RCA: 186] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Visualization of dynamic functional and molecular events in an unperturbed in vivo environment is essential for understanding the complex biology of living organisms and of disease state and progression. To this end, optoacoustic (photoacoustic) sensing and imaging have demonstrated the exclusive capacity to maintain excellent optical contrast and high resolution in deep-tissue observations, far beyond the penetration limits of modern microscopy. Yet, the time domain is paramount for the observation and study of complex biological interactions that may be invisible in single snapshots of living systems. This review focuses on the recent advances in optoacoustic imaging assisted by smart molecular labeling and dynamic contrast enhancement approaches that enable new types of multiscale dynamic observations not attainable with other bio-imaging modalities. A wealth of investigated new research topics and clinical applications is further discussed, including imaging of large-scale brain activity patterns, volumetric visualization of moving organs and contrast agent kinetics, molecular imaging using targeted and genetically expressed labels, as well as three-dimensional handheld diagnostics of human subjects.
Collapse
Affiliation(s)
- X L Deán-Ben
- Institute for Biological and Medical Imaging (IBMI), Helmholtz Center Munich, Ingolstädter Landstr. 1, 85764 Neuherberg, Germany.
| | - S Gottschalk
- Institute for Biological and Medical Imaging (IBMI), Helmholtz Center Munich, Ingolstädter Landstr. 1, 85764 Neuherberg, Germany.
| | - B Mc Larney
- Institute for Biological and Medical Imaging (IBMI), Helmholtz Center Munich, Ingolstädter Landstr. 1, 85764 Neuherberg, Germany. and Faculty of Medicine, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - S Shoham
- Department of Biomedical Engineering, Technion - Israel Institute of Technology, 32000 Haifa, Israel
| | - D Razansky
- Institute for Biological and Medical Imaging (IBMI), Helmholtz Center Munich, Ingolstädter Landstr. 1, 85764 Neuherberg, Germany. and Faculty of Medicine, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| |
Collapse
|
53
|
Kim WJ, Kim BS, Cho YD, Yoon WJ, Baek JH, Woo KM, Ryoo HM. Fibroin particle-supported cationic lipid layers for highly efficient intracellular protein delivery. Biomaterials 2017; 122:154-162. [DOI: 10.1016/j.biomaterials.2017.01.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Revised: 01/10/2017] [Accepted: 01/13/2017] [Indexed: 12/31/2022]
|
54
|
Basal LA, Yan Y, Shen Y, Haacke EM, Mehrmohammadi M, Allen MJ. Oxidation-Responsive, Eu II/III-Based, Multimodal Contrast Agent for Magnetic Resonance and Photoacoustic Imaging. ACS OMEGA 2017; 2:800-805. [PMID: 28393130 PMCID: PMC5377279 DOI: 10.1021/acsomega.6b00514] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 02/21/2017] [Indexed: 05/08/2023]
Abstract
We report, for the first time, a multimodal, oxidation-responsive contrast agent for magnetic resonance imaging and photoacoustic imaging that uses the differences in the properties between Eu in the +2 and +3 oxidation states. The enhancement of contrast in T1-weighted magnetic resonance and photoacoustic imaging was observed in the +2 but not in the +3 oxidation state, and the complex is a known chemical exchange saturation transfer agent for magnetic resonance imaging in the +3 oxidation state.
Collapse
Affiliation(s)
- Lina A. Basal
- Department
of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, Michigan 48202, United States
| | - Yan Yan
- Department
of Biomedical Engineering, Wayne State University, 818 W. Hancock, Detroit, Michigan 48201, United States
| | - Yimin Shen
- Department
of Radiology, Wayne State University, Detroit, Michigan 48201, United States
| | - E. Mark Haacke
- Department
of Radiology, Wayne State University, Detroit, Michigan 48201, United States
- Barbara
Ann Karmanos Cancer Institute, Detroit, Michigan 48201, United States
| | - Mohammad Mehrmohammadi
- Department
of Biomedical Engineering, Wayne State University, 818 W. Hancock, Detroit, Michigan 48201, United States
- Barbara
Ann Karmanos Cancer Institute, Detroit, Michigan 48201, United States
| | - Matthew J. Allen
- Department
of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, Michigan 48202, United States
- Barbara
Ann Karmanos Cancer Institute, Detroit, Michigan 48201, United States
| |
Collapse
|
55
|
Pelaz B, Alexiou C, Alvarez-Puebla RA, Alves F, Andrews AM, Ashraf S, Balogh LP, Ballerini L, Bestetti A, Brendel C, Bosi S, Carril M, Chan WCW, Chen C, Chen X, Chen X, Cheng Z, Cui D, Du J, Dullin C, Escudero A, Feliu N, Gao M, George M, Gogotsi Y, Grünweller A, Gu Z, Halas NJ, Hampp N, Hartmann RK, Hersam MC, Hunziker P, Jian J, Jiang X, Jungebluth P, Kadhiresan P, Kataoka K, Khademhosseini A, Kopeček J, Kotov NA, Krug HF, Lee DS, Lehr CM, Leong KW, Liang XJ, Ling Lim M, Liz-Marzán LM, Ma X, Macchiarini P, Meng H, Möhwald H, Mulvaney P, Nel AE, Nie S, Nordlander P, Okano T, Oliveira J, Park TH, Penner RM, Prato M, Puntes V, Rotello VM, Samarakoon A, Schaak RE, Shen Y, Sjöqvist S, Skirtach AG, Soliman MG, Stevens MM, Sung HW, Tang BZ, Tietze R, Udugama BN, VanEpps JS, Weil T, Weiss PS, Willner I, Wu Y, Yang L, Yue Z, Zhang Q, Zhang Q, Zhang XE, Zhao Y, Zhou X, Parak WJ. Diverse Applications of Nanomedicine. ACS NANO 2017; 11:2313-2381. [PMID: 28290206 PMCID: PMC5371978 DOI: 10.1021/acsnano.6b06040] [Citation(s) in RCA: 784] [Impact Index Per Article: 112.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Indexed: 04/14/2023]
Abstract
The design and use of materials in the nanoscale size range for addressing medical and health-related issues continues to receive increasing interest. Research in nanomedicine spans a multitude of areas, including drug delivery, vaccine development, antibacterial, diagnosis and imaging tools, wearable devices, implants, high-throughput screening platforms, etc. using biological, nonbiological, biomimetic, or hybrid materials. Many of these developments are starting to be translated into viable clinical products. Here, we provide an overview of recent developments in nanomedicine and highlight the current challenges and upcoming opportunities for the field and translation to the clinic.
Collapse
Affiliation(s)
- Beatriz Pelaz
- Fachbereich Physik, Fachbereich Medizin, Fachbereich Pharmazie, and Department of Chemistry, Philipps Universität Marburg, 35037 Marburg, Germany
| | - Christoph Alexiou
- ENT-Department, Section of Experimental Oncology & Nanomedicine
(SEON), Else Kröner-Fresenius-Stiftung-Professorship for Nanomedicine, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Ramon A. Alvarez-Puebla
- Department of Physical Chemistry, Universitat Rovira I Virgili, 43007 Tarragona, Spain
- ICREA, Pg. Lluís Companys 23, 08010 Barcelona, Spain
| | - Frauke Alves
- Department of Haematology and Medical Oncology, Department of Diagnostic
and Interventional Radiology, University
Medical Center Göttingen, 37075 Göttingen Germany
- Department of Molecular Biology of Neuronal Signals, Max-Planck-Institute for Experimental Medicine, 37075 Göttingen, Germany
| | - Anne M. Andrews
- California NanoSystems Institute, Department of Chemistry
and Biochemistry and Department of Psychiatry and Semel Institute
for Neuroscience and Human Behavior, Division of NanoMedicine and Center
for the Environmental Impact of Nanotechnology, and Department of Materials Science
and Engineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Sumaira Ashraf
- Fachbereich Physik, Fachbereich Medizin, Fachbereich Pharmazie, and Department of Chemistry, Philipps Universität Marburg, 35037 Marburg, Germany
| | - Lajos P. Balogh
- AA Nanomedicine & Nanotechnology Consultants, North Andover, Massachusetts 01845, United States
| | - Laura Ballerini
- International School for Advanced Studies (SISSA/ISAS), 34136 Trieste, Italy
| | - Alessandra Bestetti
- School of Chemistry & Bio21 Institute, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Cornelia Brendel
- Fachbereich Physik, Fachbereich Medizin, Fachbereich Pharmazie, and Department of Chemistry, Philipps Universität Marburg, 35037 Marburg, Germany
| | - Susanna Bosi
- Department of Chemical
and Pharmaceutical Sciences, University
of Trieste, 34127 Trieste, Italy
| | - Monica Carril
- CIC biomaGUNE, Paseo de Miramón 182, 20014, Donostia - San Sebastián, Spain
- Ikerbasque, Basque Foundation
for Science, 48013 Bilbao, Spain
| | - Warren C. W. Chan
- Institute of Biomaterials
and Biomedical Engineering, University of
Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Chunying Chen
- CAS Center for Excellence in Nanoscience and CAS Key
Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of
China, Beijing 100190, China
| | - Xiaodong Chen
- School of Materials
Science and Engineering, Nanyang Technological
University, Singapore 639798
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine,
National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Zhen Cheng
- Molecular
Imaging Program at Stanford and Bio-X Program, Canary Center at Stanford
for Cancer Early Detection, Stanford University, Stanford, California 94305, United States
| | - Daxiang Cui
- Institute of Nano Biomedicine and Engineering, Department of Instrument
Science and Engineering, School of Electronic Information and Electronical
Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, 200240 Shanghai, China
| | - Jianzhong Du
- Department of Polymeric Materials, School of Materials
Science and Engineering, Tongji University, Shanghai, China
| | - Christian Dullin
- Department of Haematology and Medical Oncology, Department of Diagnostic
and Interventional Radiology, University
Medical Center Göttingen, 37075 Göttingen Germany
| | - Alberto Escudero
- Fachbereich Physik, Fachbereich Medizin, Fachbereich Pharmazie, and Department of Chemistry, Philipps Universität Marburg, 35037 Marburg, Germany
- Instituto
de Ciencia de Materiales de Sevilla. CSIC, Universidad de Sevilla, 41092 Seville, Spain
| | - Neus Feliu
- Department of Clinical Science, Intervention, and Technology (CLINTEC), Karolinska Institutet, 141 86 Stockholm, Sweden
| | - Mingyuan Gao
- Institute of Chemistry, Chinese
Academy of Sciences, 100190 Beijing, China
| | | | - Yury Gogotsi
- Department of Materials Science and Engineering and A.J. Drexel Nanomaterials
Institute, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Arnold Grünweller
- Fachbereich Physik, Fachbereich Medizin, Fachbereich Pharmazie, and Department of Chemistry, Philipps Universität Marburg, 35037 Marburg, Germany
| | - Zhongwei Gu
- College of Polymer Science and Engineering, Sichuan University, 610000 Chengdu, China
| | - Naomi J. Halas
- Departments of Physics and Astronomy, Rice
University, Houston, Texas 77005, United
States
| | - Norbert Hampp
- Fachbereich Physik, Fachbereich Medizin, Fachbereich Pharmazie, and Department of Chemistry, Philipps Universität Marburg, 35037 Marburg, Germany
| | - Roland K. Hartmann
- Fachbereich Physik, Fachbereich Medizin, Fachbereich Pharmazie, and Department of Chemistry, Philipps Universität Marburg, 35037 Marburg, Germany
| | - Mark C. Hersam
- Departments of Materials Science and Engineering, Chemistry,
and Medicine, Northwestern University, Evanston, Illinois 60208, United States
| | - Patrick Hunziker
- University Hospital, 4056 Basel, Switzerland
- CLINAM,
European Foundation for Clinical Nanomedicine, 4058 Basel, Switzerland
| | - Ji Jian
- Department of Polymer Science and Engineering and Center for
Bionanoengineering and Department of Chemical and Biological Engineering, Zhejiang University, 310027 Hangzhou, China
| | - Xingyu Jiang
- CAS Center for Excellence in Nanoscience and CAS Key
Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of
China, Beijing 100190, China
| | - Philipp Jungebluth
- Thoraxklinik Heidelberg, Universitätsklinikum
Heidelberg, 69120 Heidelberg, Germany
| | - Pranav Kadhiresan
- Institute of Biomaterials
and Biomedical Engineering, University of
Toronto, Toronto, Ontario M5S 3G9, Canada
| | | | | | - Jindřich Kopeček
- Biomedical Polymers Laboratory, University of Utah, Salt Lake City, Utah 84112, United States
| | - Nicholas A. Kotov
- Emergency Medicine, University of Michigan, Ann Arbor, Michigan 48019, United States
| | - Harald F. Krug
- EMPA, Federal Institute for Materials
Science and Technology, CH-9014 St. Gallen, Switzerland
| | - Dong Soo Lee
- Department of Molecular Medicine and Biopharmaceutical
Sciences and School of Chemical and Biological Engineering, Seoul National University, Seoul, South Korea
| | - Claus-Michael Lehr
- Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
- HIPS - Helmhotz Institute for Pharmaceutical Research Saarland, Helmholtz-Center for Infection Research, 66123 Saarbrücken, Germany
| | - Kam W. Leong
- Department of Biomedical Engineering, Columbia University, New York City, New York 10027, United States
| | - Xing-Jie Liang
- CAS Center for Excellence in Nanoscience and CAS Key
Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of
China, Beijing 100190, China
- Laboratory of Controllable Nanopharmaceuticals, Chinese Academy of Sciences (CAS), 100190 Beijing, China
| | - Mei Ling Lim
- Department of Clinical Science, Intervention, and Technology (CLINTEC), Karolinska Institutet, 141 86 Stockholm, Sweden
| | - Luis M. Liz-Marzán
- CIC biomaGUNE, Paseo de Miramón 182, 20014, Donostia - San Sebastián, Spain
- Ikerbasque, Basque Foundation
for Science, 48013 Bilbao, Spain
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine, Ciber-BBN, 20014 Donostia - San Sebastián, Spain
| | - Xiaowei Ma
- Laboratory of Controllable Nanopharmaceuticals, Chinese Academy of Sciences (CAS), 100190 Beijing, China
| | - Paolo Macchiarini
- Laboratory of Bioengineering Regenerative Medicine (BioReM), Kazan Federal University, 420008 Kazan, Russia
| | - Huan Meng
- California NanoSystems Institute, Department of Chemistry
and Biochemistry and Department of Psychiatry and Semel Institute
for Neuroscience and Human Behavior, Division of NanoMedicine and Center
for the Environmental Impact of Nanotechnology, and Department of Materials Science
and Engineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Helmuth Möhwald
- Department of Interfaces, Max-Planck
Institute of Colloids and Interfaces, 14476 Potsdam, Germany
| | - Paul Mulvaney
- School of Chemistry & Bio21 Institute, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Andre E. Nel
- California NanoSystems Institute, Department of Chemistry
and Biochemistry and Department of Psychiatry and Semel Institute
for Neuroscience and Human Behavior, Division of NanoMedicine and Center
for the Environmental Impact of Nanotechnology, and Department of Materials Science
and Engineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Shuming Nie
- Emory University, Atlanta, Georgia 30322, United States
| | - Peter Nordlander
- Departments of Physics and Astronomy, Rice
University, Houston, Texas 77005, United
States
| | - Teruo Okano
- Tokyo Women’s Medical University, Tokyo 162-8666, Japan
| | | | - Tai Hyun Park
- Department of Molecular Medicine and Biopharmaceutical
Sciences and School of Chemical and Biological Engineering, Seoul National University, Seoul, South Korea
- Advanced Institutes of Convergence Technology, Suwon, South Korea
| | - Reginald M. Penner
- Department of Chemistry, University of
California, Irvine, California 92697, United States
| | - Maurizio Prato
- Department of Chemical
and Pharmaceutical Sciences, University
of Trieste, 34127 Trieste, Italy
- CIC biomaGUNE, Paseo de Miramón 182, 20014, Donostia - San Sebastián, Spain
- Ikerbasque, Basque Foundation
for Science, 48013 Bilbao, Spain
| | - Victor Puntes
- ICREA, Pg. Lluís Companys 23, 08010 Barcelona, Spain
- Institut Català de Nanotecnologia, UAB, 08193 Barcelona, Spain
- Vall d’Hebron University Hospital
Institute of Research, 08035 Barcelona, Spain
| | - Vincent M. Rotello
- Department
of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Amila Samarakoon
- Institute of Biomaterials
and Biomedical Engineering, University of
Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Raymond E. Schaak
- Department of Chemistry, The
Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Youqing Shen
- Department of Polymer Science and Engineering and Center for
Bionanoengineering and Department of Chemical and Biological Engineering, Zhejiang University, 310027 Hangzhou, China
| | - Sebastian Sjöqvist
- Department of Clinical Science, Intervention, and Technology (CLINTEC), Karolinska Institutet, 141 86 Stockholm, Sweden
| | - Andre G. Skirtach
- Department of Interfaces, Max-Planck
Institute of Colloids and Interfaces, 14476 Potsdam, Germany
- Department of Molecular Biotechnology, University of Ghent, B-9000 Ghent, Belgium
| | - Mahmoud G. Soliman
- Fachbereich Physik, Fachbereich Medizin, Fachbereich Pharmazie, and Department of Chemistry, Philipps Universität Marburg, 35037 Marburg, Germany
| | - Molly M. Stevens
- Department of Materials,
Department of Bioengineering, Institute for Biomedical Engineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Hsing-Wen Sung
- Department of Chemical Engineering and Institute of Biomedical
Engineering, National Tsing Hua University, Hsinchu City, Taiwan,
ROC 300
| | - Ben Zhong Tang
- Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Hong Kong, China
| | - Rainer Tietze
- ENT-Department, Section of Experimental Oncology & Nanomedicine
(SEON), Else Kröner-Fresenius-Stiftung-Professorship for Nanomedicine, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Buddhisha N. Udugama
- Institute of Biomaterials
and Biomedical Engineering, University of
Toronto, Toronto, Ontario M5S 3G9, Canada
| | - J. Scott VanEpps
- Emergency Medicine, University of Michigan, Ann Arbor, Michigan 48019, United States
| | - Tanja Weil
- Institut für
Organische Chemie, Universität Ulm, 89081 Ulm, Germany
- Max-Planck-Institute for Polymer Research, 55128 Mainz, Germany
| | - Paul S. Weiss
- California NanoSystems Institute, Department of Chemistry
and Biochemistry and Department of Psychiatry and Semel Institute
for Neuroscience and Human Behavior, Division of NanoMedicine and Center
for the Environmental Impact of Nanotechnology, and Department of Materials Science
and Engineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Itamar Willner
- Institute of Chemistry, The Center for
Nanoscience and Nanotechnology, The Hebrew
University of Jerusalem, Jerusalem 91904, Israel
| | - Yuzhou Wu
- Max-Planck-Institute for Polymer Research, 55128 Mainz, Germany
- School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, 430074 Wuhan, China
| | | | - Zhao Yue
- Fachbereich Physik, Fachbereich Medizin, Fachbereich Pharmazie, and Department of Chemistry, Philipps Universität Marburg, 35037 Marburg, Germany
| | - Qian Zhang
- Fachbereich Physik, Fachbereich Medizin, Fachbereich Pharmazie, and Department of Chemistry, Philipps Universität Marburg, 35037 Marburg, Germany
| | - Qiang Zhang
- School of Pharmaceutical Science, Peking University, 100191 Beijing, China
| | - Xian-En Zhang
- National Laboratory of Biomacromolecules,
CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing, 100101, China
| | - Yuliang Zhao
- CAS Center for Excellence in Nanoscience and CAS Key
Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of
China, Beijing 100190, China
| | - Xin Zhou
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China
| | - Wolfgang J. Parak
- Fachbereich Physik, Fachbereich Medizin, Fachbereich Pharmazie, and Department of Chemistry, Philipps Universität Marburg, 35037 Marburg, Germany
- CIC biomaGUNE, Paseo de Miramón 182, 20014, Donostia - San Sebastián, Spain
| |
Collapse
|
56
|
Longo DL, Stefania R, Callari C, De Rose F, Rolle R, Conti L, Consolino L, Arena F, Aime S. Water Soluble Melanin Derivatives for Dynamic Contrast Enhanced Photoacoustic Imaging of Tumor Vasculature and Response to Antiangiogenic Therapy. Adv Healthc Mater 2017; 6. [PMID: 27782375 DOI: 10.1002/adhm.201600550] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 08/22/2016] [Indexed: 12/17/2022]
Abstract
A dynamic contrast enhanced (DCE) approach for tumor photoacoustic (PA) imaging is described. Novel water soluble melanin-based derivatives are synthesized that exhibit good PA properties, stability, safety and accumulation in tumor bearing mice. This melanin derivative is capable to characterize tumor vasculature and to monitor vessel permeability changes upon antiangiogenic treatment. DCE-PA imaging can assess functional response to cancer treatments.
Collapse
Affiliation(s)
- Dario L. Longo
- Institute of Biostructure and Bioimaging (CNR) c/o Molecular Biotechnology Center; Via Nizza 52 10126 Torino Italy
| | - Rachele Stefania
- Department of Molecular Biotechnology and Health Sciences; University of Torino; Via Nizza 52 10126 Torino Italy
| | - Chiara Callari
- Department of Molecular Biotechnology and Health Sciences; University of Torino; Via Nizza 52 10126 Torino Italy
| | - Francesco De Rose
- Department of Molecular Biotechnology and Health Sciences; University of Torino; Via Nizza 52 10126 Torino Italy
| | - Riccardo Rolle
- Department of Molecular Biotechnology and Health Sciences; University of Torino; Via Nizza 52 10126 Torino Italy
| | - Laura Conti
- Department of Molecular Biotechnology and Health Sciences; University of Torino; Via Nizza 52 10126 Torino Italy
| | - Lorena Consolino
- Department of Molecular Biotechnology and Health Sciences; University of Torino; Via Nizza 52 10126 Torino Italy
| | - Francesca Arena
- Department of Molecular Biotechnology and Health Sciences; University of Torino; Via Nizza 52 10126 Torino Italy
| | - Silvio Aime
- Department of Molecular Biotechnology and Health Sciences; University of Torino; Via Nizza 52 10126 Torino Italy
| |
Collapse
|
57
|
Liu C, Gong X, Lin R, Liu F, Chen J, Wang Z, Song L, Chu J. Advances in Imaging Techniques and Genetically Encoded Probes for Photoacoustic Imaging. Am J Cancer Res 2016; 6:2414-2430. [PMID: 27877244 PMCID: PMC5118604 DOI: 10.7150/thno.15878] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 05/31/2016] [Indexed: 11/05/2022] Open
Abstract
Photoacoustic (PA) imaging is a rapidly emerging biomedical imaging modality that is capable of visualizing cellular and molecular functions with high detection sensitivity and spatial resolution in deep tissue. Great efforts and progress have been made on the development of various PA imaging technologies with improved resolution and sensitivity over the past two decades. Various PA probes with high contrast have also been extensively developed, with many important biomedical applications. In comparison with chemical dyes and nanoparticles, genetically encoded probes offer easier labeling of defined cells within tissues or proteins of interest within a cell, have higher stability in vivo, and eliminate the need for delivery of exogenous substances. Genetically encoded probes have thus attracted increasing attention from researchers in engineering and biomedicine. In this review, we aim to provide an overview of the existing PA imaging technologies and genetically encoded PA probes, and describe further improvements in PA imaging techniques and the near-infrared photochromic protein BphP1, the most sensitive genetically encoded probe thus far, as well as the potential biomedical applications of BphP1-based PA imaging in vivo.
Collapse
|
58
|
Progress of Multimodal Molecular Imaging Technology in Diagnosis of Tumor. CHINESE JOURNAL OF ANALYTICAL CHEMISTRY 2016. [DOI: 10.1016/s1872-2040(16)60966-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
59
|
Cho IK, Wang S, Mao H, Chan AWS. Genetic engineered molecular imaging probes for applications in cell therapy: emphasis on MRI approach. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2016; 6:234-261. [PMID: 27766183 PMCID: PMC5069277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 08/31/2016] [Indexed: 06/06/2023]
Abstract
Recent advances in stem cell-based regenerative medicine, cell replacement therapy, and genome editing technologies (i.e. CRISPR-Cas 9) have sparked great interest in in vivo cell monitoring. Molecular imaging promises a unique approach to noninvasively monitor cellular and molecular phenomena, including cell survival, migration, proliferation, and even differentiation at the whole organismal level. Several imaging modalities and strategies have been explored for monitoring cell grafts in vivo. We begin this review with an introduction describing the progress in stem cell technology, with a perspective toward cell replacement therapy. The importance of molecular imaging in reporting and assessing the status of cell grafts and their relation to the local microenvironment is highlighted since the current knowledge gap is one of the major obstacles in clinical translation of stem cell therapy. Based on currently available imaging techniques, we provide a brief discussion on the pros and cons of each imaging modality used for monitoring cell grafts with particular emphasis on magnetic resonance imaging (MRI) and the reporter gene approach. Finally, we conclude with a comprehensive discussion of future directions of applying molecular imaging in regenerative medicine to emphasize further the importance of correlating cell graft conditions and clinical outcomes to advance regenerative medicine.
Collapse
Affiliation(s)
- In K Cho
- Department of Human Genetics, Emory University School of MedicineAtlanta, GA, USA
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research CenterAtlanta, GA, USA
| | - Silun Wang
- Department of Radiology and Imaging Sciences, Emory University School of MedicineAtlanta, GA, USA
| | - Hui Mao
- Department of Radiology and Imaging Sciences, Emory University School of MedicineAtlanta, GA, USA
| | - Anthony WS Chan
- Department of Human Genetics, Emory University School of MedicineAtlanta, GA, USA
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research CenterAtlanta, GA, USA
| |
Collapse
|
60
|
Sun Y, Hong S, Ma X, Cheng K, Wang J, Zhang Z, Yang M, Jiang Y, Hong X, Cheng Z. Recyclable Cu(i)/melanin dots for cycloaddition, bioconjugation and cell labelling. Chem Sci 2016; 7:5888-5892. [PMID: 30034730 PMCID: PMC6024301 DOI: 10.1039/c6sc01536k] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 05/18/2016] [Indexed: 12/13/2022] Open
Abstract
Development of biocompatible and high-performance heterogeneous catalysts for bioconjugation and cell labeling is highly challenging. Melanin has previously been used as a target for melanoma imaging and therapy. Herein, this important biomarker was transferred into a novel catalytic platform. A biocompatible Cu(i)/melanin dot-based catalyst [Cu(i)/M-dots] was easily prepared and exhibited high catalytic activity and excellent reusability in various Cu(i)-catalyzed azide-alkyne cycloadditions (CuAAC). Furthermore, DNA bioconjugation was carried out efficiently using Cu(i)/M-dots under ligand-free and reductant-free conditions, and the Cu(i)/M-dots could easily be removed by centrifugation. Lastly, the integrin receptor (alkyne RGD targeted) of U87MG cells was effectively labelled with a fluorescent dye (Cyanine5.5 azide) in combination with Cu(i)/M-dots. These attractive properties of Cu(i)/M-dots render it a promising catalytic platform in bioconjugation and chemical biology applications.
Collapse
Affiliation(s)
- Yao Sun
- Molecular Imaging Program at Stanford (MIPS) , Bio-X Program , Department of Radiology , Canary Center at Stanford for Cancer Early Detection , Stanford University , California 94305-5344 , USA .
- State Key Laboratory of Virology , Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) , Hubei Provincial Key Laboratory of Developmentally Originated Disease , Wuhan University School of Pharmaceutical Sciences , Wuhan 430071 , China .
| | - Suhyun Hong
- Molecular Imaging Program at Stanford (MIPS) , Bio-X Program , Department of Radiology , Canary Center at Stanford for Cancer Early Detection , Stanford University , California 94305-5344 , USA .
| | - Xiaowei Ma
- Molecular Imaging Program at Stanford (MIPS) , Bio-X Program , Department of Radiology , Canary Center at Stanford for Cancer Early Detection , Stanford University , California 94305-5344 , USA .
| | - Kai Cheng
- Molecular Imaging Program at Stanford (MIPS) , Bio-X Program , Department of Radiology , Canary Center at Stanford for Cancer Early Detection , Stanford University , California 94305-5344 , USA .
| | - Jing Wang
- Molecular Imaging Program at Stanford (MIPS) , Bio-X Program , Department of Radiology , Canary Center at Stanford for Cancer Early Detection , Stanford University , California 94305-5344 , USA .
| | - Zhe Zhang
- Molecular Imaging Program at Stanford (MIPS) , Bio-X Program , Department of Radiology , Canary Center at Stanford for Cancer Early Detection , Stanford University , California 94305-5344 , USA .
| | - Meng Yang
- Chinese Academy of Medical Science , Peking Union Medical College Hospital , Department of Ultrasound , Beijing , 100730 , China
| | - Yuxin Jiang
- Chinese Academy of Medical Science , Peking Union Medical College Hospital , Department of Ultrasound , Beijing , 100730 , China
| | - Xuechuan Hong
- State Key Laboratory of Virology , Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) , Hubei Provincial Key Laboratory of Developmentally Originated Disease , Wuhan University School of Pharmaceutical Sciences , Wuhan 430071 , China .
| | - Zhen Cheng
- Molecular Imaging Program at Stanford (MIPS) , Bio-X Program , Department of Radiology , Canary Center at Stanford for Cancer Early Detection , Stanford University , California 94305-5344 , USA .
| |
Collapse
|
61
|
Wang D, Zhou J, Chen R, Shi R, Zhao G, Xia G, Li R, Liu Z, Tian J, Wang H, Guo Z, Wang H, Chen Q. Controllable synthesis of dual-MOFs nanostructures for pH-responsive artemisinin delivery, magnetic resonance and optical dual-model imaging-guided chemo/photothermal combinational cancer therapy. Biomaterials 2016; 100:27-40. [DOI: 10.1016/j.biomaterials.2016.05.027] [Citation(s) in RCA: 182] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Revised: 04/21/2016] [Accepted: 05/17/2016] [Indexed: 01/08/2023]
|
62
|
Weber J, Beard PC, Bohndiek SE. Contrast agents for molecular photoacoustic imaging. Nat Methods 2016; 13:639-50. [PMID: 27467727 DOI: 10.1038/nmeth.3929] [Citation(s) in RCA: 799] [Impact Index Per Article: 99.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 06/25/2016] [Indexed: 02/07/2023]
Abstract
Photoacoustic imaging (PAI) is an emerging tool that bridges the traditional depth limits of ballistic optical imaging and the resolution limits of diffuse optical imaging. Using the acoustic waves generated in response to the absorption of pulsed laser light, it provides noninvasive images of absorbed optical energy density at depths of several centimeters with a resolution of ∼100 μm. This versatile and scalable imaging modality has now shown potential for molecular imaging, which enables visualization of biological processes with systemically introduced contrast agents. Understanding the relative merits of the vast range of contrast agents available, from small-molecule dyes to gold and carbon nanostructures to liposome encapsulations, is a considerable challenge. Here we critically review the physical, chemical and biochemical characteristics of the existing photoacoustic contrast agents, highlighting key applications and present challenges for molecular PAI.
Collapse
Affiliation(s)
- Judith Weber
- Department of Physics, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Paul C Beard
- Department of Medical Physics and Biomedical Engineering, University College London, London, UK
| | - Sarah E Bohndiek
- Department of Physics, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| |
Collapse
|
63
|
Huang L, Cai W, Zhao Y, Wu D, Wang L, Wang Y, Lai D, Rong J, Gao F, Jiang H. In vivo tumor detection with combined MR–Photoacoustic-Thermoacoustic imaging. JOURNAL OF INNOVATIVE OPTICAL HEALTH SCIENCES 2016. [DOI: 10.1142/s1793545816500152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Here, we report a new method using combined magnetic resonance (MR)–Photoacoustic (PA)–Thermoacoustic (TA) imaging techniques, and demonstrate its unique ability for in vivo cancer detection using tumor-bearing mice. Circular scanning TA and PA imaging systems were used to recover the dielectric and optical property distributions of three colon carcinoma bearing mice While a 7.0-T magnetic resonance imaging (MRI) unit with a mouse body volume coil was utilized for high resolution structural imaging of the same mice. Three plastic tubes filled with soybean sauce were used as fiducial markers for the co-registration of MR, PA and TA images. The resulting fused images provided both enhanced tumor margin and contrast relative to the surrounding normal tissues. In particular, some finger-like protrusions extending into the surrounding tissues were revealed in the MR/TA infused images. These results show that the tissue functional optical and dielectric properties provided by PA and TA images along with the anatomical structure by MRI in one picture make accurate tumor identification easier. This combined MR–PA–TA-imaging strategy has the potential to offer a clinically useful triple-modality tool for accurate cancer detection and for intraoperative surgical navigation.
Collapse
Affiliation(s)
- Lin Huang
- School of Physical Electronics, University of Electronic Science and Technology of China, Chengdu 610054, P. R. China
| | - Wei Cai
- Department of Biomedical Engineering, University of Florida, FL 32611, USA
| | - Yuan Zhao
- School of Physical Electronics, University of Electronic Science and Technology of China, Chengdu 610054, P. R. China
| | - Dan Wu
- School of Physical Electronics, University of Electronic Science and Technology of China, Chengdu 610054, P. R. China
| | - Lei Wang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Yuqing Wang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Dakun Lai
- School of Physical Electronics, University of Electronic Science and Technology of China, Chengdu 610054, P. R. China
| | - Jian Rong
- School of Physical Electronics, University of Electronic Science and Technology of China, Chengdu 610054, P. R. China
| | - Fabao Gao
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Huabei Jiang
- School of Physical Electronics, University of Electronic Science and Technology of China, Chengdu 610054, P. R. China
- Department of Biomedical Engineering, University of Florida, FL 32611, USA
| |
Collapse
|
64
|
Cui L, Rao J. Semiconducting polymer nanoparticles as photoacoustic molecular imaging probes. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2016; 9. [PMID: 27346564 DOI: 10.1002/wnan.1418] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 05/18/2016] [Accepted: 06/02/2016] [Indexed: 01/28/2023]
Abstract
As an emerging class of optical nanomaterials, semiconducting polymer nanoparticles (SPNs) are highly photostable, optically active and versatile in chemistry; these properties make them attractive as molecular imaging agents to enable imaging of biological events and functionalities at multiple scales. More recently, a variety of SPNs have been found to exhibit high photoacoustic properties, and further empowered photoacoustic imaging for contrast enhanced in vivo molecular imaging. Target-sensitive components can be incorporated in the SPNs to create activatable imaging probes to sense and monitor the target dynamics in living objects. Intrinsically biophotonic and biocompatible, SPNs can be further engineered for multimodal imaging and for real-time imaging of drug delivery. WIREs Nanomed Nanobiotechnol 2017, 9:e1418. doi: 10.1002/wnan.1418 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Liyang Cui
- Molecular Imaging Program at Stanford, Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Jianghong Rao
- Molecular Imaging Program at Stanford, Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA
| |
Collapse
|
65
|
Zhang C, Kimura R, Abou-Elkacem L, Levi J, Xu L, Gambhir SS. A Cystine Knot Peptide Targeting Integrin αvβ6 for Photoacoustic and Fluorescence Imaging of Tumors in Living Subjects. J Nucl Med 2016; 57:1629-1634. [PMID: 27230926 DOI: 10.2967/jnumed.115.169383] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 04/14/2016] [Indexed: 11/16/2022] Open
Abstract
Photoacoustic imaging is a nonionizing biomedical imaging modality with higher resolution and imaging depth than fluorescence imaging, which has greater sensitivity. The combination of the 2 imaging modalities could improve the detection of cancer. Integrin αvβ6 is a cell surface marker overexpressed in many different cancers. Here, we report the development and evaluation of a dye-labeled cystine knot peptide, which selectively recognizes integrin αvβ6 with high affinity, for photoacoustic and fluorescence imaging. The new dual-modality probe may find clinical application in cancer diagnosis and intraoperative imaging of integrin αvβ6-positive tumors. METHODS An engineered cystine knot peptide, R01, that recognizes integrin αvβ6 was labeled with Atto 740 (A740) and evaluated for its specific cell uptake and its sensitivity threshold. A740-R01 was injected via the tail vein into nude mice xenografted with A431 (integrin αvβ6-positive) or 293T (integrin αvβ6-negative) tumors. Photoacoustic and fluorescence scans of tumors were acquired before and at 0.5, 1, 2, and 4 h after injection of A740-R01. Dynamic photoacoustic scans of various normal organs were also acquired. Ex vivo fluorescence imaging of tissues was performed 1 h after injection. RESULTS The A740-R01 demonstrated integrin αvβ6-dependent binding to A431 cells in culture. Sensitivity studies indicated that the probe may potentially detect lesions as small as 1 or 6 mm3 by fluorescence or photoacoustic imaging, respectively. The photoacoustic and fluorescence signals of A431 xenografts at 1 h after injection were 1.87 ± 0.25 arbitrary units (AU) and 8.27 ± 0.87 AU, respectively. Target specificity was confirmed by low tumor uptake in 293T tumors at 1 h after injection (1.07 ± 0.15 AU and 1.10 ± 0.14 AU for photoacoustic and fluorescence signals, respectively). A740-R01 exhibited hepatobiliary clearance marked by high uptake in the liver, spleen, and intestine but low uptake in the kidneys. CONCLUSION A740-R01 specifically targeted integrin αvβ6 with low nanomolar affinity. A740-R01 was able to detect integrin αvβ6 both in vitro and in vivo by photoacoustic and fluorescence imaging. A740-R01 is able to detect αvβ6-positive tumors in living subjects and may have clinical application in cancer diagnosis and real-time image-guided surgery.
Collapse
Affiliation(s)
- Chao Zhang
- Department of Radiology, Molecular Imaging Program at Stanford, Canary Center for Cancer Early Detection, Stanford University, Stanford, California; and Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Richard Kimura
- Department of Radiology, Molecular Imaging Program at Stanford, Canary Center for Cancer Early Detection, Stanford University, Stanford, California; and
| | - Lotfi Abou-Elkacem
- Department of Radiology, Molecular Imaging Program at Stanford, Canary Center for Cancer Early Detection, Stanford University, Stanford, California; and
| | - Jelena Levi
- Department of Radiology, Molecular Imaging Program at Stanford, Canary Center for Cancer Early Detection, Stanford University, Stanford, California; and
| | - Lingyun Xu
- Department of Radiology, Molecular Imaging Program at Stanford, Canary Center for Cancer Early Detection, Stanford University, Stanford, California; and
| | - Sanjiv Sam Gambhir
- Department of Radiology, Molecular Imaging Program at Stanford, Canary Center for Cancer Early Detection, Stanford University, Stanford, California; and
| |
Collapse
|
66
|
MRI Reporter Genes for Noninvasive Molecular Imaging. Molecules 2016; 21:molecules21050580. [PMID: 27213309 PMCID: PMC6273230 DOI: 10.3390/molecules21050580] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 04/21/2016] [Accepted: 04/25/2016] [Indexed: 01/17/2023] Open
Abstract
Magnetic resonance imaging (MRI) is one of the most important imaging technologies used in clinical diagnosis. Reporter genes for MRI can be applied to accurately track the delivery of cell in cell therapy, evaluate the therapy effect of gene delivery, and monitor tissue/cell-specific microenvironments. Commonly used reporter genes for MRI usually include genes encoding the enzyme (e.g., tyrosinase and β-galactosidase), the receptor on the cells (e.g., transferrin receptor), and endogenous reporter genes (e.g., ferritin reporter gene). However, low sensitivity limits the application of MRI and reporter gene-based multimodal imaging strategies are common including optical imaging and radionuclide imaging. These can significantly improve diagnostic efficiency and accelerate the development of new therapies.
Collapse
|
67
|
Patrick PS, Rodrigues TB, Kettunen MI, Lyons SK, Neves AA, Brindle KM. Development of Timd2 as a reporter gene for MRI. Magn Reson Med 2016; 75:1697-707. [PMID: 25981669 PMCID: PMC4832381 DOI: 10.1002/mrm.25750] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 03/27/2015] [Accepted: 03/27/2015] [Indexed: 12/20/2022]
Abstract
PURPOSE To assess the potential of an MRI gene reporter based on the ferritin receptor Timd2 (T-cell immunoglobulin and mucin domain containing protein 2), using T1- and T2-weighted imaging. METHODS Pellets of cells that had been modified to express the Timd2 transgene, and incubated with either iron-loaded or manganese-loaded ferritin, were imaged using T1- and T2-weighted MRI. Mice were also implanted subcutaneously with Timd2-expressing cells and the resulting xenograft tissue imaged following intravenous injection of ferritin using T2-weighted imaging. RESULTS Timd2-expressing cells, but not control cells, showed a large increase in both R2 and R1 in vitro following incubation with iron-loaded and manganese-loaded ferritin, respectively. Expression of Timd2 had no effect on cell viability or proliferation; however, manganese-loaded ferritin, but not iron-loaded ferritin, was toxic to Timd2-expressing cells. Timd2-expressing xenografts in vivo showed much smaller changes in R2 following injection of iron-loaded ferritin than the same cells incubated in vitro with iron-loaded ferritin. CONCLUSION Timd2 has demonstrated potential as an MRI reporter gene, producing large increases in R2 and R1 with ferritin and manganese-loaded ferritin respectively in vitro, although more modest changes in R2 in vivo. Manganese-loaded apoferritin was not used in vivo due to the toxicity observed in vitro. Magnetic Resonance in Medicine published by Wiley Periodicals, Inc. on behalf of International Society for Magnetic Resonance.
Collapse
Affiliation(s)
- P. Stephen Patrick
- Department of BiochemistryUniversity of CambridgeCambridgeUnited Kingdom
- Cancer Research UK Cambridge Institute, University of CambridgeCambridgeUnited Kingdom
| | - Tiago B. Rodrigues
- Cancer Research UK Cambridge Institute, University of CambridgeCambridgeUnited Kingdom
| | - Mikko I. Kettunen
- Cancer Research UK Cambridge Institute, University of CambridgeCambridgeUnited Kingdom
| | - Scott K. Lyons
- Cancer Research UK Cambridge Institute, University of CambridgeCambridgeUnited Kingdom
| | - André A. Neves
- Cancer Research UK Cambridge Institute, University of CambridgeCambridgeUnited Kingdom
| | - Kevin M. Brindle
- Department of BiochemistryUniversity of CambridgeCambridgeUnited Kingdom
- Cancer Research UK Cambridge Institute, University of CambridgeCambridgeUnited Kingdom
| |
Collapse
|
68
|
Lemaster JE, Jokerst JV. What is new in nanoparticle-based photoacoustic imaging? WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2016; 9. [PMID: 27038222 DOI: 10.1002/wnan.1404] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 02/15/2016] [Accepted: 02/24/2016] [Indexed: 11/07/2022]
Abstract
Photoacoustic imaging combines the high temporal and spatial resolution of ultrasound with the good contrast and spectral tuning of optical imaging. Contrast agents are used in photoacoustic imaging to further increase the contrast and specificity of imaging or to image a specific molecular process, e.g., cell-surface proteins or small molecule biomarkers. Nanoparticle-based contrast agents are important tools in photoacoustic imaging because they offer intense and stable signal and can be targeted to specific molecular processes. In this review, we describe some of the most interesting and recent advances in nanoparticle-based photoacoustic imaging including inorganic nanoparticles, organic/polymeric nanoparticles, nanoparticle coatings, multimodality imaging, as well as emerging topics in the field. WIREs Nanomed Nanobiotechnol 2017, 9:e1404. doi: 10.1002/wnan.1404 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Jeanne E Lemaster
- Department of NanoEngineering, University of California, San Diego, CA, USA
| | - Jesse V Jokerst
- Department of NanoEngineering, University of California, San Diego, CA, USA
| |
Collapse
|
69
|
Stem Cell Imaging: Tools to Improve Cell Delivery and Viability. Stem Cells Int 2016; 2016:9240652. [PMID: 26880997 PMCID: PMC4736428 DOI: 10.1155/2016/9240652] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 10/06/2015] [Accepted: 10/15/2015] [Indexed: 01/01/2023] Open
Abstract
Stem cell therapy (SCT) has shown very promising preclinical results in a variety of regenerative medicine applications. Nevertheless, the complete utility of this technology remains unrealized. Imaging is a potent tool used in multiple stages of SCT and this review describes the role that imaging plays in cell harvest, cell purification, and cell implantation, as well as a discussion of how imaging can be used to assess outcome in SCT. We close with some perspective on potential growth in the field.
Collapse
|
70
|
Abstract
Photoacoustic tomography (PAT) combines rich optical absorption contrast with the high spatial resolution of ultrasound at depths in tissue. The high scalability of PAT has enabled anatomical imaging of biological structures ranging from organelles to organs. The inherent functional and molecular imaging capabilities of PAT have further allowed it to measure important physiological parameters and track critical cellular activities. Integration of PAT with other imaging technologies provides complementary capabilities and can potentially accelerate the clinical translation of PAT.
Collapse
Affiliation(s)
- Junjie Yao
- Optical Imaging Laboratory, Department of Biomedical Engineering, Washington University in St. Louis, MO, USA
| | - Jun Xia
- Optical Imaging Laboratory, Department of Biomedical Engineering, Washington University in St. Louis, MO, USA Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Lihong V Wang
- Optical Imaging Laboratory, Department of Biomedical Engineering, Washington University in St. Louis, MO, USA
| |
Collapse
|
71
|
He X, Cai J, Liu B, Zhong Y, Qin Y. Cellular magnetic resonance imaging contrast generated by the ferritin heavy chain genetic reporter under the control of a Tet-On switch. Stem Cell Res Ther 2015; 6:207. [PMID: 26517988 PMCID: PMC4628232 DOI: 10.1186/s13287-015-0205-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Revised: 08/30/2015] [Accepted: 10/16/2015] [Indexed: 12/26/2022] Open
Abstract
INTRODUCTION Despite the strong appeal of ferritin as a magnetic resonance imaging (MRI) reporter for stem cell research, no attempts have been made to apply this genetic imaging reporter in stem cells in an inducible manner, which is important for minimizing the potential risk related to the constitutive expression of an imaging reporter. The aim of the present study was to develop an inducible genetic MRI reporter system that enables the production of intracellular MRI contrast as needed. METHODS Ferritin heavy chain (FTH1) was genetically modified by adding a Tet-On switch. A C3H10T1/2 cell line carrying Tet-FTH1 (C3H10T1/2-FTH1) was established via lentiviral transduction. The dose- and time-dependent expression of FTH1 in C3H10T1/2 cells was assessed by western blot and immunofluorescence staining. The induced "ON" and non-induced "OFF" expressions of FTH1 were detected using a 3.0 T MRI scanner. Iron accumulation in cells was analyzed by Prussian blue staining and transmission electron microscopy (TEM). RESULTS The expression of FTH1 was both dose- and time-dependently induced, and FTH1 expression peaked in response to induction with doxycycline (Dox) at 0.2 μg/ml for 72 h. The induced expression of FTH1 resulted in a significant increase in the transverse relaxation rate of C3H10T1/2-FTH1 cells following iron supplementation. Prussian blue staining and TEM revealed extensive iron accumulation in C3H10T1/2-FTH1 cells in the presence of Dox. CONCLUSIONS Cellular MRI contrast can be produced as needed via the expression of FTH1 under the control of a Tet-On switch. This finding could lay the groundwork for the use of FTH1 to track stem cells in vivo in an inducible manner.
Collapse
Affiliation(s)
- Xiaoya He
- Department of Radiology, Children's Hospital of Chongqing Medical University, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China. .,Ministry of Education Key Laboratory of Child Development and Disorders, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China. .,Key Laboratory of Pediatrics in Chongqing, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China. .,Chongqing International Science and Technology Cooperation Center For Child Development and Disorders, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China.
| | - Jinhua Cai
- Department of Radiology, Children's Hospital of Chongqing Medical University, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China. .,Ministry of Education Key Laboratory of Child Development and Disorders, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China. .,Key Laboratory of Pediatrics in Chongqing, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China. .,Chongqing International Science and Technology Cooperation Center For Child Development and Disorders, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China.
| | - Bo Liu
- Department of Radiology, Children's Hospital of Chongqing Medical University, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China. .,Ministry of Education Key Laboratory of Child Development and Disorders, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China. .,Key Laboratory of Pediatrics in Chongqing, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China. .,Chongqing International Science and Technology Cooperation Center For Child Development and Disorders, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China.
| | - Yi Zhong
- Department of Radiology, Children's Hospital of Chongqing Medical University, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China. .,Ministry of Education Key Laboratory of Child Development and Disorders, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China. .,Key Laboratory of Pediatrics in Chongqing, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China. .,Chongqing International Science and Technology Cooperation Center For Child Development and Disorders, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China.
| | - Yong Qin
- Department of Radiology, Children's Hospital of Chongqing Medical University, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China. .,Ministry of Education Key Laboratory of Child Development and Disorders, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China. .,Key Laboratory of Pediatrics in Chongqing, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China. .,Chongqing International Science and Technology Cooperation Center For Child Development and Disorders, 136 Zhongshan 2 Road, Yuzhong District, Chongqing, 400014, China.
| |
Collapse
|
72
|
Feng H, Xia X, Li C, Song Y, Qin C, Zhang Y, Lan X. TYR as a multifunctional reporter gene regulated by the Tet-on system for multimodality imaging: an in vitro study. Sci Rep 2015; 5:15502. [PMID: 26483258 PMCID: PMC4611178 DOI: 10.1038/srep15502] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 09/24/2015] [Indexed: 12/04/2022] Open
Abstract
The human tyrosinase gene TYR is a multifunctional reporter gene with potential use in photoacoustic imaging (PAI), positron emission tomography (PET), and magnetic resonance imaging (MRI). We sought to establish and evaluate a reporter gene system using TYR under the control of the Tet-on gene expression system (gene expression induced by doxycycline [Dox]) as a multimodality imaging agent. We transfected TYR into human breast cancer cells (MDA-MB-231), naming the resulting cell line 231-TYR. Using non-transfected MDA-MB-231 cells as a control, we verified successful expression of TYR by 231-TYR after incubation with Dox using western blot, cellular tyrosinase activity, Masson-Fontana silver staining, and a cell immunofluorescence study, while the control cells and 231-TYR cells without Dox exposure revealed no TYR expression. Detected by its absorbance at 405 nm, increasing concentrations of melanin correlated positively with Dox concentration and incubation time. TYR expression by Dox-induced transfected cells shortened MRI T1 and T2 relaxation times. Photoacoustic signals were easily detected in these cells. (18)F-5-fluoro-N-(2-[diethylamino]ethyl)picolinamide ((18)F-5-FPN), which targets melanin, quickly accumulated in Dox-induced 231-TYR cells. These show that TYR induction of melanin production is regulated by the Tet-on system, and TYR-containing indicator cells may have utility in multimodality imaging.
Collapse
Affiliation(s)
- Hongyan Feng
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Xiaotian Xia
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Chongjiao Li
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Yiling Song
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Chunxia Qin
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Yongxue Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, China
| |
Collapse
|
73
|
Paproski RJ, Li Y, Barber Q, Lewis JD, Campbell RE, Zemp R. Validating tyrosinase homologue melA as a photoacoustic reporter gene for imaging Escherichia coli. JOURNAL OF BIOMEDICAL OPTICS 2015; 20:106008. [PMID: 26502231 DOI: 10.1117/1.jbo.20.10.106008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 09/22/2015] [Indexed: 06/05/2023]
Abstract
To understand the pathogenic processes for infectious bacteria, appropriate research tools are required for replicating and characterizing infections. Fluorescence and bioluminescence imaging have primarily been used to image infections in animal models, but optical scattering in tissue significantly limits imaging depth and resolution. Photoacoustic imaging, which has improved depth-to-resolution ratio compared to conventional optical imaging, could be useful for visualizing melA-expressing bacteria since melA is a bacterial tyrosinase homologue which produces melanin. Escherichia coli-expressing melA was visibly dark in liquid culture. When melA-expressing bacteria in tubes were imaged with a VisualSonics Vevo LAZR system, the signal-to-noise ratio of a 9×dilution sample was 55, suggesting that ∼20 bacteria cells could be detected with our system. Multispectral (680, 700, 750, 800, 850, and 900 nm) analysis of the photoacoustic signal allowed unmixing of melA-expressing bacteria from blood. To compare photoacoustic reporter gene melA (using Vevo system) with luminescent and fluorescent reporter gene Nano-lantern (using Bruker Xtreme In-Vivo system), tubes of bacteria expressing melA or Nano-lantern were submerged 10 mm in 1% Intralipid, spaced between <1 and 20 mm apart from each other, and imaged with the appropriate imaging modality. Photoacoustic imaging could resolve the two tubes of melA-expressing bacteria even when the tubes were less than 1 mm from each other, while bioluminescence and fluorescence imaging could not resolve the two tubes of Nano-lantern-expressing bacteria even when the tubes were spaced 10 mm from each other. After injecting 100-μL of melA-expressing bacteria in the back flank of a chicken embryo, photoacoustic imaging allowed visualization of melA-expressing bacteria up to 10-mm deep into the embryo. Photoacoustic signal from melA could also be separated from deoxy- and oxy-hemoglobin signal observed within the embryo and chorioallantoic membrane. Our results suggest that melA is a useful photoacoustic reporter gene for visualizing bacteria, and further work incorporating photoacoustic reporters into infectious bacterial strains is warranted.
Collapse
Affiliation(s)
- Robert J Paproski
- University of Alberta, Department of Electrical and Computer Engineering, Donadeo Innovation Centre for Engineering, 9211-116 Street, Edmonton, Alberta T6G 1H9, CanadabUniversity of Alberta, Department of Oncology, Katz Group Centre, 114 Street & 87 Avenu
| | - Yan Li
- University of Alberta, Department of Chemistry, 11227 Saskatchewan Drive, Edmonton, Alberta T6G 2G2, Canada
| | - Quinn Barber
- University of Alberta, Department of Electrical and Computer Engineering, Donadeo Innovation Centre for Engineering, 9211-116 Street, Edmonton, Alberta T6G 1H9, Canada
| | - John D Lewis
- University of Alberta, Department of Oncology, Katz Group Centre, 114 Street & 87 Avenue, Edmonton, Alberta T6G 2E1, Canada
| | - Robert E Campbell
- University of Alberta, Department of Chemistry, 11227 Saskatchewan Drive, Edmonton, Alberta T6G 2G2, Canada
| | - Roger Zemp
- University of Alberta, Department of Electrical and Computer Engineering, Donadeo Innovation Centre for Engineering, 9211-116 Street, Edmonton, Alberta T6G 1H9, Canada
| |
Collapse
|
74
|
Zhang R, Fan Q, Yang M, Cheng K, Lu X, Zhang L, Huang W, Cheng Z. Engineering Melanin Nanoparticles as an Efficient Drug-Delivery System for Imaging-Guided Chemotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2015. [PMID: 26222210 DOI: 10.1002/adma.201502201] [Citation(s) in RCA: 151] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
In order to promote imaging-guided chemotherapy for preclinical and clinical applications, endogenous nanosystems with both contrast and drug-delivery properties are highly desired. Here, the simple use of melanin is first reported, and this biopolymer with good biocompatibility and biodegradability, binding ability to drugs and ions, and intrinsic photoacoustic properties, can serve as an efficient endogenous nanosystem for imaging-guided tumor chemotherapy in living mice.
Collapse
Affiliation(s)
- Ruiping Zhang
- Department of Radiology, First Hospital of Shanxi Medical University, Taiyuan, 030001, China
- Molecular Imaging Program at Stanford (MIPS), Canary Center at Stanford for Cancer Early Detection, Department of Radiology and Bio-X Program, School of Medicine, Stanford University, Stanford, CA, 94305-5484, USA
| | - Quli Fan
- Key Laboratory for Organic Electronics and Information Displays and Institute of Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| | - Min Yang
- Molecular Imaging Program at Stanford (MIPS), Canary Center at Stanford for Cancer Early Detection, Department of Radiology and Bio-X Program, School of Medicine, Stanford University, Stanford, CA, 94305-5484, USA
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, China
| | - Kai Cheng
- Molecular Imaging Program at Stanford (MIPS), Canary Center at Stanford for Cancer Early Detection, Department of Radiology and Bio-X Program, School of Medicine, Stanford University, Stanford, CA, 94305-5484, USA
| | - Xiaomei Lu
- Key Laboratory for Organic Electronics and Information Displays and Institute of Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| | - Lei Zhang
- Key Laboratory for Organic Electronics and Information Displays and Institute of Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| | - Wei Huang
- Key Laboratory for Organic Electronics and Information Displays and Institute of Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| | - Zhen Cheng
- Molecular Imaging Program at Stanford (MIPS), Canary Center at Stanford for Cancer Early Detection, Department of Radiology and Bio-X Program, School of Medicine, Stanford University, Stanford, CA, 94305-5484, USA
| |
Collapse
|
75
|
Listen to the chemical and histological information in biological tissue. CHINESE CHEM LETT 2015. [DOI: 10.1016/j.cclet.2015.01.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
76
|
Badar A, Kiru L, Kalber TL, Jathoul A, Straathof K, Årstad E, Lythgoe MF, Pule M. Fluorescence-guided development of a tricistronic vector encoding bimodal optical and nuclear genetic reporters for in vivo cellular imaging. EJNMMI Res 2015; 5:18. [PMID: 25853023 PMCID: PMC4385325 DOI: 10.1186/s13550-015-0097-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 03/10/2015] [Indexed: 01/26/2023] Open
Abstract
Background In vivo imaging using genetic reporters is a central supporting tool in the development of cell and gene therapies affording us the ability to selectively track the therapeutic indefinitely. Previous studies have demonstrated the utility of the human norepinephrine transporter (hNET) as a positron emission tomography/single photon emission computed tomography (PET/SPECT) genetic reporter for in vivo cellular imaging. Here, our aim was to extend on this work and construct a tricistronic vector with dual optical (firefly luciferase) and nuclear (hNET) in vivo imaging and ex vivo histochemical capabilities. Guiding this development, we describe how a fluorescent substrate for hNET, 4-(4-(dimethylamino)styryl)-N-methylpyridinium (ASP+), can be used to optimise vector design and serve as an in vitro functional screen. Methods Vectors were designed to co-express a bright red-shifted firefly luciferase (FLuc), hNET and a small marker gene RQR8. Genes were co-expressed using 2A peptide linkage, and vectors were transduced into a T cell line, SupT1. Two vectors were constructed with different gene orientations; FLuc.2A.RQR8.2A.hNET and hNET.2A.FLuc.2A.RQR8. hNET function was assessed using ASP+-guided flow cytometry. In vivo cellular conspicuity was confirmed using sequential bioluminescence imaging (BLI) and SPECT imaging of transduced SupT1 cells injected into the flanks of mice. Results SupT1/FLuc.2A.RQR8.2A.hNET cells resulted in >4-fold higher ASP+ uptake compared to SupT1/hNET.2A.FLuc.2A.RQR8, suggesting that 2A orientation effected hNET function. SupT1/FLuc.2A.RQR8.2A.hNET cells were readily visualised with both BLI and SPECT, demonstrating high signal to noise at 24 h post 123I-meta-iodobenzylguanidine (MIBG) administration. Conclusions In this study, a pre-clinical tricistronic vector with flow cytometry, BLI, SPECT and histochemical capabilities was constructed, which can be widely applied in cell tracking studies supporting the development of cell therapies. The study further demonstrates that hNET function in engineered cells can be assessed using ASP+-guided flow cytometry in place of costly radiosubstrate methodologies. This fluorogenic approach is unique to the hNET PET/SPECT reporter and may prove valuable when screening large numbers of cell lines or vector/mutant constructs. Electronic supplementary material The online version of this article (doi:10.1186/s13550-015-0097-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Adam Badar
- Division of Medicine, Centre for Advanced Biomedical Imaging (CABI), University College London, 72 Huntley Street, London, WC1E 6DD UK
| | - Louise Kiru
- Division of Medicine, Centre for Advanced Biomedical Imaging (CABI), University College London, 72 Huntley Street, London, WC1E 6DD UK ; UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD UK
| | - Tammy L Kalber
- Division of Medicine, Centre for Advanced Biomedical Imaging (CABI), University College London, 72 Huntley Street, London, WC1E 6DD UK
| | - Amit Jathoul
- UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD UK
| | - Karin Straathof
- UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD UK
| | - Erik Årstad
- Department of Chemistry and Institute of Nuclear Medicine, University College London, 235 Euston Road (T-5), London, NW1 2BU UK
| | - Mark F Lythgoe
- Division of Medicine, Centre for Advanced Biomedical Imaging (CABI), University College London, 72 Huntley Street, London, WC1E 6DD UK
| | - Martin Pule
- UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD UK
| |
Collapse
|
77
|
Liopo A, Su R, Oraevsky AA. Melanin nanoparticles as a novel contrast agent for optoacoustic tomography. PHOTOACOUSTICS 2015; 3:35-43. [PMID: 25893172 PMCID: PMC4398798 DOI: 10.1016/j.pacs.2015.02.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 01/30/2015] [Accepted: 02/02/2015] [Indexed: 05/04/2023]
Abstract
We describe the synthesis and characterization of melanin-like nanoparticles (MNP) as novel contrast agents for optoacoustic tomography. Good dispersion stability of high concentration MNPs in different biological media was achieved with thiol-terminated methoxy-poly(ethylene glycol), which can be used for further functional conjugation. MNP-PEG were found biocompatible with human MCF-7 and 3T3 cells. Cell toxicity of MNPs was found lower than that of gold nanorods for concentrations that provide equal optical absorbance. Optoacoustic tomography images were obtained with Laser Optoacoustic Imaging System (LOIS-3D) from tubes filled with contrast agents and live mice. Imaging of tubes permitted verification of the system resolution <300 μm and sensitivity Δμa=0.03/cm under safe laser fluence of 20 mJ/cm(2). Water suspensions of MNP demonstrated optoacoustic efficiency that is about equal to that of gold nanorods under conditions of equal optical absorption. We conclude that MNPs have the potential for biomedical imaging applications as optoacoustic contrast agents.
Collapse
|
78
|
Xia J, Yao J, Wang LV. Photoacoustic tomography: principles and advances. ELECTROMAGNETIC WAVES (CAMBRIDGE, MASS.) 2015; 147:1-22. [PMID: 25642127 PMCID: PMC4311576 DOI: 10.2528/pier14032303] [Citation(s) in RCA: 309] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Photoacoustic tomography (PAT) is an emerging imaging modality that shows great potential for preclinical research and clinical practice. As a hybrid technique, PAT is based on the acoustic detection of optical absorption from either endogenous chromophores, such as oxy-hemoglobin and deoxy-hemoglobin, or exogenous contrast agents, such as organic dyes and nanoparticles. Because ultrasound scatters much less than light in tissue, PAT generates high-resolution images in both the optical ballistic and diffusive regimes. Over the past decade, the photoacoustic technique has been evolving rapidly, leading to a variety of exciting discoveries and applications. This review covers the basic principles of PAT and its different implementations. Strengths of PAT are highlighted, along with the most recent imaging results.
Collapse
Affiliation(s)
- Jun Xia
- Optical Imaging Laboratory, Department of Biomedical Engineering, Washington University in St. Louis, One Brookings Drive, St. Louis, Missouri 63130, USA
| | - Junjie Yao
- Optical Imaging Laboratory, Department of Biomedical Engineering, Washington University in St. Louis, One Brookings Drive, St. Louis, Missouri 63130, USA
| | - Lihong V. Wang
- Optical Imaging Laboratory, Department of Biomedical Engineering, Washington University in St. Louis, One Brookings Drive, St. Louis, Missouri 63130, USA
| |
Collapse
|
79
|
Liu T, Shi S, Liang C, Shen S, Cheng L, Wang C, Song X, Goel S, Barnhart TE, Cai W, Liu Z. Iron oxide decorated MoS2 nanosheets with double PEGylation for chelator-free radiolabeling and multimodal imaging guided photothermal therapy. ACS NANO 2015; 9:950-60. [PMID: 25562533 PMCID: PMC4351725 DOI: 10.1021/nn506757x] [Citation(s) in RCA: 335] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Theranostics for in vivo cancer diagnosis and treatment generally requires well-designed nanoscale platforms with multiple integrated functionalities. In this study, we uncover that functionalized iron oxide nanoparticles (IONPs) could be self-assembled on the surface of two-dimensional MoS2 nanosheets via sulfur chemistry, forming MoS2-IO nanocomposites, which are then modified with two types of polyethylene glycol (PEG) to acquire enhanced stability in physiological environments. Interestingly, (64)Cu, a commonly used positron-emitting radioisotope, could be firmly adsorbed on the surface of MoS2 without the need of chelating molecules, to enable in vivo positron emission tomography (PET) imaging. On the other hand, the strong near-infrared (NIR) and superparamagnetism of MoS2-IO-PEG could also be utilized for photoacoustic tomography (PAT) and magnetic resonance (MR) imaging, respectively. Under the guidance by such triple-modal imaging, which uncovers efficient tumor retention of MoS2-IO-(d)PEG upon intravenous injection, in vivo photothermal therapy is finally conducted, achieving effective tumor ablation in an animal tumor model. Our study highlights the promise of constructing multifunctional theranostic nanocomposites based on 2D transitional metal dichalcogenides for multimodal imaging-guided cancer therapy.
Collapse
Affiliation(s)
- Teng Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, the Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Sixiang Shi
- Materials Science Program, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | - Chao Liang
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, the Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Sida Shen
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, the Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Liang Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, the Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Chao Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, the Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Xuejiao Song
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, the Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Shreya Goel
- Materials Science Program, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | - Todd E. Barnhart
- Department of Medical Physics, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | - Weibo Cai
- Materials Science Program, University of Wisconsin–Madison, Madison, Wisconsin, United States
- Department of Medical Physics, University of Wisconsin–Madison, Madison, Wisconsin, United States
- Department of Radiology, University of Wisconsin–Madison, Madison, Wisconsin, United States
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin, United States
- Address correspondence to ,
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, the Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu 215123, China
- Address correspondence to ,
| |
Collapse
|
80
|
|
81
|
Fan Q, Cheng K, Hu X, Ma X, Zhang R, Yang M, Lu X, Xing L, Huang W, Gambhir SS, Cheng Z. Transferring biomarker into molecular probe: melanin nanoparticle as a naturally active platform for multimodality imaging. J Am Chem Soc 2014; 136:15185-94. [PMID: 25292385 PMCID: PMC4227813 DOI: 10.1021/ja505412p] [Citation(s) in RCA: 284] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Indexed: 12/12/2022]
Abstract
Developing multifunctional and easily prepared nanoplatforms with integrated different modalities is highly challenging for molecular imaging. Here, we report the successful transfer of an important molecular target, melanin, into a novel multimodality imaging nanoplatform. Melanin is abundantly expressed in melanotic melanomas and thus has been actively studied as a target for melanoma imaging. In our work, the multifunctional biopolymer nanoplatform based on ultrasmall (<10 nm) water-soluble melanin nanoparticle (MNP) was developed and showed unique photoacoustic property and natural binding ability with metal ions (for example, (64)Cu(2+), Fe(3+)). Therefore, MNP can serve not only as a photoacoustic contrast agent, but also as a nanoplatform for positron emission tomography (PET) and magnetic resonance imaging (MRI). Traditional passive nanoplatforms require complicated and time-consuming processes for prebuilding reporting moieties or chemical modifications using active groups to integrate different contrast properties into one entity. In comparison, utilizing functional biomarker melanin can greatly simplify the building process. We further conjugated αvβ3 integrins, cyclic c(RGDfC) peptide, to MNPs to allow for U87MG tumor accumulation due to its targeting property combined with the enhanced permeability and retention (EPR) effect. The multimodal properties of MNPs demonstrate the high potential of endogenous materials with multifunctions as nanoplatforms for molecular theranostics and clinical translation.
Collapse
Affiliation(s)
- Quli Fan
- Molecular
Imaging Program at Stanford (MIPS), Canary Center at Stanford for
Cancer Early Detection, Department of Radiology and Bio-X Program,
School of Medicine, Stanford University, Stanford, California 94305-5484, United States
- Key
Laboratory for Organic Electronics & Information Displays and
Institute of Advanced Materials, Nanjing
University of Posts & Telecommunications, Nanjing 210046, China
| | - Kai Cheng
- Molecular
Imaging Program at Stanford (MIPS), Canary Center at Stanford for
Cancer Early Detection, Department of Radiology and Bio-X Program,
School of Medicine, Stanford University, Stanford, California 94305-5484, United States
| | - Xiang Hu
- Molecular
Imaging Program at Stanford (MIPS), Canary Center at Stanford for
Cancer Early Detection, Department of Radiology and Bio-X Program,
School of Medicine, Stanford University, Stanford, California 94305-5484, United States
| | - Xiaowei Ma
- Molecular
Imaging Program at Stanford (MIPS), Canary Center at Stanford for
Cancer Early Detection, Department of Radiology and Bio-X Program,
School of Medicine, Stanford University, Stanford, California 94305-5484, United States
| | - Ruiping Zhang
- Molecular
Imaging Program at Stanford (MIPS), Canary Center at Stanford for
Cancer Early Detection, Department of Radiology and Bio-X Program,
School of Medicine, Stanford University, Stanford, California 94305-5484, United States
| | - Min Yang
- Molecular
Imaging Program at Stanford (MIPS), Canary Center at Stanford for
Cancer Early Detection, Department of Radiology and Bio-X Program,
School of Medicine, Stanford University, Stanford, California 94305-5484, United States
| | - Xiaomei Lu
- Key
Laboratory for Organic Electronics & Information Displays and
Institute of Advanced Materials, Nanjing
University of Posts & Telecommunications, Nanjing 210046, China
| | - Lei Xing
- Department
of Radiation Oncology, School of Medicine, Stanford University, Stanford, California 94305, United States
| | - Wei Huang
- Key
Laboratory for Organic Electronics & Information Displays and
Institute of Advanced Materials, Nanjing
University of Posts & Telecommunications, Nanjing 210046, China
| | - Sanjiv Sam Gambhir
- Molecular
Imaging Program at Stanford (MIPS), Canary Center at Stanford for
Cancer Early Detection, Department of Radiology and Bio-X Program,
School of Medicine, Stanford University, Stanford, California 94305-5484, United States
| | - Zhen Cheng
- Molecular
Imaging Program at Stanford (MIPS), Canary Center at Stanford for
Cancer Early Detection, Department of Radiology and Bio-X Program,
School of Medicine, Stanford University, Stanford, California 94305-5484, United States
| |
Collapse
|
82
|
Cheng K, Yang M, Zhang R, Qin C, Su X, Cheng Z. Hybrid nanotrimers for dual T1 and T2-weighted magnetic resonance imaging. ACS NANO 2014; 8:9884-9896. [PMID: 25283972 PMCID: PMC4334264 DOI: 10.1021/nn500188y] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Accepted: 10/03/2014] [Indexed: 05/28/2023]
Abstract
Development of multifunctional nanoparticle-based probes for dual T1- and T2-weighted magnetic resonance imaging (MRI) could allow us to image and diagnose the tumors or other abnormalities in an exceptionally accurate and reliable manner. In this study, by fusing distinct nanocrystals via solid-state interfaces, we built hybrid heteronanostructures to combine both T1 and T2- weighted contrast agents together for MRI with high accuracy and reliability. The resultant hybrid heterotrimers showed high stability in physiological conditions and could induce both simultaneous positive and negative contrast enhancements in MR images. Small animal positron emission tomography imaging study revealed that the hybrid heterostructures displayed favorable biodistribution and were suitable for in vivo imaging. Their potential as dual contrast agents for T1 and T2-weighted MRI was further demonstrated by in vitro and in vivo imaging and relaxivity measurements.
Collapse
Affiliation(s)
- Kai Cheng
- Molecular Imaging Program at Stanford (MIPS), Canary Center at Stanford for Cancer Early Detection, Department of Radiology and Bio-X Program, School of Medicine, Stanford University, 1201 Welch Road, Lucas P095, Stanford, California 94305-5484, United States
| | - Meng Yang
- Molecular Imaging Program at Stanford (MIPS), Canary Center at Stanford for Cancer Early Detection, Department of Radiology and Bio-X Program, School of Medicine, Stanford University, 1201 Welch Road, Lucas P095, Stanford, California 94305-5484, United States
| | - Ruiping Zhang
- Molecular Imaging Program at Stanford (MIPS), Canary Center at Stanford for Cancer Early Detection, Department of Radiology and Bio-X Program, School of Medicine, Stanford University, 1201 Welch Road, Lucas P095, Stanford, California 94305-5484, United States
- Department of Radiology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Chunxia Qin
- Molecular Imaging Program at Stanford (MIPS), Canary Center at Stanford for Cancer Early Detection, Department of Radiology and Bio-X Program, School of Medicine, Stanford University, 1201 Welch Road, Lucas P095, Stanford, California 94305-5484, United States
| | - Xinhui Su
- Molecular Imaging Program at Stanford (MIPS), Canary Center at Stanford for Cancer Early Detection, Department of Radiology and Bio-X Program, School of Medicine, Stanford University, 1201 Welch Road, Lucas P095, Stanford, California 94305-5484, United States
| | - Zhen Cheng
- Molecular Imaging Program at Stanford (MIPS), Canary Center at Stanford for Cancer Early Detection, Department of Radiology and Bio-X Program, School of Medicine, Stanford University, 1201 Welch Road, Lucas P095, Stanford, California 94305-5484, United States
| |
Collapse
|
83
|
Abstract
Molecular imaging non-invasively visualizes and characterizes the biologic functions and mechanisms in living organisms at a molecular level. In recent years, advances in imaging instruments, imaging probes, assay methods, and quantification techniques have enabled more refined and reliable images for more accurate diagnoses. Multimodal imaging combines two or more imaging modalities into one system to produce details in clinical diagnostic imaging that are more precise than conventional imaging. Multimodal imaging offers complementary advantages: high spatial resolution, soft tissue contrast, and biological information on the molecular level with high sensitivity. However, combining all modalities into a single imaging probe involves problems yet to be solved due to the requirement of high dose contrast agents for a component of imaging modality with low sensitivity. The introduction of targeting moieties into the probes enhances the specific binding of targeted multimodal imaging modalities and selective accumulation of the imaging agents at a disease site to provide more accurate diagnoses. An extensive list of prior reports on the targeted multimodal imaging probes categorized by each modality is presented and discussed. In addition to accurate diagnosis, targeted multimodal imaging agents carrying therapeutic medications make it possible to visualize the theranostic effect and the progress of disease. This will facilitate the development of an imaging-guided therapy, which will widen the application of the targeted multimodal imaging field to experiments in vivo.
Collapse
|
84
|
Kiessling F. Science to practice: Cellular therapy of Parkinson disease--a new radiotracer to target transplanted dopaminergic cells with PET. Radiology 2014; 272:1-3. [PMID: 24956043 DOI: 10.1148/radiol.14140548] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The article by Bu and colleagues (1) introduces N-[2-(diethylamino)ethyl]-(18)F-5-fluoropicolinamide ((18)F-P3BZA) as a promising positron emission tomography (PET) radiotracer to target transplanted porcine retinal pigment epithelial (pRPE) cells in the striatum. This strategy to monitor cellular therapy of Parkinson disease has a high translational potential and in the future may help us to better understand some of the controversial results reported in clinical trials.
Collapse
Affiliation(s)
- Fabian Kiessling
- Department of Experimental Molecular Imaging, RWTH Aachen University Pauwelsstrasse 20 52074 Aachen, Germany
| |
Collapse
|
85
|
Vande Velde G, Himmelreich U, Neeman M. Reporter gene approaches for mapping cell fate decisions by MRI: promises and pitfalls. CONTRAST MEDIA & MOLECULAR IMAGING 2014; 8:424-31. [PMID: 24375898 DOI: 10.1002/cmmi.1590] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 11/29/2013] [Indexed: 12/15/2022]
Abstract
The central dogma of molecular biology, namely the process by which information encoded in the DNA serves as the template for transcriptional activation of specific mRNA resulting in temporal and spatial control of the translation of specific proteins, stands at the basis of normal and pathological cellular processes. Serving as the primary mechanism linking genotype to phenotype, it is clearly of significant interest for in vivo imaging. While classically, imaging revolutionized the ability to phenotype the anatomical and physiological impact of induction of changes in gene expression, the preceding molecular events remained invisible. Reporter gene-based imaging techniques provide a window for in vivo visualization of such transcriptional activation events. In addition to the widespread use of fluorescent and bioluminescent reporter genes and development of a number of reporter genes for positron emission tomography (PET) imaging, there has been significant progress in the development of reporter genes for MRI. With the development of strategies for cellular based therapies, such imaging tools could become central components for personalized patient monitoring.
Collapse
Affiliation(s)
- Greetje Vande Velde
- Biomedical MRI, KU Leuven, O&N I Herestraat 49 - box 505, 3000, Leuven, Belgium
| | | | | |
Collapse
|
86
|
Multi-wavelength photoacoustic imaging of inducible tyrosinase reporter gene expression in xenograft tumors. Sci Rep 2014; 4:5329. [PMID: 24936769 PMCID: PMC4060505 DOI: 10.1038/srep05329] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 06/02/2014] [Indexed: 12/17/2022] Open
Abstract
Photoacoustic imaging is an emerging hybrid imaging technology capable of breaking through resolution limits of pure optical imaging technologies imposed by optical-scattering to provide fine-resolution optical contrast information in deep tissues. We demonstrate the ability of multi-wavelength photoacoustic imaging to estimate relative gene expression distributions using an inducible expression system and co-register images with hemoglobin oxygen saturation estimates and micro-ultrasound data. Tyrosinase, the rate-limiting enzyme in melanin production, is used as a reporter gene owing to its strong optical absorption and enzymatic amplification mechanism. Tetracycline-inducible melanin expression is turned on via doxycycline treatment in vivo. Serial multi-wavelength imaging reveals very low estimated melanin expression in tumors prior to doxycycline treatment or in tumors with no tyrosinase gene present, but strong signals after melanin induction in tumors tagged with the tyrosinase reporter. The combination of new inducible reporters and high-resolution photoacoustic and micro-ultrasound technology is poised to bring a new dimension to the study of gene expression in vivo.
Collapse
|
87
|
Yao J, Wang LV. Photoacoustic Brain Imaging: from Microscopic to Macroscopic Scales. NEUROPHOTONICS 2014; 1:1877516. [PMID: 25401121 PMCID: PMC4232215 DOI: 10.1117/1.nph.1.1.011003] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 03/28/2014] [Accepted: 03/31/2014] [Indexed: 05/12/2023]
Abstract
Human brain mapping has become one of the most exciting contemporary research areas, with major breakthroughs expected in the following decades. Modern brain imaging techniques have allowed neuroscientists to gather a wealth of anatomic and functional information about the brain. Among these techniques, by virtue of its rich optical absorption contrast, high spatial and temporal resolutions, and deep penetration, photoacoustic tomography (PAT) has attracted more and more attention, and is playing an increasingly important role in brain studies. In particular, PAT complements other brain imaging modalities by providing high-resolution functional and metabolic imaging. More importantly, PAT's unique scalability enables scrutinizing the brain at both microscopic and macroscopic scales, using the same imaging contrast. In this Review, we present the state-of-the-art PAT techniques for brain imaging, summarize representative neuroscience applications, outline the technical challenges in translating PAT to human brain imaging, and envision potential technological deliverables.
Collapse
Affiliation(s)
- Junjie Yao
- Washington University in St. Louis, Department of Biomedical Engineering, Optical Imaging Laboratory, One Brookings Drive, St. Louis, Missouri 63130
| | - Lihong V. Wang
- Washington University in St. Louis, Department of Biomedical Engineering, Optical Imaging Laboratory, One Brookings Drive, St. Louis, Missouri 63130
| |
Collapse
|
88
|
Bu L, Li R, Liu H, Feng W, Xiong X, Zhao H, Vollrath D, Shen B, Cheng Z. Intrastriatal transplantation of retinal pigment epithelial cells for the treatment of Parkinson disease: in vivo longitudinal molecular imaging with 18F-P3BZA PET/CT. Radiology 2014; 272:174-83. [PMID: 24758555 DOI: 10.1148/radiol.14132042] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
PURPOSE To evaluate the performance of N-[2-(diethylamino)ethyl]-(18)F-5-fluoropicolinamide ((18)F-P3BZA) for visualizing porcine retinal pigment epithelium (pRPE) cells transplanted in the striatum for the treatment of Parkinson disease and to monitor the long-term activity of implanted pRPE cells by means of (18)F-P3BZA positron emission tomography (PET)/computed tomography (CT) in vivo. MATERIALS AND METHODS Animal work was conducted in accordance with the administrative panel on laboratory animal care. In vitro cell uptake of (18)F-P3BZA was determined with incubation of melanotic pRPE or amelanotic ARPE-19 cells with (18)F-P3BZA. To visualize the implanted pRPE cells in vivo, normal rats (four per group) were injected with pRPE or ARPE-19 cells attached to gelatin microcarriers in the left striatum and with control gelatin microcarriers in the right striatum and followed up with small animal PET/CT. Longitudinal PET/CT scans were acquired in 12 rats up to 16 days after surgery. Postmortem analysis, which included autoradiography and hematoxylin-eosin, Fontana-Masson, and immunofluorescence staining, was performed. Data were compared with the Student t test, analysis of variance, and regression analysis. RESULTS (18)F-P3BZA accumulated in pRPE cells effectively (3.48% of the injected dose [ID] per gram of brain tissue ± 0.58 at 1 hour after injection of the probe at 2 days after surgery in vivo) but not in control ARPE-19 cells (P < .05). Longitudinal PET/CT scans revealed that the activity of implanted pRPE cells decreased over time, as evidenced by a reduction in (18)F-P3BZA uptake (3.39% ID/g ± 0.18, 2.49% ID/g ± 0.41, and 1.20% ID/g ± 0.13 at days 2, 9, and 16, respectively; P < .05). Postmortem analysis helped confirm the results of in vivo imaging. CONCLUSION (18)F-P3BZA PET/CT is a feasible technique for visualizing and detecting the activity of implanted RPE cells in vivo.
Collapse
Affiliation(s)
- Lihong Bu
- From the Molecular Imaging Program at Stanford (MIPS), Department of Radiology and Bio-X Program, Stanford University, 1201 Welch Rd, Lucas Center, Room P095, Stanford, CA 94305-5484 (L.B., R.L., H.L., Z.C.); Molecular Imaging Center, Department of Radiology, The 4th Hospital of Harbin Medical University, Harbin, Heilongjiang, P.R. China (L.B., R.L., B.S.); and Departments of Genetics (W.F., D.V.) and Neurosurgery (X.X., H.Z.), School of Medicine, Stanford University, Stanford, Calif
| | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Cheng K, Kothapalli SR, Liu H, Koh AL, Jokerst JV, Jiang H, Yang M, Li J, Levi J, Wu JC, Gambhir SS, Cheng Z. Construction and validation of nano gold tripods for molecular imaging of living subjects. J Am Chem Soc 2014; 136:3560-71. [PMID: 24495038 PMCID: PMC3985880 DOI: 10.1021/ja412001e] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Indexed: 12/20/2022]
Abstract
Anisotropic colloidal hybrid nanoparticles exhibit superior optical and physical properties compared to their counterparts with regular architectures. We herein developed a controlled, stepwise strategy to build novel, anisotropic, branched, gold nanoarchitectures (Au-tripods) with predetermined composition and morphology for bioimaging. The resultant Au-tripods with size less than 20 nm showed great promise as contrast agents for in vivo photoacoustic imaging (PAI). We further identified Au-tripods with two possible configurations as high-absorbance nanomaterials from various gold multipods using a numerical simulation analysis. The PAI signals were linearly correlated with their concentrations after subcutaneous injection. The in vivo biodistribution of Au-tripods favorable for molecular imaging was confirmed using small animal positron emission tomography (PET). Intravenous administration of cyclic Arg-Gly-Asp-d-Phe-Cys (RGDfC) peptide conjugated Au-tripods (RGD-Au-tripods) to U87MG tumor-bearing mice showed PAI contrasts in tumors almost 3-fold higher than for the blocking group. PAI results correlated well with the corresponding PET images. Quantitative biodistribution data revealed that 7.9% ID/g of RGD-Au-tripods had accumulated in the U87MG tumor after 24 h post-injection. A pilot mouse toxicology study confirmed that no evidence of significant acute or systemic toxicity was observed in histopathological examination. Our study suggests that Au-tripods can be reliably synthesized through stringently controlled chemical synthesis and could serve as a new generation of platform with high selectivity and sensitivity for multimodality molecular imaging.
Collapse
Affiliation(s)
- Kai Cheng
- Molecular Imaging Program at Stanford (MIPS), Canary Center at
Stanford for Cancer
Early Detection, Department of Radiology and Bio-X Program, School of Medicine, Stanford Nanocharacterization
Laboratory, Stanford University, 1201 Welch Road, Lucas P095, Stanford, California 94305-5484, United States
| | - Sri-Rajasekhar Kothapalli
- Molecular Imaging Program at Stanford (MIPS), Canary Center at
Stanford for Cancer
Early Detection, Department of Radiology and Bio-X Program, School of Medicine, Stanford Nanocharacterization
Laboratory, Stanford University, 1201 Welch Road, Lucas P095, Stanford, California 94305-5484, United States
| | - Hongguang Liu
- Molecular Imaging Program at Stanford (MIPS), Canary Center at
Stanford for Cancer
Early Detection, Department of Radiology and Bio-X Program, School of Medicine, Stanford Nanocharacterization
Laboratory, Stanford University, 1201 Welch Road, Lucas P095, Stanford, California 94305-5484, United States
| | - Ai Leen Koh
- Molecular Imaging Program at Stanford (MIPS), Canary Center at
Stanford for Cancer
Early Detection, Department of Radiology and Bio-X Program, School of Medicine, Stanford Nanocharacterization
Laboratory, Stanford University, 1201 Welch Road, Lucas P095, Stanford, California 94305-5484, United States
| | - Jesse V. Jokerst
- Molecular Imaging Program at Stanford (MIPS), Canary Center at
Stanford for Cancer
Early Detection, Department of Radiology and Bio-X Program, School of Medicine, Stanford Nanocharacterization
Laboratory, Stanford University, 1201 Welch Road, Lucas P095, Stanford, California 94305-5484, United States
| | - Han Jiang
- Molecular Imaging Program at Stanford (MIPS), Canary Center at
Stanford for Cancer
Early Detection, Department of Radiology and Bio-X Program, School of Medicine, Stanford Nanocharacterization
Laboratory, Stanford University, 1201 Welch Road, Lucas P095, Stanford, California 94305-5484, United States
| | - Meng Yang
- Molecular Imaging Program at Stanford (MIPS), Canary Center at
Stanford for Cancer
Early Detection, Department of Radiology and Bio-X Program, School of Medicine, Stanford Nanocharacterization
Laboratory, Stanford University, 1201 Welch Road, Lucas P095, Stanford, California 94305-5484, United States
| | - Jinbo Li
- Molecular Imaging Program at Stanford (MIPS), Canary Center at
Stanford for Cancer
Early Detection, Department of Radiology and Bio-X Program, School of Medicine, Stanford Nanocharacterization
Laboratory, Stanford University, 1201 Welch Road, Lucas P095, Stanford, California 94305-5484, United States
| | - Jelena Levi
- Molecular Imaging Program at Stanford (MIPS), Canary Center at
Stanford for Cancer
Early Detection, Department of Radiology and Bio-X Program, School of Medicine, Stanford Nanocharacterization
Laboratory, Stanford University, 1201 Welch Road, Lucas P095, Stanford, California 94305-5484, United States
| | - Joseph C. Wu
- Molecular Imaging Program at Stanford (MIPS), Canary Center at
Stanford for Cancer
Early Detection, Department of Radiology and Bio-X Program, School of Medicine, Stanford Nanocharacterization
Laboratory, Stanford University, 1201 Welch Road, Lucas P095, Stanford, California 94305-5484, United States
| | - Sanjiv S. Gambhir
- Molecular Imaging Program at Stanford (MIPS), Canary Center at
Stanford for Cancer
Early Detection, Department of Radiology and Bio-X Program, School of Medicine, Stanford Nanocharacterization
Laboratory, Stanford University, 1201 Welch Road, Lucas P095, Stanford, California 94305-5484, United States
| | - Zhen Cheng
- Molecular Imaging Program at Stanford (MIPS), Canary Center at
Stanford for Cancer
Early Detection, Department of Radiology and Bio-X Program, School of Medicine, Stanford Nanocharacterization
Laboratory, Stanford University, 1201 Welch Road, Lucas P095, Stanford, California 94305-5484, United States
| |
Collapse
|
90
|
Zackrisson S, van de Ven SMWY, Gambhir SS. Light in and sound out: emerging translational strategies for photoacoustic imaging. Cancer Res 2014; 74:979-1004. [PMID: 24514041 DOI: 10.1158/0008-5472.can-13-2387] [Citation(s) in RCA: 318] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Photoacoustic imaging (PAI) has the potential for real-time molecular imaging at high resolution and deep inside the tissue, using nonionizing radiation and not necessarily depending on exogenous imaging agents, making this technique very promising for a range of clinical applications. The fact that PAI systems can be made portable and compatible with existing imaging technologies favors clinical translation even more. The breadth of clinical applications in which photoacoustics could play a valuable role include: noninvasive imaging of the breast, sentinel lymph nodes, skin, thyroid, eye, prostate (transrectal), and ovaries (transvaginal); minimally invasive endoscopic imaging of gastrointestinal tract, bladder, and circulating tumor cells (in vivo flow cytometry); and intraoperative imaging for assessment of tumor margins and (lymph node) metastases. In this review, we describe the basics of PAI and its recent advances in biomedical research, followed by a discussion of strategies for clinical translation of the technique.
Collapse
Affiliation(s)
- S Zackrisson
- Departments of Radiology, Bioengineering, and Department of Materials Science & Engineering. Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, CA, USA.,Diagnostic Radiology, Department of Clinical Sciences in Malmö, Lund University, Sweden
| | - S M W Y van de Ven
- Departments of Radiology, Bioengineering, and Department of Materials Science & Engineering. Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, CA, USA
| | - S S Gambhir
- Departments of Radiology, Bioengineering, and Department of Materials Science & Engineering. Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
91
|
Kooreman NG, Ransohoff JD, Wu JC. Tracking gene and cell fate for therapeutic gain. NATURE MATERIALS 2014; 13:106-9. [PMID: 24452344 PMCID: PMC4892936 DOI: 10.1038/nmat3868] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Parallel advances in molecular imaging modalities and in gene- and cell-based therapeutics have significantly advanced their respective fields. Here we discuss how the collaborative, preclinical intersection of these technologies will facilitate more informed and effective clinical translation of relevant therapies.
Collapse
Affiliation(s)
- Nigel G. Kooreman
- Stanford Cardiovascular Institute
- Departments of Medicine and Radiology (Molecular Imaging Program)
| | - Julia D. Ransohoff
- Stanford Cardiovascular Institute
- Departments of Medicine and Radiology (Molecular Imaging Program)
| | - Joseph C. Wu
- Stanford Cardiovascular Institute
- Departments of Medicine and Radiology (Molecular Imaging Program)
- Institute of Stem Cell Biology and Regenerative Medicine
| |
Collapse
|
92
|
Minn I, Menezes ME, Sarkar S, Yarlagadda K, Das SK, Emdad L, Sarkar D, Fisher PB, Pomper MG. Molecular-genetic imaging of cancer. Adv Cancer Res 2014; 124:131-69. [PMID: 25287688 PMCID: PMC4339000 DOI: 10.1016/b978-0-12-411638-2.00004-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Molecular-genetic imaging of cancer using nonviral delivery systems has great potential for clinical application as a safe, efficient, noninvasive tool for visualization of various cellular processes including detection of cancer, and its attendant metastases. In recent years, significant effort has been expended in overcoming technical hurdles to enable clinical adoption of molecular-genetic imaging. This chapter will provide an introduction to the components of molecular-genetic imaging and recent advances on each component leading to safe, efficient clinical applications for detecting cancer. Combination with therapy, namely, generating molecular-genetic theranostic constructs, will provide further impetus for clinical translation of this promising technology.
Collapse
Affiliation(s)
- Il Minn
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Siddik Sarkar
- Department of Human and Molecular Genetics, Richmond, Virginia, USA
| | - Keerthi Yarlagadda
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland, USA
| | - Swadesh K Das
- Department of Human and Molecular Genetics, Richmond, Virginia, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Luni Emdad
- Department of Human and Molecular Genetics, Richmond, Virginia, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Devanand Sarkar
- Department of Human and Molecular Genetics, Richmond, Virginia, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Richmond, Virginia, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Martin G Pomper
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland, USA; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA.
| |
Collapse
|
93
|
Nie L, Chen X. Structural and functional photoacoustic molecular tomography aided by emerging contrast agents. Chem Soc Rev 2014; 43:7132-70. [PMID: 24967718 PMCID: PMC4569000 DOI: 10.1039/c4cs00086b] [Citation(s) in RCA: 269] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Photoacoustic tomography (PAT) can offer structural, functional and molecular contrasts at scalable observation level. By ultrasonically overcoming the strong optical scattering, this imaging technology can reach centimeters penetration depth while retaining high spatial resolution in biological tissue. Recent extensive research has been focused on developing new contrast agents to improve the imaging sensitivity, specificity and efficiency. These emerging materials have substantially accelerated PAT applications in signal sensing, functional imaging, biomarker labeling and therapy monitoring etc. Here, the potentials of different optical probes as PAT contrast agents were elucidated. We first describe the instrumental embodiments and the measured functional parameters, then focus on emerging contrast agent-based PAT applications, and finally discuss the challenges and prospects.
Collapse
Affiliation(s)
- Liming Nie
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA.
| | | |
Collapse
|
94
|
Multimodality molecular imaging of stem cells therapy for stroke. BIOMED RESEARCH INTERNATIONAL 2013; 2013:849819. [PMID: 24222920 PMCID: PMC3816035 DOI: 10.1155/2013/849819] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2013] [Accepted: 08/21/2013] [Indexed: 12/03/2022]
Abstract
Stem cells have been proposed as a promising therapy for treating stroke. While several studies have demonstrated the therapeutic benefits of stem cells, the exact mechanism remains elusive. Molecular imaging provides the possibility of the visual representation of biological processes at the cellular and molecular level. In order to facilitate research efforts to understand the stem cells therapeutic mechanisms, we need to further develop means of monitoring these cells noninvasively, longitudinally and repeatedly. Because of tissue depth and the blood-brain barrier (BBB), in vivo imaging of stem cells therapy for stroke has unique challenges. In this review, we describe existing methods of tracking transplanted stem cells in vivo, including magnetic resonance imaging (MRI), nuclear medicine imaging, and optical imaging (OI). Each of the imaging techniques has advantages and drawbacks. Finally, we describe multimodality imaging strategies as a more comprehensive and potential method to monitor transplanted stem cells for stroke.
Collapse
|
95
|
Liu H, Liu S, Miao Z, Jiang H, Deng Z, Hong X, Cheng Z. A novel aliphatic 18F-labeled probe for PET imaging of melanoma. Mol Pharm 2013; 10:3384-91. [PMID: 23927458 DOI: 10.1021/mp400225s] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Radiofluorinated benzamide and nicotinamide analogues are promising molecular probes for the positron emission tomography (PET) imaging of melanoma. Compounds containing aromatic (benzene or pyridine) and N,N-diethylethylenediamine groups have been successfully used for development of melanin targeted PET and single-photon emission computed tomography (SPECT) imaging agents for melanoma. The objective of this study was to determine the feasibility of using aliphatic compounds as a molecular platform for the development of a new generation of PET probes for melanoma detection. An aliphatic N,N-diethylethylenediamine precursor was directly coupled to a radiofluorination synthon, p-nitrophenyl 2-(18)F-fluoropropionate ((18)F-NFP), to produce the probe N-(2-(diethylamino)ethyl)-2-(18)F-fluoropropanamide ((18)F-FPDA). The melanoma-targeting ability of (18)F-FPDA was further evaluated both in vitro and in vivo through cell uptake assays, biodistribution studies, and small animal PET imaging in C57BL/6 mice bearing B16F10 murine melanoma tumors. Beginning with the precursor (18)F-NFP, the total preparation time for (18)F-FPDA, including the final high-performance liquid chromatography purification step, was approximately 30 min, with a decay-corrected radiochemical yield of 79.8%. The melanin-targeting specificity of (18)F-FPDA was demonstrated by significantly different uptake rates in tyrosine-treated and untreated B16F10 cells in vitro. The tumor uptake of (18)F-FPDA in vivo reached 2.65 ± 0.48 %ID/g at 2 h postinjection (p.i.) in pigment-enriched B16F10 xenografts, whereas the tumor uptake of (18)F-FPDA was close to the background levels, with rates of only 0.37 ± 0.07 %ID/g at 2 h p.i. in the nonpigmented U87MG tumor mouse model. Furthermore, small animal PET imaging studies revealed that (18)F-FPDA specifically targeted the melanotic B16F10 tumor, yielding a tumor-to-muscle ratio of approximately 4:1 at 1 h p.i. and 7:1 at 2 h p.i. In summary, we report the development of a novel (18)F-labeled aliphatic compound for melanoma imaging that can be easily synthesized in high yields using the radiosynthon (18)F-NFP. The PET probe (18)F-FPDA exhibits high B16F10 tumor-targeting efficacy and favorable in vivo pharmacokinetics. Our study demonstrates that aliphatic compounds can be used as a new generation molecular platform for the development of novel melanoma targeting agents. Further evaluation and optimization of (18)F-FPDA for melanin targeted molecular imaging are therefore warranted.
Collapse
Affiliation(s)
- Hongguang Liu
- Molecular Imaging Program at Stanford (MIPS), Bio-X Program, and Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University , California, 94305-5344, United States
| | | | | | | | | | | | | |
Collapse
|