1
|
Ji H, Hu C, Yang X, Liu Y, Ji G, Ge S, Wang X, Wang M. Lymph node metastasis in cancer progression: molecular mechanisms, clinical significance and therapeutic interventions. Signal Transduct Target Ther 2023; 8:367. [PMID: 37752146 PMCID: PMC10522642 DOI: 10.1038/s41392-023-01576-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 07/04/2023] [Accepted: 07/26/2023] [Indexed: 09/28/2023] Open
Abstract
Lymph nodes (LNs) are important hubs for metastatic cell arrest and growth, immune modulation, and secondary dissemination to distant sites through a series of mechanisms, and it has been proved that lymph node metastasis (LNM) is an essential prognostic indicator in many different types of cancer. Therefore, it is important for oncologists to understand the mechanisms of tumor cells to metastasize to LNs, as well as how LNM affects the prognosis and therapy of patients with cancer in order to provide patients with accurate disease assessment and effective treatment strategies. In recent years, with the updates in both basic and clinical studies on LNM and the application of advanced medical technologies, much progress has been made in the understanding of the mechanisms of LNM and the strategies for diagnosis and treatment of LNM. In this review, current knowledge of the anatomical and physiological characteristics of LNs, as well as the molecular mechanisms of LNM, are described. The clinical significance of LNM in different anatomical sites is summarized, including the roles of LNM playing in staging, prognostic prediction, and treatment selection for patients with various types of cancers. And the novel exploration and academic disputes of strategies for recognition, diagnosis, and therapeutic interventions of metastatic LNs are also discussed.
Collapse
Affiliation(s)
- Haoran Ji
- Department of Thoracic Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Chuang Hu
- Department of Thoracic Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Xuhui Yang
- Department of Thoracic Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yuanhao Liu
- Department of Thoracic Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Guangyu Ji
- Department of Thoracic Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Shengfang Ge
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiansong Wang
- Department of Thoracic Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Mingsong Wang
- Department of Thoracic Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| |
Collapse
|
2
|
Azari F, Zhang K, Kennedy GT, Chang A, Nadeem B, Delikatny EJ, Singhal S. Precision Surgery Guided by Intraoperative Molecular Imaging. J Nucl Med 2022; 63:1620-1627. [PMID: 35953303 PMCID: PMC9635678 DOI: 10.2967/jnumed.121.263409] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 08/11/2022] [Indexed: 11/16/2022] Open
Abstract
Intraoperative molecular imaging (IMI) has recently emerged as an important tool in the armamentarium of surgical oncologists. IMI allows real-time assessment of oncologic resection quality, margin assessment, and occult disease detection during real-time surgery. Numerous tracers have now been developed for use in IMI-guided tissue sampling. Fluorochromes localize to the tumor by taking advantage of their disorganized capillary milieu, overexpressed receptors, or upregulated enzymes. Although fluorescent tracers can suffer from issues of autofluorescence and lack of depth penetration, these challenges are being addressed through hybrid radioactive/fluorescent tracers and new tracers that fluoresce in the near-infrared (NIR-II [wavelength > 1,000 nm]) range. IMI is already being used to treat numerous cancers, with demonstrated improvement in cancer recurrence and patient outcomes without incurring significant burden on either clinicians or patients. In this comprehensive review, we discuss history, mechanism, current oncologic applications, and future directions of IMI-guided optical biopsy.
Collapse
Affiliation(s)
- Feredun Azari
- Department of Thoracic Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Kevin Zhang
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - Gregory T. Kennedy
- Department of Thoracic Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Ashley Chang
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - Bilal Nadeem
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - Edward J. Delikatny
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sunil Singhal
- Department of Thoracic Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania;
| |
Collapse
|
3
|
Dual-Labelling Strategies for Nuclear and Fluorescence Molecular Imaging: Current Status and Future Perspectives. Pharmaceuticals (Basel) 2022; 15:ph15040432. [PMID: 35455430 PMCID: PMC9028399 DOI: 10.3390/ph15040432] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 03/27/2022] [Accepted: 03/28/2022] [Indexed: 12/13/2022] Open
Abstract
Molecular imaging offers the possibility to investigate biological and biochemical processes non-invasively and to obtain information on both anatomy and dysfunctions. Based on the data obtained, a fundamental understanding of various disease processes can be derived and treatment strategies can be planned. In this context, methods that combine several modalities in one probe are increasingly being used. Due to the comparably high sensitivity and provided complementary information, the combination of nuclear and optical probes has taken on a special significance. In this review article, dual-labelled systems for bimodal nuclear and optical imaging based on both modular ligands and nanomaterials are discussed. Particular attention is paid to radiometal-labelled molecules for single-photon emission computed tomography (SPECT) and positron emission tomography (PET) and metal complexes combined with fluorescent dyes for optical imaging. The clinical potential of such probes, especially for fluorescence-guided surgery, is assessed.
Collapse
|
4
|
Ariztia J, Solmont K, Moïse NP, Specklin S, Heck MP, Lamandé-Langle S, Kuhnast B. PET/Fluorescence Imaging: An Overview of the Chemical Strategies to Build Dual Imaging Tools. Bioconjug Chem 2022; 33:24-52. [PMID: 34994545 DOI: 10.1021/acs.bioconjchem.1c00503] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Molecular imaging is a biomedical research discipline that has quickly emerged to afford the observation, characterization, monitoring, and quantification of biomarkers and biological processes in living organism. It covers a large array of imaging techniques, each of which provides anatomical, functional, or metabolic information. Multimodality, as the combination of two or more of these techniques, has proven to be one of the best options to boost their individual properties, hence offering unprecedented tools for human health. In this review, we will focus on the combination of positron emission tomography and fluorescence imaging from the specific perspective of the chemical synthesis of dual imaging agents. Based on a detailed analysis of the literature, this review aims at giving a comprehensive overview of the chemical strategies implemented to build adequate imaging tools considering radiohalogens and radiometals as positron emitters, fluorescent dyes mostly emitting in the NIR window and all types of targeting vectors.
Collapse
Affiliation(s)
- Julen Ariztia
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay, 91401, Orsay, France
| | - Kathleen Solmont
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay, 91401, Orsay, France
| | | | - Simon Specklin
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay, 91401, Orsay, France
| | - Marie Pierre Heck
- Université Paris-Saclay, INRAE, Département Médicaments et Technologies pour la santé (DMTS), SCBM, 91191, Gif-sur-Yvette cedex, France
| | | | - Bertrand Kuhnast
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay, 91401, Orsay, France
| |
Collapse
|
5
|
Oksel Karakus C, Bilgi E, Winkler DA. Biomedical nanomaterials: applications, toxicological concerns, and regulatory needs. Nanotoxicology 2020; 15:331-351. [PMID: 33337941 DOI: 10.1080/17435390.2020.1860265] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Advances in cutting-edge technologies such as nano- and biotechnology have created an opportunity for re-engineering existing materials and generating new nano-scale products that can function beyond the limits of conventional ones. While the step change in the properties and functionalities of these new materials opens up new possibilities for a broad range of applications, it also calls for structural modifications to existing safety assessment processes that are primarily focused on bulk material properties. Decades after the need to modify existing risk management practices to include nano-specific behaviors and exposure pathways was recognized, relevant policies for evaluating, and controlling health risks of nano-enabled materials is still lacking. This review provides an overview of current progress in the field of nanobiotechnology rather than intentions and aspirations, summarizes long-recognized but still unresolved issues surrounding materials safety at the nanoscale, and discusses key barriers preventing generation and integration of reliable data in bio/nano-safety domain. Particular attention is given to nanostructured materials that are commonly used in biomedical applications.
Collapse
Affiliation(s)
| | - Eyup Bilgi
- Department of Bioengineering, Izmir Institute of Technology, Izmir, Turkey
| | - David A Winkler
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia.,Latrobe Institute for Molecular Science, La Trobe University, Bundoora, Australia.,School of Pharmacy, University of Nottingham, Nottingham, UK.,CSIRO Data61, Pullenvale, Australia
| |
Collapse
|
6
|
Klenner MA, Pascali G, Massi M, Fraser BH. Fluorine‐18 Radiolabelling and Photophysical Characteristics of Multimodal PET–Fluorescence Molecular Probes. Chemistry 2020; 27:861-876. [DOI: 10.1002/chem.202001402] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Indexed: 12/22/2022]
Affiliation(s)
- Mitchell A. Klenner
- Human Health and National Deuteration Facility (NDF) Australian Nuclear Science and Technology Organisation (ANSTO) New Illawarra Road Lucas Heights NSW 2234 Australia
- School of Molecular and Life Sciences Curtin University Kent Street Bentley WA 6102 Australia
| | - Giancarlo Pascali
- Human Health and National Deuteration Facility (NDF) Australian Nuclear Science and Technology Organisation (ANSTO) New Illawarra Road Lucas Heights NSW 2234 Australia
- Prince of Wales Hospital Barker St Randwick NSW 2031 Australia
- University of New South Wales Sydney (UNSW) Kensington NSW 2052 Australia
| | - Massimiliano Massi
- School of Molecular and Life Sciences Curtin University Kent Street Bentley WA 6102 Australia
| | - Benjamin H. Fraser
- Human Health and National Deuteration Facility (NDF) Australian Nuclear Science and Technology Organisation (ANSTO) New Illawarra Road Lucas Heights NSW 2234 Australia
| |
Collapse
|
7
|
Chen C, Yao Y, Wang W, Duan L, Zhang W, Qian J. Selective bioimaging of cancer cells and detection of HSA with indomethacin-based fluorescent probes. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2020; 241:118685. [PMID: 32653821 DOI: 10.1016/j.saa.2020.118685] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 06/25/2020] [Accepted: 07/03/2020] [Indexed: 06/11/2023]
Abstract
Two fluorescent probes were designed by connecting indomethacin to coumarin through different linkers. The introduction of indomethacin quenched the fluorescence of coumarin-based probes with apparent red-shifts in the absorption and emission maxima, probably due to the photoinduced electron transfer (PET) from the indomethacin to the fluorophore and the formation of folding conformation. The addition of human serum albumin (HSA) triggered about 40-fold fluorescence enhancements of ADC-IMC-2 and ADC-IMC-6 with 85 nm blue-shifts. The probe with longer spacer ADC-IMC-6 exhibited ratiometric fluorescent response toward HSA, and that with shorter linker showed "off-on" fluorescence response to HSA. However, insignificant spectral changes of the reference compounds (ADC-6 and ADC-2) initiated by HSA implied that indomethacin played critical role in the identification of HSA. The competitive assays and molecular docking results reveal that the indomethacin in ADC-IMC-6 could tightly combine at drug site I of HSA. Fluorescence bio-imaging experiments show that both probes could distinguish cancer cells from normal cells.
Collapse
Affiliation(s)
- Cong Chen
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry & Molecular Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Yuhua Yao
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry & Molecular Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Weisi Wang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai 200032, China
| | - Liping Duan
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai 200032, China.
| | - Weibing Zhang
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry & Molecular Engineering, East China University of Science and Technology, Shanghai 200237, China.
| | - Junhong Qian
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry & Molecular Engineering, East China University of Science and Technology, Shanghai 200237, China.
| |
Collapse
|
8
|
Focused ultrasound for opening blood-brain barrier and drug delivery monitored with positron emission tomography. J Control Release 2020; 324:303-316. [DOI: 10.1016/j.jconrel.2020.05.020] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 05/13/2020] [Accepted: 05/14/2020] [Indexed: 12/14/2022]
|
9
|
Dammes N, Peer D. Monoclonal antibody-based molecular imaging strategies and theranostic opportunities. Theranostics 2020; 10:938-955. [PMID: 31903161 PMCID: PMC6929980 DOI: 10.7150/thno.37443] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 09/26/2019] [Indexed: 01/13/2023] Open
Abstract
Molecular imaging modalities hold great potential as less invasive techniques for diagnosis and management of various diseases. Molecular imaging combines imaging agents with targeting moieties to specifically image diseased sites in the body. Monoclonal antibodies (mAbs) have become increasingly popular as novel therapeutics against a variety of diseases due to their specificity, affinity and serum stability. Because of the same properties, mAbs are also exploited in molecular imaging to target imaging agents such as radionuclides to the cell of interest in vivo. Many studies investigated the use of mAb-targeted imaging for a variety of purposes, for instance to monitor disease progression and to predict response to a specific therapeutic agent. Herein, we highlighted the application of mAb-targeted imaging in three different types of pathologies: autoimmune diseases, oncology and cardiovascular diseases. We also described the potential of molecular imaging strategies in theranostics and precision medicine. Due to the nearly infinite repertoire of mAbs, molecular imaging can change the future of modern medicine by revolutionizing diagnostics and response prediction in practically any disease.
Collapse
Affiliation(s)
- Niels Dammes
- Laboratory of Precision NanoMedicine, Tel Aviv University, Tel Aviv 69978, Israel
- School of Molecular Cell Biology and Biotechnology, George S Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, and Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Dan Peer
- Laboratory of Precision NanoMedicine, Tel Aviv University, Tel Aviv 69978, Israel
- School of Molecular Cell Biology and Biotechnology, George S Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, and Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
10
|
He M, Jiang Z, Wang C, Hao Z, An J, Shen J. Diagnostic value of near‐infrared or fluorescent indocyanine green guided sentinel lymph node mapping in gastric cancer: A systematic review and meta‐analysis. J Surg Oncol 2018; 118:1243-1256. [DOI: 10.1002/jso.25285] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 10/05/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Meifeng He
- Chengde Medical UniversityChengde Hebei China
| | - Zhanwu Jiang
- Baoding First Central HospitalBaoding Hebei China
| | | | - Zhiwei Hao
- Baoding First Central HospitalBaoding Hebei China
| | - Jie An
- Baoding First Central HospitalBaoding Hebei China
| | - Jiankai Shen
- Baoding First Central HospitalBaoding Hebei China
| |
Collapse
|
11
|
Zhu S, Yung BC, Chandra S, Niu G, Antaris AL, Chen X. Near-Infrared-II (NIR-II) Bioimaging via Off-Peak NIR-I Fluorescence Emission. Theranostics 2018; 8:4141-4151. [PMID: 30128042 PMCID: PMC6096392 DOI: 10.7150/thno.27995] [Citation(s) in RCA: 179] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 06/20/2018] [Indexed: 12/15/2022] Open
Abstract
Significantly reduced photon scattering and minimal tissue autofluorescence levels in the second biological transparency window (NIR-II; 1000-1700 nm) facilitate higher resolution in vivo biological imaging compared to tradition NIR fluorophores (~700-900 nm). However, the existing palette of NIR-II fluorescent agents including semiconducting inorganic nanomaterials and recently introduced small-molecule organic dyes face significant technical and regulatory hurdles prior to clinical translation. Fortunately, recent spectroscopic characterization of NIR-I dyes (e.g., indocyanine green (ICG), IRDye800CW and IR-12N3) revealed long non-negligible emission tails reaching past 1500 nm. Repurposing the most widely used NIR dye in medicine, in addition to those in the midst of clinical trials creates an accelerated pathway for NIR-II clinical translation. This review focuses on the significant advantage of imaging past 1000 nm with NIR-I fluorophores from both a basic and clinical viewpoint. We further discuss optimizing NIR-I dyes around their NIR-II/shortwave infrared (SWIR) emission, NIR-II emission tail characteristics and prospects of NIR-II imaging with clinically available and commercially available dyes.
Collapse
Affiliation(s)
- Shoujun Zhu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), 35A Convent Dr, Bethesda, Maryland 20892, United States
| | - Bryant C. Yung
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), 35A Convent Dr, Bethesda, Maryland 20892, United States
| | - Swati Chandra
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), 35A Convent Dr, Bethesda, Maryland 20892, United States
| | - Gang Niu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), 35A Convent Dr, Bethesda, Maryland 20892, United States
| | | | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), 35A Convent Dr, Bethesda, Maryland 20892, United States
| |
Collapse
|
12
|
Chen Q, Shang W, Zeng C, Wang K, Liang X, Chi C, Liang X, Yang J, Fang C, Tian J. Theranostic imaging of liver cancer using targeted optical/MRI dual-modal probes. Oncotarget 2018; 8:32741-32751. [PMID: 28416757 PMCID: PMC5464824 DOI: 10.18632/oncotarget.15642] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 12/05/2016] [Indexed: 12/15/2022] Open
Abstract
The accurate preoperative detection and intraoperative navigation afforded by imaging techniques have had significant impact on the success of liver cancer surgeries. However, it is difficult to achieve satisfactory performance in both diagnosis and surgical treatment processes using any single modality imaging method. Here, we report the synthesis and characteristics of a novel dual-modality magnetic resonance imaging (MRI) and near-infrared fluorescence (NIRF) probe and verify its feasibility in nude mouse models with liver cancer. The probes are comprised of superparamagnetic iron oxide (SPIO) nanoparticles coated with liposomes to which a tumor-targeted agent, Arg-Gly-Asp peptides (RGD), and a NIRF dye (indocyanine green, ICG) have been conjugated. Specific targeting, biodistribution, and the imaging ability of the probes for MRI-NIRF were examined. Furthermore, we applied the dual-modality methodology toward the preoperative diagnosis and intraoperative guidance of radical resection in mouse models with both orthotopic liver tumors and intrahepatic tumor metastasis. The study demonstrated that both MRI and fluorescent images showed clear tumor delineation after probe injection (SPIO@Liposome-ICG-RGD). The contrast-to-noise ratio obtained from MRI was 31.9 ± 25.4 at post-injection for the preoperative diagnosis, which is helpful for detecting small tumors (0.9 ± 0.5 mm). The maximum tumor to background ratio of NIRF imaging was 2.5 ± 0.3 at 72 h post-injection for effectively capturing miniscule tumor lesions (0.6 ± 0.3 mm) intraoperatively. The novel MRI-NIRF dual modality probes are promising for the achievement of more accurate liver tumor detection and resection.
Collapse
Affiliation(s)
- Qingshan Chen
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.,Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Wenting Shang
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China.,Beijing Key Laboratory of Molecular Imaging, Beijing 100190, China
| | - Chaoting Zeng
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.,Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Kun Wang
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China.,Beijing Key Laboratory of Molecular Imaging, Beijing 100190, China
| | - Xiaoyuan Liang
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China.,Beijing Key Laboratory of Molecular Imaging, Beijing 100190, China
| | - Chongwei Chi
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China.,Beijing Key Laboratory of Molecular Imaging, Beijing 100190, China
| | - Xiao Liang
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China.,Beijing Key Laboratory of Molecular Imaging, Beijing 100190, China
| | - Jian Yang
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.,Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Chihua Fang
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Jie Tian
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China.,Beijing Key Laboratory of Molecular Imaging, Beijing 100190, China
| |
Collapse
|
13
|
NIRF Optical/PET Dual-Modal Imaging of Hepatocellular Carcinoma Using Heptamethine Carbocyanine Dye. CONTRAST MEDIA & MOLECULAR IMAGING 2018; 2018:4979746. [PMID: 29706843 PMCID: PMC5863326 DOI: 10.1155/2018/4979746] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 01/13/2018] [Accepted: 02/05/2018] [Indexed: 12/17/2022]
Abstract
Combining near-infrared fluorescence (NIRF) and nuclear imaging techniques provides a novel approach for hepatocellular carcinoma (HCC) diagnosis. Here, we report the synthesis and characteristics of a dual-modality NIRF optical/positron emission tomography (PET) imaging probe using heptamethine carbocyanine dye and verify its feasibility in both nude mice and rabbits with orthotopic xenograft liver cancer. This dye, MHI-148, is an effective cancer-specific NIRF imaging agent and shows preferential uptake and retention in liver cancer. The corresponding NIRF imaging intensity reaches 109/cm2 tumor area at 24 h after injection in mice with HCC subcutaneous tumors. The dye can be further conjugated with radionuclide 68Ga (68Ga-MHI-148) for PET tracing. We applied the dual-modality methodology toward the detection of HCC in both patient-derived orthotopic xenograft (PDX) models and rabbit orthotopic transplantation models. NIRF/PET images showed clear tumor delineation after probe injection (MHI-148 and 68Ga-MHI-148). The tumor-to-muscle (T/M) standardized uptake value (SUV) ratios were obtained from PET at 1 h after injection of 68Ga-MHI-148, which was helpful for effectively capturing small tumors in mice (0.5 cm × 0.3 cm) and rabbits (1.2 cm × 1.8 cm). This cancer-targeting NIRF/PET dual-modality imaging probe provides a proof of principle for noninvasive detection of deep-tissue tumors in mouse and rabbit and is a promising technique for more accurate and early detection of HCC.
Collapse
|
14
|
Liu D, Liu X, Zhang Y, Wang Q, Lu J, Sun J. Imitation-tumor targeting based on continuous-wave near-infrared tomography. Comput Assist Surg (Abingdon) 2017; 22:157-162. [PMID: 29041839 DOI: 10.1080/24699322.2017.1389393] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Continuous-wave Near-Infrared (NIR) optical spectroscopy has shown great diagnostic capability in the early tumor detection with advantages of low-cost, portable, non-invasive, and non-radiative. In this paper, Modified Lambert-Beer Theory is deployed to address the low-resolution issues of the NIR technique and to design the tumor detecting and imaging system. Considering that tumor tissues have features such as high blood flow and hypoxia, the proposed technique can detect the location, size, and other information of the tumor tissues by comparing the absorbance between pathological and normal tissues. Finally, the tumor tissues can be imaged through tomographic method. The simulation experiments prove that the proposed technique and designed system can efficiently detect the tumor tissues, achieving imaging precision within 1 mm. The work of the paper has shown great potential in the diagnosis of tumor close to body surface.
Collapse
Affiliation(s)
- Dan Liu
- a School of Electrical Engineering & Automation , Harbin Institute of Technology , Harbin , China
| | - Xin Liu
- b School of Transportation Science and Engineering , Harbin Institute of Technology , Harbin , China
| | - Yan Zhang
- a School of Electrical Engineering & Automation , Harbin Institute of Technology , Harbin , China
| | - Qisong Wang
- a School of Electrical Engineering & Automation , Harbin Institute of Technology , Harbin , China
| | - Jingyang Lu
- c Intelligent Fusion Technology, Inc , Germantown , MD , USA
| | - Jinwei Sun
- a School of Electrical Engineering & Automation , Harbin Institute of Technology , Harbin , China
| |
Collapse
|
15
|
Bugby SL, Lees JE, Perkins AC. Hybrid intraoperative imaging techniques in radioguided surgery: present clinical applications and future outlook. Clin Transl Imaging 2017; 5:323-341. [PMID: 28804703 PMCID: PMC5532406 DOI: 10.1007/s40336-017-0235-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/10/2017] [Indexed: 12/26/2022]
Abstract
PURPOSE This review aims to summarise the hybrid modality radioguidance techniques currently in clinical use and development, and to discuss possible future avenues of research. Due to the novelty of these approaches, evidence of their clinical relevance does not yet exist. The purpose of this review is to inform nuclear medicine practitioners of current cutting edge research in radioguided surgery which may enter standard clinical practice within the next 5-10 years. Hybrid imaging is of growing importance to nuclear medicine diagnostics, but it is only with recent advances in technology that hybrid modalities are being investigated for use during radioguided surgery. These modalities aim to overcome some of the difficulties of surgical imaging while maintaining many benefits, or providing entirely new information unavailable to surgeons with traditional radioguidance. METHODS A literature review was carried out using online reference databases (Scopus, PubMed). Review articles obtained using this technique were citation mined to obtain further references. RESULTS In total, 2367 papers were returned, with 425 suitable for further assessment. 60 papers directly related to hybrid intraoperative imaging in radioguided surgery are reported on. Of these papers, 25 described the clinical use of hybrid imaging, 22 described the development of new hybrid probes and tracers, and 13 described the development of hybrid technologies for future clinical use. Hybrid gamma-NIR fluorescence was found to be the most common clinical technique, with 35 papers associated with these modalities. Other hybrid combinations include gamma-bright field imaging, gamma-ultrasound imaging, gamma-β imaging and β-OCT imaging. The combination of preoperative and intraoperative images is also discussed. CONCLUSION Hybrid imaging offers new possibilities for assisting clinicians and surgeons in localising the site of uptake in procedures such as in sentinel node detection.
Collapse
Affiliation(s)
- S L Bugby
- Space Research Centre, Michael Atiyah Building, University of Leicester, Leicester, LE1 7RH UK
| | - J E Lees
- Space Research Centre, Michael Atiyah Building, University of Leicester, Leicester, LE1 7RH UK
| | - A C Perkins
- Radiological Sciences, Division of Clinical Neuroscience, School of Medical, University of Nottingham, Nottingham, NG7 2UH UK.,Medical Physics and Clinical Engineering, Nottingham University Hospitals NHS Trust, Nottingham, NH7 2UH UK
| |
Collapse
|
16
|
Radioiodination of BODIPY and its application to a nuclear and optical dual functional labeling agent for proteins and peptides. Sci Rep 2017; 7:3337. [PMID: 28611426 PMCID: PMC5469783 DOI: 10.1038/s41598-017-03419-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 04/28/2017] [Indexed: 11/20/2022] Open
Abstract
In molecular imaging research, the development of multimodal imaging probes has recently attracted much attention. In the present study, we prepared radioiodinated BODIPY and applied it as a nuclear and optical dual functional labeling agent for proteins and peptides. We designed and synthesized [125I]BODIPY with a N-hydroxysuccinimide (NHS) ester, and evaluated its utility as a nuclear and fluorescent dual labeling agent for proteins and peptides. In the radioiodination reaction of BODIPY-NHS with [125I]NaI, [125I]BODIPY-NHS was obtained at a 48% radiochemical yield. When we carried out the conjugation reaction of [125I]BODIPY-NHS with bovine serum albumin (BSA) and RGD (Arg-Gly-Asp) peptide as a model protein and peptide, respectively, [125I]BODIPY-BSA and [125I]BODIPY-RGD peptide were successfully prepared at 98 and 82% radiochemical yields, respectively. Furthermore, we prepared [123I]BODIPY-trastuzumab by this conjugation reaction and successfully applied it to single photon emission computed tomography (SPECT) imaging studies using tumor-bearing mice, suggesting that radioiodinated BODIPY-NHS serves as a dual functional labeling agent for proteins and peptides. Since iodine has various radioisotopes that can be used for SPECT and positron emission tomography (PET) imaging, biological research, and radiotherapy, the radioiodinated BODIPY may be extensively applicable from basic to clinical research.
Collapse
|
17
|
Hu Z, Chi C, Liu M, Guo H, Zhang Z, Zeng C, Ye J, Wang J, Tian J, Yang W, Xu W. Nanoparticle-mediated radiopharmaceutical-excited fluorescence molecular imaging allows precise image-guided tumor-removal surgery. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 13:1323-1331. [PMID: 28115248 DOI: 10.1016/j.nano.2017.01.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 12/29/2016] [Accepted: 01/02/2017] [Indexed: 01/16/2023]
Abstract
Fluorescent molecular imaging technique has been actively explored for optical image-guided cancer surgery in pre-clinical and clinical research and has attracted many attentions. However, the efficacy of the fluorescent image-guided cancer surgery can be compromised by the low signal-to-noise ratio caused by the external light excitation. This study presents a novel nanoparticle-mediated radiopharmaceutical-excited fluorescent (REF) image-guided cancer surgery strategy, which employs the internal dual-excitation of europium oxide nanoparticles through both gamma rays and Cerenkov luminescence emitted from radiopharmaceuticals. The performance of the novel image-guided surgery technique was systematically evaluated using subcutaneous breast cancer 4 T1 tumor models, orthotropic and orthotropic-ectopic hepatocellular carcinoma tumor-bearing mice. The results reveal that the novel REF image-guided cancer surgery technique exhibits high performance of detecting invisible ultra-small size tumor (even less than 1 mm) and residual tumor tissue. Our study demonstrates the high potential of the novel image-guided cancer surgery for precise tumor resection.
Collapse
Affiliation(s)
- Zhenhua Hu
- Key Laboratory of Molecular Imaging of Chinese Academy of Sciences, Institute of Automation, Chinese Academy of Sciences, Beijing, China; The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, China.
| | - Chongwei Chi
- Key Laboratory of Molecular Imaging of Chinese Academy of Sciences, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Muhan Liu
- Key Laboratory of Molecular Imaging of Chinese Academy of Sciences, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Hongbo Guo
- Key Laboratory of Molecular Imaging of Chinese Academy of Sciences, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Zeyu Zhang
- Key Laboratory of Molecular Imaging of Chinese Academy of Sciences, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Chaoting Zeng
- Key Laboratory of Molecular Imaging of Chinese Academy of Sciences, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Jinzuo Ye
- Key Laboratory of Molecular Imaging of Chinese Academy of Sciences, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Jing Wang
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jie Tian
- Key Laboratory of Molecular Imaging of Chinese Academy of Sciences, Institute of Automation, Chinese Academy of Sciences, Beijing, China; The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, China.
| | - Weidong Yang
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| | - Wanhai Xu
- Department of Urinary Surgery, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
18
|
Adumeau P, Carnazza KE, Brand C, Carlin SD, Reiner T, Agnew BJ, Lewis JS, Zeglis BM. A Pretargeted Approach for the Multimodal PET/NIRF Imaging of Colorectal Cancer. Theranostics 2016; 6:2267-2277. [PMID: 27924162 PMCID: PMC5135447 DOI: 10.7150/thno.16744] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 09/10/2016] [Indexed: 01/15/2023] Open
Abstract
The complementary nature of positron emission tomography (PET) and near-infrared fluorescence (NIRF) imaging makes the development of strategies for the multimodal PET/NIRF imaging of cancer a very enticing prospect. Indeed, in the context of colorectal cancer, a single multimodal PET/NIRF imaging agent could be used to stage the disease, identify candidates for surgical intervention, and facilitate the image-guided resection of the disease. While antibodies have proven to be highly effective vectors for the delivery of radioisotopes and fluorophores to malignant tissues, the use of radioimmunoconjugates labeled with long-lived nuclides such as 89Zr poses two important clinical complications: high radiation doses to the patient and the need for significant lag time between imaging and surgery. In vivo pretargeting strategies that decouple the targeting vector from the radioactivity at the time of injection have the potential to circumvent these issues by facilitating the use of positron-emitting radioisotopes with far shorter half-lives. Here, we report the synthesis, characterization, and in vivo validation of a pretargeted strategy for the multimodal PET and NIRF imaging of colorectal carcinoma. This approach is based on the rapid and bioorthogonal ligation between a trans-cyclooctene- and fluorophore-bearing immunoconjugate of the huA33 antibody (huA33-Dye800-TCO) and a 64Cu-labeled tetrazine radioligand (64Cu-Tz-SarAr). In vivo imaging experiments in mice bearing A33 antigen-expressing SW1222 colorectal cancer xenografts clearly demonstrate that this approach enables the non-invasive visualization of tumors and the image-guided resection of malignant tissue, all at only a fraction of the radiation dose created by a directly labeled radioimmunoconjugate. Additional in vivo experiments in peritoneal and patient-derived xenograft models of colorectal carcinoma reinforce the efficacy of this methodology and underscore its potential as an innovative and useful clinical tool.
Collapse
|
19
|
Liu D, Liu X, Zhang Y, Wang Q, Lu J. Tissue phantom-based breast cancer detection using continuous near-infrared sensor. Bioengineered 2016; 7:321-326. [PMID: 27459672 DOI: 10.1080/21655979.2016.1197747] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Women's health is seriously threatened by breast cancer. Taking advantage of efficient diagnostic instruments to identify the disease is very meaningful in prolonging life. As a cheap noninvasive radiation-free technology, Near-infrared Spectroscopy is suitable for general breast cancer examination. A discrimination method of breast cancer is presented using the deference between absorption coefficients and applied to construct a blood oxygen detection device based on Modified Lambert-Beer theory. Combined with multi-wavelength multi-path near-infrared sensing technology, the proposed method can quantitatively distinguish the normal breast from the abnormal one by measuring the absorption coefficients of breast tissue and the blood oxygen saturation. An objective judgment about the breast tumor is made according to its high absorption of near-infrared light. The phantom experiment is implemented to show the presented method is able to recognize the absorption differences between phantoms and demonstrates its feasibility in the breast tumor detection.
Collapse
Affiliation(s)
- Dan Liu
- a Harbin Institute of Technology, School of Electrical Engineering & Automation , Harbin , China
| | - Xin Liu
- a Harbin Institute of Technology, School of Electrical Engineering & Automation , Harbin , China
| | - Yan Zhang
- a Harbin Institute of Technology, School of Electrical Engineering & Automation , Harbin , China
| | - Qisong Wang
- a Harbin Institute of Technology, School of Electrical Engineering & Automation , Harbin , China
| | - Jingyang Lu
- b Virginia Commonwealth University , Department of Electrical and Computer Engineering , Richmond , Virginia , USA
| |
Collapse
|
20
|
Wang X, Aldrich MB, Yang Z, Zhou N, Xie Q, Liu C, Sevick-Muraca E. Influence of chelator and near-infrared dye labeling on biocharacteristics of dual-labeled trastuzumab-based imaging agents. Chin J Cancer Res 2016; 28:362-9. [PMID: 27478322 PMCID: PMC4949282 DOI: 10.21147/j.issn.1000-9604.2016.03.11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Objective To investigate the effect of fluorescent dye labeling on the targeting capabilities of 111In-
(DTPA)n-trastuzumab-(IRDye 800)m. Methods Trastuzumab-based conjugates were synthesized and conjugated with diethylenetriaminepentaacetic acid (DTPA) at molar ratios of 1, 2, 3 and 5 and with a fluorescent dye (IRDye 800CW) at molar ratios of 1, 3 and 5. Immunoreactivity and internalization were assessed on SKBR-3 cells, overexpressing human epidermal growth factor receptor 2. The stability in human serum and phosphate-buffered saline (PBS) was evaluated. The biodistribution of dual-labeled conjugates was compared with that of 111In-(DTPA)2-trastuzumab in a SKBR-3 xenograft model to evaluate the effect of dye-to-protein ratio. Results All trastuzumab-based conjugates exhibited a high level of chemical and optical purity. Flow cytometry results showed that increasing dye-to-protein ratios were associated with decreased immunoreactivity. Stability studies revealed that the conjugate was stable in PBS, while in human serum, increased degradation and protein precipitation were observed with increasing dye-to-protein ratios. At 4 h, the percentages of internalization of dual-labeled conjugates normalized by dye-to-protein ratio (m) were 24.88%±2.10%, 19.99%±0.59%, and 17.47%±1.26% for "m" equal to 1, 3, and 5, respectively. A biodistribution study revealed a progressive decrease in tumor uptake with an increase in the dye-to-protein ratios. The liver, spleen and kidney showed a marked uptake with increased dye-to-protein ratios, particularly in the latter. Conclusions With non-specific-site conjugation of the fluorescent dye with a protein based on imaging agent, the increase in dye-to-protein ratios negatively impacted the immunoreactivity and stability, indicating a reduced tumor uptake.
Collapse
Affiliation(s)
- Xuejuan Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Melissa B Aldrich
- Center for Molecular Imaging, the Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston TX 77030, USA
| | - Zhi Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Nina Zhou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Qing Xie
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Chen Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Eva Sevick-Muraca
- Center for Molecular Imaging, the Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston TX 77030, USA
| |
Collapse
|
21
|
Dual PET and Near-Infrared Fluorescence Imaging Probes as Tools for Imaging in Oncology. AJR Am J Roentgenol 2016; 207:266-73. [PMID: 27223168 DOI: 10.2214/ajr.16.16181] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
OBJECTIVE The purpose of this article is to summarize advances in PET fluorescence resolution, agent design, and preclinical imaging that make a growing case for clinical PET fluorescence imaging. CONCLUSION Existing SPECT, PET, fluorescence, and MRI contrast imaging techniques are already deeply integrated into the management of cancer, from initial diagnosis to the observation and management of metastases. Combined positron-emitting fluorescent contrast agents can convey new or substantial benefits that improve on these proven clinical contrast agents.
Collapse
|
22
|
Hekman MCH, Boerman OC, de Weijert M, Bos DL, Oosterwijk E, Langenhuijsen HF, Mulders PFA, Rijpkema M. Targeted Dual-Modality Imaging in Renal Cell Carcinoma: An Ex Vivo Kidney Perfusion Study. Clin Cancer Res 2016; 22:4634-42. [PMID: 27103404 DOI: 10.1158/1078-0432.ccr-15-2937] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 04/12/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE Antibodies labeled with both a near-infrared fluorescent dye and a radionuclide can be used for tumor-targeted intraoperative dual-modality imaging. Girentuximab is a chimeric monoclonal antibody against carbonic anhydrase IX (CAIX), an antigen expressed in 95% of clear cell renal cell carcinoma (ccRCC). This study aimed to assess the feasibility of targeted dual-modality imaging with (111)In-girentuximab-IRDye800CW using ex vivo perfusion of human tumorous kidneys. EXPERIMENTAL DESIGN Seven radical nephrectomy specimens from patients with ccRCC were perfused during 11 to 15 hours with dual-labeled girentuximab and subsequently rinsed during 2.5 to 4 hours with Ringer's Lactate solution. Then, dual-modality imaging was performed on a 5- to 10-mm-thick lamella of the kidney. Fluorescence imaging was performed with a clinical fluorescence camera set-up as applied during image-guided surgery. The distribution of Indium-111 in the slice of tumor tissue was visualized by autoradiography. In two perfusions, an additional dual-labeled control antibody was added to demonstrate specific accumulation of dual-labeled girentuximab in CAIX-expressing tumor tissue. RESULTS Both radionuclide and fluorescence imaging clearly visualized uptake in tumor tissue and tumor-to-normal tissue borders, as confirmed (immuno)histochemically and by gamma counting. Maximum uptake of girentuximab in tumor tissue was 0.33% of the injected dose per gram (mean, 0.12 %ID/g; range, 0.01-0.33 %ID/g), whereas maximum uptake in the normal kidney tissue was 0.04 %ID/g (mean, 0.02 %ID/g; range, 0.00-0.04 %ID/g). CONCLUSIONS Dual-labeled girentuximab accumulated specifically in ccRCC tissue, indicating the feasibility of dual-modality imaging to detect ccRCC. A clinical study to evaluate intraoperative dual-modality imaging in patients with ccRCC has been initiated. Clin Cancer Res; 22(18); 4634-42. ©2016 AACR.
Collapse
Affiliation(s)
- Marlène C H Hekman
- Department of Radiology and Nuclear Medicine, Radboudumc, Nijmegen, the Netherlands. Department of Urology, Radboudumc, Nijmegen, the Netherlands.
| | - Otto C Boerman
- Department of Radiology and Nuclear Medicine, Radboudumc, Nijmegen, the Netherlands
| | | | - Desirée L Bos
- Department of Radiology and Nuclear Medicine, Radboudumc, Nijmegen, the Netherlands
| | | | | | | | - Mark Rijpkema
- Department of Radiology and Nuclear Medicine, Radboudumc, Nijmegen, the Netherlands
| |
Collapse
|
23
|
Cisneros BT, Law JJ, Matson ML, Azhdarinia A, Sevick-Muraca EM, Wilson LJ. Stable confinement of positron emission tomography and magnetic resonance agents within carbon nanotubes for bimodal imaging. Nanomedicine (Lond) 2015; 9:2499-509. [PMID: 24628687 DOI: 10.2217/nnm.14.26] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
AIMS Simultaneous positron emission tomography/MRI has recently been introduced to the clinic and dual positron emission tomography/MRI probes are rare and of growing interest. We have developed a strategy for producing multimodal probes based on a carbon nanotube platform without the use of chelating ligands. MATERIALS & METHODS Gd(3+) and (64)Cu(2+) ions were loaded into ultra-short single-walled carbon nanotubes by sonication. Normal, tumor-free athymic nude mice were injected intravenously with the probe and imaged over 48 h. RESULTS & CONCLUSION The probe was stable for up to 24 h when challenged with phosphate-buffered saline and mouse serum. Positron emission tomography imaging also confirmed the stability of the probe in vivo for up to 48 h. The probe was quickly cleared from circulation, with enhanced accumulation in the lungs. Stable encapsulation of contrast agents within ultra-short single-walled carbon nanotubes represents a new strategy for the design of advanced imaging probes with variable multimodal imaging capabilities.
Collapse
Affiliation(s)
- Brandon T Cisneros
- Department of Chemistry & Richard E Smalley Institute for Nanoscale Science & Technology, Rice University, 1900 Rice Blvd, Houston, TX 77005, USA
| | | | | | | | | | | |
Collapse
|
24
|
Zelken JA, Tufaro AP. Current Trends and Emerging Future of Indocyanine Green Usage in Surgery and Oncology: An Update. Ann Surg Oncol 2015; 22 Suppl 3:S1271-83. [PMID: 26193966 DOI: 10.1245/s10434-015-4743-5] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Indexed: 12/21/2022]
Abstract
BACKGROUND Indocyanine green (ICG) is a widely available dye of clinical importance that has been used for more than 50 years. Near-infrared (NIR) ICG fluorescence imaging has found a niche in cancer care since 2005, and was reviewed in 2011. There is a need for a comprehensive update and we aim to provide this through a review of the most recent literature. METHODS A systematic review of the literature using PubMed, EMBASE, and MEDLINE databases of articles published from 2000 to June 2015 evaluated topics pertinent to NIR fluorescence imaging with ICG in the diagnosis and surgical treatment of cancer. Articles previously referenced in a 2011 review and a 2015 meta-analysis were excluded, while articles that referenced future directions and economics were included in this current review. RESULTS Since 2011, the literature has grown exponentially, with significant advances at the molecular level. Significant findings from 89 select articles and 10 reviews, most of which were published between 2011 and 2015, are summarized. Preclinical studies are currently underway investigating tumor-specific fluorescence and targeted therapeutic delivery. The potential for ICG exists at every level of cancer care, from diagnosis to surveillance. CONCLUSION The indications, applications, and potential for ICG have grown exponentially in the past decade; an updated review of the literature is overdue and we present the most comprehensive review to date.
Collapse
Affiliation(s)
- Jonathan A Zelken
- Finesse Plastic Surgery, Orange, CA, USA. .,Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan.
| | - Anthony P Tufaro
- Department of Plastic and Reconstructive Surgery, The Johns Hopkins Hospital, Baltimore, MD, USA.,Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, MD, USA
| |
Collapse
|
25
|
Rijpkema M, Bos DL, Cornelissen AS, Franssen GM, Goldenberg DM, Oyen WJ, Boerman OC. Optimization of Dual-Labeled Antibodies for Targeted Intraoperative Imaging of Tumors. Mol Imaging 2015. [DOI: 10.2310/7290.2015.00015] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Mark Rijpkema
- From the Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, the Netherlands; Immunomedics, Inc., Morris Plains, NJ; and Garden State Cancer Center, Center for Molecular Medicine and Immunology, Morris Plains, NJ
| | - Desirée L. Bos
- From the Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, the Netherlands; Immunomedics, Inc., Morris Plains, NJ; and Garden State Cancer Center, Center for Molecular Medicine and Immunology, Morris Plains, NJ
| | - Alex S. Cornelissen
- From the Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, the Netherlands; Immunomedics, Inc., Morris Plains, NJ; and Garden State Cancer Center, Center for Molecular Medicine and Immunology, Morris Plains, NJ
| | - Gerben M. Franssen
- From the Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, the Netherlands; Immunomedics, Inc., Morris Plains, NJ; and Garden State Cancer Center, Center for Molecular Medicine and Immunology, Morris Plains, NJ
| | - David M. Goldenberg
- From the Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, the Netherlands; Immunomedics, Inc., Morris Plains, NJ; and Garden State Cancer Center, Center for Molecular Medicine and Immunology, Morris Plains, NJ
| | - Wim J. Oyen
- From the Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, the Netherlands; Immunomedics, Inc., Morris Plains, NJ; and Garden State Cancer Center, Center for Molecular Medicine and Immunology, Morris Plains, NJ
| | - Otto C. Boerman
- From the Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, the Netherlands; Immunomedics, Inc., Morris Plains, NJ; and Garden State Cancer Center, Center for Molecular Medicine and Immunology, Morris Plains, NJ
| |
Collapse
|
26
|
Xi L, Jiang H. Image-guided surgery using multimodality strategy and molecular probes. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2015; 8:46-60. [PMID: 26053199 DOI: 10.1002/wnan.1352] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 03/23/2015] [Accepted: 04/19/2015] [Indexed: 12/15/2022]
Abstract
The ultimate goal of cancer surgery is to maximize the excision of tumorous tissue with minimal damage to the collateral normal tissues, reduce the postoperative recurrence, and improve the survival rate of patients. In order to locate tumor lesions, highlight tumor margins, visualize residual disease in the surgical wound, and map potential lymph node metastasis, various imaging techniques and molecular probes have been investigated to assist surgeons to perform more complete tumor resection. Combining imaging techniques with molecular probes is particularly promising as a new approach for image-guided surgery. Considering inherent limitations of different imaging techniques and insufficient sensitivity of nonspecific molecular probes, image-guided surgery with multimodality strategy and specific molecular probes appears to be an optimal choice. In this article, we briefly describe typical imaging techniques and molecular probes followed by a focused review on the current progress of multimodal image-guided surgery with specific molecular navigation. We also discuss optimal strategy that covers all stages of image-guided surgery including preoperative scanning of tumors, intraoperative inspection of surgical bed and postoperative care of patients.
Collapse
Affiliation(s)
- Lei Xi
- School of Physical Electronics, University of Electronic Science and Technology of China, Chengdu, China
| | - Hubei Jiang
- School of Physical Electronics, University of Electronic Science and Technology of China, Chengdu, China.,Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| |
Collapse
|
27
|
Ogawa R, Watanabe H, Yazaki K, Fujita K, Tsunoda Y, Nakazawa K, Homma S, Kagohashi K, Satoh H, Hizawa N. Lung cancer with spontaneous regression of primary and metastatic sites: A case report. Oncol Lett 2015; 10:550-552. [PMID: 26171067 DOI: 10.3892/ol.2015.3243] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 04/30/2015] [Indexed: 12/15/2022] Open
Abstract
Partial or complete spontaneous cancer regression is a rare phenomenon, particularly in patients with lung cancer. This is the case report of a patient with lung cancer who exhibited spontaneous regression of the primary as well as metastatic lesions, without receiving any treatment. Spontaneous regression commenced within a week of obtaining pathological specimens by transbronchial and percutaneous biopsies from the primary lesion and metastatic lymph nodes of the left side of the neck. The reason for this phenomenon is unknown; however, we hypothesized that there may be an immunological association between the stimulus of the biopsies and the spontaneous regression. This patient should be closely followed up to monitor the clinical course of this unusual case.
Collapse
Affiliation(s)
- Ryoko Ogawa
- Division of Respiratory Medicine, Faculty of Medicine, University of Tsukuba, Mito, Ibaraki 310-0015, Japan
| | - Hiroko Watanabe
- Division of Respiratory Medicine, Faculty of Medicine, University of Tsukuba, Mito, Ibaraki 310-0015, Japan
| | - Kai Yazaki
- Division of Respiratory Medicine, Faculty of Medicine, University of Tsukuba, Mito, Ibaraki 310-0015, Japan
| | - Kazutaka Fujita
- Division of Respiratory Medicine, Faculty of Medicine, University of Tsukuba, Mito, Ibaraki 310-0015, Japan
| | - Yoshiya Tsunoda
- Division of Respiratory Medicine, Faculty of Medicine, University of Tsukuba, Mito, Ibaraki 310-0015, Japan
| | - Kensuke Nakazawa
- Division of Respiratory Medicine, Faculty of Medicine, University of Tsukuba, Mito, Ibaraki 310-0015, Japan
| | - Shinsuke Homma
- Division of Respiratory Medicine, Faculty of Medicine, University of Tsukuba, Mito, Ibaraki 310-0015, Japan
| | - Katsunori Kagohashi
- Division of Respiratory Medicine, Mito Medical Center, University of Tsukuba, Mito, Ibaraki 310-0015, Japan
| | - Hiroaki Satoh
- Division of Respiratory Medicine, Mito Medical Center, University of Tsukuba, Mito, Ibaraki 310-0015, Japan
| | - Nobuyuki Hizawa
- Division of Respiratory Medicine, Faculty of Medicine, University of Tsukuba, Mito, Ibaraki 310-0015, Japan
| |
Collapse
|
28
|
Wang X, Aldrich MB, Marshall MV, Sevick-Muraca EM. Preclinical characterization and validation of a dual-labeled trastuzumab-based imaging agent for diagnosing breast cancer. Chin J Cancer Res 2015; 27:74-82. [PMID: 25717229 DOI: 10.3978/j.issn.1000-9604.2015.01.02] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 01/09/2015] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE The combination of both nuclear and fluorescent reporters provides unique opportunities for noninvasive nuclear imaging with subsequent fluorescence image-guided resection and pathology. Our objective was to synthesize and optimize a dual-labeled trastuzumab-based imaging agent that can be used to validate an optical imaging agent with potential use in identifying tumor metastases in human epidermal growth factor receptor 2 (HER2) positive breast cancer patients. METHODS [(111)In]-DTPA-trastuzumab-IRDye 800 was synthesized by a three-step procedure. Purity, stability, immunoreactivity, internalization and biodistribution were explored in HER2+ SKBR-3 cells. Biodistribution of [(111)In]-DTPA-trastuzumab-IRDye 800 was performed in a SKBR-3 xenograft model. RESULTS [(111)In]-DTPA-trastuzumab-IRDye 800 demonstrated high purity by both chemical and fluorometric determinations. Both flow cytometry and the Lindmo assay demonstrated a high binding affinity of [(111)In]-DTPA-trastuzumab-IRDye 800 to HER2-overexpressing cells. The dual-labeled conjugate was stable in PBS, but not in serum after 24 h at 37 °C. Larger molecules (>150 kD) were seen after a 24 h-incubation in human serum. Biodistribution studies revealed tumor-specific accumulation of [(111)In]-DTPA-trastuzumab-IRDye 800 in SKBR-3 tumors, and tumor uptakes at 24 and 48 h were (12.42±1.72)% and (9.96±1.05)%, respectively, following intravenous administration. The tumor-to-muscle ratio was 9.13±1.68 at 24 h, and increased to 12.79±2.13 at 48 h. Liver and kidney showed marked uptake of the dual-labeled imaging agent. CONCLUSIONS [(111)In]-DTPA-trastuzumab-IRDye 800 is an effective diagnostic biomarker that can be used to validate dual-labeled, molecularly targeted imaging agents and can allow these agents to be translated into clinical practice for identifying HER2+ lesions.
Collapse
Affiliation(s)
- Xuejuan Wang
- 1 Department of Nuclear Medicine, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing 100142, China ; 2 Division of Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Melissa B Aldrich
- 1 Department of Nuclear Medicine, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing 100142, China ; 2 Division of Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Milton V Marshall
- 1 Department of Nuclear Medicine, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing 100142, China ; 2 Division of Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Eva M Sevick-Muraca
- 1 Department of Nuclear Medicine, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing 100142, China ; 2 Division of Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
29
|
Yao N, Yan P, Wang RF, Zhang CL, Ma C, Chen XQ, Zhao Q, Hao P. Detection of pulmonary metastases with the novel radiolabeled molecular probe, (99m)Tc-RRL. Int J Clin Exp Med 2015; 8:1726-36. [PMID: 25932101 PMCID: PMC4402748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 02/12/2015] [Indexed: 06/04/2023]
Abstract
BACKGROUND To improve the detection of pulmonary metastases, experimental blood-borne pulmonary metastasis mouse models were established using three intravenously administered cell lines. In a previous study we demonstrated that (99m)Tc-radiolabeled arginine-arginine-leucine (RRL) could be used to non-invasively image malignant tumors. METHODS (99m)Tc-RRL was prepared and injected intravenously in mice with pulmonary metastases that arose from the intravenous injection of HepG2, B16, and Hela cells. The bio-distribution and imaging of (99m)Tc-RRL were determined in different pulmonary metastases mouse models and in normal mice. RESULTS (99m)Tc-RRL exhibited higher uptake values in the lungs of pulmonary metastatic mice compared to normal mice (P<0.05; 3.92±0.48% ID/g 2 h post-injection and 3.89±0.36% ID/g 4 h post-injection in metastatic hepatic carcinoma [HepG2]-bearing lungs; 5.49±0.84% ID/g 2 h post-injection and 5.11±0.75% ID/g 4 h post-injection in metastatic melanoma [B16]-bearing lungs; 3.72±0.52% ID/g 2 h post-injection and 3.51±0.35% ID/g 4 h post-injection in metastatic cervical carcinoma [Hela]-bearing lungs; 2.38±0.20% ID/g 2 h post-injection and 2.11±0.24% ID/g 4 h post-injection in normal lungs). The pulmonary metastatic lesions were clearly visualized using (99m)Tc-RRL. CONCLUSIONS (99m)Tc-RRL exhibited favorable metastatic tumor targeting and imaging properties, thus highlighting its potential as an effective imaging probe for detection of pulmonary metastases. (99m)Tc-RRL can be used as a reasonable supplement to (18)F-FDG imaging in the non-invasive imaging of tumor angiogenesis.
Collapse
Affiliation(s)
- Ning Yao
- Department of Nuclear Medicine, Peking University First HospitalWest District, Beijing 100034, China
| | - Ping Yan
- Department of Nuclear Medicine, Peking University First HospitalWest District, Beijing 100034, China
| | - Rong-Fu Wang
- Department of Nuclear Medicine, Peking University First HospitalWest District, Beijing 100034, China
| | - Chun-Li Zhang
- Department of Nuclear Medicine, Peking University First HospitalWest District, Beijing 100034, China
| | - Chao Ma
- Department of Nuclear Medicine, Peking University First HospitalWest District, Beijing 100034, China
| | - Xue-Qi Chen
- Department of Nuclear Medicine, Peking University First HospitalWest District, Beijing 100034, China
| | - Qian Zhao
- Department of Nuclear Medicine, General Hospital of Ningxia Medical UniversityXingqing District, Yinchuan 750004, Ningxia, China
| | - Pan Hao
- Department of Nuclear Medicine, Peking University First HospitalWest District, Beijing 100034, China
| |
Collapse
|
30
|
Lee ES, Kim TS, Kim SK. Current status of optical imaging for evaluating lymph nodes and lymphatic system. Korean J Radiol 2015; 16:21-31. [PMID: 25598672 PMCID: PMC4296273 DOI: 10.3348/kjr.2015.16.1.21] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2014] [Accepted: 07/30/2014] [Indexed: 12/26/2022] Open
Abstract
Optical imaging techniques use visual and near infrared rays. Despite their considerably poor penetration depth, they are widely used due to their safe and intuitive properties and potential for intraoperative usage. Optical imaging techniques have been actively investigated for clinical imaging of lymph nodes and lymphatic system. This article summarizes a variety of optical tracers and techniques used for lymph node and lymphatic imaging, and reviews their clinical applications. Emerging new optical imaging techniques and their potential are also described.
Collapse
Affiliation(s)
- Eun Seong Lee
- Department of Nuclear Medicine, Research Institute and Hospital, National Cancer Center, Goyang 410-769, Korea
| | - Tae Sung Kim
- Department of Nuclear Medicine, Research Institute and Hospital, National Cancer Center, Goyang 410-769, Korea
| | - Seok-Ki Kim
- Department of Nuclear Medicine, Research Institute and Hospital, National Cancer Center, Goyang 410-769, Korea
| |
Collapse
|
31
|
Pandya DN, Bhatt N, Dale AV, Kim JY, Lee H, Ha YS, Lee JE, An GI, Yoo J. New bifunctional chelator for 64Cu-immuno-positron emission tomography. Bioconjug Chem 2014; 24:1356-66. [PMID: 23883075 DOI: 10.1021/bc400192a] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
A new tetraazamacrocyclic bifunctional chelator, TE2A-Bn-NCS, was synthesized in high overall yield from cyclam. An extra functional group (NCS) was introduced to the N-atom of TE2A for specific conjugation with antibody. The Cu complex of TE2A-Bn-NCS showed high kinetic stability in acidic decomplexation and cyclic voltammetry studies. X-ray structure determination of the Cu-TE2A-Bn-NH2 complex confirmed octahedral geometry, in which copper atom is strongly coordinated by four macrocyclic nitrogens in equatorial positions and two carboxylate oxygen atoms occupy the elongated axial positions. Trastuzumab was conjugated with TE2A-Bn-NCS and then radiolabeled with 64Cu quantitatively at room temperature within 10 min. Biodistribution studies showed that the 64Cu-labeled TE2A-Bn-NCS-trastuzumab conjugates maintain high stability in physiological conditions, and NIH3T6.7 tumors were clearly visualized up to 3 days by 64Cu-immuno-positron emission tomography imaging in animal models.
Collapse
|
32
|
Zeglis BM, Davis CB, Abdel-Atti D, Carlin SD, Chen A, Aggeler R, Agnew BJ, Lewis JS. Chemoenzymatic strategy for the synthesis of site-specifically labeled immunoconjugates for multimodal PET and optical imaging. Bioconjug Chem 2014; 25:2123-8. [PMID: 25418333 PMCID: PMC4334285 DOI: 10.1021/bc500499h] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
![]()
The complementary nature of positron
emission tomography (PET) and optical imaging (OI) has fueled increasing
interest in the development of multimodal PET/OI probes that can be
employed during the diagnosis, staging, and surgical treatment of
cancer. Due to their high selectivity and affinity, antibodies have
emerged as promising platforms for the development of hybrid PET/OI
agents. However, the lack of specificity of many bioconjugation reactions
can threaten immunoreactivity and lead to poorly defined constructs.
To circumvent this issue, we have developed a chemoenzymatic strategy
for the construction of multimodal PET/OI immunoconjugates that have
been site-specifically labeled on the heavy chain glycans. The methodology
consists of four steps: (1) the enzymatic removal of the terminal
galactose residues on the heavy chain glycans; (2) the enzymatic incorporation
of azide-bearing galactose (GalNAz) residues into the heavy chain
glycans; (3) the strain-promoted click conjugation of chelator- and
fluorophore-modified dibenzocyclooctynes to the azide-modified sugars;
and (4) the radiolabeling of the immunoconjugate. For proof-of-concept,
a model system was created using the colorectal cancer-targeting antibody
huA33, the chelator desferrioxamine (DFO), the positron-emitting radiometal 89Zr, and the near-infrared fluorescent dye Alexa Fluor 680.
The bioconjugation strategy is robust and reproducible, reliably producing
well-defined and immunoreactive conjugates labeled with 89Zr, Alexa Fluor 680, or an easily and precisely tuned mixture of
the two reporters. In in vivo PET and fluorescence
imaging experiments, a hybrid 89Zr- and Alexa Fluor 680-labeled
huA33 conjugate displayed high levels of specific uptake (>45%
ID/g) in athymic nude mice bearing A33 antigen-expressing SW1222 colorectal
cancer xenografts.
Collapse
Affiliation(s)
- Brian M Zeglis
- Department of Radiology, Memorial Sloan Kettering Cancer Center , New York, New York 10065, United States
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Stephan H, Walther M, Fähnemann S, Ceroni P, Molloy JK, Bergamini G, Heisig F, Müller CE, Kraus W, Comba P. Bispidines for Dual Imaging. Chemistry 2014; 20:17011-8. [DOI: 10.1002/chem.201404086] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Indexed: 11/08/2022]
|
34
|
Bu L, Shen B, Cheng Z. Fluorescent imaging of cancerous tissues for targeted surgery. Adv Drug Deliv Rev 2014; 76:21-38. [PMID: 25064553 DOI: 10.1016/j.addr.2014.07.008] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Revised: 05/29/2014] [Accepted: 07/10/2014] [Indexed: 12/18/2022]
Abstract
To maximize tumor excision and minimize collateral damage are the primary goals of cancer surgery. Emerging molecular imaging techniques have made "image-guided surgery" developed into "molecular imaging-guided surgery", which is termed as "targeted surgery" in this review. Consequently, the precision of surgery can be advanced from tissue-scale to molecule-scale, enabling "targeted surgery" to be a component of "targeted therapy". Evidence from numerous experimental and clinical studies has demonstrated significant benefits of fluorescent imaging in targeted surgery with preoperative molecular diagnostic screening. Fluorescent imaging can help to improve intraoperative staging and enable more radical cytoreduction, detect obscure tumor lesions in special organs, highlight tumor margins, better map lymph node metastases, and identify important normal structures intraoperatively. Though limited tissue penetration of fluorescent imaging and tumor heterogeneity are two major hurdles for current targeted surgery, multimodality imaging and multiplex imaging may provide potential solutions to overcome these issues, respectively. Moreover, though many fluorescent imaging techniques and probes have been investigated, targeted surgery remains at a proof-of-principle stage. The impact of fluorescent imaging on cancer surgery will likely be realized through persistent interdisciplinary amalgamation of research in diverse fields.
Collapse
|
35
|
Brizet B, Goncalves V, Bernhard C, Harvey PD, Denat F, Goze C. DMAP-BODIPY Alkynes: A Convenient Tool for Labeling Biomolecules for Bimodal PET-Optical Imaging. Chemistry 2014; 20:12933-44. [DOI: 10.1002/chem.201402379] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Indexed: 11/11/2022]
|
36
|
Lütje S, Rijpkema M, Helfrich W, Oyen WJG, Boerman OC. Targeted Radionuclide and Fluorescence Dual-modality Imaging of Cancer: Preclinical Advances and Clinical Translation. Mol Imaging Biol 2014; 16:747-55. [DOI: 10.1007/s11307-014-0747-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
37
|
Bimodal imaging probes for combined PET and OI: recent developments and future directions for hybrid agent development. BIOMED RESEARCH INTERNATIONAL 2014; 2014:153741. [PMID: 24822177 PMCID: PMC4009187 DOI: 10.1155/2014/153741] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 03/18/2014] [Indexed: 12/02/2022]
Abstract
Molecular imaging—and especially positron emission tomography (PET)—has gained increasing importance for diagnosis of various diseases and thus experiences an increasing dissemination. Therefore, there is also a growing demand for highly affine PET tracers specifically accumulating and visualizing target structures in the human body. Beyond the development of agents suitable for PET alone, recent tendencies aim at the synthesis of bimodal imaging probes applicable in PET as well as optical imaging (OI), as this combination of modalities can provide clinical advantages. PET, due to the high tissue penetration of the γ-radiation emitted by PET nuclides, allows a quantitative imaging able to identify and visualize tumors and metastases in the whole body. OI on the contrary visualizes photons exhibiting only a limited tissue penetration but enables the identification of tumor margins and infected lymph nodes during surgery without bearing a radiation burden for the surgeon. Thus, there is an emerging interest in bimodal agents for PET and OI in order to exploit the potential of both imaging techniques for the imaging and treatment of tumor diseases. This short review summarizes the available hybrid probes developed for dual PET and OI and discusses future directions for hybrid agent development.
Collapse
|
38
|
Abstract
Passive protection, the administration of antibodies to prevent infection, has garnered significant interest in recent years as a potential prophylactic countermeasure to decrease the prevalence of hospital-acquired infections. Pili, polymerized protein structures covalently anchored to the peptidoglycan wall of many Gram-positive pathogens, are ideal targets for antibody intervention, given their importance in establishing infection and their accessibility to antibody interactions. In this work, we demonstrated that a monoclonal antibody to the major component of Enterococcus faecalis pili, EbpC, labels polymerized pilus structures, diminishes biofilm formation, and significantly prevents the establishment of a rat endocarditis infection. The effectiveness of this anti-EbpC monoclonal provides strong evidence in support of its potential as a preventative. In addition, after radiolabeling, this monoclonal identified the site of enterococcal infection, providing a rare example of molecularly specific imaging of an established bacterial infection and demonstrating the versatility of this agent for use in future diagnostic and therapeutic applications.
Collapse
|
39
|
Grootendorst DJ, Steenbergen W, Manohar S, Ruers TJM. Optical techniques for the intraoperative assessment of nodal status. Future Oncol 2013; 9:1741-55. [PMID: 24156334 DOI: 10.2217/fon.13.125] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The lymphatic system is an important pathway in the metastatic spread of many malignancies and a key prognostic indicator. Nondestructive assessment of the nodal status during surgery could limit the amount of lymph nodes that need to be resected and allow for immediate regional lymphadenectomy during sentinel lymph node biopsy procedures. This review looks into the possibilities of conventional medical imaging methods that are capable of intraoperative nodal assessment and discusses multiple newly developed optical techniques. The physical background behind these techniques is reviewed and a concise overview of their main advantages and disadvantages is provided. These recent innovations show that while the application of optical modalities for intraoperative nodal staging is not yet applied routinely, there is reason enough to expect their introduction in the near future.
Collapse
Affiliation(s)
- Diederik J Grootendorst
- Biomedical Photonic Imaging Group, MIRA Institute for Biomedical Technology & Technical Medicine, Science & Technology, University of Twente, PO Box 217, 7500 AE Enschede, The Netherlands
| | | | | | | |
Collapse
|
40
|
Inert coupling of IRDye800CW and zirconium-89 to monoclonal antibodies for single- or dual-mode fluorescence and PET imaging. Nat Protoc 2013; 8:1010-8. [PMID: 23619892 DOI: 10.1038/nprot.2013.054] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
IRDye800CW and zirconium-89 ((89)Zr) have very attractive properties for optical imaging and positron emission tomography (PET) imaging, respectively. Here we describe a procedure for dual labeling of mAbs with IRDye800CW and (89)Zr in a current good manufacturing practice (cGMP)-compliant way. IRDye800CW and (89)Zr are coupled inertly, without impairment of immunoreactivity and pharmacokinetics of the mAb. Organ and whole-body distribution of the final product can be assessed by optical and PET imaging, respectively. For this purpose, a minimal amount of the chelate N-succinyldesferrioxamine (N-sucDf) is first conjugated to the mAb. Next, N-sucDf-mAb is conjugated with IRDye800CW, after which the N-sucDf-mAb-IRDye800CW is labeled with (89)Zr. After each of these three steps, the product is purified by gel filtration. The sequence of this process avoids unnecessary radiation exposure to personnel and takes about 5 h. The process can be scaled up by the production of large batches of premodified mAbs that can be dispensed and stored until they are labeled with (89)Zr.
Collapse
|
41
|
Ghosh SC, Ghosh P, Wilganowski N, Robinson H, Hall MA, Dickinson G, Pinkston KL, Harvey BR, Sevick-Muraca EM, Azhdarinia A. Multimodal chelation platform for near-infrared fluorescence/nuclear imaging. J Med Chem 2013; 56:406-16. [PMID: 23214723 DOI: 10.1021/jm300906g] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Dual-labeled compounds containing nuclear and near-infrared fluorescence contrast have the potential to molecularly guide surgical resection of cancer by extending whole-body diagnostic imaging findings into the surgical suite. To simplify the dual labeling process for antibody-based agents, we designed a multimodality chelation (MMC) scaffold which combined a radiometal chelating agent and fluorescent dye into a single moiety. Three dye-derivatized MMC compounds were synthesized and radiolabeled. The IRDye 800CW conjugate, 4, had favorable optical properties and showed rapid clearance in vivo. Using 4, an epithelial cell adhesion molecule (EpCAM) targeting MMC-immunoconjugate was prepared and dual-labeled with (64)Cu. In vitro binding activity was confirmed after MMC conjugation. Multimodal imaging studies showed higher tumor accumulation of (64)Cu-7 compared to nontargeted (64)Cu-4 in a prostate cancer model. Further evaluation in different EpCAM-expressing cell lines is warranted as well as application of the MMC dual labeling approach with other monoclonal antibodies.
Collapse
Affiliation(s)
- Sukhen C Ghosh
- Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston , Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Sevick-Muraca EM, Akers WJ, Joshi BP, Luker GD, Cutler CS, Marnett LJ, Contag CH, Wang TD, Azhdarinia A. Advancing the translation of optical imaging agents for clinical imaging. BIOMEDICAL OPTICS EXPRESS 2013; 4:160-170. [PMID: 23304655 PMCID: PMC3539189 DOI: 10.1364/boe.4.000160] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Revised: 11/13/2012] [Accepted: 11/23/2012] [Indexed: 05/29/2023]
Abstract
Despite the development of a large number of promising candidates, few contrast agents for established medical imaging modalities have successfully been translated over the past decade. The emergence of new imaging contrast agents that employ biomedical optics is further complicated by the relative infancy of the field and the lack of approved imaging devices compared to more established clinical modalities such as nuclear medicine. Herein, we propose a navigational approach (as opposed to a fixed "roadmap") for translation of optical imaging agents that is (i) proposed through consensus by four academic research programs that are part of the cooperative U54 NCI Network for Translational Research, (ii) developed through early experiences for translating optical imaging agents in order to meet distinctly varied needs in cancer diagnostics, and (iii) adaptable to the rapidly changing environment of academic medicine. We describe the pathways by which optical imaging agents are synthesized, qualified, and validated for preclinical testing, and ultimately translated for "first-in-humans" studies using investigational optical imaging devices. By identifying and adopting consensus approaches for seemingly disparate optical imaging modalities and clinical indications, we seek to establish a systematic method for navigating the ever-changing "roadmap" to most efficiently arrive at the destination of clinical adoption and improved outcome and survivorship for cancer patients.
Collapse
Affiliation(s)
- Eva M. Sevick-Muraca
- The University of Texas Health Science Center at Houston, Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, 1825 Pressler Street, Houston, TX 77030, USA
| | - Walter J. Akers
- Washington University School of Medicine, Department of Radiology, St. Louis, MO 63110, USA
| | - Bishnu P. Joshi
- The University of Michigan, School of Medicine, Department of Internal-Medicine-Division of Gastroenterology, Ann Arbor, MI 48109, USA
| | - Gary D. Luker
- The University of Michigan, School of Medicine, Department of Internal-Medicine-Division of Gastroenterology, Ann Arbor, MI 48109, USA
| | - Cathy S. Cutler
- University of Missouri Research Reactor Center (MURR), Radiopharmaceutical Sciences Institute, Nuclear Engineering and Sciences Institute, Nuclear Engineering, Columbia, MO 65211, USA
| | | | - Christopher H. Contag
- Stanford University, School of Medicine, Department of Pediatrics, Stanford, CA 94305, USA
| | - Thomas D. Wang
- The University of Michigan, School of Medicine, Department of Internal-Medicine-Division of Gastroenterology, Ann Arbor, MI 48109, USA
| | - Ali Azhdarinia
- The University of Texas Health Science Center at Houston, Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, 1825 Pressler Street, Houston, TX 77030, USA
| |
Collapse
|
43
|
Tang L, Yang X, Dobrucki LW, Chaudhury I, Yin Q, Yao C, Lezmi S, Helferich WG, Fan TM, Cheng J. Aptamer-functionalized, ultra-small, monodisperse silica nanoconjugates for targeted dual-modal imaging of lymph nodes with metastatic tumors. Angew Chem Int Ed Engl 2012; 51:12721-6. [PMID: 23136130 PMCID: PMC4486261 DOI: 10.1002/anie.201205271] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Indexed: 11/07/2022]
Affiliation(s)
- Li Tang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, 1304 W. Green Street, Urbana, IL, 61801 (USA)
| | - Xujuan Yang
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign
| | | | - Isthier Chaudhury
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, 1304 W. Green Street, Urbana, IL, 61801 (USA)
| | - Qian Yin
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, 1304 W. Green Street, Urbana, IL, 61801 (USA)
| | - Catherine Yao
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, 1304 W. Green Street, Urbana, IL, 61801 (USA)
| | - Stéphane Lezmi
- Department of Pathobiology, University of Illinois at Urbana-Champaign
| | - William G. Helferich
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign
| | - Timothy M. Fan
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign
| | - Jianjun Cheng
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, 1304 W. Green Street, Urbana, IL, 61801 (USA)
| |
Collapse
|
44
|
Szymański P, Frączek T, Markowicz M, Mikiciuk-Olasik E. Development of copper based drugs, radiopharmaceuticals and medical materials. Biometals 2012; 25:1089-112. [PMID: 22914969 PMCID: PMC3496555 DOI: 10.1007/s10534-012-9578-y] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 08/03/2012] [Indexed: 01/23/2023]
Abstract
Copper is one of the most interesting elements for various biomedical applications. Copper compounds show vast array of biological actions, including anti-inflammatory, anti-proliferative, biocidal and other. It also offers a selection of radioisotopes, suitable for nuclear imaging and radiotherapy. Quick progress in nanotechnology opened new possibilities for design of copper based drugs and medical materials. To date, copper has not found many uses in medicine, but number of ongoing research, as well as preclinical and clinical studies, will most likely lead to many novel applications of copper in the near future.
Collapse
Affiliation(s)
- Paweł Szymański
- Department of Pharmaceutical Chemistry and Drug Analysis, Medical University of Lodz, Muszyńskiego 1, 90-151, Lodz, Poland.
| | | | | | | |
Collapse
|
45
|
Tang L, Yang X, Dobrucki LW, Chaudhury I, Yin Q, Yao C, Lezmi S, Helferich WG, Fan TM, Cheng J. Aptamer-Functionalized, Ultra-Small, Monodisperse Silica Nanoconjugates for Targeted Dual-Modal Imaging of Lymph Nodes with Metastatic Tumors. Angew Chem Int Ed Engl 2012. [DOI: 10.1002/ange.201205271] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
46
|
Hong H, Zhang Y, Severin GW, Yang Y, Engle JW, Niu G, Nickles RJ, Chen X, Leigh BR, Barnhart TE, Cai W. Multimodality imaging of breast cancer experimental lung metastasis with bioluminescence and a monoclonal antibody dual-labeled with 89Zr and IRDye 800CW. Mol Pharm 2012; 9:2339-49. [PMID: 22784250 DOI: 10.1021/mp300277f] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Metastatic breast cancer is incurable. The goal of this study was to develop a positron emission tomography (PET)/near-infrared fluorescent (NIRF) probe for imaging CD105 expression in breast cancer experimental lung metastasis. TRC105, a chimeric anti-CD105 antibody, was dual-labeled with a NIRF dye (IRDye 800CW) and (89)Zr to yield (89)Zr-Df-TRC105-800CW. Luciferase-transfected 4T1 murine breast cancer cells were injected intravenously into female mice to establish the tumor model. Bioluminescence imaging (BLI) was carried out to noninvasively monitor the lung tumor burden. PET imaging revealed that 4T1 lung tumor uptake of (89)Zr-Df-TRC105-800CW was 8.7 ± 1.4, 10.9 ± 0.5, and 9.7 ± 1.1% ID/g at 4, 24, and 48 h postinjection (n = 4), with excellent tumor contrast. Biodistribution studies, blocking, control studies with (89)Zr-Df-cetuximab-800CW, ex vivo BLI/PET/NIRF imaging, and histology all confirmed CD105 specificity of the tracer. Broad clinical potential of TRC105-based agents was shown in many tumor types, which also enabled early detection of small metastasis and intraoperative guidance for tumor removal.
Collapse
Affiliation(s)
- Hao Hong
- Department of Radiology, University of Wisconsin-Madison , Madison, Wisconsin 53705, United States
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Hellebust A, Richards-Kortum R. Advances in molecular imaging: targeted optical contrast agents for cancer diagnostics. Nanomedicine (Lond) 2012; 7:429-45. [PMID: 22385200 DOI: 10.2217/nnm.12.12] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Over the last three decades, our understanding of the molecular changes associated with cancer development and progression has advanced greatly. This has led to new cancer therapeutics targeted against specific molecular pathways; such therapies show great promise to reduce mortality, in part by enabling physicians to tailor therapy for patients based on a molecular profile of their tumor. Unfortunately, the tools for definitive cancer diagnosis - light microscopic examination of biopsied tissue stained with nonspecific dyes - remain focused on the analysis of tissue ex vivo. There is an important need for new clinical tools to support the molecular diagnosis of cancer. Optical molecular imaging is emerging as a technique to help meet this need. Targeted, optically active contrast agents can specifically label extra- and intracellular biomarkers of cancer. Optical images can be acquired in real time with high spatial resolution to image-specific molecular targets, while still providing morphologic context. This article reviews recent advances in optical molecular imaging, highlighting the advances in technology required to improve early cancer detection, guide selection of targeted therapy and rapidly evaluate therapeutic efficacy.
Collapse
Affiliation(s)
- Anne Hellebust
- Rice University, Bioengineering Department, 6100 Main Street, Bioengineering, MS 142, Houston, TX 77005-1892, USA
| | | |
Collapse
|
48
|
Azhdarinia A, Ghosh P, Ghosh S, Wilganowski N, Sevick-Muraca EM. Dual-labeling strategies for nuclear and fluorescence molecular imaging: a review and analysis. Mol Imaging Biol 2012; 14:261-76. [PMID: 22160875 PMCID: PMC3346941 DOI: 10.1007/s11307-011-0528-9] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Molecular imaging is used for the detection of biochemical processes through the development of target-specific contrast agents. Separately, modalities such as nuclear and near-infrared fluorescence (NIRF) imaging have been shown to non-invasively monitor disease. More recently, merging of these modalities has shown promise owing to their comparable detection sensitivity and benefited from the development of dual-labeled imaging agents. Dual-labeled agents hold promise for whole-body and intraoperative imaging and could bridge the gap between surgical planning and image-guided resection with a single, molecularly targeted agent. In this review, we summarized the literature for dual-labeled antibodies and peptides that have been developed and have highlighted key considerations for incorporating NIRF dyes into nuclear labeling strategies. We also summarized our findings on several commercially available NIRF dyes and offer perspectives for developing a toolkit to select the optimal NIRF dye and radiometal combination for multimodality imaging.
Collapse
Affiliation(s)
- Ali Azhdarinia
- Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX 77030, USA.
| | | | | | | | | |
Collapse
|
49
|
Kramer-Marek G, Bernardo M, Kiesewetter DO, Bagci U, Kuban M, Aras O, Omer A, Zielinski R, Seidel J, Choyke P, Capala J. PET of HER2-positive pulmonary metastases with 18F-ZHER2:342 affibody in a murine model of breast cancer: comparison with 18F-FDG. J Nucl Med 2012; 53:939-46. [PMID: 22582046 PMCID: PMC7497802 DOI: 10.2967/jnumed.111.100354] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
UNLABELLED Targeted therapies often depend on the expression of the target present in the tumor. This expression can be difficult to ascertain in widespread metastases. (18)F-FDG PET/CT, although sensitive, is nonspecific for particular tumor markers. Here, we compare the use of a human epidermal growth factor receptor 2 (HER2)-specific (18)F-Z(HER2)(:342)-Affibody and (18)F-FDG in HER2-expressing pulmonary metastases in a murine model of breast cancer. METHODS The lung metastasis model was established by intravenous injection of MDA-MB-231(HER2)-Luc human breast cancer cells into the tail vein. Bioluminescence imaging was used to evaluate metastasis progression. Uptake of (18)F-Z(HER2)(:342)-Affibody and (18)F-FDG was confirmed by coregistration of the PET images with MR and CT images. At the end of the study, the presence of neoplastic cells and HER2 expression in lung tissues, and distribution of the tracer, were assessed ex vivo by immunohistochemistry and autoradiography. RESULTS (18)F-Z(HER2)(:342)-Affibody successfully targeted HER2-positive lesions in the lung and allowed detection of metastases as early as 9 wk after injection of cells. In contrast, (18)F-FDG uptake was often masked by surrounding inflammatory changes and was nonspecific for HER2 expression. HER2 expression at a cellular level correlated well with tracer uptake on autoradiography. CONCLUSION (18)F-Z(HER2)(:342)-Affibody is a promising tracer for evaluation of HER2 status of breast cancer metastases and is more specific for detecting HER2-positive lesions than (18)F-FDG.
Collapse
Affiliation(s)
- Gabriela Kramer-Marek
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Stroud MR, Hansen SJ, Olson JM. In vivo bio-imaging using chlorotoxin-based conjugates. Curr Pharm Des 2012; 17:4362-71. [PMID: 22204434 DOI: 10.2174/138161211798999375] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Accepted: 11/02/2011] [Indexed: 01/10/2023]
Abstract
Surgical resection remains the primary component of cancer therapy. The precision required to successfully separate cancer tissue from normal tissue relies heavily on the surgeon's ability to delineate the tumor margins. Despite recent advances in surgical guidance and monitoring systems, intra-operative identification of these margins remains imprecise and directly influences patient prognosis. If the surgeon had improved tools to distinguish these margins, tumor progression and unacceptable morbidity could be avoided. In this article, we review the history of chlorotoxin and its tumor specificity and discuss the research currently being generated to target optical imaging agents to cancer tissue.
Collapse
Affiliation(s)
- Mark R Stroud
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | | |
Collapse
|