1
|
Jin C, Luo X, Li X, Zhou R, Zhong Y, Xu Z, Cui C, Xing X, Zhang H, Tian M. Positron emission tomography molecular imaging-based cancer phenotyping. Cancer 2022; 128:2704-2716. [PMID: 35417604 PMCID: PMC9324101 DOI: 10.1002/cncr.34228] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/06/2022] [Accepted: 03/09/2022] [Indexed: 12/28/2022]
Abstract
During the past several decades, numerous studies have provided insights into biological characteristics of cancer cells and identified various hallmarks of cancer acquired in the tumorigenic processes. However, it is still challenging to image these distinctive traits of cancer to facilitate the management of patients in clinical settings. The rapidly evolving field of positron emission tomography (PET) imaging has provided opportunities to investigate cancer's biological characteristics in vivo. This article reviews the current status of PET imaging on characterizing hallmarks of cancer and discusses the future directions of PET imaging strategies facilitating in vivo cancer phenotyping.
Collapse
Affiliation(s)
- Chentao Jin
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
| | - Xiaoyun Luo
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
| | - Xiaoyi Li
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
| | - Rui Zhou
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
| | - Yan Zhong
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
| | - Zhoujiao Xu
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
| | - Chunyi Cui
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
| | - Xiaoqing Xing
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
| | - Hong Zhang
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
- College of Biomedical Engineering and Instrument ScienceZhejiang UniversityHangzhouChina
- Key Laboratory for Biomedical Engineering of Ministry of EducationZhejiang UniversityHangzhouChina
| | - Mei Tian
- Department of Nuclear Medicine and Positron Emission Tomography CenterThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- Institute of Nuclear Medicine and Molecular ImagingZhejiang UniversityHangzhouChina
- Key Laboratory of Medical Molecular Imaging of Zhejiang ProvinceHangzhouChina
| |
Collapse
|
2
|
Ayo A, Laakkonen P. Peptide-Based Strategies for Targeted Tumor Treatment and Imaging. Pharmaceutics 2021; 13:pharmaceutics13040481. [PMID: 33918106 PMCID: PMC8065807 DOI: 10.3390/pharmaceutics13040481] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 02/03/2023] Open
Abstract
Cancer is one of the leading causes of death worldwide. The development of cancer-specific diagnostic agents and anticancer toxins would improve patient survival. The current and standard types of medical care for cancer patients, including surgery, radiotherapy, and chemotherapy, are not able to treat all cancers. A new treatment strategy utilizing tumor targeting peptides to selectively deliver drugs or applicable active agents to solid tumors is becoming a promising approach. In this review, we discuss the different tumor-homing peptides discovered through combinatorial library screening, as well as native active peptides. The different structure–function relationship data that have been used to improve the peptide’s activity and conjugation strategies are highlighted.
Collapse
Affiliation(s)
- Abiodun Ayo
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland;
| | - Pirjo Laakkonen
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland;
- Laboratory Animal Center, HiLIFE—Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland
- Correspondence: ; Tel.: +358-50-4489100
| |
Collapse
|
3
|
Marcu LG, Moghaddasi L, Bezak E. Imaging of Tumor Characteristics and Molecular Pathways With PET: Developments Over the Last Decade Toward Personalized Cancer Therapy. Int J Radiat Oncol Biol Phys 2018; 102:1165-1182. [PMID: 29907486 DOI: 10.1016/j.ijrobp.2018.04.055] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 04/09/2018] [Accepted: 04/19/2018] [Indexed: 02/08/2023]
Abstract
PURPOSE Improvements in personalized therapy are made possible by the advances in molecular biology that led to developments in molecular imaging, allowing highly specific in vivo imaging of biological processes. Positron emission tomography (PET) is the most specific and sensitive imaging technique for in vivo molecular targets and pathways, offering quantification and evaluation of functional properties of the targeted anatomy. MATERIALS AND METHODS This work is an integrative research review that summarizes and evaluates the accumulated current status of knowledge of recent advances in PET imaging for cancer diagnosis and treatment, concentrating on novel radiotracers and evaluating their advantages and disadvantages in cancer characterization. Medline search was conducted, limited to English publications from 2007 onward. Identified manuscripts were evaluated for most recent developments in PET imaging of cancer hypoxia, angiogenesis, proliferation, and clonogenic cancer stem cells (CSC). RESULTS There is an expansion observed from purely metabolic-based PET imaging toward antibody-based PET to achieve more information on cancer characteristics to identify hypoxia, proangiogenic factors, CSC, and others. 64Cu-ATSM, for example, can be used both as a hypoxia and a CSC marker. CONCLUSIONS Progress in the field of functional imaging will possibly lead to more specific tumor targeting and personalized treatment, increasing tumor control and improving quality of life.
Collapse
Affiliation(s)
- Loredana Gabriela Marcu
- Faculty of Science, University of Oradea, Oradea, Romania; Cancer Research Institute and School of Health Sciences, University of South Australia, Adelaide SA, Australia
| | - Leyla Moghaddasi
- GenesisCare, Tennyson Centre, Adelaide SA, Australia; Department of Physics, University of Adelaide, Adelaide SA, Australia
| | - Eva Bezak
- Cancer Research Institute and School of Health Sciences, University of South Australia, Adelaide SA, Australia; Department of Physics, University of Adelaide, Adelaide SA, Australia.
| |
Collapse
|
4
|
Butsch V, Börgel F, Galla F, Schwegmann K, Hermann S, Schäfers M, Riemann B, Wünsch B, Wagner S. Design, (Radio)Synthesis, and in Vitro and in Vivo Evaluation of Highly Selective and Potent Matrix Metalloproteinase 12 (MMP-12) Inhibitors as Radiotracers for Positron Emission Tomography. J Med Chem 2018; 61:4115-4134. [DOI: 10.1021/acs.jmedchem.8b00200] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Viktoria Butsch
- Department of Nuclear Medicine, University Hospital Münster, Albert-Schweitzer-Campus 1, Building A1, 48149 Münster, Germany
| | - Frederik Börgel
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Corrensstraße 48, 48149 Münster, Germany
| | - Fabian Galla
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Corrensstraße 48, 48149 Münster, Germany
| | - Katrin Schwegmann
- European Institute for Molecular Imaging, University of Münster, Waldeyerstraße 15, 48149 Münster, Germany
| | - Sven Hermann
- European Institute for Molecular Imaging, University of Münster, Waldeyerstraße 15, 48149 Münster, Germany
| | - Michael Schäfers
- Department of Nuclear Medicine, University Hospital Münster, Albert-Schweitzer-Campus 1, Building A1, 48149 Münster, Germany
- European Institute for Molecular Imaging, University of Münster, Waldeyerstraße 15, 48149 Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003 − CiM), University of Münster, 48149 Münster, Germany
| | - Burkhard Riemann
- Department of Nuclear Medicine, University Hospital Münster, Albert-Schweitzer-Campus 1, Building A1, 48149 Münster, Germany
| | - Bernhard Wünsch
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Corrensstraße 48, 48149 Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003 − CiM), University of Münster, 48149 Münster, Germany
| | - Stefan Wagner
- Department of Nuclear Medicine, University Hospital Münster, Albert-Schweitzer-Campus 1, Building A1, 48149 Münster, Germany
| |
Collapse
|
5
|
Lu L, Qi H, Zhu J, Sun WX, Zhang B, Tang CY, Cheng Q. Vascular-homing peptides for cancer therapy. Biomed Pharmacother 2017; 92:187-195. [PMID: 28544932 DOI: 10.1016/j.biopha.2017.05.054] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 05/09/2017] [Accepted: 05/10/2017] [Indexed: 12/11/2022] Open
Abstract
In the past 30 years, a variety of phage libraries have been extensively utilized to identify and develop tumor homing peptides (THPs). THPs specifically bind to tumor cells or elements of the tumor microenvironment while no or low affinity to normal cells. In this regard, the efficacy of therapeutic agents in cancer therapy can be enhanced by targeting strategies based on coupling with THPs that recognize receptors expressed by tumor cells or tumor vasculature. Especially, vascular-homing peptides, targeting tumor vasculature, have their receptors expressed on or around the blood vessel including pro-angiogenic factors, metalloproteinase, integrins, fibrin-fibronectin complexes, etc. This review briefly summarizes recent studies on identification and therapeutic applications of vascular-homing peptides targeting common angiogenic markers or with unknown vascular targets in some certain types of cancers. These newly discovered vascular-homing peptides are promising candidates which could provide novel strategies for cancer therapy.
Collapse
Affiliation(s)
- Lan Lu
- Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, PR China; Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, PR China.
| | - Huan Qi
- School of Life Science and Engineering, Southwest University of Science and Technology, PR China
| | - Jie Zhu
- Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, PR China
| | - Wen Xia Sun
- Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, PR China
| | - Bin Zhang
- Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, PR China
| | - Chun Yan Tang
- Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, PR China
| | - Qiang Cheng
- Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, PR China.
| |
Collapse
|
6
|
Abstract
Molecular imaging allows for the visualization of changes at the cellular level in diseases such as cancer. A successful molecular imaging agent must rely on disease-selective targets and ligands that specifically interact with those targets. Unfortunately, the translation of novel target-specific ligands into the clinic has been frustratingly slow with limitations including the complex design and screening approaches for ligand identification, as well as their subsequent optimization into useful imaging agents. This review focuses on combinatorial library approaches towards addressing these two challenges, with particular focus on phage display and one-bead one-compound (OBOC) libraries. Both of these peptide-based techniques have proven successful in identifying new ligands for cancer-specific targets and some of the success stories will be highlighted. New developments in screening methodology and sequencing technology have pushed the bounds of phage display and OBOC even further, allowing for even faster and more robust discovery of novel ligands. The combination of multiple high-throughput technologies will not only allow for more accurate identification, but also faster affinity maturation, while overall streamlining the process of translating novel ligands into clinical imaging agents.
Collapse
|
7
|
Liu R, Li X, Xiao W, Lam KS. Tumor-targeting peptides from combinatorial libraries. Adv Drug Deliv Rev 2017; 110-111:13-37. [PMID: 27210583 DOI: 10.1016/j.addr.2016.05.009] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 05/10/2016] [Accepted: 05/11/2016] [Indexed: 02/07/2023]
Abstract
Cancer is one of the major and leading causes of death worldwide. Two of the greatest challenges in fighting cancer are early detection and effective treatments with no or minimum side effects. Widespread use of targeted therapies and molecular imaging in clinics requires high affinity, tumor-specific agents as effective targeting vehicles to deliver therapeutics and imaging probes to the primary or metastatic tumor sites. Combinatorial libraries such as phage-display and one-bead one-compound (OBOC) peptide libraries are powerful approaches in discovering tumor-targeting peptides. This review gives an overview of different combinatorial library technologies that have been used for the discovery of tumor-targeting peptides. Examples of tumor-targeting peptides identified from each combinatorial library method will be discussed. Published tumor-targeting peptide ligands and their applications will also be summarized by the combinatorial library methods and their corresponding binding receptors.
Collapse
Affiliation(s)
- Ruiwu Liu
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA 95817, USA; University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA
| | - Xiaocen Li
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA 95817, USA; University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA
| | - Wenwu Xiao
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA 95817, USA; University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA
| | - Kit S Lam
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA 95817, USA; University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA; Division of Hematology & Oncology, Department of Internal Medicine, University of California Davis, Sacramento, CA 95817, USA
| |
Collapse
|
8
|
Baldoneschi V, Cerofolini L, Dragoni E, Storai A, Luchinat C, Fragai M, Richichi B, Nativi C. Active-Site Targeting Paramagnetic Probe for Matrix Metalloproteinases. Chempluschem 2016; 81:1333-1338. [DOI: 10.1002/cplu.201600375] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 09/23/2016] [Indexed: 11/11/2022]
Affiliation(s)
- Veronica Baldoneschi
- Department of Chemistry “Ugo Schiff”; University of Florence; Via della Lastruccia 3-13 50019 Sesto Fiorentino Italy
| | - Linda Cerofolini
- Department of Chemistry “Ugo Schiff”; University of Florence; Via della Lastruccia 3-13 50019 Sesto Fiorentino Italy
- CERM; Univeristy of Florence; Via Sacconi 6 50019 Sesto Fiorentino Italy
| | - Elisa Dragoni
- Department of Chemistry “Ugo Schiff”; University of Florence; Via della Lastruccia 3-13 50019 Sesto Fiorentino Italy
| | - Andrea Storai
- Department of Chemistry “Ugo Schiff”; University of Florence; Via della Lastruccia 3-13 50019 Sesto Fiorentino Italy
| | - Claudio Luchinat
- Department of Chemistry “Ugo Schiff”; University of Florence; Via della Lastruccia 3-13 50019 Sesto Fiorentino Italy
- CERM; Univeristy of Florence; Via Sacconi 6 50019 Sesto Fiorentino Italy
| | - Marco Fragai
- Department of Chemistry “Ugo Schiff”; University of Florence; Via della Lastruccia 3-13 50019 Sesto Fiorentino Italy
- CERM; Univeristy of Florence; Via Sacconi 6 50019 Sesto Fiorentino Italy
| | - Barbara Richichi
- Department of Chemistry “Ugo Schiff”; University of Florence; Via della Lastruccia 3-13 50019 Sesto Fiorentino Italy
| | - Cristina Nativi
- Department of Chemistry “Ugo Schiff”; University of Florence; Via della Lastruccia 3-13 50019 Sesto Fiorentino Italy
- CERM; Univeristy of Florence; Via Sacconi 6 50019 Sesto Fiorentino Italy
| |
Collapse
|
9
|
Jackson SR, Wong AC, Travis AR, Catrina IE, Bratu DP, Wright DW, Jayagopal A. Applications of Hairpin DNA-Functionalized Gold Nanoparticles for Imaging mRNA in Living Cells. Methods Enzymol 2016; 572:87-103. [PMID: 27241751 DOI: 10.1016/bs.mie.2016.03.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Molecular imaging agents are useful for imaging molecular processes in living systems in order to elucidate the function of molecular mediators in health and disease. Here, we demonstrate a technique for the synthesis, characterization, and application of hairpin DNA-functionalized gold nanoparticles (hAuNPs) as fluorescent hybridization probes for imaging mRNA expression and spatiotemporal dynamics in living cells. These imaging probes feature gold colloids linked to fluorophores via engineered oligonucleotides to resemble a molecular beacon in which the gold colloid serves as the fluorescence quencher in a fluorescence resonance energy transfer system. Target-specific hybridization of the hairpin oligonucleotide enables fluorescence de-quenching and subsequent emission with high signal to noise ratios. hAuNPs exhibit high specificity without adverse toxicity or the need for transfection reagents. Furthermore, tunability of hAuNP emission profiles by selection of spectrally distinct fluorophores enables multiplexed mRNA imaging applications. Therefore, hAuNPs are promising tools for imaging gene expression in living cells. As a representative application of this technology, we discuss the design and applications of hAuNP targeted against distinct matrix metalloproteinase enzymes for the multiplexed detection of mRNA expression in live breast cancer cells using flow cytometry and fluorescence microscopy.
Collapse
Affiliation(s)
- S R Jackson
- Vanderbilt University, Nashville, TN, United States
| | - A C Wong
- Vanderbilt University, Nashville, TN, United States
| | - A R Travis
- Vanderbilt University, Nashville, TN, United States
| | - I E Catrina
- Hunter College, City University of New York, New York, NY, United States
| | - D P Bratu
- Hunter College, City University of New York, New York, NY, United States; Program in Molecular, Cellular, and Developmental Biology, and Program in Biochemistry, The Graduate Center, City University of New York, New York, NY, United States
| | - D W Wright
- Vanderbilt University, Nashville, TN, United States
| | - A Jayagopal
- Pharma Research and Early Development (pRED), F. Hoffman-La Roche Ltd., Basel, Switzerland.
| |
Collapse
|
10
|
Fields GB, Stawikowski MJ. Imaging Matrix Metalloproteinase Activity Implicated in Breast Cancer Progression. Methods Mol Biol 2016; 1406:303-29. [PMID: 26820965 DOI: 10.1007/978-1-4939-3444-7_25] [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: 01/08/2023]
Abstract
Proteolysis has been cited as an important contributor to cancer initiation and progression. One can take advantage of tumor-associated proteases to selectively deliver imaging agents. Protease-activated imaging systems have been developed using substrates designed for hydrolysis by members of the matrix metalloproteinase (MMP) family. We presently describe approaches by which one can optically image matrix metalloproteinase activity implicated in breast cancer progression, with consideration of selective versus broad protease probes.
Collapse
Affiliation(s)
- Gregg B Fields
- Department of Chemistry & Biochemistry, Florida Atlantic University, Jupiter, FL, 33458, USA. .,Department of Chemistry, The Scripps Research Institute/Scripps Florida, Jupiter, FL, 33458, USA. .,Departments of Chemistry and Biology, Torrey Pines Institute for Molecular Studies, Port St. Lucie, FL, 34987, USA.
| | - Maciej J Stawikowski
- Department of Chemistry & Biochemistry, Florida Atlantic University, Jupiter, FL, 33458, USA
| |
Collapse
|
11
|
Lebel R, Lepage M. A comprehensive review on controls in molecular imaging: lessons from MMP-2 imaging. CONTRAST MEDIA & MOLECULAR IMAGING 2015; 9:187-210. [PMID: 24700747 DOI: 10.1002/cmmi.1555] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 06/11/2013] [Accepted: 06/19/2013] [Indexed: 12/31/2022]
Abstract
Metalloproteinases (MMPs), including MMP-2, play critical roles in tissue remodeling and are involved in a large array of pathologies, including cancer, arthritis and atherosclerosis. Their prognostic value warranted a large investment or resources in the development of noninvasive detection methods, based on probes for many current clinical and pre-clinical imaging modalities. However, the potential of imaging techniques is only matched by the complexity of the data they generate. This complexity must be properly assessed and accounted for in the early steps of probe design and testing in order to accurately determine the efficacy and efficiency of an imaging strategy. This review proposes basic rules for the evaluation of novel probes by addressing the specific case of MMP targeted probes.
Collapse
Affiliation(s)
- Réjean Lebel
- Centre d'imagerie moléculaire de Sherbrooke, Département de médecine nucléaire et radiobiologie, Université de Sherbrooke, Sherbrooke, QC, Canada
| | | |
Collapse
|
12
|
Liu Q, Pan D, Cheng C, Zhang D, Zhang A, Wang L, Jiang H, Wang T, Liu H, Xu Y, Yang R, Chen F, Yang M, Zuo C. Development of a Novel PET Tracer [18F]AlF-NOTA-C6 Targeting MMP2 for Tumor Imaging. PLoS One 2015; 10:e0141668. [PMID: 26540114 PMCID: PMC4634933 DOI: 10.1371/journal.pone.0141668] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 10/12/2015] [Indexed: 12/28/2022] Open
Abstract
Background and Objective The overexpression of gelatinases, that is, matrix metalloproteinase MMP2 and MMP9, has been associated with tumor progression, invasion, and metastasis. To image MMP2 in tumors, we developed a novel ligand termed [18F]AlF-NOTA-C6, with consideration that: c(KAHWGFTLD)NH2 (herein, C6) is a selective gelatinase inhibitor; Cy5.5-C6 has been visualized in many in vivo tumor models; positron emission tomography (PET) has a higher detection sensitivity and a wider field of view than optical imaging; fluorine-18 (18F) is the optimal PET radioisotope, and the creation of a [18F]AlF-peptide complex is a simple procedure. Methods C6 was conjugated to the bifunctional chelator NOTA (1, 4, 7-triazacyclononanetriacetic acid) for radiolabeling [18F]AlF conjugation. The MMP2-binding characteristics and tumor-targeting efficacy of [18F]AlF-NOTA-C6 were tested in vitro and in vivo. Results The non-decay corrected yield of [18F]AlF-NOTA-C6 was 46.2–64.2%, and the radiochemical purity exceeded 95%. [18F]AlF-NOTA-C6 was favorably retained in SKOV3 and PC3 cells, determined by cell uptake. Using NOTA-C6 as a competitive ligand, the uptake of [18F]AlF-NOTA-C6 in SKOV3 cells decreased in a dose-dependent manner. In biodistribution and PET imaging studies, higher radioactivity concentrations were observed in tumors. Pre-injection of C6 caused a marked reduction in tumor tissue uptake. Immunohistochemistry showed MMP2 in tumor tissues. Conclusions [18F]AlF-NOTA-C6 was easy to synthesize and has substantial potential as an imaging agent that targets MMP2 in tumors.
Collapse
Affiliation(s)
- Qinghua Liu
- Department of Nuclear Medicine, Changhai Hospital, the Second Military Medical University, Shanghai, 200433, China
- * E-mail: (QHL); (MY); (CJZ)
| | - Donghui Pan
- Jiangsu Institute of Nuclear Medicine, Key Laboratory of Nuclear Medicine, Ministry of Health, Wuxi, 214063, China
| | - Chao Cheng
- Department of Nuclear Medicine, Changhai Hospital, the Second Military Medical University, Shanghai, 200433, China
| | - Dazhi Zhang
- Department of Organic Chemistry, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Anyu Zhang
- Department of Nuclear Medicine, Changhai Hospital, the Second Military Medical University, Shanghai, 200433, China
| | - Lizhen Wang
- Jiangsu Institute of Nuclear Medicine, Key Laboratory of Nuclear Medicine, Ministry of Health, Wuxi, 214063, China
| | - Hongdie Jiang
- Department of Nuclear Medicine, Changhai Hospital, the Second Military Medical University, Shanghai, 200433, China
| | - Tao Wang
- Department of Nuclear Medicine, Changhai Hospital, the Second Military Medical University, Shanghai, 200433, China
| | - Hongrui Liu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Yuping Xu
- Jiangsu Institute of Nuclear Medicine, Key Laboratory of Nuclear Medicine, Ministry of Health, Wuxi, 214063, China
| | - Runlin Yang
- Jiangsu Institute of Nuclear Medicine, Key Laboratory of Nuclear Medicine, Ministry of Health, Wuxi, 214063, China
| | - Fei Chen
- Jiangsu Institute of Nuclear Medicine, Key Laboratory of Nuclear Medicine, Ministry of Health, Wuxi, 214063, China
| | - Min Yang
- Jiangsu Institute of Nuclear Medicine, Key Laboratory of Nuclear Medicine, Ministry of Health, Wuxi, 214063, China
- * E-mail: (QHL); (MY); (CJZ)
| | - Changjing Zuo
- Department of Nuclear Medicine, Changhai Hospital, the Second Military Medical University, Shanghai, 200433, China
- * E-mail: (QHL); (MY); (CJZ)
| |
Collapse
|
13
|
Mahajan A, Goh V, Basu S, Vaish R, Weeks AJ, Thakur MH, Cook GJ. Bench to bedside molecular functional imaging in translational cancer medicine: to image or to imagine? Clin Radiol 2015; 70:1060-82. [PMID: 26187890 DOI: 10.1016/j.crad.2015.06.082] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 06/03/2015] [Accepted: 06/08/2015] [Indexed: 02/05/2023]
Abstract
Ongoing research on malignant and normal cell biology has substantially enhanced the understanding of the biology of cancer and carcinogenesis. This has led to the development of methods to image the evolution of cancer, target specific biological molecules, and study the anti-tumour effects of novel therapeutic agents. At the same time, there has been a paradigm shift in the field of oncological imaging from purely structural or functional imaging to combined multimodal structure-function approaches that enable the assessment of malignancy from all aspects (including molecular and functional level) in a single examination. The evolving molecular functional imaging using specific molecular targets (especially with combined positron-emission tomography [PET] computed tomography [CT] using 2- [(18)F]-fluoro-2-deoxy-D-glucose [FDG] and other novel PET tracers) has great potential in translational research, giving specific quantitative information with regard to tumour activity, and has been of pivotal importance in diagnoses and therapy tailoring. Furthermore, molecular functional imaging has taken a key place in the present era of translational cancer research, producing an important tool to study and evolve newer receptor-targeted therapies, gene therapies, and in cancer stem cell research, which could form the basis to translate these agents into clinical practice, popularly termed "theranostics". Targeted molecular imaging needs to be developed in close association with biotechnology, information technology, and basic translational scientists for its best utility. This article reviews the current role of molecular functional imaging as one of the main pillars of translational research.
Collapse
Affiliation(s)
- A Mahajan
- Division of Imaging Sciences and Biomedical Engineering, King's College London, UK; Department of Radiodiagnosis, Tata Memorial Centre, Mumbai, 400012, India.
| | - V Goh
- Division of Imaging Sciences and Biomedical Engineering, King's College London, UK
| | - S Basu
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital Annexe, Mumbai, 400 012, India
| | - R Vaish
- Department of Head and Neck Surgical Oncology, Tata Memorial Centre, Mumbai, 400012, India
| | - A J Weeks
- Division of Imaging Sciences and Biomedical Engineering, King's College London, UK
| | - M H Thakur
- Department of Radiodiagnosis, Tata Memorial Centre, Mumbai, 400012, India
| | - G J Cook
- Division of Imaging Sciences and Biomedical Engineering, King's College London, UK; Department of Nuclear Medicine, Guy's and St Thomas NHS Foundation Trust Hospital, London, UK
| |
Collapse
|
14
|
Liu Q, Pan D, Cheng C, Zhang A, Ma C, Wang L, Zhang D, Liu H, Jiang H, Wang T, Xu Y, Yang R, Chen F, Yang M, Zuo C. Targeting of MMP2 activity in malignant tumors with a 68Ga-labeled gelatinase inhibitor cyclic peptide. Nucl Med Biol 2015; 42:939-44. [PMID: 26344861 DOI: 10.1016/j.nucmedbio.2015.07.013] [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: 04/30/2015] [Revised: 07/30/2015] [Accepted: 07/31/2015] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Elevated levels of gelatinases (matrix metalloproteinases 2/9, i.e., MMP2 and MMP9) are associated with tumor progression, invasion and metastasis, so these enzymes are potential targets for tumor imaging. The peptide c(KAHWGFTLD)NH2 (herein, C6) is a selective gelatinase inhibitor. Cy5.5-C6 has been visualized in many tumor models in vivo. However, the sensitivity and penetrance of optical imaging are poor. It is well known that positron emission tomography (PET) has a high detection sensitivity and Gallium-68 ((68)Ga) is an optimal PET radioisotope. Thus, in the present study, we developed a novel ligand, (68)Ga-NOTA-C6, to image MMP2 activity in tumors. METHODS C6 was conjugated with the bifunctional chelator NOTA (1,4,7-triazacyclononanetriacetic acid) and labeled with (68)Ga. In vitro uptake and binding analyses were performed by using SKOV3 cell lines, coincubating with or without the MMP inhibitor doxycycline. The biodistribution and PET imaging were conducted on SKOV3 ovarian tumor models. MMP2 expression in tumors was analyzed by immunohistochemistry (IHC). RESULTS The non-decay corrected yield of (68)Ga-NOTA-C6 was 61.8%-63.3%. (68)Ga-NOTA-C6 was stable in both physiological saline and human serum. The uptake of (68)Ga-NOTA-C6 in SKOV3 cells increased with time, and could be blocked by doxycycline in a dose dependent manner. The results of biodistribution and PET imaging showed that high radioactivity concentrations of (68)Ga-NOTA-C6 occurred in tumors. The ratios of tumor to blood, muscle and ovary and oviduct at 30, 60 and 120min p.i. were 2.78±0.54, 3.86±0.65, 0.48±0.14, and 1.73±0.36, 10.31±3.12, 1.22±0.10, and 2.50±0.78, 7.03±1.85, 0.97±0.25, respectively. The tracer was excreted mainly through the renal system, as evidenced by high levels of radioactivity in the kidneys. These data support the possibility of using (68)Ga-NOTA-C6 in PET to visualize tumors that overexpress MMP2. CONCLUSIONS (68)Ga-NOTA-C6 is a potential radiopharmaceutical for the imaging of in vivo MMP2 activity in tumors.
Collapse
Affiliation(s)
- Qinghua Liu
- Department of Nuclear Medicine, Changhai Hospital, the Second Military Medical University, Shanghai, China, 200433.
| | - Donghui Pan
- Jiangsu Institute of Nuclear Medicine, Key Laboratory of Nuclear Medicine, Ministry of Health, Wuxi, Jiangsu, China, 214063
| | - Chao Cheng
- Department of Nuclear Medicine, Changhai Hospital, the Second Military Medical University, Shanghai, China, 200433
| | - Anyu Zhang
- Department of Nuclear Medicine, Changhai Hospital, the Second Military Medical University, Shanghai, China, 200433
| | - Chao Ma
- Department of Nuclear Medicine, Zhongshan Hospital, Xiamen University, Xiamen, Fujian, China, 361004
| | - Lizhen Wang
- Jiangsu Institute of Nuclear Medicine, Key Laboratory of Nuclear Medicine, Ministry of Health, Wuxi, Jiangsu, China, 214063
| | - Dazhi Zhang
- Department of Organic Chemistry, School of Pharmacy, the Second Military Medical University, Shanghai, China, 200433
| | - Hongrui Liu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China, 201203
| | - Hongdie Jiang
- Department of Nuclear Medicine, Changhai Hospital, the Second Military Medical University, Shanghai, China, 200433
| | - Tao Wang
- Department of Nuclear Medicine, Changhai Hospital, the Second Military Medical University, Shanghai, China, 200433
| | - Yuping Xu
- Jiangsu Institute of Nuclear Medicine, Key Laboratory of Nuclear Medicine, Ministry of Health, Wuxi, Jiangsu, China, 214063
| | - Runlin Yang
- Jiangsu Institute of Nuclear Medicine, Key Laboratory of Nuclear Medicine, Ministry of Health, Wuxi, Jiangsu, China, 214063
| | - Fei Chen
- Jiangsu Institute of Nuclear Medicine, Key Laboratory of Nuclear Medicine, Ministry of Health, Wuxi, Jiangsu, China, 214063
| | - Min Yang
- Jiangsu Institute of Nuclear Medicine, Key Laboratory of Nuclear Medicine, Ministry of Health, Wuxi, Jiangsu, China, 214063.
| | - Changjing Zuo
- Department of Nuclear Medicine, Changhai Hospital, the Second Military Medical University, Shanghai, China, 200433.
| |
Collapse
|
15
|
Kawano T, Murata M, Piao JS, Narahara S, Hamano N, Kang JH, Hashizume M. Systemic delivery of protein nanocages bearing CTT peptides for enhanced imaging of MMP-2 expression in metastatic tumor models. Int J Mol Sci 2014; 16:148-58. [PMID: 25547485 PMCID: PMC4307240 DOI: 10.3390/ijms16010148] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 12/15/2014] [Indexed: 11/16/2022] Open
Abstract
Matrix metalloproteinase 2 (MMP-2) in metastatic cancer tissue, which is associated with a poor prognosis, is a potential target for tumor imaging in vivo. Here, we describe a metastatic cancer cell-targeted protein nanocage. An MMP-2-binding peptide, termed CTT peptide (CTTHWGFTLC), was conjugated to the surface of a naturally occurring heat shock protein nanocage by genetic modification. The engineered protein nanocages showed a binding affinity for MMP-2 and selective uptake in cancer cells that highly expressed MMP-2 in vitro. In near-infrared fluorescence imaging, the nanocages showed specific and significant accumulation in tumor tissue after intravenous injection in vivo. These protein nanocages conjugated with CTT peptide could be potentially applied to a noninvasive near-infrared fluorescence detection method for imaging gelatinase activity in metastatic tumors in vivo.
Collapse
Affiliation(s)
- Takahito Kawano
- Innovation Center for Medical Redox Navigation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.
| | - Masaharu Murata
- Innovation Center for Medical Redox Navigation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.
| | - Jing Shu Piao
- Department of Advanced Medical Initiatives, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.
| | - Sayoko Narahara
- Innovation Center for Medical Redox Navigation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.
| | - Nobuhito Hamano
- Department of Advanced Medical Initiatives, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.
| | - Jeong-Hun Kang
- Division of Biopharmaceutics and Pharmacokinetics, Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka 565-8565, Japan.
| | - Makoto Hashizume
- Innovation Center for Medical Redox Navigation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.
| |
Collapse
|
16
|
Lin M, Welch MJ, Lapi SE. Effects of chelator modifications on (68)Ga-labeled [Tyr (3)]octreotide conjugates. Mol Imaging Biol 2014; 15:606-13. [PMID: 23529373 DOI: 10.1007/s11307-013-0627-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE Somatostatin receptors (SSTR) have been reported as promising targets for imaging agents for cancer. Recently, (68)Ga-DOTATOC-based PET imaging has been used successfully for diagnosis and management of SSTR-expressing tumors. The purpose of this study was to evaluate the influence of chelator modifications and charge on (68)Ga-labeled peptide conjugates. PROCEDURES We have synthesized a series of [Tyr(3)]octreotide conjugates that consisted of different NOTA-based chelators with two to five carboxylate moieties, and compared our results with (68)Ga-DOTATOC in both in vitro and in vivo studies. RESULTS With the exception of (68)Ga-1 (three carboxylates), the increased number of carboxylates on the NOTA-based chelators resulted in a reduced binding affinity and internalization. Additionally, the tumor uptake for (68)Ga-2 (four carboxylates) and (68)Ga-3 (five carboxylates) was reduced compared to that of (68)Ga-DOTATOC (three carboxylates) and (68)Ga-NO2ATOC (two carboxylates) and (68)Ga-1 (three carboxylates) at 2 h p.i. suggesting the presence of an optimal charge for this compound. CONCLUSIONS Chelator modifications can lead to the altered pharmacokinetics. These results may impact further design considerations for peptide-based imaging agents.
Collapse
Affiliation(s)
- Mai Lin
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Campus Box.8225, 510 South Kingshighway Blvd, St. Louis, MO, 63110, USA
| | | | | |
Collapse
|
17
|
Altıparmak B, Lambrecht FY, Citak A. Design of radiolabeled gelatinase inhibitor peptide ((99m)Tc-CLP) and evaluation in rats. Appl Radiat Isot 2014; 89:130-3. [PMID: 24631744 DOI: 10.1016/j.apradiso.2014.02.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 01/16/2014] [Accepted: 02/13/2014] [Indexed: 11/15/2022]
Abstract
In malignant tissues, MMP-9 (gelatinase B, 92 kDa type IV collagenase) and MMP-2 (gelatinase A, 72 kDa type IV collagenase) are the most prevalent matrix metalloproteinases related to the tumor aggressiveness and metastatic potential. Since elevated levels of gelatinases are associated with poor prognosis in cancer patients, these enzymes are potential targets for tumor imaging to possibly predict metastases. In the present study, a cyclic decapeptide, CLP (Cys-Leu-Pro-Gly-His-Trp-Gly-Phe-Pro-Ser-Cys), was selected as a basic peptide because of its selective inhibitory activity toward gelatinases. The peptide was labelled with (99m)Tc with a radiolabelling efficiency of 94.6±4.1%. After determining the appropriate conditions for radiolabelling, a biodistribution study of radiolabelled peptide in Albino Wistar rats was done. According to biodistribution data, (99m)Tc-CLP showed high uptake in the lung, liver, uterus and spleen. The amount of normal tissue MMPs enzymes is known to be lower than a tumor tissue. In this connection, our findings show that matrix metalloproteinases inhibitory peptide which is CLP is labeled with (99m)Tc with high yield and radiolabeled peptide might be might be utilized for the imaging of gelatinase activity due to overexpression of MMP-2 and MMP-9 in tumor tissue.
Collapse
Affiliation(s)
- Burcu Altıparmak
- Department of Nuclear Applications, Institute of Nuclear Science, Ege University, Bornova, 35100 Izmir, Turkey
| | - Fatma Yurt Lambrecht
- Department of Nuclear Applications, Institute of Nuclear Science, Ege University, Bornova, 35100 Izmir, Turkey.
| | - Asli Citak
- Department of Nuclear Applications, Institute of Nuclear Science, Ege University, Bornova, 35100 Izmir, Turkey
| |
Collapse
|
18
|
Altıparmak B, Lambrecht FY, Er O. Design of (99m) Tc-DTPA-CLP and preliminary evaluation in rats. Chem Biol Drug Des 2014; 83:362-6. [PMID: 24148110 DOI: 10.1111/cbdd.12253] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 09/12/2013] [Accepted: 10/15/2013] [Indexed: 11/28/2022]
Abstract
Radiopharmaceuticals are localized in (malignant) tumor tissues by different mechanisms. One of these mechanisms, gelatinase enzyme activity, is associated with poor prognosis in cancer patients and potential targets for tumor imaging. There are some gelatinases to be associated with metastatic potential for tumor imaging to possibly predict metastases. In this study, a cyclic decapeptide conjugate, DTPA-CLP (DTPA-Cys-Leu-Pro-Gly-His-Trp-Gly-Phe-Pro-Ser-Cys), was selected as a peptide conjugate because of its selective inhibitory activity toward gelatinases. Peptide-conjugated DTPA-CLP was labeled with (99m) Tc with a radiolabeling efficiency of 97.0 ± 2.8%. After determining optimization conditions for radiolabeling, a biodistribution study of radiolabeled peptide in albino Wistar rats was performed. According to biodistribution data, (99m) Tc-DTPA-CLP showed high uptake in the lung, liver, uterus, and spleen. These results show that (99m) Tc-DTPA-CLP may be used for the imaging of gelatinase activity in metastatic tumors.
Collapse
Affiliation(s)
- Burcu Altıparmak
- Department of Nuclear Applications, Institute of Nuclear Science, Ege University, Bornova, Izmir, 35100, Turkey
| | | | | |
Collapse
|
19
|
Affiliation(s)
- Bethany Powell Gray
- Department of Internal Medicine and The Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390-8807, United States
| | - Kathlynn C. Brown
- Department of Internal Medicine and The Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390-8807, United States
| |
Collapse
|
20
|
Abstract
Neuroinflammation plays a central role in a variety of neurological diseases, including stroke, multiple sclerosis, Alzheimer’s disease, and malignant CNS neoplasms, among many other. Different cell types and molecular mediators participate in a cascade of events in the brain that is ultimately aimed at control, regeneration and repair, but leads to damage of brain tissue under pathological conditions. Non-invasive molecular imaging of key players in the inflammation cascade holds promise for identification and quantification of the disease process before it is too late for effective therapeutic intervention. In this review, we focus on molecular imaging techniques that target inflammatory cells and molecules that are of interest in neuroinflammation, especially those with high translational potential. Over the past decade, a plethora of molecular imaging agents have been developed and tested in animal models of (neuro)inflammation, and a few have been translated from bench to bedside. The most promising imaging techniques to visualize neuroinflammation include MRI, positron emission tomography (PET), single photon emission computed tomography (SPECT), and optical imaging methods. These techniques enable us to image adhesion molecules to visualize endothelial cell activation, assess leukocyte functions such as oxidative stress, granule release, and phagocytosis, and label a variety of inflammatory cells for cell tracking experiments. In addition, several cell types and their activation can be specifically targeted in vivo, and consequences of neuroinflammation such as neuronal death and demyelination can be quantified. As we continue to make progress in utilizing molecular imaging technology to study and understand neuroinflammation, increasing efforts and investment should be made to bring more of these novel imaging agents from the “bench to bedside.”
Collapse
Affiliation(s)
- Benjamin Pulli
- Center for Systems Biology and Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 185 Cambridge Street, Boston, MA 02114, USA
| | - John W Chen
- Center for Systems Biology and Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 185 Cambridge Street, Boston, MA 02114, USA
| |
Collapse
|
21
|
Jacobson O, Chen X. Interrogating tumor metabolism and tumor microenvironments using molecular positron emission tomography imaging. Theranostic approaches to improve therapeutics. Pharmacol Rev 2013; 65:1214-56. [PMID: 24064460 DOI: 10.1124/pr.113.007625] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Positron emission tomography (PET) is a noninvasive molecular imaging technology that is becoming increasingly important for the measurement of physiologic, biochemical, and pharmacological functions at cellular and molecular levels in patients with cancer. Formation, development, and aggressiveness of tumor involve a number of molecular pathways, including intrinsic tumor cell mutations and extrinsic interaction between tumor cells and the microenvironment. Currently, evaluation of these processes is mainly through biopsy, which is invasive and limited to the site of biopsy. Ongoing research on specific target molecules of the tumor and its microenvironment for PET imaging is showing great potential. To date, the use of PET for diagnosing local recurrence and metastatic sites of various cancers and evaluation of treatment response is mainly based on [(18)F]fluorodeoxyglucose ([(18)F]FDG), which measures glucose metabolism. However, [(18)F]FDG is not a target-specific PET tracer and does not give enough insight into tumor biology and/or its vulnerability to potential treatments. Hence, there is an increasing need for the development of selective biologic radiotracers that will yield specific biochemical information and allow for noninvasive molecular imaging. The possibility of cancer-associated targets for imaging will provide the opportunity to use PET for diagnosis and therapy response monitoring (theranostics) and thus personalized medicine. This article will focus on the review of non-[(18)F]FDG PET tracers for specific tumor biology processes and their preclinical and clinical applications.
Collapse
Affiliation(s)
- Orit Jacobson
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD.
| | | |
Collapse
|
22
|
Wu C, Li F, Niu G, Chen X. PET imaging of inflammation biomarkers. Theranostics 2013; 3:448-66. [PMID: 23843893 PMCID: PMC3706689 DOI: 10.7150/thno.6592] [Citation(s) in RCA: 131] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 05/24/2013] [Indexed: 01/04/2023] Open
Abstract
Inflammation plays a significant role in many disease processes. Development in molecular imaging in recent years provides new insight into the diagnosis and treatment evaluation of various inflammatory diseases and diseases involving inflammatory process. Positron emission tomography using (18)F-FDG has been successfully applied in clinical oncology and neurology and in the inflammation realm. In addition to glucose metabolism, a variety of targets for inflammation imaging are being discovered and utilized, some of which are considered superior to FDG for imaging inflammation. This review summarizes the potential inflammation imaging targets and corresponding PET tracers, and the applications of PET in major inflammatory diseases and tumor associated inflammation. Also, the current attempt in differentiating inflammation from tumor using PET is also discussed.
Collapse
|
23
|
Casalini F, Fugazza L, Esposito G, Cabella C, Brioschi C, Cordaro A, D’Angeli L, Bartoli A, Filannino AM, Gringeri CV, Longo DL, Muzio V, Nuti E, Orlandini E, Figlia G, Quattrini A, Tei L, Digilio G, Rossello A, Maiocchi A. Synthesis and Preliminary Evaluation in Tumor Bearing Mice of New 18F-Labeled Arylsulfone Matrix Metalloproteinase Inhibitors as Tracers for Positron Emission Tomography. J Med Chem 2013; 56:2676-89. [DOI: 10.1021/jm4001743] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Francesca Casalini
- Department of Science and Technological
Innovation, Università del Piemonte Orientale “Amedeo Avogadro”, Viale T. Michel 11,
I-15121 Alessandria, Italy
| | - Lorenza Fugazza
- Research and Development, Advanced Accelerator Applications, Via Ribes 5, I-10010
Colleretto Giacosa (TO), Italy
| | - Giovanna Esposito
- Molecular Imaging Centre, University of Torino, Via Nizza 52, I-10126 Torino,
Italy
| | - Claudia Cabella
- Centro Ricerche Bracco, Bracco Imaging S.p.A., Via Ribes 5, I-10010 Colleretto
Giacosa (TO), Italy
| | - Chiara Brioschi
- Centro Ricerche Bracco, Bracco Imaging S.p.A., Via Ribes 5, I-10010 Colleretto
Giacosa (TO), Italy
| | - Alessia Cordaro
- Centro Ricerche Bracco, Bracco Imaging S.p.A., Via Ribes 5, I-10010 Colleretto
Giacosa (TO), Italy
| | - Luca D’Angeli
- Molecular Imaging Centre, University of Torino, Via Nizza 52, I-10126 Torino,
Italy
| | - Antonietta Bartoli
- Molecular Imaging Centre, University of Torino, Via Nizza 52, I-10126 Torino,
Italy
| | - Azzurra M. Filannino
- Research and Development, Advanced Accelerator Applications, Via Ribes 5, I-10010
Colleretto Giacosa (TO), Italy
| | - Concetta V. Gringeri
- Department of Science and Technological
Innovation, Università del Piemonte Orientale “Amedeo Avogadro”, Viale T. Michel 11,
I-15121 Alessandria, Italy
| | - Dario L. Longo
- Molecular Imaging Centre, University of Torino, Via Nizza 52, I-10126 Torino,
Italy
| | - Valeria Muzio
- Research and Development, Advanced Accelerator Applications, Via Ribes 5, I-10010
Colleretto Giacosa (TO), Italy
| | - Elisa Nuti
- Department
of Pharmacy, University of Pisa, Via Bonanno
6, I-56126 Pisa, Italy
| | | | - Gianluca Figlia
- Institute of Experimental Neurology,
Division of Neuroscience, San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy
| | - Angelo Quattrini
- Institute of Experimental Neurology,
Division of Neuroscience, San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy
| | - Lorenzo Tei
- Department of Science and Technological
Innovation, Università del Piemonte Orientale “Amedeo Avogadro”, Viale T. Michel 11,
I-15121 Alessandria, Italy
| | - Giuseppe Digilio
- Department of Science and Technological
Innovation, Università del Piemonte Orientale “Amedeo Avogadro”, Viale T. Michel 11,
I-15121 Alessandria, Italy
- Molecular Imaging Centre, University of Torino, Via Nizza 52, I-10126 Torino,
Italy
| | - Armando Rossello
- Department
of Pharmacy, University of Pisa, Via Bonanno
6, I-56126 Pisa, Italy
| | - Alessandro Maiocchi
- Centro Ricerche Bracco, Bracco Imaging S.p.A., Via Ribes 5, I-10010 Colleretto
Giacosa (TO), Italy
| |
Collapse
|
24
|
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
|
25
|
Abstract
The matrix metalloproteinases (MMPs) exhibit a broad array of activities, some catalytic and some non-catalytic in nature. An overall lack of selectivity has rendered small molecule, active site targeted MMP inhibitors problematic in execution. Inhibitors that favor few or individual members of the MMP family often take advantage of interactions outside the enzyme active site. We presently focus on peptide-based MMP inhibitors and probes that do not incorporate conventional Zn2+ binding groups. In some cases, these inhibitors and probes function by binding only secondary binding sites (exosites), while others bind both exosites and the active site. A myriad of MMP mediated-activities beyond selective catalysis can be inhibited by peptides, particularly cell adhesion, proliferation, motility, and invasion. Selective MMP binding peptides comprise highly customizable, unique imaging agents. Areas of needed improvement for MMP targeting peptides include binding affinity and stability.
Collapse
|
26
|
Da Rocha Gomes S, Miguel J, Azéma L, Eimer S, Ries C, Dausse E, Loiseau H, Allard M, Toulmé JJ. (99m)Tc-MAG3-aptamer for imaging human tumors associated with high level of matrix metalloprotease-9. Bioconjug Chem 2012; 23:2192-200. [PMID: 23043415 DOI: 10.1021/bc300146c] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The human matrix metalloprotease 9 (hMMP-9) is involved in many physiological processes such as tissue remodeling. Its overexpression in tumors promotes the release of cancer cells thus contributing to tumor metastasis. It is a relevant marker of malignant tumors. We selected an RNA aptamer containing 2'-fluoro, pyrimidine ribonucleosides, that exhibits a strong affinity for hMMP-9 (K(d) = 20 nM) and that discriminates other human MMPs: no binding was detected to either hMMP-2 or -7. Investigating the binding properties of different MMP-9 aptamer variants by surface plasmon resonance allowed the determination of recognition elements. As a result, a truncated aptamer, 36 nucleotides long, was made fully resistant to nuclease following the substitution of every purine ribonucleoside residue by 2'-O-methyl analogues and was conjugated to S-acetylmercaptoacetyltriglycine for imaging purposes. The resulting modified aptamer retained the binding properties of the originally selected sequence. Following (99m)Tc labeling, this aptamer was used for ex vivo imaging slices of human brain tumors. We were able to specifically detect the presence of hMMP-9 in such tissues.
Collapse
Affiliation(s)
- Sonia Da Rocha Gomes
- INSERM U869, ARNA, Institut Européen de Chimie et Biologie, 33607 Pessac, France
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Quillard T, Libby P. Molecular imaging of atherosclerosis for improving diagnostic and therapeutic development. Circ Res 2012; 111:231-44. [PMID: 22773426 DOI: 10.1161/circresaha.112.268144] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Despite recent progress, cardiovascular and allied metabolic disorders remain a worldwide health challenge. We must identify new targets for therapy, develop new agents for clinical use, and deploy them in a clinically effective and cost-effective manner. Molecular imaging of atherosclerotic lesions has become a major experimental tool in the last decade, notably by providing a direct gateway to the processes involved in atherogenesis and its complications. This review summarizes the current status of molecular imaging approaches that target the key processes implicated in plaque formation, development, and disruption and highlights how the refinement and application of such tools might aid the development and evaluation of novel therapeutics.
Collapse
Affiliation(s)
- Thibaut Quillard
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
28
|
Chuang CH, Chuang KH, Wang HE, Roffler SR, Shiea JT, Tzou SC, Cheng TC, Kao CH, Wu SY, Tseng WL, Cheng CM, Hou MF, Wang JM, Cheng TL. In vivo positron emission tomography imaging of protease activity by generation of a hydrophobic product from a noninhibitory protease substrate. Clin Cancer Res 2011; 18:238-47. [PMID: 22019516 DOI: 10.1158/1078-0432.ccr-11-0608] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To develop an imaging technology for protease activities in patients that could help in prognosis prediction and in design of personalized, protease-based inhibitors and prodrugs for targeted therapy. EXPERIMENTAL DESIGN Polyethylene glycol (PEG) was covalently attached to the N-terminus of a hydrophilic peptide substrate (GPLGVR) for matrix metalloproteinase (MMP) to increase hydrophilicity. PEG-peptide was then linked to a hydrophobic tetramethylrhodamine (TMR) domain and labeled with (18)F to form a PEG-peptide-(18)F-TMR probe. Specific cleavage of the probe by MMP2 was tested in vitro by matrix-assisted laser desorption/ionization-time-of-flight (MALDI-TOF). In vivo imaging of MMP2-expressing tumors was evaluated by micro-PET. RESULTS The hydrophobic TMR fragment (948 Da) was specifically generated by MMP2 enzymes and MMP-expressing HT1080 cells but not control MCF-7 cells. MMP-expressing HT1080 cells and tumors selectively accumulated the hydrolyzed, hydrophobic TMR fragment at sites of protease activity. Importantly, we found that (18)F-labeled probe ((18)F-TMR) preferentially localized in HT1080 tumors but not control MCF-7 tumors as shown by micro-PET. Uptake of the probe in HT1080 tumors was 18.4 ± 1.9-fold greater than in the MCF-7 tumors 30 minutes after injection. These results suggest that the PEG-peptide-(18)F-TMR probe displays high selectivity for imaging MMP activity. CONCLUSIONS This strategy successfully images MMP expression in vivo and may be extended to other proteases to predict patient prognosis and to design personalized, protease-based inhibitors and prodrug-targeted therapies.
Collapse
Affiliation(s)
- Chih-Hung Chuang
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Animal tumor models for PET in drug development. Ann Nucl Med 2011; 25:717-31. [DOI: 10.1007/s12149-011-0531-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 08/16/2011] [Indexed: 10/17/2022]
|
30
|
Rodenberg E, Azhdarinia A, Lazard ZW, Hall M, Kwon SK, Wilganowski N, Salisbury EA, Merched-Sauvage M, Olmsted-Davis EA, Sevick-Muraca EM, Davis AR. Matrix metalloproteinase-9 is a diagnostic marker of heterotopic ossification in a murine model. Tissue Eng Part A 2011; 17:2487-96. [PMID: 21599541 DOI: 10.1089/ten.tea.2011.0007] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Heterotopic ossification (HO) is a serious disorder that occurs when there is aberrant bone morphogenic protein (BMP) signaling in soft tissues. Currently, there are no methods to detect HO before mineralization occurs. Yet once mineralization occurs, there are no effective treatments, short of surgery, to reverse HO. Herein, we used in vivo molecular imaging and confirmatory ex vivo tissue analyses of an established murine animal model of BMP-induced HO to show that matrix metalloproteinase-9 (MMP-9) can be detected as an early-stage biomarker before mineralization. Ex vivo analyses show that active MMP-9 protein is significantly elevated within tissues undergoing HO as early as 48 h after BMP induction, with its expression co-localizing to nerves and vessels. In vivo molecular imaging with a dual-labeled near-infrared fluorescence and micro-positron emission tomography (μPET) agent specific to MMP-2/-9 expression paralleled the ex vivo observations and reflected the site of HO formation as detected from microcomputed tomography 7 days later. The results suggest that the MMP-9 is a biomarker of the early extracellular matrix (ECM) re-organization and could be used as an in vivo diagnostic with confirmatory ex vivo tissue analysis for detecting HO or conversely for monitoring the success of tissue-engineered bone implants that employ ECM biology for engraftment.
Collapse
Affiliation(s)
- Eric Rodenberg
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Azhdarinia A, Wilganowski N, Robinson H, Ghosh P, Kwon S, Lazard ZW, Davis AR, Olmsted-Davis E, Sevick-Muraca EM. Characterization of chemical, radiochemical and optical properties of a dual-labeled MMP-9 targeting peptide. Bioorg Med Chem 2011; 19:3769-76. [PMID: 21612930 DOI: 10.1016/j.bmc.2011.04.054] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Revised: 04/25/2011] [Accepted: 04/30/2011] [Indexed: 10/18/2022]
Abstract
Optical imaging possesses similar sensitivity to nuclear imaging and has led to the emergence of multimodal approaches with dual-labeled nuclear/near-infrared (NIR) agents. The growing impact of (68)Ga (t(1/2)=68 min) labeled peptides on preclinical and clinical research offers a promising opportunity to merge the high spatial resolution of NIR imaging with the clinically-accepted positron emission tomography (PET). Previously, dual-labeled agents have been prepared with longer-lived radiometals and showed no detrimental effects on optical properties as a result of radiolabeling. In this study, we selected a peptide (M(2)) that targets MMP-2/9 and is dual-labeled with IRDye 800 CW and (68)Ga. Since (68)Ga chelation typically requires low pH (3.5-4) and elevated heating temperatures (95 °C), we sought to evaluate the impact of (68)Ga labeling on the optical properties of M(2). An efficient method for preparation of (68)Ga-M(2) was developed and reaction conditions were optimized. Stability studies in PBS, DTPA, and serum were performed and high levels of intact agent were evident under each condition. The addition of multiple reporters to a targeting agent adds further complexity to the characterization and validation and thus requires not only testing to ensure the agent is stable chemically and radiochemically, but also optically. Therefore, fluorescence properties were evaluated using a spectrofluorometer as well as by fluorescence detection via HPLC. It was determined that (68)Ga-labeling conditions did not impair the fluorescent properties of the agent. The agent was then used for in vivo imaging in a mouse model of heterotopic ossification (HO) with activated MMP-9 expression as an early biomarker which precedes mineralization. Although (68)Ga-complexation greatly reduced binding affinity of the peptide and negated tracer uptake on PET, NIR imaging showed consistent fluorescent signal that correlated to MMP-9 expression. This attests to the feasibility of using (68)Ga/NIR for dual-labeling of other peptides or small molecules for multimodality molecular 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
|
32
|
Cochran R, Cochran F. Phage display and molecular imaging: expanding fields of vision in living subjects. Biotechnol Genet Eng Rev 2011; 27:57-94. [PMID: 21415893 DOI: 10.1080/02648725.2010.10648145] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In vivo molecular imaging enables non-invasive visualization of biological processes within living subjects, and holds great promise for diagnosis and monitoring of disease. The ability to create new agents that bind to molecular targets and deliver imaging probes to desired locations in the body is critically important to further advance this field. To address this need, phage display, an established technology for the discovery and development of novel binding agents, is increasingly becoming a key component of many molecular imaging research programs. This review discusses the expanding role played by phage display in the field of molecular imaging with a focus on in vivo applications. Furthermore, new methodological advances in phage display that can be directly applied to the discovery and development of molecular imaging agents are described. Various phage library selection strategies are summarized and compared, including selections against purified target, intact cells, and ex vivo tissue, plus in vivo homing strategies. An outline of the process for converting polypeptides obtained from phage display library selections into successful in vivo imaging agents is provided, including strategies to optimize in vivo performance. Additionally, the use of phage particles as imaging agents is also described. In the latter part of the review, a survey of phage-derived in vivo imaging agents is presented, and important recent examples are highlighted. Other imaging applications are also discussed, such as the development of peptide tags for site-specific protein labeling and the use of phage as delivery agents for reporter genes. The review concludes with a discussion of how phage display technology will continue to impact both basic science and clinical applications in the field of molecular imaging.
Collapse
Affiliation(s)
- R Cochran
- Department of Bioengineering, Cancer Center, Bio-X Program, Stanford University, Stanford CA, USA
| | | |
Collapse
|
33
|
van Oosten M, Crane LM, Bart J, van Leeuwen FW, van Dam GM. Selecting Potential Targetable Biomarkers for Imaging Purposes in Colorectal Cancer Using TArget Selection Criteria (TASC): A Novel Target Identification Tool. Transl Oncol 2011; 4:71-82. [PMID: 21461170 PMCID: PMC3069650 DOI: 10.1593/tlo.10220] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Revised: 10/23/2010] [Accepted: 11/01/2010] [Indexed: 12/19/2022] Open
Abstract
Peritoneal carcinomatosis (PC) of colorectal origin is associated with a poor prognosis. However, cytoreductive surgery combined with hyperthermic intraperitoneal chemotherapy is available for a selected group of PC patients, which significantly increases overall survival rates up to 30%. As a consequence, there is substantial room for improvement. Tumor targeting is expected to improve the treatment efficacy of colorectal cancer (CRC) further through 1) more sensitive preoperative tumor detection, thus reducing overtreatment; 2) better intraoperative detection and surgical elimination of residual disease using tumor-specific intraoperative imaging; and 3) tumor-specific targeted therapeutics. This review focuses, in particular, on the development of tumor-targeted imaging agents. A large number of biomarkers are known to be upregulated in CRC. However, to date, no validated criteria have been described for the selection of the most promising biomarkers for tumor targeting. Such a scoring system might improve the selection of the correct biomarker for imaging purposes. In this review, we present the TArget Selection Criteria (TASC) scoring system for selection of potential biomarkers for tumor-targeted imaging. By applying TASC to biomarkers for CRC, we identified seven biomarkers (carcinoembryonic antigen, CXC chemokine receptor 4, epidermal growth factor receptor, epithelial cell adhesion molecule, matrix metalloproteinases, mucin 1, and vascular endothelial growth factor A) that seem most suitable for tumor-targeted imaging applications in colorectal cancer. Further cross-validation studies in CRC and other tumor types are necessary to establish its definitive value.
Collapse
Affiliation(s)
- Marleen van Oosten
- Department of Surgery, Division of Surgical Oncology, Surgical Research Laboratory/BioOptical Imaging Center, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | | | | | | |
Collapse
|
34
|
Quillard T, Croce K, Jaffer FA, Weissleder R, Libby P. Molecular imaging of macrophage protease activity in cardiovascular inflammation in vivo. Thromb Haemost 2011; 105:828-36. [PMID: 21225096 DOI: 10.1160/th10-09-0589] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Accepted: 11/21/2010] [Indexed: 01/01/2023]
Abstract
Macrophages contribute pivotally to cardiovascular diseases (CVD), notably to atherosclerosis. Imaging of macrophages in vivo could furnish new tools to advance evaluation of disease and therapies. Proteolytic enzymes serve as key effectors of many macrophage contributions to CVD. Therefore, intravital imaging of protease activity could aid evaluation of the progress and outcome of atherosclerosis, aortic aneurysm formation, or rejection of cardiac allografts. Among the large families of proteases, matrix metalloproteinases (MMPs) and cysteinyl cathepsins have garnered the most interest because of their participation in extracellular matrix remodelling. These considerations have spurred the development of dedicated imaging agents for protease activity detection. Activatable fluorescent probes, radiolabelled inhibitors, and nanoparticles are currently under exploration for this purpose. While some agents and technologies may soon see clinical use, others will require further refinement. Imaging of macrophages and protease activity should provide an important adjunct to understanding pathophysiology in vivo, evaluating the effects of interventions, and ultimately aiding clinical care.
Collapse
Affiliation(s)
- T Quillard
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | |
Collapse
|
35
|
Li Y, Ting R, Harwig CW, auf dem Keller U, Bellac CL, Lange PF, Inkster JAH, Schaffer P, Adam MJ, Ruth TJ, Overall CM, Perrin DM. Towards kit-like 18F-labeling of marimastat, a noncovalent inhibitor drug for in vivo PET imaging cancer associated matrix metalloproteases. MEDCHEMCOMM 2011. [DOI: 10.1039/c1md00117e] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
36
|
Liu Y, Solomon M, Achilefu S. Perspectives and potential applications of nanomedicine in breast and prostate cancer. Med Res Rev 2010; 33:3-32. [PMID: 23239045 DOI: 10.1002/med.20233] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Nanomedicine is a branch of nanotechnology that includes the development of nanostructures and nanoanalytical systems for various medical applications. Among these applications, utilization of nanotechnology in oncology has captivated the attention of many research endeavors in recent years. The rapid development of nano-oncology raises new possibilities in cancer diagnosis and treatment. It also holds great promise for realization of point-of-care, theranostics, and personalized medicine. In this article, we review advances in nano-oncology, with an emphasis on breast and prostate cancer because these organs are amenable to the translation of nanomedicine from small animals to humans. As new drugs are developed, the incorporation of nanotechnology approaches into medicinal research becomes critical. Diverse aspects of nano-oncology are discussed, including nanocarriers, targeting strategies, nanodevices, as well as nanomedical diagnostics, therapeutics, and safety. The review concludes by identifying some limitations and future perspectives of nano-oncology in breast and prostate cancer management.
Collapse
Affiliation(s)
- Yang Liu
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | | |
Collapse
|
37
|
Ujula T, Huttunen M, Luoto P, Peräkylä H, Simpura I, Wilson I, Bergman M, Roivainen A. Matrix Metalloproteinase 9 Targeting Peptides: Syntheses, 68Ga-labeling, and Preliminary Evaluation in a Rat Melanoma Xenograft Model. Bioconjug Chem 2010; 21:1612-21. [DOI: 10.1021/bc1000643] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Tiina Ujula
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland, Karyon-CTT Ltd., Helsinki, Finland, Turku Imanet, GE Healthcare Medical Diagnostics, Turku, Finland, and Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Merja Huttunen
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland, Karyon-CTT Ltd., Helsinki, Finland, Turku Imanet, GE Healthcare Medical Diagnostics, Turku, Finland, and Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Pauliina Luoto
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland, Karyon-CTT Ltd., Helsinki, Finland, Turku Imanet, GE Healthcare Medical Diagnostics, Turku, Finland, and Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Hannu Peräkylä
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland, Karyon-CTT Ltd., Helsinki, Finland, Turku Imanet, GE Healthcare Medical Diagnostics, Turku, Finland, and Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Ilkka Simpura
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland, Karyon-CTT Ltd., Helsinki, Finland, Turku Imanet, GE Healthcare Medical Diagnostics, Turku, Finland, and Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Ian Wilson
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland, Karyon-CTT Ltd., Helsinki, Finland, Turku Imanet, GE Healthcare Medical Diagnostics, Turku, Finland, and Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Mathias Bergman
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland, Karyon-CTT Ltd., Helsinki, Finland, Turku Imanet, GE Healthcare Medical Diagnostics, Turku, Finland, and Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Anne Roivainen
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland, Karyon-CTT Ltd., Helsinki, Finland, Turku Imanet, GE Healthcare Medical Diagnostics, Turku, Finland, and Turku Center for Disease Modeling, University of Turku, Turku, Finland
| |
Collapse
|
38
|
Michalski MH, Chen X. Molecular imaging in cancer treatment. Eur J Nucl Med Mol Imaging 2010; 38:358-77. [PMID: 20661557 DOI: 10.1007/s00259-010-1569-z] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2010] [Accepted: 07/12/2010] [Indexed: 12/19/2022]
Abstract
The success of cancer therapy can be difficult to predict, as its efficacy is often predicated upon characteristics of the cancer, treatment, and individual that are not fully understood or are difficult to ascertain. Monitoring the response of disease to treatment is therefore essential and has traditionally been characterized by changes in tumor volume. However, in many instances, this singular measure is insufficient for predicting treatment effects on patient survival. Molecular imaging allows repeated in vivo measurement of many critical molecular features of neoplasm, such as metabolism, proliferation, angiogenesis, hypoxia, and apoptosis, which can be employed for monitoring therapeutic response. In this review, we examine the current methods for evaluating response to treatment and provide an overview of emerging PET molecular imaging methods that will help guide future cancer therapies.
Collapse
|
39
|
auf dem Keller U, Schilling O. Proteomic techniques and activity-based probes for the system-wide study of proteolysis. Biochimie 2010; 92:1705-14. [PMID: 20493233 DOI: 10.1016/j.biochi.2010.04.027] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2010] [Accepted: 04/29/2010] [Indexed: 11/17/2022]
Abstract
Proteolysis constitutes a major post-translational modification but specificity and substrate selectivity of numerous proteases have remained elusive. In this review, we highlight how advanced techniques in the areas of proteomics and activity-based probes can be used to investigate i) protease active site specificity; ii) protease in vivo substrates; iii) protease contribution to proteome homeostasis and composition; and iv) detection and localization of active proteases. Peptide libraries together with genetical or biochemical selection have traditionally been used for active site profiling of proteases. These are now complemented by proteome-derived peptide libraries that simultaneously determine prime and non-prime specificity and characterize subsite cooperativity. Cell-contextual discovery of protease substrates is rendered possible by techniques that isolate and quantitate protein termini. Here, a novel approach termed Terminal Amine Isotopic Labeling of Substrates (TAILS) provides an integrated platform for substrate discovery and appropriate statistical evaluation of terminal peptide identification and quantification. Proteolytically generated carboxy-termini can now also be analyzed on a proteome-wide level. Proteolytic regulation of proteome composition is monitored by quantitative proteomic approaches employing stable isotope coding or label free quantification. Activity-based probes specifically recognize active proteases. In proteomic screens, they can be used to detect and quantitate proteolytic activity while their application in cellular histology allows to locate proteolytic activity in situ. Activity-based probes - especially in conjunction with positron emission tomography - are also promising tools to monitor proteolytic activities on an organism-wide basis with a focus on in vivo tumor imaging. Together, this array of methodological possibilities enables unveiling physiological protease substrate repertoires and defining protease function in the cellular- and organism-wide context.
Collapse
Affiliation(s)
- Ulrich auf dem Keller
- ETH Zürich Institute of Cell Biology, Schafmattstrasse 18, CH-8093 Zurich, Switzerland
| | | |
Collapse
|
40
|
Dijkgraaf I, Boerman OC. Radionuclide imaging of tumor angiogenesis. Cancer Biother Radiopharm 2010; 24:637-47. [PMID: 20025543 DOI: 10.1089/cbr.2009.0694] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Angiogenesis is a multistep process regulated by pro- and antiangiogenic factors. In order to grow and metastasize, tumors need a constant supply of oxygen and nutrients. For growth beyond 1-2 mm in size, tumors are dependent on angiogenesis. Inhibition of angiogenesis is a new cancer treatment strategy that is now widely investigated clinically. Researchers have begun to search for objective measures that indicate pharmacologic responses to antiangiogenic drugs. Therefore, there is a great interest in techniques to visualize angiogenesis in growing tumors noninvasively. Several markers have been described that are preferentially expressed on newly formed blood vessels in tumors (alpha(v)beta(3) integrin, vascular endothelial growth factor, and its receptor, prostate-specific membrane antigen) and in the extracellular matrix surrounding newly formed blood vessels (extra domain B of fibronectin, Tenascin-C, matrix metalloproteinases, and Robo-4). Several ligands targeting these markers have been tested as a radiotracer for imaging angiogenesis in tumors. The potential of some of these tracers, such as radiolabeled cyclic RGD peptides and radiolabeled anti-PSMA antibodies, has already been tested in cancer patients, while for markers such as Robo-4, the ligand has not yet been identified. In this review, an overview on the currently used nuclear imaging probes for noninvasive visualization of tumor angiogenesis is given.
Collapse
Affiliation(s)
- Ingrid Dijkgraaf
- Department of Nuclear Medicine, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands.
| | | |
Collapse
|
41
|
Chebbi I, Migianu-Griffoni E, Sainte-Catherine O, Lecouvey M, Seksek O. In vitro assessment of liposomal neridronate on MDA-MB-231 human breast cancer cells. Int J Pharm 2009; 383:116-22. [PMID: 19748562 DOI: 10.1016/j.ijpharm.2009.09.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2009] [Revised: 09/04/2009] [Accepted: 09/07/2009] [Indexed: 11/28/2022]
Abstract
Bisphosphonates have been used for decades in the standard therapy of bone-related diseases, including bone metastasis of various malignancies, and they might as well be toxic on early cancer cells themselves. In order to allow a better delivery of neridronate (a N-containing bisphosphonate with relatively poor activity), liposomes were evaluated in vitro on cancer cell lines (MDA-MB-231, U87-MG and Caco2). After chemical synthesis, this water-soluble molecule was encapsulated into liposomes containing DOPC:DOPG:Chol (72:27:1 molar ratio). The influence of neridronate (free or liposomal) on cell viability or proliferation after treatment was evaluated using the MTT method, as well as cell migration and invasion assays; these techniques showed a drastic improvement of the action of neridronate on MDA-MB-231 cells with an EC(50) 50 times lower when neridronate was encapsulated. Internalization of liposomes was followed by flow cytometry and fluorescence microscopy, demonstrating internalization via the endocytic pathway. Furthermore, since overexpression of matrix metalloproteinases (particularly MMP-2 and MMP-9) has been correlated to poor prognosis in many cancer types, detection of MMP expression is a satisfactory indication of the therapy efficiency and was then performed on treated cells. On MDA-MB-231 cells, MPPs expression was also significantly reduced by neridronate while entrapped in liposomes.
Collapse
Affiliation(s)
- Imène Chebbi
- ANBioPhy, CNRS FRE 3207, Université Pierre & Marie Curie, Génopole Campus 1, Evry, France
| | | | | | | | | |
Collapse
|
42
|
Yang Y, Hong H, Zhang Y, Cai W. Molecular Imaging of Proteases in Cancer. CANCER GROWTH AND METASTASIS 2009; 2:13-27. [PMID: 20234801 DOI: 10.4137/cgm.s2814] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Proteases play important roles during tumor angiogenesis, invasion, and metastasis. Various molecular imaging techniques have been employed for protease imaging: optical (both fluorescence and bioluminescence), magnetic resonance imaging (MRI), single-photon emission computed tomography (SPECT), and positron emission tomography (PET). In this review, we will summarize the current status of imaging proteases in cancer with these techniques. Optical imaging of proteases, in particular with fluorescence, is the most intensively validated and many of the imaging probes are already commercially available. It is generally agreed that the use of activatable probes is the most accurate and appropriate means for measuring protease activity. Molecular imaging of proteases with other techniques (i.e. MRI, SPECT, and PET) has not been well-documented in the literature which certainly deserves much future effort. Optical imaging and molecular MRI of protease activity has very limited potential for clinical investigation. PET/SPECT imaging is suitable for clinical investigation; however the optimal probes for PET/SPECT imaging of proteases in cancer have yet to be developed. Successful development of protease imaging probes with optimal in vivo stability, tumor targeting efficacy, and desirable pharmacokinetics for clinical translation will eventually improve cancer patient management. Not limited to cancer, these protease-targeted imaging probes will also have broad applications in other diseases such as arthritis, atherosclerosis, and myocardial infarction.
Collapse
|
43
|
Sun Y, Lu N, Ling Y, Gao Y, Chen Y, Wang L, Hu R, Qi Q, Liu W, Yang Y, You Q, Guo Q. Oroxylin A suppresses invasion through down-regulating the expression of matrix metalloproteinase-2/9 in MDA-MB-435 human breast cancer cells. Eur J Pharmacol 2009; 603:22-8. [DOI: 10.1016/j.ejphar.2008.12.008] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2008] [Revised: 11/18/2008] [Accepted: 12/01/2008] [Indexed: 10/21/2022]
|
44
|
Abstract
Angiogenesis is a highly-controlled process that is dependent on the intricate balance of both promoting and inhibiting factors, involved in various physiological and pathological processes. A comprehensive understanding of the molecular mechanisms that regulate angiogenesis has resulted in the design of new and more effective therapeutic strategies. Due to insufficient sensitivity to detect therapeutic effects by using standard clinical endpoints or by looking for physiological improvement, a multitude of imaging techniques have been developed to assess tissue vasculature on the structural, functional and molecular level. Imaging is expected to provide a novel approach to noninvasively monitor angiogenesis, to optimize the dose of new antiangiogenic agents and to assess the efficacy of therapies directed at modulation of the angiogenic process. All these methods have been successfully used preclinically and will hopefully aid in antiangiogenic drug development in animal studies. In this review article, the application of PET in angiogenesis imaging at both functional and molecular level will be discussed. For PET imaging of angiogenesis related molecular markers, we emphasize integrin alpha(v)beta(3), VEGF/VEGFR, and MMPs.
Collapse
|
45
|
Development of an optimized activatable MMP-14 targeted SPECT imaging probe. Bioorg Med Chem 2008; 17:653-9. [PMID: 19109023 DOI: 10.1016/j.bmc.2008.11.078] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2008] [Revised: 11/18/2008] [Accepted: 11/23/2008] [Indexed: 11/20/2022]
Abstract
Matrix metalloproteinase-14 (MT1-MMP or MMP-14) is a membrane-associated protease implicated in a variety of tissue remodeling processes and a molecular hallmark of select metastatic cancers. The ability to detect MMP-14 in vivo would be useful in studying its role in pathologic processes and may potentially serve as a guide for the development of targeted molecular therapies. Four MMP-14 specific probes containing a positively charged cell penetrating peptide (CPP) d-arginine octamer (r(8)) linked with a MMP-14 peptide substrate and attenuating sequences with glutamate (8e, 4e) or glutamate-glycine (4eg and 4egg) repeating units were modeled using an AMBER force field method. The probe with 4egg attenuating sequence exhibited the highest CPP/attenuator interaction, predicting minimized cellular uptake until cleaved. The in vitro MMP-14-mediated cleavage studies using the human recombinant MMP-14 catalytic domain revealed an enhanced cleavage rate that directly correlated with the linearity of the embedded peptide substrate sequence. Successful cleavage and uptake of a technetium-99m labeled version of the optimal probe was demonstrated in MMP-14 transfected human breast cancer cells. Two-fold reduction of cellular uptake was found in the presence of a broad spectrum MMP inhibitor. The combination of computational chemistry, parallel synthesis and biochemical screening, therefore, shows promise as a set of tools for developing new radiolabeled probes that are sensitive to protease activity.
Collapse
|
46
|
Kimura Y, Sumiyoshi M, Baba K. Antitumor activities of synthetic and natural stilbenes through antiangiogenic action. Cancer Sci 2008; 99:2083-96. [PMID: 19016770 PMCID: PMC11158896 DOI: 10.1111/j.1349-7006.2008.00948.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2008] [Revised: 06/22/2008] [Accepted: 06/27/2008] [Indexed: 01/22/2023] Open
Abstract
We reported that the antitumor and antimetastatic actions of resveratrol might be due to the inhibition of tumor-induced angiogenesis. To search for anticancer agents with stronger activity than resveratrol, we examined the antiangiogenic effects of 21 synthetic and/or natural stilbenes. Among these 21 stilbenes, 2,3-, 3,4-, and 4,4'-dihydroxystilbene inhibited the pro-matrix metalloproteinase (pro-MMP)-9 production in colon 26 cells at 5-25 microM, vascular endothelial growth factor (VEGF)-induced human umbilical vein endothelial cell (HUVEC) migration at 10 and 25 microM, and VEGF-induced angiogenesis at 5-50 microM. Resvertarol inhibited the pro-MMP-9 production and VEGF-induced angiogenesis at 25 or 50 microM. Thus, the inhibition of pro-MMP-9 production in colon 26 cells and VEGF-induced angiogenesis by three dihydroxystilbenes were greater than those of resveratrol. The three dihydroxystilbenes (8 mg/kg, intraperitoneal injection) inhibited the tumor-induced neovascularization in colon 26-packed chamber-bearing mice and the tumor growth in colon 26-bearing mice. Furthermore, the three dihydroxystilbenes inhibited VEGF-induced VEGFR-2 phosphorylation. On the other hand, the three dihydroxystilbenes had no effect on VEGFR-1 and-2 expression, and VEGF-induced VEGFR-1 phosphorylation in HUVECs. These findings suggest that the inhibition of tumor-induced neovascularization by these three dihydroxystilbenes may be due to the inhibition of VEGF-induced endothelial cell migration and VEGF-induced angiogenesis through the inhibition of VEGF-induced VEGFR-2 phosphorylation in endothelial cells and pro-MMP-9 expression in colon 26 cells.
Collapse
Affiliation(s)
- Yoshiyuki Kimura
- Division of Biochemical Pharmacology, Department of Basic Medical Research, Graduate School of Medicine, Ehime University, Toon City, Ehime, Japan.
| | | | | |
Collapse
|
47
|
Synthesis of a beta-tetrapeptide analog as a mother compound for the development of matrix metalloproteinase-2-imaging agents. Chem Pharm Bull (Tokyo) 2008; 56:260-5. [PMID: 18310933 DOI: 10.1248/cpb.56.260] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Matrix metalloproteinase-2 (MMP-2) is an attractive target for the diagnosis of cancer and atherosclerosis in nuclear imaging. A cyclic decapeptide, cCTTHWGFTLC (cCTT), has been used as the mother compound for the development of MMP-2-imaging agents with high potency and selectivity. Most of radiolabeled derivatives of cCTT currently developed for in vivo studies of MMP-2, however, suffer from low accumulation in the target tissues, such as tumors. For enhanced in vivo stability and tissue penetration, we designed a linear beta-tetrapeptide analog, H-beta 3-Phe-beta-Ala-beta 3-Trp-beta 3-His-OH (1), to mimic cCTT. The component beta-amino acids were prepared by reduction of N-protected alpha-amino acid methyl esters to the alcohols, followed by conversion into the cyanides, and subsequent hydrolysis. Compound 1 was obtained from these beta-amino acids by the conventional solution method. In MMP-2 inhibition assay, compound 1 displayed desirably significant inhibition, which was comparable to cCTT. These findings suggest that compound 1 may serve as a mother compound in the design and development of in vivo MMP-2-imaging agents.
Collapse
|
48
|
Lepage M, Dow WC, Melchior M, You Y, Fingleton B, Quarles CC, Pépin C, Gore JC, Matrisian LM, McIntyre JO. Noninvasive Detection of Matrix Metalloproteinase Activity In Vivo using a Novel Magnetic Resonance Imaging Contrast Agent with a Solubility Switch. Mol Imaging 2007. [DOI: 10.2310/7290.2007.00035] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Martin Lepage
- From the Vanderbilt University Insitute of Imaging Science, Department of Cancer Biology, and Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN; Sherbrooke Molecular Imaging Centre, Université de Sherbrooke, Sherbrooke, QC; and Alerion Biomedical Inc., San Diego, CA
| | - William C. Dow
- From the Vanderbilt University Insitute of Imaging Science, Department of Cancer Biology, and Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN; Sherbrooke Molecular Imaging Centre, Université de Sherbrooke, Sherbrooke, QC; and Alerion Biomedical Inc., San Diego, CA
| | - Marco Melchior
- From the Vanderbilt University Insitute of Imaging Science, Department of Cancer Biology, and Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN; Sherbrooke Molecular Imaging Centre, Université de Sherbrooke, Sherbrooke, QC; and Alerion Biomedical Inc., San Diego, CA
| | - Ying You
- From the Vanderbilt University Insitute of Imaging Science, Department of Cancer Biology, and Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN; Sherbrooke Molecular Imaging Centre, Université de Sherbrooke, Sherbrooke, QC; and Alerion Biomedical Inc., San Diego, CA
| | - Barbara Fingleton
- From the Vanderbilt University Insitute of Imaging Science, Department of Cancer Biology, and Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN; Sherbrooke Molecular Imaging Centre, Université de Sherbrooke, Sherbrooke, QC; and Alerion Biomedical Inc., San Diego, CA
| | - C. Chad Quarles
- From the Vanderbilt University Insitute of Imaging Science, Department of Cancer Biology, and Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN; Sherbrooke Molecular Imaging Centre, Université de Sherbrooke, Sherbrooke, QC; and Alerion Biomedical Inc., San Diego, CA
| | - Claude Pépin
- From the Vanderbilt University Insitute of Imaging Science, Department of Cancer Biology, and Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN; Sherbrooke Molecular Imaging Centre, Université de Sherbrooke, Sherbrooke, QC; and Alerion Biomedical Inc., San Diego, CA
| | - John C. Gore
- From the Vanderbilt University Insitute of Imaging Science, Department of Cancer Biology, and Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN; Sherbrooke Molecular Imaging Centre, Université de Sherbrooke, Sherbrooke, QC; and Alerion Biomedical Inc., San Diego, CA
| | - Lynn M. Matrisian
- From the Vanderbilt University Insitute of Imaging Science, Department of Cancer Biology, and Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN; Sherbrooke Molecular Imaging Centre, Université de Sherbrooke, Sherbrooke, QC; and Alerion Biomedical Inc., San Diego, CA
| | - J. Oliver McIntyre
- From the Vanderbilt University Insitute of Imaging Science, Department of Cancer Biology, and Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN; Sherbrooke Molecular Imaging Centre, Université de Sherbrooke, Sherbrooke, QC; and Alerion Biomedical Inc., San Diego, CA
| |
Collapse
|
49
|
Park JB, Lee JK, Cho ST, Park EY, Riew KD. A biochemical mechanism for resistance of intervertebral discs to metastatic cancer: Fas ligand produced by disc cells induces apoptotic cell death of cancer cells. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2007; 16:1319-24. [PMID: 17684774 PMCID: PMC2200753 DOI: 10.1007/s00586-007-0463-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2007] [Accepted: 07/23/2007] [Indexed: 11/24/2022]
Abstract
Metastatic spinal cancer is characterized by the maintenance of normal disc structure until the vertebral body is severely destroyed by cancer cells. Anatomic features of the discs have been thought to be the main factor which confer the discs their resistance to metastatic cancer. However, little is known about the biochemical mechanism to prevent or attenuate the local infiltration of cancer cells into the discs. The purpose of this study was to investigate whether Fas ligand (FasL) produced by disc cells can kill Fas-bearing breast cancer cells by Fas and FasL interaction. Two human breast cancer cells (MCF-7 and MDA-MB-231) were obtained and cultured (1 x 10(6) cells/well), and the expression of Fas was investigated by western blot analysis. Annulus fibrosus cells were isolated and cultured, and the presence of FasL was quantified in the supernatants of three different numbers of annulus fibrosus cells (1x, 2x, and 4 x 10(6) cells/well) by ELISA assay. The MCF-7 and MDA-MB-231 cancer cells were cultured with supernatants of annulus fibrosus cells for 48 h. As controls, MCF-7 and MDA-MB-231 cancer cells were also cultured by themselves for 48 h. Finally, we determined and quantified the apoptosis rates of MCF-7 and MDA-MB-231 cancer cells by Annexin V-FITC and PI and TUNEL at 48 h, respectively. The expression of Fas was identified in MCF-7 and MDA-MB-231 cancer cells. The mean concentrations of FasL in supernatants of annulus fibrosus cells (1x, 2x, and 4 x 10(6) cells/well) were 10.8, 29.6, and 56.4 pg/mL, respectively. After treatment with the supernatant of three different numbers of annulus fibrosus cells, the mean apoptosis rate of MCF-7 cancer cells was increased (2.8%, P < 0.01; 6.7%, P < 0.001; 31.0%, P < 0.001) in a dose-dependent manner of FasL compared to that of control (1.1%). The mean apoptosis rate of MDA-MB-231 cancer cells was also increased (5.7%, P < 0.01; 11.1%, P < 0.001; 25.3%, P < 0.001) in a dose-dependent manner of FasL compared to that of control (2.1%). TUNEL also demonstrated direct evidence of apoptosis of MCF-7 and MDA-MB-231 cancer cells. Our results demonstrate that Fas-bearing cancer cells undergo apoptosis by FasL produced by disc cells, which may be considered as a potential biochemical explanation for the disc's resistance to metastatic cancer.
Collapse
Affiliation(s)
- Jong-Beom Park
- Department of Orthopaedic Surgery, The Catholic University of Korea School of Medicine, Uijongbu-si, Kyunggi-do 480-717, South Korea.
| | | | | | | | | |
Collapse
|
50
|
Park JB, Lee JK, Park EY, Riew KD. Fas/FasL interaction of nucleus pulposus and cancer cells with the activation of caspases. INTERNATIONAL ORTHOPAEDICS 2007; 32:835-40. [PMID: 17589843 PMCID: PMC2898961 DOI: 10.1007/s00264-007-0410-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2007] [Revised: 05/17/2007] [Accepted: 05/17/2007] [Indexed: 10/23/2022]
Abstract
Spinal metastatic disease is characterised by the preservation of the intervertebral disc structure, even after severe destruction of the vertebral body by neoplastic tissues. Anatomical features of the discs are thought to be the reason for the disc's resistance to metastatic cancer. However, little is known about the biochemical mechanism to prevent or attenuate the local invasion of cancer cells into the discs. The purpose of this study was to investigate the hypothesis that Fas ligand (FasL) produced by nucleus pulposus cells can kill Fas-expressing cancer cells infiltrating into the discs by the activation of caspases. Fas-expressing MCF-7 breast cancer cells were cultured with (experimental group) and without (control group) supernatant of nucleus pulposus cells containing FasL (50 pg/ml) for 48 h. The apoptosis of MCF-7 breast cancer cells was determined by the TUNEL technique. In addition, the activation of caspase-8, -9 and -3 was investigated by Western blot analysis. After treatment with supernatant of the nucleus pulposus cells containing FasL, the apoptosis of MCF-7 breast cancer cells was significantly increased, along with the activation of caspase-8, -9 and -3 compared with those of the control group. Our results suggest that the Fas/FasL interaction of nucleus pulposus and cancer cells might be a potential mechanism of the disc's resistance to metastatic cancer.
Collapse
Affiliation(s)
- Jong-Beom Park
- Department of Orthopaedic Surgery, Uijeongbu St. Mary's Hospital, The Catholic University of Korea School of Medicine, 65-1 Kumho-dong, Uijeongbu-si, Kyunggi-do, 480-717, South Korea.
| | | | | | | |
Collapse
|