1
|
Yang Y, Jiang Q, Zhang F. Nanocrystals for Deep-Tissue In Vivo Luminescence Imaging in the Near-Infrared Region. Chem Rev 2024; 124:554-628. [PMID: 37991799 DOI: 10.1021/acs.chemrev.3c00506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2023]
Abstract
In vivo imaging technologies have emerged as a powerful tool for both fundamental research and clinical practice. In particular, luminescence imaging in the tissue-transparent near-infrared (NIR, 700-1700 nm) region offers tremendous potential for visualizing biological architectures and pathophysiological events in living subjects with deep tissue penetration and high imaging contrast owing to the reduced light-tissue interactions of absorption, scattering, and autofluorescence. The distinctive quantum effects of nanocrystals have been harnessed to achieve exceptional photophysical properties, establishing them as a promising category of luminescent probes. In this comprehensive review, the interactions between light and biological tissues, as well as the advantages of NIR light for in vivo luminescence imaging, are initially elaborated. Subsequently, we focus on achieving deep tissue penetration and improved imaging contrast by optimizing the performance of nanocrystal fluorophores. The ingenious design strategies of NIR nanocrystal probes are discussed, along with their respective biomedical applications in versatile in vivo luminescence imaging modalities. Finally, thought-provoking reflections on the challenges and prospects for future clinical translation of nanocrystal-based in vivo luminescence imaging in the NIR region are wisely provided.
Collapse
Affiliation(s)
- Yang Yang
- College of Energy Materials and Chemistry, State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot 010021, China
| | - Qunying Jiang
- College of Energy Materials and Chemistry, State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot 010021, China
| | - Fan Zhang
- College of Energy Materials and Chemistry, State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot 010021, China
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, China
| |
Collapse
|
2
|
Calatayud DG, Lledos M, Casarsa F, Pascu SI. Functional Diversity in Radiolabeled Nanoceramics and Related Biomaterials for the Multimodal Imaging of Tumors. ACS BIO & MED CHEM AU 2023; 3:389-417. [PMID: 37876497 PMCID: PMC10591303 DOI: 10.1021/acsbiomedchemau.3c00021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 07/18/2023] [Accepted: 07/18/2023] [Indexed: 10/26/2023]
Abstract
Nanotechnology advances have the potential to assist toward the earlier detection of diseases, giving increased accuracy for diagnosis and helping to personalize treatments, especially in the case of noncommunicative diseases (NCDs) such as cancer. The main advantage of nanoparticles, the scaffolds underpinning nanomedicine, is their potential to present multifunctionality: synthetic nanoplatforms for nanomedicines can be tailored to support a range of biomedical imaging modalities of relevance for clinical practice, such as, for example, optical imaging, computed tomography (CT), magnetic resonance imaging (MRI), single photon emission computed tomography (SPECT), and positron emission tomography (PET). A single nanoparticle has the potential to incorporate myriads of contrast agent units or imaging tracers, encapsulate, and/or be conjugated to different combinations of imaging tags, thus providing the means for multimodality diagnostic methods. These arrangements have been shown to provide significant improvements to the signal-to-noise ratios that may be obtained by molecular imaging techniques, for example, in PET diagnostic imaging with nanomaterials versus the cases when molecular species are involved as radiotracers. We surveyed some of the main discoveries in the simultaneous incorporation of nanoparticulate materials and imaging agents within highly kinetically stable radio-nanomaterials as potential tracers with (pre)clinical potential. Diversity in function and new developments toward synthesis, radiolabeling, and microscopy investigations are explored, and preclinical applications in molecular imaging are highlighted. The emphasis is on the biocompatible materials at the forefront of the main preclinical developments, e.g., nanoceramics and liposome-based constructs, which have driven the evolution of diagnostic radio-nanomedicines over the past decade.
Collapse
Affiliation(s)
- David G. Calatayud
- Department
of Inorganic Chemistry, Universidad Autónoma
de Madrid, Madrid 28049, Spain
- Department
of Electroceramics, Instituto de Cerámica
y Vidrio, Madrid 28049, Spain
| | - Marina Lledos
- Department
of Chemistry, University of Bath, Bath BA2 7AY, United Kingdom
| | - Federico Casarsa
- Department
of Chemistry, University of Bath, Bath BA2 7AY, United Kingdom
| | - Sofia I. Pascu
- Department
of Chemistry, University of Bath, Bath BA2 7AY, United Kingdom
- Centre
of Therapeutic Innovations, University of
Bath, Bath BA2 7AY, United Kingdom
| |
Collapse
|
3
|
Eom S, Kim MH, Yoo R, Choi G, Kang JH, Lee YJ, Choy JH. Dilute lattice doping of 64Cu into 2D-nanoplates: its impact on radio-labeling efficiency and stability for target selective PET imaging. J Mater Chem B 2022; 10:9389-9399. [PMID: 35929536 DOI: 10.1039/d2tb01165d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
A quintinite nanoplate (64Cu-QT-NP) isomorphically substituted with 64Cu, as the positron emission tomography (PET) imaging material, was prepared via two-step processes. A 64Cu labeling efficiency of 99% was realized, for the first time, by immobilizing the 64Cu radioisotope directly in the octahedral site of the 2-dimensional (2D) quintinite lattice. Furthermore, the 64Cu labeling stability of 64Cu-QT-NPs was also achieved to be more than ∼99% in various solutions such as saline, phosphate-buffered saline (PBS), and other biological media (mouse and human serums). In an in vivo xenograft mouse model, the passive targeting behavior of 64Cu-QT-NPs into tumor tissue based on the enhanced permeability and retention (EPR) effect was also demonstrated by parenteral administration, and successfully visualized using a PET scanner. For enhancing the tumor tissue selectivity, bovine serum albumin (BSA) was coated on 64Cu-QT-NPs to form 64Cu-QT-NPs/BSA, resulting in better colloidal stability and longer blood circulation time, which was eventually evidenced by the 2-fold higher tumor uptake rate when intravenousely injected in an animal model. It is, therefore, concluded that the present 64Cu-QT-NPs/BSA with tumor tissue selectivity could be an advanced nano-device for radio-imaging and diagnosis as well.
Collapse
Affiliation(s)
- Sairan Eom
- Center for Intelligent Nano-Bio Materials (CINBM), Department of Chemistry and Nanoscience, Ewha Womans University, Seoul 03760, Republic of Korea.,Division of Applied-RI, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea
| | - Min Hwan Kim
- Division of Applied-RI, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea
| | - Ranji Yoo
- Division of Applied-RI, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea
| | - Goeun Choi
- Intelligent Nanohybrid Materials Laboratory (INML), Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea. .,College of Science and Technology, Dankook University, Cheonan 31116, Republic of Korea.,Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Joo Hyun Kang
- Division of Applied-RI, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea
| | - Yong Jin Lee
- Division of Applied-RI, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea
| | - Jin-Ho Choy
- Intelligent Nanohybrid Materials Laboratory (INML), Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea. .,Department of Pre-medical Course, College of Medicine, Dankook University, Cheonan 31116, Republic of Korea.,International Research Frontier Initiative (IRFI), Institute of Innovative Research, Tokyo Institute of Technology, Yokohama 226-8503, Japan
| |
Collapse
|
4
|
Xulu JH, Ndongwe T, Ezealisiji KM, Tembu VJ, Mncwangi NP, Witika BA, Siwe-Noundou X. The Use of Medicinal Plant-Derived Metallic Nanoparticles in Theranostics. Pharmaceutics 2022; 14:2437. [PMID: 36365255 PMCID: PMC9698412 DOI: 10.3390/pharmaceutics14112437] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/30/2022] [Accepted: 11/01/2022] [Indexed: 08/20/2023] Open
Abstract
In the quest to effectively diagnose and treat the diseases that afflict mankind, the development of a tool capable of simultaneous detection and treatment would provide a significant cornerstone for the survival and control of these diseases. Theranostics denotes a portmanteau of therapeutics and diagnostics which simultaneously detect and treat ailments. Research advances have initiated the advent of theranostics in modern medicine. Overall, theranostics are drug delivery systems with molecular or targeted imaging agents integrated into their structure. The application of theranostics is rising exponentially due to the urgent need for treatments that can be utilized for diagnostic imaging as an aid in precision and personalised medicine. Subsequently, the emergence of nanobiotechnology and the green synthesis of metallic nanoparticles (MNPs) has provided one such avenue for nanoscale development and research. Of interest is the drastic rise in the use of medicinal plants in the synthesis of MNPs which have been reported to be potentially effective in the diagnosis and treatment of diseases. At present, medicinal plant-derived MNPs have been cited to have broad pharmacological applications and have been studied for their potential use in the treatment and management of cancer, malaria, microbial and cardiovascular diseases. The subject of this article regards the role of medicinal plants in the synthesis of MNPs and the potential role of MNPs in the field of theranostics.
Collapse
Affiliation(s)
- Jabulile Happiness Xulu
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria 0204, South Africa
| | - Tanaka Ndongwe
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria 0204, South Africa
| | - Kenneth M. Ezealisiji
- Department of Pharmaceutical and Medicinal Chemistry, Faculty of Pharmaceutical Sciences, University of Port Harcourt, PMB 5323 Choba, Rivers State, Nigeria
| | - Vuyelwa J. Tembu
- Department of Chemistry, Tshwane University of Technology, Pretoria 0001, South Africa
| | - Nontobeko P. Mncwangi
- Department of Pharmacy Practice, School of Pharmacy, Sefako Makgatho Health Sciences University, MEDUNSA, Pretoria 0204, South Africa
| | - Bwalya A. Witika
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria 0204, South Africa
| | - Xavier Siwe-Noundou
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria 0204, South Africa
| |
Collapse
|
5
|
Subasinghe SAAS, Pautler RG, Samee MAH, Yustein JT, Allen MJ. Dual-Mode Tumor Imaging Using Probes That Are Responsive to Hypoxia-Induced Pathological Conditions. BIOSENSORS 2022; 12:478. [PMID: 35884281 PMCID: PMC9313010 DOI: 10.3390/bios12070478] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/22/2022] [Accepted: 06/26/2022] [Indexed: 05/02/2023]
Abstract
Hypoxia in solid tumors is associated with poor prognosis, increased aggressiveness, and strong resistance to therapeutics, making accurate monitoring of hypoxia important. Several imaging modalities have been used to study hypoxia, but each modality has inherent limitations. The use of a second modality can compensate for the limitations and validate the results of any single imaging modality. In this review, we describe dual-mode imaging systems for the detection of hypoxia that have been reported since the start of the 21st century. First, we provide a brief overview of the hallmarks of hypoxia used for imaging and the imaging modalities used to detect hypoxia, including optical imaging, ultrasound imaging, photoacoustic imaging, single-photon emission tomography, X-ray computed tomography, positron emission tomography, Cerenkov radiation energy transfer imaging, magnetic resonance imaging, electron paramagnetic resonance imaging, magnetic particle imaging, and surface-enhanced Raman spectroscopy, and mass spectrometric imaging. These overviews are followed by examples of hypoxia-relevant imaging using a mixture of probes for complementary single-mode imaging techniques. Then, we describe dual-mode molecular switches that are responsive in multiple imaging modalities to at least one hypoxia-induced pathological change. Finally, we offer future perspectives toward dual-mode imaging of hypoxia and hypoxia-induced pathophysiological changes in tumor microenvironments.
Collapse
Affiliation(s)
| | - Robia G. Pautler
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA; (R.G.P.); (M.A.H.S.)
| | - Md. Abul Hassan Samee
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA; (R.G.P.); (M.A.H.S.)
| | - Jason T. Yustein
- Integrative Molecular and Biomedical Sciences and the Department of Pediatrics in the Texas Children’s Cancer and Hematology Centers and The Faris D. Virani Ewing Sarcoma Center, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Matthew J. Allen
- Department of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, MI 48202, USA;
| |
Collapse
|
6
|
Chen X, Niu W, Du Z, Zhang Y, Su D, Gao X. 64Cu radiolabeled nanomaterials for positron emission tomography (PET) imaging. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.02.070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
7
|
Synthesis and Characterization of Multifunctional Nanovesicles Composed of POPC Lipid Molecules for Nuclear Imaging. Molecules 2021; 26:molecules26216591. [PMID: 34770999 PMCID: PMC8587727 DOI: 10.3390/molecules26216591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 10/26/2021] [Accepted: 10/27/2021] [Indexed: 12/02/2022] Open
Abstract
The integration of nuclear imaging analysis with nanomedicine has tremendously grown and represents a valid and powerful tool for the development and clinical translation of drug delivery systems. Among the various types of nanostructures used as drug carriers, nanovesicles represent intriguing platforms due to their capability to entrap both lipophilic and hydrophilic agents, and their well-known biocompatibility and biodegradability. In this respect, here we present the development of a labelling procedure of POPC (1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine)-based liposomes incorporating an ad hoc designed lipophilic NOTA (1,4,7-triazacyclononane-1,4,7-triacetic acid) analogue, derivatized with an oleic acid residue, able to bind the positron emitter gallium-68(III). Based on POPC features, the optimal conditions for liposome labelling were studied with the aim of optimizing the Ga(III) incorporation and obtaining a significant radiochemical yield. The data presented in this work demonstrate the feasibility of the labelling procedure on POPC liposomes co-formulated with the ad hoc designed NOTA analogue. We thus provided a critical insight into the practical aspects of the development of vesicles for theranostic approaches, which in principle can be extended to other nanosystems exploiting a variety of bioconjugation protocols.
Collapse
|
8
|
Sarcan ET, Silindir-Gunay M, Ozer AY, Hartman N. 89Zr as a promising radionuclide and it’s applications for effective cancer imaging. J Radioanal Nucl Chem 2021. [DOI: 10.1007/s10967-021-07928-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
9
|
Theranostic Applications of Nanoparticle-Mediated Photoactivated Therapies. JOURNAL OF NANOTHERANOSTICS 2021. [DOI: 10.3390/jnt2030009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Nanoparticle-mediated light-activated therapies, such as photodynamic therapy and photothermal therapy, are earnestly being viewed as efficient interventional strategies against several cancer types. Theranostics is a key hallmark of cancer nanomedicine since it allows diagnosis and therapy of both primary and metastatic cancer using a single nanoprobe. Advanced in vivo diagnostic imaging using theranostic nanoparticles not only provides precise information about the location of tumor/s but also outlines the narrow time window corresponding to the maximum tumor-specific drug accumulation. Such information plays a critical role in guiding light-activated therapies with high spatio-temporal accuracy. Furthermore, theranostics facilitates monitoring the progression of therapy in real time. Herein, we provide a general review of the application of theranostic nanoparticles for in vivo image-guided light-activated therapy in cancer. The imaging modalities considered here include fluorescence imaging, photoacoustic imaging, thermal imaging, magnetic resonance imaging, X-ray computed tomography, positron emission tomography, and single-photon emission computed tomography. The review concludes with a brief discussion about the broad scope of theranostic light-activated nanomedicine.
Collapse
|
10
|
Qiu J, Liu Y, Xia Y. Radiolabeling of Gold Nanocages for Potential Applications in Tracking, Diagnosis, and Image-Guided Therapy. Adv Healthc Mater 2021; 10:e2002031. [PMID: 33470560 PMCID: PMC8289932 DOI: 10.1002/adhm.202002031] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/06/2021] [Indexed: 12/23/2022]
Abstract
Gold nanocages (AuNCs) have emerged as a novel class of multifunctional nanomaterials with an array of applications in nanomedicine, including drug delivery, controlled release, as well as disease diagnosis and treatment. Labeling AuNCs with radionuclides not only offers additional therapeutic capabilities but also makes it easy to analyze their biodistribution, monitor their uptake by the tissue or organ of interest, and optimize their performance in both diagnosis and treatment. Here, an introduction to the chemical synthesis and optical properties of AuNCs is provided in the beginning. The methods developed for their radiolabeling are then showcased, followed by the use of radiolabeled AuNCs in tracking and quantifying their pharmacokinetics, including biodistribution, tumor uptake, and intratumoral distribution. Finally, their potential applications in targeted imaging and image-guided therapy are discussed.
Collapse
Affiliation(s)
- Jichuan Qiu
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
| | - Yongjian Liu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Younan Xia
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
- School of Chemistry and Biochemistry, School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| |
Collapse
|
11
|
Mari M, Carrozza D, Ferrari E, Asti M. Applications of Radiolabelled Curcumin and Its Derivatives in Medicinal Chemistry. Int J Mol Sci 2021; 22:ijms22147410. [PMID: 34299029 PMCID: PMC8306375 DOI: 10.3390/ijms22147410] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/02/2021] [Accepted: 07/07/2021] [Indexed: 12/27/2022] Open
Abstract
Curcumin is a natural occurring molecule that has aroused much interest among researchers over the years due to its pleiotropic set of biological properties. In the nuclear medicine field, radiolabelled curcumin and curcumin derivatives have been studied as potential radiotracers for the early diagnosis of Alzheimer’s disease and cancer. In the present review, the synthetic pathways, labelling methods and the preclinical investigations involving these radioactive compounds are treated. The studies entailed chemical modifications for enhancing curcumin stability, as well as its functionalisation for the labelling with several radiohalogens or metal radionuclides (fluorine-18, technetium-99m, gallium-68, etc.). Although some drawbacks have yet to be addressed, and none of the radiolabelled curcuminoids have so far achieved clinical application, the studies performed hitherto provide useful insights and lay the foundation for further developments.
Collapse
Affiliation(s)
- Matteo Mari
- Department of Chemical and Geological Sciences, University of Modena and Reggio Emilia, Via G. Campi 103, 41125 Modena, Italy
| | - Debora Carrozza
- Department of Chemical and Geological Sciences, University of Modena and Reggio Emilia, Via G. Campi 103, 41125 Modena, Italy
| | - Erika Ferrari
- Department of Chemical and Geological Sciences, University of Modena and Reggio Emilia, Via G. Campi 103, 41125 Modena, Italy
| | - Mattia Asti
- Radiopharmaceutical Chemistry Section, Nuclear Medicine Unit, AUSL-IRCCS di Reggio Emilia, Viale Risorgimento 80, 42122 Reggio Emilia, Italy
| |
Collapse
|
12
|
Omabe K, Paris C, Lannes F, Taïeb D, Rocchi P. Nanovectorization of Prostate Cancer Treatment Strategies: A New Approach to Improved Outcomes. Pharmaceutics 2021; 13:591. [PMID: 33919150 PMCID: PMC8143094 DOI: 10.3390/pharmaceutics13050591] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/06/2021] [Accepted: 04/08/2021] [Indexed: 12/21/2022] Open
Abstract
Prostate cancer (PC) is the most frequent male cancer in the Western world. Progression to Castration Resistant Prostate Cancer (CRPC) is a known consequence of androgen withdrawal therapy, making CRPC an end-stage disease. Combination of cytotoxic drugs and hormonal therapy/or genotherapy is a recognized modality for the treatment of advanced PC. However, this strategy is limited by poor bio-accessibility of the chemotherapy to tumor sites, resulting in an increased rate of collateral toxicity and incidence of multidrug resistance (MDR). Nanovectorization of these strategies has evolved to an effective approach to efficacious therapeutic outcomes. It offers the possibility to consolidate their antitumor activity through enhanced specific and less toxic active or passive targeting mechanisms, as well as enabling diagnostic imaging through theranostics. While studies on nanomedicine are common in other cancer types, only a few have focused on prostate cancer. This review provides an in-depth knowledge of the principles of nanotherapeutics and nanotheranostics, and how the application of this rapidly evolving technology can clinically impact CRPC treatment. With particular reference to respective nanovectors, we draw clinical and preclinical evidence, demonstrating the potentials and prospects of homing nanovectorization into CRPC treatment strategies.
Collapse
Affiliation(s)
- Kenneth Omabe
- Centre de Recherche en Cancérologie de Marseille, CRCM, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University U105, Institut Paoli-Calmettes, 13273 Marseille, France; (K.O.); (C.P.); (F.L.); (D.T.)
- Department of Biochemistry & Molecular Biology, Alex Ekwueme Federal University, Ndufu-Alike Ikwo, PMB 1010, Abakaliki 84001, Nigeria
| | - Clément Paris
- Centre de Recherche en Cancérologie de Marseille, CRCM, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University U105, Institut Paoli-Calmettes, 13273 Marseille, France; (K.O.); (C.P.); (F.L.); (D.T.)
| | - François Lannes
- Centre de Recherche en Cancérologie de Marseille, CRCM, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University U105, Institut Paoli-Calmettes, 13273 Marseille, France; (K.O.); (C.P.); (F.L.); (D.T.)
| | - David Taïeb
- Centre de Recherche en Cancérologie de Marseille, CRCM, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University U105, Institut Paoli-Calmettes, 13273 Marseille, France; (K.O.); (C.P.); (F.L.); (D.T.)
- Biophysics and Nuclear Medicine, La Timone University Hospital, European Center for Research in Medical Imaging, Aix-Marseille University, 13005 Marseille, France
| | - Palma Rocchi
- Centre de Recherche en Cancérologie de Marseille, CRCM, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University U105, Institut Paoli-Calmettes, 13273 Marseille, France; (K.O.); (C.P.); (F.L.); (D.T.)
| |
Collapse
|
13
|
Zhang X, Detering L, Sultan D, Luehmann H, Li L, Heo GS, Zhang X, Lou L, Grierson PM, Greco S, Ruzinova M, Laforest R, Dehdashti F, Lim KH, Liu Y. CC Chemokine Receptor 2-Targeting Copper Nanoparticles for Positron Emission Tomography-Guided Delivery of Gemcitabine for Pancreatic Ductal Adenocarcinoma. ACS NANO 2021; 15:1186-1198. [PMID: 33406361 PMCID: PMC7846978 DOI: 10.1021/acsnano.0c08185] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a deadly malignancy with dire prognosis due to aggressive biology, lack of effective tools for diagnosis at an early stage, and limited treatment options. Detection of PDAC using conventional radiographic imaging is limited by the dense, hypovascular stromal component and relatively scarce neoplastic cells within the tumor microenvironment (TME). The CC motif chemokine 2 (CCL2) and its cognate receptor CCR2 (CCL2/CCR2) axis are critical in fostering and maintaining this kind of TME by recruiting immunosuppressive myeloid cells such as the tumor-associated macrophages, thereby presenting an opportunity to exploit this axis for both diagnostic and therapeutic purposes. We engineered CCR2-targeting ultrasmall copper nanoparticles (Cu@CuOx) as nanovehicles not only for targeted positron emission tomography imaging by intrinsic radiolabeling with 64Cu but also for loading and delivery of the chemotherapy drug gemcitabine to PDAC. This 64Cu-radiolabeled nanovehicle allowed sensitive and accurate detection of PDAC malignancy in autochthonous genetically engineered mouse models. The ultrasmall Cu@CuOx showed efficient renal clearance, favorable pharmacokinetics, and minimal in vivo toxicity. Systemic administration of gemcitabine-loaded Cu@CuOx effectively suppressed the progression of PDAC tumors in a syngeneic xenograft mouse model and prolonged survival. These CCR2-targeted ultrasmall nanoparticles offer a promising image-guided therapeutic agent and show great potential for translation.
Collapse
Affiliation(s)
- Xiaohui Zhang
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Lisa Detering
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Deborah Sultan
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Hannah Luehmann
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Lin Li
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Gyu Seong Heo
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Xiuli Zhang
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Lanlan Lou
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Patrick M. Grierson
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Suellen Greco
- Division of Comparative Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Marianna Ruzinova
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Richard Laforest
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Farrokh Dehdashti
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Kian-Huat Lim
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Yongjian Liu
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| |
Collapse
|
14
|
Munch M, Rotstein BH, Ulrich G. Fluorine-18-Labeled Fluorescent Dyes for Dual-Mode Molecular Imaging. Molecules 2020; 25:E6042. [PMID: 33371284 PMCID: PMC7766373 DOI: 10.3390/molecules25246042] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 12/16/2020] [Indexed: 12/27/2022] Open
Abstract
Recent progress realized in the development of optical imaging (OPI) probes and devices has made this technique more and more affordable for imaging studies and fluorescence-guided surgery procedures. However, this imaging modality still suffers from a low depth of penetration, thus limiting its use to shallow tissues or endoscopy-based procedures. In contrast, positron emission tomography (PET) presents a high depth of penetration and the resulting signal is less attenuated, allowing for imaging in-depth tissues. Thus, association of these imaging techniques has the potential to push back the limits of each single modality. Recently, several research groups have been involved in the development of radiolabeled fluorophores with the aim of affording dual-mode PET/OPI probes used in preclinical imaging studies of diverse pathological conditions such as cancer, Alzheimer's disease, or cardiovascular diseases. Among all the available PET-active radionuclides, 18F stands out as the most widely used for clinical imaging thanks to its advantageous characteristics (t1/2 = 109.77 min; 97% β+ emitter). This review focuses on the recent efforts in the synthesis and radiofluorination of fluorescent scaffolds such as 4,4-difluoro-4-bora-diazaindacenes (BODIPYs), cyanines, and xanthene derivatives and their use in preclinical imaging studies using both PET and OPI technologies.
Collapse
Affiliation(s)
- Maxime Munch
- University of Ottawa Heart Institute, Ottawa, ON K1Y 4W7, Canada;
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Benjamin H. Rotstein
- University of Ottawa Heart Institute, Ottawa, ON K1Y 4W7, Canada;
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Gilles Ulrich
- Institut de Chimie et Procédés pour l’Énergie, l’Environnement et la Santé (ICPEES), UMR CNRS 7515, École Européenne de Chimie, Polymères et Matériaux (ECPM), 25 rue Becquerel, CEDEX 02, 67087 Strasbourg, France;
| |
Collapse
|
15
|
Badeggi UM, Ismail E, Adeloye AO, Botha S, Badmus JA, Marnewick JL, Cupido CN, Hussein AA. Green Synthesis of Gold Nanoparticles Capped with Procyanidins from Leucosidea sericea as Potential Antidiabetic and Antioxidant Agents. Biomolecules 2020; 10:biom10030452. [PMID: 32183213 PMCID: PMC7175165 DOI: 10.3390/biom10030452] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 02/28/2020] [Accepted: 03/02/2020] [Indexed: 01/02/2023] Open
Abstract
In this study, procyanidins fractions of dimers and trimers (F1-F2) from the Leucosidea sericea total extract (LSTE) were investigated for their chemical constituents. The total extract and the procyanidins were employed in the synthesis of gold nanoparticles (Au NPs) and fully characterized. Au NPs of 6, 24 and 21 nm were obtained using LSTE, F1 and F2 respectively. Zeta potential and in vitro stability studies confirmed the stability of the particles. The enzymatic activity of LSTE, F1, F2 and their corresponding Au NPs showed strong inhibitory alpha-amylase activity where F1 Au NPs demonstrated the highest with IC50 of 1.88 µg/mL. On the other hand, F2 Au NPs displayed the strongest alpha-glucosidase activity at 4.5 µg/mL. F2 and F2 Au NPs also demonstrated the highest antioxidant activity, 1834.0 ± 4.7 μM AAE/g and 1521.9 ± 3.0 μM TE/g respectively. The study revealed not only the ability of procyanidins dimers (F1 and F2) in forming biostable and bioactive Au NPs but also, a significant enhancement of the natural products activities, which could improve the smart delivery in future biomedical applications.
Collapse
Affiliation(s)
- Umar M. Badeggi
- Chemistry Department, Cape Peninsula University of Technology, Symphony Rd., Bellville 7535, South Africa; (U.M.B.); (E.I.); (A.O.A.)
| | - Enas Ismail
- Chemistry Department, Cape Peninsula University of Technology, Symphony Rd., Bellville 7535, South Africa; (U.M.B.); (E.I.); (A.O.A.)
| | - Adewale O. Adeloye
- Chemistry Department, Cape Peninsula University of Technology, Symphony Rd., Bellville 7535, South Africa; (U.M.B.); (E.I.); (A.O.A.)
| | - Subelia Botha
- Electron Microscope Unit, University of the Western Cape, Bellville 7535, South Africa;
| | - Jelili A. Badmus
- Oxidative Stress Research Centre, Institute of Biomedical and Microbial Biotechnology, Cape Peninsula University of Technology, Symphony Rd., Bellville 7535, South Africa; (J.A.B.); (J.L.M.)
| | - Jeanine L. Marnewick
- Oxidative Stress Research Centre, Institute of Biomedical and Microbial Biotechnology, Cape Peninsula University of Technology, Symphony Rd., Bellville 7535, South Africa; (J.A.B.); (J.L.M.)
| | - Christopher N. Cupido
- Department of Botany, University of Fort Hare, Private Bag X1314, Alice 5700, South Africa;
| | - Ahmed A. Hussein
- Chemistry Department, Cape Peninsula University of Technology, Symphony Rd., Bellville 7535, South Africa; (U.M.B.); (E.I.); (A.O.A.)
- Correspondence: ; Tel.: +27-21-959-6193; Fax: +27-21-959-3055
| |
Collapse
|
16
|
Pérez-Medina C, Teunissen AJ, Kluza E, Mulder WJ, van der Meel R. Nuclear imaging approaches facilitating nanomedicine translation. Adv Drug Deliv Rev 2020; 154-155:123-141. [PMID: 32721459 DOI: 10.1016/j.addr.2020.07.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/08/2020] [Accepted: 07/17/2020] [Indexed: 02/07/2023]
Abstract
Nanomedicine approaches can effectively modulate the biodistribution and bioavailability of therapeutic agents, improving their therapeutic index. However, despite the ever-increasing amount of literature reporting on preclinical nanomedicine, the number of nanotherapeutics receiving FDA approval remains relatively low. Several barriers exist that hamper the effective preclinical evaluation and clinical translation of nanotherapeutics. Key barriers include insufficient understanding of nanomedicines' in vivo behavior, inadequate translation from murine models to larger animals, and a lack of patient stratification strategies. Integrating quantitative non-invasive imaging techniques in nanomedicine development offers attractive possibilities to address these issues. Among the available imaging techniques, nuclear imaging by positron emission tomography (PET) and single-photon emission computed tomography (SPECT) are highly attractive in this context owing to their quantitative nature and uncontested sensitivity. In basic and translational research, nuclear imaging techniques can provide critical quantitative information about pharmacokinetic parameters, biodistribution profiles or target site accumulation of nanocarriers and their associated payload. During clinical evaluation, nuclear imaging can be used to select patients amenable to nanomedicine treatment. Here, we review how nuclear imaging-based approaches are increasingly being integrated into nanomedicine development and discuss future developments that will accelerate their clinical translation.
Collapse
|
17
|
Ghiassian S, Yu L, Gobbo P, Nazemi A, Romagnoli T, Luo W, Luyt LG, Workentin MS. Nitrone-Modified Gold Nanoparticles: Synthesis, Characterization, and Their Potential as 18F-Labeled Positron Emission Tomography Probes via I-SPANC. ACS OMEGA 2019; 4:19106-19115. [PMID: 31763533 PMCID: PMC6868604 DOI: 10.1021/acsomega.9b02322] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 10/28/2019] [Indexed: 05/11/2023]
Abstract
A novel bioorthogonal gold nanoparticle (AuNP) template displaying interfacial nitrone functional groups for bioorthogonal interfacial strain-promoted alkyne-nitrone cycloaddition reactions has been synthesized. These nitrone-AuNPs were characterized in detail using 1H nuclear magnetic resonance spectroscopy, transmission electron microscopy, thermogravimetric analysis, and X-ray photoelectron spectroscopy, and a nanoparticle raw formula was calculated. The ability to control the conjugation of molecules of interest at the molecular level onto the nitrone-AuNP template allowed us to create a novel methodology for the synthesis of AuNP-based radiolabeled probes.
Collapse
Affiliation(s)
- Sara Ghiassian
- Department
of Chemistry and the Center for Materials and Biomaterials
Research and Department of Oncology, The University
of Western Ontario, London N6A 5B7, Ontario, Canada
| | - Lihai Yu
- London
Regional Cancer Program, 800 Commissioners Rd. E., London N6A 5W9, Ontario, Canada
| | - Pierangelo Gobbo
- Department
of Chemistry and the Center for Materials and Biomaterials
Research and Department of Oncology, The University
of Western Ontario, London N6A 5B7, Ontario, Canada
| | - Ali Nazemi
- Department
of Chemistry and the Center for Materials and Biomaterials
Research and Department of Oncology, The University
of Western Ontario, London N6A 5B7, Ontario, Canada
| | - Tommaso Romagnoli
- Department
of Chemistry and the Center for Materials and Biomaterials
Research and Department of Oncology, The University
of Western Ontario, London N6A 5B7, Ontario, Canada
| | - Wilson Luo
- Department
of Chemistry and the Center for Materials and Biomaterials
Research and Department of Oncology, The University
of Western Ontario, London N6A 5B7, Ontario, Canada
| | - Leonard G. Luyt
- Department
of Chemistry and the Center for Materials and Biomaterials
Research and Department of Oncology, The University
of Western Ontario, London N6A 5B7, Ontario, Canada
- London
Regional Cancer Program, 800 Commissioners Rd. E., London N6A 5W9, Ontario, Canada
| | - Mark S. Workentin
- Department
of Chemistry and the Center for Materials and Biomaterials
Research and Department of Oncology, The University
of Western Ontario, London N6A 5B7, Ontario, Canada
| |
Collapse
|
18
|
Szkop M, Brygoła K, Janczewska M, Ciach T. A simple time-resolved fluorescence assay for quantitative determination of DOTA chelator. Anal Biochem 2019; 584:113384. [PMID: 31356774 DOI: 10.1016/j.ab.2019.113384] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 07/14/2019] [Accepted: 07/25/2019] [Indexed: 01/01/2023]
Abstract
DOTA (1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetate) is one of the preeminent metal chelator applied for diagnostic and therapeutic purposes, however to date there is no versatile and reliable nonradioisotopic method for its precise determination. In this technical note, we present a novel and sensitive fluorimetric assay for quantitative determination of DOTA based on the luminescence quenching of the highly luminescent europium ions complex with trioctyl phosphine oxide and naphthoyl trifluoroacetone sensitizing activators. The assay is carried out in two simple steps and enables the determination of DOTA in the nanomolar range providing a superior tool compared to commonly applied spectrophotometric assay with Arsenazo-III reagent.
Collapse
Affiliation(s)
- Michał Szkop
- NanoThea Inc., Waryńskiego 1, 00-645, Warsaw, Poland.
| | - Kamil Brygoła
- NanoThea Inc., Waryńskiego 1, 00-645, Warsaw, Poland
| | | | - Tomasz Ciach
- NanoThea Inc., Waryńskiego 1, 00-645, Warsaw, Poland; Faculty of Chemical and Process Engineering, Warsaw University of Technology, Waryńskiego 1, 00-645, Warsaw, Poland
| |
Collapse
|
19
|
Park JY, Park S, Lee TS, Hwang YH, Kim JY, Kang WJ, Key J. Biodegradable micro-sized discoidal polymeric particles for lung-targeted delivery system. Biomaterials 2019; 218:119331. [PMID: 31299455 DOI: 10.1016/j.biomaterials.2019.119331] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 06/24/2019] [Accepted: 07/01/2019] [Indexed: 12/21/2022]
Abstract
Various types of particle-based drug delivery systems have been explored for the treatment of pulmonary diseases; however, bio-distribution and elimination of the particles should be monitored for better understanding of their therapeutic efficacy and safety. This study aimed to characterize the biological properties of micro-sized discoidal polymeric particles (DPPs) as lung-targeted drug delivery carriers. DPPs were prepared using a top-down fabrication approach and characterized by assessing size and zeta potential. They were labeled with zirconium-89 (89Zr), and bio-distribution studies and PET imaging were performed for 7 days after intravenous administration. Their hydrodynamic size was 2.8 ± 6.1 μm and average zeta potential was -39.9 ± 5.39 mV. At doses of 5, 12.5, and 25 mg/kg, they showed no acute toxicity in nude mice. Desferrioxamine (DFO)-functionalized 89Zr-labeled DPPs gave a decay-corrected radiochemical yield of 82.1 ± 0.2%. Furthermore, 89Zr-DPPs, from chelate-free labeling methods, showed a yield of 48.5 ± 0.9%. Bio-distribution studies and PET imaging showed 89Zr-DFO-DPPs to be mainly accumulated in the lungs and degraded within 3 d of injection. However, 89Zr-DFO-DPPs showed significantly low uptake in the bone. Overall, our results suggested micro-sized DPPs as promising drug delivery carriers for the targeted treatment of various pulmonary diseases.
Collapse
Affiliation(s)
- Jun Young Park
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Sanghyo Park
- Department of Biomedical Engineering, Yonsei University, Wonju, Gangwon-do, 26493, Republic of Korea
| | - Tae Sup Lee
- Division of RI Application, Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul, 01812, Republic of Korea
| | - Yong Hwa Hwang
- Division of RI Application, Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul, 01812, Republic of Korea
| | - Jung Young Kim
- Division of RI Application, Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul, 01812, Republic of Korea
| | - Won Jun Kang
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea.
| | - Jaehong Key
- Department of Biomedical Engineering, Yonsei University, Wonju, Gangwon-do, 26493, Republic of Korea.
| |
Collapse
|
20
|
Lobatto ME, Binderup T, Robson PM, Giesen LFP, Calcagno C, Witjes J, Fay F, Baxter S, Wessel CH, Eldib M, Bini J, Carlin SD, Stroes ESG, Storm G, Kjaer A, Lewis JS, Reiner T, Fayad ZA, Mulder WJM, Pérez-Medina C. Multimodal Positron Emission Tomography Imaging to Quantify Uptake of 89Zr-Labeled Liposomes in the Atherosclerotic Vessel Wall. Bioconjug Chem 2019; 31:360-368. [PMID: 31095372 DOI: 10.1021/acs.bioconjchem.9b00256] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Nanotherapy has recently emerged as an experimental treatment option for atherosclerosis. To fulfill its promise, robust noninvasive imaging approaches for subject selection and treatment evaluation are warranted. To that end, we present here a positron emission tomography (PET)-based method for quantification of liposomal nanoparticle uptake in the atherosclerotic vessel wall. We evaluated a modular procedure to label liposomal nanoparticles with the radioisotope zirconium-89 (89Zr). Their biodistribution and vessel wall targeting in a rabbit atherosclerosis model was evaluated up to 15 days after intravenous injection by PET/computed tomography (CT) and PET/magnetic resonance imaging (PET/MRI). Vascular permeability was assessed in vivo using three-dimensional dynamic contrast-enhanced MRI (3D DCE-MRI) and ex vivo using near-infrared fluorescence (NIRF) imaging. The 89Zr-radiolabeled liposomes displayed a biodistribution pattern typical of long-circulating nanoparticles. Importantly, they markedly accumulated in atherosclerotic lesions in the abdominal aorta, as evident on PET/MRI and confirmed by autoradiography, and this uptake moderately correlated with vascular permeability. The method presented herein facilitates the development of nanotherapy for atherosclerotic disease as it provides a tool to screen for nanoparticle targeting in individual subjects' plaques.
Collapse
Affiliation(s)
- Mark E Lobatto
- Translational and Molecular Imaging Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States.,Department of Radiology , Spaarne Gasthuis , 2035 RC Haarlem , The Netherlands
| | - Tina Binderup
- Translational and Molecular Imaging Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States.,Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging , Rigshospitalet & University of Copenhagen , 2100 Copenhagen , Denmark
| | - Philip M Robson
- Translational and Molecular Imaging Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Luuk F P Giesen
- Translational and Molecular Imaging Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Claudia Calcagno
- Translational and Molecular Imaging Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Julia Witjes
- Translational and Molecular Imaging Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Francois Fay
- Translational and Molecular Imaging Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States.,Institut Galien Paris Sud UMR 8612, Faculté de Pharmacie, CNRS, Univ. Paris-Sud Université Paris-Saclay , 92290 Châtenay-Malabry , France
| | - Samantha Baxter
- Translational and Molecular Imaging Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Chang Ho Wessel
- Translational and Molecular Imaging Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Mootaz Eldib
- Translational and Molecular Imaging Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Jason Bini
- Translational and Molecular Imaging Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Sean D Carlin
- Department of Radiology , Memorial Sloan Kettering Cancer Center , New York , New York 10065 , United States
| | - Erik S G Stroes
- Department of Vascular Medicine , Academic Medical Center , 1105 AZ Amsterdam , The Netherlands
| | - Gert Storm
- Department of Targeted Therapeutics, MIRA Institute , University of Twente , 7522 NB Enschede , The Netherlands.,Utrecht Institute for Pharmaceutical Sciences , Utrecht University , 3512 JE Utrecht , The Netherlands
| | - Andreas Kjaer
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging , Rigshospitalet & University of Copenhagen , 2100 Copenhagen , Denmark
| | - Jason S Lewis
- Department of Radiology , Memorial Sloan Kettering Cancer Center , New York , New York 10065 , United States.,Program in Molecular Pharmacology , Memorial Sloan Kettering Cancer Center , New York , New York 10065 , United States
| | - Thomas Reiner
- Department of Radiology , Memorial Sloan Kettering Cancer Center , New York , New York 10065 , United States.,Chemical Biology Program , Memorial Sloan Kettering Cancer Center , New York , New York 10065 , United States
| | - Zahi A Fayad
- Translational and Molecular Imaging Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Willem J M Mulder
- Translational and Molecular Imaging Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States.,Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems , Eindhoven University of Technology , 5612 AZ Eindhoven , The Netherlands.,Department of Oncological Sciences , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Carlos Pérez-Medina
- Translational and Molecular Imaging Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States.,Centro Nacional de Investigaciones Cardiovasculares Carlos III , 28029 Madrid , Spain
| |
Collapse
|
21
|
Pretze M, van der Meulen N, Wängler C, Schibli R, Wängler B. Targeted 64
Cu-labeled gold nanoparticles for dual imaging with positron emission tomography and optical imaging. J Labelled Comp Radiopharm 2019; 62:471-482. [DOI: 10.1002/jlcr.3736] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 03/29/2019] [Accepted: 04/04/2019] [Indexed: 11/11/2022]
Affiliation(s)
- Marc Pretze
- Molecular Imaging and Radiochemistry, Department of Clinical Radiology and Nuclear Medicine, Medical Faculty Mannheim; Heidelberg University; Mannheim Germany
| | - Nick P. van der Meulen
- Laboratory of Radiochemistry (LRC), Center of Radiopharmaceutical Sciences; PSI; Villigen Switzerland
| | - Carmen Wängler
- Biomedical Chemistry, Department of Clinical Radiology and Nuclear Medicine, Medical Faculty Mannheim; Heidelberg University; Mannheim Germany
| | - Roger Schibli
- Laboratory of Radiochemistry (LRC), Center of Radiopharmaceutical Sciences; PSI; Villigen Switzerland
| | - Björn Wängler
- Molecular Imaging and Radiochemistry, Department of Clinical Radiology and Nuclear Medicine, Medical Faculty Mannheim; Heidelberg University; Mannheim Germany
| |
Collapse
|
22
|
Yang CT, Ghosh KK, Padmanabhan P, Langer O, Liu J, Eng DNC, Halldin C, Gulyás B. PET-MR and SPECT-MR multimodality probes: Development and challenges. Theranostics 2018; 8:6210-6232. [PMID: 30613293 PMCID: PMC6299694 DOI: 10.7150/thno.26610] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 06/08/2018] [Indexed: 12/22/2022] Open
Abstract
Positron emission tomography (PET)-magnetic resonance (MR) or single photon emission computed tomography (SPECT)-MR hybrid imaging is being used in daily clinical practice. Due to its advantages over stand-alone PET, SPECT or MR imaging, in many areas such as oncology, the demand for hybrid imaging techniques is increasing dramatically. The use of multimodal imaging probes or biomarkers in a single molecule or particle to characterize the imaging subjects such as disease tissues certainly provides us with more accurate diagnosis and promotes therapeutic accuracy. A limited number of multimodal imaging probes are being used in preclinical and potential clinical investigations. The further development of multimodal PET-MR and SPECT-MR imaging probes includes several key elements: novel synthetic strategies, high sensitivity for accurate quantification and high anatomic resolution, favourable pharmacokinetic profile and target-specific binding of a new probe. This review thoroughly summarizes all recently available and noteworthy PET-MR and SPECT-MR multimodal imaging probes including small molecule bimodal probes, nano-sized bimodal probes, small molecular trimodal probes and nano-sized trimodal probes. To the best of our knowledge, this is the first comprehensive overview of all PET-MR and SPECT-MR multimodal probes. Since the development of multimodal PET-MR and SPECT-MR imaging probes is an emerging research field, a selection of 139 papers were recognized following the literature review. The challenges for designing multimodal probes have also been addressed in order to offer some future research directions for this novel interdisciplinary research field.
Collapse
Affiliation(s)
- Chang-Tong Yang
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore 636921
- Cixi Institute of Biomedical Engineering, Ningbo Institute of Industrial Technology and Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, P.R. China, 315201
- Department of Nuclear Medicine and Molecular Imaging, Radiological Sciences Division, Singapore General Hospital, Outram Road, Singapore 169608
| | - Krishna K. Ghosh
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore 636921
| | - Parasuraman Padmanabhan
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore 636921
| | - Oliver Langer
- Department of Clinical Pharmacology and Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, A-1090, Vienna, Austria
- Center for Health and Bioresources, Biomedical Systems, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Jiang Liu
- Cixi Institute of Biomedical Engineering, Ningbo Institute of Industrial Technology and Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, P.R. China, 315201
| | - David Ng Chee Eng
- Department of Nuclear Medicine and Molecular Imaging, Radiological Sciences Division, Singapore General Hospital, Outram Road, Singapore 169608
- Duke-NUS Medical School, 8 College Road, Singapore 169857
| | - Christer Halldin
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore 636921
- Karolinska Institutet, Department of Clinical Neuroscience, S-171 76, Stockholm, Sweden
| | - Balázs Gulyás
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore 636921
- Karolinska Institutet, Department of Clinical Neuroscience, S-171 76, Stockholm, Sweden
| |
Collapse
|
23
|
Hajdu I, Makhlouf A, Solomon VR, Michel D, Al-Dulaymi M, Wasan KM, Fonge H, Badea I. A 89Zr-labeled lipoplex nanosystem for image-guided gene delivery: design, evaluation of stability and in vivo behavior. Int J Nanomedicine 2018; 13:7801-7818. [PMID: 30538460 PMCID: PMC6257135 DOI: 10.2147/ijn.s179806] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background With the advances in radiopharmaceutical research, the development of image-guided therapy has become a major interest. While the development of theranostic nanotherapeutics is frequently associated with cancer chemotherapy, phototherapy and radiotherapy, there is little information available on the in vivo monitoring of gene delivery systems and the application of image-guided approach in gene therapy. The goal of this work was to determine the in vivo behavior of DNA delivery nanosystems - based on cationic gemini surfactants – designed for image-guided gene therapy. We tested the feasibility of monitoring tumor accumulation of gene delivery nanoparticles by positron emission tomography. Methods To be able to conjugate radiotracers to the nanoparticles, a deferoxamine-modified gemini surfactant was synthesized, DNA-containing lipoplex nanoparticles were formulated, and radiolabeled with Zirconium-89 (89Zr). The pharmacokinetics and biodistribution of 89Zr labeled surfactant and 89Zr labeled nanoparticles were monitored in mice by microPET/CT imaging and ex vivo gamma counting. Results Modification of the nanoparticles with deferoxamine did not alter their physicochemical properties. The radiolabeled nanoparticles (labeling efficiency of 95±3%) were stable in PBS and serum. The biological half-life of the 89Zr labeled nanoparticles was significantly higher compared to 89Zr labeled surfactant. As expected, the nanoparticles had significantly higher liver accumulation than the radiolabeled surfactant alone and lower kidney accumulation. Tumor uptake was detected at 2 hours post injection and decreased throughout the 3-day monitoring. Conclusion We propose that radiolabeling DNA delivery lipoplex nanosystems is a promising approach for the design and optimization of image-guided nanomedicines, especially in the context of cancer gene therapy.
Collapse
Affiliation(s)
- Istvan Hajdu
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada,
| | - Amal Makhlouf
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada, .,Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, 12411 Cairo, Egypt
| | - Viswas Raja Solomon
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada,
| | - Deborah Michel
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada,
| | - Mays Al-Dulaymi
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada,
| | - Kishor M Wasan
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada,
| | - Humphrey Fonge
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada, .,Department of Medical Imaging, Royal University Hospital Saskatoon, SK S7N 0W8, Canada,
| | - Ildiko Badea
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada,
| |
Collapse
|
24
|
Arms L, Smith DW, Flynn J, Palmer W, Martin A, Woldu A, Hua S. Advantages and Limitations of Current Techniques for Analyzing the Biodistribution of Nanoparticles. Front Pharmacol 2018; 9:802. [PMID: 30154715 PMCID: PMC6102329 DOI: 10.3389/fphar.2018.00802] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 07/03/2018] [Indexed: 12/22/2022] Open
Abstract
Nanomedicines are typically submicrometer-sized carrier materials (nanoparticles) encapsulating therapeutic and/or imaging compounds that are used for the prevention, diagnosis and treatment of diseases. They are increasingly being used to overcome biological barriers in the body to improve the way we deliver compounds to specific tissues and organs. Nanomedicine technology aims to improve the balance between the efficacy and the toxicity of therapeutic compounds. Nanoparticles, one of the key technologies of nanomedicine, can exhibit a combination of physical, chemical and biological characteristics that determine their in vivo behavior. A key component in the translational assessment of nanomedicines is determining the biodistribution of the nanoparticles following in vivo administration in animals and humans. There are a range of techniques available for evaluating nanoparticle biodistribution, including histology, electron microscopy, liquid scintillation counting (LSC), indirectly measuring drug concentrations, in vivo optical imaging, computed tomography (CT), magnetic resonance imaging (MRI), and nuclear medicine imaging. Each technique has its own advantages and limitations, as well as capabilities for assessing real-time, whole-organ and cellular accumulation. This review will address the principles and methodology of each technique and their advantages and limitations for evaluating in vivo biodistribution of nanoparticles.
Collapse
Affiliation(s)
- Lauren Arms
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
| | - Doug W. Smith
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
| | - Jamie Flynn
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - William Palmer
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW, Australia
| | - Antony Martin
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW, Australia
| | - Ameha Woldu
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Susan Hua
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| |
Collapse
|
25
|
Sultan D, Ye D, Heo GS, Zhang X, Luehmann H, Yue Y, Detering L, Komarov S, Taylor S, Tai YC, Rubin JB, Chen H, Liu Y. Focused Ultrasound Enabled Trans-Blood Brain Barrier Delivery of Gold Nanoclusters: Effect of Surface Charges and Quantification Using Positron Emission Tomography. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1703115. [PMID: 29966035 DOI: 10.1002/smll.201703115] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 04/30/2018] [Indexed: 05/27/2023]
Abstract
Focused ultrasound (FUS) technology is reported to enhance the delivery of 64 Cu-integrated ultrasmall gold nanoclusters (64 Cu-AuNCs) across the blood-brain barrier (BBB) as measured by positron emission tomography (PET). To better define the optimal physical properties for brain delivery, 64 Cu-AuNCs with different surface charges are synthesized and characterized. In vivo biodistribution studies are performed to compare the individual organ uptake of each type of 64 Cu-AuNCs. Quantitative PET imaging post-FUS treatment shows site-targeted brain penetration, retention, and diffusion of the negative, neutral, and positive 64 Cu-AuNCs. Autoradiography is performed to compare the intrabrain distribution of these nanoclusters. PET Imaging demonstrates the effective BBB opening and successful delivery of 64 Cu-AuNCs into the brain. Of the three 64 Cu-AuNCs investigated, the neutrally charged nanostructure performs the best and is the candidate platform for future theranostic applications in neuro-oncology.
Collapse
Affiliation(s)
- Deborah Sultan
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Dezhuang Ye
- Department of Mechanical Engineering and Material Science, Washington University in St. Louis, St. Louis, MO, 63130, USA
| | - Gyu Seong Heo
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Xiaohui Zhang
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Hannah Luehmann
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Yimei Yue
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, 63130, USA
| | - Lisa Detering
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Sergey Komarov
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Sara Taylor
- Department of Pediatrics and Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Yuan-Chuan Tai
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Joshua B Rubin
- Department of Pediatrics and Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Hong Chen
- Department of Biomedical Engineering and Department of Radiation Oncology, Washington University in St. Louis, St. Louis, MO, 63130, USA
| | - Yongjian Liu
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| |
Collapse
|
26
|
Stéen EJL, Edem PE, Nørregaard K, Jørgensen JT, Shalgunov V, Kjaer A, Herth MM. Pretargeting in nuclear imaging and radionuclide therapy: Improving efficacy of theranostics and nanomedicines. Biomaterials 2018; 179:209-245. [PMID: 30007471 DOI: 10.1016/j.biomaterials.2018.06.021] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 06/13/2018] [Accepted: 06/14/2018] [Indexed: 01/18/2023]
Abstract
Pretargeted nuclear imaging and radiotherapy have recently attracted increasing attention for diagnosis and treatment of cancer with nanomedicines. This is because it conceptually offers better imaging contrast and therapeutic efficiency while reducing the dose to radiosensitive tissues compared to conventional strategies. In conventional imaging and radiotherapy, a directly radiolabeled nano-sized vector is administered and allowed to accumulate in the tumor, typically on a timescale of several days. In contrast, pretargeting is based on a two-step approach. First, a tumor-accumulating vector carrying a tag is administered followed by injection of a fast clearing radiolabeled agent that rapidly recognizes the tag of the tumor-bound vector in vivo. Therefore, pretargeting circumvents the use of long-lived radionuclides that is a necessity for sufficient tumor accumulation and target-to-background ratios using conventional approaches. In this review, we give an overview of recent advances in pretargeted imaging strategies. We will critically reflect on the advantages and disadvantages of current state-of-the-art conventional imaging approaches and compare them to pretargeted strategies. We will discuss the pretargeted imaging concept and the involved chemistry. Finally, we will discuss the steps forward in respect to clinical translation, and how pretargeted strategies could be applied to improve state-of-the-art radiotherapeutic approaches.
Collapse
Affiliation(s)
- E Johanna L Stéen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 160, DK-2100 Copenhagen, Denmark; Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - Patricia E Edem
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 160, DK-2100 Copenhagen, Denmark; Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark; Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2100 Copenhagen, Denmark
| | - Kamilla Nørregaard
- Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark; Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2100 Copenhagen, Denmark
| | - Jesper T Jørgensen
- Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark; Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2100 Copenhagen, Denmark
| | - Vladimir Shalgunov
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 160, DK-2100 Copenhagen, Denmark
| | - Andreas Kjaer
- Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark; Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2100 Copenhagen, Denmark
| | - Matthias M Herth
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 160, DK-2100 Copenhagen, Denmark; Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark.
| |
Collapse
|
27
|
Zhang P, Cui Y, Anderson CF, Zhang C, Li Y, Wang R, Cui H. Peptide-based nanoprobes for molecular imaging and disease diagnostics. Chem Soc Rev 2018; 47:3490-3529. [PMID: 29497722 DOI: 10.1039/c7cs00793k] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Pathological changes in a diseased site are often accompanied by abnormal activities of various biomolecules in and around the involved cells. Identifying the location and expression levels of these biomolecules could enable early-stage diagnosis of the related disease, the design of an appropriate treatment strategy, and the accurate assessment of the treatment outcomes. Over the past two decades, a great diversity of peptide-based nanoprobes (PBNs) have been developed, aiming to improve the in vitro and in vivo performances of water-soluble molecular probes through engineering of their primary chemical structures as well as the physicochemical properties of their resultant assemblies. In this review, we introduce strategies and approaches adopted for the identification of functional peptides in the context of molecular imaging and disease diagnostics, and then focus our discussion on the design and construction of PBNs capable of navigating through physiological barriers for targeted delivery and improved specificity and sensitivity in recognizing target biomolecules. We highlight the biological and structural roles that low-molecular-weight peptides play in PBN design and provide our perspectives on the future development of PBNs for clinical translation.
Collapse
Affiliation(s)
- Pengcheng Zhang
- State Key Laboratory of Drug Research & Center for Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China.
| | | | | | | | | | | | | |
Collapse
|
28
|
Affiliation(s)
- Lingzhou Zhao
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Xiangyang Shi
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, People’s Republic of China
- CQM-Centro de Química da Madeira, Universidade da Madeira, Funchal, Portugal
| | - Jinhua Zhao
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| |
Collapse
|
29
|
Lamb J, Holland JP. Advanced Methods for Radiolabeling Multimodality Nanomedicines for SPECT/MRI and PET/MRI. J Nucl Med 2017; 59:382-389. [PMID: 29025988 DOI: 10.2967/jnumed.116.187419] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 09/30/2017] [Indexed: 12/17/2022] Open
Abstract
The advent of hybrid cameras that combine MRI with either SPECT or PET has stimulated growing interest in developing multimodality imaging probes. Countless options are available for fusing magnetically active species with positron- or γ-ray-emitting radionuclides. The initial problem is one of choice: which chemical systems are a suitable basis for developing hybrid imaging agents? Any attempt to answer this question must also address how the physical, chemical, and biologic properties of a unified imaging agent can be tailored to ensure that optimum specificity and contrast are achieved simultaneously for both imaging modalities. Nanoparticles have emerged as attractive platforms for building multimodality radiotracers for SPECT/MRI and PET/MRI. A wide variety of nanoparticle constructs have been utilized as radiotracers, but irrespective of the particle class, radiolabeling remains a key step. Classic methods for radiolabeling nanoparticles involve functionalization of the particle surface, core, or coating. These modifications typically rely on using traditional metal ion chelate or prosthetic group chemistries. Though seemingly innocuous, appending nanoparticles with these radiolabeling handles can have dramatic effects on important properties such as particle size, charge, and solubility. In turn, alterations in the chemical and physical properties of the nanoparticle often have a negative impact on their pharmacologic profile. A central challenge in radiolabeling nanoparticles is to identify alternative chemical methods that facilitate the introduction of a radioactive nuclide without detrimental effects on the pharmacokinetic and toxicologic properties of the construct. Efforts to solve this challenge have generated a range of innovative chelate-free radiolabeling methods that exploit intrinsic chemical features of nanoparticles. Here, the chemistry of 9 mechanistically distinct methods for radiolabeling nanoparticles is presented. This discourse illustrates the evolution of nanoparticle radiochemistry from classic approaches to modern chelate-free or intrinsic methods.
Collapse
Affiliation(s)
- Jennifer Lamb
- Department of Chemistry, University of Zurich, Zurich, Switzerland
| | - Jason P Holland
- Department of Chemistry, University of Zurich, Zurich, Switzerland
| |
Collapse
|
30
|
Zhao L, Zhu M, Li Y, Xing Y, Zhao J. Radiolabeled Dendrimers for Nuclear Medicine Applications. Molecules 2017; 22:E1350. [PMID: 28841180 PMCID: PMC6151832 DOI: 10.3390/molecules22091350] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 08/10/2017] [Accepted: 08/10/2017] [Indexed: 12/11/2022] Open
Abstract
Recent advances in nuclear medicine have explored nanoscale carriers for targeted delivery of various radionuclides in specific manners to improve the effect of diagnosis and therapy of diseases. Due to the unique molecular architecture allowing facile attachment of targeting ligands and radionuclides, dendrimers provide versatile platforms in this filed to build abundant multifunctional radiolabeled nanoparticles for nuclear medicine applications. This review gives special focus to recent advances in dendrimer-based nuclear medicine agents for the imaging and treatment of cancer, cardiovascular and other diseases. Radiolabeling strategies for different radionuclides and several challenges involved in clinical translation of radiolabeled dendrimers are extensively discussed.
Collapse
Affiliation(s)
- Lingzhou Zhao
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.
| | - Meilin Zhu
- Basic Medical College, Ningxia Medical University, Yinchuan 750004, Ningxia, China.
| | - Yujie Li
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.
| | - Yan Xing
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.
| | - Jinhua Zhao
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.
| |
Collapse
|
31
|
Huang L, Hu J, Huang S, Wang B, Siaw-Debrah F, Nyanzu M, Zhang Y, Zhuge Q. Nanomaterial applications for neurological diseases and central nervous system injury. Prog Neurobiol 2017; 157:29-48. [PMID: 28743465 DOI: 10.1016/j.pneurobio.2017.07.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 07/18/2017] [Accepted: 07/18/2017] [Indexed: 12/20/2022]
Abstract
The effectiveness of noninvasive treatment for neurological disease is generally limited by the poor entry of therapeutic agents into the central nervous system (CNS). Most CNS drugs cannot permeate into the brain parenchyma because of the blood-brain barrier thus, overcoming this problem has become one of the most significant challenges in the development of neurological therapeutics. Nanotechnology has emerged as an innovative alternative for treating neurological diseases. In fact, rapid advances in nanotechnology have provided promising solutions to this challenge. This review highlights the applications of nanomaterials in the developing neurological field and discusses the evidence for their efficacies.
Collapse
Affiliation(s)
- Lijie Huang
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China; Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China
| | - Jiangnan Hu
- Center for Neuroscience Discovery, Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China
| | - Shengwei Huang
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China; Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China
| | - Brian Wang
- Center for Neuroscience Discovery, Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Felix Siaw-Debrah
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China
| | - Mark Nyanzu
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China
| | - Yu Zhang
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China
| | - Qichuan Zhuge
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China; Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China.
| |
Collapse
|
32
|
Montiel Schneider MG, Lassalle VL. Magnetic iron oxide nanoparticles as novel and efficient tools for atherosclerosis diagnosis. Biomed Pharmacother 2017; 93:1098-1115. [PMID: 28738519 DOI: 10.1016/j.biopha.2017.07.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 06/14/2017] [Accepted: 07/05/2017] [Indexed: 01/09/2023] Open
Abstract
Cardiovascular complications derivate from atherosclerosis are the main cause of death in western world. An early detection of vulnerable atherosclerotic plaques is primordial for a better care of patients suffering the pathology. In this context nanotechnology has emerged as a promising tool to achieve this goal. Nanoparticles based on magnetic iron oxide (MNPs) have been extensively studied in cardiovascular diseases diagnosis, as well as in the treatment and diagnostic of other pathologies. The present review aims to describe and analyze the most current literature regarding to this topic, offering the level of detail required to reproduce the experimental tasks providing a critical input of the latest available reports. The current diagnostic features are presented and compared, highlighting their advantages and disadvantages. Information on novel technology intended to this purpose is also recompiled and in deep analyzed. Special emphasis is placed in magnetic nanotechnology, remarking the possibility to assess selective and multifunctional systems to the early detection of artherosclerotic pathologies. Finally, in view of the state of the art, the future perspectives about the trends on MNPs in artherosclerorsis diagnostic and treatment have also been addressed.
Collapse
Affiliation(s)
| | - Verónica Leticia Lassalle
- INQUISUR, Departamento de Química, Universidad Nacional del Sur (UNS)-CONICET, Av. Alem 1253, 8000 Bahía Blanca, Argentina.
| |
Collapse
|
33
|
Real-time liver uptake and biodistribution of magnetic nanoparticles determined by AC biosusceptometry. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 13:1519-1529. [DOI: 10.1016/j.nano.2017.02.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 01/30/2017] [Accepted: 02/05/2017] [Indexed: 01/05/2023]
|
34
|
Pant K, Sedláček O, Nadar RA, Hrubý M, Stephan H. Radiolabelled Polymeric Materials for Imaging and Treatment of Cancer: Quo Vadis? Adv Healthc Mater 2017; 6. [PMID: 28218487 DOI: 10.1002/adhm.201601115] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/24/2016] [Indexed: 12/15/2022]
Abstract
Owing to their tunable blood circulation time and suitable plasma stability, polymer-based nanomaterials hold a great potential for designing and utilising multifunctional nanocarriers for efficient imaging and effective treatment of cancer. When tagged with appropriate radionuclides, they may allow for specific detection (diagnosis) as well as the destruction of tumours (therapy) or even customization of materials, aiming to both diagnosis and therapy (theranostic approach). This review provides an overview of recent developments of radiolabelled polymeric nanomaterials (natural and synthetic polymers) for molecular imaging of cancer, specifically, applying nuclear techniques such as positron emission tomography (PET) and single-photon emission computed tomography (SPECT). Different approaches to radiolabel polymers are evaluated from the methodical radiochemical point of view. This includes new bifunctional chelating agents (BFCAs) for radiometals as well as novel labelling methods. Special emphasis is given to eligible strategies employed to evade the mononuclear phagocytic system (MPS) in view of efficient targeting. The discussion encompasses promising strategies currently employed as well as emerging possibilities in radionuclide-based cancer therapy. Key issues involved in the clinical translation of radiolabelled polymers and future scopes of this intriguing research field are also discussed.
Collapse
Affiliation(s)
- Kritee Pant
- Helmholtz-Zentrum Dresden-Rossendorf; Institute of Radiopharmaceutical Cancer Research; Bautzner Landstraße 400 01328 Dresden Germany
| | - Ondřej Sedláček
- Institute of Macromolecular Chemistry; The Academy of Sciences of the Czech Republic; Heyrovského námeˇstí 2 16206 Prague 6 Czech Republic
| | - Robin A. Nadar
- Helmholtz-Zentrum Dresden-Rossendorf; Institute of Radiopharmaceutical Cancer Research; Bautzner Landstraße 400 01328 Dresden Germany
| | - Martin Hrubý
- Institute of Macromolecular Chemistry; The Academy of Sciences of the Czech Republic; Heyrovského námeˇstí 2 16206 Prague 6 Czech Republic
| | - Holger Stephan
- Helmholtz-Zentrum Dresden-Rossendorf; Institute of Radiopharmaceutical Cancer Research; Bautzner Landstraße 400 01328 Dresden Germany
| |
Collapse
|
35
|
Hong SC, Yoo SY, Kim H, Lee J. Chitosan-Based Multifunctional Platforms for Local Delivery of Therapeutics. Mar Drugs 2017; 15:md15030060. [PMID: 28257059 PMCID: PMC5367017 DOI: 10.3390/md15030060] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 02/21/2017] [Accepted: 02/24/2017] [Indexed: 12/24/2022] Open
Abstract
Chitosan has been widely used as a key biomaterial for the development of drug delivery systems intended to be administered via oral and parenteral routes. In particular, chitosan-based microparticles are the most frequently employed delivery system, along with specialized systems such as hydrogels, nanoparticles and thin films. Based on the progress made in chitosan-based drug delivery systems, the usefulness of chitosan has further expanded to anti-cancer chemoembolization, tissue engineering, and stem cell research. For instance, chitosan has been used to develop embolic materials designed to efficiently occlude the blood vessels by which the oxygen and nutrients are supplied. Indeed, it has been reported to be a promising embolic material. For better anti-cancer effect, embolic materials that can locally release anti-cancer drugs were proposed. In addition, a complex of radioactive materials and chitosan to be locally injected into the liver has been investigated as an efficient therapeutic tool for hepatocellular carcinoma. In line with this, a number of attempts have been explored to use chitosan-based carriers for the delivery of various agents, especially to the site of interest. Thus, in this work, studies where chitosan-based drug delivery systems have successfully been used for local delivery will be presented along with future perspectives.
Collapse
Affiliation(s)
- Seong-Chul Hong
- College of Pharmacy, Chung-Ang University, Seoul 06974, Korea.
| | - Seung-Yup Yoo
- College of Pharmacy, Chung-Ang University, Seoul 06974, Korea.
| | - Hyeongmin Kim
- College of Pharmacy, Chung-Ang University, Seoul 06974, Korea.
| | - Jaehwi Lee
- College of Pharmacy, Chung-Ang University, Seoul 06974, Korea.
| |
Collapse
|
36
|
Richards DA, Maruani A, Chudasama V. Antibody fragments as nanoparticle targeting ligands: a step in the right direction. Chem Sci 2017; 8:63-77. [PMID: 28451149 PMCID: PMC5304706 DOI: 10.1039/c6sc02403c] [Citation(s) in RCA: 172] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 09/05/2016] [Indexed: 12/13/2022] Open
Abstract
Recent advances in nanomedicine have shown that dramatic improvements in nanoparticle therapeutics and diagnostics can be achieved through the use of disease specific targeting ligands. Although immunoglobulins have successfully been employed for the generation of actively targeted nanoparticles, their use is often hampered by the suboptimal characteristics of the resulting complexes. Emerging data suggest that a switch in focus from full antibodies to antibody derived fragments could help to alleviate these problems and expand the potential of antibody-nanoparticle conjugates as biomedical tools. This review aims to highlight how antibody derived fragments have been utilised to overcome both fundamental and practical issues encountered during the design and application of antibody-targeted nanoparticles.
Collapse
Affiliation(s)
- Daniel A Richards
- Department of Chemistry , University College London , 20 Gordon Street , London , WC1H 0AJ , UK . ; ; Tel: +44 (0)207 679 2077
| | - Antoine Maruani
- Department of Chemistry , University College London , 20 Gordon Street , London , WC1H 0AJ , UK . ; ; Tel: +44 (0)207 679 2077
| | - Vijay Chudasama
- Department of Chemistry , University College London , 20 Gordon Street , London , WC1H 0AJ , UK . ; ; Tel: +44 (0)207 679 2077
| |
Collapse
|
37
|
Liu JM, Liu YY, Zhang DD, Fang GZ, Wang S. Synthesis of GdAlO 3:Mn 4+,Ge 4+@Au Core-Shell Nanoprobes with Plasmon-Enhanced Near-Infrared Persistent Luminescence for in Vivo Trimodality Bioimaging. ACS APPLIED MATERIALS & INTERFACES 2016; 8:29939-29949. [PMID: 27759378 DOI: 10.1021/acsami.6b09580] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
The rise of multimodal nanoprobes has promoted the development of new methods to explore multiple molecular targets simultaneously or to combine various bioimaging tools in one assay to more clearly delineate localization and expression of biomarkers. Persistent luminescence nanophosphors (PLNPs) have been qualified as a promising contrast agent for in vivo imaging. The easy surface modification and proper nanostructure design strategy would favor the fabrication of PLNP-based multifunctional nanoprobes for biological application. In this paper, we have proposed novel multifunctional core-shell nanomaterials, applying the Mn4+ and Ge4+ co-doped gadolinium aluminate (GdAlO3:Mn4+,Ge4+) PLNPs as the near-infrared persistent luminescence emission center and introducing the gold nanoshell coated on the PLNPs to enhance the luminescence efficiency via plasmon resonance. Our developed core-shell nanoprobes have demonstrated the excellent features of ultrabrightness, superlong afterglow, good monodispersity, low toxicity, and excellent biocompatibility. The well-characterized nanoprobes have been utilized for trimodality in vivo imaging, with near-infrared persistent luminescence for optical imaging, Gd element for magnetic resonance imaging, and Au element for computed tomography imaging.
Collapse
Affiliation(s)
- Jing-Min Liu
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin University of Science and Technology , Tianjin, 300457, China
| | - Yao-Yao Liu
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin University of Science and Technology , Tianjin, 300457, China
| | - Dong-Dong Zhang
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin University of Science and Technology , Tianjin, 300457, China
| | - Guo-Zhen Fang
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin University of Science and Technology , Tianjin, 300457, China
| | - Shuo Wang
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin University of Science and Technology , Tianjin, 300457, China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology & Business University (BTBU) , Beijing, 100048, China
| |
Collapse
|
38
|
Woodard PK, Liu Y, Pressly ED, Luehmann HP, Detering L, Sultan DE, Laforest R, McGrath AJ, Gropler RJ, Hawker CJ. Design and Modular Construction of a Polymeric Nanoparticle for Targeted Atherosclerosis Positron Emission Tomography Imaging: A Story of 25% (64)Cu-CANF-Comb. Pharm Res 2016; 33:2400-10. [PMID: 27286872 PMCID: PMC5096390 DOI: 10.1007/s11095-016-1963-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 06/01/2016] [Indexed: 01/21/2023]
Abstract
PURPOSE To assess the physicochemical properties, pharmacokinetic profiles, and in vivo positron emission tomography (PET) imaging of natriuretic peptide clearance receptors (NPRC) expressed on atherosclerotic plaque of a series of targeted, polymeric nanoparticles. METHODS To control their structure, non-targeted and targeted polymeric (comb) nanoparticles, conjugated with various amounts of c-atrial natriuretic peptide (CANF, 0, 5, 10 and 25%), were synthesized by controlled and modular chemistry. In vivo pharmacokinetic evaluation of these nanoparticles was performed in wildtype (WT) C57BL/6 mice after (64)Cu radiolabeling. PET imaging was performed on an apolipoprotein E-deficient (ApoE(-/-)) mouse atherosclerosis model to assess the NPRC targeting efficiency. For comparison, an in vivo blood metabolism study was carried out in WT mice. RESULTS All three (64)Cu-CANF-comb nanoparticles showed improved biodistribution profiles, including significantly reduced accumulation in both liver and spleen, compared to the non-targeted (64)Cu-comb. Of the three nanoparticles, the 25% (64)Cu-CANF-comb demonstrated the best NPRC targeting specificity and sensitivity in ApoE(-/-) mice. Metabolism studies showed that the radiolabeled CANF-comb was stable in blood up to 9 days. Histopathological analyses confirmed the up-regulation of NPRC along the progression of atherosclerosis. CONCLUSION The 25% (64)Cu-CANF-comb demonstrated its potential as a PET imaging agent to detect atherosclerosis progression and status.
Collapse
Affiliation(s)
- Pamela K Woodard
- Department of Radiology, Washington University, St. Louis, Missouri, USA
| | - Yongjian Liu
- Department of Radiology, Washington University, St. Louis, Missouri, USA
| | - Eric D Pressly
- Materials Research Laboratory, University of California, Santa Barbara, California,, USA
| | - Hannah P Luehmann
- Department of Radiology, Washington University, St. Louis, Missouri, USA
| | - Lisa Detering
- Department of Radiology, Washington University, St. Louis, Missouri, USA
| | - Deborah E Sultan
- Department of Radiology, Washington University, St. Louis, Missouri, USA
| | - Richard Laforest
- Department of Radiology, Washington University, St. Louis, Missouri, USA
| | - Alaina J McGrath
- Materials Research Laboratory, University of California, Santa Barbara, California,, USA
| | - Robert J Gropler
- Department of Radiology, Washington University, St. Louis, Missouri, USA
| | - Craig J Hawker
- Materials Research Laboratory, University of California, Santa Barbara, California,, USA.
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, California,, USA.
- Materials Department, University of California, Santa Barbara, California, USA.
| |
Collapse
|
39
|
Aanei IL, ElSohly AM, Farkas ME, Netirojjanakul C, Regan M, Taylor Murphy S, O'Neil JP, Seo Y, Francis MB. Biodistribution of Antibody-MS2 Viral Capsid Conjugates in Breast Cancer Models. Mol Pharm 2016; 13:3764-3772. [PMID: 27611245 DOI: 10.1021/acs.molpharmaceut.6b00566] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A variety of nanoscale scaffolds, including virus-like particles (VLPs), are being developed for biomedical applications; however, little information is available about their in vivo behavior. Targeted nanoparticles are particularly valuable as diagnostic and therapeutic carriers because they can increase the signal-to-background ratio of imaging agents, improve the efficacy of drugs, and reduce adverse effects by concentrating the therapeutic molecule in the region of interest. The genome-free capsid of bacteriophage MS2 has several features that make it well-suited for use in delivery applications, such as facile production and modification, the ability to display multiple copies of targeting ligands, and the capacity to deliver large payloads. Anti-EGFR antibodies were conjugated to MS2 capsids to construct nanoparticles targeted toward receptors overexpressed on breast cancer cells. The MS2 agents showed good stability in physiological conditions up to 2 days and specific binding to the targeted receptors in in vitro experiments. Capsids radiolabeled with 64Cu isotopes were injected into mice possessing tumor xenografts, and both positron emission tomography-computed tomography (PET/CT) and scintillation counting of the organs ex vivo were used to determine the localization of the agents. The capsids exhibit surprisingly long circulation times (10-15% ID/g in blood at 24 h) and moderate tumor uptake (2-5% ID/g). However, the targeting antibodies did not lead to increased uptake in vivo despite in vitro enhancements, suggesting that extravasation is a limiting factor for delivery to tumors by these particles.
Collapse
Affiliation(s)
- Ioana L Aanei
- Department of Chemistry, University of California, Berkeley , Berkeley, California 94720, United States.,Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratories , Berkeley, California 94720, United States
| | - Adel M ElSohly
- Department of Chemistry, University of California, Berkeley , Berkeley, California 94720, United States
| | - Michelle E Farkas
- Department of Chemistry, University of California, Berkeley , Berkeley, California 94720, United States
| | - Chawita Netirojjanakul
- Department of Chemistry, University of California, Berkeley , Berkeley, California 94720, United States
| | - Melanie Regan
- Department of Radiology and Biomedical Imaging, University of California, San Francisco , San Francisco, California 94143, United States
| | - Stephanie Taylor Murphy
- Department of Radiology and Biomedical Imaging, University of California, San Francisco , San Francisco, California 94143, United States
| | - James P O'Neil
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratories , Berkeley, California 94720, United States
| | - Youngho Seo
- Department of Radiology and Biomedical Imaging, University of California, San Francisco , San Francisco, California 94143, United States
| | - Matthew B Francis
- Department of Chemistry, University of California, Berkeley , Berkeley, California 94720, United States.,Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratories , Berkeley, California 94720, United States
| |
Collapse
|
40
|
Luehmann HP, Detering L, Fors BP, Pressly ED, Woodard PK, Randolph GJ, Gropler RJ, Hawker CJ, Liu Y. PET/CT Imaging of Chemokine Receptors in Inflammatory Atherosclerosis Using Targeted Nanoparticles. J Nucl Med 2016; 57:1124-9. [PMID: 26795285 PMCID: PMC5088780 DOI: 10.2967/jnumed.115.166751] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Accepted: 12/07/2015] [Indexed: 12/17/2022] Open
Abstract
UNLABELLED Atherosclerosis is inherently an inflammatory process that is strongly affected by the chemokine-chemokine receptor axes regulating the trafficking of inflammatory cells at all stages of the disease. Of the chemokine receptor family, some specifically upregulated on macrophages play a critical role in plaque development and may have the potential to track plaque progression. However, the diagnostic potential of these chemokine receptors has not been fully realized. On the basis of our previous work using a broad-spectrum peptide antagonist imaging 8 chemokine receptors together, the purpose of this study was to develop a targeted nanoparticle for sensitive and specific detection of these chemokine receptors in both a mouse vascular injury model and a spontaneously developed mouse atherosclerosis model. METHODS The viral macrophage inflammatory protein-II (vMIP-II) was conjugated to a biocompatible poly(methyl methacrylate)-core/polyethylene glycol-shell amphiphilic comblike nanoparticle through controlled conjugation and polymerization before radiolabeling with (64)Cu for PET imaging in an apolipoprotein E-deficient (ApoE(-/-)) mouse vascular injury model and a spontaneous ApoE(-/-) mouse atherosclerosis model. Histology, immunohistochemistry, and real-time reverse transcription polymerase chain reaction were performed to assess the plaque progression and upregulation of chemokine receptors. RESULTS The chemokine receptor-targeted (64)Cu-vMIP-II-comb showed extended blood retention and improved biodistribution. PET imaging showed specific tracer accumulation at plaques in ApoE(-/-) mice, confirmed by competitive receptor blocking studies and assessment in wild-type mice. Histopathologic characterization showed the progression of plaque including size and macrophage population, corresponding to the elevated concentration of chemokine receptors and more importantly increased PET signals. CONCLUSION This work provides a useful nanoplatform for sensitive and specific detection of chemokine receptors to assess plaque progression in mouse atherosclerosis models.
Collapse
Affiliation(s)
- Hannah P. Luehmann
- Department of Radiology, Washington University, Campus Box 8225, 510
S. Kingshighway Blvd., St. Louis, MO 63110
| | - Lisa Detering
- Department of Radiology, Washington University, Campus Box 8225, 510
S. Kingshighway Blvd., St. Louis, MO 63110
| | - Brett P. Fors
- Department of Materials, Chemistry and Biochemistry, University of
California, Santa Barbara, California
| | - Eric D. Pressly
- Department of Materials, Chemistry and Biochemistry, University of
California, Santa Barbara, California
| | - Pamela K. Woodard
- Department of Radiology, Washington University, Campus Box 8225, 510
S. Kingshighway Blvd., St. Louis, MO 63110
| | | | - Robert J. Gropler
- Department of Radiology, Washington University, Campus Box 8225, 510
S. Kingshighway Blvd., St. Louis, MO 63110
| | - Craig J. Hawker
- Department of Materials, Chemistry and Biochemistry, University of
California, Santa Barbara, California
| | - Yongjian Liu
- Department of Radiology, Washington University, Campus Box 8225, 510
S. Kingshighway Blvd., St. Louis, MO 63110
| |
Collapse
|
41
|
Zhao Y, Detering L, Sultan D, Cooper ML, You M, Cho S, Meier SL, Luehmann H, Sun G, Rettig M, Dehdashti F, Wooley KL, DiPersio JF, Liu Y. Gold Nanoclusters Doped with (64)Cu for CXCR4 Positron Emission Tomography Imaging of Breast Cancer and Metastasis. ACS NANO 2016; 10:5959-70. [PMID: 27159079 PMCID: PMC5479491 DOI: 10.1021/acsnano.6b01326] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
As an emerging class of nanomaterial, nanoclusters hold great potential for biomedical applications due to their unique sizes and related properties. Herein, we prepared a (64)Cu doped gold nanocluster ((64)CuAuNC, hydrodynamic size: 4.2 ± 0.5 nm) functionalized with AMD3100 (or Plerixafor) for targeted positron emission tomography (PET) imaging of CXCR4, an up-regulated receptor on primary tumor and lung metastasis in a mouse 4T1 orthotopic breast cancer model. The preparation of targeted (64)CuAuNCs-AMD3100 (4.5 ± 0.4 nm) was done via one-step reaction with controlled conjugation of AMD3100 and specific activity, as well as improved colloid stability. In vivo pharmacokinetic evaluation showed favorable organ distribution and significant renal and fecal clearance within 48 h post injection. The expression of CXCR4 in tumors and metastasis was characterized by immunohistochemistry, Western blot, and reverse transcription polymerase chain reaction analysis. PET imaging with (64)CuAuNCs-AMD3100 demonstrated sensitive and accurate detection of CXCR4 in engineered tumors expressing various levels of the receptor, while competitive receptor blocking studies confirmed targeting specificity of the nanoclusters. In contrast to nontargeted (64)CuAuNCs and (64)Cu-AMD3100 alone, the targeted (64)CuAuNCs-AMD3100 detected up-regulated CXCR4 in early stage tumors and premetastatic niche of lung earlier and with greater sensitivity. Taken together, we believe that (64)CuAuNCs-AMD3100 could serve as a useful platform for early and accurate detection of breast cancer and metastasis providing an essential tool to guide the treatment.
Collapse
Affiliation(s)
- Yongfeng Zhao
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, Missouri, 63110, United States
| | - Lisa Detering
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, Missouri, 63110, United States
| | - Deborah Sultan
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, Missouri, 63110, United States
| | - Matthew L Cooper
- Department of Medicine, Washington University, St. Louis, Missouri, 63110, United States
| | - Meng You
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, Missouri, 63110, United States
| | - Sangho Cho
- Department of Chemistry, Department of Chemical Engineering, and Department of Materials Science and Engineering, Texas A&M University, College Station, TX 77842, United States
| | - Stephanie L. Meier
- Department of Medicine, Washington University, St. Louis, Missouri, 63110, United States
| | - Hannah Luehmann
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, Missouri, 63110, United States
| | - Guorong Sun
- Department of Chemistry, Department of Chemical Engineering, and Department of Materials Science and Engineering, Texas A&M University, College Station, TX 77842, United States
| | - Michael Rettig
- Department of Medicine, Washington University, St. Louis, Missouri, 63110, United States
| | - Farrokh Dehdashti
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, Missouri, 63110, United States
| | - Karen L. Wooley
- Department of Chemistry, Department of Chemical Engineering, and Department of Materials Science and Engineering, Texas A&M University, College Station, TX 77842, United States
| | - John F. DiPersio
- Department of Medicine, Washington University, St. Louis, Missouri, 63110, United States
| | - Yongjian Liu
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, Missouri, 63110, United States
- Corresponding Author: Address correspondence to:
| |
Collapse
|
42
|
Badun GA, Chernysheva MG, Aldobaev VN. Preparation of tritium-labeled modified single-walled carbon nanotubes for pharmacokinetic studies. RADIOCHEMISTRY 2016. [DOI: 10.1134/s1066362216030139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
43
|
Zhao Y, Pang B, Luehmann H, Detering L, Yang X, Sultan D, Harpstrite S, Sharma V, Cutler CS, Xia Y, Liu Y. Gold Nanoparticles Doped with (199) Au Atoms and Their Use for Targeted Cancer Imaging by SPECT. Adv Healthc Mater 2016; 5:928-35. [PMID: 26865221 PMCID: PMC4836969 DOI: 10.1002/adhm.201500992] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 01/09/2016] [Indexed: 12/18/2022]
Abstract
Gold nanoparticles have been labeled with various radionuclides and extensively explored for single photon emission computed tomography (SPECT) in the context of cancer diagnosis. The stability of most radiolabels, however, still needs to be improved for accurate detection of cancer biomarkers and thereby monitoring of tumor progression and metastasis. Here, the first synthesis of Au nanoparticles doped with (199)Au atoms for targeted SPECT tumor imaging in a mouse triple negative breast cancer (TNBC) model is reported. By directly incorporating (199)Au atoms into the crystal lattice of each Au nanoparticle, the stability of the radiolabel can be ensured. The synthetic procedure also allows for a precise control over both the radiochemistry and particle size. When conjugated with D-Ala1-peptide T-amide, the Au nanoparticles doped with (199)Au atoms can serve as a C-C chemokine receptor 5 (CCR5)-targeted nanoprobe for the sensitive and specific detection of both TNBC and its metastasis in a mouse tumor model.
Collapse
Affiliation(s)
- Yongfeng Zhao
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Bo Pang
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
- Department of Biomedical Engineering, Peking University, Beijing, 100871, P. R. China
| | - Hannah Luehmann
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Lisa Detering
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Xuan Yang
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
| | - Deborah Sultan
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Scott Harpstrite
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Vijay Sharma
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Cathy S Cutler
- Research Reactor Center, University of Missouri, Columbia, MO, 65211, USA
| | - Younan Xia
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
| | - Yongjian Liu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| |
Collapse
|
44
|
Revia RA, Zhang M. Magnetite nanoparticles for cancer diagnosis, treatment, and treatment monitoring: recent advances. MATERIALS TODAY (KIDLINGTON, ENGLAND) 2016; 19:157-168. [PMID: 27524934 PMCID: PMC4981486 DOI: 10.1016/j.mattod.2015.08.022] [Citation(s) in RCA: 353] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
The development of nanoparticles (NPs) for use in all facets of oncological disease detection and therapy has shown great progress over the past two decades. NPs have been tailored for use as contrast enhancement agents for imaging, drug delivery vehicles, and most recently as a therapeutic component in initiating tumor cell death in magnetic and photonic ablation therapies. Of the many possible core constituents of NPs, such as gold, silver, carbon nanotubes, fullerenes, manganese oxide, lipids, micelles, etc., iron oxide (or magnetite) based NPs have been extensively investigated due to their excellent superparamagnetic, biocompatible, and biodegradable properties. This review addresses recent applications of magnetite NPs in diagnosis, treatment, and treatment monitoring of cancer. Finally, some views will be discussed concerning the toxicity and clinical translation of iron oxide NPs and the future outlook of NP development to facilitate multiple therapies in a single formulation for cancer theranostics.
Collapse
Affiliation(s)
- Richard A. Revia
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | - Miqin Zhang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
45
|
Rokka J, Snellman A, Kaasalainen M, Salonen J, Zona C, La Ferla B, Nicotra F, Re F, Masserini M, Forsback S, Lopez-Picon F, Rinne JO, Haaparanta-Solin M, Solin O. (18)F-labeling syntheses and preclinical evaluation of functionalized nanoliposomes for Alzheimer's disease. Eur J Pharm Sci 2016; 88:257-66. [PMID: 26993963 DOI: 10.1016/j.ejps.2016.03.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 02/17/2016] [Accepted: 03/14/2016] [Indexed: 01/06/2023]
Abstract
The aim of the present study was to synthesize functionalized (18)F-labeled NLs ((18)F-NLs) and evaluate their biological behavior in mouse models of Alzheimer's disease (AD) using positron emission tomography (PET) and ex vivo brain autoradiography. (18)F-fluorine was introduced to (18)F-NLs either by using a core forming (18)F-lipid or by encapsulating a (18)F-tracer, (18)F-treg-curcumin inside the NLs. Phosphatidic acid (PA) and curcumin derivative (Curc) functionalized (18)F-NLs with or without additional mApoE functionalization were produced using thin film hydration. The biodistribution and β-amyloid plaque-binding ability of (18)F-NLs were studied in wild type mice and AD mouse models using in vivo PET imaging and ex vivo brain autoradiography at 60min after (18)F-NL injection. Functionalized (18)F-NLs were successfully synthesized. The preclinical evaluation in mice showed that the functional group affected the biodistribution of (18)F-NLs. Further functionalization with mApoE increased the brain-to-blood ratio of (18)F-NLs but the overall brain uptake remained low with all functionalized (18)F-NLs. The liposomal encapsulation of (18)F-treg-curcumin was not successful and preclinical results of encapsulated (18)F-treg-curcumin and plain (18)F-treg-curcumin were identical. Although the studied functionalized (18)F-NLs were not suitable for PET imaging as such, the synthesis techniques introduced in this study can be utilized to modify the biological behavior of (18)F-labeled NLs.
Collapse
Affiliation(s)
- Johanna Rokka
- Turku PET Centre, Radiopharmaceutical Chemistry Laboratory, University of Turku, Turku, Finland.
| | - Anniina Snellman
- Turku PET Centre, Preclinical Imaging, University of Turku, Turku, Finland; MediCity Research Laboratory, University of Turku, Turku, Finland
| | | | - Jarno Salonen
- Laboratory of Industrial Physics, University of Turku, Finland
| | - Cristiano Zona
- Department of Biotechnology and Bioscience, University of Milano-Bicocca, Milano, Italy
| | - Barbara La Ferla
- Department of Biotechnology and Bioscience, University of Milano-Bicocca, Milano, Italy
| | - Francesco Nicotra
- Department of Biotechnology and Bioscience, University of Milano-Bicocca, Milano, Italy
| | - Francesca Re
- Department of Health Science, University of Milano-Bicocca, Monza, Italy
| | - Massimo Masserini
- Department of Health Science, University of Milano-Bicocca, Monza, Italy
| | - Sarita Forsback
- Turku PET Centre, Radiopharmaceutical Chemistry Laboratory, University of Turku, Turku, Finland
| | - Francisco Lopez-Picon
- Turku PET Centre, Preclinical Imaging, University of Turku, Turku, Finland; MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Juha O Rinne
- Turku PET Centre, University of Turku, Turku, Finland; Division of Clinical Neurosciences, Turku University Hospital, Turku, Finland
| | - Merja Haaparanta-Solin
- Turku PET Centre, Preclinical Imaging, University of Turku, Turku, Finland; MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Olof Solin
- Turku PET Centre, Radiopharmaceutical Chemistry Laboratory, University of Turku, Turku, Finland; Turku PET Centre, Accelerator Laboratory, Åbo Akademi University, Turku, Finland; Department of Chemistry, University of Turku, Turku, Finland
| |
Collapse
|
46
|
Sillerud LO. Quantitative [Fe]MRI of PSMA-targeted SPIONs specifically discriminates among prostate tumor cell types based on their PSMA expression levels. Int J Nanomedicine 2016; 11:357-71. [PMID: 26855574 PMCID: PMC4725637 DOI: 10.2147/ijn.s93409] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
We report the development, experimental verification, and application of a general theory called [Fe]MRI (pronounced fem-ree) for the non-invasive, quantitative molecular magnetic resonance imaging (MRI) of added magnetic nanoparticles or other magnetic contrast agents in biological tissues and other sites. [Fe]MRI can easily be implemented on any MRI instrument, requiring only measurements of the background nuclear magnetic relaxation times (T1, T2) of the tissue of interest, injection of the magnetic particles, and the subsequent acquisition of a pair of T1-weighted and T2-weighted images. These images, converted into contrast images, are subtracted to yield a contrast difference image proportional to the absolute nanoparticle, iron concentration, ([Fe]) image. [Fe]MRI was validated with the samples of superparamagnetic iron oxide nanoparticles (SPIONs) both in agarose gels and bound to human prostate tumor cells. The [Fe]MRI measurement of the binding of anti-prostate specific membrane antigen (PSMA) conjugated SPIONs to PSMA-positive LNCaP and PSMA-negative DU145 cells in vitro allowed a facile discrimination among prostate tumor cell types based on their PSMA expression level. The low [Fe] detection limit of ~2 μM for SPIONs allows sensitive MRI of added iron at concentrations considerably below the US Food and Drug Administration’s human iron dosage guidelines (<90 μM, 5 mg/kg).
Collapse
Affiliation(s)
- Laurel O Sillerud
- BRaIN Center, Department of Neurology, University of New Mexico School of Medicine, Albuquerque, NM, USA
| |
Collapse
|
47
|
Kalidasan V, Liu XL, Herng TS, Yang Y, Ding J. Bovine Serum Albumin-Conjugated Ferrimagnetic Iron Oxide Nanoparticles to Enhance the Biocompatibility and Magnetic Hyperthermia Performance. NANO-MICRO LETTERS 2016; 8:80-93. [PMID: 30464997 PMCID: PMC6223930 DOI: 10.1007/s40820-015-0065-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 09/14/2015] [Indexed: 05/09/2023]
Abstract
ABSTRACT Magnetic hyperthermia is a fast emerging, non-invasive cancer treatment method which is used synergistically with the existing cancer therapeutics. We have attempted to address the current challenges in clinical magnetic hyperthermia-improved biocompatibility and enhanced heating characteristics, through a single combinatorial approach. Both superparamagnetic iron oxide nanoparticles (SPIONs) of size 10 nm and ferrimagnetic iron oxide nanoparticles (FIONs) of size 30 nm were synthesized by thermal decomposition method for comparison studies. Two different surface modifying agents, viz, Cetyl Trimethyl Ammonium Bromide and 3-Aminopropyltrimethoxysilane, were used to conjugate Bovine Serum Albumin (BSA) over the iron oxide nanoparticles via two different methods-surface charge adsorption and covalent amide bonding, respectively. The preliminary haemolysis and cell viability experiments show that BSA conjugation mitigates the haemolytic effect of the iron oxide nanoparticles on erythrocytes and is non-cytotoxic to the healthy Baby Hamster Kidney cells. It was observed from the results that due to better colloidal stability, the SAR value of the BSA-iron oxide nanoparticles is higher than the iron oxide nanoparticles without BSA, irrespective of the size of the iron oxide nanoparticles and method of conjugation. The BSA-FIONs seem to show improved biocompatibility, as the haemolytic index is less than 2 % and cell viability is up to 120 %, when normalized with the control. The SAR value of BSA-FIONs is 2300 W g-1 when compared to 1700 W g-1 of FIONs without BSA conjugation. Thus, we report here that BSA conjugation over FIONs (with a high saturation magnetization of 87 emu g-1) provide a single combinatorial approach to improve the biocompatibility and enhance the SAR value for magnetic hyperthermia, thus addressing both the current challenges of the same.
Collapse
Affiliation(s)
- Viveka Kalidasan
- Department of Materials Science and Engineering, Faculty of Engineering, National University of Singapore, 7 Engineering Drive 1, Singapore, 117574 Singapore
| | - Xiao Li Liu
- Department of Materials Science and Engineering, Faculty of Engineering, National University of Singapore, 7 Engineering Drive 1, Singapore, 117574 Singapore
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, Xi’an, 710069 Shaanxi People’s Republic of China
| | - Tun Seng Herng
- Department of Materials Science and Engineering, Faculty of Engineering, National University of Singapore, 7 Engineering Drive 1, Singapore, 117574 Singapore
| | - Yong Yang
- Department of Materials Science and Engineering, Faculty of Engineering, National University of Singapore, 7 Engineering Drive 1, Singapore, 117574 Singapore
| | - Jun Ding
- Department of Materials Science and Engineering, Faculty of Engineering, National University of Singapore, 7 Engineering Drive 1, Singapore, 117574 Singapore
| |
Collapse
|
48
|
Paik T, Chacko AM, Mikitsh JL, Friedberg JS, Pryma DA, Murray CB. Shape-Controlled Synthesis of Isotopic Yttrium-90-Labeled Rare Earth Fluoride Nanocrystals for Multimodal Imaging. ACS NANO 2015; 9:8718-8728. [PMID: 26257288 DOI: 10.1021/acsnano.5b03355] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Isotopically labeled nanomaterials have recently attracted much attention in biomedical research, environmental health studies, and clinical medicine because radioactive probes allow the elucidation of in vitro and in vivo cellular transport mechanisms, as well as the unambiguous distribution and localization of nanomaterials in vivo. In addition, nanocrystal-based inorganic materials have a unique capability of customizing size, shape, and composition; with the potential to be designed as multimodal imaging probes. Size and shape of nanocrystals can directly influence interactions with biological systems, hence it is important to develop synthetic methods to design radiolabeled nanocrystals with precise control of size and shape. Here, we report size- and shape-controlled synthesis of rare earth fluoride nanocrystals doped with the β-emitting radioisotope yttrium-90 ((90)Y). Size and shape of nanocrystals are tailored via tight control of reaction parameters and the type of rare earth hosts (e.g., Gd or Y) employed. Radiolabeled nanocrystals are synthesized in high radiochemical yield and purity as well as excellent radiolabel stability in the face of surface modification with different polymeric ligands. We demonstrate the Cerenkov radioluminescence imaging and magnetic resonance imaging capabilities of (90)Y-doped GdF3 nanoplates, which offer unique opportunities as a promising platform for multimodal imaging and targeted therapy.
Collapse
Affiliation(s)
- Taejong Paik
- Department of Chemistry, ‡Division of Nuclear Medicine and Clinical Molecular Imaging, Department of Radiology, §Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, ∥Division of Thoracic Surgery, and ⊥Department of Materials Science and Engineering, University of Pennsylvania , Philadelphia, Pennsylvania 19104, United States
| | - Ann-Marie Chacko
- Department of Chemistry, ‡Division of Nuclear Medicine and Clinical Molecular Imaging, Department of Radiology, §Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, ∥Division of Thoracic Surgery, and ⊥Department of Materials Science and Engineering, University of Pennsylvania , Philadelphia, Pennsylvania 19104, United States
| | - John L Mikitsh
- Department of Chemistry, ‡Division of Nuclear Medicine and Clinical Molecular Imaging, Department of Radiology, §Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, ∥Division of Thoracic Surgery, and ⊥Department of Materials Science and Engineering, University of Pennsylvania , Philadelphia, Pennsylvania 19104, United States
| | - Joseph S Friedberg
- Department of Chemistry, ‡Division of Nuclear Medicine and Clinical Molecular Imaging, Department of Radiology, §Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, ∥Division of Thoracic Surgery, and ⊥Department of Materials Science and Engineering, University of Pennsylvania , Philadelphia, Pennsylvania 19104, United States
| | - Daniel A Pryma
- Department of Chemistry, ‡Division of Nuclear Medicine and Clinical Molecular Imaging, Department of Radiology, §Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, ∥Division of Thoracic Surgery, and ⊥Department of Materials Science and Engineering, University of Pennsylvania , Philadelphia, Pennsylvania 19104, United States
| | - Christopher B Murray
- Department of Chemistry, ‡Division of Nuclear Medicine and Clinical Molecular Imaging, Department of Radiology, §Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, ∥Division of Thoracic Surgery, and ⊥Department of Materials Science and Engineering, University of Pennsylvania , Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
49
|
Abstract
Theranostics is a promising field that combines therapeutics and diagnostics into single multifunctional formulations. This field is driven by advancements in nanoparticle systems capable of providing the necessary functionalities. By utilizing these powerful nanomedicines, the concept of personalized medicine can be realized by tailoring treatment strategies to the individual. This review gives a brief overview of the components of a theranostic system and the challenges that designing truly multifunctional nanoparticles present. Considerations when choosing a class of nanoparticle include the size, shape, charge, and surface chemistry, while classes of nanoparticles discussed are polymers, liposomes, dendrimers, and polymeric micelles. Targeting to disease states can be achieved either through passive or active targeting which uses specific ligands to target receptors that are overexpressed in tumors and common targeting elements are presented. To image the interactions with disease states, contrast agents are included in the nanoparticle formulation. Imaging options include optical imaging techniques, computed tomography, nuclear based, and magnetic resonance imaging. The interplay between all of these components needs to be carefully considered when designing a theranostic system.
Collapse
|
50
|
Burke BP, Baghdadi N, Clemente GS, Camus N, Guillou A, Kownacka AE, Domarkas J, Halime Z, Tripier R, Archibald SJ. Final step gallium-68 radiolabelling of silica-coated iron oxide nanorods as potential PET/MR multimodal imaging agents. Faraday Discuss 2015; 175:59-71. [PMID: 25325197 DOI: 10.1039/c4fd00137k] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The investigation of iron oxide-based positron emission tomography/magnetic resonance (PET/MR) multimodal imaging agents is an expanding field in which a variety of nanoparticle sizes, shapes, surface coatings and radioisotopes are open for exploration. This study develops iron oxide nanorods which are coated with various mixtures of poly(ethylene glycol) (PEG) and macrocyclic ligand (DO3A) via the formation of a silica layer on the surface. Gallium-68 radiolabelling of the nanorods was carried out in high radiochemical yields (RCY) and their stability in human serum was demonstrated for all constructs, even in the absence of the macrocyclic chelating unit. Further studies were carried out in an attempt to determine the appropriate amount of PEG coating to give optimal properties for future in vivo studies.
Collapse
Affiliation(s)
- Benjamin P Burke
- Department of Chemistry, University of Hull, Cottingham Road, Hull, HU6 7RX, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|