1
|
Low HY, Yang CT, Xia B, He T, Lam WWC, Ng DCE. Radiolabeled Liposomes for Nuclear Imaging Probes. Molecules 2023; 28:molecules28093798. [PMID: 37175207 PMCID: PMC10180453 DOI: 10.3390/molecules28093798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/17/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
Quantitative nuclear imaging techniques are in high demand for various disease diagnostics and cancer theranostics. The non-invasive imaging modality requires radiotracing through the radioactive decay emission of the radionuclide. Current preclinical and clinical radiotracers, so-called nuclear imaging probes, are radioisotope-labeled small molecules. Liposomal radiotracers have been rapidly developing as novel nuclear imaging probes. The physicochemical properties and structural characteristics of liposomes have been elucidated to address their long circulation and stability as radiopharmaceuticals. Various radiolabeling methods for synthesizing radionuclides onto liposomes and synthesis strategies have been summarized to render them biocompatible and enable specific targeting. Through a variety of radionuclide labeling methods, radiolabeled liposomes for use as nuclear imaging probes can be obtained for in vivo biodistribution and specific targeting studies. The advantages of radiolabeled liposomes including their use as potential clinical nuclear imaging probes have been highlighted. This review is a comprehensive overview of all recently published liposomal SPECT and PET imaging probes.
Collapse
Affiliation(s)
- Ho Ying Low
- Department of Nuclear Medicine and Molecular Imaging, Radiological Sciences Division, Singapore General Hospital, Outram Road, Singapore 169608, Singapore
| | - Chang-Tong Yang
- Department of Nuclear Medicine and Molecular Imaging, Radiological Sciences Division, Singapore General Hospital, Outram Road, Singapore 169608, Singapore
- Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Bin Xia
- School of Chemistry and Chemical Engineering, Hefei University of Technology, Hefei 230009, China
| | - Tao He
- School of Chemistry and Chemical Engineering, Hefei University of Technology, Hefei 230009, China
| | - Winnie Wing Chuen Lam
- Department of Nuclear Medicine and Molecular Imaging, Radiological Sciences Division, Singapore General Hospital, Outram Road, Singapore 169608, Singapore
- Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - David Chee Eng Ng
- Department of Nuclear Medicine and Molecular Imaging, Radiological Sciences Division, Singapore General Hospital, Outram Road, Singapore 169608, Singapore
- Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| |
Collapse
|
2
|
Bordeianu G, Filip N, Cernomaz A, Veliceasa B, Hurjui LL, Pinzariu AC, Pertea M, Clim A, Marinca MV, Serban IL. The Usefulness of Nanotechnology in Improving the Prognosis of Lung Cancer. Biomedicines 2023; 11:biomedicines11030705. [PMID: 36979684 PMCID: PMC10045176 DOI: 10.3390/biomedicines11030705] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/20/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
Lung cancer remains a major public health problem both in terms of incidence and specific mortality despite recent developments in terms of prevention, such as smoking reduction policies and clinical management advances. Better lung cancer prognosis could be achieved by early and accurate diagnosis and improved therapeutic interventions. Nanotechnology is a dynamic and fast-developing field; various medical applications have been developed and deployed, and more exist as proofs of concepts or experimental models. We aim to summarize current knowledge relevant to the use of nanotechnology in lung cancer management. Starting from the chemical structure-based classification of nanoparticles, we identify and review various practical implementations roughly organized as diagnostic or therapeutic in scope, ranging from innovative contrast agents to targeted drug carriers. Available data are presented starting with standards of practice and moving to highly experimental methods and proofs of concept; particularities, advantages, limits and future directions are explored, focusing on the potential impact on lung cancer clinical prognosis.
Collapse
Affiliation(s)
- Gabriela Bordeianu
- Department of Morpho-Functional Sciences (II), Discipline of Biochemistry, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Nina Filip
- Department of Morpho-Functional Sciences (II), Discipline of Biochemistry, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Correspondence: (N.F.); (A.C.)
| | - Andrei Cernomaz
- III-rd Medical Department, Discipline of Pneumology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Correspondence: (N.F.); (A.C.)
| | - Bogdan Veliceasa
- Department of Orthopedics and Traumatology, Surgical Science (II), Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Loredana Liliana Hurjui
- Department of Morpho-Functional Sciences (II), Discipline of Physiology, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Alin Constantin Pinzariu
- Department of Morpho-Functional Sciences (II), Discipline of Physiology, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Mihaela Pertea
- Department of Plastic Surgery and Reconstructive Microsurgery, “Sf. Spiridon” Emergency County Hospital, 700111 Iasi, Romania
| | - Andreea Clim
- Department of Morpho-Functional Sciences (II), Discipline of Physiology, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Mihai Vasile Marinca
- III-rd Medical Department, Discipline of Oncology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Ionela Lacramioara Serban
- Department of Morpho-Functional Sciences (II), Discipline of Physiology, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| |
Collapse
|
3
|
Vitale G, Carra S, Alessi Y, Campolo F, Pandozzi C, Zanata I, Colao A, Faggiano A. Carcinoid Syndrome: Preclinical Models and Future Therapeutic Strategies. Int J Mol Sci 2023; 24:ijms24043610. [PMID: 36835022 PMCID: PMC9961914 DOI: 10.3390/ijms24043610] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/27/2023] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
Carcinoid syndrome represents a debilitating paraneoplastic disease, caused by the secretion of several substances, occurring in about 10-40% of patients with well-differentiated neuroendocrine tumors (NETs). The main signs and symptoms associated with carcinoid syndrome are flushing, diarrhea, hypotension, tachycardia, bronchoconstriction, venous telangiectasia, dyspnea and fibrotic complications (mesenteric and retroperitoneal fibrosis, and carcinoid heart disease). Although there are several drugs available for the treatment of carcinoid syndrome, the lack of therapeutic response, poor tolerance or resistance to drugs are often reported. Preclinical models are indispensable tools for investigating the pathogenesis, mechanisms for tumor progression and new therapeutic approaches for cancer. This paper provides a state-of-the-art overview of in vitro and in vivo models in NETs with carcinoid syndrome, highlighting the future developments and therapeutic approaches in this field.
Collapse
Affiliation(s)
- Giovanni Vitale
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20122 Milan, Italy
- Laboratory of Geriatric and Oncologic Neuroendocrinology Research, IRCCS, Istituto Auxologico Italiano, 20100 Milan, Italy
- Correspondence: ; Tel.: +39-02-6191-12023; Fax: +39-02-6191-13033
| | - Silvia Carra
- Laboratory of Endocrine and Metabolic Research, IRCCS, Istituto Auxologico Italiano, 20100 Milan, Italy
| | - Ylenia Alessi
- Endocrine Unit, University Hospital “Gaetano Martino” of Messina, 98125 Messina, Italy
| | - Federica Campolo
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Carla Pandozzi
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Isabella Zanata
- Section of Endocrinology and Internal Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Annamaria Colao
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80138 Naples, Italy
| | - Antongiulio Faggiano
- Endocrinology Unit, Department of Clinical and Molecular Medicine, Sant’Andrea Hospital, ENETS Center of Excellence, Sapienza University of Rome, 00189 Rome, Italy
| | | |
Collapse
|
4
|
Horváth D, Vágner A, Szikra D, Trencsényi G, Demitri N, Guidolin N, Maiocchi A, Ghiani S, Travagin F, Giovenzana GB, Baranyai Z. Boosting Bismuth(III) Complexation for Targeted α-Therapy (TAT) Applications with the Mesocyclic Chelating Agent AAZTA. Angew Chem Int Ed Engl 2022; 61:e202207120. [PMID: 36073561 PMCID: PMC9828418 DOI: 10.1002/anie.202207120] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Indexed: 01/12/2023]
Abstract
Targeted α therapy (TAT) is a promising tool in the therapy of cancer. The radionuclide 213 BiIII shows favourable physical properties for this application, but the fast and stable chelation of this metal ion remains challenging. Herein, we demonstrate that the mesocyclic chelator AAZTA quickly coordinates BiIII at room temperature, leading to a robust complex. A comprehensive study of the structural, thermodynamic and kinetic properties of [Bi(AAZTA)]- is reported, along with bifunctional [Bi(AAZTA-C4-COO- )]2- and the targeted agent [Bi(AAZTA-C4-TATE)]- , which incorporates the SSR agonist Tyr3 -octreotate. An unexpected increase in the stability and kinetic inertness of the metal chelate was observed for the bifunctional derivative and was maintained for the peptide conjugate. A cyclotron-produced 205/206 Bi mixture was used as a model of 213 Bi in labelling, stability, and biodistribution experiments, allowing the efficiency of [213 Bi(AAZTA-C4-TATE)]- to be estimated. High accumulation in AR42J tumours and reduced kidney uptake were observed with respect to the macrocyclic chelate [213 Bi(DOTA-TATE)]- .
Collapse
Affiliation(s)
- Dávid Horváth
- Department of Physical ChemistryUniversity of DebrecenEgyetem tér 14010DebrecenHungary
| | | | - Dezsö Szikra
- Scanomed Ltd.Nagyerdei Krt. 984032DebrecenHungary,Medical Imaging ClinicUniversity of DebrecenNagyerdei krt. 984032DebrecenHungary
| | - György Trencsényi
- Scanomed Ltd.Nagyerdei Krt. 984032DebrecenHungary,Medical Imaging ClinicUniversity of DebrecenNagyerdei krt. 984032DebrecenHungary
| | - Nicola Demitri
- Elettra-Sincrotrone TriesteS.S. 14 Km 163.5 in Area Science Park34149Basovizza (TS)Italy
| | - Nicol Guidolin
- Bracco Imaging SpaBracco Research CentreVia Ribes 510010Colleretto Giacosa (TO)Italy
| | - Alessandro Maiocchi
- Bracco Imaging SpaBracco Research CentreVia Ribes 510010Colleretto Giacosa (TO)Italy
| | - Simona Ghiani
- Bracco Imaging SpaBracco Research CentreVia Ribes 510010Colleretto Giacosa (TO)Italy
| | - Fabio Travagin
- Dipartimento di Scienze del FarmacoUniversità del Piemonte OrientaleLargo Donegani 2/328100NovaraItaly
| | - Giovanni B. Giovenzana
- Dipartimento di Scienze del FarmacoUniversità del Piemonte OrientaleLargo Donegani 2/328100NovaraItaly
| | - Zsolt Baranyai
- Bracco Imaging SpaBracco Research CentreVia Ribes 510010Colleretto Giacosa (TO)Italy
| |
Collapse
|
5
|
Horváth D, Vágner A, Szikra D, Trencsényi G, Demitri N, Guidolin N, Maiocchi A, Ghiani S, Travagin F, Giovenzana GB, Baranyai Z. Boosting Bismuth(III) Complexation for Targeted α‐Therapy (TAT) Applications with the Mesocyclic Chelating Agent AAZTA. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202207120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Dávid Horváth
- University of Debrecen Department of Physical Chemistry HUNGARY
| | | | | | | | - Nicola Demitri
- Elettra Sincrotrone Trieste SCpA Elettra Sincrotrone Trieste SCpA ITALY
| | | | | | | | - Fabio Travagin
- Universita degli Studi del Piemonte Orientale Amedeo Avogadro Dipartimento di Scienze del Farmaco ITALY
| | - Giovanni Battista Giovenzana
- Università degli Studi del Piemonte Orientale Amedeo Avogadro Facoltà di Farmacia: Universita degli Studi del Piemonte Orientale Amedeo Avogadro Dipartimento di Scienze del Farmaco Dipartimento di Scienze del Farmaco Largo Donegani 2/3Via Bovio 6 28100 Novara ITALY
| | | |
Collapse
|
6
|
Tuguntaev RG, Hussain A, Fu C, Chen H, Tao Y, Huang Y, Liu L, Liang XJ, Guo W. Bioimaging guided pharmaceutical evaluations of nanomedicines for clinical translations. J Nanobiotechnology 2022; 20:236. [PMID: 35590412 PMCID: PMC9118863 DOI: 10.1186/s12951-022-01451-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 05/05/2022] [Indexed: 11/25/2022] Open
Abstract
Nanomedicines (NMs) have emerged as an efficient approach for developing novel treatment strategies against a variety of diseases. Over the past few decades, NM formulations have received great attention, and a large number of studies have been performed in this field. Despite this, only about 60 nano-formulations have received industrial acceptance and are currently available for clinical use. Their in vivo pharmaceutical behavior is considered one of the main challenges and hurdles for the effective clinical translation of NMs, because it is difficult to monitor the pharmaceutic fate of NMs in the biological environment using conventional pharmaceutical evaluations. In this context, non-invasive imaging modalities offer attractive solutions, providing the direct monitoring and quantification of the pharmacokinetic and pharmacodynamic behavior of labeled NMs in a real-time manner. Imaging evaluations have great potential for revealing the relationship between the physicochemical properties of NMs and their pharmaceutical profiles in living subjects. In this review, we introduced imaging techniques that can be used for in vivo NM evaluations. We also provided an overview of various studies on the influence of key parameters on the in vivo pharmaceutical behavior of NMs that had been visualized in a non-invasive and real-time manner.
Collapse
Affiliation(s)
- Ruslan G Tuguntaev
- Department of Minimally Invasive Interventional Radiology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, People's Republic of China
| | - Abid Hussain
- Advanced Research Institute of Multidisciplinary Science, School of Life Science, School of Medical Technology (Institute of Engineering Medicine), Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecular Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Chenxing Fu
- Department of Minimally Invasive Interventional Radiology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, People's Republic of China
| | - Haoting Chen
- Department of Minimally Invasive Interventional Radiology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, People's Republic of China
| | - Ying Tao
- Department of Minimally Invasive Interventional Radiology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, People's Republic of China
| | - Yan Huang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Lu Liu
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, People's Republic of China.
| | - Xing-Jie Liang
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, People's Republic of China.
| | - Weisheng Guo
- Department of Minimally Invasive Interventional Radiology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, People's Republic of China.
| |
Collapse
|
7
|
Wang R, Zhang Z, Liu B, Xue J, Liu F, Tang T, Liu W, Feng F, Qu W. Strategies for the design of nanoparticles: starting with long-circulating nanoparticles, from lab to clinic. Biomater Sci 2021; 9:3621-3637. [PMID: 34008587 DOI: 10.1039/d0bm02221g] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Short half-life is one of the main causes of drug attrition in clinical development, which also leads to the failure of many leading compounds and hits to become drug candidates. Nowadays, nanomaterials have been applied to drug development to address this problem. In fact, the clinical application of nanoparticles (NPs) is severely limited due to their rapid elimination by the reticuloendothelial system (RES) in vivo. In this paper, we aim to summarize representative strategies on prolonging the circulation time for bridging the gap between excellent pharmaceutics and proper half-life and encourage clinical translation.
Collapse
Affiliation(s)
- Ruyi Wang
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| | - Zhongtao Zhang
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| | - Bowen Liu
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| | - Jingwei Xue
- The Joint Laboratory of China Pharmaceutical University and Taian City Central Hospital, Taian City Central Hospital, Taian, 271000, China and Taian City institute of Digestive Disease, Taian City Central Hospital, Taian, 271000, China
| | - Fulei Liu
- The Joint Laboratory of China Pharmaceutical University and Taian City Central Hospital, Taian City Central Hospital, Taian, 271000, China and Pharmaceutical Department, Taian City Central Hospital, Taian, 271000, China
| | - Tongzhong Tang
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| | - Wenyuan Liu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 210009, China and Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, 210009, China
| | - Feng Feng
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, People's Republic of China. and Jiangsu Food and Pharmaceutical Science College, Huaian, 223003, China.
| | - Wei Qu
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| |
Collapse
|
8
|
Lima PHCD, Butera AP, Cabeça LF, Ribeiro-Viana RM. Liposome surface modification by phospholipid chemical reactions. Chem Phys Lipids 2021; 237:105084. [PMID: 33891960 DOI: 10.1016/j.chemphyslip.2021.105084] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/17/2021] [Accepted: 04/16/2021] [Indexed: 12/13/2022]
Abstract
Liposomal systems are well known for playing an important role as drug carriers, presenting several therapeutic applications in different sectors, such as in drug delivery, diagnosis, and in many other academic areas. A novel class of this nanoparticle is the actively target liposome, which is constructed with the surface modified with appropriated molecules (or ligands) to actively bind a target molecule of certain cells, system, or tissue. There are many ways to functionalize these nanostructures, from non-covalent adsorption to covalent bond formation. In this review, we focus on the strategies of modifying liposomes by glycerophospholipid covalent chemical reaction. The approach used in this text summarizes the main reactions and strategies used in phospholipid modification that can be carried out by chemists and researchers from other areas. The knowledge of these methodologies is of great importance for planning new studies using this material and also for manipulating its properties.
Collapse
Affiliation(s)
- Pedro Henrique Correia de Lima
- Programa de Pós-graduação em Ciências e Engenharia de Materiais (PPGCEM-UTFPR), Universidade Tecnológica Federal do Paraná, UTFPR-Ld, CEP 86036-370, Londrina, PR, Brazil
| | - Anna Paola Butera
- Departamento de Química, Universidade Estadual de Londrina, UEL, CEP 86051-980, Londrina, PR, Brazil
| | - Luis Fernando Cabeça
- Programa de Pós-graduação em Ciências e Engenharia de Materiais (PPGCEM-UTFPR), Universidade Tecnológica Federal do Paraná, UTFPR-Ld, CEP 86036-370, Londrina, PR, Brazil; Departamento Acadêmico de Química, Universidade Tecnológica Federal do Paraná, UTFPR-Ld, CEP 86036-370, Londrina, PR, Brazil
| | - Renato Márcio Ribeiro-Viana
- Programa de Pós-graduação em Ciências e Engenharia de Materiais (PPGCEM-UTFPR), Universidade Tecnológica Federal do Paraná, UTFPR-Ld, CEP 86036-370, Londrina, PR, Brazil; Departamento Acadêmico de Química, Universidade Tecnológica Federal do Paraná, UTFPR-Ld, CEP 86036-370, Londrina, PR, Brazil.
| |
Collapse
|
9
|
Pellico J, Gawne PJ, T M de Rosales R. Radiolabelling of nanomaterials for medical imaging and therapy. Chem Soc Rev 2021; 50:3355-3423. [PMID: 33491714 DOI: 10.1039/d0cs00384k] [Citation(s) in RCA: 112] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Nanomaterials offer unique physical, chemical and biological properties of interest for medical imaging and therapy. Over the last two decades, there has been an increasing effort to translate nanomaterial-based medicinal products (so-called nanomedicines) into clinical practice and, although multiple nanoparticle-based formulations are clinically available, there is still a disparity between the number of pre-clinical products and those that reach clinical approval. To facilitate the efficient clinical translation of nanomedicinal-drugs, it is important to study their whole-body biodistribution and pharmacokinetics from the early stages of their development. Integrating this knowledge with that of their therapeutic profile and/or toxicity should provide a powerful combination to efficiently inform nanomedicine trials and allow early selection of the most promising candidates. In this context, radiolabelling nanomaterials allows whole-body and non-invasive in vivo tracking by the sensitive clinical imaging techniques positron emission tomography (PET), and single photon emission computed tomography (SPECT). Furthermore, certain radionuclides with specific nuclear emissions can elicit therapeutic effects by themselves, leading to radionuclide-based therapy. To ensure robust information during the development of nanomaterials for PET/SPECT imaging and/or radionuclide therapy, selection of the most appropriate radiolabelling method and knowledge of its limitations are critical. Different radiolabelling strategies are available depending on the type of material, the radionuclide and/or the final application. In this review we describe the different radiolabelling strategies currently available, with a critical vision over their advantages and disadvantages. The final aim is to review the most relevant and up-to-date knowledge available in this field, and support the efficient clinical translation of future nanomedicinal products for in vivo imaging and/or therapy.
Collapse
Affiliation(s)
- Juan Pellico
- School of Biomedical Engineering & Imaging Sciences, King's College London, St. Thomas' Hospital, London SE1 7EH, UK.
| | | | | |
Collapse
|
10
|
Henriksen JR, Engel TB, Petersen AL, Kempen PJ, Melander F, Roos P, Jølck RI, Andresen TL. Elucidating the anomalous membrane permeability of Ag(I), Cu(II), Zn(II) and Au(III) towards new nanoreactor strategies for synthesizing metal nanoparticles. NANOSCALE 2020; 12:22298-22306. [PMID: 33146209 DOI: 10.1039/d0nr04655h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The main structural element defining the cell is the lipid membrane, which is an integral part of regulating the fluxes of ion and nutrition molecules in and out of the cell. Surprisingly, copper ions were found to have anomalous membrane permeability. This led us to consider a broader spectrum of cations and further a new approach for using liposomes as nanoreactors for synthesis of metal and metal alloy nanoparticles. In the present study, the high membrane permeability of Cu2+ and its neighbouring transition elements in the periodic table was investigated. The permeability of Ni2+, Cu2+, Zn2+, Ag+, Au3+, Mg2+, Ca2+ and Lu3+ was assessed, and we report that Zn2+, Cu2+, Ag+ and Au3+ surprisingly are able to cross lipid bilayers. This knowledge is highly relevant for understanding trafficking of cations in biological systems, as well as for design of novel nanoparticle and nanoreactor systems. An example of its use is presented as a platform for synthesizing single highly uniform gold nanoparticles inside liposomal nanoreactors. We envision that this approach could provide a new nanoreactor methodology for forming highly structurally constrained uniform metal and metal alloy nanoparticles, as well as new methods for in vivo tracking of liposomes.
Collapse
Affiliation(s)
- Jonas R Henriksen
- Department of Health Technology, Technical University of Denmark, Building 423, DK-2800 Lyngby, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Capriotti G, Varani M, Lauri C, Franchi G, Pizzichini P, Signore A. Copper-64 labeled nanoparticles for positron emission tomography imaging: a review of the recent literature. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF RADIOPHARMACEUTICAL CHEMISTRY AND BIOLOGY 2020; 64:346-355. [PMID: 33073558 DOI: 10.23736/s1824-4785.20.03315-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Nuclear medicine plays a crucial role for personalized therapy, mainly in oncology. Chemotherapy and radiotherapy present some disadvantages and research is shifting toward nanotechnology with significant improvements in therapy and diagnosis of several cancers. Indeed, nanoparticles can be tagged with different radioisotopes for single photon emission computed tomography (SPECT) and positron emission tomography (PET) imaging and for therapy. This review describes the current state of the art of 64Copper-labeled nanoparticles for PET imaging of cancer. EVIDENCE ACQUISITION We performed a systematic analysis of literature using the terms "64CuCl<inf>2</inf>," "64Cu," "Copper" AND "nanoparticle" AND "PET" in online databases: i.e. PubMed/MEDLINE and Scopus. The search was limited to English papers and original articles. We excluded articles not in English language, abstracts, case reports, review articles and meeting presentations. EVIDENCE SYNTHESIS Amongst the 116 articles retrieved, 88 were excluded because reviews, or not in English, or only in-vitro studies or meeting presentations. We considered only 28 original papers. The most used nanoparticles are liposomes and they are mainly used in breast cancer although other animal models of cancer have been also investigated. CONCLUSIONS The results showed that nanoparticles can be considered a promising radiopharmaceutical for PET imaging of different type of cancer.
Collapse
Affiliation(s)
- Gabriela Capriotti
- Unit of Nuclear Medicine, Department of Medical-Surgical Sciences and Translational Medicine, Sapienza University, Rome, Italy - .,Unit of Nuclear Medicine, Sant'Andrea University Hospital, Rome, Italy -
| | - Michela Varani
- Unit of Nuclear Medicine, Department of Medical-Surgical Sciences and Translational Medicine, Sapienza University, Rome, Italy
| | - Chiara Lauri
- Unit of Nuclear Medicine, Department of Medical-Surgical Sciences and Translational Medicine, Sapienza University, Rome, Italy
| | - Gabriele Franchi
- Unit of Nuclear Medicine, Sant'Andrea University Hospital, Rome, Italy
| | | | - Alberto Signore
- Unit of Nuclear Medicine, Department of Medical-Surgical Sciences and Translational Medicine, Sapienza University, Rome, Italy.,Unit of Nuclear Medicine, Sant'Andrea University Hospital, Rome, Italy
| |
Collapse
|
12
|
Clausen AS, Østergaard DE, Holmberg P, Henriksen JR, Tham J, Damborg PP, Jensen AI, Kjaer A, Hansen AE, Andresen TL. Quantitative determination of 64Cu-liposome accumulation at inflammatory and infectious sites: Potential for future theranostic system. J Control Release 2020; 327:737-746. [PMID: 32920081 DOI: 10.1016/j.jconrel.2020.09.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/14/2020] [Accepted: 09/08/2020] [Indexed: 10/23/2022]
Affiliation(s)
- Anne Skovsbo Clausen
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Department of Biomedical Sciences, Rigshospitalet and University of Copenhagen, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - Daniella Elisabet Østergaard
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Department of Biomedical Sciences, Rigshospitalet and University of Copenhagen, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - Petter Holmberg
- Department of Translational Medicine, Clinical Infection Medicine, Lund University, Skåne University Hospital, Ruth Lundskogs gata 3, SE-214 28 Malmö, Sweden
| | - Jonas Rosager Henriksen
- Department of Health Technology, Section for Biotherapeutic Engineering and Drug Targeting, Technical University of Denmark, Anker Engelundsvej 1, DK-2800 Kgs. Lyngby, Denmark
| | - Johan Tham
- Department of Translational Medicine, Clinical Infection Medicine, Lund University, Skåne University Hospital, Ruth Lundskogs gata 3, SE-214 28 Malmö, Sweden
| | - Peter Panduro Damborg
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Bülowsvej 7, DK-1870 Frederiksberg, Denmark
| | - Andreas I Jensen
- Department of Health Technology, The Hevesy Laboratory, Technical University of Denmark, Frederiksborgvej 399, DK-4000 Roskilde, Denmark
| | - Andreas Kjaer
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Department of Biomedical Sciences, Rigshospitalet and University of Copenhagen, Blegdamsvej 9, DK-2100 Copenhagen, Denmark.
| | - Anders Elias Hansen
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Department of Biomedical Sciences, Rigshospitalet and University of Copenhagen, Blegdamsvej 9, DK-2100 Copenhagen, Denmark; Department of Health Technology, Section for Biotherapeutic Engineering and Drug Targeting, Technical University of Denmark, Anker Engelundsvej 1, DK-2800 Kgs. Lyngby, Denmark
| | - Thomas Lars Andresen
- Department of Health Technology, Section for Biotherapeutic Engineering and Drug Targeting, Technical University of Denmark, Anker Engelundsvej 1, DK-2800 Kgs. Lyngby, Denmark
| |
Collapse
|
13
|
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: 7.5] [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
|
14
|
d'Angelo M, Castelli V, Benedetti E, Antonosante A, Catanesi M, Dominguez-Benot R, Pitari G, Ippoliti R, Cimini A. Theranostic Nanomedicine for Malignant Gliomas. Front Bioeng Biotechnol 2019; 7:325. [PMID: 31799246 PMCID: PMC6868071 DOI: 10.3389/fbioe.2019.00325] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 10/28/2019] [Indexed: 12/14/2022] Open
Abstract
Brain tumors mainly originate from glial cells and are classified as gliomas. Malignant gliomas represent an incurable disease; indeed, after surgery and chemotherapy, recurrence appears within a few months, and mortality has remained high in the last decades. This is mainly due to the heterogeneity of malignant gliomas, indicating that a single therapy is not effective for all patients. In this regard, the advent of theranostic nanomedicine, a combination of imaging and therapeutic agents, represents a strategic tool for the management of malignant brain tumors, allowing for the detection of therapies that are specific to the single patient and avoiding overdosing the non-responders. Here, recent theranostic nanomedicine approaches for glioma therapy are described.
Collapse
Affiliation(s)
- Michele d'Angelo
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Elisabetta Benedetti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Andrea Antonosante
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Mariano Catanesi
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Reyes Dominguez-Benot
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Giuseppina Pitari
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Rodolfo Ippoliti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
- Department of Biology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
15
|
Ge J, Zhang Q, Zeng J, Gu Z, Gao M. Radiolabeling nanomaterials for multimodality imaging: New insights into nuclear medicine and cancer diagnosis. Biomaterials 2019; 228:119553. [PMID: 31689672 DOI: 10.1016/j.biomaterials.2019.119553] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 10/15/2019] [Accepted: 10/15/2019] [Indexed: 12/22/2022]
Abstract
Nuclear medicine imaging has been developed as a powerful diagnostic approach for cancers by detecting gamma rays directly or indirectly from radionuclides to construct images with beneficial characteristics of high sensitivity, infinite penetration depth and quantitative capability. Current nuclear medicine imaging modalities mainly include single-photon emission computed tomography (SPECT) and positron emission tomography (PET) that require administration of radioactive tracers. In recent years, a vast number of radioactive tracers have been designed and constructed to improve nuclear medicine imaging performance toward early and accurate diagnosis of cancers. This review will discuss recent progress of nuclear medicine imaging tracers and associated biomedical imaging applications. Radiolabeling nanomaterials for rational development of tracers will be comprehensively reviewed with highlights on radiolabeling approaches (surface coupling, inner incorporation and interface engineering), providing profound understanding on radiolabeling chemistry and the associated imaging functionalities. The applications of radiolabeled nanomaterials in nuclear medicine imaging-related multimodality imaging will also be summarized with typical paradigms described. Finally, key challenges and new directions for future research will be discussed to guide further advancement and practical use of radiolabeled nanomaterials for imaging of cancers.
Collapse
Affiliation(s)
- Jianxian Ge
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Qianyi Zhang
- School of Chemical Engineering and Australian Centre for NanoMedicine (ACN), University of New South Wales, Sydney, NSW, 2052, Australia
| | - Jianfeng Zeng
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China.
| | - Zi Gu
- School of Chemical Engineering and Australian Centre for NanoMedicine (ACN), University of New South Wales, Sydney, NSW, 2052, Australia.
| | - Mingyuan Gao
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China; Institute of Chemistry, Chinese Academy of Sciences/School of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100190, China
| |
Collapse
|
16
|
Man F, Gawne PJ, T M de Rosales R. Nuclear imaging of liposomal drug delivery systems: A critical review of radiolabelling methods and applications in nanomedicine. Adv Drug Deliv Rev 2019; 143:134-160. [PMID: 31170428 PMCID: PMC6866902 DOI: 10.1016/j.addr.2019.05.012] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/25/2019] [Accepted: 05/29/2019] [Indexed: 12/14/2022]
Abstract
The integration of nuclear imaging with nanomedicine is a powerful tool for efficient development and clinical translation of liposomal drug delivery systems. Furthermore, it may allow highly efficient imaging-guided personalised treatments. In this article, we critically review methods available for radiolabelling liposomes. We discuss the influence that the radiolabelling methods can have on their biodistribution and highlight the often-overlooked possibility of misinterpretation of results due to decomposition in vivo. We stress the need for knowing the biodistribution/pharmacokinetics of both the radiolabelled liposomal components and free radionuclides in order to confidently evaluate the images, as they often share excretion pathways with intact liposomes (e.g. phospholipids, metallic radionuclides) and even show significant tumour uptake by themselves (e.g. some radionuclides). Finally, we describe preclinical and clinical studies using radiolabelled liposomes and discuss their impact in supporting liposomal drug development and clinical translation in several diseases, including personalised nanomedicine approaches.
Collapse
Affiliation(s)
- Francis Man
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London SE1 7EH, United Kingdom
| | - Peter J Gawne
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London SE1 7EH, United Kingdom
| | - Rafael T M de Rosales
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London SE1 7EH, United Kingdom; London Centre for Nanotechnology, King's College London, Strand Campus, London WC2R 2LS, United Kingdom.
| |
Collapse
|
17
|
Tesauro D, Accardo A, Diaferia C, Milano V, Guillon J, Ronga L, Rossi F. Peptide-Based Drug-Delivery Systems in Biotechnological Applications: Recent Advances and Perspectives. Molecules 2019; 24:E351. [PMID: 30669445 PMCID: PMC6359574 DOI: 10.3390/molecules24020351] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 01/09/2019] [Accepted: 01/18/2019] [Indexed: 12/30/2022] Open
Abstract
Peptides of natural and synthetic sources are compounds operating in a wide range of biological interactions. They play a key role in biotechnological applications as both therapeutic and diagnostic tools. They are easily synthesized thanks to solid-phase peptide devices where the amino acid sequence can be exactly selected at molecular levels, by tuning the basic units. Recently, peptides achieved resounding success in drug delivery and in nanomedicine smart applications. These applications are the most significant challenge of recent decades: they can selectively deliver drugs to only pathological tissues whilst saving the other districts of the body. This specific feature allows a reduction in the drug side effects and increases the drug efficacy. In this context, peptide-based aggregates present many advantages, including biocompatibility, high drug loading capacities, chemical diversity, specific targeting, and stimuli responsive drug delivery. A dual behavior is observed: on the one hand they can fulfill a structural and bioactive role. In this review, we focus on the design and the characterization of drug delivery systems using peptide-based carriers; moreover, we will also highlight the peptide ability to self-assemble and to actively address nanosystems toward specific targets.
Collapse
Affiliation(s)
- Diego Tesauro
- Department of Pharmacy and CIRPeB, Università Federico II, 80134 Naples, Italy.
| | - Antonella Accardo
- Department of Pharmacy and CIRPeB, Università Federico II, 80134 Naples, Italy.
| | - Carlo Diaferia
- Department of Pharmacy and CIRPeB, Università Federico II, 80134 Naples, Italy.
| | - Vittoria Milano
- Department of Pharmacy and CIRPeB, Università Federico II, 80134 Naples, Italy.
- ARNA, INSERM U1212/UMR CNRS 5320, UFR des Sciences Pharmaceutiques, Université de Bordeaux, F-33000 Bordeaux, France.
| | - Jean Guillon
- ARNA, INSERM U1212/UMR CNRS 5320, UFR des Sciences Pharmaceutiques, Université de Bordeaux, F-33000 Bordeaux, France.
| | - Luisa Ronga
- Institute of Analytical Sciences, IPREM, UMR 5254, CNRS-University of Pau, 64000 Pau, France.
| | - Filomena Rossi
- Department of Pharmacy and CIRPeB, Università Federico II, 80134 Naples, Italy.
| |
Collapse
|
18
|
Hervella P, Dam JH, Thisgaard H, Baun C, Olsen BB, Høilund-Carlsen PF, Needham D. Chelation, formulation, encapsulation, retention, and in vivo biodistribution of hydrophobic nanoparticles labelled with 57Co-porphyrin: Oleylamine ensures stable chelation of cobalt in nanoparticles that accumulate in tumors. J Control Release 2018; 291:11-25. [PMID: 30291986 DOI: 10.1016/j.jconrel.2018.09.027] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 07/02/2018] [Accepted: 09/30/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND MOTIVATION While small molecules can be used in cancer diagnosis there is a need for imageable diagnostic NanoParticles (NPs) that act as surrogates for the therapeutic NPs. Many NPs are composed of hydrophobic materials so the challenge is to formulate hydrophobic imaging agents. To develop individualized medical treatments based on NP, a first step should be the selection of patients who are likely responders to the treatment as judged by imaging tumor accumulation of NPs. This requires NPs with the same size and structure as the subsequent therapeutic NPs but labelled with a long-lived radionuclide. Cobalt isotopes are good candidates for NP labelling since 55Co has half-life of 17.5 h and positron energy of 570 keV while 57Co (t1/2 271.6 d) is an isotope suited for preclinical single photon emission tomography (SPECT) to visualize biodistribution and pharmacokinetics of NPs. We used the hydrophobic octaethyl porphyrin (OEP) to chelate cobalt and to encapsulate it inside hydrophobic liquid NPs (LNPs). We hypothesized that at least two additional hydrophobic axial ligands (oleylamine, OA) must be provided to the OEP-Co complex in order to encapsulate and retain Co inside LNP. RESULTS 1. Cobalt chelation by OEP and OA. The association constant of cobalt to OEP was 2.49 × 105 M-1 and the formation of the hexacoordinate complex OEP-Co-4OA was measured by spectroscopy. 2. NP formulation and characterization: LNPs were prepared by the fast ethanol injection method and were composed of a liquid core (triolein) surrounded by a lipid monolayer (DSPC:Cholesterol:DSPE-PEG2000). The size of the LNPs loaded with the cobalt complex was 40 ± 5 nm, 3. Encapsulation of OEP-Co-OA: The loading capacity of OEP-Co-OA in LNP was 5 mol%. 4. Retention of OEP-57Co-4OA complex in the LNPs: the positive effect of the OA ligands was demonstrated on the stability of the OEP-57Co-4OA complex, providing a half-life for retention in PBS of 170 h (7 days) while in the absence of the axial OA ligands was only 22 h. 5 Biodistribution Study: the in vivo biodistribution of LNP was studied in AR42J pancreatic tumor-bearing mice. The estimated half-life of LNPs in blood was about 7.2 h. Remarkably, the accumulation of LNPs in the tumor was as high as 9.4% ID/g 24 h after injection with a doubling time for tumor accumulation of 3.22 h. The most important result was that the nanoparticles could indeed accumulate in the AR42J tumors up to levels greater than those of other NPs previously measured in the same tumor model, and at about half the values reported for the molecular agent 57Co-DOTATATE. CONCLUSIONS The additional hydrophobic chelator OA was indeed needed to obtain a stable octahedral OEP-Co-4OA. Cobalt was actually well-retained inside LNP in the OEP-Co-4OA complex. The method described in the present work for the core-labelling of LNPs with cobalt is now ready for labeling of NPs with 55Co, or indeed other hexadentate radionuclides of interest for preclinical in vivo PET-imaging and radio-therapeutics.
Collapse
Affiliation(s)
- Pablo Hervella
- Center for Single Particle Science and Engineering (SPSE), Institute for Molecular Medicine, Health Sciences, University Southern Denmark, Campusvej 55, Odense DK-5230, Denmark; Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Travesa da Choupana s/n, Santiago de Compostela 15706, Spain.
| | - Johan Hygum Dam
- Department of Nuclear Medicine, Odense University Hospital, Sdr. Boulevard 29, Odense 5000, Denmark
| | - Helge Thisgaard
- Department of Nuclear Medicine, Odense University Hospital, Sdr. Boulevard 29, Odense 5000, Denmark
| | - Christina Baun
- Department of Nuclear Medicine, Odense University Hospital, Sdr. Boulevard 29, Odense 5000, Denmark
| | - Birgitte Brinkmann Olsen
- Department of Nuclear Medicine, Odense University Hospital, Sdr. Boulevard 29, Odense 5000, Denmark
| | | | - David Needham
- Center for Single Particle Science and Engineering (SPSE), Institute for Molecular Medicine, Health Sciences, University Southern Denmark, Campusvej 55, Odense DK-5230, Denmark; Department of Mechanical Engineering and Material Science, Duke University, Durham, NC 27708,USA; School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK.
| |
Collapse
|
19
|
Christensen E, Henriksen JR, Jørgensen JT, Amitay Y, Shmeeda H, Gabizon AA, Kjær A, Andresen TL, Hansen AE. Folate receptor targeting of radiolabeled liposomes reduces intratumoral liposome accumulation in human KB carcinoma xenografts. Int J Nanomedicine 2018; 13:7647-7656. [PMID: 30538449 PMCID: PMC6251465 DOI: 10.2147/ijn.s182579] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background Active, ligand-mediated, targeting of functionalized liposomes to folate receptors (FRs) overexpressed on cancer cells could potentially improve drug delivery and specificity. Studies on folate-targeting liposomes (FTLs) have, however, yielded varying results and generally fail to display a clear benefit of FR targeting. Method Tumor accumulating potential of FTLs and NTLs were investigated in a FR overex-pressing xenograft model by positron emission tomography/computed tomography imaging. Results Tumors displayed significantly lower activity of FTLs than NTLs. Furthermore, FTLs displayed worse circulating properties and increased liver-accumulation than NTLs. Conclusion This study underlines that long-circulating properties of liposomes must be achieved to take advantage of EPR-dependent tumor accumulation which may be lost by functionalization. FR-functionalization negatively affected both tumor accumulation and circulation properties.
Collapse
Affiliation(s)
- Esben Christensen
- Department of Micro- and Nanotechnology, DTU Nanotech, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark, .,Center for Nanomedicine and Theranostics, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark, .,Cluster for Molecular Imaging, Department of Biomedical Sciences and Department of Clinical Physiology, Nuclear Medicine & PET, University of Copenhagen and Rigshospitalet, DK-2200 & DK-2100, Copenhagen, Denmark,
| | - Jonas R Henriksen
- Center for Nanomedicine and Theranostics, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark, .,Department of Chemistry, DTU Chemistry, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark
| | - Jesper T Jørgensen
- Cluster for Molecular Imaging, Department of Biomedical Sciences and Department of Clinical Physiology, Nuclear Medicine & PET, University of Copenhagen and Rigshospitalet, DK-2200 & DK-2100, Copenhagen, Denmark,
| | - Yasmine Amitay
- Shaare Zedek Medical Center and Hebrew University - School of Medicine, Jerusalem, Israel
| | - Hilary Shmeeda
- Shaare Zedek Medical Center and Hebrew University - School of Medicine, Jerusalem, Israel
| | - Alberto A Gabizon
- Shaare Zedek Medical Center and Hebrew University - School of Medicine, Jerusalem, Israel
| | - Andreas Kjær
- Cluster for Molecular Imaging, Department of Biomedical Sciences and Department of Clinical Physiology, Nuclear Medicine & PET, University of Copenhagen and Rigshospitalet, DK-2200 & DK-2100, Copenhagen, Denmark,
| | - Thomas L Andresen
- Department of Micro- and Nanotechnology, DTU Nanotech, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark, .,Center for Nanomedicine and Theranostics, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark,
| | - Anders E Hansen
- Department of Micro- and Nanotechnology, DTU Nanotech, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark, .,Center for Nanomedicine and Theranostics, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark, .,Cluster for Molecular Imaging, Department of Biomedical Sciences and Department of Clinical Physiology, Nuclear Medicine & PET, University of Copenhagen and Rigshospitalet, DK-2200 & DK-2100, Copenhagen, Denmark,
| |
Collapse
|
20
|
Gahete MD, Jimenez-Vacas JM, Alors-Perez E, Herrero-Aguayo V, Fuentes-Fayos AC, Pedraza-Arevalo S, Castaño JP, Luque RM. Mouse models in endocrine tumors. J Endocrinol 2018; 240:JOE-18-0571.R1. [PMID: 30475226 DOI: 10.1530/joe-18-0571] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 11/26/2018] [Indexed: 12/14/2022]
Abstract
Endocrine and neuroendocrine tumors comprise a highly heterogeneous group of neoplasms that can arise from (neuro)endocrine cells, either from endocrine glands or from the widespread diffuse neuroendocrine system, and, consequently, are widely distributed throughout the body. Due to their diversity, heterogeneity and limited incidence, studying in detail the molecular and genetic alterations that underlie their development and progression is still a highly elusive task. This, in turn, hinders the discovery of novel therapeutic options for these tumors. To circumvent these limitations, numerous mouse models of endocrine and neuroendocrine tumors have been developed, characterized and used in pre-clinical, co-clinical (implemented in mouse models and patients simultaneously) and post-clinical studies, for they represent powerful and necessary tools in basic and translational tumor biology research. Indeed, different in vivo mouse models, including cell line-based xenografts (CDXs), patient-derived xenografts (PDXs) and genetically engineered mouse models (GEMs), have been used to delineate the development, progression and behavior of human tumors. Results gained with these in vivo models have facilitated the clinical application in patients of diverse breakthrough discoveries made in this field. Herein, we review the generation, characterization and translatability of the most prominent mouse models of endocrine and neuroendocrine tumors reported to date, as well as the most relevant clinical implications obtained for each endocrine and neuroendocrine tumor type.
Collapse
Affiliation(s)
- Manuel D Gahete
- M Gahete, Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, 14011, Spain
| | - Juan M Jimenez-Vacas
- J Jimenez-Vacas, Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, Spain
| | - Emilia Alors-Perez
- E Alors-Perez, Department of Cell Biology, Physiology and Inmunology, Maimonides Institute for Biomedical Research of Cordoba (IMIBIC) / University of Cordoba, Cordoba, Spain
| | - Vicente Herrero-Aguayo
- V Herrero-Aguayo, Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, Spain
| | - Antonio C Fuentes-Fayos
- A Fuentes-Fayos, Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, Spain
| | - Sergio Pedraza-Arevalo
- S Pedraza-Arevalo, Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, Spain
| | - Justo P Castaño
- J Castaño, Dpt. of Cell Biology-University of Córdoba, IMIBIC-Maimonides Biomedical Research Institute of Cordoba, Cordoba, E-14004, Spain
| | - Raul M Luque
- R Luque, Dept of Cell Biology, Phisiology and Inmunology, Section of Cell Biology, University of Cordoba, Cordoba, Spain, Cordoba, 14014, Spain
| |
Collapse
|
21
|
Farzin L, Sheibani S, Moassesi ME, Shamsipur M. An overview of nanoscale radionuclides and radiolabeled nanomaterials commonly used for nuclear molecular imaging and therapeutic functions. J Biomed Mater Res A 2018; 107:251-285. [PMID: 30358098 DOI: 10.1002/jbm.a.36550] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 08/08/2018] [Accepted: 09/03/2018] [Indexed: 02/06/2023]
Abstract
Recent advances in the field of nanotechnology applications in nuclear medicine offer the promise of better diagnostic and therapeutic options. In recent years, increasing efforts have been focused on developing nanoconstructs that can be used as core platforms for attaching medical radionuclides with different strategies for the purposes of molecular imaging and targeted drug delivery. This review article presents an introduction to some commonly used nanomaterials with zero-dimensional, one-dimensional, two-dimensional, and three-dimensional structures, describes the various methods applied to radiolabeling of nanomaterials, and provides illustrative examples of application of the nanoscale radionuclides or radiolabeled nanocarriers in nuclear nanomedicine. Especially, the passive and active nanotargeting delivery of radionuclides with illustrating examples for tumor imaging and therapy was reviewed and summarized. The accurate and early diagnosis of cancer can lead to increased survival rates for different types of this disease. Although, the conventional single-modality diagnostic methods such as positron emission tomography/single photon emission computed tomography or MRI used for such purposes are powerful means; most of these are limited by sensitivity or resolution. By integrating complementary signal reporters into a single nanoparticulate contrast agent, multimodal molecular imaging can be performed as scalable images with high sensitivity, resolution, and specificity. The advent of radiolabeled nanocarriers or radioisotope-loaded nanomaterials with magnetic, plasmonic, or fluorescent properties has stimulated growing interest in the developing multimodality imaging probes. These new developments in nuclear nanomedicine are expected to introduce a paradigm shift in multimodal molecular imaging and thereby opening up an era of new diagnostic medical imaging agents. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 251-285, 2019.
Collapse
Affiliation(s)
- Leila Farzin
- Radiation Application Research School, Nuclear Science and Technology Research Institute, Tehran, Iran
| | - Shahab Sheibani
- Radiation Application Research School, Nuclear Science and Technology Research Institute, Tehran, Iran
| | - Mohammad Esmaeil Moassesi
- Radiation Application Research School, Nuclear Science and Technology Research Institute, Tehran, Iran
| | | |
Collapse
|
22
|
Kleynhans J, Grobler AF, Ebenhan T, Sathekge MM, Zeevaart JR. Radiopharmaceutical enhancement by drug delivery systems: A review. J Control Release 2018; 287:177-193. [DOI: 10.1016/j.jconrel.2018.08.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 08/02/2018] [Accepted: 08/03/2018] [Indexed: 12/17/2022]
|
23
|
Ahmedova A, Todorov B, Burdzhiev N, Goze C. Copper radiopharmaceuticals for theranostic applications. Eur J Med Chem 2018; 157:1406-1425. [DOI: 10.1016/j.ejmech.2018.08.051] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 08/15/2018] [Accepted: 08/18/2018] [Indexed: 12/12/2022]
|
24
|
Paramonov VM, Desai D, Kettiger H, Mamaeva V, Rosenholm JM, Sahlgren C, Rivero-Müller A. Targeting Somatostatin Receptors By Functionalized Mesoporous Silica Nanoparticles - Are We Striking Home? Nanotheranostics 2018; 2:320-346. [PMID: 30148051 PMCID: PMC6107779 DOI: 10.7150/ntno.23826] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 06/25/2018] [Indexed: 12/02/2022] Open
Abstract
The concept of delivering nanoformulations to desired tissues by means of targeting membrane receptors of high local abundance by ligands anchored to the nanocarrier has gained a lot of attention over the last decade. Currently, there is no unanimous opinion on whether surface functionalization of nanocarriers by targeting ligands translates into any real benefit in terms of pharmacokinetics or treatment outcomes. Having examined the published nanocarriers designed to engage with somatostatin receptors, we realized that in the majority of cases targetability claims were not supported by solid evidence of targeting ligand-targeted receptor coupling, which is the very crux of a targetability concept. Here, we present an approach to characterize targetability of mesoporous silica-based nanocarriers functionalized with ligands of somatostatin receptors. The targetability proof in our case comes from a functional assay based on a genetically-encoded cAMP probe, which allows for real-time capture of receptor activation in living cells, triggered by targeting ligands on nanoparticles. We elaborate on the development and validation of the assay, highlighting the power of proper functional tests in the characterization pipeline of targeted nanoformulations.
Collapse
Affiliation(s)
- Valeriy M Paramonov
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Finland.,Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Finland.,Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Finland
| | - Diti Desai
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Finland
| | - Helene Kettiger
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Finland
| | - Veronika Mamaeva
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Finland.,Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Finland
| | - Jessica M Rosenholm
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Finland
| | - Cecilia Sahlgren
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Finland.,Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Finland.,Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Adolfo Rivero-Müller
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Finland.,Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Finland.,Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Finland.,Department of Biochemistry and Molecular Biology, Medical University of Lublin, Poland
| |
Collapse
|
25
|
Jensen AI, Severin GW, Hansen AE, Fliedner FP, Eliasen R, Parhamifar L, Kjær A, Andresen TL, Henriksen JR. Remote-loading of liposomes with manganese-52 and in vivo evaluation of the stabilities of 52Mn-DOTA and 64Cu-DOTA using radiolabelled liposomes and PET imaging. J Control Release 2018; 269:100-109. [DOI: 10.1016/j.jconrel.2017.11.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Revised: 10/25/2017] [Accepted: 11/04/2017] [Indexed: 10/18/2022]
|
26
|
Dearling JL, Packard AB. Molecular imaging in nanomedicine – A developmental tool and a clinical necessity. J Control Release 2017. [DOI: 10.1016/j.jconrel.2017.06.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
27
|
North AJ, Karas JA, Ma MT, Blower PJ, Ackermann U, White JM, Donnelly PS. Rhenium and Technetium-oxo Complexes with Thioamide Derivatives of Pyridylhydrazine Bifunctional Chelators Conjugated to the Tumour Targeting Peptides Octreotate and Cyclic-RGDfK. Inorg Chem 2017; 56:9725-9741. [PMID: 28766938 PMCID: PMC5569669 DOI: 10.1021/acs.inorgchem.7b01247] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Indexed: 11/29/2022]
Abstract
This research aimed to develop new tumor targeted theranostic agents taking advantage of the similarities in coordination chemistry between technetium and rhenium. A γ-emitting radioactive isotope of technetium is commonly used in diagnostic imaging, and there are two β- emitting radioactive isotopes of rhenium that have the potential to be of use in radiotherapy. Variants of the 6-hydrazinonicotinamide (HYNIC) bifunctional ligands have been prepared by appending thioamide functional groups to 6-hydrazinonicotinamide to form pyridylthiosemicarbazide ligands (SHYNIC). The new bidentate ligands were conjugated to the tumor targeting peptides Tyr3-octreotate and cyclic-RGD. The new ligands and conjugates were used to prepare well-defined {M═O}3+ complexes (where M = 99mTc or natRe or 188Re) that feature two targeting peptides attached to the single metal ion. These new SHYNIC ligands are capable of forming well-defined rhenium and technetium complexes and offer the possibility of using the 99mTc imaging and 188/186Re therapeutic matched pairs.
Collapse
Affiliation(s)
- Andrea J. North
- The School of Chemistry
and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, 3010, Victorria, Australia
| | - John A. Karas
- The School of Chemistry
and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, 3010, Victorria, Australia
| | - Michelle T. Ma
- Division of Imaging Sciences and Biomedical Engineering, King’s College London, Fourth Floor Lambeth Wing, St Thomas’ Hospital, London SE1 7EH, U.K.
| | - Philip J. Blower
- Division of Imaging Sciences and Biomedical Engineering, King’s College London, Fourth Floor Lambeth Wing, St Thomas’ Hospital, London SE1 7EH, U.K.
| | - Uwe Ackermann
- Department of Molecular Imaging and Therapy, Department
of Medicine, University of Melbourne, Austin Health, Studley Road, Heidelberg, Victoria 3010, Australia
| | - Jonathan M. White
- The School of Chemistry
and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, 3010, Victorria, Australia
| | - Paul S. Donnelly
- The School of Chemistry
and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, 3010, Victorria, Australia
| |
Collapse
|
28
|
Frellsen AF, Hansen AE, Jølck RI, Kempen PJ, Severin GW, Rasmussen PH, Kjær A, Jensen ATI, Andresen TL. Mouse Positron Emission Tomography Study of the Biodistribution of Gold Nanoparticles with Different Surface Coatings Using Embedded Copper-64. ACS NANO 2016; 10:9887-9898. [PMID: 27754658 DOI: 10.1021/acsnano.6b03144] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
By taking advantage of the ability of 64Cu to bind nonspecifically to gold surfaces, we have developed a methodology to embed this radionuclide inside gold nanoparticles (AuNPs). 64Cu enables the in vivo imaging of AuNPs by positron emission tomography (PET). AuNPs have a multitude of uses within health technology and are useful tools for general nanoparticle research. 64Cu-AuNPs were prepared by incubating AuNP seeds with 64Cu2+, followed by the entrapment of the radionuclide by grafting on a second layer of gold. This resulted in radiolabeling efficiencies of 53 ± 6%. The radiolabel showed excellent stability when incubated with EDTA for 2 days (95% radioactivity retention) and showed no loss of 64Cu when incubated with 50% mouse serum for 2 days. The methodology was chelator-free, removing traditional concerns over chelator instability and altered AuNP properties due to surface modification. Radiolabeled 64Cu-AuNP cores were prepared in biomedically relevant sizes of 20-30 nm and used to investigate the in vivo stability of three different AuNP coatings by PET imaging in a murine xenograft tumor model. We found the longest plasma half-life (T1/2 about 9 h) and tumor accumulation (3.9%ID/g) to result from a polyethylene glycol coating, while faster elimination from the bloodstream was observed with both a Tween 20-stabilized coating and a zwitterionic coating based on a mixture of sulfonic acids and quaternary amines. In the in vivo model, the 64Cu was observed to closely follow the AuNPs for each coating, again attributed to the excellent stability of the radiolabel.
Collapse
Affiliation(s)
- Anders F Frellsen
- The Hevesy Laboratory-Center for Nuclear Technologies, Technical University of Denmark , 4000 Roskilde, Denmark
- DTU Nanotech, Center for Nanomedicine and Theranostics, Technical University of Denmark , 2800 Lyngby, Denmark
| | - Anders E Hansen
- DTU Nanotech, Center for Nanomedicine and Theranostics, Technical University of Denmark , 2800 Lyngby, Denmark
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen , 2100 Copenhagen, Denmark
| | - Rasmus I Jølck
- DTU Nanotech, Center for Nanomedicine and Theranostics, Technical University of Denmark , 2800 Lyngby, Denmark
| | - Paul J Kempen
- DTU Nanotech, Center for Nanomedicine and Theranostics, Technical University of Denmark , 2800 Lyngby, Denmark
| | - Gregory W Severin
- The Hevesy Laboratory-Center for Nuclear Technologies, Technical University of Denmark , 4000 Roskilde, Denmark
| | - Palle H Rasmussen
- The Hevesy Laboratory-Center for Nuclear Technologies, Technical University of Denmark , 4000 Roskilde, Denmark
| | - Andreas Kjær
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen , 2100 Copenhagen, Denmark
| | - Andreas T I Jensen
- The Hevesy Laboratory-Center for Nuclear Technologies, Technical University of Denmark , 4000 Roskilde, Denmark
| | - Thomas L Andresen
- DTU Nanotech, Center for Nanomedicine and Theranostics, Technical University of Denmark , 2800 Lyngby, Denmark
| |
Collapse
|
29
|
Cancer theranostics with ⁶⁴Cu/¹⁷⁷Lu-loaded liposomes. Eur J Nucl Med Mol Imaging 2016; 43:938-940. [PMID: 26743898 DOI: 10.1007/s00259-015-3299-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Accepted: 12/22/2015] [Indexed: 12/24/2022]
|
30
|
Rauscher A, Frindel M, Rajerison H, Gouard S, Maurel C, Barbet J, Faivre-Chauvet A, Mougin-Degraef M. Improvement of the Targeting of Radiolabeled and Functionalized Liposomes with a Two-Step System Using a Bispecific Monoclonal Antibody (Anti-CEA × Anti-DTPA-In). Front Med (Lausanne) 2015; 2:83. [PMID: 26636087 PMCID: PMC4658472 DOI: 10.3389/fmed.2015.00083] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 11/06/2015] [Indexed: 01/16/2023] Open
Abstract
This study proposes liposomes as a new tool for pretargeted radioimmunotherapy (RIT) in solid tumors. Tumor pretargeting is obtained by using a bispecific monoclonal antibody [BsmAb, anti-CEA × anti-DTPA-indium complex (DTPA–In)] and pegylated radioactive liposomes containing a lipid-hapten conjugate (DSPE–PEG–DTPA–In). In this work, the immunospecificity of tumor targeting is demonstrated both in vitro by fluorescence microscopy and in vivo by biodistribution studies.
Collapse
Affiliation(s)
- Aurore Rauscher
- Centre de Recherche en Cancérologie Nantes-Angers (CRCNA), 6299 CNRS, UMR 892 - INSERM, Université de Nantes , Nantes , France ; Nuclear Medicine Department, Institut de Cancérologie de l'Ouest , Saint Herblain , France
| | - Mathieu Frindel
- Centre de Recherche en Cancérologie Nantes-Angers (CRCNA), 6299 CNRS, UMR 892 - INSERM, Université de Nantes , Nantes , France ; Nuclear Medicine Department, University Hospital Nantes , Nantes , France
| | - Holisoa Rajerison
- Centre de Recherche en Cancérologie Nantes-Angers (CRCNA), 6299 CNRS, UMR 892 - INSERM, Université de Nantes , Nantes , France
| | - Sébastien Gouard
- Centre de Recherche en Cancérologie Nantes-Angers (CRCNA), 6299 CNRS, UMR 892 - INSERM, Université de Nantes , Nantes , France
| | - Catherine Maurel
- Centre de Recherche en Cancérologie Nantes-Angers (CRCNA), 6299 CNRS, UMR 892 - INSERM, Université de Nantes , Nantes , France
| | - Jacques Barbet
- Centre de Recherche en Cancérologie Nantes-Angers (CRCNA), 6299 CNRS, UMR 892 - INSERM, Université de Nantes , Nantes , France ; GIP Arronax , Saint Herblain , France
| | - Alain Faivre-Chauvet
- Centre de Recherche en Cancérologie Nantes-Angers (CRCNA), 6299 CNRS, UMR 892 - INSERM, Université de Nantes , Nantes , France ; Nuclear Medicine Department, University Hospital Nantes , Nantes , France
| | - Marie Mougin-Degraef
- Centre de Recherche en Cancérologie Nantes-Angers (CRCNA), 6299 CNRS, UMR 892 - INSERM, Université de Nantes , Nantes , France ; Nuclear Medicine Department, University Hospital Nantes , Nantes , France
| |
Collapse
|
31
|
Henriksen JR, Petersen AL, Hansen AE, Frankær CG, Harris P, Elema DR, Kristensen AT, Kjær A, Andresen TL. Remote Loading of (64)Cu(2+) into Liposomes without the Use of Ion Transport Enhancers. ACS APPLIED MATERIALS & INTERFACES 2015; 7:22796-22806. [PMID: 26426093 DOI: 10.1021/acsami.5b04612] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Due to low ion permeability of lipid bilayers, it has been and still is common practice to use transporter molecules such as ionophores or lipophilic chelators to increase transmembrane diffusion rates and loading efficiencies of radionuclides into liposomes. Here, we report a novel and very simple method for loading the positron emitter (64)Cu(2+) into liposomes, which is important for in vivo positron emission tomography (PET) imaging. By this approach, copper is added to liposomes entrapping a chelator, which causes spontaneous diffusion of copper across the lipid bilayer where it is trapped. Using this method, we achieve highly efficient (64)Cu(2+) loading (>95%), high radionuclide retention (>95%), and favorable loading kinetics, excluding the use of transporter molecule additives. Therefore, clinically relevant activities of 200-400 MBq/patient can be loaded fast (60-75 min) and efficiently into preformed stealth liposomes avoiding subsequent purification steps. We investigate the molecular coordination of entrapped copper using X-ray absorption spectroscopy and demonstrate high adaptability of the loading method to pegylated, nonpegylated, gel- or fluid-like, cholesterol rich or cholesterol depleted, cationic, anionic, and zwitterionic lipid compositions. We demonstrate high in vivo stability of (64)Cu-liposomes in a large canine model observing a blood circulation half-life of 24 h and show a tumor accumulation of 6% ID/g in FaDu xenograft mice using PET imaging. With this work, it is demonstrated that copper ions are capable of crossing a lipid membrane unassisted. This method is highly valuable for characterizing the in vivo performance of liposome-based nanomedicine with great potential in diagnostic imaging applications.
Collapse
Affiliation(s)
- Jonas R Henriksen
- Department of Chemistry, Technical University of Denmark , Building 206, 2800 Lyngby, Denmark
- Center for Nanomedicine and Theranostics, Technical University of Denmark , 2800 Lyngby, Denmark
| | - Anncatrine L Petersen
- Department of Micro- and Nanotechnology, Technical University of Denmark , Building 423, 2800 Lyngby, Denmark
- Center for Nanomedicine and Theranostics, Technical University of Denmark , 2800 Lyngby, Denmark
| | - Anders E Hansen
- Department of Micro- and Nanotechnology, Technical University of Denmark , Building 423, 2800 Lyngby, Denmark
- Department of Clinical Physiology, Nuclear Medicine & PET, Faculty of Health Sciences and Cluster for Molecular Imaging, Rigshospitalet, University of Copenhagen , Blegdamsvej 3, 2200 Copenhagen N, Denmark
- Center for Nanomedicine and Theranostics, Technical University of Denmark , 2800 Lyngby, Denmark
| | - Christian G Frankær
- Department of Chemistry, Technical University of Denmark , Building 206, 2800 Lyngby, Denmark
| | - Pernille Harris
- Department of Chemistry, Technical University of Denmark , Building 206, 2800 Lyngby, Denmark
| | - Dennis R Elema
- DTU Nutech, Hevesy Laboratory, Technical University of Denmark , Frederiksborgvej 399, 4000 Roskilde, Denmark
- Center for Nanomedicine and Theranostics, Technical University of Denmark , 2800 Lyngby, Denmark
| | - Annemarie T Kristensen
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen , Dyrlægevej 16, 1870 Frederiksberg C, Denmark
| | - Andreas Kjær
- Department of Clinical Physiology, Nuclear Medicine & PET, Faculty of Health Sciences and Cluster for Molecular Imaging, Rigshospitalet, University of Copenhagen , Blegdamsvej 3, 2200 Copenhagen N, Denmark
| | - Thomas L Andresen
- Department of Micro- and Nanotechnology, Technical University of Denmark , Building 423, 2800 Lyngby, Denmark
- Center for Nanomedicine and Theranostics, Technical University of Denmark , 2800 Lyngby, Denmark
| |
Collapse
|
32
|
Miura Y, Tsuji AB, Sugyo A, Sudo H, Aoki I, Inubushi M, Yashiro M, Hirakawa K, Cabral H, Nishiyama N, Saga T, Kataoka K. Polymeric Micelle Platform for Multimodal Tomographic Imaging to Detect Scirrhous Gastric Cancer. ACS Biomater Sci Eng 2015; 1:1067-1076. [PMID: 33429548 DOI: 10.1021/acsbiomaterials.5b00142] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Scirrhous gastric cancer (SGC) is a recalcitrant tumor, which is among the most lethal cancers. A critical issue for the improvement of SGC prognosis is the lack of an effective imaging method for accurate detection and diagnosis. Because combined nuclear medicine imaging with magnetic resonance imaging (MRI) has the ability to detect cancer with high sensitivity, and quantitation and spatial resolution, it has potential to overcome the issues with SGC detection. Herein, we designed and synthesized a new block copolymer poly(ethylene glycol)-b-poly(γ-benzyl l-glutamate) linked with a chelator 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA-PEG-b-PBLG) to provide a platform for multimodal tomographic imaging. We then successfully prepared DOTA-functionalized polymeric micelles (DOTA/m) measuring 30 nm in diameter, which is an appropriate size to penetrate deeply into tumors with thick fibrosis, including SGC. 111In-labeled DOTA/m highly accumulated in Colon-26 tumors (mouse colon cancer with hyperpermeability), but also in OCUM-2 M LN tumors (SGC with hypopermeability), clearly depicting both tumors by single photon emission computed tomography (SPECT). Gd-labeled DOTA/m clearly visualized OCUM-2 M LN tumors by MRI with high spatial resolution. Moreover, 111In/Gd-labeled micelles, as well as the mixture of 111In- and Gd-labeled DOTA/m demonstrated the capability of this system for selective multimodal SPECT/MR imaging of SCG. Our findings support 111In/Gd-DOTA-labeled micelles as a clinical translationable modality for multimodal tomographic imaging capable of detecting SGC.
Collapse
Affiliation(s)
- Yutaka Miura
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Atsushi B Tsuji
- Diagnostic Imaging Program, Molecular Imaging Center, National Institute of Radiological Sciences, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Aya Sugyo
- Diagnostic Imaging Program, Molecular Imaging Center, National Institute of Radiological Sciences, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Hitomi Sudo
- Diagnostic Imaging Program, Molecular Imaging Center, National Institute of Radiological Sciences, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Ichio Aoki
- Diagnostic Imaging Program, Molecular Imaging Center, National Institute of Radiological Sciences, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Masayuki Inubushi
- Diagnostic Imaging Program, Molecular Imaging Center, National Institute of Radiological Sciences, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Masakazu Yashiro
- Department of Surgical Oncology, Osaka City University, Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Japan
| | - Kosei Hirakawa
- Department of Surgical Oncology, Osaka City University, Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Japan
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Nobuhiro Nishiyama
- Polymer Chemistry Division, Chemical Resources Laboratory, Tokyo Institute of Technology, R1-11, 4259 Nagatsuta, Midori-ku, Yokohama 226-8503, Japan
| | - Tsuneo Saga
- Diagnostic Imaging Program, Molecular Imaging Center, National Institute of Radiological Sciences, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Kazunori Kataoka
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.,Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.,Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| |
Collapse
|
33
|
Hansen AE, Petersen AL, Henriksen JR, Boerresen B, Rasmussen P, Elema DR, af Rosenschöld PM, Kristensen AT, Kjær A, Andresen TL. Positron Emission Tomography Based Elucidation of the Enhanced Permeability and Retention Effect in Dogs with Cancer Using Copper-64 Liposomes. ACS NANO 2015; 9:6985-6995. [PMID: 26022907 DOI: 10.1021/acsnano.5b01324] [Citation(s) in RCA: 200] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Since the first report of the enhanced permeability and retention (EPR) effect, the research in nanocarrier based antitumor drugs has been intense. The field has been devoted to treatment of cancer by exploiting EPR-based accumulation of nanocarriers in solid tumors, which for many years was considered to be a ubiquitous phenomenon. However, the understanding of differences in the EPR-effect between tumor types, heterogeneities within each patient group, and dependency on tumor development stage in humans is sparse. It is therefore important to enhance our understanding of the EPR-effect in large animals and humans with spontaneously developed cancer. In the present paper, we describe a novel loading method of copper-64 into PEGylated liposomes and use these liposomes to evaluate the EPR-effect in 11 canine cancer patients with spontaneous solid tumors by PET/CT imaging. We thereby provide the first high-resolution analysis of EPR-based tumor accumulation in large animals. We find that the EPR-effect is strong in some tumor types but cannot be considered a general feature of solid malignant tumors since we observed a high degree of accumulation heterogeneity between tumors. Six of seven included carcinomas displayed high uptake levels of liposomes, whereas one of four sarcomas displayed signs of liposome retention. We conclude that nanocarrier-radiotracers could be important in identifying cancer patients that will benefit from nanocarrier-based therapeutics in clinical practice.
Collapse
Affiliation(s)
- Anders E Hansen
- †Center for Nanomedicine and Theranostics, DTU Nanotech, Technical University of Denmark, Building 423, DK-2800 Lyngby, Denmark
| | - Anncatrine L Petersen
- †Center for Nanomedicine and Theranostics, DTU Nanotech, Technical University of Denmark, Building 423, DK-2800 Lyngby, Denmark
| | - Jonas R Henriksen
- †Center for Nanomedicine and Theranostics, DTU Nanotech, Technical University of Denmark, Building 423, DK-2800 Lyngby, Denmark
- §DTU Chemistry, Technical University of Denmark, Building 206, DK-2800 Lyngby, Denmark
| | - Betina Boerresen
- ∥Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Dyrlaegevej 16, DK-1870 Frederiksberg, Denmark
| | - Palle Rasmussen
- †Center for Nanomedicine and Theranostics, DTU Nanotech, Technical University of Denmark, Building 423, DK-2800 Lyngby, Denmark
- ⊥DTU Nutech, Hevesy Laboratory, Technical University of Denmark, Frederiksborgvej 399, DK-4000 Roskilde, Denmark
| | - Dennis R Elema
- †Center for Nanomedicine and Theranostics, DTU Nanotech, Technical University of Denmark, Building 423, DK-2800 Lyngby, Denmark
- ⊥DTU Nutech, Hevesy Laboratory, Technical University of Denmark, Frederiksborgvej 399, DK-4000 Roskilde, Denmark
| | - Per Munck af Rosenschöld
- #Radiation Medicine Research Center, Department of Radiation Oncology, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - Annemarie T Kristensen
- ∥Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Dyrlaegevej 16, DK-1870 Frederiksberg, Denmark
| | | | - Thomas L Andresen
- †Center for Nanomedicine and Theranostics, DTU Nanotech, Technical University of Denmark, Building 423, DK-2800 Lyngby, Denmark
| |
Collapse
|
34
|
Abou DS, Pickett JE, Thorek DLJ. Nuclear molecular imaging with nanoparticles: radiochemistry, applications and translation. Br J Radiol 2015; 88:20150185. [PMID: 26133075 PMCID: PMC4730968 DOI: 10.1259/bjr.20150185] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Molecular imaging provides considerable insight into biological processes for greater understanding of health and disease. Numerous advances in medical physics, chemistry and biology have driven the growth of this field in the past two decades. With exquisite sensitivity, depth of detection and potential for theranostics, radioactive imaging approaches have played a major role in the emergence of molecular imaging. At the same time, developments in materials science, characterization and synthesis have led to explosive progress in the nanoparticle (NP) sciences. NPs are generally defined as particles with a diameter in the nanometre size range. Unique physical, chemical and biological properties arise at this scale, stimulating interest for applications as diverse as energy production and storage, chemical catalysis and electronics. In biomedicine, NPs have generated perhaps the greatest attention. These materials directly interface with life at the subcellular scale of nucleic acids, membranes and proteins. In this review, we will detail the advances made in combining radioactive imaging and NPs. First, we provide an overview of the NP platforms and their properties. This is followed by a look at methods for radiolabelling NPs with gamma-emitting radionuclides for use in single photon emission CT and planar scintigraphy. Next, utilization of positron-emitting radionuclides for positron emission tomography is considered. Finally, recent advances for multimodal nuclear imaging with NPs and efforts for clinical translation and ongoing trials are discussed.
Collapse
Affiliation(s)
- D S Abou
- 1 Division of Nuclear Medicine, Russell H. Morgan Department of Radiology and Radiological Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - J E Pickett
- 1 Division of Nuclear Medicine, Russell H. Morgan Department of Radiology and Radiological Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - D L J Thorek
- 1 Division of Nuclear Medicine, Russell H. Morgan Department of Radiology and Radiological Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,2 Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
35
|
Seo JW, Mahakian LM, Tam S, Qin S, Ingham ES, Meares CF, Ferrara KW. The pharmacokinetics of Zr-89 labeled liposomes over extended periods in a murine tumor model. Nucl Med Biol 2014; 42:155-63. [PMID: 25451215 DOI: 10.1016/j.nucmedbio.2014.09.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Revised: 08/31/2014] [Accepted: 09/02/2014] [Indexed: 01/07/2023]
Abstract
(89)Zr (t1/2=78.4h), a positron-emitting metal, has been exploited for PET studies of antibodies because of its relatively long decay time and facile labeling procedures. Here, we used (89)Zr to evaluate the pharmacokinetics of long-circulating liposomes over 168h (1week). We first developed a liposomal-labeling method using p-isothiocyanatobenzyl-desferrioxamine (df-Bz-NCS) and df-PEG1k-DSPE. Df-Bz-NCS was conjugated to 1mol% amino- and amino-PEG2k-DSPE, where the 1mol% df-PEG1k-DSPE was incorporated when the liposomes were formulated. Incubation of (89)Zr with df, df-PEG1k, and df-PEG2k liposomes for one hour resulted in greater than 68% decay-corrected yield. The loss of the (89)Zr label from liposomes after incubation in 50% human serum for 48h ranged from ~1 to 3% across the three formulations. Tail vein administration of the three liposomal formulations in NDL tumor-bearing mice showed that the (89)Zr label at the end of the PEG2k brush was retained in the tumor, liver, spleen and whole body for a longer time interval than (89)Zr labels located under the PEG2k brush. The blood clearance rate of all three liposomal formulations was similar. Overall, the results indicate that the location of the (89)Zr label altered the clearance rate of intracellularly-trapped radioactivity and that df-PEG1k-DSPE provides a stable chelation site for liposomal or lipid-based particle studies over extended periods of time.
Collapse
Affiliation(s)
- Jai Woong Seo
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA.
| | - Lisa M Mahakian
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - Sarah Tam
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - Shengping Qin
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - Elizabeth S Ingham
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - Claude F Meares
- Department of Chemistry, University of California, Davis, CA 95616, USA
| | - Katherine W Ferrara
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA.
| |
Collapse
|
36
|
Lee H, Zheng J, Gaddy D, Orcutt KD, Leonard S, Geretti E, Hesterman J, Harwell C, Hoppin J, Jaffray DA, Wickham T, Hendriks BS, Kirpotin D. A gradient-loadable (64)Cu-chelator for quantifying tumor deposition kinetics of nanoliposomal therapeutics by positron emission tomography. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2014; 11:155-65. [PMID: 25200610 DOI: 10.1016/j.nano.2014.08.011] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 08/26/2014] [Accepted: 08/28/2014] [Indexed: 01/13/2023]
Abstract
Effective drug delivery to tumors is a barrier to treatment with nanomedicines. Non-invasively tracking liposome biodistribution and tumor deposition in patients may provide insight into identifying patients that are well-suited for liposomal therapies. We describe a novel gradient-loadable chelator, 4-DEAP-ATSC, for incorporating (64)Cu into liposomal therapeutics for positron emission tomographic (PET). (64)Cu chelated to 4-DEAP-ATSC (>94%) was loaded into PEGylated liposomal doxorubicin (PLD) and HER2-targeted PLD (MM-302) with efficiencies >90%. (64)Cu-MM-302 was stable in human plasma for at least 48h. PET/CT imaging of xenografts injected with (64)Cu-MM-302 revealed biodistribution profiles that were quantitatively consistent with tissue-based analysis, and tumor (64)Cu positively correlated with liposomal drug deposition. This loading technique transforms liposomal therapeutics into theranostics and is currently being applied in a clinical trial (NCT01304797) to non-invasively quantify MM-302 tumor deposition, and evaluate its potential as a prognostic tool for predicting treatment outcome of nanomedicines.
Collapse
Affiliation(s)
- Helen Lee
- Merrimack Pharmaceuticals, Cambridge, MA, USA.
| | - Jinzi Zheng
- STTARR Innovation Centre, Radiation Medicine Program, Princess Margaret Hospital, University Health Network, Toronto, ON, Canada
| | | | | | | | | | | | | | | | - David A Jaffray
- STTARR Innovation Centre, Radiation Medicine Program, Princess Margaret Hospital, University Health Network, Toronto, ON, Canada
| | | | | | | |
Collapse
|
37
|
Dawidczyk CM, Russell LM, Searson PC. Nanomedicines for cancer therapy: state-of-the-art and limitations to pre-clinical studies that hinder future developments. Front Chem 2014; 2:69. [PMID: 25202689 PMCID: PMC4142601 DOI: 10.3389/fchem.2014.00069] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 08/05/2014] [Indexed: 01/31/2023] Open
Abstract
The ability to efficiently deliver a drug or gene to a tumor site is dependent on a wide range of factors including circulation time, interactions with the mononuclear phagocyte system, extravasation from circulation at the tumor site, targeting strategy, release from the delivery vehicle, and uptake in cancer cells. Nanotechnology provides the possibility of creating delivery systems where the design constraints are decoupled, allowing new approaches for reducing the unwanted side effects of systemic delivery, increasing tumor accumulation, and improving efficacy. The physico-chemical properties of nanoparticle-based delivery platforms introduce additional complexity associated with pharmacokinetics, tumor accumulation, and biodistribution. To assess the impact of nanoparticle-based delivery systems, we first review the design strategies and pharmacokinetics of FDA-approved nanomedicines. Next we review nanomedicines under development, summarizing the range of nanoparticle platforms, strategies for targeting, and pharmacokinetics. We show how the lack of uniformity in preclinical trials prevents systematic comparison and hence limits advances in the field.
Collapse
Affiliation(s)
- Charlene M Dawidczyk
- Institute for Nanobiotechnology, Johns Hopkins University Baltimore, MD, USA ; Johns Hopkins Center of Cancer Nanotechnology Excellence, Johns Hopkins University Baltimore, MD, USA ; Department of Materials Science and Engineering, Johns Hopkins University Baltimore, MD, USA
| | - Luisa M Russell
- Institute for Nanobiotechnology, Johns Hopkins University Baltimore, MD, USA ; Johns Hopkins Center of Cancer Nanotechnology Excellence, Johns Hopkins University Baltimore, MD, USA ; Department of Materials Science and Engineering, Johns Hopkins University Baltimore, MD, USA
| | - Peter C Searson
- Institute for Nanobiotechnology, Johns Hopkins University Baltimore, MD, USA ; Johns Hopkins Center of Cancer Nanotechnology Excellence, Johns Hopkins University Baltimore, MD, USA ; Department of Materials Science and Engineering, Johns Hopkins University Baltimore, MD, USA
| |
Collapse
|
38
|
Azhdarinia A, Ghosh S. Nuclear Imaging with Nanoparticles. Nanomedicine (Lond) 2014. [DOI: 10.1201/b17246-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
39
|
Chakravarty R, Hong H, Cai W. Positron emission tomography image-guided drug delivery: current status and future perspectives. Mol Pharm 2014; 11:3777-97. [PMID: 24865108 PMCID: PMC4218872 DOI: 10.1021/mp500173s] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
![]()
Positron
emission tomography (PET) is an important modality in
the field of molecular imaging, which is gradually impacting patient
care by providing safe, fast, and reliable techniques that help to
alter the course of patient care by revealing invasive, de facto procedures
to be unnecessary or rendering them obsolete. Also, PET provides a
key connection between the molecular mechanisms involved in the pathophysiology
of disease and the according targeted therapies. Recently, PET imaging
is also gaining ground in the field of drug delivery. Current drug
delivery research is focused on developing novel drug delivery systems
with emphasis on precise targeting, accurate dose delivery, and minimal
toxicity in order to achieve maximum therapeutic efficacy. At the
intersection between PET imaging and controlled drug delivery, interest
has grown in combining both these paradigms into clinically effective
formulations. PET image-guided drug delivery has great potential to
revolutionize patient care by in vivo assessment
of drug biodistribution and accumulation at the target site and real-time
monitoring of the therapeutic outcome. The expected end point of this
approach is to provide fundamental support for the optimization of
innovative diagnostic and therapeutic strategies that could contribute
to emerging concepts in the field of “personalized medicine”.
This review focuses on the recent developments in PET image-guided
drug delivery and discusses intriguing opportunities for future development.
The preclinical data reported to date are quite promising, and it
is evident that such strategies in cancer management hold promise
for clinically translatable advances that can positively impact the
overall diagnostic and therapeutic processes and result in enhanced
quality of life for cancer patients.
Collapse
Affiliation(s)
- Rubel Chakravarty
- Department of Radiology, University of Wisconsin-Madison , Madison, Wisconsin 53705-2275, United States
| | | | | |
Collapse
|
40
|
Jensen AI, Binderup T, Kumar EK P, Kjær A, Rasmussen PH, Andresen TL. Positron Emission Tomography Based Analysis of Long-Circulating Cross-Linked Triblock Polymeric Micelles in a U87MG Mouse Xenograft Model and Comparison of DOTA and CB-TE2A as Chelators of Copper-64. Biomacromolecules 2014; 15:1625-33. [DOI: 10.1021/bm401871w] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Andreas I. Jensen
- The
Hevesy Laboratory, DTU Nutech, Technical University of Denmark, Roskilde, Denmark
- Department
of Micro- and Nanotechnology, Center for Nanomedicine and Theranostics,
DTU Nanotech, Technical University of Denmark, Building 423, 2800 Lyngby, Denmark
| | - Tina Binderup
- Cluster
for Molecular Imaging and Department of Clinical Physiology, Nuclear
Medicine, and PET, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Pramod Kumar EK
- Department
of Micro- and Nanotechnology, Center for Nanomedicine and Theranostics,
DTU Nanotech, Technical University of Denmark, Building 423, 2800 Lyngby, Denmark
| | - Andreas Kjær
- Cluster
for Molecular Imaging and Department of Clinical Physiology, Nuclear
Medicine, and PET, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Palle H. Rasmussen
- The
Hevesy Laboratory, DTU Nutech, Technical University of Denmark, Roskilde, Denmark
| | - Thomas L. Andresen
- Department
of Micro- and Nanotechnology, Center for Nanomedicine and Theranostics,
DTU Nanotech, Technical University of Denmark, Building 423, 2800 Lyngby, Denmark
| |
Collapse
|
41
|
Accardo A, Aloj L, Aurilio M, Morelli G, Tesauro D. Receptor binding peptides for target-selective delivery of nanoparticles encapsulated drugs. Int J Nanomedicine 2014; 9:1537-57. [PMID: 24741304 PMCID: PMC3970945 DOI: 10.2147/ijn.s53593] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Active targeting by means of drug encapsulated nanoparticles decorated with targeting bioactive moieties represents the next frontier in drug delivery; it reduces drug side effects and increases the therapeutic index. Peptides, based on their chemical and biological properties, could have a prevalent role to direct drug encapsulated nanoparticles, such as liposomes, micelles, or hard nanoparticles, toward the tumor tissues. A considerable number of molecular targets for peptides are either exclusively expressed or overexpressed on both cancer vasculature and cancer cells. They can be classified into three wide categories: integrins; growth factor receptors (GFRs); and G-protein coupled receptors (GPCRs). Therapeutic agents based on nanovectors decorated with peptides targeting membrane receptors belonging to the GPCR family overexpressed by cancer cells are reviewed in this article. The most studied targeting membrane receptors are considered: somatostatin receptors; cholecystokinin receptors; receptors associated with the Bombesin like peptides family; luteinizing hormone-releasing hormone receptors; and neurotensin receptors. Nanovectors of different sizes and shapes (micelles, liposomes, or hard nanoparticles) loaded with doxorubicin or other cytotoxic drugs and externally functionalized with natural or synthetic peptides are able to target the overexpressed receptors and are described based on their formulation and in vitro and in vivo behaviors.
Collapse
Affiliation(s)
- Antonella Accardo
- Centro interuniversitario di Ricerca sui Peptidi Bioattivi (CIRPeB), Department of Pharmacy and Istituto di Biostrutture e Bioimmagini -Consiglio Nazionale delle Ricerche (IBB CNR), University of Naples "Federico II", Napoli, Italy
| | - Luigi Aloj
- Department of Nuclear Medicine, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione "G. Pascale", Napoli, Italy
| | - Michela Aurilio
- Department of Nuclear Medicine, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione "G. Pascale", Napoli, Italy
| | - Giancarlo Morelli
- Centro interuniversitario di Ricerca sui Peptidi Bioattivi (CIRPeB), Department of Pharmacy and Istituto di Biostrutture e Bioimmagini -Consiglio Nazionale delle Ricerche (IBB CNR), University of Naples "Federico II", Napoli, Italy
| | - Diego Tesauro
- Centro interuniversitario di Ricerca sui Peptidi Bioattivi (CIRPeB), Department of Pharmacy and Istituto di Biostrutture e Bioimmagini -Consiglio Nazionale delle Ricerche (IBB CNR), University of Naples "Federico II", Napoli, Italy
| |
Collapse
|
42
|
Theranostic nanoparticles for cancer and cardiovascular applications. Pharm Res 2014; 31:1390-406. [PMID: 24595494 DOI: 10.1007/s11095-013-1277-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 12/31/2013] [Indexed: 01/15/2023]
Abstract
Theranostics have received enormous attentions for individualized diagnosis and treatment in the past few years. Especially, the availability of various nanoplatforms provides great potentials for designing of sophisticated theranostic agents including imaging, targeting and therapeutic functions. Numerous reports have been published on how to construct multifunctional nanoparticles for the targeted diagnosis and therapy simultaneously since the concept of "theranostics". This review presents recent advances of molecular imaging and nanoplatform technology, and their applications in drug discovery and development. Applications of nanoplatform-based theranostics in cancer and cardiovascular diseases will also be covered including diagnosis, assessment of drug biodistribution, and visualization of drug release from nanoparticles, as well as monitoring of therapeutic effects.
Collapse
|
43
|
Abstract
Nuclear imaging techniques that include positron emission tomography (PET) and single-photon computed tomography have found great success in the clinic because of their inherent high sensitivity. Radionuclide imaging is the most popular form of imaging to be used for molecular imaging in oncology. While many types of molecules have been used for radionuclide-based molecular imaging, there has been a great interest in developing newer nanomaterials for use in clinic, especially for cancer diagnosis and treatment. Nanomaterials have unique physical properties which allow them to be used as imaging probes to locate and identify cancerous lesions. Over the past decade, a great number of nanoparticles have been developed for radionuclide imaging of cancer. This chapter reviews the different kinds of nanomaterials, both organic and inorganic, which are currently being researched for as potential agents for nuclear imaging of variety of cancers. Several radiolabeled multifunctional nanocarriers have been extremely successful for the detection of cancer in preclinical models. So far, significant progress has been achieved in nanoparticle structure design, in vitro/in vivo trafficking, and in vivo fate mapping by using PET. There is a great need for the development of newer nanoparticles, which improve active targeting and quantify new biomarkers for early disease detection and possible prevention of cancer.
Collapse
|
44
|
Hendriks BS, Klinz SG, Reynolds JG, Espelin CW, Gaddy DF, Wickham TJ. Impact of tumor HER2/ERBB2 expression level on HER2-targeted liposomal doxorubicin-mediated drug delivery: multiple low-affinity interactions lead to a threshold effect. Mol Cancer Ther 2013; 12:1816-28. [PMID: 23723124 DOI: 10.1158/1535-7163.mct-13-0180] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Numerous targeted nanotherapeutics have been described for potential treatment of solid tumors. Although attention has focused on antigen selection and molecular design of these systems, there has been comparatively little study of how cellular heterogeneity influences interaction of targeted nanoparticles with tumor cells. Antigens, such as HER2/ERBB2, are heterogeneously expressed across different indications, across patients, and within individual tumors. Furthermore, antigen expression in nontarget tissues necessitates optimization of the therapeutic window. Understanding the performance of a given nanoparticle under different regimens of antigen expression has the ability to inform patient selection and clinical development decisions. In this work, HER2-targeted liposomal doxorubicin was used as a model-targeted nanoparticle to quantitatively investigate the effect of HER2 expression levels on delivery of doxorubicin to the nucleus. We find quantitatively greater nuclear doxorubicin delivery with increasing HER2 expression, exhibiting a threshold effect at approximately 2 × 10(5) HER2 receptors/cell. Kinetic modeling indicated that the threshold effect arises from multiple low-affinity interactions between the targeted liposome and HER2. These results support previous data showing little or no uptake into human cardiomyocytes, which express levels of HER2 below the threshold. Finally, these results suggest that HER2-targeted liposomal doxorubicin may effectively target tumors that fall below traditional definitions of HER2-positive tumors, thereby expanding the potential population of patients that might benefit from this agent.
Collapse
Affiliation(s)
- Bart S Hendriks
- Corresponding Author: Bart Hendriks, Merrimack Pharmaceuticals, 1 Kendall Square, Suite B7201, Cambridge, MA 02139.
| | | | | | | | | | | |
Collapse
|
45
|
Sen K, Mandal M. Second generation liposomal cancer therapeutics: transition from laboratory to clinic. Int J Pharm 2013; 448:28-43. [PMID: 23500602 DOI: 10.1016/j.ijpharm.2013.03.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 03/03/2013] [Accepted: 03/06/2013] [Indexed: 01/10/2023]
Abstract
Recent innovations and developments in nanotechnology have revolutionized cancer therapeutics. Engineered nanomaterials are the current workhorses in the emerging field of cancer nano-therapeutics. Lipid vesicles bearing anti-tumor drugs have turned out to be a clinically feasible and promising nano-therapeutic approach to treat cancer. Efficient entrapment of therapeutics, biocompatibility, biodegradability, low systemic toxicity, low immunogenicity and ability to bypass multidrug resistance mechanisms has made liposomes a versatile drug/gene delivery system in cancer chemotherapy. The present review attempts to explore the recent key advances in liposomal research and the vast arsenal of liposomal formulations currently being utilized in treatment and diagnosis of cancer.
Collapse
Affiliation(s)
- Kacoli Sen
- School of Medical Science and Technology, Indian Institute of Technology, Kharagpur 721302, India
| | | |
Collapse
|
46
|
Patent Highlights. Pharm Pat Anal 2013. [DOI: 10.4155/ppa.13.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Snapshot of recent key developments in the patent literature of relevance to the advancement of pharmaceutical and medical R&D
Collapse
|
47
|
Feldborg LN, Jølck RI, Andresen TL. Quantitative Evaluation of Bioorthogonal Chemistries for Surface Functionalization of Nanoparticles. Bioconjug Chem 2012; 23:2444-50. [DOI: 10.1021/bc3005057] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Lise N. Feldborg
- DTU Nanotech,
Department of Micro- and Nanotechnology, Technical University of Denmark, Building 423 2800
Lyngby, Denmark
| | - Rasmus I. Jølck
- DTU Nanotech,
Department of Micro- and Nanotechnology, Technical University of Denmark, Building 423 2800
Lyngby, Denmark
| | - Thomas L. Andresen
- DTU Nanotech,
Department of Micro- and Nanotechnology, Technical University of Denmark, Building 423 2800
Lyngby, Denmark
| |
Collapse
|
48
|
Rangger C, Helbok A, von Guggenberg E, Sosabowski J, Radolf T, Prassl R, Andreae F, Thurner GC, Haubner R, Decristoforo C. Influence of PEGylation and RGD loading on the targeting properties of radiolabeled liposomal nanoparticles. Int J Nanomedicine 2012; 7:5889-900. [PMID: 23226020 PMCID: PMC3512544 DOI: 10.2147/ijn.s36847] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Purpose Liposomes have been proposed to be a means of selectively targeting cancer sites for diagnostic and therapeutic applications. The focus of this work was the evaluation of radiolabeled PEGylated liposomes derivatized with varying amounts of a cyclic arginyl–glycyl–aspartic acid (RGD) peptide. RGD peptides are known to bind to αvβ3 integrin receptors overexpressed during tumor-induced angiogenesis. Methods Several liposomal nanoparticles carrying the RGD peptide targeting sequence (RLPs) were synthesized using a combination of 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine, cholesterol, diethylenetriaminepentaacetic acid-derivatized lipids for radiolabeling, a polyethylene glycol (PEG) building block, and a lipid-based RGD building block. Relative amounts of RGD and PEG building blocks were varied. In vitro binding affinities were determined using isolated αvβ3 integrin receptors incubated with different concentrations of RLPs in competition with iodine-125-labeled cyclo-(-RGDyV-). Binding of the indium-111-labeled RLPs was also evaluated. Biodistribution and micro single photon emission computed tomography/computed tomography imaging studies were performed in nude mice using different tumor xenograft models. Results RLPs were labeled with indium-111 with high radiochemical yields. In vitro binding studies of RLPs with different RGD/PEG loading revealed good binding to isolated receptors, which was dependent on the extent of RGD and PEG loading. Binding increased with higher RGD loading, whereas reduced binding was found with higher PEG loading. Biodistribution showed increased circulating time for PEGylated RLPs, but no dependence on RGD loading. Both biodistribution and micro single photon emission computed tomography/computed tomography imaging studies revealed low, nonspecific tumor uptake values. Conclusion In this study, RLPs for targeting angiogenesis were described. Even though good binding to αvβ3 integrin receptors was found in vitro, the balance between PEGylation and RGD loading clearly requires optimization to achieve targeting in vivo. These data form the basis for future development and provide a platform for the investigation of multimodal approaches.
Collapse
Affiliation(s)
- Christine Rangger
- Department of Nuclear Medicine, Innsbruck Medical University, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Sun H, Benjaminsen RV, Almdal K, Andresen TL. Hyaluronic Acid Immobilized Polyacrylamide Nanoparticle Sensors for CD44 Receptor Targeting and pH Measurement in Cells. Bioconjug Chem 2012; 23:2247-55. [DOI: 10.1021/bc300349n] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Honghao Sun
- Department
of Pharmacy, College
of Bioengineering, Hubei University of Technology, Wuhan 430068, China
| | | | | | | |
Collapse
|
50
|
Ding H, Wu F. Image guided biodistribution and pharmacokinetic studies of theranostics. Am J Cancer Res 2012; 2:1040-53. [PMID: 23227121 PMCID: PMC3516836 DOI: 10.7150/thno.4652] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Accepted: 06/17/2012] [Indexed: 11/05/2022] Open
Abstract
Image guided technique is playing an increasingly important role in the investigation of the biodistribution and pharmacokinetics of drugs or drug delivery systems in various diseases, especially cancers. Besides anatomical imaging modalities such as computed tomography (CT), magnetic resonance imaging (MRI), molecular imaging strategy including optical imaging, positron emission tomography (PET) and single-photon emission computed tomography (SPECT) will facilitate the localization and quantization of radioisotope or optical probe labeled nanoparticle delivery systems in the category of theranostics. The quantitative measurement of the bio-distribution and pharmacokinetics of theranostics in the fields of new drug/probe development, diagnosis and treatment process monitoring as well as tracking the brain-blood-barrier (BBB) breaking through by high sensitive imaging method, and the applications of the representative imaging modalities are summarized in this review.
Collapse
|