1
|
Ahmadi M, Emzhik M, Mosayebnia M. Nanoparticles labeled with gamma-emitting radioisotopes: an attractive approach for in vivo tracking using SPECT imaging. Drug Deliv Transl Res 2023; 13:1546-1583. [PMID: 36811810 DOI: 10.1007/s13346-023-01291-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/03/2023] [Indexed: 02/24/2023]
Abstract
Providing accurate molecular imaging of the body and biological process is critical for diagnosing disease and personalizing treatment with the minimum side effects. Recently, diagnostic radiopharmaceuticals have gained more attention in precise molecular imaging due to their high sensitivity and appropriate tissue penetration depth. The fate of these radiopharmaceuticals throughout the body can be traced using nuclear imaging systems, including single-photon emission computed tomography (SPECT) and positron emission tomography (PET) modalities. In this regard, nanoparticles are attractive platforms for delivering radionuclides into targets because they can directly interfere with the cell membranes and subcellular organelles. Moreover, applying radiolabeled nanomaterials can decrease their toxicity concerns because radiopharmaceuticals are usually administrated at low doses. Therefore, incorporating gamma-emitting radionuclides into nanomaterials can provide imaging probes with valuable additional properties compared to the other carriers. Herein, we aim to review (1) the gamma-emitting radionuclides used for labeling different nanomaterials, (2) the approaches and conditions adopted for their radiolabeling, and (3) their application. This study can help researchers to compare different radiolabeling methods in terms of stability and efficiency and choose the best way for each nanosystem.
Collapse
Affiliation(s)
- Mahnaz Ahmadi
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Marjan Emzhik
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mona Mosayebnia
- Department of Pharmaceutical Chemistry and Radiopharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Niayesh Junction, Vali-E-Asr Ave, Tehran, 14155-6153, Iran.
| |
Collapse
|
2
|
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: 8] [Impact Index Per Article: 4.0] [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
|
3
|
Zhang L, Aragon-Sanabria V, Aditya A, Marelli M, Cao T, Chen F, Yoo B, Ma K, Zhuang L, Cailleau T, Masterson L, Turker MZ, Lee R, DeLeon G, Monette S, Colombo R, Christie RJ, Zanzonico P, Wiesner U, Subramony JA, Bradbury MS. Engineered Ultrasmall Nanoparticle Drug-Immune Conjugates with "Hit and Run" Tumor Delivery to Eradicate Gastric Cancer. ADVANCED THERAPEUTICS 2023; 6:2200209. [PMID: 37007587 PMCID: PMC10061546 DOI: 10.1002/adtp.202370009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Despite advances by recently approved antibody-drug conjugates in treating advanced gastric cancer patients, substantial limitations remain. Here, several key obstacles are overcome by developing a first-in-class ultrasmall (sub-8-nanometer (nm)) anti-human epidermal growth factor receptor 2 (HER2)-targeting drug-immune conjugate nanoparticle therapy. This multivalent fluorescent core-shell silica nanoparticle bears multiple anti-HER2 single-chain variable fragments (scFv), topoisomerase inhibitors, and deferoxamine moieties. Most surprisingly, drawing upon its favorable physicochemical, pharmacokinetic, clearance, and target-specific dual-modality imaging properties in a "hit and run" approach, this conjugate eradicated HER2-expressing gastric tumors without any evidence of tumor regrowth, while exhibiting a wide therapeutic index. Therapeutic response mechanisms are accompanied by the activation of functional markers, as well as pathway-specific inhibition. Results highlight the potential clinical utility of this molecularly engineered particle drug-immune conjugate and underscore the versatility of the base platform as a carrier for conjugating an array of other immune products and payloads.
Collapse
Affiliation(s)
- Li Zhang
- Department of Radiology, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
- MSK-Cornell Center for Translation of Cancer Nanomedicines, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Virginia Aragon-Sanabria
- Department of Radiology, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
- MSK-Cornell Center for Translation of Cancer Nanomedicines, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Anusha Aditya
- Department of Radiology, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
- MSK-Cornell Center for Translation of Cancer Nanomedicines, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Marcello Marelli
- AstraZeneca, One MedImmune Way, Gaithersburg, MD 20878, United States
| | - Tianye Cao
- Department of Radiology, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
- MSK-Cornell Center for Translation of Cancer Nanomedicines, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Feng Chen
- Department of Radiology, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
- MSK-Cornell Center for Translation of Cancer Nanomedicines, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Barney Yoo
- MSK-Cornell Center for Translation of Cancer Nanomedicines, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
- Department of Chemistry, Hunter College, New York, NY 10065, USA
| | - Kai Ma
- MSK-Cornell Center for Translation of Cancer Nanomedicines, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
- Department of Materials Science & Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Li Zhuang
- AstraZeneca, One MedImmune Way, Gaithersburg, MD 20878, United States
| | - Thais Cailleau
- AstraZeneca, Spirogen, QMB Innovation Centre, 42 New Road, London E1 2AX, UK
| | - Luke Masterson
- AstraZeneca, Spirogen, QMB Innovation Centre, 42 New Road, London E1 2AX, UK
| | - Melik Z Turker
- MSK-Cornell Center for Translation of Cancer Nanomedicines, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
- Department of Materials Science & Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Rachel Lee
- MSK-Cornell Center for Translation of Cancer Nanomedicines, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
- Department of Materials Science & Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Gabriel DeLeon
- Department of Radiology, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
- MSK-Cornell Center for Translation of Cancer Nanomedicines, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Sebastien Monette
- Laboratory of Comparative Pathology, Sloan Kettering Institute for Cancer Research, Weill Cornell Medicine, The Rockefeller University, New York, NY 10065, USA
| | - Raffaele Colombo
- AstraZeneca, One MedImmune Way, Gaithersburg, MD 20878, United States
| | - Ronald J Christie
- AstraZeneca, One MedImmune Way, Gaithersburg, MD 20878, United States
| | - Pat Zanzonico
- MSK-Cornell Center for Translation of Cancer Nanomedicines, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
- Department of Medical Physics, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Ulrich Wiesner
- MSK-Cornell Center for Translation of Cancer Nanomedicines, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
- Department of Materials Science & Engineering, Cornell University, Ithaca, NY 14853, USA
- Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY 14853, USA
| | - J Anand Subramony
- AstraZeneca, One MedImmune Way, Gaithersburg, MD 20878, United States
| | - Michelle S Bradbury
- Department of Radiology, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
- MSK-Cornell Center for Translation of Cancer Nanomedicines, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
- Molecular Pharmacology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| |
Collapse
|
4
|
Wang Z, Li J, Lin G, He Z, Wang Y. Metal complex-based liposomes: Applications and prospects in cancer diagnostics and therapeutics. J Control Release 2022; 348:1066-1088. [PMID: 35718211 DOI: 10.1016/j.jconrel.2022.06.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 06/09/2022] [Indexed: 12/17/2022]
Abstract
Metal complexes are of increasing interest as pharmaceutical agents in cancer diagnostics and therapeutics, while some of them suffer from issues such as limited water solubility and severe systemic toxicity. These drawbacks severely hampered their efficacy and clinical applications. Liposomes hold promise as delivery vehicles for constructing metal complex-based liposomes to maximize the therapeutic efficacy and minimize the side effects of metal complexes. This review provides an overview on the latest advances of metal complex-based liposomal delivery systems. First, the development of metal complex-mediated liposomal encapsulation is briefly introduced. Next, applications of metal complex-based liposomes in a variety of fields are overviewed, where drug delivery, cancer imaging (single photon emission computed tomography (SPECT), positron emission tomography (PET), and magnetic resonance imaging (MRI)), and cancer therapy (chemotherapy, phototherapy, and radiotherapy) were involved. Moreover, the potential toxicity, action of toxic mechanisms, immunological effects of metal complexes as well as the advantages of metal complex-liposomes in this content are also discussed. In the end, the future expectations and challenges of metal complex-based liposomes in clinical cancer therapy are tentatively proposed.
Collapse
Affiliation(s)
- Zhaomeng Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Jinbo Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Guimei Lin
- School of Pharmacy, Shandong University, Jinan 250000, PR China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China.
| | - Yongjun Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China.
| |
Collapse
|
5
|
Aragon-Sanabria V, Aditya A, Zhang L, Chen F, Yoo B, Cao T, Madajewski B, Lee R, Turker MZ, Ma K, Monette S, Chen P, Wu J, Ruan S, Overholtzer M, Zanzonico P, Rudin CM, Brennan C, Wiesner U, Zhang L. Ultrasmall Nanoparticle Delivery of Doxorubicin Improves Therapeutic Index for High-Grade Glioma. Clin Cancer Res 2022; 28:2938-2952. [PMID: 35499557 DOI: 10.1158/1078-0432.ccr-21-4053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 01/11/2022] [Accepted: 04/27/2022] [Indexed: 11/16/2022]
Abstract
Abstract
Purpose: Despite dramatic growth in the number of small molecule drugs developed to treat solid tumors, durable therapeutic options to control primary central nervous system malignancies are relatively scarce. Chemotherapeutic agents which appear biologically potent in model systems have often been found to be marginally effective at best when given systemically in clinical trials. This work presents for the first time an ultrasmall (< 8 nm) multimodal core-shell silica nanoparticle, Cornell prime dots (or C' dots), for the efficacious treatment of high-grade gliomas. Experimental Design: This work presents first-in-kind renally-clearable ultrasmall (< 8 nm) multimodal Cornell prime dots (or C' dots) with surface-conjugated doxorubicin via pH-sensitive linkers for the efficacious treatment in two different clinically relevant high-grade glioma models. Results: Optimal drug-per-particle ratios of as-developed nanoparticle-drug conjugates were established and used to obtain favorable pharmacokinetic profiles. The in vivo efficacy results showed significantly improved biological, therapeutic, and toxicological properties over the native drug after intravenous administration in platelet-derived growth factor-driven genetically engineered mouse model, and an epidermal growth factor expressing patient-derived xenograft (EGFR PDX) model. Conclusions: Ultrasmall C' dot-drug conjugates showed great translational potential over doxorubicin for improving the therapeutic outcome of patients with high-grade gliomas, even without a cancer-targeting moiety.
Collapse
Affiliation(s)
| | - Anusha Aditya
- Memorial Sloan Kettering Cancer Center, New York, United States
| | - Li Zhang
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Feng Chen
- Memorial Sloan Kettering Cancer Center, United States
| | | | - Tianye Cao
- Memorial Sloan Kettering Cancer Center, New York, United States
| | - Brian Madajewski
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | | | | | - Kai Ma
- Cornell University, Ithaca, NY, United States
| | - Sebastien Monette
- Memorial Sloan Kettering Cancer Center, The Rockefeller University, Weill Cornell Medicine, New York, New York, United States
| | - Peiming Chen
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Jing Wu
- Hunter College, United States
| | - Shutian Ruan
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | | | - Pat Zanzonico
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Charles M. Rudin
- Memorial Sloan Kettering Cancer Center, New York, New York, United States
| | - Cameron Brennan
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | | | - Li Zhang
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
6
|
Chen X, Niu W, Du Z, Zhang Y, Su D, Gao X. 64Cu radiolabeled nanomaterials for positron emission tomography (PET) imaging. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.02.070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
7
|
Lee SG, Kalidindi TM, Lou H, Gangangari K, Punzalan B, Bitton A, Lee CJ, Vargas HA, Park S, Bodei L, Kharas MG, Singh VK, Kishore Pillarsetty NV, Larson SM. γ-Tocotrienol-Loaded Liposomes for Radioprotection from Hematopoietic Side Effects Caused by Radiotherapeutic Drugs. J Nucl Med 2021; 62:584-590. [PMID: 32826318 PMCID: PMC8049360 DOI: 10.2967/jnumed.120.244681] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 07/03/2020] [Indexed: 11/28/2022] Open
Abstract
With the successful development and increased use of targeted radionuclide therapy for treating cancer comes the increased risk of radiation injury to bone marrow-both direct suppression and stochastic effects, leading to neoplasia. Herein, we report a novel radioprotector drug, a liposomal formulation of γ-tocotrienol (GT3), or GT3-Nano for short, to mitigate bone marrow radiation damage during targeted radionuclide therapy. Methods: GT3 was loaded into liposomes using passive loading. 64Cu-GT3-Nano and 3H-GT3-Nano were synthesized to study the in vivo biodistribution profile of the liposome and GT3 individually. The radioprotection efficacy of GT3-Nano was assessed after acute 137Cs whole-body irradiation at a sublethal (4 Gy), a lethal (9 Gy), or a single high-dose administration of 153Sm-ethylenediamine-N,N,N',N'-tetrakis(methylene phosphonic acid) (EDTMP). Flow cytometry and fluorescence microscopy were used to analyze hematopoietic cell population dynamics and the cellular site of GT3-Nano localization in the spleen and bone marrow, respectively. Results: Bone marrow uptake and retention (percentage injected dose per gram of tissue) at 24 h was 6.98 ± 2.34 for 64Cu-GT3-Nano and 7.44 ± 2.52 for 3H-GT3-Nano. GT3-Nano administered 24 h before or after 4 Gy of total-body irradiation (TBI) promoted rapid and complete hematopoietic recovery, whereas recovery of controls stalled at 60%. GT3-Nano demonstrated dose-dependent radioprotection, achieving 90% survival at 50 mg/kg against lethal 9-Gy TBI. Flow cytometry of the bone marrow indicated that progenitor bone marrow cells MPP2 and CMP were upregulated in GT3-Nano-treated mice. Immunohistochemistry showed that GT3-Nano accumulates in CD105-positive sinusoid epithelial cells. Conclusion: GT3-Nano is highly effective in mitigating the marrow-suppressive effects of sublethal and lethal TBI in mice. GT3-Nano can facilitate rapid recovery of hematopoietic components in mice treated with the endoradiotherapeutic agent 153Sm-EDTMP.
Collapse
Affiliation(s)
- Sang-Gyu Lee
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Hanzhi Lou
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kishore Gangangari
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Chemistry, Hunter College, City University of New York, New York, New York
| | - Blesida Punzalan
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | | | - Hebert A Vargas
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Lisa Bodei
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael G Kharas
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vijay K Singh
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland; and
| | - Naga Vara Kishore Pillarsetty
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Steven M Larson
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Weill Cornell Medical College, New York, New York
| |
Collapse
|
8
|
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: 146] [Impact Index Per Article: 36.5] [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
|
9
|
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: 0.8] [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
|
10
|
Radiolabeled liposomes and lipoproteins as lipidic nanoparticles for imaging and therapy. Chem Phys Lipids 2020; 230:104934. [PMID: 32562666 DOI: 10.1016/j.chemphyslip.2020.104934] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 06/09/2020] [Accepted: 06/15/2020] [Indexed: 02/06/2023]
Abstract
Radiolabeled lipidic nanoparticles, particularly liposomes and lipoproteins, are of great interest as agents for imaging and therapy, due not only to their peculiar physicochemical and biological properties, but also to their great versatility and the ability to manipulate them to obtain the desired properties. This review provides an overview of radionuclide labeling strategies for preparing diagnostic and therapeutic nanoparticles based on liposomes and lipoproteins that have been developed to date, as well as the main quality control methods and in vivo applications.
Collapse
|
11
|
Williams KM, Chakrabarty JH. Imaging haemopoietic stem cells and microenvironment dynamics through transplantation. Lancet Haematol 2020; 7:e259-e269. [PMID: 32109406 PMCID: PMC7820939 DOI: 10.1016/s2352-3026(20)30003-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 12/13/2019] [Accepted: 01/03/2020] [Indexed: 11/19/2022]
Abstract
Understanding the subclinical pathway to cellular engraftment following haemopoietic stem cell transplantation (HSCT) has historically been limited by infrequent marrow biopsies, which increase the risk of infections and might poorly represent the health of the marrow space. Nuclear imaging could represent an opportunity to evaluate the entire medullary space non-invasively, yielding information about cell number, proliferation, or metabolism. Because imaging is not associated with infectious risk, it permits assessment of neutropenic timepoints that were previously inaccessible. This Viewpoint summarises the data regarding the use of nuclear medicine techniques to assess the phases of HSCT: pre-transplant homoeostasis, induced aplasia, early settling and engraftment of infused cells, and later recovery of lymphocytes that target cancers or mediate tolerance. Although these data are newly emerging and preliminary, nuclear medicine imaging approaches might advance our understanding of HSCT events and lead to novel recommendations to enhance outcomes.
Collapse
Affiliation(s)
- Kirsten M Williams
- Department of Pediatrics, Emory University and the Children's Healthcare of Atlanta, Atlanta, GA, USA; Division of Blood and Marrow Transplantation, AFLAC Cancer and Blood disorder Center, Atlanta, GA, USA.
| | - Jennifer Holter Chakrabarty
- Department of Medicine, Division of Marrow Transplantation and Cell Therapy, Stephenson Cancer Center, Oklahoma CIty, OK, USA
| |
Collapse
|
12
|
Radiolabeled PET/MRI Nanoparticles for Tumor Imaging. J Clin Med 2019; 9:jcm9010089. [PMID: 31905769 PMCID: PMC7019574 DOI: 10.3390/jcm9010089] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/23/2019] [Accepted: 12/24/2019] [Indexed: 02/07/2023] Open
Abstract
The development of integrated positron emission tomography (PET)/magnetic resonance imaging (MRI) scanners opened a new scenario for cancer diagnosis, treatment, and follow-up. Multimodal imaging combines functional and morphological information from different modalities, which, singularly, cannot provide a comprehensive pathophysiological overview. Molecular imaging exploits multimodal imaging in order to obtain information at a biological and cellular level; in this way, it is possible to track biological pathways and discover many typical tumoral features. In this context, nanoparticle-based contrast agents (CAs) can improve probe biocompatibility and biodistribution, prolonging blood half-life to achieve specific target accumulation and non-toxicity. In addition, CAs can be simultaneously delivered with drugs or, in general, therapeutic agents gathering a dual diagnostic and therapeutic effect in order to perform cancer diagnosis and treatment simultaneous. The way for personalized medicine is not so far. Herein, we report principles, characteristics, applications, and concerns of nanoparticle (NP)-based PET/MRI CAs.
Collapse
|
13
|
El-Hammadi MM, Arias JL. An update on liposomes in drug delivery: a patent review (2014-2018). Expert Opin Ther Pat 2019; 29:891-907. [DOI: 10.1080/13543776.2019.1679767] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Mazen M. El-Hammadi
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Seville, Seville, Spain
| | - José L. Arias
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Granada, Granada, Spain
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain
- Biosanitary Research Institute of Granada (ibs.GRANADA), Andalusian Health Service (SAS), University of Granada, Granada, Spain
| |
Collapse
|
14
|
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: 105] [Impact Index Per Article: 17.5] [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
|
15
|
Medved'ko AV, Dalinger AI, Nuriev VN, Semashko VS, Filatov AV, Ezhov AA, Churakov AV, Howard JAK, Shiryaev AA, Baranchikov AE, Ivanov VK, Vatsadze SZ. Supramolecular Organogels Based on N-Benzyl, N'-Acylbispidinols. NANOMATERIALS (BASEL, SWITZERLAND) 2019; 9:E89. [PMID: 30641896 PMCID: PMC6359647 DOI: 10.3390/nano9010089] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 12/24/2018] [Accepted: 01/03/2019] [Indexed: 12/12/2022]
Abstract
The acylation of unsymmetrical N-benzylbispidinols in aromatic solvents without an external base led to the formation of supramolecular gels, which possess different thicknesses and degrees of stability depending on the substituents in para-positions of the benzylic group as well as on the nature of the acylating agent and of the solvent used. Structural features of the native gels as well as of their dried forms were studied by complementary techniques including Fourier-transform infrared (FTIR) and attenuated total reflection (ATR) spectroscopy, atomic force microscopy (AFM), transmission electron microscopy (TEM), scanning electron microscopy (SEM), and small-angle X-ray scattering and diffraction (SAXS). Structures of the key crystalline compounds were established by X-ray diffraction. An analysis of the obtained data allowed speculation on the crucial structural and condition factors that governed the gel formation. The most important factors were as follows: (i) absence of base, either external or internal; (ii) presence of HCl; (iii) presence of carbonyl and hydroxyl groups to allow hydrogen bonding; and (iv) presence of two (hetero)aromatic rings at both sides of the molecule. The hydrogen bonding involving amide carbonyl, hydroxyl at position 9, and, very probably, ammonium N-H⁺ and Cl- anion appears to be responsible for the formation of infinite molecular chains required for the first step of gel formation. Subsequent lateral cooperation of molecular chains into fibers occurred, presumably, due to the aromatic π-π-stacking interactions. Supercritical carbon dioxide drying of the organogels gave rise to aerogels with morphologies different from that of air-dried samples.
Collapse
Affiliation(s)
- Alexey V Medved'ko
- Faculty of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia.
| | | | - Vyacheslav N Nuriev
- Faculty of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia.
| | - Vera S Semashko
- Faculty of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia.
| | - Andrei V Filatov
- Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, 119991 Moscow, Russia.
| | - Alexander A Ezhov
- Faculty of Physics, Lomonosov Moscow State University, 119991 Moscow, Russia.
- Topchiev Institute of Petrochemical Synthesis, Russian Academy of Sciences, 119991 Moscow, Russia.
| | - Andrei V Churakov
- Kurnakov Institute of General and Inorganic Chemistry of the Russian Academy of Sciences, 119991 Moscow, Russia.
| | | | - Andrey A Shiryaev
- Frumkin Institute of Physical Chemistry and Electrochemistry, Russian Academy of Sciences, 119071 Moscow, Russia.
- Institute of Geology of Ore Deposits, Petrography, Mineralogy and Geochemistry, Russian Academy of Sciences, 119017 Moscow, Russia.
| | - Alexander E Baranchikov
- Faculty of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia.
- Kurnakov Institute of General and Inorganic Chemistry of the Russian Academy of Sciences, 119991 Moscow, Russia.
| | - Vladimir K Ivanov
- Kurnakov Institute of General and Inorganic Chemistry of the Russian Academy of Sciences, 119991 Moscow, Russia.
- Faculty of Material Science, Lomonosov Moscow State University, 119991 Moscow, Russia.
| | - Sergey Z Vatsadze
- Faculty of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia.
| |
Collapse
|
16
|
Wagner M, Wuest M, Hamann I, Lopez-Campistrous A, McMullen TPW, Wuest F. Molecular imaging of platelet-derived growth factor receptor-alpha (PDGFRα) in papillary thyroid cancer using immuno-PET. Nucl Med Biol 2017; 58:51-58. [PMID: 29367096 DOI: 10.1016/j.nucmedbio.2017.12.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 12/11/2017] [Accepted: 12/13/2017] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Receptor tyrosine kinase (RTK) platelet-derived growth factor receptor-alpha (PDGFRα) was recently identified as a molecular switch for dedifferentiation in thyroid cancer that predicts resistance to therapy as well as recurrence of disease in papillary thyroid cancer. Here we describe the radiolabeling and functional characterization of an imaging probe based on a PDGFRα-specific monoclonal antibody (mAb) for immuno-PET imaging of PDGFRα in papillary thyroid cancer. METHODS Antibody D13C6 (Cell Signaling) was decorated with chelator NOTA using bioconjugation reaction with 2-(p-NCS-Bz)-NOTA. Radiolabeling was carried out using 40 μg of antibody-NOTA conjugate with 143-223 MBq of [64Cu]CuCl2 in 0.25 M NaOAc (pH 5.5) at 30 °C for 1 h. The reaction mixture was purified with size-exclusion chromatography (PD-10 column). PDGFRα and mock transfected B-CPAP thyroid cancer cells lines for validation of 64Cu-labeled immuno-conjugates were generated using LVX-Tet-On technology. PET imaging was performed in NSG mice bearing bilaterally-induced PDGFRα (+/-) B-CPAP tumors. RESULTS Bioconjugation of NOTA chelator to monoclonal antibody D13C6 resulted in 2.8 ± 1.3 chelator molecules per antibody as determined by radiometric titration with 64Cu. [64Cu]Cu-NOTA-D13C6 was isolated in high radiochemical purity (>98%) and good radiochemical yields (19-61%). The specific activity was 0.9-5.1 MBq/μg. Cellular uptake studies revealed a specific radiotracer uptake in PDGFRα expressing cells compared to control cells. PET imaging resulted in SUVmean values of ~5.5 for PDGFRα (+) and ~2 for PDGFRα (-) tumors, after 48 h p.i.. After 1 h, radiotracer uptake was also observed in the bone marrow (SUVmean ~5) and spleen (SUVmean ~8.5). CONCLUSION Radiolabeled antibody [64Cu]Cu-NOTA-D13C6 represents a novel and promising radiotracer for immuno-PET imaging of PDGFRα in metastatic papillary thyroid cancer. ADVANCES IN KNOWLEDGE AND IMPLICATIONS FOR PATIENT CARE The presented work has the potential to allow physicians to identify papillary thyroid cancer patients at risk of metastases by using the novel immuno-PET imaging assay based on PDGFRα-targeting antibody [64Cu]Cu-NOTA-D13C6.
Collapse
Affiliation(s)
- Michael Wagner
- University of Alberta, Department of Oncology, 11560 University Ave, Edmonton, AB T6G 1Z2, Canada
| | - Melinda Wuest
- University of Alberta, Department of Oncology, 11560 University Ave, Edmonton, AB T6G 1Z2, Canada
| | - Ingrit Hamann
- University of Alberta, Department of Oncology, 11560 University Ave, Edmonton, AB T6G 1Z2, Canada
| | - Ana Lopez-Campistrous
- University of Alberta, Department of Surgery, 2D4.41 Walter Mackenzie Centre 8440- 112 Street, Edmonton, AB T6G 2B7, Canada
| | - Todd P W McMullen
- University of Alberta, Department of Oncology, 11560 University Ave, Edmonton, AB T6G 1Z2, Canada; University of Alberta, Department of Surgery, 2D4.41 Walter Mackenzie Centre 8440- 112 Street, Edmonton, AB T6G 2B7, Canada.
| | - Frank Wuest
- University of Alberta, Department of Oncology, 11560 University Ave, Edmonton, AB T6G 1Z2, Canada.
| |
Collapse
|
17
|
Brand C, Iacono P, Pérez-Medina C, Mulder WJM, Kircher MF, Reiner T. Specific Binding of Liposomal Nanoparticles through Inverse Electron-Demand Diels-Alder Click Chemistry. ChemistryOpen 2017; 6:615-619. [PMID: 29046855 PMCID: PMC5641912 DOI: 10.1002/open.201700105] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Indexed: 12/12/2022] Open
Abstract
Here, we report a method to specifically bind liposomal radiopharmaceuticals to a CoCrMo alloy, which can be used in arterial stents, via an irreversible inverse electron‐demand Diels–Alder reaction. Inspired by recent accomplishments in pre‐targeted imaging using tetrazine‐trans‐cyclooctene click chemistry, we synthesized 89Zr‐labeled trans‐cyclooctene‐functionalized liposomal nanoparticles, which were validated on a tetrazine‐appended polydopamine‐coated CoCrMo surface. In efforts to ultimately translate this new material to biomedical applications, we compared the ability of 89Zr‐TCO–liposomal nanoparticles (89Zr‐TCO‐LNP) to be immobilized on the tetrazine surface to the control suspensions of non‐TCO functionalized 89Zr‐liposomal nanoparticles. Ultimately, this platform technology could result in a systemic decrease of the radiotherapeutic dose deposited in non‐targeted tissues by specific removal of long‐circulating liposomal radiopharmaceuticals from the blood pool.
Collapse
Affiliation(s)
- Christian Brand
- Department of Radiology Memorial Sloan Kettering Cancer Center 1275 York Avenue New York NY 10065 USA
| | - Pasquale Iacono
- Department of Radiology Memorial Sloan Kettering Cancer Center 1275 York Avenue New York NY 10065 USA
| | - Carlos Pérez-Medina
- Translational and Molecular Imaging InstituteIcahn School of Medicine at Mount Sinai 1470 Madison Ave New York NY 10029 USA
| | - Willem J M Mulder
- Translational and Molecular Imaging InstituteIcahn School of Medicine at Mount Sinai 1470 Madison Ave New York NY 10029 USA.,Department of Medical Biochemistry Academic Medical Center Meibergdreef 91105 AZ Amsterdam The Netherlands
| | - Moritz F Kircher
- Department of Radiology Memorial Sloan Kettering Cancer Center 1275 York Avenue New York NY 10065 USA.,Department of Radiology Weill Cornell Medical College 1300 York Avenue New York NY 10065 USA.,Center for Molecular Imaging & Nanotechnology (CMINT) Memorial Sloan Kettering Cancer Center 1275 York Avenue New York NY 10065 USA.,Molecular Pharmacology Program Memorial Sloan Kettering Cancer Center 1275 York Avenue New York NY 10065 USA
| | - Thomas Reiner
- Department of Radiology Memorial Sloan Kettering Cancer Center 1275 York Avenue New York NY 10065 USA.,Department of Radiology Weill Cornell Medical College 1300 York Avenue New York NY 10065 USA.,Center for Molecular Imaging & Nanotechnology (CMINT) Memorial Sloan Kettering Cancer Center 1275 York Avenue New York NY 10065 USA
| |
Collapse
|
18
|
Lamichhane N, Dewkar GK, Sundaresan G, Mahon RN, Zweit J. [ 18F]-Fluorinated Carboplatin and [ 111In]-Liposome for Image-Guided Drug Delivery. Int J Mol Sci 2017; 18:E1079. [PMID: 28524076 PMCID: PMC5454988 DOI: 10.3390/ijms18051079] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 05/02/2017] [Accepted: 05/08/2017] [Indexed: 11/16/2022] Open
Abstract
Radiolabeled liposomes have been employed as diagnostic tools to monitor in vivo distribution of liposomes in real-time, which helps in optimizing the therapeutic efficacy of the liposomal drug delivery. This work utilizes the platform of [111In]-Liposome as a drug delivery vehicle, encapsulating a novel 18F-labeled carboplatin drug derivative ([18F]-FCP) as a dual-molecular imaging tool as both a radiolabeled drug and radiolabeled carrier. The approach has the potential for clinical translation in individual patients using a dual modal approach of clinically-relevant radionuclides of 18F positron emission tomography (PET) and 111In single photon emission computed tomography (SPECT). [111In]-Liposome was synthesized and evaluated in vivo by biodistribution and SPECT imaging. The [18F]-FCP encapsulated [111In]-Liposome nano-construct was investigated, in vivo, using an optimized dual-tracer PET and SPECT imaging in a nude mouse. The biodistribution data and SPECT imaging showed spleen and liver uptake of [111In]-Liposome and the subsequent clearance of activity with time. Dual-modality imaging of [18F]-FCP encapsulated [111In]-Liposome showed significant uptake in liver and spleen in both PET and SPECT images. Qualitative analysis of SPECT images and quantitative analysis of PET images showed the same pattern of activity during the imaging period and demonstrated the feasibility of dual-tracer imaging of a single dual-labeled nano-construct.
Collapse
Affiliation(s)
- Narottam Lamichhane
- Center for Molecular Imaging, Department of Radiology, Virginia Commonwealth University, 1101 E. Marshall Street, Richmond, VA 23298-0031, USA.
| | - Gajanan K Dewkar
- Center for Molecular Imaging, Department of Radiology, Virginia Commonwealth University, 1101 E. Marshall Street, Richmond, VA 23298-0031, USA.
| | - Gobalakrishnan Sundaresan
- Center for Molecular Imaging, Department of Radiology, Virginia Commonwealth University, 1101 E. Marshall Street, Richmond, VA 23298-0031, USA.
| | - Rebecca N Mahon
- Center for Molecular Imaging, Department of Radiology, Virginia Commonwealth University, 1101 E. Marshall Street, Richmond, VA 23298-0031, USA.
| | - Jamal Zweit
- Center for Molecular Imaging, Department of Radiology, Virginia Commonwealth University, 1101 E. Marshall Street, Richmond, VA 23298-0031, USA.
| |
Collapse
|