1
|
Dixit T, Dave N, Basu K, Sonawane P, Gawas T, Ravindran S. Nano-radiopharmaceuticals as therapeutic agents. Front Med (Lausanne) 2024; 11:1355058. [PMID: 38560384 PMCID: PMC10978739 DOI: 10.3389/fmed.2024.1355058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 02/26/2024] [Indexed: 04/04/2024] Open
Abstract
In recent years, there has been an increased interest in exploring the potential synergy between nanotechnology and nuclear medicine. The application of radioactive isotopes, commonly referred to as radiopharmaceuticals, is recognized in nuclear medicine for diagnosing and treating various diseases. Unlike conventional pharmaceutical agents, radiopharmaceuticals are designed to work without any pharmacological impact on the body. Nevertheless, the radiation dosage employed in radiopharmaceuticals is often sufficiently high to elicit adverse effects associated with radiation exposure. Exploiting their capacity for selective accumulation on specific organ targets, radiopharmaceuticals have utility in treating diverse disorders. The incorporation of nanosystems may additionally augment the targeting capability of radiopharmaceuticals, leveraging their distinct pharmacokinetic characteristics. Conversely, radionuclides could be used in research to assess nanosystems pharmacologically. However, more investigation is needed to verify the safety and effectiveness of radiopharmaceutical applications mediated by nanosystems. The use of nano-radiopharmaceuticals as therapeutic agents to treat various illnesses and disorders is majorly covered in this review. The targeted approach to cancer therapy and various types of nanotools for nano-radiopharmaceutical delivery, is also covered in this article.
Collapse
Affiliation(s)
| | | | | | | | | | - Selvan Ravindran
- Symbiosis School of Biological Sciences, Faculty of Medical and Health Sciences, Symbiosis International (Deemed University), Lavale, Pune, India
| |
Collapse
|
2
|
Ozolmez N, Silindir-Gunay M, Volkan-Salanci B. An overview: Radiotracers and nano-radiopharmaceuticals for diagnosis of Parkinson's disease. Appl Radiat Isot 2024; 203:111110. [PMID: 37989065 DOI: 10.1016/j.apradiso.2023.111110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 11/23/2023]
Abstract
Parkinson's disease (PD) is a widespread progressive neurodegenerative disease. Clinical diagnosis approaches are insufficient to provide an early and accurate diagnosis before a substantial of loss of dopaminergic neurons. PET and SPECT can be used for accurate and early diagnosis of PD by using target-specific radiotracers. Additionally, the importance of BBB penetrating targeted nanosystems has increased in recent years. This article reviews targeted radiopharmaceuticals used in clinics and novel nanocarriers for research purposes of PD imaging.
Collapse
Affiliation(s)
- Nur Ozolmez
- Hacettepe University, Faculty of Pharmacy, Department of Radiopharmacy, Ankara, Turkey.
| | - Mine Silindir-Gunay
- Hacettepe University, Faculty of Pharmacy, Department of Radiopharmacy, Ankara, Turkey.
| | - Bilge Volkan-Salanci
- Hacettepe University, Faculty of Medicine, Department of Nuclear Medicine, Ankara, Turkey.
| |
Collapse
|
3
|
Suman SK, Mukherjee A, Sharma RK. A liposomal radionanoformulation for targeted drug delivery and real time monitoring by radionuclide imaging for HER2 overexpressing cancers. Drug Dev Res 2023; 84:1553-1563. [PMID: 37578143 DOI: 10.1002/ddr.22106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/21/2023] [Accepted: 07/28/2023] [Indexed: 08/15/2023]
Abstract
Liposomal formulations carrying chemotherapeutic drugs have demonstrated great potential as effective drug delivery systems. Smart nanoformulations decorated with targeting agents and probes are desired for site specific delivery of drugs and real time monitoring. In this study, we aimed to develop liposomal formulation loaded with doxorubicin and tagged with trastuzumab antibody (Ab) for targeting human epidermal growth factor receptor 2 (HER2) positive tumors. Liposomes were prepared by ethanol injection method using modified lipids to conjugate trastuzumab and radiolabel with Tc-99m radioisotope using DTPA for imaging by single photon emission computed tomography (SPECT). Doxorubicin was loaded using the active pH gradient method. The conjugation of Ab to liposomes was validated by SDS-PAGE and MALDI-MS. 99m Tc labeled liposomes encapsulating doxorubicin conjugated with antibody (99m Tc-Lip-Ab-Dox) and 99m Tc labeled liposomes encapsulating doxorubicin (99m Tc-Lip-Dox) were found to be stable in blood plasma and saline using chromatography method. The specificity of 99m Tc-Lip-Ab-Dox against HER2 receptor was evident from cell uptake and inhibition studies. Results also corroborated with confocal microscopy studies. In vivo studies in tumor bearing severe combined immunodeficient mice by SPECT imaging and biodistribution studies revealed higher uptake of 99m Tc-Lip-Ab-Dox in tumor and less accumulation in the liver compared to 99m Tc-Lip-Dox. In conclusion, liposomal nanoformulation for immunotargeting and monitoring of drug delivery was successfully formulated and evaluated. Encouraging results in preclinical studies were obtained with the radioformulation. Such smart radioformulations will not only serve the purpose of site-specific controlled release of drugs at the target site but also aid in optimizing the drug doses and schedule of cancer treatment by monitoring pharmacokinetics.
Collapse
Affiliation(s)
- Shishu Kant Suman
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre (BARC), Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Archana Mukherjee
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre (BARC), Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Rohit Kumar Sharma
- Department of Chemistry and Centre of Advanced Studies, Panjab University, Chandigarh, India
| |
Collapse
|
4
|
Wen H, Poutiainen P, Batnasan E, Latonen L, Lehto VP, Xu W. Biomimetic Inorganic Nanovectors as Tumor-Targeting Theranostic Platform against Triple-Negative Breast Cancer. Pharmaceutics 2023; 15:2507. [PMID: 37896267 PMCID: PMC10610067 DOI: 10.3390/pharmaceutics15102507] [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] [Received: 09/06/2023] [Revised: 10/18/2023] [Accepted: 10/20/2023] [Indexed: 10/29/2023] Open
Abstract
Mesoporous silicon nanoparticles (PSi NPs) are promising platforms of nanomedicine because of their good compatibility, high payload capacities of anticancer drugs, and easy chemical modification. Here, PSi surfaces were functionalized with bisphosphonates (BP) for radiolabeling, loaded with doxorubicin (DOX) for chemotherapy, and the NPs were coated with cancer cell membrane (CCm) for homotypic cancer targeting. To enhance the CCm coating, the NP surfaces were covered with polyethylene glycol prior to the CCm coating. The effects of the BP amount and pH conditions on the radiolabeling efficacy were studied. The maximum BP was (2.27 wt%) on the PSi surfaces, and higher radiochemical yields were obtained for 99mTc (97% ± 2%) and 68Ga (94.6% ± 0.2%) under optimized pH conditions (pH = 5). The biomimetic NPs exhibited a good radiochemical and colloidal stability in phosphate-buffered saline and cell medium. In vitro studies demonstrated that the biomimetic NPs exhibited an enhanced cellular uptake and increased delivery of DOX to cancer cells, resulting in better chemotherapy than free DOX or pure NPs. Altogether, these findings indicate the potential of the developed platform for cancer treatment and diagnosis.
Collapse
Affiliation(s)
- Huang Wen
- Department of Technical Physics, University of Eastern Finland, Yliopistonranta 1F, 70211 Kuopio, Finland;
| | - Pekka Poutiainen
- Kuopio University Hospital, University of Eastern Finland, Puijonlaaksontie 2, 70210 Kuopio, Finland;
| | - Enkhzaya Batnasan
- School of Medicine, University of Eastern Finland, Yliopistonranta 1F, 70211 Kuopio, Finland; (E.B.); (L.L.)
| | - Leena Latonen
- School of Medicine, University of Eastern Finland, Yliopistonranta 1F, 70211 Kuopio, Finland; (E.B.); (L.L.)
| | - Vesa-Pekka Lehto
- Department of Technical Physics, University of Eastern Finland, Yliopistonranta 1F, 70211 Kuopio, Finland;
| | - Wujun Xu
- Department of Technical Physics, University of Eastern Finland, Yliopistonranta 1F, 70211 Kuopio, Finland;
| |
Collapse
|
5
|
Umeda IO, Koike Y, Ogata M, Kaneko E, Hamamichi S, Uehara T, Moribe K, Arano Y, Takahashi T, Fujii H. New liposome-radionuclide-chelate combination for tumor targeting and rapid healthy tissue clearance. J Control Release 2023; 361:847-855. [PMID: 37543291 DOI: 10.1016/j.jconrel.2023.07.060] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 07/19/2023] [Accepted: 07/31/2023] [Indexed: 08/07/2023]
Abstract
Radionuclide imaging and therapy are promising methods for controlling systemic cancers; however, their clinical application has been limited by excessive radionuclide accumulation in healthy tissues. To minimize radionuclide accumulation in non-cancerous tissues while ensuring sufficient build up in tumors, we aimed to develop a method that controlled the in vivo dynamics of radionuclides post-administration. To this end, we describe a novel strategy that combines liposomes, a potent carrier system for drug delivery, with unique radionuclide-ligand complexes based on 111In-ethylenedicysteine. Conventional 111In-ligand-complexes-carrying liposomes delivered substantial amounts of radionuclides to tumors; however, they also accumulated in the liver and spleen. In contrast, 111In-ethylenedicysteine-carrying liposomes greatly reduced non-specific accumulation, while being retained selectively at high doses within tumors. Liposomes were rapidly broken down in the liver, releasing encapsulated 111In-ligand complexes. Among the chelates used, only 111In-ethylenedicysteine could escape from the liver and be excreted in the urine. Instead, most liposomes remained intact in tumors, retaining the radionuclide-ligand complexes within them. Therefore, high tumor accumulation was obtained regardless of the type of 111In-ligand complexes in the liposomes. In vivo single photon emission computed tomography/computed tomography imaging with 111In-ethylenedicysteine-carrying liposomes accurately revealed tumor-selective radionuclide retention with little background. Hence, our new strategy could greatly enhance tumor-to-healthy tissue ratios, improve diagnostic imaging, boost therapeutic efficacy, reduce toxicity to healthy tissues, and facilitate radionuclide imaging and therapy.
Collapse
Affiliation(s)
- Izumi O Umeda
- Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, 6-5-1, Kashiwanoha, Kashiwa, Chiba 277-8577, Japan; Kavli Institute for the Physics and Mathematics of the Universe (WPI), The University of Tokyo Institutes for Advanced Study, 5-1-5, Kashiwanoha, Kashiwa, Chiba 277-8583, Japan; Kyoto College of Medical Science, 1-3, Imakita, Oyama-higashi, Sonobe, Nantan, Kyoto 622-0041, Japan.
| | - Yusuke Koike
- Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, 6-5-1, Kashiwanoha, Kashiwa, Chiba 277-8577, Japan; Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1, Inohana, Cyuo-ku, Chiba, Chiba 260-8675, Japan
| | - Mayumi Ogata
- Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, 6-5-1, Kashiwanoha, Kashiwa, Chiba 277-8577, Japan; Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1, Inohana, Cyuo-ku, Chiba, Chiba 260-8675, Japan
| | - Emi Kaneko
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1, Inohana, Cyuo-ku, Chiba, Chiba 260-8675, Japan
| | - Shusei Hamamichi
- Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, 6-5-1, Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| | - Tomoya Uehara
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1, Inohana, Cyuo-ku, Chiba, Chiba 260-8675, Japan
| | - Kunikazu Moribe
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1, Inohana, Cyuo-ku, Chiba, Chiba 260-8675, Japan
| | - Yasushi Arano
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1, Inohana, Cyuo-ku, Chiba, Chiba 260-8675, Japan
| | - Tadayuki Takahashi
- Kavli Institute for the Physics and Mathematics of the Universe (WPI), The University of Tokyo Institutes for Advanced Study, 5-1-5, Kashiwanoha, Kashiwa, Chiba 277-8583, Japan
| | - Hirofumi Fujii
- Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, 6-5-1, Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| |
Collapse
|
6
|
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
|
7
|
Image-guided drug delivery in nanosystem-based cancer therapies. Adv Drug Deliv Rev 2023; 192:114621. [PMID: 36402247 DOI: 10.1016/j.addr.2022.114621] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/18/2022] [Accepted: 11/13/2022] [Indexed: 11/18/2022]
Abstract
The past decades have shown significant advancements in the development of solid tumor treatment. For instance, implementation of nanosystems for drug delivery has led to a reduction in side effects and improved delivery to the tumor region. However, clinical translation has faced challenges, as tumor drug levels are still considered to be inadequate. Interdisciplinary research has resulted in the development of more advanced drug delivery systems. These are coined "smart" due to the ability to be followed and actively manipulated in order to have better control over local drug release. Therefore, image-guided drug delivery can be a powerful strategy to improve drug activity at the target site. Being able to visualize the inflow of the administered smart nanosystem within the tumor gives the potential to determine the right moment to apply the facilitator to initiate drug release. Here we provide an overview of available nanosystems, imaging moieties, and imaging techniques. We discuss preclinical application of these smart drug delivery systems, the strength of image-guided drug delivery, and the future of personalized treatment.
Collapse
|
8
|
Fahmy S, Preis E, Dayyih AA, Alawak M, El-Said Azzazy HM, Bakowsky U, Shoeib T. Thermosensitive Liposomes Encapsulating Nedaplatin and Picoplatin Demonstrate Enhanced Cytotoxicity against Breast Cancer Cells. ACS OMEGA 2022; 7:42115-42125. [PMID: 36440163 PMCID: PMC9686199 DOI: 10.1021/acsomega.2c04525] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 10/13/2022] [Indexed: 06/16/2023]
Abstract
Thermosensitive liposomes (TSL) have been used for localized temperature-responsive release of chemotherapeutics into solid cancers, with a minimum of one invention currently in clinical trials (phase III). In this study, TSL was designed using a lipid blend comprising 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), cholesterol, and 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[maleimide(polyethylene glycol)-2000] (DSPE-PEG-2000) (molar ratio of 88:9:2.8:0.2). Either nedaplatin (ND) or p-sulfonatocalix[4]arene-nedaplatin was encapsulated in the aqueous inner layer of TSL to form (ND-TSL) or p-SC4-ND-TSL, respectively. The hydrophobic platinum-based drug picoplatin (P) was loaded into the external lipid bilayer of the TSL to develop P-TSL. The three nanosystems were studied in terms of size, PDI, surface charge, and on-shelf stability. Moreover, the entrapment efficiency (EE%) and release % at 37 and 40 °C were evaluated. In a 30 min in vitro release study, the maximum release of ND, p-SC4-ND, and picoplatin at 40 °C reached 74, 79, and 75%, respectively, compared to approximately 10% at 37 °C. This demonstrated temperature-triggered drug release from the TSL in all three developed systems. The designed TSL exhibited significant in vitro anticancer activity at 40 °C when tested on human mammary gland/breast adenocarcinoma cells (MDA-MB-231). The cytotoxicity of ND-TSL, p-SC4-ND-TSL, and P-TSL at 40 °C was approximately twice those observed at 37 °C. This study suggests that TSL is a promising nanoplatform for the temperature-triggered release of platinum-based drugs into cancer cells.
Collapse
Affiliation(s)
- Sherif
Ashraf Fahmy
- Department
of Chemistry, American University in Cairo
(AUC), AUC Avenue, P.O. Box 74, New Cairo11835, Egypt
- Department
of Chemistry, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation, R5 New Garden City, New Administrative
Capital, AL109AB, Cairo11835, Egypt
| | - Eduard Preis
- Department
of Pharmaceutics and Biopharmaceutics, University
of Marburg, Robert-Koch-Str. 4, 35037Marburg, Germany
| | - Alice Abu Dayyih
- Department
of Pharmaceutics and Biopharmaceutics, University
of Marburg, Robert-Koch-Str. 4, 35037Marburg, Germany
| | - Mohamed Alawak
- Department
of Pharmaceutics and Biopharmaceutics, University
of Marburg, Robert-Koch-Str. 4, 35037Marburg, Germany
| | | | - Udo Bakowsky
- Department
of Pharmaceutics and Biopharmaceutics, University
of Marburg, Robert-Koch-Str. 4, 35037Marburg, Germany
| | - Tamer Shoeib
- Department
of Chemistry, American University in Cairo
(AUC), AUC Avenue, P.O. Box 74, New Cairo11835, Egypt
| |
Collapse
|
9
|
Bhargava S, de la Puente-Secades S, Schurgers L, Jankowski J. Lipids and lipoproteins in cardiovascular diseases: a classification. Trends Endocrinol Metab 2022; 33:409-423. [PMID: 35370062 DOI: 10.1016/j.tem.2022.02.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 02/04/2022] [Accepted: 02/07/2022] [Indexed: 12/15/2022]
Abstract
Lipids and lipoproteins, their metabolism, and their transport are essential contributing factors of cardiovascular disease (CVD) as they regulate plasma cholesterol concentration, enhancing cholesterol uptake by macrophages, leading to foam cell formation and ultimately resulting in plaque formation and inflammation. However, lipids and lipoproteins have cardioprotective functions as well, such as preventing oxidation of proatherogenic molecules and downregulating inflammatory proteins.
Collapse
Affiliation(s)
- Shruti Bhargava
- Institute of Molecular Cardiovascular Research, Medical Faculty, RWTH Aachen University, Aachen, Germany; Experimental Vascular Pathology, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands
| | - Sofia de la Puente-Secades
- Institute of Molecular Cardiovascular Research, Medical Faculty, RWTH Aachen University, Aachen, Germany; Experimental Vascular Pathology, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands
| | - Leon Schurgers
- Experimental Vascular Pathology, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands
| | - Joachim Jankowski
- Institute of Molecular Cardiovascular Research, Medical Faculty, RWTH Aachen University, Aachen, Germany; Experimental Vascular Pathology, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands.
| |
Collapse
|
10
|
Qian J, Wen H, Tamarov K, Xu W, Lehto V. Recent Developments in Porous Silicon Nanovectors with Various Imaging Modalities in the Framework of Theranostics. ChemMedChem 2022; 17:e202200004. [PMID: 35212460 PMCID: PMC9314675 DOI: 10.1002/cmdc.202200004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 02/24/2022] [Indexed: 11/17/2022]
Abstract
The number of in vitro, ex vivo, and in vivo studies on porous silicon (PSi) nanoparticles for biomedical applications has increased extensively over the last decade. The focus of the reports has been on the carrier properties of PSi concerning the therapeutic aspect due to several beneficial nanovector characteristics including high payload capacity, biocompatibility, and versatile surface chemistry. Recently, increasing attention has been paid to the diagnostic aspects of PSi, which is typically attributed to the biotraceability of the nanovector. Also, PSi has been studied as a contrast agent. When both these aspects, therapy and diagnosis, are integrated into one nanovector, we can discuss a real nanotheranostics approach. Herein, we review the recent progress developing PSi for various imaging modalities, specifically focusing on optical imaging, magnetic resonance imaging, and nuclear medicine imaging. Furthermore, we summarized the knowledge gaps that must be covered before applying PSi in clinical imaging, highlighting future research trends.
Collapse
Affiliation(s)
- Jing Qian
- Department of Applied PhysicsUniversity of Eastern FinlandYliopistonranta 170211KuopioFinland
- College of Chemistry and Chemical EngineeringLanzhou UniversityLanzhou730000China
| | - Huang Wen
- Department of Applied PhysicsUniversity of Eastern FinlandYliopistonranta 170211KuopioFinland
| | - Konstantin Tamarov
- Department of Applied PhysicsUniversity of Eastern FinlandYliopistonranta 170211KuopioFinland
| | - Wujun Xu
- Department of Applied PhysicsUniversity of Eastern FinlandYliopistonranta 170211KuopioFinland
| | - Vesa‐Pekka Lehto
- Department of Applied PhysicsUniversity of Eastern FinlandYliopistonranta 170211KuopioFinland
| |
Collapse
|
11
|
Detiger SE, Kremer TM, A.S.H. Dalm V, de Keizer RO, Wubbels RJ, Metselaar J, van Hagen PM, Peeters RP, Paridaens D. A pilot study on the use of prednisolone-encapsulated liposomes for the treatment of moderate-to-severe Graves' orbitopathy with reduced systemic steroid exposure. Acta Ophthalmol 2021; 99:797-804. [PMID: 33423386 DOI: 10.1111/aos.14751] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/29/2020] [Accepted: 12/07/2020] [Indexed: 12/17/2022]
Abstract
PURPOSE To demonstrate that long-circulating PEGylated liposomal prednisolone is a safe and effective therapy in patients with active moderate-to-severe Graves' orbitopathy. METHODS Open-label, proof-of-concept, multicentre pilot study. Ten patients with moderate-to-severe Graves's orbitopathy, who were euthyroid for at least three months. Long-circulating PEGylated liposomal prednisolone 150 mg was administered intravenously twice, with 2-week interval. Total follow-up was 12 months, with visits at baseline, week 2, 6, 13, 26 and 52. Physical, laboratory and ophthalmological examinations were performed. Response to treatment was defined as a reduction in Clinical Activity Score by ≥2 points; palpebral aperture by ≥3 mm; soft tissue signs by ≥2 grades; exophthalmos by ≥2 mm; and motility by >8 degrees or improvement in diplopia score. A response was sustained when equally observed at weeks 6 and 13. RESULTS One patient achieved a sustained response according to the predetermined definition. All patients showed a decrease in Clinical Activity Score after one infusion, with a mean decrease of two points. The Clinical Activity Score was ≤1 at week 52 for all patients. Improvement was also observed in the soft tissue signs. Most of the adverse events were mild and of a transient nature. Two patients required further treatment with intravenous methylprednisolone. CONCLUSION This pilot study showed a positive effect of long-circulating PEGylated liposomal prednisolone on the Clinical Activity Score in patients with moderate-to-severe Graves's orbitopathy, resulting in fewer hospital visits and possibly less glucocorticoid-related side-effects.
Collapse
Affiliation(s)
| | | | - Virgil A.S.H. Dalm
- Department of Internal Medicine Division of Clinical Immunology & Department of Immunology Erasmus University Medical Center Rotterdam Rotterdam the Netherlands
| | - Ronald O.B. de Keizer
- The Rotterdam Eye Hospital Division of Oculoplastic, Orbital and Lacrimal Surgery Rotterdam the Netherlands
| | - Rene J. Wubbels
- The Rotterdam Ophthalmic Institute Rotterdam the Netherlands
| | - J.M. Metselaar
- Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Forckenbeckstrasse 55, 52074 Aachen, Germany
| | - P. Martin van Hagen
- Department of Internal Medicine Division of Clinical Immunology & Department of Immunology Erasmus University Medical Center Rotterdam Rotterdam the Netherlands
| | - Robin P. Peeters
- Department of Internal Medicine Division of Endocrinology Erasmus University Medical Center Rotterdam Rotterdam the Netherlands
| | - Dion Paridaens
- The Rotterdam Eye Hospital Division of Oculoplastic, Orbital and Lacrimal Surgery Rotterdam the Netherlands
- Department of Ophthalmology Erasmus Medical Center Rotterdam Rotterdam the Netherlands
| |
Collapse
|
12
|
Synthesis and Characterization of Multifunctional Nanovesicles Composed of POPC Lipid Molecules for Nuclear Imaging. Molecules 2021; 26:molecules26216591. [PMID: 34770999 PMCID: PMC8587727 DOI: 10.3390/molecules26216591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 10/26/2021] [Accepted: 10/27/2021] [Indexed: 12/02/2022] Open
Abstract
The integration of nuclear imaging analysis with nanomedicine has tremendously grown and represents a valid and powerful tool for the development and clinical translation of drug delivery systems. Among the various types of nanostructures used as drug carriers, nanovesicles represent intriguing platforms due to their capability to entrap both lipophilic and hydrophilic agents, and their well-known biocompatibility and biodegradability. In this respect, here we present the development of a labelling procedure of POPC (1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine)-based liposomes incorporating an ad hoc designed lipophilic NOTA (1,4,7-triazacyclononane-1,4,7-triacetic acid) analogue, derivatized with an oleic acid residue, able to bind the positron emitter gallium-68(III). Based on POPC features, the optimal conditions for liposome labelling were studied with the aim of optimizing the Ga(III) incorporation and obtaining a significant radiochemical yield. The data presented in this work demonstrate the feasibility of the labelling procedure on POPC liposomes co-formulated with the ad hoc designed NOTA analogue. We thus provided a critical insight into the practical aspects of the development of vesicles for theranostic approaches, which in principle can be extended to other nanosystems exploiting a variety of bioconjugation protocols.
Collapse
|
13
|
Barani M, Sangiovanni E, Angarano M, Rajizadeh MA, Mehrabani M, Piazza S, Gangadharappa HV, Pardakhty A, Mehrbani M, Dell’Agli M, Nematollahi MH. Phytosomes as Innovative Delivery Systems for Phytochemicals: A Comprehensive Review of Literature. Int J Nanomedicine 2021; 16:6983-7022. [PMID: 34703224 PMCID: PMC8527653 DOI: 10.2147/ijn.s318416] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 08/13/2021] [Indexed: 12/12/2022] Open
Abstract
Nowadays, medicinal herbs and their phytochemicals have emerged as a great therapeutic option for many disorders. However, poor bioavailability and selectivity might limit their clinical application. Therefore, bioavailability is considered a notable challenge to improve bio-efficacy in transporting dietary phytochemicals. Different methods have been proposed for generating effective carrier systems to enhance the bioavailability of phytochemicals. Among them, nano-vesicles have been introduced as promising candidates for the delivery of insoluble phytochemicals. Due to the easy preparation of the bilayer vesicles and their adaptability, they have been widely used and approved by the scientific literature. The first part of the review is focused on introducing phytosome technology as well as its applications, with emphasis on principles of formulations and characterization. The second part provides a wide overview of biological activities of commercial and non-commercial phytosomes, divided by systems and related pathologies. These results confirm the greater effectiveness of phytosomes, both in terms of biological activity or reduced dosage, highlighting curcumin and silymarin as the most formulated compounds. Finally, we describe the promising clinical and experimental findings regarding the applications of phytosomes. The conclusion of this study encourages the researchers to transfer their knowledge from laboratories to market, for a further development of these products.
Collapse
Affiliation(s)
- Mahmood Barani
- Medical Mycology and Bacteriology Research Center, Kerman University of Medical Sciences, Kerman, 76169-13555, Iran
| | - Enrico Sangiovanni
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, 20133, Italy
| | - Marco Angarano
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, 20133, Italy
| | | | - Mehrnaz Mehrabani
- Physiology Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Stefano Piazza
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, 20133, Italy
| | | | - Abbas Pardakhty
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mehrzad Mehrbani
- Department of Traditional Medicine, Faculty of Traditional Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mario Dell’Agli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, 20133, Italy
| | - Mohammad Hadi Nematollahi
- Herbal and Traditional Medicines Research Center, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
14
|
Ruiz-Pulido G, Medina DI, Barani M, Rahdar A, Sargazi G, Baino F, Pandey S. Nanomaterials for the Diagnosis and Treatment of Head and Neck Cancers: A Review. MATERIALS (BASEL, SWITZERLAND) 2021; 14:3706. [PMID: 34279276 PMCID: PMC8269895 DOI: 10.3390/ma14133706] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/25/2021] [Accepted: 06/29/2021] [Indexed: 12/12/2022]
Abstract
Head and neck cancer (HNC) is a category of cancers that typically arise from the nose-, mouth-, and throat-lining squamous cells. The later stage of HNC diagnosis significantly affects the patient's survival rate. This makes it mandatory to diagnose this cancer with a suitable biomarker and imaging techniques at the earlier stages of growth. There are limitations to traditional technologies for early detection of HNC. Furthermore, the use of nanocarriers for delivering chemo-, radio-, and phototherapeutic drugs represents a promising approach for improving the outcome of HNC treatments. Several studies with nanostructures focus on the development of a targeted and sustained release of anticancer molecules with reduced side effects. Besides, nanovehicles could allow co-delivering of anticancer drugs for synergistic activity to counteract chemo- or radioresistance. Additionally, a new generation of smart nanomaterials with stimuli-responsive properties have been developed to distinguish between unique tumor conditions and healthy tissue. In this light, the present article reviews the mechanisms used by different nanostructures (metallic and metal oxide nanoparticles, polymeric nanoparticles, quantum dots, liposomes, nanomicelles, etc.) to improve cancer diagnosis and treatment, provides an up-to-date picture of the state of the art in this field, and highlights the major challenges for future improvements.
Collapse
Affiliation(s)
- Gustavo Ruiz-Pulido
- Tecnologico de Monterrey, School of Engineering and Sciences, Atizapan de Zaragoza 52926, Mexico
| | - Dora I Medina
- Tecnologico de Monterrey, School of Engineering and Sciences, Atizapan de Zaragoza 52926, Mexico
| | - Mahmood Barani
- Medical Mycology and Bacteriology Research Center, Kerman University of Medical Sciences, Kerman 76169-14115, Iran
| | - Abbas Rahdar
- Department of Physics, Faculty of Science, University of Zabol, Zabol 538-98615, Iran
| | - Ghasem Sargazi
- Noncommunicable Diseases Research Center, Bam University of Medical Science, Bam 76617-71967, Iran
| | - Francesco Baino
- Department of Applied Science and Technology, Institute of Materials Physics and Engineering, Politecnico di Torino, 10129 Torino, Italy
| | - Sadanand Pandey
- Department of Chemistry, College of Natural Science, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Korea
- Particulate Matter Research Center, Research Institute of Industrial Science & Technology (RIST), 187-12, Geumho-ro, Gwangyang-si 57801, Korea
| |
Collapse
|
15
|
Translating Research for the Radiotheranostics of Nanotargeted 188Re-Liposome. Int J Mol Sci 2021; 22:ijms22083868. [PMID: 33918011 PMCID: PMC8068325 DOI: 10.3390/ijms22083868] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 12/11/2022] Open
Abstract
Nanoliposomes are one of the leading potential nano drug delivery systems capable of targeting chemotherapeutics to tumor sites because of their passive nano-targeting capability through the enhanced permeability and retention (EPR) effect for cancer patients. Recent advances in nano-delivery systems have inspired the development of a wide range of nanotargeted materials and strategies for applications in preclinical and clinical usage in the cancer field. Nanotargeted 188Re-liposome is a unique internal passive radiotheranostic agent for nuclear imaging and radiotherapeutic applications in various types of cancer. This article reviews and summarizes our multi-institute, multidiscipline, and multi-functional studied results and achievements in the research and development of nanotargeted 188Re-liposome from preclinical cells and animal models to translational clinical investigations, including radionuclide nanoliposome formulation, targeted nuclear imaging, biodistribution, pharmacokinetics, radiation dosimetry, radiation tumor killing effects in animal models, nanotargeted radionuclide and radio/chemo-combination therapeutic effects, and acute toxicity in various tumor animal models. The systemic preclinical and clinical studied results suggest 188Re-liposome is feasible and promising for in vivo passive nanotargeted radionuclide theranostics in future cancer care applications.
Collapse
|
16
|
Cabrera M, Lecot N, Fernández M, Gambini JP, Porcal W, Cabral P. 99mTc Stearyl 6-(benzylidenehydrazinyl) nicotinamide Liposomes as Tumor Permeability Evaluation Tracer. AAPS PharmSciTech 2021; 22:115. [PMID: 33763814 DOI: 10.1208/s12249-021-01984-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 03/08/2021] [Indexed: 12/13/2022] Open
Abstract
Nanomedicine is a highly demanded discipline. Liposomes have seen an increased attention due to their physicochemical properties that allow them to act as nanocarriers of drugs and also of radioisotopes that can be used to diagnose and treat cancer. In order to obtain a novel permeability cancer imaging agent based on 99mTc-labeled liposomes, we describe microwave-assisted synthesis of stearyl 6-(benzylidenehydrazinyl) nicotinamide lipid, which was included in two formulations: nanometric hydrazinonicotinic acid (HYNIC) liposome and its PEGylated coated analogue, HYNIC-PEG liposome. Radiolabeling with 99mTc via stearyl 6-(benzylidenehydrazinyl) nicotinamide was found to be easy, reproducible, and stable, revealing high radiochemical purity (94 ± 1.7%) for both liposomal formulations. Biodistribution at 4 h and 24 h and scintigraphic images at 4 h were performed in normal and melanoma-bearing C57BL/6 mice. Biodistribution studies at 4 h showed tumor uptake of 99mTc-HYNIC liposome and 99mTc-HYNIC-PEG liposome (1.1 ± 0.6 and 2.5 ± 0.4, respectively) and also at 24 h p.i. (1.8 ± 0.5 and 3.0 ± 1.1, respectively). Scintigraphic images showed appreciable tumor uptake in melanoma tumor-bearing mice with both liposomal formulations. Our results show that 99mTc stearyl 6-(benzylidenehydrazinyl) nicotinamide liposomes can be used as diagnostic noninvasive in vivo tumor-targeting agents capable of evaluating tumor permeability and development who can be used in personalized chemotherapy planning.
Collapse
|
17
|
Bi-Functional Radiotheranostics of 188Re-Liposome-Fcy-hEGF for Radio- and Chemo-Therapy of EGFR-Overexpressing Cancer Cells. Int J Mol Sci 2021; 22:ijms22041902. [PMID: 33672989 PMCID: PMC7918434 DOI: 10.3390/ijms22041902] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 12/14/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) specific therapeutics is of great importance in cancer treatment. Fcy-hEGF fusion protein, composed of yeast cytosine deaminase (Fcy) and human EGF (hEGF), is capable of binding to EGFR and enzymatically convert 5-fluorocytosine (5-FC) to 1000-fold toxic 5-fluorocuracil (5-FU), thereby inhibiting the growth of EGFR-expressing tumor cells. To develop EGFR-specific therapy, 188Re-liposome-Fcy-hEGF was constructed by insertion of Fcy-hEGF fusion protein onto the surface of liposomes encapsulating of 188Re. Western blotting, MALDI-TOF, column size exclusion and flow cytometry were used to confirm the conjugation and bio-activity of 188Re-liposome-Fcy-hEGF. Cell lines with EGFR expression were subjected to treat with 188Re-liposome-Fcy-hEGF/5-FC in the presence of 5-FC. The 188Re-liposome-Fcy-hEGF/5-FC revealed a better cytotoxic effect for cancer cells than the treatment of liposome-Fcy-hEGF/5-FC or 188Re-liposome-Fcy-hEGF alone. The therapeutics has radio- and chemo-toxicity simultaneously and specifically target to EGFR-expression tumor cells, thereby achieving synergistic anticancer activity.
Collapse
|
18
|
Delivery of the Radionuclide 131I Using Cationic Fusogenic Liposomes as Nanocarriers. Int J Mol Sci 2021; 22:ijms22010457. [PMID: 33466417 PMCID: PMC7796481 DOI: 10.3390/ijms22010457] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/30/2020] [Accepted: 12/31/2020] [Indexed: 12/12/2022] Open
Abstract
Liposomes are highly biocompatible and versatile drug carriers with an increasing number of applications in the field of nuclear medicine and diagnostics. So far, only negatively charged liposomes with intercalated radiometals, e.g., 64Cu, 99mTc, have been reported. However, the process of cellular uptake of liposomes by endocytosis is rather slow. Cellular uptake can be accelerated by recently developed cationic liposomes, which exhibit extraordinarily high membrane fusion ability. The aim of the present study was the development of the formulation and the characterization of such cationic fusogenic liposomes with intercalated radioactive [131I]I- for potential use in therapeutic applications. The epithelial human breast cancer cell line MDA-MB-231 was used as a model for invasive cancer cells and cellular uptake of [131I]I- was monitored in vitro. Delivery efficiencies of cationic and neutral liposomes were compared with uptake of free iodide. The best cargo delivery efficiency (~10%) was achieved using cationic fusogenic liposomes due to their special delivery pathway of membrane fusion. Additionally, human blood cells were also incubated with cationic control liposomes and free [131I]I-. In these cases, iodide delivery efficiencies remained below 3%.
Collapse
|
19
|
Cho E, Lu Y. Compartmentalizing Cell-Free Systems: Toward Creating Life-Like Artificial Cells and Beyond. ACS Synth Biol 2020; 9:2881-2901. [PMID: 33095011 DOI: 10.1021/acssynbio.0c00433] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Building an artificial cell is a research area that is rigorously studied in the field of synthetic biology. It has brought about much attention with the aim of ultimately constructing a natural cell-like structure. In particular, with the more mature cell-free platforms and various compartmentalization methods becoming available, achieving this aim seems not far away. In this review, we discuss the various types of artificial cells capable of hosting several cellular functions. Different compartmental boundaries and the mature and evolving technologies that are used for compartmentalization are examined, and exciting recent advances that overcome or have the potential to address current challenges are discussed. Ultimately, we show how compartmentalization and cell-free systems have, and will, come together to fulfill the goal to assemble a fully synthetic cell that displays functionality and complexity as advanced as that in nature. The development of such artificial cell systems will offer insight into the fundamental study of evolutionary biology and the sea of applications as a result. Although several challenges remain, emerging technologies such as artificial intelligence also appear to help pave the way to address them and achieve the ultimate goal.
Collapse
Affiliation(s)
- Eunhee Cho
- Key Lab of Industrial Biocatalysis, Ministry of Education, Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
| | - Yuan Lu
- Key Lab of Industrial Biocatalysis, Ministry of Education, Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
| |
Collapse
|
20
|
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: 16] [Impact Index Per Article: 3.2] [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
|
21
|
Datta P, Ray S. Nanoparticulate formulations of radiopharmaceuticals: Strategy to improve targeting and biodistribution properties. J Labelled Comp Radiopharm 2020; 63:333-355. [PMID: 32220029 DOI: 10.1002/jlcr.3839] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/17/2020] [Accepted: 03/08/2020] [Indexed: 02/06/2023]
Abstract
Application of nanotechnology principles in drug delivery has created opportunities for treatment of several diseases. Nanotechnology offers the advantage of overcoming the adverse biopharmaceutics or pharmacokinetic properties of drug molecules, to be determined by the transport properties of the particles themselves. Through the manipulation of size, shape, charge, and type of nanoparticle delivery system, variety of distribution profiles may be obtained. However, there still exists greater need to derive and standardize definitive structure property relationships for the distribution profiles of the delivery system. When applied to radiopharmaceuticals, the delivery systems assume greater significance. For the safety and efficacy of both diagnostics and therapeutic radiopharmaceuticals, selective localization in target tissue is even more important. At the same time, the synthesis and fabrication reactions of radiolabelled nanoparticles need to be completed in much shorter time. Moreover, the extensive understanding of the several interesting optical and magnetic properties of materials in nanoscale provides for achieving multiple objectives in nuclear medicine. This review discusses the various nanoparticle systems, which are applied for radionuclides and analyses the important bottlenecks that are required to be overcome for their more widespread clinical adaptation.
Collapse
Affiliation(s)
- Pallab Datta
- Centre for Healthcare Science and Technology, Indian Institute of Engineering Science and Technology Shibpur, Howrah, India
| | | |
Collapse
|
22
|
Xia Y, Xu C, Zhang X, Ning P, Wang Z, Tian J, Chen X. Liposome-based probes for molecular imaging: from basic research to the bedside. NANOSCALE 2019; 11:5822-5838. [PMID: 30888379 DOI: 10.1039/c9nr00207c] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Molecular imaging is very important in disease diagnosis and prognosis. Liposomes are excellent carriers for different types of molecular imaging probes. In this work, we summarize current developments in liposome-based probes used for molecular imaging and their applications in image-guided drug delivery and tumour surgery, including computed tomography (CT), ultrasound imaging (USI), magnetic resonance imaging (MRI), positron emission tomography (PET), fluorescence imaging (FLI) and photoacoustic imaging (PAI). We also summarized liposome-based multimodal imaging probes and new targeting strategies for liposomes. This work will offer guidance for the design of liposome-based imaging probes for future clinical applications.
Collapse
Affiliation(s)
- Yuqiong Xia
- Engineering Research Center of Molecular-imaging and Neuro-imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China.
| | | | | | | | | | | | | |
Collapse
|
23
|
Vitamin E-inspired multi-scale imaging agent. Bioorg Med Chem Lett 2019; 29:107-114. [PMID: 30459096 DOI: 10.1016/j.bmcl.2018.10.052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 09/13/2018] [Accepted: 10/31/2018] [Indexed: 12/18/2022]
Abstract
The production and use of multi-modal imaging agents is on the rise. The vast majority of these imaging agents are limited to a single length scale for the agent (e.g. tissues only), which is typically at the organ or tissue scale. This work explores the synthesis of such an imaging agent and discusses the applications of our vitamin E-inspired multi-modal and multi-length scale imaging agents TB-Toc ((S,E)-5,5-difluoro-7-(2-(5-((6-hydroxy-2,5,7,8-tetramethylchroman-2-yl) methyl) thiophen-2-yl) vinyl)-9-methyl-5H-dipyrrolo-[1,2-c:2',1'-f][1,3,2]diazaborinin-4-ium-5-uide). We investigate the toxicity of TB-Toc along with the starting materials and lipid based delivery vehicle in mouse myoblasts and fibroblasts. Further we investigate the uptake of TB-Toc delivered to cultured cells in both solvent and liposomes. TB-Toc has low toxicity, and no change in cell viability was observed up to concentrations of 10 mM. TB-Toc shows time-dependent cellular uptake that is complete in about 30 min. This work is the first step in demonstrating our vitamin E derivatives are viable multi-modal and length scale diagnostic tools.
Collapse
|
24
|
Thakur A, Rodríguez-Rodríguez C, Saatchi K, Rose F, Esposito T, Nosrati Z, Andersen P, Christensen D, Häfeli UO, Foged C. Dual-Isotope SPECT/CT Imaging of the Tuberculosis Subunit Vaccine H56/CAF01: Induction of Strong Systemic and Mucosal IgA and T-Cell Responses in Mice Upon Subcutaneous Prime and Intrapulmonary Boost Immunization. Front Immunol 2018; 9:2825. [PMID: 30555488 PMCID: PMC6284049 DOI: 10.3389/fimmu.2018.02825] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 11/15/2018] [Indexed: 12/20/2022] Open
Abstract
Pulmonary tuberculosis (TB), which is caused by Mycobacterium tuberculosis (Mtb), remains a global pandemic, despite the widespread use of the parenteral live attenuated Bacillus Calmette–Guérin (BCG) vaccine during the past decades. Mucosal administration of next generation TB vaccines has great potential, but developing a safe and efficacious mucosal vaccine is challenging. Hence, understanding the in vivo biodistribution and pharmacokinetics of mucosal vaccines is essential for shaping the desired immune response and for optimal spatiotemporal targeting of the appropriate effector cells in the lungs. A subunit vaccine consisting of the fusion antigen H56 (Ag85B-ESAT-6-Rv2660) and the liposome-based cationic adjuvant formulation (CAF01) confers efficient protection in preclinical animal models. In this study, we devise a novel immunization strategy for the H56/CAF01 vaccine, which comply with the intrapulmonary (i.pulmon.) route of immunization. We also describe a novel dual-isotope (111In/67Ga) radiolabeling approach, which enables simultaneous non-invasive and longitudinal SPECT/CT imaging and quantification of H56 and CAF01 upon parenteral prime and/or i.pulmon. boost immunization. Our results demonstrate that the vaccine is distributed evenly in the lungs, and there are pronounced differences in the pharmacokinetics of H56 and CAF01. We provide convincing evidence that the H56/CAF01 vaccine is not only well-tolerated when administered to the respiratory tract, but it also induces strong lung mucosal and systemic IgA and polyfunctional Th1 and Th17 responses after parenteral prime and i.pulmon. boost immunization. The study furthermore evaluate the application of SPECT/CT imaging for the investigation of vaccine biodistribution after parenteral and i.pulmon. immunization of mice.
Collapse
Affiliation(s)
- Aneesh Thakur
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Cristina Rodríguez-Rodríguez
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada.,Department of Physics and Astronomy, The University of British Columbia, Vancouver, BC, Canada
| | - Katayoun Saatchi
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Fabrice Rose
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tullio Esposito
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Zeynab Nosrati
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Peter Andersen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Dennis Christensen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Urs O Häfeli
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Camilla Foged
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
25
|
Wang D, Sun Y, Liu Y, Meng F, Lee RJ. Clinical translation of immunoliposomes for cancer therapy: recent perspectives. Expert Opin Drug Deliv 2018; 15:893-903. [PMID: 30169978 DOI: 10.1080/17425247.2018.1517747] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Liposomes have been extensively investigated as drug delivery vehicles. Immunoliposomes (ILs) are antibody-conjugated liposomes designed to selectively target antigen-expressing cells. ILs can be used to deliver drugs to tumor cells for improving efficacy and reducing toxicity. In addition, ILs can be used in immunoassays, immunotherapy, and imaging. Although there has been extensive coverage on ILs in the literature, only a limited number of clinical trials have been reported and no IL drug has been approved by the FDA. AREAS COVERED Factors to consider in developing ILs are discussed, including the choice of antibody or antibody fragment, the formulation of liposomes, and the conjugation chemistry. In addition, challenges and opportunities in clinical development of ILs are discussed. The purpose of this review is to provide an overview on the state of the art of ILs and to discuss potential future developments. EXPERT OPINION IL research has had a lengthy history and numerous preclinical studies have yielded encouraging results. However, there are a number of obstacles to clinical translation of ILs. Given the unique capabilities of ILs, its potential for clinical application is underexplored. There is great potential for expanded role for ILs in the clinic and further efforts to this end are warranted. ABBREVIATIONS Ab: antibody; ADCs: antibody-drug conjugates; API: active pharmaceutical ingredient; ADCC: antibody-dependent cellular cytotoxicity; CR: complete remission; cGMP: current good manufacturing practice; DSPE: distearoyl phosphatidylethanolamine; EGF: epidermal growth factor; EGFR: epidermal growth factor receptor; EPR: enhanced permeability and retention; Fc: fragment crystalline; Tf: transferrin; HACA: human-anti-chimeric antibody; HAHA: human-anti-human antibody; HAMA: human-anti-mouse antibody; HER2: human epidermal growth factor 2; IL: immunoliposome; LNPs: lipid nanoparticles; MRI: magnetic resonance imaging; MTD: maximum tolerated dose; PEG: polyethylene glycol; PET: positron emission tomography; PR: partial response; PSMA: prostate-specific membrane antigen; scFv: single-chain variable fragment; SPECT: single photon emission computed tomography; TTR: transthyretin.
Collapse
Affiliation(s)
- Di Wang
- a School of Life Sciences , Jilin University , Changchun , China
| | - Yating Sun
- a School of Life Sciences , Jilin University , Changchun , China
| | - Yange Liu
- a School of Life Sciences , Jilin University , Changchun , China
| | - Fanchao Meng
- a School of Life Sciences , Jilin University , Changchun , China
| | - Robert J Lee
- a School of Life Sciences , Jilin University , Changchun , China.,b Division of Pharmaceutics and Pharmaceutical Chemistry , The Ohio State University , Columbus , OH , USA
| |
Collapse
|
26
|
Wong P, Li L, Chea J, Delgado MK, Poku E, Szpikowska B, Bowles N, Minnix M, Colcher D, Wong JYC, Shively JE, Yazaki PJ. Synthesis, Positron Emission Tomography Imaging, and Therapy of Diabody Targeted Drug Lipid Nanoparticles in a Prostate Cancer Murine Model. Cancer Biother Radiopharm 2018; 32:247-257. [PMID: 28910151 DOI: 10.1089/cbr.2017.2253] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The blood clearance of chemotherapeutic drugs such as doxorubicin (Dox) can be extended by incorporation into lipid nanoparticles (LNPs) and further improved by tumor targeting with antibody fragments. We used positron emission tomography (PET) imaging in a murine prostate cancer model to evaluate tumor targeting of LNPs incorporating Dox and antiprostate-specific membrane antigen (PSMA) diabodies. Dox-LNPs were generated by mixing or covalent attachment to water soluble distearoylphosphatidyl ethanolamine-polyethylene glycol (DSPE-PEG)2000. Cu-64 PET imaging was performed with DOTA-conjugated Dox, PEG-LNP, or an anti-PSMA site-specific cysteine-diabody. Since the mixture Dox+PEG-LNP was unstable in serum, further studies utilized Dox covalently bound to LNP ± covalently bound DOTA-cys-diabody (cys-DB)-LNP. Blood clearance of covalent Dox-PEG-LNP was slower than Dox alone or Dox+PEG-LNP. PET imaging of 64Cu-DOTA-Dox-PEG-LNP reached a maximum of 10% ID/g in tumors compared with 3% ID/g of 64Cu-DOTA-Dox, due to the prolonged blood clearance. Mixing 64Cu-DOTA-cys-DB-PEG-LNP with covalent Dox-PEG-LNP gave LNPs containing both drug and tumor targeting cys-DB. The mixed LNPs exhibited increased tumor uptake (15% ID/g) versus untargeted 64Cu-DOTA-Dox-PEG-LNPs (10% ID/g) demonstrating feasibility of the approach. Based on these results, a therapy study with mixed LNPs containing cys-DB-LNP and either Dox-LNP or the antitubulin drug auristatin-LNP showed significant reduction of tumor growth with the auristatin-diabody-LNP mixture, but not the Dox-diabody-LNP mixture.
Collapse
Affiliation(s)
- Patty Wong
- 1 Department of Radiation Oncology, City of Hope National Medical Center , Duarte, California
| | - Lin Li
- 2 Department of Molecular Immunology, Beckman Research Institute , City of Hope, Duarte, California
| | - Junie Chea
- 2 Department of Molecular Immunology, Beckman Research Institute , City of Hope, Duarte, California
| | - Melissa K Delgado
- 2 Department of Molecular Immunology, Beckman Research Institute , City of Hope, Duarte, California
| | - Erasmus Poku
- 2 Department of Molecular Immunology, Beckman Research Institute , City of Hope, Duarte, California
| | - Barbara Szpikowska
- 2 Department of Molecular Immunology, Beckman Research Institute , City of Hope, Duarte, California
| | - Nicole Bowles
- 2 Department of Molecular Immunology, Beckman Research Institute , City of Hope, Duarte, California
| | - Megan Minnix
- 2 Department of Molecular Immunology, Beckman Research Institute , City of Hope, Duarte, California
| | - David Colcher
- 2 Department of Molecular Immunology, Beckman Research Institute , City of Hope, Duarte, California
| | - Jeffrey Y C Wong
- 1 Department of Radiation Oncology, City of Hope National Medical Center , Duarte, California
| | - John E Shively
- 2 Department of Molecular Immunology, Beckman Research Institute , City of Hope, Duarte, California
| | - Paul J Yazaki
- 2 Department of Molecular Immunology, Beckman Research Institute , City of Hope, Duarte, California
| |
Collapse
|
27
|
Belfiore L, Saunders DN, Ranson M, Thurecht KJ, Storm G, Vine KL. Towards clinical translation of ligand-functionalized liposomes in targeted cancer therapy: Challenges and opportunities. J Control Release 2018; 277:1-13. [PMID: 29501721 DOI: 10.1016/j.jconrel.2018.02.040] [Citation(s) in RCA: 191] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 02/26/2018] [Accepted: 02/27/2018] [Indexed: 01/03/2023]
Abstract
The development of therapeutic resistance to targeted anticancer therapies remains a significant clinical problem, with intratumoral heterogeneity playing a key role. In this context, improving the therapeutic outcome through simultaneous targeting of multiple tumor cell subtypes within a heterogeneous tumor is a promising approach. Liposomes have emerged as useful drug carriers that can reduce systemic toxicity and increase drug delivery to the tumor site. While clinically used liposomal drug formulations show marked therapeutic advantages over free drug formulations, ligand-functionalized liposomes that can target multiple tumor cell subtypes may further improve the therapeutic efficacy by facilitating drug delivery to a broader population of tumor cells making up the heterogeneous tumor tissue. Ligand-directed liposomes enable the so-called active targeting of cell receptors via surface-attached ligands that direct drug uptake into tumor cells or tumor-associated stromal cells, and so can increase the selectivity of drug delivery. Despite promising preclinical results demonstrating improved targeting and anti-tumor effects of ligand-directed liposomes, there has been limited translation of this approach to the clinic. Key challenges for translation include the lack of established methods to scale up production and comprehensively characterize ligand-functionalized liposome formulations, as well as the inadequate recapitulation of in vivo tumors in the preclinical models currently used to evaluate their performance. Herein, we discuss the utility of recent ligand-directed liposome approaches, with a focus on dual-ligand liposomes, for the treatment of solid tumors and examine the drawbacks limiting their progression to clinical adoption.
Collapse
Affiliation(s)
- Lisa Belfiore
- Illawarra Health and Medical Research Institute, Centre for Medical and Molecular Bioscience, School of Biological Sciences, University of Wollongong, Wollongong, Australia
| | - Darren N Saunders
- School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Marie Ranson
- Illawarra Health and Medical Research Institute, Centre for Medical and Molecular Bioscience, School of Biological Sciences, University of Wollongong, Wollongong, Australia
| | - Kristofer J Thurecht
- Australian Institute for Bioengineering and Nanotechnology (AIBN), Centre for Advanced Imaging (CAI), Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, Brisbane, Australia
| | - Gert Storm
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, CG, The Netherlands
| | - Kara L Vine
- Illawarra Health and Medical Research Institute, Centre for Medical and Molecular Bioscience, School of Biological Sciences, University of Wollongong, Wollongong, Australia.
| |
Collapse
|
28
|
Lamichhane N, Udayakumar TS, D'Souza WD, Simone CB, Raghavan SR, Polf J, Mahmood J. Liposomes: Clinical Applications and Potential for Image-Guided Drug Delivery. Molecules 2018; 23:molecules23020288. [PMID: 29385755 PMCID: PMC6017282 DOI: 10.3390/molecules23020288] [Citation(s) in RCA: 164] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 01/22/2018] [Accepted: 01/26/2018] [Indexed: 01/16/2023] Open
Abstract
Liposomes have been extensively studied and are used in the treatment of several diseases. Liposomes improve the therapeutic efficacy by enhancing drug absorption while avoiding or minimizing rapid degradation and side effects, prolonging the biological half-life and reducing toxicity. The unique feature of liposomes is that they are biocompatible and biodegradable lipids, and are inert and non-immunogenic. Liposomes can compartmentalize and solubilize both hydrophilic and hydrophobic materials. All these properties of liposomes and their flexibility for surface modification to add targeting moieties make liposomes more attractive candidates for use as drug delivery vehicles. There are many novel liposomal formulations that are in various stages of development, to enhance therapeutic effectiveness of new and established drugs that are in preclinical and clinical trials. Recent developments in multimodality imaging to better diagnose disease and monitor treatments embarked on using liposomes as diagnostic tool. Conjugating liposomes with different labeling probes enables precise localization of these liposomal formulations using various modalities such as PET, SPECT, and MRI. In this review, we will briefly review the clinical applications of liposomal formulation and their potential imaging properties.
Collapse
Affiliation(s)
- Narottam Lamichhane
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | | | - Warren D D'Souza
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Charles B Simone
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Srinivasa R Raghavan
- Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, MD 20742, USA.
| | - Jerimy Polf
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Javed Mahmood
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
29
|
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: 4.9] [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]
|
30
|
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
|
31
|
Wong P, Li L, Chea J, Delgado MK, Crow D, Poku E, Szpikowska B, Bowles N, Channappa D, Colcher D, Wong JYC, Shively JE, Yazaki PJ. PET imaging of 64Cu-DOTA-scFv-anti-PSMA lipid nanoparticles (LNPs): Enhanced tumor targeting over anti-PSMA scFv or untargeted LNPs. Nucl Med Biol 2017; 47:62-68. [PMID: 28126683 DOI: 10.1016/j.nucmedbio.2017.01.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 11/24/2016] [Accepted: 01/12/2017] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Single chain (scFv) antibodies are ideal targeting ligands due to their modular structure, high antigen specificity and affinity. These monovalent ligands display rapid tumor targeting but have limitations due to their fast urinary clearance. METHODS An anti-prostate membrane antigen (PSMA) scFv with a site-specific cysteine was expressed and evaluated in a prostate cancer xenograft model by Cu-64 PET imaging. To enhance tumor accumulation, the scFv-cys was conjugated to the co-polymer DSPE-PEG-maleimide that spontaneously assembled into a homogeneous multivalent lipid nanoparticle (LNP). RESULTS The targeted LNP exhibited a 2-fold increase in tumor uptake compared to the scFv alone using two different thiol ester chemistries. The anti-PSMA scFv-LNP exhibited a 1.6 fold increase in tumor targeting over the untargeted LNP. CONCLUSIONS The targeted anti-PSMA scFv-LNP showed enhanced tumor accumulation over the scFv alone or the untargeted DOTA-micelle providing evidence for the development of this system for drug delivery. ADVANCES IN KNOWLEDGE AND IMPLICATIONS FOR PATIENT CARE Anti-tumor scFv antibody fragments have not achieved their therapeutic potential due to their fast blood clearance. Conjugation to an LNP enables multivalency to the tumor antigen as well as increased molecular size for chemotherapy drug delivery.
Collapse
Affiliation(s)
- Patty Wong
- Department of Radiation Oncology, City of Hope Medical Center, City of Hope, Duarte, CA, 91010-3000, USA
| | - Lin Li
- Department of Molecular Immunology, Beckman Research Institute, City of Hope, Duarte, CA, 91010-3000, USA
| | - Junie Chea
- Department of Molecular Immunology, Beckman Research Institute, City of Hope, Duarte, CA, 91010-3000, USA
| | - Melissa K Delgado
- Department of Molecular Immunology, Beckman Research Institute, City of Hope, Duarte, CA, 91010-3000, USA
| | - Desiree Crow
- Department of Molecular Immunology, Beckman Research Institute, City of Hope, Duarte, CA, 91010-3000, USA
| | - Erasmus Poku
- Department of Molecular Immunology, Beckman Research Institute, City of Hope, Duarte, CA, 91010-3000, USA
| | - Barbara Szpikowska
- Department of Molecular Immunology, Beckman Research Institute, City of Hope, Duarte, CA, 91010-3000, USA
| | - Nicole Bowles
- Department of Molecular Immunology, Beckman Research Institute, City of Hope, Duarte, CA, 91010-3000, USA
| | - Divya Channappa
- Department of Molecular Immunology, Beckman Research Institute, City of Hope, Duarte, CA, 91010-3000, USA
| | - David Colcher
- Department of Molecular Immunology, Beckman Research Institute, City of Hope, Duarte, CA, 91010-3000, USA
| | - Jeffrey Y C Wong
- Department of Radiation Oncology, City of Hope Medical Center, City of Hope, Duarte, CA, 91010-3000, USA
| | - John E Shively
- Department of Molecular Immunology, Beckman Research Institute, City of Hope, Duarte, CA, 91010-3000, USA
| | - Paul J Yazaki
- Department of Molecular Immunology, Beckman Research Institute, City of Hope, Duarte, CA, 91010-3000, USA.
| |
Collapse
|