1
|
Temme S, Kleimann P, Tiren ZB, Bouvain P, Zielinski A, Dollmeyer W, Poth S, Görges J, Flögel U. Imaging of Thromboinflammation by Multispectral 19F MRI. Int J Mol Sci 2025; 26:2462. [PMID: 40141106 PMCID: PMC11942564 DOI: 10.3390/ijms26062462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/24/2025] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
The close interplay between thrombotic and immunologic processes plays an important physiological role in the immune defence after tissue injury and has the aim to reduce damage and to prevent the spread of invading pathogens. However, the uncontrolled or exaggerated activation of these processes can lead to pathological thromboinflammation. Thromboinflammation has been shown to worsen the outcome of cardiovascular, autoinflammatory, or even infectious diseases. Imaging of thromboinflammation is difficult because many clinically relevant imaging techniques can only visualize either inflammatory or thrombotic processes. One interesting option for the noninvasive imaging of thromboinflammation is multispectral 19F magnetic resonance imaging (MRI). Due to the large chemical shift range of the 19F atoms, it is possible to simultaneously visualize immune cells as well as thrombus components with specific 19F tracer that have individual spectral 19F signatures. Of note, the 19F signal can be easily quantified and a merging of the 19F datasets with the anatomical 1H MRI images enables precise anatomical localization. In this review, we briefly summarize the background of 19F MRI for inflammation imaging, active targeting approaches to visualize thrombi and specific immune cells, introduce studies about multispectral 19F MRI, and summarize one study that imaged thromboinflammation by multispectral 19F MRI.
Collapse
Affiliation(s)
- Sebastian Temme
- Department of Anesthesiology, Faculty of Medicine, University Hospital, Heinrich-Heine-University, 40225 Düsseldorf, Germany; (Z.-B.T.); (A.Z.); (W.D.); (J.G.)
| | - Patricia Kleimann
- Experimental Cardiovascular Imaging, Institute of Molecular Cardiology, Faculty of Medicine, University Hospital, Heinrich-Heine-University, 40225 Düsseldorf, Germany; (P.K.); (P.B.); (S.P.); (U.F.)
| | - Zeynep-Büsra Tiren
- Department of Anesthesiology, Faculty of Medicine, University Hospital, Heinrich-Heine-University, 40225 Düsseldorf, Germany; (Z.-B.T.); (A.Z.); (W.D.); (J.G.)
| | - Pascal Bouvain
- Experimental Cardiovascular Imaging, Institute of Molecular Cardiology, Faculty of Medicine, University Hospital, Heinrich-Heine-University, 40225 Düsseldorf, Germany; (P.K.); (P.B.); (S.P.); (U.F.)
| | - Arthur Zielinski
- Department of Anesthesiology, Faculty of Medicine, University Hospital, Heinrich-Heine-University, 40225 Düsseldorf, Germany; (Z.-B.T.); (A.Z.); (W.D.); (J.G.)
| | - William Dollmeyer
- Department of Anesthesiology, Faculty of Medicine, University Hospital, Heinrich-Heine-University, 40225 Düsseldorf, Germany; (Z.-B.T.); (A.Z.); (W.D.); (J.G.)
| | - Sarah Poth
- Experimental Cardiovascular Imaging, Institute of Molecular Cardiology, Faculty of Medicine, University Hospital, Heinrich-Heine-University, 40225 Düsseldorf, Germany; (P.K.); (P.B.); (S.P.); (U.F.)
| | - Juliana Görges
- Department of Anesthesiology, Faculty of Medicine, University Hospital, Heinrich-Heine-University, 40225 Düsseldorf, Germany; (Z.-B.T.); (A.Z.); (W.D.); (J.G.)
| | - Ulrich Flögel
- Experimental Cardiovascular Imaging, Institute of Molecular Cardiology, Faculty of Medicine, University Hospital, Heinrich-Heine-University, 40225 Düsseldorf, Germany; (P.K.); (P.B.); (S.P.); (U.F.)
| |
Collapse
|
2
|
Zimmer O, Goepferich A. On the uncertainty of the correlation between nanoparticle avidity and biodistribution. Eur J Pharm Biopharm 2024; 198:114240. [PMID: 38437906 DOI: 10.1016/j.ejpb.2024.114240] [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: 11/17/2023] [Revised: 02/05/2024] [Accepted: 02/28/2024] [Indexed: 03/06/2024]
Abstract
The specific delivery of a drug to its site of action also known as targeted drug delivery is a topic in the field of pharmaceutics studied for decades. One approach extensively investigated in this context is the use ligand functionalized nanoparticles. These particles are modified to carry receptor specific ligands, enabling them to accumulate at a desired target site. However, while this concept initially appears straightforward to implement, in-depth research has revealed several challenges hindering target site specific particle accumulation - some of which remain unresolved to this day. One of these challenges consists in the still incomplete understanding of how nanoparticles interact with biological systems. This knowledge gap significantly compromises the predictability of particle distribution in biological systems, which is critical for therapeutic efficacy. One of the most crucial steps in delivery is the attachment of nanoparticles to cells at the target site. This attachment occurs via the formation of multiple ligand receptor bonds. A process also referred to as multivalent interaction. While multivalency has been described extensively for individual molecules and macromolecules respectively, little is known on the multivalent binding of nanoparticles to cells. Here, we will specifically introduce the concept of avidity as a measure for favorable particle membrane interactions. Also, an overview about nanoparticle and membrane properties affecting avidity will be given. Thereafter, we provide a thorough review on literature investigating the correlation between nanoparticle avidity and success in targeted particle delivery. In particular, we want to analyze the currently uncertain data on the existence and nature of the correlation between particle avidity and biodistribution.
Collapse
Affiliation(s)
- Oliver Zimmer
- Department of Pharmaceutical Technology, University of Regensburg, Regensburg, Bavaria 93053, Germany
| | - Achim Goepferich
- Department of Pharmaceutical Technology, University of Regensburg, Regensburg, Bavaria 93053, Germany.
| |
Collapse
|
3
|
van Heeswijk RB, Bauer WR, Bönner F, Janjic JM, Mulder WJM, Schreiber LM, Schwitter J, Flögel U. Cardiovascular Molecular Imaging With Fluorine-19 MRI: The Road to the Clinic. Circ Cardiovasc Imaging 2023; 16:e014742. [PMID: 37725674 DOI: 10.1161/circimaging.123.014742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/21/2023]
Abstract
Fluorine-19 (19F) magnetic resonance imaging is a unique quantitative molecular imaging modality that makes use of an injectable fluorine-containing tracer that generates the only visible 19F signal in the body. This hot spot imaging technique has recently been used to characterize a wide array of cardiovascular diseases and seen a broad range of technical improvements. Concurrently, its potential to be translated to the clinical setting is being explored. This review provides an overview of this emerging field and demonstrates its diagnostic potential, which shows promise for clinical translation. We will describe 19F magnetic resonance imaging hardware, pulse sequences, and tracers, followed by an overview of cardiovascular applications. Finally, the challenges on the road to clinical translation are discussed.
Collapse
Affiliation(s)
- Ruud B van Heeswijk
- Department of Radiology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Switzerland (R.B.v.H.)
| | - Wolfgang R Bauer
- Department of Internal Medicine I, Universitätsklinikum Würzburg, Germany (W.R.B.)
| | - Florian Bönner
- Department of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty of Heinrich Heine University, University Hospital Düsseldorf, Germany (F.B.)
| | - Jelena M Janjic
- Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, PA (J.M.J.)
| | - Willem J M Mulder
- Laboratory of Chemical Biology, Department of Biochemical Engineering, Eindhoven University of Technology, the Netherlands (W.J.M.M.)
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands (W.J.M.M.)
| | - Laura M Schreiber
- Chair of Molecular and Cellular Imaging, Comprehensive Heart Failure Center (CHFC), Wuerzburg University Hospitals, Germany (L.M.S.)
| | - Juerg Schwitter
- Division of Cardiology, Cardiovascular Department (J.S.), Lausanne University Hospital (CHUV), Switzerland
- CMR Center (J.S.), Lausanne University Hospital (CHUV), Switzerland
- Faculty of Biology and Medicine, University of Lausanne (UNIL), Switzerland (J.S.)
| | - Ulrich Flögel
- Experimental Cardiovascular Imaging (U.F.), Heinrich Heine University, Germany
- Cardiovascular Research Institute Düsseldorf (CARID) (U.F.), Heinrich Heine University, Germany
| |
Collapse
|
4
|
Gao W, Lai JCK, Leung SW. Co-Culturing Rat Dorsal Root Ganglion Neurons With Rat Schwann Cells Protects Them Against the Cytotoxic Effects of Silver and Gold Nanoparticles. Int J Toxicol 2023; 42:4-18. [PMID: 36308016 DOI: 10.1177/10915818221133508] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Previous studies using monotypic nerve cell cultures have shown that nanoparticles induced neurotoxic effects on nerve cells. Interactions between neurons and Schwann cells may protect against the neurotoxicity of nanoparticles. In this study, we developed a co-culture model consisting of immortalized rat dorsal root ganglion (DRG) neurons and rat Schwann cells and employed it to investigate our hypothesis that co-culturing DRG neurons with Schwann cells imparts protection on them against neurotoxicity induced by silver or gold nanoparticles. Our results indicated that neurons survived better in co-cultures when they were exposed to these nanoparticles at the higher concentrations compared to when they were exposed to these nanoparticles at the same concentrations in monotypic cultures. Synapsin I expression was increased in DRG neurons when they were co-cultured with Schwann cells and treated with or without nanoparticles. Glial fibrillary acidic protein (GFAP) expression was increased in Schwann cells when they were co-cultured with DRG neurons and treated with nanoparticles. Furthermore, we found co-culturing with Schwann cells stimulated neurofilament polymerization in DRG neurons and produced the morphological differentiation. Silver nanoparticles induced morphological disorganization in monotypic cultures. However, there were more cells displaying normal morphology in co-cultures than in monotypic cultures. All of these results suggested that co-culturing DRG neurons with Schwann cells imparted some protection on them against neurotoxicity induced by silver or gold nanoparticles, and altering the expression of neurofilament-L, synapsin I, and GFAP could account for the phenomenon of protection in co-cultures.
Collapse
Affiliation(s)
- Wenjuan Gao
- Department of Civil & Environmental Engineering, College of Science & Engineering, 6640Idaho State University, Pocatello, ID, USA
| | - James C K Lai
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, Division of Health Sciences, 6640Idaho State University, Pocatello, ID, USA
| | - Solomon W Leung
- Department of Civil & Environmental Engineering, College of Science & Engineering, 6640Idaho State University, Pocatello, ID, USA
| |
Collapse
|
5
|
Rahiminezhad Z, Tamaddon A, Dehshahri A, Borandeh S, Abolmaali SS, Najafi H, Azarpira N. PLGA-graphene quantum dot nanocomposites targeted against α vβ 3 integrin receptor for sorafenib delivery in angiogenesis. BIOMATERIALS ADVANCES 2022; 137:212851. [PMID: 35929279 DOI: 10.1016/j.bioadv.2022.212851] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/13/2022] [Accepted: 05/09/2022] [Indexed: 06/15/2023]
Abstract
Angiogenesis is a vital step in many severe diseases such as cancer, diabetic retinopathy, and rheumatoid arthritis. Sorafenib (SFB), a multi-tyrosine kinase inhibitor, has recently been shown to inhibit tumor progression and suppress angiogenesis. Its narrow therapeutic window, however, has limited its clinical application and therapeutic efficacy. Accordingly, in this study, a nanocomposite formulation comprising of graphene quantum dots (GQDs) and poly (D, l-lactide-co-glycolide) (PLGA) nanoparticles was functionalized with an integrin-targeting ligand (RGD peptide) to improve SFB delivery for the treatment of angiogenesis. Physicochemical and biological properties of the targeted nanocomposite were evaluated in terms of chemical structure, morphology, particle size, zeta potential, photoluminescence, and cell toxicity. The loading capacity of the nanocomposite was optimized at different drug-to-PLGA ratios. Drug release behavior was also investigated at 37 °C in pH = 7.4. The SFB-to-PLGA ratio of 1:3 was selected as the optimum condition which resulted in the encapsulation efficiency and encapsulation capacity of 68.93 ± 1.39 and 18.77 ± 0.46, respectively. Photoluminescence properties of GQD in nanocomposite were used to track the delivery system. The results indicated that conjugating targeting ligand could enhance cellular uptake of nanocomposite in cells overexpressing integrin receptors. In vivo anti-angiogenesis activity of targeted nanocomposite was investigated in chick chorioallantoic membrane (CAM). The findings showed that SFB loaded in the targeted nanocomposite reduced VEGF secretion in vitro and its anti-angiogenic effect surpass free SFB. Thanks to its unique therapeutic and bioimaging properties, the developed nanocomposite could be an effective drug delivery system for poorly water-soluble therapeutic agents.
Collapse
Affiliation(s)
- Zahra Rahiminezhad
- Pharmaceutical Nanotechnology Department, School of Pharmay, Shiraz University of Medical Sciences, Shiraz 71345, Iran
| | - AliMohammad Tamaddon
- Pharmaceutical Nanotechnology Department, School of Pharmay, Shiraz University of Medical Sciences, Shiraz 71345, Iran; Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz 71345, Iran.
| | - Ali Dehshahri
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz 71345, Iran
| | - Sedigheh Borandeh
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz 71345, Iran
| | - Samira Sadat Abolmaali
- Pharmaceutical Nanotechnology Department, School of Pharmay, Shiraz University of Medical Sciences, Shiraz 71345, Iran; Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz 71345, Iran
| | - Haniyeh Najafi
- Pharmaceutical Nanotechnology Department, School of Pharmay, Shiraz University of Medical Sciences, Shiraz 71345, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
6
|
Rasool M, Malik A, Waquar S, Arooj M, Zahid S, Asif M, Shaheen S, Hussain A, Ullah H, Gan SH. New challenges in the use of nanomedicine in cancer therapy. Bioengineered 2022; 13:759-773. [PMID: 34856849 PMCID: PMC8805951 DOI: 10.1080/21655979.2021.2012907] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/26/2021] [Accepted: 11/27/2021] [Indexed: 11/02/2022] Open
Abstract
Nanomedicines are applied as alternative treatments for anticancer agents. For the treatment of cancer, due to the small size in nanometers (nm), specific site targeting can be achieved with the use of nanomedicines, increasing their bioavailability and conferring fewer toxic side effects. Additionally, the use of minute amounts of drugs can lead to cost savings. In addition, nanotechnology is effectively applied in the preparation of such drugs as they are in nm sizes, considered one of the earliest cutoff values for the production of products utilized in nanotechnology. Early concepts described gold nanoshells as one of the successful therapies for cancer and associated diseases where the benefits of nanomedicine include effective active or passive targeting. Common medicines are degraded at a higher rate, whereas the degradation of macromolecules is time-consuming. All of the discussed properties are responsible for executing the physiological behaviors occurring at the following scale, depending on the geometry. Finally, large nanomaterials based on organic, lipid, inorganic, protein, and synthetic polymers have also been utilized to develop novel cancer cures.
Collapse
Affiliation(s)
- Mahmood Rasool
- Center of Excellence in Genomic Medicine Research, Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Arif Malik
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, Pakistan
| | - Sulayman Waquar
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, Pakistan
| | - Mahwish Arooj
- University College of Medicine and Dentistry (UCMD), Lahore, Pakistan
| | - Sara Zahid
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, Pakistan
| | - Muhammad Asif
- Department of Biotechnology and ORIC, BUITEMS, Quetta, Pakistan
- Department of Biotechnology, BUITEMS, Quetta, Pakistan
| | - Sumaira Shaheen
- Centre for Research in Molecular Medicine (CRiMM), The University of Lahore, Lahore, Pakistan
| | - Abrar Hussain
- Department of Biotechnology, BUITEMS, Quetta, Pakistan
| | - Hamid Ullah
- Department of Chemistry, BUITEMS, Quetta, Pakistan
| | - Siew Hua Gan
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Malaysia
| |
Collapse
|
7
|
Nichols JM, Crelli CV, Liu L, Pham HV, Janjic JM, Shepherd AJ. Tracking macrophages in diabetic neuropathy with two-color nanoemulsions for near-infrared fluorescent imaging and microscopy. J Neuroinflammation 2021; 18:299. [PMID: 34949179 PMCID: PMC8697472 DOI: 10.1186/s12974-021-02365-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 12/17/2021] [Indexed: 02/08/2023] Open
Abstract
Background The incidence of diabetes and diabetic peripheral neuropathy continues to rise, and studies have shown that macrophages play an important role in their pathogenesis. To date, macrophage tracking has largely been achieved using genetically-encoded fluorescent proteins. Here we present a novel two-color fluorescently labeled perfluorocarbon nanoemulsion (PFC-NE) designed to monitor phagocytic macrophages in diabetic neuropathy in vitro and in vivo using non-invasive near-infrared fluorescent (NIRF) imaging and fluorescence microscopy. Methods Presented PFC-NEs were formulated with perfluorocarbon oil surrounded by hydrocarbon shell carrying two fluorescent dyes and stabilized with non-ionic surfactants. In vitro assessment of nanoemulsions was performed by measuring fluorescent signal stability, colloidal stability, and macrophage uptake and subsequent viability. The two-color PFC-NE was administered to Leprdb/db and wild-type mice by tail vein injection, and in vivo tracking of the nanoemulsion was performed using both NIRF imaging and confocal microscopy to assess its biodistribution within phagocytic macrophages along the peripheral sensory apparatus of the hindlimb. Results In vitro experiments show two-color PFC-NE demonstrated high fluorescent and colloidal stability, and that it was readily incorporated into RAW 264.7 macrophages. In vivo tracking revealed distribution of the two-color nanoemulsion to macrophages within most tissues of Leprdb/db and wild-type mice which persisted for several weeks, however it did not cross the blood brain barrier. Reduced fluorescence was seen in sciatic nerves of both Leprdb/db and wild-type mice, implying that the nanoemulsion may also have difficulty crossing an intact blood nerve barrier. Additionally, distribution of the nanoemulsion in Leprdb/db mice was reduced in several tissues as compared to wild-type mice. This reduction in biodistribution appears to be caused by the increased number of adipose tissue macrophages in Leprdb/db mice. Conclusions The nanoemulsion in this study has the ability to identify phagocytic macrophages in the Leprdb/db model using both NIRF imaging and fluorescence microscopy. Presented nanoemulsions have the potential for carrying lipophilic drugs and/or fluorescent dyes, and target inflammatory macrophages in diabetes. Therefore, we foresee these agents becoming a useful tool in both imaging inflammation and providing potential treatment in diabetic peripheral neuropathy.
Collapse
Affiliation(s)
- James M Nichols
- Division of Internal Medicine, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd., Houston, TX, 77030, USA
| | - Caitlin V Crelli
- School of Pharmacy, Duquesne University, 600 Forbes Ave., Pittsburgh, PA, 15282, USA
| | - Lu Liu
- School of Pharmacy, Duquesne University, 600 Forbes Ave., Pittsburgh, PA, 15282, USA
| | - Hoang Vu Pham
- Division of Internal Medicine, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd., Houston, TX, 77030, USA
| | - Jelena M Janjic
- School of Pharmacy, Duquesne University, 600 Forbes Ave., Pittsburgh, PA, 15282, USA.
| | - Andrew J Shepherd
- Division of Internal Medicine, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd., Houston, TX, 77030, USA.
| |
Collapse
|
8
|
Bouvain P, Temme S, Flögel U. Hot spot 19 F magnetic resonance imaging of inflammation. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 12:e1639. [PMID: 32380579 DOI: 10.1002/wnan.1639] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 03/20/2020] [Accepted: 04/01/2020] [Indexed: 12/11/2022]
Abstract
Among the preclinical molecular imaging approaches, lately fluorine (19 F) magnetic resonance imaging (MRI) has garnered significant scientific interest in the biomedical research community, due to the unique properties of fluorinated materials and the 19 F nucleus. Fluorine is an intrinsically sensitive nucleus for MRI-there is negligible endogenous 19 F in the body and, thus, no background signal which allows the detection of fluorinated materials as "hot spots" by combined 1 H/19 F MRI and renders fluorine-containing molecules as ideal tracers with high specificity. In addition, perfluorocarbons are a family of compounds that exhibit a very high fluorine payload and are biochemically as well as physiologically inert. Perfluorocarbon nanoemulsions (PFCs) are well known to be readily taken up by immunocompetent cells, which can be exploited for the unequivocal identification of inflammatory foci by tracking the recruitment of PFC-loaded immune cells to affected tissues using 1 H/19 F MRI. The required 19 F labeling of immune cells can be accomplished either ex vivo by PFC incubation of isolated endogenous immune cells followed by their re-injection or by intravenous application of PFCs for in situ uptake by circulating immune cells. With both approaches, inflamed tissues can unambiguously be detected via background-free 19 F signals due to trafficking of PFC-loaded immune cells to affected organs. To extend 19 F MRI tracking beyond cells with phagocytic properties, the PFC surface can further be equipped with distinct ligands to generate specificity against epitopes and/or types of immune cells independent of phagocytosis. Recent developments also allow for concurrent detection of different PFCs with distinct spectral signatures allowing the simultaneous visualization of several targets, such as various immune cell subtypes labeled with these PFCs. Since ligands and targets can easily be adapted to a variety of problems, this approach provides a general and versatile platform for inflammation imaging which will strongly extend the frontiers of molecular MRI. This article is categorized under: Diagnostic Tools > in vivo Nanodiagnostics and Imaging Therapeutic Approaches and Drug Discovery > Emerging Technologies Therapeutic Approaches and Drug Discovery > Nanomedicine for Cardiovascular Disease.
Collapse
Affiliation(s)
- Pascal Bouvain
- Experimental Cardiovascular Imaging, Molecular Cardiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Sebastian Temme
- Experimental Cardiovascular Imaging, Molecular Cardiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Ulrich Flögel
- Experimental Cardiovascular Imaging, Molecular Cardiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
9
|
Wu PH, Opadele AE, Onodera Y, Nam JM. Targeting Integrins in Cancer Nanomedicine: Applications in Cancer Diagnosis and Therapy. Cancers (Basel) 2019; 11:E1783. [PMID: 31766201 PMCID: PMC6895796 DOI: 10.3390/cancers11111783] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 11/11/2019] [Accepted: 11/12/2019] [Indexed: 02/08/2023] Open
Abstract
Due to advancements in nanotechnology, the application of nanosized materials (nanomaterials) in cancer diagnostics and therapeutics has become a leading area in cancer research. The decoration of nanomaterial surfaces with biological ligands is a major strategy for directing the actions of nanomaterials specifically to cancer cells. These ligands can bind to specific receptors on the cell surface and enable nanomaterials to actively target cancer cells. Integrins are one of the cell surface receptors that regulate the communication between cells and their microenvironment. Several integrins are overexpressed in many types of cancer cells and the tumor microvasculature and function in the mediation of various cellular events. Therefore, the surface modification of nanomaterials with integrin-specific ligands not only increases their binding affinity to cancer cells but also enhances the cellular uptake of nanomaterials through the intracellular trafficking of integrins. Moreover, the integrin-specific ligands themselves interfere with cancer migration and invasion by interacting with integrins, and this finding provides a novel direction for new treatment approaches in cancer nanomedicine. This article reviews the integrin-specific ligands that have been used in cancer nanomedicine and provides an overview of the recent progress in cancer diagnostics and therapeutic strategies involving the use of integrin-targeted nanomaterials.
Collapse
Affiliation(s)
- Ping-Hsiu Wu
- Global Station for Quantum Medical Science and Engineering, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo 060-8638, Hokkaido, Japan
| | - Abayomi Emmanuel Opadele
- Molecular and Cellular Dynamics Research, Graduate School of Biomedical Science and Engineering, Hokkaido University, Sapporo 060-8638, Hokkaido, Japan;
| | - Yasuhito Onodera
- Global Station for Quantum Medical Science and Engineering, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo 060-8638, Hokkaido, Japan
- Department of Molecular Biology, Faculty of Medicine, Hokkaido University, Sapporo 060-8638, Hokkaido, Japan
| | - Jin-Min Nam
- Global Station for Quantum Medical Science and Engineering, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo 060-8638, Hokkaido, Japan
| |
Collapse
|
10
|
Immunological considerations and concerns as pertinent to whole eye transplantation. Curr Opin Organ Transplant 2019; 24:726-732. [PMID: 31689262 DOI: 10.1097/mot.0000000000000713] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
PURPOSE OF REVIEW The advent of clinical vascularized composite allotransplantation (VCA), offers hope for whole eye transplantation (WET) in patients with devastating vison loss that fails or defies current treatment options. Optic nerve regeneration and reintegration remain the overarching hurdles to WET. However, the realization of WET may indeed be limited by our lack of understanding of the singular immunological features of the eye as pertinent to graft survival and functional vision restoration in the setting of transplantation. RECENT FINDINGS Like other VCA, such as the hand or face, the eye includes multiple tissues with distinct embryonic lineage and differential antigenicity. The ultimate goal of vision restoration through WET requires optimal immune modulation of the graft for successful optic nerve regeneration. Our team is exploring barriers to our understanding of the immunology of the eye in the context of WET including the role of immune privilege and lymphatic drainage on rejection, as well as the effects ischemia, reperfusion injury and rejection on optic nerve regeneration. SUMMARY Elucidation of the unique immunological responses in the eye and adnexa after WET will provide foundational clues that will help inform therapies that prevent immune rejection without hindering optic nerve regeneration or reintegration.
Collapse
|
11
|
Delivery of Conjugated Silicon Dioxide Nanoparticles Show Strong Anti-Proliferative Activities. Appl Biochem Biotechnol 2019; 189:760-773. [PMID: 31119527 DOI: 10.1007/s12010-019-03030-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 04/22/2019] [Indexed: 12/29/2022]
Abstract
Conjugation of different molecules is a promising approach to enhance the drug delivery and treatment. In the present study, here, we have synthesized silica oxide (SiO2) nanoparticles conjugated with (3-Glycidyloxypropyl) trimethoxysilane (3GPS) and further reacted with 1,2,4-triazole (Tri), 3-aminotriazole (ATri), 5-aminetetrazole (Atet), imidazole (Imi). The structure, size, and morphology of nanocomposite materials were characterized by Fourier transform infrared spectroscopy (FTIR), thermogravimetric analysis (TGA), transmission electron microscopy (TEM), and scanning electron microscopy (SEM) methods. These nanocomposite materials were tested on human colorectal carcinoma cells (HCT-116) to examine their anti-cancer capabilities by using MTT assay and morphometric analysis. Our results revealed that nanocomposite materials decreased cancer cell viability and cell proliferation and caused cell death in a concentration-dependent manner. Our findings demonstrate that SiO2-conjugated nanocomposite materials possess strong anti-cancer capabilities and hold a great potential for the colon cancer treatments.
Collapse
|
12
|
Avitabile E, Bedognetti D, Ciofani G, Bianco A, Delogu LG. How can nanotechnology help the fight against breast cancer? NANOSCALE 2018; 10:11719-11731. [PMID: 29917035 DOI: 10.1039/c8nr02796j] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
In this review we provide a broad overview on the use of nanotechnology for the fight against breast cancer (BC). Nowadays, detection, diagnosis, treatment, and prevention may be possible thanks to the application of nanotechnology to clinical practice. Taking into consideration the different forms of BC and the disease status, nanomaterials can be designed to meet the most forefront objectives of modern therapy and diagnosis. We have analyzed in detail three main groups of nanomaterial applications for BC treatment and diagnosis. We have identified several types of drugs successfully conjugated with nanomaterials. We have analyzed the main important imaging techniques and all nanomaterials used to help the non-invasive, early detection of the lesions. Moreover, we have examined theranostic nanomaterials as unique tools, combining imaging, detection, and therapy for BC. This state of the art review provides a useful guide depicting how nanotechnology can be used to overcome the current barriers in BC clinical practice, and how it will shape the future scenario of treatments, prevention, and diagnosis, revolutionizing the current approaches, e.g., reducing the suffering related to chemotherapy.
Collapse
Affiliation(s)
- Elisabetta Avitabile
- Department of Chemistry and Pharmacy, University of Sassari, Via Vienna 2, 07100 Sassari, Italy.
| | | | | | | | | |
Collapse
|
13
|
Wan H, Yue J, Zhu S, Uno T, Zhang X, Yang Q, Yu K, Hong G, Wang J, Li L, Ma Z, Gao H, Zhong Y, Su J, Antaris AL, Xia Y, Luo J, Liang Y, Dai H. A bright organic NIR-II nanofluorophore for three-dimensional imaging into biological tissues. Nat Commun 2018; 9:1171. [PMID: 29563581 PMCID: PMC5862886 DOI: 10.1038/s41467-018-03505-4] [Citation(s) in RCA: 296] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 02/16/2018] [Indexed: 12/22/2022] Open
Abstract
Fluorescence imaging of biological systems in the second near-infrared (NIR-II, 1000-1700 nm) window has shown promise of high spatial resolution, low background, and deep tissue penetration owing to low autofluorescence and suppressed scattering of long wavelength photons. Here we develop a bright organic nanofluorophore (named p-FE) for high-performance biological imaging in the NIR-II window. The bright NIR-II >1100 nm fluorescence emission from p-FE affords non-invasive in vivo tracking of blood flow in mouse brain vessels. Excitingly, p-FE enables one-photon based, three-dimensional (3D) confocal imaging of vasculatures in fixed mouse brain tissue with a layer-by-layer imaging depth up to ~1.3 mm and sub-10 µm high spatial resolution. We also perform in vivo two-color fluorescence imaging in the NIR-II window by utilizing p-FE as a vasculature imaging agent emitting between 1100 and 1300 nm and single-walled carbon nanotubes (CNTs) emitting above 1500 nm to highlight tumors in mice.
Collapse
Affiliation(s)
- Hao Wan
- Department of Materials Science and Engineering, South University of Science and Technology of China, 518055, Shenzhen, China.,Department of Chemistry, Stanford University, Stanford, CA, 94305, USA
| | - Jingying Yue
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA
| | - Shoujun Zhu
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA
| | - Takaaki Uno
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA.,JSR Corporation, Advanced Materials Research Laboratories, 100 Kawajiri-Cho, Yokkaichi, Mie, 5108552, Japan
| | - Xiaodong Zhang
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Sciences, Tianjin University, 300350, Tianjin, China
| | - Qinglai Yang
- Department of Materials Science and Engineering, South University of Science and Technology of China, 518055, Shenzhen, China.,Department of Chemistry, Tsinghua University, 100084, Beijing, China
| | - Kuai Yu
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA
| | - Guosong Hong
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA
| | - Junying Wang
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Sciences, Tianjin University, 300350, Tianjin, China
| | - Lulin Li
- Palo Alto Veterans Institute for Research, VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Zhuoran Ma
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA
| | - Hongpeng Gao
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA
| | - Yeteng Zhong
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA
| | - Jessica Su
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA
| | | | - Yan Xia
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA
| | - Jian Luo
- Palo Alto Veterans Institute for Research, VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA.,Department of Neurology and Neurological Sciences, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Yongye Liang
- Department of Materials Science and Engineering, South University of Science and Technology of China, 518055, Shenzhen, China.
| | - Hongjie Dai
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
14
|
Leung SW, Williams B, De Jesus K, Lai JC. Critical Review of Removal of Nano Materials in Waste Streams. ACTA ACUST UNITED AC 2017. [DOI: 10.1088/1755-1315/68/1/012019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
15
|
Tetraiodothyroacetic acid-conjugated polyethylenimine for integrin receptor mediated delivery of the plasmid encoding IL-12 gene. Colloids Surf B Biointerfaces 2017; 150:426-436. [DOI: 10.1016/j.colsurfb.2016.11.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 10/23/2016] [Accepted: 11/03/2016] [Indexed: 11/18/2022]
|
16
|
Zhang L, Navaratna T, Thurber GM. A Helix-Stabilizing Linker Improves Subcutaneous Bioavailability of a Helical Peptide Independent of Linker Lipophilicity. Bioconjug Chem 2016; 27:1663-72. [PMID: 27327034 DOI: 10.1021/acs.bioconjchem.6b00209] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Stabilized peptides address several limitations to peptide-based imaging agents and therapeutics such as poor stability and low affinity due to conformational flexibility. There is also active research in developing these compounds for intracellular drug targeting, and significant efforts have been invested to determine the effects of helix stabilization on intracellular delivery. However, much less is known about the impact on other pharmacokinetic parameters such as plasma clearance and bioavailability. We investigated the effect of different fluorescent helix-stabilizing linkers with varying lipophilicity on subcutaneous (sc) bioavailability using the glucagon-like peptide-1 (GLP-1) receptor ligand exendin as a model system. The stabilized peptides showed significantly higher protease resistance and increased bioavailability independent of linker hydrophilicity, and all subcutaneously delivered conjugates were able to successfully target the islets of Langerhans with high specificity. The lipophilic peptide variants had slower absorption and plasma clearance than their respective hydrophilic conjugates, and the absolute bioavailability was also lower likely due to the longer residence times in the skin. Their ease and efficiency make double-click helix stabilization chemistries a useful tool for increasing the bioavailability of peptide therapeutics, many of which suffer from rapid in vivo protease degradation. Helix stabilization using linkers of varying lipophilicity can further control sc absorption and clearance rates to customize plasma pharmacokinetics.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Chemical Engineering, and ‡Department of Biomedical Engineering University of Michigan , Ann Arbor, Michigan 48109, United States
| | - Tejas Navaratna
- Department of Chemical Engineering, and ‡Department of Biomedical Engineering University of Michigan , Ann Arbor, Michigan 48109, United States
| | - Greg M Thurber
- Department of Chemical Engineering, and ‡Department of Biomedical Engineering University of Michigan , Ann Arbor, Michigan 48109, United States
| |
Collapse
|
17
|
Approaching a Unified Theory for Particle-Induced Inflammation. CURRENT TOPICS IN ENVIRONMENTAL HEALTH AND PREVENTIVE MEDICINE 2016. [DOI: 10.1007/978-4-431-55732-6_3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
18
|
Zhang W, Lian K, Yang F, Yang Y, Zhu Z, Zhu Z, Cao W, Mao R, Jin Y, He J, Guo J, Liu X, Zheng H. Establishment and evaluation of a murine ανβ3-integrin-expressing cell line with increased susceptibility to Foot-and-mouth disease virus. J Vet Sci 2015; 16:265-72. [PMID: 25643796 PMCID: PMC4588011 DOI: 10.4142/jvs.2015.16.3.265] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 01/29/2015] [Indexed: 11/20/2022] Open
Abstract
Integrin ανβ3 plays a major role in various signaling pathways, cell apoptosis, and tumor angiogenesis. To examine the functions and roles of ανβ3 integrin, a stable CHO-677 cell line expressing the murine ανβ3 heterodimer (designated as "CHO-677-mανβ3" cells) was established using a highly efficient lentiviral-mediated gene transfer technique. Integrin subunits αν and β3 were detected at the gene and protein levels by polymerase chain reaction (PCR) and indirect immunofluorescent assay (IFA), respectively, in the CHO-677-mανβ3 cell line at the 20th passage, implying that these genes were successfully introduced into the CHO-677 cells and expressed stably. A plaque-forming assay, 50% tissue culture infective dose (TCID50), real-time quantitative reverse transcription-PCR, and IFA were used to detect the replication levels of Foot-and-mouth disease virus (FMDV) in the CHO-677-mανβ3 cell line. After infection with FMDV/O/ZK/93, the cell line showed a significant increase in viral RNA and protein compared with CHO-677 cells. These findings suggest that we successfully established a stable ανβ3-receptor-expressing cell line with increased susceptibility to FMDV. This cell line will be very useful for further investigation of ανβ3 integrin, and as a cell model for FMDV research.
Collapse
Affiliation(s)
- Wei Zhang
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Zhang L, Navaratna T, Liao J, Thurber GM. Dual-purpose linker for alpha helix stabilization and imaging agent conjugation to glucagon-like peptide-1 receptor ligands. Bioconjug Chem 2015; 26:329-37. [PMID: 25594741 DOI: 10.1021/bc500584t] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Peptides display many characteristics of efficient imaging agents such as rapid targeting, fast background clearance, and low non-specific cellular uptake. However, poor stability, low affinity, and loss of binding after labeling often preclude their use in vivo. Using glucagon-like peptide-1 receptor (GLP-1R) ligands exendin and GLP-1 as a model system, we designed a novel α-helix-stabilizing linker to simultaneously address these limitations. The stabilized and labeled peptides showed an increase in helicity, improved protease resistance, negligible loss or an improvement in binding affinity, and excellent in vivo targeting. The ease of incorporating azidohomoalanine in peptides and efficient reaction with the dialkyne linker enable this technique to potentially be used as a general method for labeling α helices. This strategy should be useful for imaging beta cells in diabetes research and in developing and testing other peptide targeting agents.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Chemical Engineering, ‡Department of Biomedical Engineering, University of Michigan , Ann Arbor, Michigan 48109, United States
| | | | | | | |
Collapse
|
20
|
Balducci A, Wen Y, Zhang Y, Helfer BM, Hitchens TK, Meng WS, Wesa AK, Janjic JM. A novel probe for the non-invasive detection of tumor-associated inflammation. Oncoimmunology 2014; 2:e23034. [PMID: 23526711 PMCID: PMC3601170 DOI: 10.4161/onci.23034] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
A novel dual-mode contrast agent was formulated through the addition of an optical near infrared (NIR) probe to a perfluorocarbon (PFC)-based 19F magnetic resonance imaging (MRI) agent, which labels inflammatory cells in situ. A single PFC-NIR imaging agent enables both a qualitative, rapid optical monitoring of an inflammatory state and a quantitative, detailed and tissue-depth independent magnetic resonance imaging (MRI). The feasibility of in vivo optical imaging of the inflammatory response was demonstrated in a subcutaneous murine breast carcinoma model. Ex vivo optical imaging was used to quantify the PFC-NIR signal in the tumor and organs, and results correlated well with quantitative 19F NMR analyses of intact tissues. 19F MRI was employed to construct a three-dimensional image of the cellular microenvironment at the tumor site. Flow cytometry of isolated tumor cells was used to identify the cellular localization of the PFC-NIR probe within the tumor microenvironment. Contrast is achieved through the labeling of host cells involved in the immune response, but not tumor cells. The major cellular reservoir of the imaging agent were tumor-infiltrating CD11b+ F4/80low Gr-1low cells, a cell subset sharing immunophenotypic features with myeloid-derived suppressor cells (MDSCs). These cells are recruited to sites of inflammation and are implicated in immune evasion and tumor progression. This PFC-NIR contrast agent coupled to non-invasive, quantitative imaging techniques could serve as a valuable tool for evaluating novel anticancer agents.
Collapse
Affiliation(s)
- Anthony Balducci
- Department of Research and Development; Celsense, Inc.; Pittsburgh, PA USA
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Winter PM. Perfluorocarbon nanoparticles: evolution of a multimodality and multifunctional imaging agent. SCIENTIFICA 2014; 2014:746574. [PMID: 25024867 PMCID: PMC4082945 DOI: 10.1155/2014/746574] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2013] [Accepted: 05/20/2014] [Indexed: 06/03/2023]
Abstract
Perfluorocarbon nanoparticles offer a biologically inert, highly stable, and nontoxic platform that can be specifically designed to accomplish a range of molecular imaging and drug delivery functions in vivo. The particle surface can be decorated with targeting ligands to direct the agent to a variety of biomarkers that are associated with diseases such as cancer, cardiovascular disease, obesity, and thrombosis. The surface can also carry a high payload of imaging agents, ranging from paramagnetic metals for MRI, radionuclides for nuclear imaging, iodine for CT, and florescent tags for histology, allowing high sensitivity mapping of cellular receptors that may be expressed at very low levels in the body. In addition to these diagnostic imaging applications, the particles can be engineered to carry highly potent drugs and specifically deposit them into cell populations that display biosignatures of a variety of diseases. The highly flexible and robust nature of this combined molecular imaging and drug delivery vehicle has been exploited in a variety of animal models to demonstrate its potential impact on the care and treatment of patients suffering from some of the most debilitating diseases.
Collapse
Affiliation(s)
- Patrick M. Winter
- Department of Radiology, Cincinnati Children's Hospital, Cincinnati, OH 45229, USA
| |
Collapse
|
22
|
Janjic JM, Shao P, Zhang S, Yang X, Patel SK, Bai M. Perfluorocarbon nanoemulsions with fluorescent, colloidal and magnetic properties. Biomaterials 2014; 35:4958-68. [PMID: 24674463 DOI: 10.1016/j.biomaterials.2014.03.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 03/03/2014] [Indexed: 10/25/2022]
Abstract
Bimodal imaging agents that combine magnetic resonance imaging (MRI) and nearinfrared (NIR) imaging formulated as nanoemulsions became increasingly popular for imaging inflammation in vivo. Quality of in vivo imaging using nanoemulsions is directly dependent on their integrity and stability. Here we report the design of nanoemulsions for bimodal imaging, where both photostability and colloidal stability are equally addressed. A highly chemically and photo stable quaterrylenediimide dye was introduced into perfluoro-15-crown-5 ether (PCE) nanoemulsions. The nanoemulsions were prepared with PCE and Miglyol 812N mixed at 1:1 v/v ratio as internal phase stabilized by non-ionic surfactants. Data shows exceptional colloidal stability demonstrated as unchanged droplet size (~130 nm) and polydispersity (<0.15) after 182 days follow up at both 4 and 25 °C. Nanoemulsions also sustained the exposure to mechanical and temperature stress, and prolonged exposure to light without changes in droplet size, (19)F signal or fluorescence signal. No toxicity was observed in vitro in model inflammatory cells upon 24 h exposure while confocal microscopy showed that nanoemulsions droplets accumulated in the cytoplasm. Overall, our data demonstrates that design of bimodal imaging agents requires consideration of stability of each imaging component and that of the nanosystem as a whole to achieve excellent imaging performance.
Collapse
Affiliation(s)
- Jelena M Janjic
- Graduate School of Pharmaceutical Sciences, Mylan School of Pharmacy, Duquesne University, Pittsburgh, PA 15282, USA.
| | - Pin Shao
- Molecular Imaging Laboratory, Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Shaojuan Zhang
- Molecular Imaging Laboratory, Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; Department of Diagnostic Radiology, The First Hospital of Medical School, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Xun Yang
- Bayer School of Natural and Environmental Sciences, Duquesne University, Pittsburgh, PA 15282, USA
| | - Sravan K Patel
- Graduate School of Pharmaceutical Sciences, Mylan School of Pharmacy, Duquesne University, Pittsburgh, PA 15282, USA
| | - Mingfeng Bai
- Molecular Imaging Laboratory, Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA; University of Pittsburgh Cancer Institute, Pittsburgh, PA 15232, USA.
| |
Collapse
|
23
|
Wang S, Zhang J, Chen M, Wang Y. Delivering flavonoids into solid tumors using nanotechnologies. Expert Opin Drug Deliv 2013; 10:1411-28. [PMID: 23862581 DOI: 10.1517/17425247.2013.807795] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Long-term epidemiological studies have demonstrated that regular ingestion of flavonoids contained in dietary sources is associated with a reduced risk for many chronic diseases including cancer. However, although flavonoids are largely consumed in the diet and high concentrations may exist in the intestine after oral administration, the plasma/tissue concentrations of flavonoids are lower than their effective therapeutic doses due to poor bioavailability, resulting in the limited efficacy of flavonoids in various clinical studies. Therefore, the application of nanotechnology to deliver flavonoids to tumor sites has received considerable attention in recent years. AREAS COVERED In this review, after a general review of the potential benefits of flavonoids in cancer therapy and several key factors affecting their bioavailability, the current efforts in improving the delivery efficacy of promising candidates that are particularly important in the human diet, namely quercetin, epigallocatechin-3-gallate (EGCG) and genistein were focused on. Finally, the challenges of developing flavonoid delivery systems that improve flavonoid bioavailability and their anticancer therapy potentials were summarized. EXPERT OPINION The design of suitable molecular carriers for flavonoids is an area of research that is in rapid progress. A large number of unheeded promising favonoids are suffering from poor in vivo parameters, their potential benefits deserves further research. Furthermore, more effort should be placed on developing active targeting systems, evaluating the efficacy and toxicity of novel flavonoid delivery systems through small and large scale clinical trials.
Collapse
Affiliation(s)
- Shengpeng Wang
- Assistant Professor, University of Macau, Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine , Av. Padre Tomas Pereira S.J, Taipa, Macau, 999078 , China
| | | | | | | |
Collapse
|
24
|
Tumor angiogenesis phenotyping by nanoparticle-facilitated magnetic resonance and near-infrared fluorescence molecular imaging. Neoplasia 2013; 14:964-73. [PMID: 23097630 DOI: 10.1593/neo.121148] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Revised: 08/29/2012] [Accepted: 08/29/2012] [Indexed: 12/21/2022] Open
Abstract
One of the challenges of tailored antiangiogenic therapy is the ability to adequately monitor the angiogenic activity of a malignancy in response to treatment. The α(v)β(3) integrin, highly overexpressed on newly formed tumor vessels, has been successfully used as a target for Arg-Gly-Asp (RGD)-functionalized nanoparticle contrast agents. In the present study, an RGD-functionalized nanocarrier was used to image ongoing angiogenesis in two different xenograft tumor models with varying intensities of angiogenesis (LS174T > EW7). To that end, iron oxide nanocrystals were included in the core of the nanoparticles to provide contrast for T(2)*-weighted magnetic resonance imaging (MRI), whereas the fluorophore Cy7 was attached to the surface to enable near-infrared fluorescence (NIRF) imaging. The mouse tumor models were used to test the potential of the nanoparticle probe in combination with dual modality imaging for in vivo detection of tumor angiogenesis. Pre-contrast and post-contrast images (4 hours) were acquired at a 9.4-T MRI system and revealed significant differences in the nanoparticle accumulation patterns between the two tumor models. In the case of the highly vascularized LS174T tumors, the accumulation was more confined to the periphery of the tumors, where angiogenesis is predominantly occurring. NIRF imaging revealed significant differences in accumulation kinetics between the models. In conclusion, this technology can serve as an in vivo biomarker for antiangiogenesis treatment and angiogenesis phenotyping.
Collapse
|
25
|
Balducci A, Wen Y, Zhang Y, Helfer BM, Hitchens TK, Meng WS, Wesa AK, Janjic JM. A novel probe for the non-invasive detection of tumor-associated inflammation. Oncoimmunology 2013; 2:e23034. [PMID: 23526711 DOI: 10.4161/onci] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2023] Open
Abstract
A novel dual-mode contrast agent was formulated through the addition of an optical near infrared (NIR) probe to a perfluorocarbon (PFC)-based 19F magnetic resonance imaging (MRI) agent, which labels inflammatory cells in situ. A single PFC-NIR imaging agent enables both a qualitative, rapid optical monitoring of an inflammatory state and a quantitative, detailed and tissue-depth independent magnetic resonance imaging (MRI). The feasibility of in vivo optical imaging of the inflammatory response was demonstrated in a subcutaneous murine breast carcinoma model. Ex vivo optical imaging was used to quantify the PFC-NIR signal in the tumor and organs, and results correlated well with quantitative 19F NMR analyses of intact tissues. 19F MRI was employed to construct a three-dimensional image of the cellular microenvironment at the tumor site. Flow cytometry of isolated tumor cells was used to identify the cellular localization of the PFC-NIR probe within the tumor microenvironment. Contrast is achieved through the labeling of host cells involved in the immune response, but not tumor cells. The major cellular reservoir of the imaging agent were tumor-infiltrating CD11b+ F4/80low Gr-1low cells, a cell subset sharing immunophenotypic features with myeloid-derived suppressor cells (MDSCs). These cells are recruited to sites of inflammation and are implicated in immune evasion and tumor progression. This PFC-NIR contrast agent coupled to non-invasive, quantitative imaging techniques could serve as a valuable tool for evaluating novel anticancer agents.
Collapse
Affiliation(s)
- Anthony Balducci
- Department of Research and Development; Celsense, Inc.; Pittsburgh, PA USA
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Gustafson TP, Dergunov SA, Akers WJ, Cao Q, Magalotti S, Achilefu S, Pinkhassik E, Berezin MY. BLOOD TRIGGERED RAPID RELEASE POROUS NANOCAPSULES. RSC Adv 2013; 3:5547-5555. [PMID: 23606942 PMCID: PMC3627417 DOI: 10.1039/c3ra22693j] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Rapid-release drug delivery systems present a new paradigm in emergency care treatments. Such systems combine a long shelf life with the ability to provide a significant dose of the drug to the bloodstream in the shortest period of time. Until now, development of delivery formulations has concentrated on slow release systems to ensure a steady concentration of the drug. To address the need for quick release system, we created hollow polyacrylate nanocapsules with nanometer-thin porous walls. Burst release occurs upon interaction with blood components that leads to escape of the cargo. The likely mechanism of release involves a conformational change of the polymer shell caused by binding albumin. To demonstrate this concept, a near-infrared fluorescent dye indocyanine green (ICG) was incorporated inside the nanocapsules. ICG-loaded nanocapsules demonstrated remarkable shelf life in aqueous buffers with no release of ICG for twelve months. Rapid release of the dye was demonstrated first in vitro using albumin solution and serum. SEM and light scattering analysis demonstrated the retention of the nanocapsule architecture after the release of the dye upon contact with albumin. In vivo studies using fluorescence lifetime imaging confirmed quick discharge of ICG from the nanocapsules following intravenous injection.
Collapse
Affiliation(s)
- Tiffany P Gustafson
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Hernández-Ortiz M, Acosta-Torres LS, Hernández-Padrón G, Mendieta AI, Bernal R, Cruz-Vázquez C, Castaño VM. Biocompatibility of crystalline opal nanoparticles. Biomed Eng Online 2012; 11:78. [PMID: 23088559 PMCID: PMC3549902 DOI: 10.1186/1475-925x-11-78] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Accepted: 10/01/2012] [Indexed: 03/09/2023] Open
Abstract
BACKGROUND Silica nanoparticles are being developed as a host of biomedical and biotechnological applications. For this reason, there are more studies about biocompatibility of silica with amorphous and crystalline structure. Except hydrated silica (opal), despite is presents directly and indirectly in humans. Two sizes of crystalline opal nanoparticles were investigated in this work under criteria of toxicology. METHODS In particular, cytotoxic and genotoxic effects caused by opal nanoparticles (80 and 120 nm) were evaluated in cultured mouse cells via a set of bioassays, methylthiazolyldiphenyl-tetrazolium-bromide (MTT) and 5-bromo-2'-deoxyuridine (BrdU). RESULTS 3T3-NIH cells were incubated for 24 and 72 h in contact with nanocrystalline opal particles, not presented significant statistically difference in the results of cytotoxicity. Genotoxicity tests of crystalline opal nanoparticles were performed by the BrdU assay on the same cultured cells for 24 h incubation. The reduction of BrdU-incorporated cells indicates that nanocrystalline opal exposure did not caused unrepairable damage DNA. CONCLUSIONS There is no relationship between that particles size and MTT reduction, as well as BrdU incorporation, such that the opal particles did not induce cytotoxic effect and genotoxicity in cultured mouse cells.
Collapse
Affiliation(s)
- Marlen Hernández-Ortiz
- Programa de Posgrado en Ciencia de Materiales del, Universidad de Sonora, A P 130, Hermosillo, Sonora, 83000, México
| | - Laura S Acosta-Torres
- Escuela Nacional de Estudios Superiores, Universidad Nacional Autónoma de México, Unidad León, Boulevard UNAM No. 2011 Predio el Potrero y el Saucillo, C.P. 36969, León, Guanajuato, México
| | | | - Alicia I Mendieta
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, 76230, México
| | - Rodolfo Bernal
- Departamento de Investigación en Física, Universidad de Sonora, A. P. 5-088, Hermosillo, Sonora, 83190, México
| | - Catalina Cruz-Vázquez
- Departamento de Investigación en Polímeros y Materiales, Universidad de Sonora, A P 130, Hermosillo, Sonora, 83000, México
| | - Victor M Castaño
- Departamento de Ingeniería Molecular de Materiales, Centro de Física Aplicada y Tecnología Avanzada, Campus Juriquilla, Querétaro, 76230, México
- Centro de Física Aplicada y Tecnología Avanzada, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, 76230, México
| |
Collapse
|
28
|
Affiliation(s)
- Françoise Schrurs
- Louvain Centre of Toxicology and Applied Pharmacology, Université catholique de Louvain, Avenue E. Mounier, Box B1.52.12, 1200 Brussels, Belgium
| | | |
Collapse
|
29
|
Zamboni WC, Torchilin V, Patri AK, Hrkach J, Stern S, Lee R, Nel A, Panaro NJ, Grodzinski P. Best practices in cancer nanotechnology: perspective from NCI nanotechnology alliance. Clin Cancer Res 2012; 18:3229-41. [PMID: 22669131 PMCID: PMC3916007 DOI: 10.1158/1078-0432.ccr-11-2938] [Citation(s) in RCA: 175] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Historically, treatment of patients with cancer using chemotherapeutic agents has been associated with debilitating and systemic toxicities, poor bioavailability, and unfavorable pharmacokinetics. Nanotechnology-based drug delivery systems, on the other hand, can specifically target cancer cells while avoiding their healthy neighbors, avoid rapid clearance from the body, and be administered without toxic solvents. They hold immense potential in addressing all of these issues, which has hampered further development of chemotherapeutics. Furthermore, such drug delivery systems will lead to cancer therapeutic modalities that are not only less toxic to the patient but also significantly more efficacious. In addition to established therapeutic modes of action, nanomaterials are opening up entirely new modalities of cancer therapy, such as photodynamic and hyperthermia treatments. Furthermore, nanoparticle carriers are also capable of addressing several drug delivery problems that could not be effectively solved in the past and include overcoming formulation issues, multidrug-resistance phenomenon, and penetrating cellular barriers that may limit device accessibility to intended targets, such as the blood-brain barrier. The challenges in optimizing design of nanoparticles tailored to specific tumor indications still remain; however, it is clear that nanoscale devices carry a significant promise toward new ways of diagnosing and treating cancer. This review focuses on future prospects of using nanotechnology in cancer applications and discusses practices and methodologies used in the development and translation of nanotechnology-based therapeutics.
Collapse
Affiliation(s)
- William C Zamboni
- UNC Eshelman School of Pharmacy, UNC Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, UNC Institute for Pharmacogenomics and Individualized Therapy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Gustafson TP, Yan Y, Newton P, Hunter DA, Achilefu S, Akers WJ, Mackinnon SE, Johnson PJ, Berezin MY. A NIR Dye for Development of Peripheral Nerve Targeted Probes. MEDCHEMCOMM 2012; 3:685-690. [PMID: 24575295 PMCID: PMC3932509 DOI: 10.1039/c2md00297c] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Current imaging modalities lack the ability to quickly assess and classify nerve injury for predicting favourable versus unfavourable healing outcomes, which could minimize episodes of chronic pain and loss of function by allowing for early intervention. Thus, the development of a technique to noninvasively assess peripheral nerve damage is of critical importance. While the development of nerve specific near infrared (NIR) molecular probes capable of such diagnostics constitutes our long term goal, initial studies to identify a NIR dye for constructing such a probe are required. We have evaluated the properties of a novel highly hydrophilic and functionalizable polymethine dye, and its more hydrophobic analogue indocyanine green, within the sciatic nerve of rats following intra-nerve injection. The reporting ability of both dyes at critical depths for nerve imaging, the importance of hydrophilicity on dye transport through nervous tissue, and their toxicity - or lack thereof - to the neural environment have been evaluated. The results suggest that the novel NIR dye is an appropriate fluorescent reporter for use in designing nerve-specific optical molecular probes for non-invasive diagnosis and classification of nerve injury.
Collapse
Affiliation(s)
- Tiffany P. Gustafson
- Department of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Ying Yan
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Piyaraj Newton
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Daniel A. Hunter
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Samuel Achilefu
- Department of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Walter J. Akers
- Department of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Susan E. Mackinnon
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Philip J. Johnson
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Mikhail Y. Berezin
- Department of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
31
|
Abstract
Angiogenesis is a fundamental requirement for tumor growth and therefore it is a primary target for anti-cancer therapy. Molecular imaging of angiogenesis may provide novel opportunities for early diagnostic and for image-guided optimization and management of therapeutic regimens. Here we reviewed the advances in targeted imaging of key biomarkers of tumor angiogenesis, integrins and receptors for vascular endothelial growth factor (VEGF). Tracers for targeted imaging of these biomarkers in different imaging modalities are now reasonably well-developed and PET tracers for integrin imaging are currently in clinical trials. Molecular imaging of longitudinal responses to anti-angiogenic therapy in model tumor systems revealed a complex pattern of changes in targeted tracer accumulation in tumor, which reflects drug-induced tumor regression followed by vascular rebound. Further work will define the competitiveness of targeted imaging of key angiogenesis markers for early diagnostic and image-guided therapy.
Collapse
|
32
|
Backer MV, Backer JM. Imaging key biomarkers of tumor angiogenesis. Theranostics 2012; 2:502-15. [PMID: 22737188 PMCID: PMC3364556 DOI: 10.7150/thno.3623] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Accepted: 01/07/2012] [Indexed: 11/05/2022] Open
Abstract
Angiogenesis is a fundamental requirement for tumor growth and therefore it is a primary target for anti-cancer therapy. Molecular imaging of angiogenesis may provide novel opportunities for early diagnostic and for image-guided optimization and management of therapeutic regimens. Here we reviewed the advances in targeted imaging of key biomarkers of tumor angiogenesis, integrins and receptors for vascular endothelial growth factor (VEGF). Tracers for targeted imaging of these biomarkers in different imaging modalities are now reasonably well-developed and PET tracers for integrin imaging are currently in clinical trials. Molecular imaging of longitudinal responses to anti-angiogenic therapy in model tumor systems revealed a complex pattern of changes in targeted tracer accumulation in tumor, which reflects drug-induced tumor regression followed by vascular rebound. Further work will define the competitiveness of targeted imaging of key angiogenesis markers for early diagnostic and image-guided therapy.
Collapse
|
33
|
Zhang X, Bloch S, Akers W, Achilefu S. Near-infrared molecular probes for in vivo imaging. CURRENT PROTOCOLS IN CYTOMETRY 2012; Chapter 12:Unit12.27. [PMID: 22470154 PMCID: PMC3334312 DOI: 10.1002/0471142956.cy1227s60] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cellular and tissue imaging in the near-infrared (NIR) wavelengths between 700 and 900 nm is advantageous for in vivo imaging because of the low absorption of biological molecules in this region. This unit presents protocols for small animal imaging using planar and fluorescence lifetime imaging techniques. Included is an overview of NIR fluorescence imaging of cells and small animals using NIR organic fluorophores, nanoparticles, and multimodal imaging probes. The development, advantages, and application of NIR fluorescent probes that have been used for in vivo imaging are also summarized. The use of NIR agents in conjunction with visible dyes and considerations in selecting imaging agents are discussed. We conclude with practical considerations for the use of these dyes in cell and small animal imaging applications.
Collapse
Affiliation(s)
- Xuan Zhang
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Sharon Bloch
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Walter Akers
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Samuel Achilefu
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
34
|
Flament J, Geffroy F, Medina C, Robic C, Mayer JF, Mériaux S, Valette J, Robert P, Port M, Le Bihan D, Lethimonnier F, Boumezbeur F. In vivo CEST MR imaging of U87 mice brain tumor angiogenesis using targeted LipoCEST contrast agent at 7 T. Magn Reson Med 2012; 69:179-87. [PMID: 22378016 DOI: 10.1002/mrm.24217] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Accepted: 01/25/2012] [Indexed: 01/22/2023]
Abstract
LipoCEST are liposome-encapsulating paramagnetic contrast agents (CA) based on chemical exchange saturation transfer with applications in biomolecular MRI. Their attractive features include biocompatibility, subnanomolar sensitivity, and amenability to functionalization for targeting biomarkers. We demonstrate MR imaging using a targeted lipoCEST, injected intravenously. A lipoCEST carrying Tm(III)-complexes was conjugated to RGD tripeptide (RGD-lipoCEST), to target integrin α(ν)β(3) receptors involved in tumor angiogenesis and was compared with an unconjugated lipoCEST. Brain tumors were induced in athymic nude mice by intracerebral injection of U87MG cells and were imaged at 7 T after intravenous injection of either of the two contrast agents (n = 12 for each group). Chemical exchange saturation transfer-MSME sequence was applied over 2 h with an average acquisition time interval of 13.5 min. The chemical exchange saturation transfer signal was ∼1% in the tumor and controlateral regions, and decreased to ∼0.3% after 2 h; while RGD-lipoCEST signal was ∼1.4% in the tumor region and persisted for up to 2 h. Immunohistochemical staining revealed a persistent colocalization of RGD-lipoCEST with α(ν)β(3) receptors in the tumor region. These results constitute an encouraging step toward in vivo MRI imaging of tumor angiogenesis using intravenously injected lipoCEST.
Collapse
Affiliation(s)
- Julien Flament
- Neurospin, I2BM, Commissariat à l'Energie Atomique, Gif-sur-Yvette, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Kievit FM, Zhang M. Cancer nanotheranostics: improving imaging and therapy by targeted delivery across biological barriers. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2011; 23:H217-47. [PMID: 21842473 PMCID: PMC3397249 DOI: 10.1002/adma.201102313] [Citation(s) in RCA: 350] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 07/12/2011] [Indexed: 05/03/2023]
Abstract
Cancer nanotheranostics aims to combine imaging and therapy of cancer through use of nanotechnology. The ability to engineer nanomaterials to interact with cancer cells at the molecular level can significantly improve the effectiveness and specificity of therapy to cancers that are currently difficult to treat. In particular, metastatic cancers, drug-resistant cancers, and cancer stem cells impose the greatest therapeutic challenge for targeted therapy. Targeted therapy can be achieved with appropriately designed drug delivery vehicles such as nanoparticles, adult stem cells, or T cells in immunotherapy. In this article, we first review the different types of nanotheranostic particles and their use in imaging, followed by the biological barriers they must bypass to reach the target cancer cells, including the blood, liver, kidneys, spleen, and particularly the blood-brain barrier. We then review how nanotheranostics can be used to improve targeted delivery and treatment of cancer cells. Finally, we discuss development of nanoparticles to overcome current limitations in cancer therapy.
Collapse
Affiliation(s)
- Forrest M Kievit
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | | |
Collapse
|
36
|
Akers WJ, Kim C, Berezin M, Guo K, Fuhrhop R, Lanza GM, Fischer GM, Daltrozzo E, Zumbusch A, Cai X, Wang LV, Achilefu S. Noninvasive photoacoustic and fluorescence sentinel lymph node identification using dye-loaded perfluorocarbon nanoparticles. ACS NANO 2011; 5:173-82. [PMID: 21171567 PMCID: PMC3026895 DOI: 10.1021/nn102274q] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
The contrast mechanisms used for photoacoustic tomography (PAT) and fluorescence imaging differ in subtle, but significant, ways. The design of contrast agents for each or both modalities requires an understanding of the spectral characteristics as well as intra- and intermolecular interactions that occur during formulation. We found that fluorescence quenching that occurs in the formulation of near-infrared (NIR) fluorescent dyes in nanoparticles results in enhanced contrast for PAT. The ability of the new PAT method to utilize strongly absorbing chromophores for signal generation allowed us to convert a highly fluorescent dye into an exceptionally high PA contrast material. Spectroscopic characterization of the developed NIR dye-loaded perfluorocarbon-based nanoparticles for combined fluorescence and PA imaging revealed distinct dye-dependent photophysical behavior. We demonstrate that the enhanced contrast allows detection of regional lymph nodes of rats in vivo with time-domain optical and photoacoustic imaging methods. The results further show that the use of fluorescence lifetime imaging, which is less dependent on fluorescence intensity, provides a strategic approach to bridge the disparate contrast reporting mechanisms of fluorescence and PA imaging methods.
Collapse
Affiliation(s)
- Walter J. Akers
- Department of Radiology, Washington University School of Medicine, 4525 Scott Avenue, Saint Louis, MO 63108 (USA)
| | - Chulhong Kim
- Department of Biomedical Engineering, Washington University, One Brookings Drive, St. Louis, MO 63130 (USA)
| | - Mikhail Berezin
- Department of Radiology, Washington University School of Medicine, 4525 Scott Avenue, Saint Louis, MO 63108 (USA)
| | - Kevin Guo
- Department of Radiology, Washington University School of Medicine, 4525 Scott Avenue, Saint Louis, MO 63108 (USA)
| | - Ralph Fuhrhop
- C-TRAIN and Division of Cardiology, Washington University School of Medicine, 4320 Forest Park Avenue, Saint Louis, MO 63108 (USA)
| | - Gregory M. Lanza
- C-TRAIN and Division of Cardiology, Washington University School of Medicine, 4320 Forest Park Avenue, Saint Louis, MO 63108 (USA)
| | - Georg M. Fischer
- Department of Biophysics, Washington University School of Medicine, 4525 Scott Avenue, Saint Louis, MO 63108 (USA)
| | - Ewald Daltrozzo
- Department of Biophysics, Washington University School of Medicine, 4525 Scott Avenue, Saint Louis, MO 63108 (USA)
| | - Andreas Zumbusch
- Department of Biophysics, Washington University School of Medicine, 4525 Scott Avenue, Saint Louis, MO 63108 (USA)
| | - Xin Cai
- Department of Biomedical Engineering, Washington University, One Brookings Drive, St. Louis, MO 63130 (USA)
| | - Lihong V. Wang
- Department of Biomedical Engineering, Washington University, One Brookings Drive, St. Louis, MO 63130 (USA)
- Address correspondence to, Prof. S. Achilefu: , Prof. L. V. Wang:
| | - Samuel Achilefu
- Department of Radiology, Washington University School of Medicine, 4525 Scott Avenue, Saint Louis, MO 63108 (USA)
- Department of Biophysics, Washington University School of Medicine, 4525 Scott Avenue, Saint Louis, MO 63108 (USA)
- Address correspondence to, Prof. S. Achilefu: , Prof. L. V. Wang:
| |
Collapse
|
37
|
Licha K, Resch-Genger U. Probes for optical imaging: new developments. DRUG DISCOVERY TODAY. TECHNOLOGIES 2011; 8:e87-94. [PMID: 24990267 DOI: 10.1016/j.ddtec.2011.11.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Recent developments of fluorescent probes beyond approved indocyanine green (ICG) - itself increasingly spreading into new imaging applications like lymphatic mapping, arthritis imaging and tumor surgery - exploit various photophysical and biochemical mechanisms to monitor molecular events with higher specificity and accuracy. Emphasizing nanoparticulate formulations, targeted conjugates, activatable probes, probes with a sensor function and multimodality probes, this review discusses advantages and limitations of each type of probe, thereby critically assessing the desired translation into the clinic.:
Collapse
Affiliation(s)
- Kai Licha
- mivenion GmbH, Robert-Koch-Platz 4, D-10115 Berlin, Germany.
| | - Ute Resch-Genger
- BAM Bundesanstalt für Materialforschung, Richard-Willstaetter-Str. 11, D-12489 Berlin, Germany.
| |
Collapse
|
38
|
Lanza GM, Marsh JN, Hu G, Scott MJ, Schmieder AH, Caruthers SD, Pan D, Wickline SA. Rationale for a nanomedicine approach to thrombolytic therapy. Stroke 2010; 41:S42-4. [PMID: 20876503 DOI: 10.1161/strokeaha.110.598656] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Gregory M Lanza
- Department of Medicine, Division of Cardiology, 4320 Forest Park Ave, Suite 101, St Louis, MO 63108, USA.
| | | | | | | | | | | | | | | |
Collapse
|