Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters











Publication year range
1.
Vaccines (Basel) ; 12(7)2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39066426

ABSTRACT

BACKGROUND: This study was conducted to investigate whether mRNA vaccine technology could be adapted for the ectothermic vertebrate Atlantic salmon (Salmo salar). Lipid nanoparticle (LNP) technology has been developed and optimized for mRNA vaccines in mammals, stabilizing mRNA and facilitating its delivery into cells. However, its utility at the temperatures and specific biological environments present in ectotherms remains unclear. In addition, it is unknown if modified mRNA containing non-canonical nucleotides can correctly translate in salmonid cells. METHODS: We used an mRNA transcript coding for enhanced green fluorescence protein, flanked by the untranslated regions of the hemagglutinin-esterase gene of the infectious salmon anemia virus, and a 120-base-long poly(A) tail. The mRNA was generated via in vitro transcription where uridine residues were replaced with N1-methyl-pseudouridines, and then encapsulated in LNPs. RESULTS: When transfected into the salmonid cell line CHH-1, the mRNA-LNP construct induced expression of EGFP. Furthermore, when mRNA-LNPs were injected intramuscularly into salmon, in vivo protein expression was demonstrated via immunohistochemistry. EGFP was observed in cells infiltrating the spaces between muscle cells in a focal inflammatory response. CONCLUSION: The results indicate that N1-methyl-pseudouridine-modified mRNA encapsulated in LNPs can be used to express antigens of interest in salmonid fish.

2.
RNA Biol ; 21(1): 1-8, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38836544

ABSTRACT

Production and storage of synthetic mRNA can introduce a variety of byproducts which reduce the overall integrity and functionality of mRNA vaccines and therapeutics. mRNA integrity is therefore designated as a critical quality attribute which must be evaluated with state-of-the-art analytical methods before clinical use. The current study first demonstrates the effect of heat degradation on transcript translatability and then describes a novel enzymatic approach to assess the integrity of conventional mRNA and long self-amplifying mRNA. By first hybridizing oligo-T to the poly(A) tail of intact mRNA and subsequently digesting the unhybridized RNA fragments with a 3'-5' exoribonuclease, individual nucleotides can be selectively released from RNA fragments. The adenosine-based fraction of these nucleotides can then be converted into ATP and detected by luminescence as a sensitive indicator of mRNA byproducts. We developed a polynucleotide phosphorylase (PNPase)-based assay that offers fast and sensitive evaluation of mRNA integrity, regardless of its length, thus presenting a novel and fully scalable alternative to chromatographic-, electrophoresis-, or sequencing-based techniques.


Subject(s)
Polyribonucleotide Nucleotidyltransferase , RNA, Messenger , RNA, Messenger/genetics , RNA, Messenger/metabolism , Polyribonucleotide Nucleotidyltransferase/metabolism , Polyribonucleotide Nucleotidyltransferase/genetics , Humans , Oligonucleotides/metabolism , RNA Stability
3.
J Control Release ; 366: 611-620, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38215988

ABSTRACT

Polyethylene glycol (PEG) conjugation (PEGylation) is a well-established strategy to improve the pharmacokinetic and biocompatibility properties of a wide variety of nanomedicines and therapeutic peptides and proteins. This broad use makes PEG an attractive 'allround' candidate marker for the biodistribution of such PEGylated compounds. This paper presents the development of a novel strategy for PEG quantification in biological matrices. The methodology is based on sample hydrolysis which both decomposes the sample matrix and degrades PEGylated analytes to specific molecular fragments more suitable for detection by LC-MS/MS. Method versatility was demonstrated by applying it to a wide variety of PEGylated compounds, including polymeric poly(ethylbutyl cyanoacrylate) (PEBCA) nanoparticles, lipidic nanoparticles (Doxil®, LipImage 815™ and lipid nanoparticles for nucleic acid delivery) and the antibody Cimzia®. Method applicability was assessed by analyzing plasma and tissue samples from a comprehensive drug biodistribution study in rats, of both PEBCA and LipImage 815™ nanoparticles. The results demonstrated the method's utility for biodistribution studies on PEG. Importantly, by using the method described herein in tandem with quantification of nanoparticle payloads, we showed that this approach can provide detailed understanding of various critical aspects of the in vivo behavior of PEGylated nanomedicines, such as drug release and particle stability. Together, the presented results demonstrate the novel method as a robust, versatile and generic approach for biodistribution analysis of PEGylated therapeutics.


Subject(s)
Cyanoacrylates , Liquid Chromatography-Mass Spectrometry , Nanomedicine , Rats , Animals , Tissue Distribution , Chromatography, Liquid , Tandem Mass Spectrometry , Polyethylene Glycols/chemistry
4.
Cancer Cell Int ; 23(1): 279, 2023 Nov 18.
Article in English | MEDLINE | ID: mdl-37980483

ABSTRACT

BACKGROUND: Myeloid cells play an essential role in cancer metastasis. The phenotypic diversity of these cells during cancer development has attracted great interest; however, their functional heterogeneity and plasticity have limited their role as prognostic markers and therapeutic targets. METHODS: To identify markers associated with myeloid cells in metastatic tumours, we compared transcriptomic data from immune cells sorted from metastatic and non-metastatic mammary tumours grown in BALB/cJ mice. To assess the translational relevance of our in vivo findings, we assessed human breast cancer biopsies and evaluated the association between arginase 1 protein expression in breast cancer tissues with tumour characteristics and patient outcomes. RESULTS: Among the differentially expressed genes, arginase 1 (ARG1) showed a unique expression pattern in tumour-infiltrating myeloid cells that correlated with the metastatic capacity of the tumour. Even though ARG1-positive cells were found almost exclusively inside the metastatic tumour, ARG1 protein was also present in the plasma. In human breast cancer biopsies, the presence of ARG1-positive cells was strongly correlated with high-grade proliferating tumours, poor prognosis, and low survival. CONCLUSION: Our findings highlight the potential use of ARG1-positive myeloid cells as an independent prognostic marker to evaluate the risk of metastasis in breast cancer patients.

5.
Nanomedicine ; 54: 102712, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37838100

ABSTRACT

Nanomedicine holds immense potential for therapeutic manipulation of phagocytic immune cells. However, in vitro studies often fail to accurately translate to the complex in vivo environment. To address this gap, we employed an ex vivo human whole-blood assay to evaluate liposome interactions with immune cells. We systematically varied liposome size, PEG-surface densities and sphingomyelin and ganglioside content. We observed differential uptake patterns of the assessed liposomes by neutrophils and monocytes, emphasizing the importance of liposome design. Interestingly, our results aligned closely with published in vivo observations in mice and patients. Moreover, liposome exposure induced changes in cytokine release and cellular responses, highlighting the potential modulation of immune system. Our study highlights the utility of human whole-blood models in assessing nanoparticle-immune cell interactions and provides insights into liposome design for modulating immune responses.


Subject(s)
Liposomes , Phagocytes , Humans , Animals , Mice , Monocytes , Sphingomyelins , Cytokines
6.
Adv Drug Deliv Rev ; 189: 114528, 2022 10.
Article in English | MEDLINE | ID: mdl-36067968

ABSTRACT

Intravital microscopy (IVM) expands our understanding of cellular and molecular processes, with applications ranging from fundamental biology to (patho)physiology and immunology, as well as from drug delivery to drug processing and drug efficacy testing. In this review, we highlight modalities, methods and model organisms that make up today's IVM landscape, and we present how IVM - via its high spatiotemporal resolution - enables analysis of metabolites, small molecules, nanoparticles, immune cells, and the (tumor) tissue microenvironment. We furthermore present examples of how IVM facilitates the elucidation of nanomedicine kinetics and targeting mechanisms, as well as of biological processes such as immune cell death, host-pathogen interactions, metabolic states, and disease progression. We conclude by discussing the prospects of IVM clinical translation and examining the integration of machine learning in future IVM practice.


Subject(s)
Intravital Microscopy , Neoplasms , Drug Delivery Systems , Humans , Neoplasms/drug therapy , Tumor Microenvironment
7.
Cell ; 183(3): 786-801.e19, 2020 10 29.
Article in English | MEDLINE | ID: mdl-33125893

ABSTRACT

Trained immunity, a functional state of myeloid cells, has been proposed as a compelling immune-oncological target. Its efficient induction requires direct engagement of myeloid progenitors in the bone marrow. For this purpose, we developed a bone marrow-avid nanobiologic platform designed specifically to induce trained immunity. We established the potent anti-tumor capabilities of our lead candidate MTP10-HDL in a B16F10 mouse melanoma model. These anti-tumor effects result from trained immunity-induced myelopoiesis caused by epigenetic rewiring of multipotent progenitors in the bone marrow, which overcomes the immunosuppressive tumor microenvironment. Furthermore, MTP10-HDL nanotherapy potentiates checkpoint inhibition in this melanoma model refractory to anti-PD-1 and anti-CTLA-4 therapy. Finally, we determined MTP10-HDL's favorable biodistribution and safety profile in non-human primates. In conclusion, we show that rationally designed nanobiologics can promote trained immunity and elicit a durable anti-tumor response either as a monotherapy or in combination with checkpoint inhibitor drugs.


Subject(s)
Immune Checkpoint Inhibitors/therapeutic use , Immunity , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , Nanotechnology , Acetylmuramyl-Alanyl-Isoglutamine/metabolism , Animals , Behavior, Animal , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Proliferation/drug effects , Cholesterol/metabolism , Female , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Immune Checkpoint Inhibitors/pharmacology , Immunity/drug effects , Immunotherapy , Lipoproteins, HDL/metabolism , Mice, Inbred C57BL , Primates , Tissue Distribution/drug effects , Tumor Microenvironment/drug effects
8.
ACS Nano ; 14(7): 7832-7846, 2020 07 28.
Article in English | MEDLINE | ID: mdl-32413260

ABSTRACT

Although the first nanomedicine was clinically approved more than two decades ago, nanoparticles' (NP) in vivo behavior is complex and the immune system's role in their application remains elusive. At present, only passive-targeting nanoformulations have been clinically approved, while more complicated active-targeting strategies typically fail to advance from the early clinical phase stage. This absence of clinical translation is, among others, due to the very limited understanding for in vivo targeting mechanisms. Dynamic in vivo phenomena such as NPs' real-time targeting kinetics and phagocytes' contribution to active NP targeting remain largely unexplored. To better understand in vivo targeting, monitoring NP accumulation and distribution at complementary levels of spatial and temporal resolution is imperative. Here, we integrate in vivo positron emission tomography/computed tomography imaging with intravital microscopy and flow cytometric analyses to study αvß3-integrin-targeted cyclic arginine-glycine-aspartate decorated liposomes and oil-in-water nanoemulsions in tumor mouse models. We observed that ligand-mediated accumulation in cancerous lesions is multifaceted and identified "NP hitchhiking" with phagocytes to contribute considerably to this intricate process. We anticipate that this understanding can facilitate rational improvement of nanomedicine applications and that immune cell-NP interactions can be harnessed to develop clinically viable nanomedicine-based immunotherapies.


Subject(s)
Nanoparticles , Neoplasms , Animals , Integrin alphaV , Integrin alphaVbeta3 , Lipids , Mice , Neoplasms/drug therapy , Phagocytes
9.
Eur J Pharm Biopharm ; 152: 248-256, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32439308

ABSTRACT

Melatonin is a neurohormone with potenial therapeutic effects in many diseases including neonatal hypoxic-ischemic (HI) brain injury. Due to limited solubility in water there is currently no clinically available melatonin formulation for parenteral use. Clinical use of melatonin has thus relied on oral administration, which in many cases is hampered by low and variable bioavailability. In animal treatment studies of neonatal HI, this issue have been circumvented by using parenteral administration of melatonin dissolved in ethanol (EtOH) or dimethyl sulfoxide (DMSO), solvents that are potentially neurotoxic, especially to the newborn brain. Thus, there is an urgent need for a non-toxic injectable melatonin formulation. The aim of this study was to develop such a formulation comprised of melatonin and biocompatible lipid-based nanoparticles with improved melatonin bioavailability. We herein report the development and characterization of an injectable system composed of melatonin and liposomes (LP) or oil-in-water nanoemulsions (NE). Nanoparticle characterization confirmed physicochemical stability over a week and an improvement with respect to melatonin solubilization in water (2.6 mg/mL in our injectable system). Determination of the in vitro release kinetics showed a prolonged release when melatonin is solubilized in nanoparticles (T1/2: 81 min vs 50 min vs 26 min for melatonin-LP, melatonin-NE, and melatonin-EtOH respectively). The pharmacokinetic (PK) parameters were confirmed in vivo in adult rats as similar melatonin levels detected in blood and indicated higher bioavailability in brain after intravenous administration of melatonin nanoformulations (10 mg/kg) in comparison to the free-melatonin administration. In conclusion, we have developed an organic solvent-free injectable formulation for melatonin by utilizing FDA-approved components, as a safe alternative for facilitating the potential of melatonin against variety of pathological conditions.


Subject(s)
Melatonin/chemistry , Nanoparticles/chemistry , Solvents/chemistry , Animals , Animals, Newborn , Biological Availability , Brain/drug effects , Brain/metabolism , Chemistry, Pharmaceutical/methods , Disease Models, Animal , Emulsions/chemistry , Female , Hypoxia-Ischemia, Brain/drug therapy , Hypoxia-Ischemia, Brain/metabolism , Liposomes/chemistry , Melatonin/pharmacokinetics , Melatonin/pharmacology , Rats , Rats, Sprague-Dawley , Solubility
10.
Nat Nanotechnol ; 15(5): 398-405, 2020 05.
Article in English | MEDLINE | ID: mdl-32313216

ABSTRACT

Ischaemic heart disease evokes a complex immune response. However, tools to track the systemic behaviour and dynamics of leukocytes non-invasively in vivo are lacking. Here, we present a multimodal hot-spot imaging approach using an innovative high-density lipoprotein-derived nanotracer with a perfluoro-crown ether payload (19F-HDL) to allow myeloid cell tracking by 19F magnetic resonance imaging. The 19F-HDL nanotracer can additionally be labelled with zirconium-89 and fluorophores to detect myeloid cells by in vivo positron emission tomography imaging and optical modalities, respectively. Using our nanotracer in atherosclerotic mice with myocardial infarction, we observed rapid myeloid cell egress from the spleen and bone marrow by in vivo 19F-HDL magnetic resonance imaging. Concurrently, using ex vivo techniques, we showed that circulating pro-inflammatory myeloid cells accumulated in atherosclerotic plaques and at the myocardial infarct site. Our multimodality imaging approach is a valuable addition to the immunology toolbox, enabling the study of complex myeloid cell behaviour dynamically.


Subject(s)
Myeloid Cells/pathology , Myocardial Ischemia/diagnostic imaging , Animals , Atherosclerosis/diagnostic imaging , Atherosclerosis/pathology , Cell Tracking/methods , Crown Ethers/analysis , Female , Fluorescent Dyes/analysis , Fluorine/analysis , Magnetic Resonance Imaging/methods , Mice , Mice, Inbred C57BL , Multimodal Imaging/methods , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/pathology , Myocardial Ischemia/pathology , Optical Imaging/methods , Positron-Emission Tomography/methods , Radioisotopes/analysis , Zirconium/analysis
11.
Mol Imaging Biol ; 22(3): 486-493, 2020 06.
Article in English | MEDLINE | ID: mdl-31650483

ABSTRACT

PURPOSE: The endeavor of deciphering intricate phenomena within the field of molecular medicine dictates the necessity to investigate tumor/disease microenvironment real-time on cellular level. We, hereby, design simple and robust intravital microscopy strategies, which can be used to elucidate cellular or molecular interactions in a fluorescent mouse model. PROCEDURES: We crossbred transgenic TIE2GFP mice with nude BALB/c mice, allowing the breeding of immunocompetent and immunodeficient mouse models expressing green fluorescent protein (GFP) in vascular endothelium. Then, we surgically exposed various tissues of interest to perform intravital microscopy. RESULTS: By utilizing simple tissue preparation procedures and confocal or two-photon microscopy, we produced high-resolution static snapshots, dynamic sequences, and 3D reconstructions of orthotopically grown mammary tumor, skin inflammation, brain, and muscle. The homogenous detection of GFP expressed by endothelial cells and a combination of fluorescence agents enabled landmarking of tumor microenvironment and precise molecular tagging. CONCLUSION: Simple intravital microscopy procedures on TIE2GFP mice allowed a real-time multi-color visualization of tissue microenvironment, underlining that robust microscopy strategies are relatively simple and can be readily available for many tissues of interest.


Subject(s)
Breast Neoplasms/pathology , Intravital Microscopy/methods , Microscopy, Confocal/methods , Receptor, TIE-2/genetics , Animals , Breast Neoplasms/diagnostic imaging , Cell Line, Tumor , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, Transgenic , Receptor, TIE-2/chemistry , Receptor, TIE-2/metabolism , Tumor Microenvironment
12.
Cancers (Basel) ; 11(11)2019 Nov 08.
Article in English | MEDLINE | ID: mdl-31717301

ABSTRACT

Tumor associated macrophages are an essential part of the tumor microenvironment. Consequently, bone marrow-derived monocytes (BMDMs) are continuously recruited to tumors and are therefore seen as ideal delivery vehicles with tumor-targeting properties. By using immune cell depleting agents and macroscopic in vivo fluorescence imaging, we demonstrated that removal of endogenous monocytes and macrophages (but not neutrophils) leads to an increased tumor accumulation of exogenously administered BMDMs. By means of intravital microscopy (IVM), we confirmed our macroscopic findings on a cellular level and visualized in real time the migration of the donor BMDMs in the tumors of living animals. Moreover, IVM also revealed that clodronate-mediated depletion drastically increases the circulation time of the exogenously administered BMDMs. In summary, these new insights illustrate that impairment of the mononuclear phagocyte system increases the circulation time and tumor accumulation of donor BMDMs.

14.
Mol Pharm ; 15(12): 5754-5761, 2018 12 03.
Article in English | MEDLINE | ID: mdl-30376341

ABSTRACT

Ligand-decorated nanoparticles are extensively studied and applied for in vivo drug delivery and molecular imaging. Generally, two different ligand-decoration procedures are utilized; ligands are either conjugated with nanoparticle ingredients and incorporated during nanoparticle preparation, or they are attached to preformed nanoparticles by utilizing functionalized reactive surface groups (e.g., maleimide). Although the two procedures result in nanoparticles with very similar physicochemical properties, formulations obtained through the latter manufacturing process typically contain nonconjugated reactive surface groups. In the current study, we hypothesized that the different ligand-decoration procedures might affect the extent of interaction between nanoparticles and immune cells (especially phagocytes). In order to investigate our hypothesis, we decorated lipidic nanoparticles with a widely used cyclic Arg-Gly-Asp (cRGD) peptide using the two different procedures. As proven from in vivo experiments in mice, the presence of nonconjugated surface moieties results in increased recognition by the immune system. This is important knowledge considering the emerging focus on understanding and optimizing ways to target and track immune cells and the development of nanomedicine-based strategies in the field of immunotherapy.


Subject(s)
Drug Compounding/methods , Nanoconjugates/administration & dosage , Oligopeptides/administration & dosage , Phagocytes/drug effects , Animals , Drug Evaluation, Preclinical , Immunotherapy/methods , Ligands , Liposomes , Maleimides/chemistry , Mice , Mice, Inbred BALB C , Nanoconjugates/chemistry , Nanomedicine/methods , Oligopeptides/chemistry , Phagocytes/immunology
15.
Philos Trans A Math Phys Eng Sci ; 375(2107)2017 Nov 28.
Article in English | MEDLINE | ID: mdl-29038380

ABSTRACT

Biomedical engineering and its associated disciplines play a pivotal role in improving our understanding and management of disease. Motivated by past accomplishments, such as the clinical implementation of coronary stents, pacemakers or recent developments in antibody therapies, disease management now enters a new era in which precision imaging and nanotechnology-enabled therapeutics are maturing to clinical translation. Preclinical molecular imaging increasingly focuses on specific components of the immune system that drive disease progression and complications, allowing the in vivo study of potential therapeutic targets. The first multicentre trials highlight the potential of clinical multimodality imaging for more efficient drug development. In this perspective, the role of integrating engineering, nanotechnology, molecular imaging and immunology to yield precision medicine is discussed.This article is part of the themed issue 'Challenges for chemistry in molecular imaging'.


Subject(s)
Molecular Imaging/methods , Nanomedicine/methods , Animals , Biomedical Engineering , Drug Development , Humans , Molecular Imaging/trends , Nanomedicine/trends , Nanotechnology , Precision Medicine , Translational Research, Biomedical
16.
Angew Chem Int Ed Engl ; 56(11): 2923-2926, 2017 03 06.
Article in English | MEDLINE | ID: mdl-28112478

ABSTRACT

Understanding the formation process of nanoparticles is of the utmost importance to improve their design and production. This especially holds true for self-assembled nanoparticles whose formation processes have been largely overlooked. Herein, we present a new technology that integrates a microfluidic-based nanoparticle synthesis method and Förster resonance energy transfer (FRET) microscopy imaging to visualize nanoparticle self-assembly in real time. Applied to different nanoparticle systems, for example, nanoemulsions, drug-loaded block-copolymer micelles, and nanocrystal-core reconstituted high-density lipoproteins, we have shown the approach's unique ability to investigate key parameters affecting nanoparticle formation.


Subject(s)
Fluorescence Resonance Energy Transfer , Nanoparticles/chemistry , Time Factors
17.
J Mater Chem B ; 5(3): 418-422, 2017 Jan 21.
Article in English | MEDLINE | ID: mdl-32263656

ABSTRACT

Herein, we report the synthesis of differently sized gadolinium oxide nanodisks and gadolinium doped iron oxide spherical and cubic nanoparticles through the thermal decomposition of an oleate precursor. We also demonstrate that these nanoparticles are promising candidates for MR contrast agents.

18.
Cytometry A ; 91(8): 760-766, 2017 08.
Article in English | MEDLINE | ID: mdl-27077940

ABSTRACT

In vitro and in vivo behavior of nanoparticles (NPs) is often studied by tracing the NPs with fluorescent dyes. This requires stable incorporation of dyes within the NPs, as dye leakage may give a wrong interpretation of NP biodistribution, cellular uptake, and intracellular distribution. Furthermore, NP labeling with trace amounts of dye should not alter NP properties such as interactions with cells or tissues. To allow for versatile NP studies with a variety of fluorescence-based assays, labeling of NPs with different dyes is desirable. Hence, when new dyes are introduced, simple and fast screening methods to assess labeling stability and NP-cell interactions are needed. For this purpose, we have used a previously described generic flow cytometry assay; incubation of cells with NPs at 4 and 37°C. Cell-NP interaction is confirmed by cellular fluorescence after 37°C incubation, and NP-dye retention is confirmed when no cellular fluorescence is detected at 4°C. Three different NP-platforms labeled with six different dyes were screened, and a great variability in dye retention was observed. Surprisingly, incorporation of trace amounts of certain dyes was found to reduce or even inhibit NP uptake. This work highlights the importance of thoroughly evaluating every dye-NP combination before pursuing NP-based applications. © 2016 International Society for Advancement of Cytometry.


Subject(s)
Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Nanoparticles/chemistry , Nanoparticles/metabolism , Animals , Biological Transport/physiology , Cell Line , Cell Line, Tumor , Flow Cytometry/methods , Fluorescence , Humans , Rats , Staining and Labeling/methods , Tissue Distribution/physiology
19.
Nat Commun ; 7: 11221, 2016 Apr 13.
Article in English | MEDLINE | ID: mdl-27071376

ABSTRACT

A major goal of cancer nanotherapy is to use nanoparticles as carriers for targeted delivery of anti-tumour agents. The drug-carrier association after intravenous administration is essential for efficient drug delivery to the tumour. However, a large number of currently available nanocarriers are self-assembled nanoparticles whose drug-loading stability is critically affected by the in vivo environment. Here we used in vivo FRET imaging to systematically investigate how drug-carrier compatibility affects drug release in a tumour mouse model. We found the drug's hydrophobicity and miscibility with the nanoparticles are two independent key parameters that determine its accumulation in the tumour. Next, we applied these findings to improve chemotherapeutic delivery by augmenting the parent drug's compatibility; as a result, we achieved better antitumour efficacy. Our results help elucidate nanomedicines' in vivo fate and provide guidelines for efficient drug delivery.


Subject(s)
Drug Carriers/chemistry , Nanomedicine , Neoplasms/therapy , Animals , Cell Line, Tumor , Computer Simulation , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Drug Delivery Systems , Drug Liberation , Female , Fluorescence Resonance Energy Transfer , Humans , Mice, Inbred BALB C , Mice, Nude , Microscopy , Nanoparticles/administration & dosage , Neoplasms/drug therapy , Spectroscopy, Near-Infrared , Treatment Outcome
20.
Small ; 12(3): 301-6, 2016 Jan 20.
Article in English | MEDLINE | ID: mdl-26619158

ABSTRACT

Manganese oxide nanoparticles (MONPs) are capable of time-dependent magnetic resonance imaging contrast switching as well as releasing a surface-bound drug. MONPs give T2/T2* contrast, but dissolve and release T1-active Mn(2+) and L-3,4-dihydroxyphenylalanine. Complementary images are acquired with a single contrast agent, and applications toward Parkinson's disease are suggested.


Subject(s)
Coated Materials, Biocompatible/chemistry , Contrast Media/chemistry , Drug Delivery Systems/methods , Levodopa/pharmacology , Magnetic Resonance Imaging/methods , Manganese Compounds/chemistry , Nanoparticles/chemistry , Oxides/chemistry , Animals , Nanoparticles/ultrastructure , Rats , Sus scrofa , Water/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL