1
|
Schraven S, Brück R, Rosenhain S, Lemainque T, Heines D, Noormohammadian H, Pabst O, Lederle W, Gremse F, Kiessling F. CT- and MRI-Aided Fluorescence Tomography Reconstructions for Biodistribution Analysis. Invest Radiol 2024; 59:504-512. [PMID: 38038691 DOI: 10.1097/rli.0000000000001052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
OBJECTIVES Optical fluorescence imaging can track the biodistribution of fluorophore-labeled drugs, nanoparticles, and antibodies longitudinally. In hybrid computed tomography-fluorescence tomography (CT-FLT), CT provides the anatomical information to generate scattering and absorption maps supporting a 3-dimensional reconstruction from the raw optical data. However, given the CT's limited soft tissue contrast, fluorescence reconstruction and quantification can be inaccurate and not sufficiently detailed. Magnetic resonance imaging (MRI) can overcome these limitations and extend the options for tissue characterization. Thus, we aimed to establish a hybrid CT-MRI-FLT approach for whole-body imaging and compared it with CT-FLT. MATERIALS AND METHODS The MRI-based hybrid imaging approaches were established first by scanning a water and coconut oil-filled phantom, second by quantifying Cy7 concentrations of inserts in dead mice, and finally by analyzing the biodistribution of AF750-labeled immunoglobulins (IgG, IgA) in living SKH1 mice. Magnetic resonance imaging, acquired with a fat-water-separated mDixon sequence, CT, and FLT were co-registered using markers in the mouse holder frame filled with white petrolatum, which was solid, stable, and visible in both modalities. RESULTS Computed tomography-MRI fusion was confirmed by comparing the segmentation agreement using Dice scores. Phantom segmentations showed good agreement, after correction for gradient linearity distortion and chemical shift. Organ segmentations in dead and living mice revealed adequate agreement for fusion. Marking the mouse holder frame and the successful CT-MRI fusion enabled MRI-FLT as well as CT-MRI-FLT reconstructions. Fluorescence tomography reconstructions supported by CT, MRI, or CT-MRI were comparable in dead mice with 60 pmol fluorescence inserts at different locations. Although standard CT-FLT reconstruction only considered general values for soft tissue, skin, lung, fat, and bone scattering, MRI's more versatile soft tissue contrast enabled the additional consideration of liver, kidneys, and brain. However, this did not change FLT reconstructions and quantifications significantly, whereas for extending scattering maps, it was important to accurately segment the organs and the entire mouse body. The various FLT reconstructions also provided comparable results for the in vivo biodistribution analyses with fluorescent immunoglobulins. However, MRI additionally enabled the visualization of gallbladder, thyroid, and brain. Furthermore, segmentations of liver, spleen, and kidney were more reliable due to better-defined contours than in CT. Therefore, the improved segmentations enabled better assignment of fluorescence signals and more differentiated conclusions with MRI-FLT. CONCLUSIONS Whole-body CT-MRI-FLT was implemented as a novel trimodal imaging approach, which allowed to more accurately assign fluorescence signals, thereby significantly improving pharmacokinetic analyses.
Collapse
Affiliation(s)
- Sarah Schraven
- From the Institute for Experimental Molecular Imaging, RWTH Aachen University, Aachen, Germany (S.S., R.B., S.R., T.L., D.H., W.L., F.G., F.K.); Institute of Molecular Medicine, RWTH Aachen University, Aachen, Germany (H.N., O.P.); Gremse-IT GmbH, Aachen, Germany (S.R., F.G.); Department for Diagnostic and Interventional Radiology, RWTH Aachen University, Aachen, Germany (T.L.); Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany (F.K.); and Fraunhofer MEVIS, Institute for Medical Image Computing, Aachen, Germany (F.K.)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
2
|
Schraven S, Rosenhain S, Brueck R, Wiechmann TM, Pola R, Etrych T, Lederle W, Lammers T, Gremse F, Kiessling F. Dye labeling for optical imaging biases drug carriers' biodistribution and tumor uptake. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 48:102650. [PMID: 36623712 DOI: 10.1016/j.nano.2023.102650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/25/2022] [Accepted: 12/27/2022] [Indexed: 01/08/2023]
Abstract
Biodistribution analyses of nanocarriers are often performed with optical imaging. Though dye tags can interact with transporters, e.g., organic anion transporting polypeptides (OATPs), their influence on biodistribution was hardly studied. Therefore, this study compared tumor cell uptake and biodistribution (in A431 tumor-bearing mice) of four near-infrared fluorescent dyes (AF750, IRDye750, Cy7, DY-750) and dye-labeled poly(N-(2-hydroxypropyl)methacrylamide)-based nanocarriers (dye-pHPMAs). Tumor cell uptake of hydrophobic dyes (Cy7, DY-750) was higher than that of hydrophilic dyes (AF750, IRDye750), and was actively mediated but not related to OATPs. Free dyes' elimination depended on their hydrophobicity, and tumor uptake correlated with blood circulation times. Dye-pHPMAs circulated longer and accumulated stronger in tumors than free dyes. Dye labeling significantly influenced nanocarriers' tumor accumulation and biodistribution. Therefore, low-interference dyes and further exploration of dye tags are required to achieve the most unbiased results possible. In our assessment, AF750 and IRDye750 best qualified for labeling hydrophilic nanocarriers.
Collapse
Affiliation(s)
- Sarah Schraven
- Institute for Experimental Molecular Imaging, RWTH Aachen University, Forckenbeckstrasse 55, 52074 Aachen, Germany
| | - Stefanie Rosenhain
- Institute for Experimental Molecular Imaging, RWTH Aachen University, Forckenbeckstrasse 55, 52074 Aachen, Germany; Gremse-IT GmbH, Dennewartstrasse 25, 52068 Aachen, Germany
| | - Ramona Brueck
- Institute for Experimental Molecular Imaging, RWTH Aachen University, Forckenbeckstrasse 55, 52074 Aachen, Germany
| | - Tim Marvin Wiechmann
- Institute for Experimental Molecular Imaging, RWTH Aachen University, Forckenbeckstrasse 55, 52074 Aachen, Germany
| | - Robert Pola
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, 162 06 Prague, Czech Republic
| | - Tomáš Etrych
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, 162 06 Prague, Czech Republic
| | - Wiltrud Lederle
- Institute for Experimental Molecular Imaging, RWTH Aachen University, Forckenbeckstrasse 55, 52074 Aachen, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, RWTH Aachen University, Forckenbeckstrasse 55, 52074 Aachen, Germany
| | - Felix Gremse
- Institute for Experimental Molecular Imaging, RWTH Aachen University, Forckenbeckstrasse 55, 52074 Aachen, Germany; Gremse-IT GmbH, Dennewartstrasse 25, 52068 Aachen, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, RWTH Aachen University, Forckenbeckstrasse 55, 52074 Aachen, Germany; Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany; Fraunhofer MEVIS, Institute for Medical Image Computing, Aachen, Germany.
| |
Collapse
|
3
|
Hu Y, Lafci B, Luzgin A, Wang H, Klohs J, Dean-Ben XL, Ni R, Razansky D, Ren W. Deep learning facilitates fully automated brain image registration of optoacoustic tomography and magnetic resonance imaging. BIOMEDICAL OPTICS EXPRESS 2022; 13:4817-4833. [PMID: 36187259 PMCID: PMC9484422 DOI: 10.1364/boe.458182] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 07/14/2022] [Accepted: 07/17/2022] [Indexed: 06/16/2023]
Abstract
Multispectral optoacoustic tomography (MSOT) is an emerging optical imaging method providing multiplex molecular and functional information from the rodent brain. It can be greatly augmented by magnetic resonance imaging (MRI) which offers excellent soft-tissue contrast and high-resolution brain anatomy. Nevertheless, registration of MSOT-MRI images remains challenging, chiefly due to the entirely different image contrast rendered by these two modalities. Previously reported registration algorithms mostly relied on manual user-dependent brain segmentation, which compromised data interpretation and quantification. Here we propose a fully automated registration method for MSOT-MRI multimodal imaging empowered by deep learning. The automated workflow includes neural network-based image segmentation to generate suitable masks, which are subsequently registered using an additional neural network. The performance of the algorithm is showcased with datasets acquired by cross-sectional MSOT and high-field MRI preclinical scanners. The automated registration method is further validated with manual and half-automated registration, demonstrating its robustness and accuracy.
Collapse
Affiliation(s)
- Yexing Hu
- School of Information Science and Technology, ShanghaiTech University, Shanghai 201210, China
- contributed equally
| | - Berkan Lafci
- Institute of Pharmacology and Toxicology and Institute for Biomedical Engineering, Faculty of Medicine, University of Zurich, Zurich 8052, Switzerland
- Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich 8093, Switzerland
- contributed equally
| | - Artur Luzgin
- Institute of Pharmacology and Toxicology and Institute for Biomedical Engineering, Faculty of Medicine, University of Zurich, Zurich 8052, Switzerland
- Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich 8093, Switzerland
| | - Hao Wang
- Institute of Pharmacology and Toxicology and Institute for Biomedical Engineering, Faculty of Medicine, University of Zurich, Zurich 8052, Switzerland
- Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich 8093, Switzerland
| | - Jan Klohs
- Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich 8093, Switzerland
| | - Xose Luis Dean-Ben
- Institute of Pharmacology and Toxicology and Institute for Biomedical Engineering, Faculty of Medicine, University of Zurich, Zurich 8052, Switzerland
- Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich 8093, Switzerland
| | - Ruiqing Ni
- Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich 8093, Switzerland
- Institute for Regenerative Medicine, University of Zurich, Zurich 8952, Switzerland
| | - Daniel Razansky
- Institute of Pharmacology and Toxicology and Institute for Biomedical Engineering, Faculty of Medicine, University of Zurich, Zurich 8052, Switzerland
- Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich 8093, Switzerland
| | - Wuwei Ren
- School of Information Science and Technology, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
4
|
Abstract
Targeting residual cardiovascular risk in primary and secondary prevention, would allow deployment of novel therapeutic agents, facilitating precision medicine. For example, lowering vascular inflammation is a promising strategy to reduce the residual inflammatory cardiovascular risk in patients already receiving optimal medical therapy, but prescribing novel anti-inflammatory treatments will be problematic due to the lack of specific companion diagnostic tests, to guide their targeted use in clinical practice. Currently available tests for the detection of coronary inflammation are either non-specific for the cardiovascular system (e.g. plasma biomarkers) or expensive and not readily available (e.g. hybrid positron emission tomography imaging). Recent technological advancements in coronary computed tomography angiography (CCTA) allow non-invasive detection of high-risk plaque features (positive remodelling, spotty calcification, low attenuation plaque, and napkin-ring sign) and help identify the vulnerable patient, but they provide only indirectly information about coronary inflammation. Perivascular fat attenuation index (FAI), a novel method for assessing coronary inflammation by analysing routine CCTA, captures changes in the perivascular adipose tissue composition driven by inflammatory signals coming from the inflamed coronary artery, by analysing the three-dimensional gradients of perivascular attenuation, followed by adjustments for technical, anatomical, and biological factors. By detecting vascular inflammation, perivascular FAI enhances cardiovascular risk discrimination which could aid more cost-effective deployment of novel therapeutic agents. In this article, we present the existing non-invasive modalities for the detection of coronary inflammation and provide a practical guide for their use in clinical practice.
Collapse
Affiliation(s)
- Charalambos Antoniades
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, UK
| | - Alexios S Antonopoulos
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, UK
| | | |
Collapse
|
5
|
Gao Y, Chu C, Jablonska A, Bulte JWM, Walczak P, Janowski M. Imaging as a tool to accelerate the translation of extracellular vesicle-based therapies for central nervous system diseases. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 13:e1688. [PMID: 33336512 DOI: 10.1002/wnan.1688] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/19/2020] [Accepted: 11/18/2020] [Indexed: 12/13/2022]
Abstract
Extracellular vesicles (EVs) are natural and diverse lipid bilayer-enclosed particles originating from various cellular components and containing an abundance of cargoes. Due to their unique properties, EVs have gained considerable interest as therapeutic agents for a variety of diseases, including central nervous system (CNS) disorders. Their therapeutic value depends on cell origin but can be further enhanced by enrichment of cargo when used as drug carriers. Therefore, there has been significant effort directed toward introducing them to clinical practice. However, it is essential to avoid the failures we have seen with whole-cell therapy, in particular for the treatment of the CNS. Successful launching of clinical studies is contingent upon the understanding of the biodistribution of EVs, including their uptake and clearance from organs and specific homing into the region of interest. A multitude of noninvasive imaging methods has been explored in vitro to investigate the spatio-temporal dynamics of EVs administered in vivo. However, only a few studies have been performed to track the delivery of EVs, especially delivery to the brain, which is the most therapeutically challenging organ. We focus here on the use of advanced imaging techniques as an essential tool to facilitate the acceleration of clinical translation of EV-based therapeutics, especially in the CNS arena. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Neurological Disease Diagnostic Tools > in vivo Nanodiagnostics and Imaging.
Collapse
Affiliation(s)
- Yue Gao
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Chengyan Chu
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Anna Jablonska
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jeff W M Bulte
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Piotr Walczak
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Miroslaw Janowski
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
6
|
Natarajan A. Copper-64-immunoPET imaging: bench to bedside. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF RADIOPHARMACEUTICAL CHEMISTRY AND BIOLOGY 2020; 64:356-363. [PMID: 33045821 DOI: 10.23736/s1824-4785.20.03310-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Positron emission tomography (PET) is a growing non-invasive diagnostic and molecular imaging tool in nuclear medicine, that is used to identify several diseases including cancer. The immunoPET probe is made up of monoclonal antibodies (mAbs) or its fragments or similar molecules that tagged with positron radioisotopes (68Ga, 64Cu, 89Zr) bound together by a bifunctional chelator (BFC). This probe is designed to identify a specific disease. Currently, several immunoPET probes are being developed for preclinical as well as for clinical applications. These studies are showing promising results, both in preclinical and patients, using mostly 64Cu, 89Zr isotopes. This review elucidates the 64Cu based immunoPET applications, their pipelines and the emerging scope of this technique within the nuclear medicine and molecular imaging clinics from bench to bedside. Recently, immunoPET research have sharply increased especially after a big surge in approval of oncology antibodies by the FDA for immune checkpoint-blockade cancer immunotherapies. Currently, preclinical to clinical translations of immunoPET has several challenges, including designing probes, choice of radioisotopes, selection of stable BFC, and size of antibody and its tracer kinetics. All these obstacles will be addressed eventually by improving PET scanner sensitivity, designing appropriate size of imaging probe, and combining immunoPET with specific targeting antibodies. These improvements should contribute to the immunoPET becoming more applicable in clinics, which, in turn, will provide critical information for correct patient selection, for right dosing, and for the right time/staging of treatment.
Collapse
|
7
|
Pirovano G, Roberts S, Kossatz S, Reiner T. Optical Imaging Modalities: Principles and Applications in Preclinical Research and Clinical Settings. J Nucl Med 2020; 61:1419-1427. [PMID: 32764124 DOI: 10.2967/jnumed.119.238279] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 06/30/2020] [Indexed: 12/25/2022] Open
Abstract
With the ability to noninvasively image and monitor molecular processes within tumors, molecular imaging represents a fundamental tool for cancer scientists. In the current review, we describe emergent optical technologies for molecular imaging. We aim to provide the reader with an overview of the fundamental principles on which each imaging strategy is based, to introduce established and future applications, and to provide a rationale for selecting optical technologies for molecular imaging depending on disease location, biology, and anatomy. To accelerate clinical translation of imaging techniques, we also describe examples of practical applications in patients. Elevating these techniques into standard-of-care tools will transform patient stratification, disease monitoring, and response evaluation.
Collapse
Affiliation(s)
- Giacomo Pirovano
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sheryl Roberts
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Susanne Kossatz
- Department of Nuclear Medicine, University Hospital Klinikum Rechts der Isar, Technical University Munich, Munich, Germany.,Central Institute for Translational Cancer Research, Technical University of Munich, Munich, Germany.,Department of Chemistry, Technical University of Munich, Munich, Germany
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York .,Department of Radiology, Weill Cornell Medical College, New York, New York; and.,Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
8
|
Eisenblätter M, Wildgruber M. Optical and Optoacoustic Imaging Probes. Recent Results Cancer Res 2020; 216:337-355. [PMID: 32594392 DOI: 10.1007/978-3-030-42618-7_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Tissue has characteristic properties when it comes to light absorption and scattering. For optical (OI) and optoacoustic imaging (OAI) these properties can be utilised to visualise biological tissue characteristics, as, for example, the oxygenation state of haemoglobin alters the optical and optoacoustic properties of the molecule.
Collapse
Affiliation(s)
- Michel Eisenblätter
- Department of Diagnostic and Interventional Radiology, University Medical Center Freiburg, Freiburg im Breisgau, Germany.
| | - Moritz Wildgruber
- Department of Radiology, Ludwig Maximilians-University of Munich, Munich, Germany
| |
Collapse
|
9
|
Bai H, Yuan M, Wang X, Wang X, Che J. Development of a Gold Nanoparticle-Functionalized Surface Plasmon Resonance Assay for the Sensitive Detection of Monoclonal Antibodies and Its Application in Pharmacokinetics. Drug Metab Dispos 2019; 47:1361-1367. [PMID: 31324700 DOI: 10.1124/dmd.119.086249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Accepted: 07/15/2019] [Indexed: 11/22/2022] Open
Abstract
As a prominent human therapeutic, therapeutic monoclonal antibodies (mAbs) have attracted increasing attention in the past decade due to their high-targeting specificity, low toxicity, and prolonged efficacy. Systematic pharmacokinetic analysis of mAbs not only largely facilitates the understanding of their biologic functions but also promotes the development of therapeutic drug discovery, early clinical trial implementation, and therapeutic monitoring. However, the extremely complex nature of biomatrices and the especially low dosages of mAbs make their detection in biomatrices and further pharmacokinetic analysis highly challenging. Therefore, a method capable of reliably, quickly, and sensitively quantifying mAbs in biomatrices is urgently needed. In this work, we developed and evaluated an gold nanoparticle-functionalized surface plasmon resonance assay for cetuximab (C225) detection and pharmacokinetic analysis in rhesus monkeys. Combining its advantages of label-free pretreatment and amplified signal response, the lower limit of quantitation of C225 in monkey serum was reduced to 0.0125 μg/ml, and the linear range had an order of magnitude comparable to that of an ELISA-based method. Furthermore, the pharmacokinetics of C225 in rhesus monkeys was studied after intravenous infusions of single doses at 7.5, 24, and 75 mg/kg. The concentration of C225 in monkey serum was detectable after dosing for 720 hours. We believe that this new strategy will be applicable as a general protocol for mAb quantification, pharmacokinetic characteristic determination, and toxicokinetic analysis during drug development.
Collapse
Affiliation(s)
- Haihong Bai
- Phase I Clinical Trial Center, Beijing Shijitan Hospital of Capital Medical University, Beijing, PR China (H.B., Xin.W.); State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, PR China (M.Y., J.C.); and Chinese Pharmaceutical Association, Beijing, PR China (Xia.W.)
| | - Mei Yuan
- Phase I Clinical Trial Center, Beijing Shijitan Hospital of Capital Medical University, Beijing, PR China (H.B., Xin.W.); State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, PR China (M.Y., J.C.); and Chinese Pharmaceutical Association, Beijing, PR China (Xia.W.)
| | - Xiaojing Wang
- Phase I Clinical Trial Center, Beijing Shijitan Hospital of Capital Medical University, Beijing, PR China (H.B., Xin.W.); State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, PR China (M.Y., J.C.); and Chinese Pharmaceutical Association, Beijing, PR China (Xia.W.)
| | - Xinghe Wang
- Phase I Clinical Trial Center, Beijing Shijitan Hospital of Capital Medical University, Beijing, PR China (H.B., Xin.W.); State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, PR China (M.Y., J.C.); and Chinese Pharmaceutical Association, Beijing, PR China (Xia.W.)
| | - Jinjing Che
- Phase I Clinical Trial Center, Beijing Shijitan Hospital of Capital Medical University, Beijing, PR China (H.B., Xin.W.); State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, PR China (M.Y., J.C.); and Chinese Pharmaceutical Association, Beijing, PR China (Xia.W.)
| |
Collapse
|
10
|
Ergen C, Niemietz PM, Heymann F, Baues M, Gremse F, Pola R, van Bloois L, Storm G, Kiessling F, Trautwein C, Luedde T, Lammers T, Tacke F. Liver fibrosis affects the targeting properties of drug delivery systems to macrophage subsets in vivo. Biomaterials 2019; 206:49-60. [PMID: 30925288 DOI: 10.1016/j.biomaterials.2019.03.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/14/2019] [Accepted: 03/18/2019] [Indexed: 12/13/2022]
Abstract
Myeloid immune cells promote inflammation and fibrosis in chronic liver diseases. Drug delivery systems, such as polymers, liposomes and microbubbles, efficiently target myeloid cells in healthy liver, but their targeting properties in hepatic fibrosis remain elusive. We therefore studied the biodistribution of three intravenously injected carrier material, i.e. 10 nm poly(N-(2-hydroxypropyl)methacrylamide) polymers, 100 nm PEGylated liposomes and 2000 nm poly(butyl cyanoacrylate) microbubbles, in two fibrosis models in immunocompetent mice. While whole-body imaging confirmed preferential hepatic uptake even after induction of liver fibrosis, flow cytometry and immunofluorescence analysis revealed markedly decreased carrier uptake by liver macrophage subsets in fibrosis, particularly for microbubbles and polymers. Importantly, carrier uptake co-localized with immune infiltrates in fibrotic livers, corroborating the intrinsic ability of the carriers to target myeloid cells in areas of inflammation. Of the tested carrier systems liposomes had the highest uptake efficiency among hepatic myeloid cells, but the lowest specificity for cellular subsets. Hepatic fibrosis affected carrier uptake in liver and partially in spleen, but not in other tissues (blood, bone marrow, lung, kidney). In conclusion, while drug carrier systems target distinct myeloid cell populations in diseased and healthy livers, hepatic fibrosis profoundly affects their targeting efficiency, supporting the need to adapt nanomedicine-based approaches in chronic liver disease.
Collapse
Affiliation(s)
- Can Ergen
- Department of Medicine I, University Hospital Hamburg-Eppendorf, Hamburg, Germany; Department of Medicine III, University Hospital Aachen, Aachen, Germany
| | | | - Felix Heymann
- Department of Medicine III, University Hospital Aachen, Aachen, Germany; Department of Hepatology and Gastroenterology, Charité University Medicine Berlin, Berlin, Germany
| | - Maike Baues
- Department of Experimental Molecular Imaging, University Clinic and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Felix Gremse
- Department of Experimental Molecular Imaging, University Clinic and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Robert Pola
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Louis van Bloois
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - Gert Storm
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - Fabian Kiessling
- Department of Experimental Molecular Imaging, University Clinic and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | | | - Tom Luedde
- Department of Medicine III, University Hospital Aachen, Aachen, Germany
| | - Twan Lammers
- Department of Experimental Molecular Imaging, University Clinic and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany; Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands; Department of Targeted Therapeutics, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, the Netherlands
| | - Frank Tacke
- Department of Medicine III, University Hospital Aachen, Aachen, Germany; Department of Hepatology and Gastroenterology, Charité University Medicine Berlin, Berlin, Germany.
| |
Collapse
|
11
|
Gordon MR, Zhuang J, Ventura J, Li L, Raghupathi K, Thayumanavan S. Biodistribution Analysis of NIR-Labeled Nanogels Using in Vivo FMT Imaging in Triple Negative Human Mammary Carcinoma Models. Mol Pharm 2018; 15:1180-1191. [PMID: 29378144 DOI: 10.1021/acs.molpharmaceut.7b01011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The purpose of this study is to evaluate the biodistribution properties of random-copolymer-based core-cross-linked nanogels of various sizes and surface poly(ethylene glycol) composition. Systematic variations of near-IR labeled nanogels, comprising varying particle sizes (28-135 nm), PEG corona quantity (0-50 mol %), and PEG length (PEG Mn 1000, 2000, and 5000), were prepared and injected in mice that had been subcutaneously implanted with MDA-MB-231-luc-D3H2LN human mammary carcinoma. In vivo biodistribution was obtained using fluorescence molecular tomography imaging at 0, 6, 24, 48, and 72 h postinjection. Retention of total body probe and percentages of total injected dose in the tumor, liver, spleen, lungs, heart, intestines, and kidneys were obtained. Smaller nanogels (∼30-40 nm) with a high PEG conjugation (∼43-46 mol %) of Mn 2000 on their coronas achieved the highest tumor specificity with peak maximum 27% ID/g, a statistically significant propensity toward accumulation with 16.5% ID/g increase from 0 to 72 h of imaging, which constitutes a 1.5-fold increase. Nanogels with greater tumor localization also had greater retention of total body probe over 72 h. Nanogels without extensive PEGylation were rapidly excreted, even at similar sizes to PEGylated nanogels exhibiting whole body retention. Of all tissues, the liver had the highest % ID, however, like other tissues, it displayed a monotonic decrease over time, suggesting nanogel clearance by hepatic metabolism. Ex vivo quantification of individual tissues from gross necropsy at 72 h postinjection generally correlated with the FMT analysis, providing confidence in tissue signal segmentation in vivo. The parameters determined to most significantly direct a nanogel to the desired tumor target can lead to improve effectiveness for nanogels as therapeutic delivery vehicles.
Collapse
|