1
|
Li J, Zhu Z, Xue Y, Downes A. Analysis of Drug Molecules in Living Cells. Crit Rev Anal Chem 2025:1-16. [PMID: 39854220 DOI: 10.1080/10408347.2025.2453431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2025]
Abstract
Cells are the fundamental units of life, comprising a highly concentrated and complex assembly of biomolecules that interact dynamic ally across spatial and temporal scales. Living cells are constantly undergoing dynamic processes, therefore, to understand the interactions between drug molecules and living cells is of paramount importance in the biomedical sciences and pharmaceutical development. Compared with traditional end-point assays and fixed cell analysis, analysis of drug molecules in living cells can provide more insight into the effects of drugs on cells in real-time and allowing for a better understanding of drug mechanisms and effects, which will contribute to the development of drug developing and testing and personalize medicine. However, the high demands of living cell analysis, including high costs, technical complexity, and throughput limitations, hinder the widespread application of this technology. In recent years, the rapid development of analytical methods such as Raman spectroscopy and fluorescence has made the in situ and real-time detection possible, allowing the analysis of single cell or cell populations at various conditions. In this review, we summarize the advanced analytical methods and technologies from last few years for drug detection in living cells.
Collapse
Affiliation(s)
- Jiaming Li
- Department of Bioengineering, Faculty of Engineering, The University of Edinburgh, Edinburgh, UK
| | - Zhiyuan Zhu
- Department of Infection Medicine, Faculty of Medicine, The University of Edinburgh, Edinburgh, UK
| | - Yuxiang Xue
- Department of Bioengineering, Faculty of Engineering, The University of Edinburgh, Edinburgh, UK
| | - Andrew Downes
- Department of Bioengineering, Faculty of Engineering, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
2
|
Bocci M, Principi L, Gilardoni E, Neri D, Cazzamalli S, Galbiati A. Delivery of Monomethyl Auristatin F to the Tumor Microenvironment with Noninternalizing Fibroblast Activation Protein-Cleavable Small Molecule-Drug Conjugates Elicits Potent In Vivo Anticancer Activity. Bioconjug Chem 2025; 36:15-19. [PMID: 39718420 DOI: 10.1021/acs.bioconjchem.4c00503] [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: 12/25/2024]
Abstract
OncoFAP is an ultrahigh affinity ligand of fibroblast activation protein (FAP), a tumor-associated antigen overexpressed in the stroma of the majority of solid tumors. OncoFAP has been previously implemented as a tumor-homing moiety for the development of small molecule drug conjugates (SMDCs). In the same context, the glycine--proline dipeptide was included with the aim to selectively undergo cleavage only in the presence of the target FAP, triggering the consequent release of the cytotoxic payload in the tumor microenvironment. In this work, we evaluate the use of monomethyl auristatin F (MMAF) as a payload, a close derivative of MMAE bearing a charged carboxylic acid that hampers its cellular permeability, typically employed in the development of internalizing antibody-drug conjugates. The novel OncoFAP-GlyPro-MMAF and the previously described OncoFAP-GlyPro-MMAE were compared in a head-to-head therapeutic experiment in mice bearing FAP-positive tumors. Surprisingly, the MMAF conjugate mediated potent antitumor activity, despite its poor cellular permeability.
Collapse
Affiliation(s)
- Matilde Bocci
- Philochem AG, R&D Department, CH-8112 Otelfingen, Switzerland
| | | | | | - Dario Neri
- Swiss Federal Institute of Technology, Department of Chemistry and Applied Biosciences, Zurich CH-8093, Switzerland
- Philogen S.p.A., Siena I-53100, Italy
| | | | - Andrea Galbiati
- Philochem AG, R&D Department, CH-8112 Otelfingen, Switzerland
| |
Collapse
|
3
|
Liang Z, Guo Y, Sharma A, McCurdy CR, Prentice BM. Multimodal Image Fusion Workflow Incorporating MALDI Imaging Mass Spectrometry and Microscopy for the Study of Small Pharmaceutical Compounds. Anal Chem 2024; 96:11869-11880. [PMID: 38982936 PMCID: PMC11649305 DOI: 10.1021/acs.analchem.4c01553] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2024]
Abstract
Multimodal imaging analyses of dosed tissue samples can provide more comprehensive insights into the effects of a therapeutically active compound on a target tissue compared to single-modal imaging. For example, simultaneous spatial mapping of pharmaceutical compounds and endogenous macromolecule receptors is difficult to achieve in a single imaging experiment. Herein, we present a multimodal workflow combining imaging mass spectrometry with immunohistochemistry (IHC) fluorescence imaging and brightfield microscopy imaging. Imaging mass spectrometry enables direct mapping of pharmaceutical compounds and metabolites, IHC fluorescence imaging can visualize large proteins, and brightfield microscopy imaging provides tissue morphology information. Single-cell resolution images are generally difficult to acquire using imaging mass spectrometry but are readily acquired with IHC fluorescence and brightfield microscopy imaging. Spatial sharpening of mass spectrometry images would thus allow for higher fidelity coregistration with other higher-resolution microscopy images. Imaging mass spectrometry spatial resolution can be predicted to a finer value via a computational image fusion workflow, which models the relationship between the intensity values in the mass spectrometry image and the features of a high-spatial resolution microscopy image. As a proof of concept, our multimodal workflow was applied to brain tissue extracted from a Sprague-Dawley rat dosed with a kratom alkaloid, corynantheidine. Four candidate mathematical models, including linear regression, partial least-squares regression, random forest regression, and two-dimensional convolutional neural network (2-D CNN), were tested. The random forest and 2-D CNN models most accurately predicted the intensity values at each pixel as well as the overall patterns of the mass spectrometry images, while also providing the best spatial resolution enhancements. Herein, image fusion enabled predicted mass spectrometry images of corynantheidine, GABA, and glutamine to approximately 2.5 μm spatial resolutions, a significant improvement compared to the original images acquired at 25 μm spatial resolution. The predicted mass spectrometry images were then coregistered with an H&E image and IHC fluorescence image of the μ-opioid receptor to assess colocalization of corynantheidine with brain cells. Our study also provides insights into the different evaluation parameters to consider when utilizing image fusion for biological applications.
Collapse
Affiliation(s)
- Zhongling Liang
- Department of Chemistry, University of Florida, Gainesville, FL 32611
| | - Yingchan Guo
- Department of Chemistry, University of Florida, Gainesville, FL 32611
| | - Abhisheak Sharma
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610
| | - Christopher R. McCurdy
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610
| | - Boone M. Prentice
- Department of Chemistry, University of Florida, Gainesville, FL 32611
| |
Collapse
|
4
|
Sun T, Zhao H, Hu L, Shao X, Lu Z, Wang Y, Ling P, Li Y, Zeng K, Chen Q. Enhanced optical imaging and fluorescent labeling for visualizing drug molecules within living organisms. Acta Pharm Sin B 2024; 14:2428-2446. [PMID: 38828150 PMCID: PMC11143489 DOI: 10.1016/j.apsb.2024.01.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/07/2024] [Accepted: 01/25/2024] [Indexed: 06/05/2024] Open
Abstract
The visualization of drugs in living systems has become key techniques in modern therapeutics. Recent advancements in optical imaging technologies and molecular design strategies have revolutionized drug visualization. At the subcellular level, super-resolution microscopy has allowed exploration of the molecular landscape within individual cells and the cellular response to drugs. Moving beyond subcellular imaging, researchers have integrated multiple modes, like optical near-infrared II imaging, to study the complex spatiotemporal interactions between drugs and their surroundings. By combining these visualization approaches, researchers gain supplementary information on physiological parameters, metabolic activity, and tissue composition, leading to a comprehensive understanding of drug behavior. This review focuses on cutting-edge technologies in drug visualization, particularly fluorescence imaging, and the main types of fluorescent molecules used. Additionally, we discuss current challenges and prospects in targeted drug research, emphasizing the importance of multidisciplinary cooperation in advancing drug visualization. With the integration of advanced imaging technology and molecular design, drug visualization has the potential to redefine our understanding of pharmacology, enabling the analysis of drug micro-dynamics in subcellular environments from new perspectives and deepening pharmacological research to the levels of the cell and organelles.
Collapse
Affiliation(s)
- Ting Sun
- School of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery System, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
- Institute of Biochemical and Biotechnological Drugs, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Huanxin Zhao
- School of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery System, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
| | - Luyao Hu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xintian Shao
- School of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery System, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
- School of Life Sciences, Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
| | - Zhiyuan Lu
- School of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery System, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
| | - Yuli Wang
- Tianjin Pharmaceutical DA REN TANG Group Corporation Limited Traditional Chinese Pharmacy Research Institute, Tianjin 300457, China
- Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemistry Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Peixue Ling
- Institute of Biochemical and Biotechnological Drugs, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Key Laboratory of Biopharmaceuticals, Postdoctoral Scientific Research Workstation, Shandong Academy of Pharmaceutical Science, Jinan 250098, China
| | - Yubo Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Kewu Zeng
- School of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery System, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Qixin Chen
- School of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery System, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
| |
Collapse
|
5
|
Rubahamya B, Dong S, Thurber GM. Clinical translation of antibody drug conjugate dosing in solid tumors from preclinical mouse data. SCIENCE ADVANCES 2024; 10:eadk1894. [PMID: 38820153 PMCID: PMC11141632 DOI: 10.1126/sciadv.adk1894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 04/29/2024] [Indexed: 06/02/2024]
Abstract
Antibody drug conjugates (ADCs) have made impressive strides in the clinic in recent years with 11 Food and Drug Administration approvals, including 6 for the treatment of patients with solid tumors. Despite this success, the development of new agents remains challenging with a high failure rate in the clinic. Here, we show that current approved ADCs for the treatment of patients with solid tumors can all show substantial efficacy in some mouse models when administered at a similar weight-based [milligrams per kilogram (mg/kg)] dosing in mice that is tolerated in the clinic. Mechanistically, equivalent mg/kg dosing results in a similar drug concentration in the tumor and a similar tissue penetration into the tumor due to the unique delivery features of ADCs. Combined with computational approaches, which can account for the complex distribution within the tumor microenvironment, these scaling concepts may aid in the evaluation of new agents and help design therapeutics with maximum clinical efficacy.
Collapse
Affiliation(s)
- Baron Rubahamya
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shujun Dong
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Greg M. Thurber
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
6
|
Kawaguchi M, Yonetani Y, Mizuguchi T, Spratt SJ, Asanuma M, Shimizu H, Sasaki M, Ozeki Y. Visualization of Modified Bisarylbutadiyne-Tagged Small Molecules in Live-Cell Nuclei by Stimulated Raman Scattering Microscopy. Anal Chem 2024; 96:6643-6651. [PMID: 38626411 DOI: 10.1021/acs.analchem.3c05946] [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: 04/18/2024]
Abstract
Visualizing the distribution of small-molecule drugs in living cells is an important strategy for developing specific, effective, and minimally toxic drugs. As an alternative to fluorescence imaging using bulky fluorophores or cell fixation, stimulated Raman scattering (SRS) imaging combined with bisarylbutadiyne (BADY) tagging enables the observation of small molecules closer to their native intracellular state. However, there is evidence that the physicochemical properties of BADY-tagged analogues of small-molecule drugs differ significantly from those of their parent drugs, potentially affecting their intracellular distribution. Herein, we developed a modified BADY to reduce deviations in physicochemical properties (in particular, lipophilicity and membrane permeability) between tagged and parent drugs, while maintaining high Raman activity in live-cell SRS imaging. We highlight the practical application of this approach by revealing the nuclear distribution of a modified BADY-tagged analogue of JQ1, a bromodomain and extra-terminal motif inhibitor with applications in targeted cancer therapy, in living HeLa cells. The modified BADY, methoxypyridazyl pyrimidyl butadiyne (MPDY), revealed intranuclear JQ1, while BADY-tagged JQ1 did not show a clear nuclear signal. We anticipate that the present approach combining MPDY tagging with live-cell SRS imaging provides important insight into the behavior of intracellular drugs and represents a promising avenue for improving drug development.
Collapse
Affiliation(s)
| | - Yuki Yonetani
- Future Technology R&D Center, Canon Inc., Tokyo 146-8501, Japan
| | - Takaha Mizuguchi
- Research Center for Advanced Science and Technology (RCAST), The University of Tokyo, Tokyo 153-8904, Japan
| | - Spencer J Spratt
- Research Center for Advanced Science and Technology (RCAST), The University of Tokyo, Tokyo 153-8904, Japan
| | - Masato Asanuma
- Department of Electrical Engineering and Information Systems, The University of Tokyo, Tokyo 113-8656, Japan
| | - Hiroki Shimizu
- Organic & Biomolecular Chemistry Department, Daiichi Sankyo RD Novare Co., Ltd., Tokyo 134-8630, Japan
| | - Masato Sasaki
- Organic & Biomolecular Chemistry Department, Daiichi Sankyo RD Novare Co., Ltd., Tokyo 134-8630, Japan
| | - Yasuyuki Ozeki
- Research Center for Advanced Science and Technology (RCAST), The University of Tokyo, Tokyo 153-8904, Japan
- Department of Electrical Engineering and Information Systems, The University of Tokyo, Tokyo 113-8656, Japan
| |
Collapse
|
7
|
Kinnunen PC, Humphries BA, Luker GD, Luker KE, Linderman JJ. Characterizing heterogeneous single-cell dose responses computationally and experimentally using threshold inhibition surfaces and dose-titration assays. NPJ Syst Biol Appl 2024; 10:42. [PMID: 38637530 PMCID: PMC11026493 DOI: 10.1038/s41540-024-00369-x] [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: 12/16/2023] [Accepted: 04/08/2024] [Indexed: 04/20/2024] Open
Abstract
Single cancer cells within a tumor exhibit variable levels of resistance to drugs, ultimately leading to treatment failures. While tumor heterogeneity is recognized as a major obstacle to cancer therapy, standard dose-response measurements for the potency of targeted kinase inhibitors aggregate populations of cells, obscuring intercellular variations in responses. In this work, we develop an analytical and experimental framework to quantify and model dose responses of individual cancer cells to drugs. We first explore the connection between population and single-cell dose responses using a computational model, revealing that multiple heterogeneous populations can yield nearly identical population dose responses. We demonstrate that a single-cell analysis method, which we term a threshold inhibition surface, can differentiate among these populations. To demonstrate the applicability of this method, we develop a dose-titration assay to measure dose responses in single cells. We apply this assay to breast cancer cells responding to phosphatidylinositol-3-kinase inhibition (PI3Ki), using clinically relevant PI3Kis on breast cancer cell lines expressing fluorescent biosensors for kinase activity. We demonstrate that MCF-7 breast cancer cells exhibit heterogeneous dose responses with some cells requiring over ten-fold higher concentrations than the population average to achieve inhibition. Our work reimagines dose-response relationships for cancer drugs in an emerging paradigm of single-cell tumor heterogeneity.
Collapse
Affiliation(s)
- Patrick C Kinnunen
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Brock A Humphries
- Department of Radiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Gary D Luker
- Department of Radiology, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kathryn E Luker
- Department of Radiology, University of Michigan, Ann Arbor, MI, 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jennifer J Linderman
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
8
|
Liang Z, Guo Y, Sharma A, McCurdy CR, Prentice BM. A multi-modal image fusion workflow incorporating MALDI imaging mass spectrometry and microscopy for the study of small pharmaceutical compounds. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.12.584673. [PMID: 38559145 PMCID: PMC10980041 DOI: 10.1101/2024.03.12.584673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Multi-modal imaging analyses of dosed tissue samples can provide more comprehensive insight into the effects of a therapeutically active compound on a target tissue compared to single-modal imaging. For example, simultaneous spatial mapping of pharmaceutical compounds and endogenous macromolecule receptors is difficult to achieve in a single imaging experiment. Herein, we present a multi-modal workflow combining imaging mass spectrometry with immunohistochemistry (IHC) fluorescence imaging and brightfield microscopy imaging. Imaging mass spectrometry enables direct mapping of pharmaceutical compounds and metabolites, IHC fluorescence imaging can visualize large proteins, and brightfield microscopy imaging provides tissue morphology information. Single-cell resolution images are generally difficult to acquire using imaging mass spectrometry, but are readily acquired with IHC fluorescence and brightfield microscopy imaging. Spatial sharpening of mass spectrometry images would thus allow for higher fidelity co-registration with higher resolution microscopy images. Imaging mass spectrometry spatial resolution can be predicted to a finer value via a computational image fusion workflow, which models the relationship between the intensity values in the mass spectrometry image and the features of a high spatial resolution microscopy image. As a proof of concept, our multi-modal workflow was applied to brain tissue extracted from a Sprague Dawley rat dosed with a kratom alkaloid, corynantheidine. Four candidate mathematical models including linear regression, partial least squares regression (PLS), random forest regression, and two-dimensional convolutional neural network (2-D CNN), were tested. The random forest and 2-D CNN models most accurately predicted the intensity values at each pixel as well as the overall patterns of the mass spectrometry images, while also providing the best spatial resolution enhancements. Herein, image fusion enabled predicted mass spectrometry images of corynantheidine, GABA, and glutamine to approximately 2.5 μm spatial resolutions, a significant improvement compared to the original images acquired at 25 μm spatial resolution. The predicted mass spectrometry images were then co-registered with an H&E image and IHC fluorescence image of the μ-opioid receptor to assess co-localization of corynantheidine with brain cells. Our study also provides insight into the different evaluation parameters to consider when utilizing image fusion for biological applications.
Collapse
Affiliation(s)
- Zhongling Liang
- Department of Chemistry, University of Florida, Gainesville, FL 32611
| | - Yingchan Guo
- Department of Chemistry, University of Florida, Gainesville, FL 32611
| | - Abhisheak Sharma
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610
| | - Christopher R. McCurdy
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610
| | - Boone M. Prentice
- Department of Chemistry, University of Florida, Gainesville, FL 32611
| |
Collapse
|
9
|
Sarkar B, Arlauckas SP, Cuccarese MF, Garris CS, Weissleder R, Rodell CB. Host-functionalization of macrin nanoparticles to enable drug loading and control tumor-associated macrophage phenotype. Front Immunol 2024; 15:1331480. [PMID: 38545103 PMCID: PMC10965546 DOI: 10.3389/fimmu.2024.1331480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 02/26/2024] [Indexed: 04/09/2024] Open
Abstract
Macrophages are critical regulators of the tumor microenvironment and often present an immuno-suppressive phenotype, supporting tumor growth and immune evasion. Promoting a robust pro-inflammatory macrophage phenotype has emerged as a therapeutic modality that supports tumor clearance, including through synergy with immune checkpoint therapies. Polyglucose nanoparticles (macrins), which possess high macrophage affinity, are useful vehicles for delivering drugs to macrophages, potentially altering their phenotype. Here, we examine the potential of functionalized macrins, synthesized by crosslinking carboxymethyl dextran with L-lysine, as effective carriers of immuno-stimulatory drugs to tumor-associated macrophages (TAMs). Azide groups incorporated during particle synthesis provided a handle for click-coupling of propargyl-modified β-cyclodextrin to macrins under mild conditions. Fluorescence-based competitive binding assays revealed the ability of β-cyclodextrin to non-covalently bind to hydrophobic immuno-stimulatory drug candidates (Keq ~ 103 M-1), enabling drug loading within nanoparticles. Furthermore, transcriptional profiles of macrophages indicated robust pro-inflammatory reprogramming (elevated Nos2 and Il12; suppressed Arg1 and Mrc1 expression levels) for a subset of these immuno-stimulatory agents (UNC2025 and R848). Loading of R848 into the modified macrins improved the drug's effect on primary murine macrophages by three-fold in vitro. Intravital microscopy in IL-12-eYFP reporter mice (24 h post-injection) revealed a two-fold enhancement in mean YFP fluorescence intensity in macrophages targeted with R848-loaded macrins, relative to vehicle controls, validating the desired pro-inflammatory reprogramming of TAMs in vivo by cell-targeted drug delivery. Finally, in an intradermal MC38 tumor model, cyclodextrin-modified macrin NPs loaded with immunostimulatory drugs significantly reduced tumor growth. Therefore, efficient and effective repolarization of tumor-associated macrophages to an M1-like phenotype-via drug-loaded macrins-inhibits tumor growth and may be useful as an adjuvant to existing immune checkpoint therapies.
Collapse
Affiliation(s)
- Biplab Sarkar
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, United States
| | - Sean P. Arlauckas
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, United States
| | - Michael F. Cuccarese
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, United States
| | - Christopher S. Garris
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, United States
- Department of Pathology, Harvard Medical School, Boston, MA, United States
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, United States
- Department of Systems Biology, Harvard Medical School, Boston, MA, United States
| | - Christopher B. Rodell
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, United States
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
10
|
Rezaeian M, Heidari H, Raahemifar K, Soltani M. Image-Based Modeling of Drug Delivery during Intraperitoneal Chemotherapy in a Heterogeneous Tumor Nodule. Cancers (Basel) 2023; 15:5069. [PMID: 37894436 PMCID: PMC10604968 DOI: 10.3390/cancers15205069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/12/2023] [Accepted: 10/18/2023] [Indexed: 10/29/2023] Open
Abstract
Intraperitoneal (IP) chemotherapy is a promising treatment approach for patients diagnosed with peritoneal carcinomatosis, allowing the direct delivery of therapeutic agents to the tumor site within the abdominal cavity. Nevertheless, limited drug penetration into the tumor remains a primary drawback of this method. The process of delivering drugs to the tumor entails numerous complications, primarily stemming from the specific pathophysiology of the tumor. Investigating drug delivery during IP chemotherapy and studying the parameters affecting it are challenging due to the limitations of experimental studies. In contrast, mathematical modeling, with its capabilities such as enabling single-parameter studies, and cost and time efficiency, emerges as a potent tool for this purpose. In this study, we developed a numerical model to investigate IP chemotherapy by incorporating an actual image of a tumor with heterogeneous vasculature. The tumor's geometry is reconstructed using image processing techniques. The model also incorporates drug binding and uptake by cancer cells. After 60 min of IP treatment with Doxorubicin, the area under the curve (AUC) of the average free drug concentration versus time curve, serving as an indicator of drug availability to the tumor, reached 295.18 mol·m-3·s-1. Additionally, the half-width parameter W1/2, which reflects drug penetration into the tumor, ranged from 0.11 to 0.14 mm. Furthermore, the treatment resulted in a fraction of killed cells reaching 20.4% by the end of the procedure. Analyzing the spatial distribution of interstitial fluid velocity, pressure, and drug concentration in the tumor revealed that the heterogeneous distribution of tumor vasculature influences the drug delivery process. Our findings underscore the significance of considering the specific vascular network of a tumor when modeling intraperitoneal chemotherapy. The proposed methodology holds promise for application in patient-specific studies.
Collapse
Affiliation(s)
- Mohsen Rezaeian
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran 19967-15433, Iran;
| | - Hamidreza Heidari
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, University Heights, Newark, NJ 07102, USA;
| | - Kaamran Raahemifar
- Data Science and Artificial Intelligence Program, College of Information Sciences and Technology (IST), Penn State University, State College, PA 16801, USA;
- School of Optometry and Vision Science, Faculty of Science, University of Waterloo, Waterloo, ON N2L 3G1, Canada
- Department of Chemical Engineering, Faculty of Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Madjid Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran 19967-15433, Iran;
- Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada
- Advanced Bioengineering Initiative Center, Multidisciplinary International Complex, K. N. Toosi University of Technology, Tehran 19967-15433, Iran
- Computational Medicine Center, K. N. Toosi University of Technology, Tehran 19967-15433, Iran
- Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, ON N2L 3G1, Canada
| |
Collapse
|
11
|
Zhang C, Tian Z, Chen R, Rowan F, Qiu K, Sun Y, Guan JL, Diao J. Advanced imaging techniques for tracking drug dynamics at the subcellular level. Adv Drug Deliv Rev 2023; 199:114978. [PMID: 37385544 PMCID: PMC10527994 DOI: 10.1016/j.addr.2023.114978] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/17/2023] [Accepted: 06/26/2023] [Indexed: 07/01/2023]
Abstract
Optical microscopes are an important imaging tool that have effectively advanced the development of modern biomedicine. In recent years, super-resolution microscopy (SRM) has become one of the most popular techniques in the life sciences, especially in the field of living cell imaging. SRM has been used to solve many problems in basic biological research and has great potential in clinical application. In particular, the use of SRM to study drug delivery and kinetics at the subcellular level enables researchers to better study drugs' mechanisms of action and to assess the efficacy of their targets in vivo. The purpose of this paper is to review the recent advances in SRM and to highlight some of its applications in assessing subcellular drug dynamics.
Collapse
Affiliation(s)
- Chengying Zhang
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Zhiqi Tian
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Rui Chen
- Department of Chemistry, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Fiona Rowan
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Kangqiang Qiu
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Yujie Sun
- Department of Chemistry, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Jun-Lin Guan
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Jiajie Diao
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
| |
Collapse
|
12
|
Zana A, Puig-Moreno C, Bocci M, Gilardoni E, Di Nitto C, Principi L, Ravazza D, Rotta G, Prodi E, De Luca R, Neri D, Cazzamalli S. A Comparative Analysis of Fibroblast Activation Protein-Targeted Small Molecule-Drug, Antibody-Drug, and Peptide-Drug Conjugates. Bioconjug Chem 2023. [PMID: 37399501 DOI: 10.1021/acs.bioconjchem.3c00244] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2023]
Abstract
We present the first in vivo comparative evaluation of chemically defined antibody-drug conjugates (ADCs), small molecule-drug conjugates (SMDCs), and peptide-drug conjugates (PDCs) targeting and activated by fibroblast activation protein (FAP) in solid tumors. Both the SMDC (OncoFAP-Gly-Pro-MMAE) and the ADC (7NP2-Gly-Pro-MMAE) candidates delivered high amounts of active payload (i.e., MMAE) selectively at the tumor site, thus producing a potent antitumor activity in a preclinical cancer model.
Collapse
Affiliation(s)
- Aureliano Zana
- Philochem AG, R&D Department, Libernstrasse 3, CH-8112 Otelfingen, Zürich, Switzerland
| | - Claudia Puig-Moreno
- Philochem AG, R&D Department, Libernstrasse 3, CH-8112 Otelfingen, Zürich, Switzerland
- Swiss Federal Institute of Technology, Department of Chemistry and Applied Biosciences, CH-8093 Zürich, Switzerland
| | - Matilde Bocci
- Philochem AG, R&D Department, Libernstrasse 3, CH-8112 Otelfingen, Zürich, Switzerland
| | - Ettore Gilardoni
- Philochem AG, R&D Department, Libernstrasse 3, CH-8112 Otelfingen, Zürich, Switzerland
| | - Cesare Di Nitto
- Philochem AG, R&D Department, Libernstrasse 3, CH-8112 Otelfingen, Zürich, Switzerland
| | - Lucrezia Principi
- Philochem AG, R&D Department, Libernstrasse 3, CH-8112 Otelfingen, Zürich, Switzerland
| | - Domenico Ravazza
- Philochem AG, R&D Department, Libernstrasse 3, CH-8112 Otelfingen, Zürich, Switzerland
| | - Giulia Rotta
- Philochem AG, R&D Department, Libernstrasse 3, CH-8112 Otelfingen, Zürich, Switzerland
| | - Eleonora Prodi
- Philochem AG, R&D Department, Libernstrasse 3, CH-8112 Otelfingen, Zürich, Switzerland
| | - Roberto De Luca
- Philochem AG, R&D Department, Libernstrasse 3, CH-8112 Otelfingen, Zürich, Switzerland
| | - Dario Neri
- Philochem AG, R&D Department, Libernstrasse 3, CH-8112 Otelfingen, Zürich, Switzerland
- Swiss Federal Institute of Technology, Department of Chemistry and Applied Biosciences, CH-8093 Zürich, Switzerland
- Philogen S.p.A., 53100 Siena, Italy
| | - Samuele Cazzamalli
- Philochem AG, R&D Department, Libernstrasse 3, CH-8112 Otelfingen, Zürich, Switzerland
| |
Collapse
|
13
|
Rios AC, van Rheenen J, Scheele CLGJ. Multidimensional Imaging of Breast Cancer. Cold Spring Harb Perspect Med 2023; 13:a041330. [PMID: 36167726 PMCID: PMC10153799 DOI: 10.1101/cshperspect.a041330] [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: 11/25/2022]
Abstract
Breast cancer is a pathological condition characterized by high morphological and molecular heterogeneity. Not only the breast cancer cells, but also their tumor micro-environment consists of a multitude of cell types and states, which continuously evolve throughout progression of the disease. To understand breast cancer evolution within this complex environment, in situ analysis of breast cancer and their co-evolving cells and structures in space and time are essential. In this review, recent technical advances in three-dimensional (3D) and intravital imaging of breast cancer are discussed. Moreover, we highlight the resulting new knowledge on breast cancer biology obtained through these innovative imaging technologies. Finally, we discuss how multidimensional imaging technologies can be integrated with molecular profiling to understand the full complexity of breast cancer and the tumor micro-environment during tumor progression and treatment response.
Collapse
Affiliation(s)
- Anne C Rios
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
- Oncode Institute, 3521 AL Utrecht, The Netherlands
| | - Jacco van Rheenen
- Oncode Institute, 3521 AL Utrecht, The Netherlands
- Department of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Colinda L G J Scheele
- Laboratory for Intravital Imaging and Dynamics of Tumor Progression, VIB Center for Cancer Biology, KU Leuven, 3000 Leuven, Belgium
- Department of Oncology, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
14
|
Demétrio de Souza França P, Viray T, Roberts S, Michel A, Abrahão M, Patel SG, Ganly I, Schöder H, Brand C, Reiner T, Pillarsetty NVK. Polyethylene Glycol 3350 (PEG 3350) as a Practical Vehicle for Rapid Reconstitution of PARPi-FL Formulations for Clinical Use. Mol Imaging Biol 2023; 25:294-302. [PMID: 35882728 PMCID: PMC11225571 DOI: 10.1007/s11307-022-01756-8] [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: 04/26/2022] [Revised: 07/07/2022] [Accepted: 07/08/2022] [Indexed: 01/18/2023]
Abstract
PARPi-FL is a molecularly specific fluorescent agent that targets poly ADP-ribose polymerase 1, a DNA repair enzyme overexpressed in the nuclei of tumor cells. This imaging agent is being investigated in a clinical trial (NCT03085147) for the detection of oral cancer. The PARPi-FL mouthwash formulation currently being used in the phase I/II clinical trial comprises 1,000 nM of PARPi-FL dissolved first in 4.5 ml of polyethylene glycol (PEG) 300 and then in 9.5 ml of water. This formulation requires a 2-step process that can be cumbersome for routine clinical use. To minimize errors and simplify the formulation process, we have developed a new one-step formulation, which requires only the direct addition of water into a vial containing a mixture of the PARPi-FL and PEG 3350, which is also a powder. In a series of analytical and preclinical studies, we demonstrate that the new formulation of PARPi-FL is stable over 365 days, sustains its characteristics, and performs similar to the previous formulation. Moving forward, the new formulation of the PARPi-FL will be used for patients accrued in the phase II clinical trial.
Collapse
Affiliation(s)
- Paula Demétrio de Souza França
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Otorhinolaryngology and Head and Neck Surgery, Federal University of São Paulo, São Paulo, SP, Brazil.
| | - Tara Viray
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sheryl Roberts
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alexa Michel
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marcio Abrahão
- Department of Otorhinolaryngology and Head and Neck Surgery, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Snehal G Patel
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ian Ganly
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Heiko Schöder
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Thomas Reiner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Weill Cornell Medical College, New York, NY, USA
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Naga Vara Kishore Pillarsetty
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Radiology, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
15
|
Zana A, Galbiati A, Gilardoni E, Bocci M, Millul J, Sturm T, Stucchi R, Elsayed A, Nadal L, Cirillo M, Roll W, Stegger L, Asmus I, Backhaus P, Schäfers M, Neri D, Cazzamalli S. Fibroblast Activation Protein Triggers Release of Drug Payload from Non-internalizing Small Molecule Drug Conjugates in Solid Tumors. Clin Cancer Res 2022; 28:5440-5454. [PMID: 36215129 DOI: 10.1158/1078-0432.ccr-22-1788] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 09/08/2022] [Accepted: 10/06/2022] [Indexed: 01/24/2023]
Abstract
PURPOSE Small molecule drug conjugates (SMDC) are modular anticancer prodrugs that include a tumor-targeting small organic ligand, a cleavable linker, and a potent cytotoxic agent. Most of the SMDC products that have been developed for clinical applications target internalizing tumor-associated antigens on the surface of tumor cells. We have recently described a novel non-internalizing small organic ligand (named OncoFAP) of fibroblast activation protein (FAP), a tumor-associated antigen highly expressed in the stroma of most solid human malignancies. EXPERIMENTAL DESIGN In this article, we describe a new series of OncoFAP-Drug derivatives based on monomethyl auristatin E (MMAE; a potent cytotoxic tubulin poison) and dipeptide linkers that are selectively cleaved by FAP in the tumor microenvironment. RESULTS The tumor-targeting potential of OncoFAP was confirmed in patients with cancer using nuclear medicine procedures. We used mass spectrometry methodologies to quantify the amount of prodrug delivered to tumors and normal organs, as well as the efficiency of the drug release process. Linkers previously exploited for anticancer drug conjugates were used as benchmark. We identified OncoFAP-Gly-Pro-MMAE as the best performing SMDC, which has now been prioritized for further clinical development. OncoFAP-Gly-Pro-MMAE selectively delivered more than 10% injected dose per gram of MMAE to FAP-positive tumors, with a tumor-to-kidney ratio of 16:1 at 24 hours post-injection. CONCLUSIONS The FAP-specific drug conjugates described in this article promise to be efficacious for the targeting of human malignancies. The extracellular release of potent anticancer payloads mediates durable complete remission in difficult-to-treat animal models of cancer.
Collapse
Affiliation(s)
- Aureliano Zana
- R&D Department, Philochem AG, Otelfingen, Zurich, Switzerland
| | - Andrea Galbiati
- R&D Department, Philochem AG, Otelfingen, Zurich, Switzerland
| | | | - Matilde Bocci
- R&D Department, Philochem AG, Otelfingen, Zurich, Switzerland
| | - Jacopo Millul
- R&D Department, Philochem AG, Otelfingen, Zurich, Switzerland
| | - Theo Sturm
- R&D Department, Philochem AG, Otelfingen, Zurich, Switzerland
| | | | - Abdullah Elsayed
- R&D Department, Philochem AG, Otelfingen, Zurich, Switzerland.,Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology, Zürich, Switzerland
| | - Lisa Nadal
- R&D Department, Philochem AG, Otelfingen, Zurich, Switzerland
| | - Martina Cirillo
- Department of Chemistry "G. Ciamician," University of Bologna, Bologna, Italy
| | - Wolfgang Roll
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
| | - Lars Stegger
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
| | - Inga Asmus
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
| | - Philipp Backhaus
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany.,European Institute for Molecular Imaging, University of Münster, Münster, Germany
| | - Michael Schäfers
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany.,European Institute for Molecular Imaging, University of Münster, Münster, Germany
| | - Dario Neri
- R&D Department, Philochem AG, Otelfingen, Zurich, Switzerland.,Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology, Zürich, Switzerland.,Philogen S.p.A., Siena, Italy
| | | |
Collapse
|
16
|
Poitout-Belissent F, Vitsky A, Smith MA, Sirivelu MP. Methodologies and Emerging Technologies for the Evaluation of the Hematopoietic System. Toxicol Pathol 2022; 50:867-870. [DOI: 10.1177/01926233221128755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Hematology and bone marrow analysis is central to our understanding of the hematopoietic system and how it responds to insults, and this session presented during the 2022 STP symposium provided a review of current and novel approaches for the evaluation of the hematopoietic system in the context of nonclinical investigations. This publication summarizes the information presented on novel approaches for evaluation of the hematopoietic system using automated hematology analyzers, including details around the quantitative assessment of bone marrow cell suspensions as well as introducing several newly available hematology parameters. It was followed by a discussion on intravital microscopy and live cell imaging and how these methods can assist with de-risking hematopoiesis-associated safety concerns, and a review of recent assays using artificial intelligence for the evaluation of bone marrow.
Collapse
|
17
|
Intravital microscopy for real-time monitoring of drug delivery and nanobiological processes. Adv Drug Deliv Rev 2022; 189:114528. [PMID: 36067968 DOI: 10.1016/j.addr.2022.114528] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 08/10/2022] [Accepted: 08/30/2022] [Indexed: 01/24/2023]
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.
Collapse
|
18
|
Ko J, Lucas K, Kohler R, Halabi EA, Wilkovitsch M, Carlson JCT, Weissleder R. In Vivo Click Chemistry Enables Multiplexed Intravital Microscopy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200064. [PMID: 35750648 PMCID: PMC9405492 DOI: 10.1002/advs.202200064] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 04/25/2022] [Indexed: 05/14/2023]
Abstract
The ability to observe cells in live organisms is essential for understanding their function in complex in vivo milieus. A major challenge today has been the limited ability to perform higher multiplexing beyond four to six colors to define cell subtypes in vivo. Here, a click chemistry-based strategy is presented for higher multiplexed in vivo imaging in mouse models. The method uses a scission-accelerated fluorophore exchange (SAFE), which exploits a highly efficient bioorthogonal mechanism to completely remove fluorescent signal from antibody-labeled cells in vivo. It is shown that the SAFE-intravital microscopy imaging method allows 1) in vivo staining of specific cell types in dorsal and cranial window chambers of mice, 2) complete un-staining in minutes, 3) in vivo click chemistries at lower (µm) and thus non-toxic concentrations, and 4) the ability to perform in vivo cyclic imaging. The potential utility of the method is demonstrated by 12 color imaging of immune cells in live mice.
Collapse
Affiliation(s)
- Jina Ko
- Center for Systems BiologyMassachusetts General Hospital185 Cambridge St, CPZN 5206BostonMA02114USA
| | - Kilean Lucas
- Center for Systems BiologyMassachusetts General Hospital185 Cambridge St, CPZN 5206BostonMA02114USA
| | - Rainer Kohler
- Center for Systems BiologyMassachusetts General Hospital185 Cambridge St, CPZN 5206BostonMA02114USA
| | - Elias A. Halabi
- Center for Systems BiologyMassachusetts General Hospital185 Cambridge St, CPZN 5206BostonMA02114USA
| | - Martin Wilkovitsch
- Center for Systems BiologyMassachusetts General Hospital185 Cambridge St, CPZN 5206BostonMA02114USA
| | - Jonathan C. T. Carlson
- Center for Systems BiologyMassachusetts General Hospital185 Cambridge St, CPZN 5206BostonMA02114USA
- Department of MedicineMassachusetts General HospitalHarvard Medical SchoolBostonMA02114USA
| | - Ralph Weissleder
- Center for Systems BiologyMassachusetts General Hospital185 Cambridge St, CPZN 5206BostonMA02114USA
- Department of Systems BiologyHarvard Medical School200 Longwood AveBostonMA02115USA
| |
Collapse
|
19
|
Spruill ML, Maletic-Savatic M, Martin H, Li F, Liu X. Spatial analysis of drug absorption, distribution, metabolism, and toxicology using mass spectrometry imaging. Biochem Pharmacol 2022; 201:115080. [PMID: 35561842 PMCID: PMC9744413 DOI: 10.1016/j.bcp.2022.115080] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/03/2022] [Accepted: 05/04/2022] [Indexed: 12/14/2022]
Abstract
Mass spectrometry imaging (MSI) is emerging as a powerful analytical tool for detection, quantification, and simultaneous spatial molecular imaging of endogenous and exogenous molecules via in situ mass spectrometry analysis of thin tissue sections without the requirement of chemical labeling. The MSI generates chemically specific and spatially resolved ion distribution information for administered drugs and metabolites, which allows numerous applications for studies involving various stages of drug absorption, distribution, metabolism, excretion, and toxicity (ADMET). MSI-based pharmacokinetic imaging analysis provides a histological context and cellular environment regarding dynamic drug distribution and metabolism processes, and facilitates the understanding of the spatial pharmacokinetics and pharmacodynamic properties of drugs. Herein, we discuss the MSI's current technological developments that offer qualitative, quantitative, and spatial location information of small molecule drugs, antibody, and oligonucleotides macromolecule drugs, and their metabolites in preclinical and clinical tissue specimens. We highlight the macro and micro drug-distribution in the whole-body, brain, lung, liver, kidney, stomach, intestine tissue sections, organoids, and the latest applications of MSI in pharmaceutical ADMET studies.
Collapse
Affiliation(s)
- Michelle L Spruill
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA
| | - Mirjana Maletic-Savatic
- Department of Pediatrics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | | | - Feng Li
- Center for Drug Discovery and Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA; NMR and Drug Metabolism Core, Advanced Technology Cores, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Xinli Liu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA.
| |
Collapse
|
20
|
Abstract
Molecular imaging is used to improve the disease diagnosis, prognosis, monitoring of treatment in living subjects. Numerous molecular targets have been developed for various cellular and molecular processes in genetic, metabolic, proteomic, and cellular biologic level. Molecular imaging modalities such as Optical Imaging, Magnetic Resonance Imaging (MRI), Positron Emission Tomography (PET), Single Photon Emission Computed Tomography (SPECT), and Computed Tomography (CT) can be used to visualize anatomic, genetic, biochemical, and physiologic changes in vivo. For in vivo cell imaging, certain cells such as cancer cells, immune cells, stem cells could be labeled by direct and indirect labeling methods to monitor cell migration, cell activity, and cell effects in cell-based therapy. In case of cancer, it could be used to investigate biological processes such as cancer metastasis and to analyze the drug treatment process. In addition, transplanted stem cells and immune cells in cell-based therapy could be visualized and tracked to confirm the fate, activity, and function of cells. In conventional molecular imaging, cells can be monitored in vivo in bulk non-invasively with optical imaging, MRI, PET, and SPECT imaging. However, single cell imaging in vivo has been a great challenge due to an extremely high sensitive detection of single cell. Recently, there has been great attention for in vivo single cell imaging due to the development of single cell study. In vivo single imaging could analyze the survival or death, movement direction, and characteristics of a single cell in live subjects. In this article, we reviewed basic principle of in vivo molecular imaging and introduced recent studies for in vivo single cell imaging based on the concept of in vivo molecular imaging.
Collapse
Affiliation(s)
- Seongje Hong
- Department of Anatomy, College of Medicine, Chung-Ang University, Seoul 06974, Korea
| | - Siyeon Rhee
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kyung Oh Jung
- Department of Anatomy, College of Medicine, Chung-Ang University, Seoul 06974, Korea
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
21
|
Hong S, Rhee S, Jung KO. In vivo molecular and single cell imaging. BMB Rep 2022; 55:267-274. [PMID: 35651326 PMCID: PMC9252890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 04/11/2022] [Accepted: 04/29/2022] [Indexed: 02/21/2025] Open
Abstract
Molecular imaging is used to improve the disease diagnosis, prognosis, monitoring of treatment in living subjects. Numerous molecular targets have been developed for various cellular and molecular processes in genetic, metabolic, proteomic, and cellular biologic level. Molecular imaging modalities such as Optical Imaging, Magnetic Resonance Imaging (MRI), Positron Emission Tomography (PET), Single Photon Emission Computed Tomography (SPECT), and Computed Tomography (CT) can be used to visualize anatomic, genetic, biochemical, and physiologic changes in vivo. For in vivo cell imaging, certain cells such as cancer cells, immune cells, stem cells could be labeled by direct and indirect labeling methods to monitor cell migration, cell activity, and cell effects in cell-based therapy. In case of cancer, it could be used to investigate biological processes such as cancer metastasis and to analyze the drug treatment process. In addition, transplanted stem cells and immune cells in cell-based therapy could be visualized and tracked to confirm the fate, activity, and function of cells. In conventional molecular imaging, cells can be monitored in vivo in bulk non-invasively with optical imaging, MRI, PET, and SPECT imaging. However, single cell imaging in vivo has been a great challenge due to an extremely high sensitive detection of single cell. Recently, there has been great attention for in vivo single cell imaging due to the development of single cell study. In vivo single imaging could analyze the survival or death, movement direction, and characteristics of a single cell in live subjects. In this article, we reviewed basic principle of in vivo molecular imaging and introduced recent studies for in vivo single cell imaging based on the concept of in vivo molecular imaging. [BMB Reports 2022; 55(6): 267-274].
Collapse
Affiliation(s)
- Seongje Hong
- Department of Anatomy, College of Medicine, Chung-Ang University, Seoul 06974, Korea, CA 94305, USA
| | - Siyeon Rhee
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kyung Oh Jung
- Department of Anatomy, College of Medicine, Chung-Ang University, Seoul 06974, Korea, CA 94305, USA
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
22
|
Kaynak A, Davis HW, Kogan AB, Lee JH, Narmoneva DA, Qi X. Phosphatidylserine: The Unique Dual-Role Biomarker for Cancer Imaging and Therapy. Cancers (Basel) 2022; 14:2536. [PMID: 35626139 PMCID: PMC9139557 DOI: 10.3390/cancers14102536] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/06/2022] [Accepted: 05/17/2022] [Indexed: 01/27/2023] Open
Abstract
Cancer is among the leading causes of death worldwide. In recent years, many cancer-associated biomarkers have been identified that are used for cancer diagnosis, prognosis, screening, and early detection, as well as for predicting and monitoring carcinogenesis and therapeutic effectiveness. Phosphatidylserine (PS) is a negatively charged phospholipid which is predominantly located in the inner leaflet of the cell membrane. In many cancer cells, PS externalizes to the outer cell membrane, a process regulated by calcium-dependent flippases and scramblases. Saposin C coupled with dioleoylphosphatidylserine (SapC-DOPS) nanovesicle (BXQ-350) and bavituximab, (Tarvacin, human-mouse chimeric monoclonal antibodies) are cell surface PS-targeting drugs being tested in clinical trial for treating a variety of cancers. Additionally, a number of other PS-selective agents have been used to trigger cytotoxicity in tumor-associated endothelial cells or cancer cells in pre-clinical studies. Recent studies have demonstrated that upregulation of surface PS exposure by chemodrugs, radiation, and external electric fields can be used as a novel approach to sensitize cancer cells to PS-targeting anticancer drugs. The objectives of this review are to provide an overview of a unique dual-role of PS as a biomarker/target for cancer imaging and therapy, and to discuss PS-based anticancer strategies that are currently under active development.
Collapse
Affiliation(s)
- Ahmet Kaynak
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, USA; (A.K.); (J.-H.L.); (D.A.N.)
| | - Harold W. Davis
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA;
| | - Andrei B. Kogan
- Physics Department, University of Cincinnati, Cincinnati, OH 45221, USA;
| | - Jing-Huei Lee
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, USA; (A.K.); (J.-H.L.); (D.A.N.)
| | - Daria A. Narmoneva
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, USA; (A.K.); (J.-H.L.); (D.A.N.)
| | - Xiaoyang Qi
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, USA; (A.K.); (J.-H.L.); (D.A.N.)
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA;
| |
Collapse
|
23
|
Ng TSC, Hu H, Kronister S, Lee C, Li R, Gerosa L, Stopka SA, Burgenske DM, Khurana I, Regan MS, Vallabhaneni S, Putta N, Scott E, Matvey D, Giobbie-Hurder A, Kohler RH, Sarkaria JN, Parangi S, Sorger PK, Agar NYR, Jacene HA, Sullivan RJ, Buchbinder E, Mikula H, Weissleder R, Miller MA. Overcoming differential tumor penetration of BRAF inhibitors using computationally guided combination therapy. SCIENCE ADVANCES 2022; 8:eabl6339. [PMID: 35486732 PMCID: PMC9054019 DOI: 10.1126/sciadv.abl6339] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 03/15/2022] [Indexed: 05/02/2023]
Abstract
BRAF-targeted kinase inhibitors (KIs) are used to treat malignancies including BRAF-mutant non-small cell lung cancer, colorectal cancer, anaplastic thyroid cancer, and, most prominently, melanoma. However, KI selection criteria in patients remain unclear, as are pharmacokinetic/pharmacodynamic (PK/PD) mechanisms that may limit context-dependent efficacy and differentiate related drugs. To address this issue, we imaged mouse models of BRAF-mutant cancers, fluorescent KI tracers, and unlabeled drug to calibrate in silico spatial PK/PD models. Results indicated that drug lipophilicity, plasma clearance, faster target dissociation, and, in particular, high albumin binding could limit dabrafenib action in visceral metastases compared to other KIs. This correlated with retrospective clinical observations. Computational modeling identified a timed strategy for combining dabrafenib and encorafenib to better sustain BRAF inhibition, which showed enhanced efficacy in mice. This study thus offers principles of spatial drug action that may help guide drug development, KI selection, and combination.
Collapse
Affiliation(s)
- Thomas S. C. Ng
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Huiyu Hu
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Stefan Kronister
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Institute of Applied Synthetic Chemistry, Technische Universität Wien, Vienna, Austria
| | - Chanseo Lee
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Ran Li
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Luca Gerosa
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Sylwia A. Stopka
- Department of Neurosurgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Ishaan Khurana
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Michael S. Regan
- Department of Neurosurgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Sreeram Vallabhaneni
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Niharika Putta
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Ella Scott
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Dylan Matvey
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Anita Giobbie-Hurder
- Division of Biostatistics, Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Rainer H. Kohler
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Jann N. Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - Sareh Parangi
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Peter K. Sorger
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Nathalie Y. R. Agar
- Department of Neurosurgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Heather A. Jacene
- Department of Radiology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Ryan J. Sullivan
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Hannes Mikula
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Institute of Applied Synthetic Chemistry, Technische Universität Wien, Vienna, Austria
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Miles A. Miller
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
24
|
Abstract
Over the years, the engineering aspect of nanotechnology has been significantly exploited. Medical intervention strategies have been developed by leveraging existing molecular biology knowledge and combining it with nanotechnology tools to improve outcomes. However, little attention has been paid to harnessing the strengths of nanotechnology as a biological discovery tool. Fundamental understanding of controlling dynamic biological processes at the subcellular level is key to developing personalized therapeutic and diagnostic interventions. Single-cell analyses using intravital microscopy, expansion microscopy, and microfluidic-based platforms have been helping to better understand cell heterogeneity in healthy and diseased cells, a major challenge in oncology. Also, single-cell analysis has revealed critical signaling pathways and biological intracellular components with key biological functions. The physical manipulation enabled by nanotools can allow real-time monitoring of biological changes at a single-cell level by sampling intracellular fluid from the same cell. The formation of intercellular highways by nanotube-like structures has important clinical implications such as metastasis development. The integration of nanomaterials into optical and molecular imaging techniques has rendered valuable morphological, structural, and biological information. Nanoscale imaging unravels mechanisms of temporality by enabling the visualization of nanoscale dynamics never observed or measured between individual cells with standard biological techniques. The exceptional sensitivity of nanozymes, artificial enzymes, make them perfect components of the next-generation mobile diagnostics devices. Here, we highlight these impactful cancer-related biological discoveries enabled by nanotechnology and producing a paradigm shift in cancer research and oncology.
Collapse
Affiliation(s)
- Carolina Salvador-Morales
- Nanodelivery Systems and Devices Branch, Cancer Imaging Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Piotr Grodzinski
- Nanodelivery Systems and Devices Branch, Cancer Imaging Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Rockville, Maryland 20850, United States
| |
Collapse
|
25
|
Thapaliya ER, Usama SM, Patel NL, Feng Y, Kalen JD, St Croix B, Schnermann MJ. Cyanine Masking: A Strategy to Test Functional Group Effects on Antibody Conjugate Targeting. Bioconjug Chem 2022; 33:718-725. [PMID: 35389618 PMCID: PMC10506421 DOI: 10.1021/acs.bioconjchem.2c00083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Conjugates of small molecules and antibodies are broadly employed diagnostic and therapeutic agents. Appending a small molecule to an antibody often significantly impacts the properties of the resulting conjugate. Here, we detail a systematic study investigating the effect of various functional groups on the properties of antibody-fluorophore conjugates. This was done through the preparation and analysis of a series of masked heptamethine cyanines (CyMasks)-bearing amides with varied functional groups. These were designed to exhibit a broad range of physical properties, and include hydrophobic (-NMe2), pegylated (NH-PEG-8 or NH-PEG-24), cationic (NH-(CH2)2NMe3+), anionic (NH-(CH2)2SO3-), and zwitterionic (N-(CH2)2NMe3+)-(CH2)3SO3-) variants. The CyMask series was appended to monoclonal antibodies (mAbs) and analyzed for the effects on tumor targeting, clearance, and non-specific organ uptake. Among the series, zwitterionic and pegylated dye conjugates had the highest tumor-to-background ratio (TBR) and a low liver-to-background ratio. By contrast, the cationic and zwitterionic probes had high tumor signal and high TBR, although the latter also exhibited an elevated liver-to-background ratio (LBR). Overall, these studies provide a strategy to test the functional group effects and suggest that zwitterionic substituents possess an optimal combination of high tumor signal, TBR, and low LBR. These results suggest an appealing strategy to mask hydrophobic payloads, with the potential to improve the properties of bioconjugates in vivo.
Collapse
Affiliation(s)
- Ek Raj Thapaliya
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Syed Muhammad Usama
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Nimit L Patel
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, Maryland 21702, United States
| | - Yang Feng
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute, NIH, Frederick, Maryland 21702, United States
| | - Joseph D Kalen
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, Maryland 21702, United States
| | - Brad St Croix
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute, NIH, Frederick, Maryland 21702, United States
| | - Martin J Schnermann
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| |
Collapse
|
26
|
Kim J, Kim HC, Kowsari K, Yoon K, Yoo SS. Transcutaneous application of ultrasound enhances the effects of finasteride in a murine model of androgenic alopecia. Ultrasonography 2022; 41:382-393. [PMID: 34963257 PMCID: PMC8942741 DOI: 10.14366/usg.21186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 11/05/2021] [Accepted: 11/10/2021] [Indexed: 11/16/2022] Open
Abstract
PURPOSE The purpose of this study was to evaluate if transcutaneous application of low-intensity ultrasound can locally enhance the effects of finasteride on hair growth in a murine model of androgenic alopecia (AA). METHODS AA mice (injected twice per week with testosterone enanthate, n=11), under daily oral administration of finasteride, received 1-MHz ultrasound for 1 hour at the unilateral thigh area five times per week for 5 weeks. Non-thermal and non-cavitational ultrasound was delivered in a pulsed manner (55-ms pulse duration with a repetition frequency of 4 Hz). Skin temperature was measured during sonication, and the measurements were validated with numerical simulations of sonication-induced tissue temperature changes. Hair growth was assessed both photographically and histologically. RESULTS We found more hair growth on the sonicated thigh area than on the unsonicated thigh, beginning from week 3 through the end of the experiment. Histological analyses showed that the number of hair follicles doubled in the skin sections that received sonication compared to the unsonicated zone, with thicker follicular diameter and skin. An over five-fold increase was also observed in the anagen/telogen ratio in the sonicated area, suggesting an enhanced anagen phase. Skin temperature was unaltered by the administered sonication. CONCLUSION The findings of the present study suggest that pulsed application of ultrasound promotes hair growth, potentially by disrupting the binding of albumin to finasteride. This may suggest further applications to enhance the pharmacological effects of other relevant drugs exhibiting high plasma protein binding.
Collapse
Affiliation(s)
- Jaeho Kim
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Hwaseong, Korea
| | - Hyun-Chul Kim
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Kavin Kowsari
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Kyungho Yoon
- Center for Healthcare Robotics, Korea Institute of Science and Technology, Seoul, Korea
| | - Seung-Schik Yoo
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
27
|
Yoon HY, Lee D, Lim DK, Koo H, Kim K. Copper-Free Click Chemistry: Applications in Drug Delivery, Cell Tracking, and Tissue Engineering. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2107192. [PMID: 34752658 DOI: 10.1002/adma.202107192] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/27/2021] [Indexed: 06/13/2023]
Abstract
Traditionally, organic chemical reactions require organic solvents, toxic catalysts, heat, or high pressure. However, copper-free click chemistry has been shown to have favorable reaction rates and orthogonality in water, buffer solutions, and physiological conditions without toxic catalysts. Strain-promoted azide-alkyne cycloaddition and inverse electron-demand Diels-Alder reactions are representative of copper-free click chemistry. Artificial chemical reactions via click chemistry can also be used outside of the laboratory in a controllable manner on live cell surfaces, in the cytosol, and in living bodies. Consequently, copper-free click chemistry has many features that are of interest in biomedical research, and various new materials and strategies for its use have been proposed. Herein, recent remarkable trials that have used copper-free click chemistry are described, focusing on their applications in molecular imaging and therapy. The research is categorized as nanoparticles for drug delivery, imaging agents for cell tracking, and hydrogels for tissue engineering, which are rapidly advancing fields based on click chemistry. The content is based primarily on the experience with click chemistry-based biomaterials over the last 10 years.
Collapse
Affiliation(s)
- Hong Yeol Yoon
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul, 02792, South Korea
| | - Donghyun Lee
- Department of Medical Life Sciences, Department of Biomedicine & Health Sciences, and Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Dong-Kwon Lim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145, Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Heebeom Koo
- Department of Medical Life Sciences, Department of Biomedicine & Health Sciences, and Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Kwangmeyung Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul, 02792, South Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145, Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| |
Collapse
|
28
|
Barouki R, Audouze K, Becker C, Blaha L, Coumoul X, Karakitsios S, Klanova J, Miller GW, Price EJ, Sarigiannis D. The Exposome and Toxicology: A Win-Win Collaboration. Toxicol Sci 2022; 186:1-11. [PMID: 34878125 PMCID: PMC9019839 DOI: 10.1093/toxsci/kfab149] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The development of the exposome concept has been one of the hallmarks of environmental and health research for the last decade. The exposome encompasses the life course environmental exposures including lifestyle factors from the prenatal period onwards. It has inspired many research programs and is expected to influence environmental and health research, practices, and policies. Yet, the links bridging toxicology and the exposome concept have not been well developed. In this review, we describe how the exposome framework can interface with and influence the field of toxicology, as well as how the field of toxicology can help advance the exposome field by providing the needed mechanistic understanding of the exposome impacts on health. Indeed, exposome-informed toxicology is expected to emphasize several orientations including (1) developing approaches integrating multiple stressors, in particular chemical mixtures, as well as the interaction of chemicals with other stressors, (2) using mechanistic frameworks such as the adverse outcome pathways to link the different stressors with toxicity outcomes, (3) characterizing the mechanistic basis of long-term effects by distinguishing different patterns of exposures and further exploring the environment-DNA interface through genetic and epigenetic studies, and (4) improving the links between environmental and human health, in particular through a stronger connection between alterations in our ecosystems and human toxicology. The exposome concept provides the linkage between the complex environment and contemporary mechanistic toxicology. What toxicology can bring to exposome characterization is a needed framework for mechanistic understanding and regulatory outcomes in risk assessment.
Collapse
Affiliation(s)
- Robert Barouki
- Inserm UMR S-1124, Université de Paris, T3S, Paris F-75006, France
- Service de Biochimie métabolomique et protéomique, Hôpital Necker enfants malades, AP-HP, Paris, France
| | - Karine Audouze
- Inserm UMR S-1124, Université de Paris, T3S, Paris F-75006, France
| | - Christel Becker
- Inserm UMR S-1124, Université de Paris, T3S, Paris F-75006, France
| | - Ludek Blaha
- RECETOX, Faculty of Science, Masaryk University, Brno 60200, Czech Republic
| | - Xavier Coumoul
- Inserm UMR S-1124, Université de Paris, T3S, Paris F-75006, France
| | - Spyros Karakitsios
- Center for Interdisciplinary Research and Innovation, HERACLES Research Center on the Exposome and Health, Aristotle University of Thessaloniki, Thessaloniki 57001, Greece
- Enve.X, Thessaloniki 55133, Greece
| | - Jana Klanova
- RECETOX, Faculty of Science, Masaryk University, Brno 60200, Czech Republic
| | - Gary W Miller
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - Elliott J Price
- RECETOX, Faculty of Science, Masaryk University, Brno 60200, Czech Republic
- Faculty of Sports Studies, Masaryk University, Brno 62500, Czech Republic
| | - Denis Sarigiannis
- Center for Interdisciplinary Research and Innovation, HERACLES Research Center on the Exposome and Health, Aristotle University of Thessaloniki, Thessaloniki 57001, Greece
- Enve.X, Thessaloniki 55133, Greece
| |
Collapse
|
29
|
Analysis of the intracellular localization of amiodarone using live single-cell mass spectrometry. J Pharm Biomed Anal 2021; 205:114318. [PMID: 34418674 DOI: 10.1016/j.jpba.2021.114318] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/06/2021] [Accepted: 08/07/2021] [Indexed: 01/08/2023]
Abstract
Amiodarone is a well-known antiarrhythmic drug with side effects including phospholipidosis. However, it is not clear how amiodarone and its metabolites are localized in the cell. In the present study, the localization of amiodarone in the cytosol, vacuoles, and lipid droplets of a single HepG2 human hepatocellular carcinoma cell was determined directly using live single-cell mass spectrometry. The cytosol, vacuoles, and lipid droplets of a single HepG2 cell treated with amiodarone were separately captured using a nano-spray tip under a fluorescence microscope after visualizing the lipid droplets using a fluorescent probe. This assay showed a linearity in the measurement of amiodarone levels with R2 values of 0.9996 and 0.9998 in the cell lysates and serum, respectively. The peak intensities of amiodarone and its metabolites in lipid droplets and vacuoles were significantly higher than those in the cytosol, while those in lipid droplets were higher than those in vacuoles. Amiodarone metabolites were detected in both lipid droplets and the cytosol. Live single-cell mass spectrometry combined with fluorescence imaging demonstrated clear localization of amiodarone and its metabolites in lipid droplets separately from the vacuole. This assay system combined with fluorescence imaging could be useful for investigating the intracellular localization of various drugs and their metabolites.
Collapse
|
30
|
Sahu A, Cordero J, Wu X, Kossatz S, Harris U, Demetrio Desouza Franca P, Kurtansky NR, Everett N, Dusza S, Monnier J, Kumar P, Alessi-Fox C, Brand C, Roberts S, Kose K, Phillip W, Lee E, Jason Chen CS, Rossi A, Nehal K, Pulitzer M, Longo C, Halpern A, Reiner T, Rajadhyaksha M, Jain M. Combined PARP1-targeted nuclear contrast and reflectance contrast enhances confocal microscopic detection of basal cell carcinoma. J Nucl Med 2021; 63:912-918. [PMID: 34649941 DOI: 10.2967/jnumed.121.262600] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/26/2021] [Indexed: 11/16/2022] Open
Abstract
Reflectance confocal microscopy (RCM) with endogenous backscattered contrast can noninvasively image basal cell carcinomas (BCCs) in skin. However, BCCs present with high nuclear density and the relatively weak backscattering from nuclei impose a fundamental limit on contrast, detectability, and diagnostic accuracy. We investigated PARPi-FL, an exogenous nuclear poly (ADP-ribose) polymerase (PARP1)-targeted fluorescent contrast agent and fluorescence confocal microscopy (FCM) towards improving BCC diagnosis. Methods: We tested PARP1 expression in 95 BCC tissues using immunohistochemistry, followed by PARPi-FL staining in 32 fresh surgical BCC specimens. Diagnostic accuracy of PARPi-FL contrast was evaluated in 83 surgical specimens. Optimal parameters for trans-epidermal permeability of PARPi-FL through intact skin was tested ex vivo on 5 human skin specimens and in vivo in 3 adult Yorkshire pigs. Results: We found significantly higher PARP1 expression and PARPi-FL binding in BCCs, as compared to normal skin structures. Blinded reading of RCM-and-FCM images by two experts demonstrated a higher diagnostic accuracy for BCCs with combined fluorescence and reflectance contrast, as compared to RCM-alone. Optimal parameters (time and concentration) for PARPi-FL trans-epidermal permeation through intact skin were successfully determined. Conclusion: Combined fluorescence and reflectance contrast may improve noninvasive BCC diagnosis with confocal microscopy.
Collapse
Affiliation(s)
- Aditi Sahu
- Memorial Sloan Kettering Cancer Center, United States
| | - Jose Cordero
- University of Puerto Rico - Medical Sciences Campus
| | | | | | | | | | | | | | - Stephen Dusza
- Memorial Sloan Kettering Cancer Center, United States
| | | | | | | | | | | | - Kivanc Kose
- Memorial Sloan Kettering Cancer Center, United States
| | | | - Erica Lee
- Memorial Sloan Kettering Cancer Center, United States
| | | | - Anthony Rossi
- Memorial Sloan Kettering Cancer Center, United States
| | - Kishwer Nehal
- Memorial Sloan Kettering Cancer Center, United States
| | | | | | - Allan Halpern
- Memorial Sloan Kettering Cancer Center, United States
| | | | | | - Manu Jain
- Memorial Sloan Kettering Cancer Center, United States
| |
Collapse
|
31
|
Kuzma BA, Pence IJ, Greenfield DA, Ho A, Evans CL. Visualizing and quantifying antimicrobial drug distribution in tissue. Adv Drug Deliv Rev 2021; 177:113942. [PMID: 34437983 DOI: 10.1016/j.addr.2021.113942] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/11/2021] [Accepted: 08/18/2021] [Indexed: 12/15/2022]
Abstract
The biodistribution and pharmacokinetics of drugs are vital to the mechanistic understanding of their efficacy. Measuring antimicrobial drug efficacy has been challenging as plasma drug concentration is used as a surrogate for tissue drug concentration, yet typically does not reflect that at the intended site(s) of action. Utilizing an image-guided approach, it is feasible to accurately quantify the biodistribution and pharmacokinetics within the desired site(s) of action. We outline imaging modalities used in visualizing drug distribution with examples ranging from in vitro cellular drug uptake to clinical treatment of microbial infections. The imaging modalities of interest are: radio-labeling, magnetic resonance, mass spectrometry imaging, computed tomography, fluorescence, and Raman spectroscopy. We outline the progress, limitations, and future outlook for each methodology. Further advances in these optical approaches would benefit patients and researchers alike, as non-invasive imaging could yield more profound insights with a lower clinical burden than invasive measurement approaches used today.
Collapse
Affiliation(s)
- Benjamin A Kuzma
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston 02114, USA
| | - Isaac J Pence
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston 02114, USA
| | - Daniel A Greenfield
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston 02114, USA
| | - Alexander Ho
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston 02114, USA
| | - Conor L Evans
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston 02114, USA.
| |
Collapse
|
32
|
Weigelin B, den Boer AT, Wagena E, Broen K, Dolstra H, de Boer RJ, Figdor CG, Textor J, Friedl P. Cytotoxic T cells are able to efficiently eliminate cancer cells by additive cytotoxicity. Nat Commun 2021; 12:5217. [PMID: 34471116 PMCID: PMC8410835 DOI: 10.1038/s41467-021-25282-3] [Citation(s) in RCA: 137] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 07/19/2021] [Indexed: 02/07/2023] Open
Abstract
Lethal hit delivery by cytotoxic T lymphocytes (CTL) towards B lymphoma cells occurs as a binary, "yes/no" process. In non-hematologic solid tumors, however, CTL often fail to kill target cells during 1:1 conjugation. Here we describe a mechanism of "additive cytotoxicity" by which time-dependent integration of sublethal damage events, delivered by multiple CTL transiting between individual tumor cells, mediates effective elimination. Reversible sublethal damage includes perforin-dependent membrane pore formation, nuclear envelope rupture and DNA damage. Statistical modeling reveals that 3 serial hits delivered with decay intervals below 50 min discriminate between tumor cell death or survival after recovery. In live melanoma lesions in vivo, sublethal multi-hit delivery is most effective in interstitial tissue where high CTL densities and swarming support frequent serial CTL-tumor cell encounters. This identifies CTL-mediated cytotoxicity by multi-hit delivery as an incremental and tunable process, whereby accelerating damage magnitude and frequency may improve immune efficacy.
Collapse
Affiliation(s)
- Bettina Weigelin
- Department of Cell Biology, RIMLS, Radboud University Medical Center, Nijmegen, The Netherlands.
- David H. Koch Center for Applied Research of Genitourinary Cancers, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany.
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tuebingen, Tübingen, Germany.
| | | | - Esther Wagena
- Department of Cell Biology, RIMLS, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Kelly Broen
- Department of Laboratory Medicine - Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Harry Dolstra
- Department of Laboratory Medicine - Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Rob J de Boer
- Theoretical Biology and Bioinformatics, Utrecht University, Utrecht, The Netherlands
| | - Carl G Figdor
- Department of Tumor Immunology, RIMLS, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Johannes Textor
- Department of Tumor Immunology, RIMLS, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Peter Friedl
- Department of Cell Biology, RIMLS, Radboud University Medical Center, Nijmegen, The Netherlands.
- David H. Koch Center for Applied Research of Genitourinary Cancers, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Cancer Genomics Centre Netherlands (CGC.nl), Utrecht, The Netherlands.
| |
Collapse
|
33
|
Drug transport kinetics of intravascular triggered drug delivery systems. Commun Biol 2021; 4:920. [PMID: 34321602 PMCID: PMC8319190 DOI: 10.1038/s42003-021-02428-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 07/05/2021] [Indexed: 01/17/2023] Open
Abstract
Intravascular triggered drug delivery systems (IV-DDS) for local drug delivery include various stimuli-responsive nanoparticles that release the associated agent in response to internal (e.g., pH, enzymes) or external stimuli (e.g., temperature, light, ultrasound, electromagnetic fields, X-rays). We developed a computational model to simulate IV-DDS drug delivery, for which we quantified all model parameters in vivo in rodent tumors. The model was validated via quantitative intravital microscopy studies with unencapsulated fluorescent dye, and with two formulations of temperature-sensitive liposomes (slow, and fast release) encapsulating a fluorescent dye as example IV-DDS. Tumor intra- and extravascular dye concentration dynamics were extracted from the intravital microscopy data by quantitative image processing, and were compared to computer model results. Via this computer model we explain IV-DDS delivery kinetics and identify parameters of IV-DDS, of drug, and of target tissue for optimal delivery. Two parameter ratios were identified that exclusively dictate how much drug can be delivered with IV-DDS, indicating the importance of IV-DDS with fast drug release (~sec) and choice of a drug with rapid tissue uptake (i.e., high first-pass extraction fraction). The computational model thus enables engineering of improved future IV-DDS based on tissue parameters that can be quantified by imaging. Ten Hagen et al. developed a computational model to simulate intravascular triggered drug delivery systems (IV-DDS), which they validated using quantitative intravital microscopy in mice. Their model potentially enables the engineering of more efficacious IV-DDS based on parameters that can be quantified by imaging.
Collapse
|
34
|
Giampetraglia M, Weigelin B. Recent advances in intravital microscopy for preclinical research. Curr Opin Chem Biol 2021; 63:200-208. [PMID: 34274700 DOI: 10.1016/j.cbpa.2021.05.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 05/08/2021] [Accepted: 05/14/2021] [Indexed: 12/14/2022]
Abstract
Intravital microscopy (IVM) has revolutionized our understanding of single-cell behavior in complex tissues by enabling real-time observation of molecular and cellular processes in their natural environment. In preclinical research, IVM has emerged as a standard tool for mechanistic studies of therapy response and the rational design of new treatment strategies. Technological developments keep expanding the imaging depth and quality that can be achieved in living tissue, and the maturation of imaging modalities such as fluorescence and phosphorescence lifetime imaging facilitates co-registration of individual cell dynamics with metabolic tissue states. Correlation of IVM with mesoscopic and macroscopic imaging modalities further promotes the translation of mechanistic insights gained by IVM into clinically relevant information. This review highlights some of the recent advances in IVM that have made the transition from experimental optical techniques to practical applications in basic and preclinical research.
Collapse
Affiliation(s)
- Martina Giampetraglia
- David H. Koch Center for Applied Research of Genitourinary Cancers, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Bettina Weigelin
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany; Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Germany.
| |
Collapse
|
35
|
de Souza França PD, Guru N, Kostolansky AR, Mauguen A, Pirovano G, Kossatz S, Roberts S, Abrahão M, Patel SG, Park KJ, Reiner T, Jewell E. PARP1: A Potential Molecular Marker to Identify Cancer During Colposcopy Procedures. J Nucl Med 2021; 62:941-948. [PMID: 33188153 PMCID: PMC8882878 DOI: 10.2967/jnumed.120.253575] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 10/16/2020] [Indexed: 11/16/2022] Open
Abstract
Despite efforts in prevention, cervical cancer still presents with a high worldwide incidence and remains a great problem in public health, especially in low-income countries. Screening programs, such as colposcopy with Papanicolaou testing, have greatly improved mortality rates. However, the agents currently used to delineate those lesions (topical application of acetic acid or Lugol iodine) lack specificity and sometimes can lead to unnecessary biopsies or even cervical excisions. A tool to enable in vivo histology to quickly and quantitatively distinguish between tumor, dysplastic tissue, and healthy tissue would be of great clinical interest. Methods: Here, we describe the use of PARPi-FL, a fluorescent inhibitor of poly[adenosine diphosphate-ribose]polymerase 1 (PARP1), which is a nuclear enzyme that is overexpressed in cancer when compared with the normal surrounding tissues. We exploit its use as an optical imaging agent to specifically target PARP1 expression, which was demonstrated to be higher in cervical cancer than the normal surrounding tissue. Results: After topical application of PARPi-FL on freshly excised cone biopsy samples, the nuclei of tumor cells emitted a specific fluorescent signal that could be visualized using a handheld fluorescence confocal microscope. Conclusion: This approach has the potential to improve in vivo identification of tumor cells during colposcopy examination, allowing a rapid, noninvasive, and accurate histopathologic assessment.
Collapse
Affiliation(s)
- Paula Demétrio de Souza França
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Otorhinolaryngology and Head and Neck Surgery, Federal University of São Paulo, São Paulo, Brazil
| | - Navjot Guru
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Abigail R Kostolansky
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Chemistry, Princeton University, Princeton, New Jersey
| | - Audrey Mauguen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Giacomo Pirovano
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Susanne Kossatz
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Nuclear Medicine, University Hospital Klinikum Rechts der Isar and TranslaTUM, Technical University Munich, Munich, Germany
| | - Sheryl Roberts
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Marcio Abrahão
- Department of Otorhinolaryngology and Head and Neck Surgery, Federal University of São Paulo, São Paulo, Brazil
| | - Snehal G Patel
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kay J Park
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - 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
| | - Elizabeth Jewell
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
36
|
Claveau S, Kindermann M, Papine A, Díaz-Riascos ZV, Délen X, Georges P, López-Alemany R, Tirado ÒM, Bertrand JR, Abasolo I, Cigler P, Treussart F. Harnessing subcellular-resolved organ distribution of cationic copolymer-functionalized fluorescent nanodiamonds for optimal delivery of active siRNA to a xenografted tumor in mice. NANOSCALE 2021; 13:9280-9292. [PMID: 33982741 DOI: 10.1039/d1nr00146a] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Diamond nanoparticles (nanodiamonds) can transport active drugs in cultured cells as well as in vivo. However, in the latter case, methods allowing the determination of their bioavailability accurately are still lacking. A nanodiamond can be made fluorescent with a perfectly stable emission and a lifetime ten times longer than that of tissue autofluorescence. Taking advantage of these properties, we present an automated quantification method of fluorescent nanodiamonds (FND) in histological sections of mouse organs and tumors, after systemic injection. We use a home-made time-delayed fluorescence microscope comprising a custom pulsed laser source synchronized on the master clock of a gated intensified array detector. This setup allows ultra-high-resolution images (120 Mpixels in size) of whole mouse organ sections to be obtained, with subcellular resolution and single-particle sensitivity. As a proof-of-principle experiment, we quantified the biodistribution and aggregation state of new cationic FNDs capable of transporting small interfering RNA inhibiting the oncogene responsible for Ewing sarcoma. Image analysis showed a low yield of nanodiamonds in the tumor after intravenous injection. Thus, for the in vivo efficacy assay, we injected the nanomedicine into the tumor. We achieved a 28-fold inhibition of the oncogene. This method can readily be applied to other nanoemitters with ≈100 ns lifetime.
Collapse
Affiliation(s)
- Sandra Claveau
- Université Paris-Saclay, ENS Paris-Saclay, CNRS, CentraleSupélec, LuMIn, 91190 Gif-sur-Yvette, France. and Université Paris-Saclay, Institut Gustave Roussy, CNRS, Metabolic and Systemic Aspects of Oncogenesis (METSY), 94805 Villejuif, France
| | - Marek Kindermann
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 166 10 Prague, Czech Republic and Department of Chemical Engineering, University of Chemistry and Technology, 166 28 Prague, Czech Republic
| | | | - Zamira V Díaz-Riascos
- Drug Delivery & Targeting, Functional Validation & Preclinical Research (FVPR), CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain and Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 08035 Barcelona, Spain
| | - Xavier Délen
- Université Paris-Saclay, Institut d'Optique Graduate School, CNRS, Laboratoire Charles Fabry, 91127 Palaiseau, France
| | - Patrick Georges
- Université Paris-Saclay, Institut d'Optique Graduate School, CNRS, Laboratoire Charles Fabry, 91127 Palaiseau, France
| | - Roser López-Alemany
- Sarcoma Research Group, Oncobell Program, CIBERONC, Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Barcelona, Spain
| | - Òscar Martínez Tirado
- Sarcoma Research Group, Oncobell Program, CIBERONC, Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Barcelona, Spain
| | - Jean-Rémi Bertrand
- Université Paris-Saclay, Institut Gustave Roussy, CNRS, Metabolic and Systemic Aspects of Oncogenesis (METSY), 94805 Villejuif, France
| | - Ibane Abasolo
- Drug Delivery & Targeting, Functional Validation & Preclinical Research (FVPR), CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain and Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 08035 Barcelona, Spain
| | - Petr Cigler
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 166 10 Prague, Czech Republic
| | - François Treussart
- Université Paris-Saclay, ENS Paris-Saclay, CNRS, CentraleSupélec, LuMIn, 91190 Gif-sur-Yvette, France.
| |
Collapse
|
37
|
A phase I study of a PARP1-targeted topical fluorophore for the detection of oral cancer. Eur J Nucl Med Mol Imaging 2021; 48:3618-3630. [PMID: 33954826 DOI: 10.1007/s00259-021-05372-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 04/15/2021] [Indexed: 02/02/2023]
Abstract
BACKGROUND Visual inspection and biopsy is the current standard of care for oral cancer diagnosis, but is subject to misinterpretation and consequently to misdiagnosis. Topically applied PARPi-FL is a molecularly specific, fluorescent contrast-based approach that may fulfill the unmet need for a simple, in vivo, non-invasive, cost-effective, point-of-care method for the early diagnosis of oral cancer. Here, we present results from a phase I safety and feasibility study on fluorescent, topically applied PARPi-FL. Twelve patients with a histologically proven oral squamous cell carcinoma (OSCC) gargled a PARPi-FL solution for 60 s (15 mL, 100 nM, 250 nM, 500 nM, or 1000 nM), followed by gargling a clearing solution for 60 s. Fluorescence measurements of the lesion and surrounding oral mucosa were taken before PARPi-FL application, after PARPi-FL application, and after clearing. Blood pressure, oxygen levels, clinical chemistry, and CBC were obtained before and after tracer administration. RESULTS PARPi-FL was well-tolerated by all patients without any safety concerns. When analyzing the fluorescence signal, all malignant lesions showed a significant differential in contrast after administration of PARPi-FL, with the highest increase occurring at the highest dose level (1000 nM), where all patients had a tumor-to-margin fluorescence signal ratio of >3. A clearing step was essential to increase signal specificity, as it clears unbound PARPi-FL trapped in normal anatomical structures. PARPi-FL tumor cell specificity was confirmed by ex vivo tabletop confocal microscopy. We have demonstrated that the fluorescence signal arose from the nuclei of tumor cells, endorsing our macroscopic findings. CONCLUSIONS A PARPi-FL swish & spit solution is a rapid and non-invasive diagnostic tool that preferentially localizes fluorescent contrast to OSCC. This technique holds promise for the early detection of OSCC based on in vivo optical evaluation and targeted biopsy of suspicious lesions in the oral cavity. TRIAL REGISTRATION Clinicaltrials.gov -NCT03085147, registered on March 21st, 2017.
Collapse
|
38
|
Moradi Kashkooli F, Soltani M, Momeni MM. Computational modeling of drug delivery to solid tumors: A pilot study based on a real image. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102347] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
39
|
Gałczyńska K, Rachuna J, Ciepluch K, Kowalska M, Wąsik S, Kosztołowicz T, Lewandowska KD, Semaniak J, Kurdziel K, Arabski M. Experimental and Theoretical Analysis of Metal Complex Diffusion through Cell Monolayer. ENTROPY 2021; 23:e23030360. [PMID: 33802897 PMCID: PMC8002612 DOI: 10.3390/e23030360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/11/2021] [Accepted: 03/15/2021] [Indexed: 01/12/2023]
Abstract
The study of drugs diffusion through different biological membranes constitutes an essential step in the development of new pharmaceuticals. In this study, the method based on the monolayer cell culture of CHO-K1 cells has been developed in order to emulate the epithelial cells barrier in permeability studies by laser interferometry. Laser interferometry was employed for the experimental analysis of nickel(II) and cobalt(II) complexes with 1-allylimidazole or their chlorides’ diffusion through eukaryotic cell monolayers. The amount (mol) of nickel(II) and cobalt(II) chlorides transported through the monolayer was greater than that of metals complexed with 1-allylimidazole by 4.34-fold and 1.45-fold, respectively, after 60 min. Thus, laser interferometry can be used for the quantitative analysis of the transport of compounds through eukaryotic cell monolayers, and the resulting parameters can be used to formulate a mathematical description of this process.
Collapse
Affiliation(s)
- Katarzyna Gałczyńska
- Institute of Biology, Jan Kochanowski University, Uniwersytecka 7, 25-406 Kielce, Poland; (K.G.); (J.R.); (K.C.); (M.K.)
| | - Jarosław Rachuna
- Institute of Biology, Jan Kochanowski University, Uniwersytecka 7, 25-406 Kielce, Poland; (K.G.); (J.R.); (K.C.); (M.K.)
| | - Karol Ciepluch
- Institute of Biology, Jan Kochanowski University, Uniwersytecka 7, 25-406 Kielce, Poland; (K.G.); (J.R.); (K.C.); (M.K.)
| | - Magdalena Kowalska
- Institute of Biology, Jan Kochanowski University, Uniwersytecka 7, 25-406 Kielce, Poland; (K.G.); (J.R.); (K.C.); (M.K.)
| | - Sławomir Wąsik
- Institute of Physics, Jan Kochanowski University, Uniwersytecka 7, 25-406 Kielce, Poland; (S.W.); (T.K.); (J.S.)
| | - Tadeusz Kosztołowicz
- Institute of Physics, Jan Kochanowski University, Uniwersytecka 7, 25-406 Kielce, Poland; (S.W.); (T.K.); (J.S.)
| | - Katarzyna D. Lewandowska
- Department of Radiological Informatics and Statistics, Medical University of Gdańsk, Tuwima 15, 80-210 Gdańsk, Poland;
| | - Jacek Semaniak
- Institute of Physics, Jan Kochanowski University, Uniwersytecka 7, 25-406 Kielce, Poland; (S.W.); (T.K.); (J.S.)
| | - Krystyna Kurdziel
- Institute of Chemistry, Jan Kochanowski University, Uniwersytecka 7, 25-406 Kielce, Poland;
| | - Michał Arabski
- Institute of Biology, Jan Kochanowski University, Uniwersytecka 7, 25-406 Kielce, Poland; (K.G.); (J.R.); (K.C.); (M.K.)
- Correspondence: ; Tel./Fax: +48-41-349-63-31
| |
Collapse
|
40
|
Guo D, Ji X, Luo J. Rational nanocarrier design towards clinical translation of cancer nanotherapy. Biomed Mater 2021; 16. [DOI: 10.1088/1748-605x/abe35a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 02/04/2021] [Indexed: 02/06/2023]
|
41
|
Zeng J, Zhao W, Yue S. Coherent Raman Scattering Microscopy in Oncology Pharmacokinetic Research. Front Pharmacol 2021; 12:630167. [PMID: 33613294 PMCID: PMC7887381 DOI: 10.3389/fphar.2021.630167] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 01/04/2021] [Indexed: 11/28/2022] Open
Abstract
The high attrition rates of anti-cancer drugs during clinical development remains a bottleneck problem in pharmaceutical industry. This is partially due to the lack of quantitative, selective, and rapid readouts of anti-cancer drug activity in situ with high resolution. Although fluorescence microscopy has been commonly used in oncology pharmacological research, fluorescent labels are often too large in size for small drug molecules, and thus may disturb the function or metabolism of these molecules. Such challenge can be overcome by coherent Raman scattering microscopy, which is capable of chemically selective, highly sensitive, high spatial resolution, and high-speed imaging, without the need of any labeling. Coherent Raman scattering microscopy has tremendously improved the understanding of pharmaceutical materials in the solid state, pharmacokinetics of anti-cancer drugs and nanocarriers in vitro and in vivo. This review focuses on the latest applications of coherent Raman scattering microscopy as a new emerging platform to facilitate oncology pharmacokinetic research.
Collapse
Affiliation(s)
- Junjie Zeng
- Institute of Medical Photonics, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Wenying Zhao
- Institute of Medical Photonics, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Shuhua Yue
- Institute of Medical Photonics, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China.,Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
42
|
Rodell CB, Baldwin P, Fernandez B, Weissleder R, Sridhar S, Dubach JM. Quantification of Cellular Drug Biodistribution Addresses Challenges in Evaluating in vitro and in vivo Encapsulated Drug Delivery. ADVANCED THERAPEUTICS 2020; 4. [PMID: 33997266 DOI: 10.1002/adtp.202000125] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Nanoencapsulated drug delivery to solid tumors is a promising approach to overcome pharmacokinetic limitations of therapeutic drugs. However, encapsulation leads to complex drug biodistribution and delivery making analysis of delivery efficacy challenging. As proxies, nanocarrier accumulation or total tumor drug uptake in the tumor are used to evaluate delivery. Yet, these measurements fail to assess delivery of active, released drug to the target, and thus it commonly remains unknown if drug-target occupancy has been achieved. Here, we develop an approach to evaluate the delivery of encapsulated drug to the target, where residual drug target vacancy is measured using a fluorescent drug analog. In vitro measurements reveal that burst release governs drug delivery independent of nanoparticle uptake, and highlight limitations of evaluating nanoencapsulated drug delivery in these models. In vivo, however, our approach captures successful nanoencapsulated delivery, finding that tumor stromal cells drive nanoparticle accumulation and mediate drug delivery to adjacent cancer cells. These results, and generalizable approach, provide a critical advance to evaluate delivery of encapsulated drug to the drug target - the central objective of nanotherapeutics.
Collapse
Affiliation(s)
- Christopher B Rodell
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA.,School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA
| | - Paige Baldwin
- Department of Bioengineering, Northeastern University, Boston, MA
| | - Bianca Fernandez
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA.,Institute for Innovation in Imaging, Massachusetts General Hospital, Boston, MA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA.,Department of Systems Biology, Harvard Medical School, Boston, MA
| | - Srinivas Sridhar
- Department of Bioengineering, Northeastern University, Boston, MA.,Department of Physics, Northeastern University, Boston, MA
| | - J Matthew Dubach
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA.,Institute for Innovation in Imaging, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
43
|
Takikawa M, Fujisawa M, Yoshino K, Takeoka S. Intracellular Distribution of Lipids and Encapsulated Model Drugs from Cationic Liposomes with Different Uptake Pathways. Int J Nanomedicine 2020; 15:8401-8409. [PMID: 33149583 PMCID: PMC7605631 DOI: 10.2147/ijn.s267638] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/31/2020] [Indexed: 01/06/2023] Open
Abstract
AIM The uptake pathway of liposomes into cells is mainly via endocytosis or membrane fusion; however, the relationship between the uptake pathway and the intracellular pharmacokinetics of the liposome components remains unclear. This study aimed at revealing the relationship by using cationic liposomes having similar physical properties and different uptake pathways. MATERIALS AND METHODS We prepared cationic liposomes composed of amino acid-type lipids, K3C14 and K3C16, which have different uptake pathways by a hydration method, and fluorescently modified them by encapsulating FITC-dextran and surface conjugation with Alexa Fluor® 488 (AF488). Then, we investigated their intracellular distribution in HeLa cells over time. RESULTS The liposomes had similar physical properties and did not cause significant cell mortality after treatment for 180 min. The delivery rate and efficiency of encapsulated FITC-dextran with the fusogenic K3C16 liposomes were 3 and 1.6 times higher, respectively, than with the endocytic K3C14 liposomes. FITC-dextran molecules delivered with K3C16 liposomes were observed throughout the cytosolic space after 10 min, while those delivered with K3C14 liposomes were mainly observed as foci and took 60 min to diffuse into the cytosolic space. K3C14 lipids modified with AF488 were distributed mostly in the cytosolic space. In contrast, fluorescently labeled K3C16 lipids were colocalized with the plasma membrane of 50% of the HeLa cells after 10 min and were gradually internalized intracellularly. CONCLUSION Fusogenic K3C16 liposomes internalized into HeLa cells faster than endocytic K3C14 liposomes, and their components differently distributed in the cells.
Collapse
Affiliation(s)
- Masato Takikawa
- Department of Advanced Science and Engineering, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo169-8555, Japan
| | - Mizuki Fujisawa
- Department of Life Science and Medical Bioscience, Graduate Schoolof Advanced Science and Engineering, Waseda University (TWIns), Tokyo162-8480, Japan
| | - Kazuma Yoshino
- Department of Life Science and Medical Bioscience, Graduate Schoolof Advanced Science and Engineering, Waseda University (TWIns), Tokyo162-8480, Japan
| | - Shinji Takeoka
- Department of Life Science and Medical Bioscience, Graduate Schoolof Advanced Science and Engineering, Waseda University (TWIns), Tokyo162-8480, Japan
- Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, Waseda University, Tokyo169-8555, Japan
| |
Collapse
|
44
|
Visualization of the distribution of nanoparticle-formulated AZD2811 in mouse tumor model using matrix-assisted laser desorption ionization mass spectrometry imaging. Sci Rep 2020; 10:15535. [PMID: 32968211 PMCID: PMC7511311 DOI: 10.1038/s41598-020-72665-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 08/03/2020] [Indexed: 02/07/2023] Open
Abstract
Penetration of nanoparticles into viable tumor regions is essential for an effective response. Mass spectrometry imaging (MSI) is a novel method for evaluating the intratumoral pharmacokinetics (PK) of a drug in terms of spatial distribution. The application of MSI for analysis of nanomedicine PK remains in its infancy. In this study, we evaluated the applicability of MALDI-MSI for nanoparticle-formulated drug visualization in tumors and biopsies, with an aim toward future application in clinical nanomedicine research. We established an analytic method for the free drug (AZD2811) and then applied it to visualize nanoparticle-formulated AZD2811. MSI analysis demonstrated heterogeneous intratumoral drug distribution in three xenograft tumors. The intensity of MSI signals correlated well with total drug concentration in tumors, indicating that drug distribution can be monitored quantitatively. Analysis of tumor biopsies indicated that MSI is applicable for analyzing the distribution of nanoparticle-formulated drugs in tumor biopsies, suggesting clinical applicability.
Collapse
|
45
|
Yano S, Tazawa H, Kagawa S, Fujiwara T, Hoffman RM. FUCCI Real-Time Cell-Cycle Imaging as a Guide for Designing Improved Cancer Therapy: A Review of Innovative Strategies to Target Quiescent Chemo-Resistant Cancer Cells. Cancers (Basel) 2020; 12:cancers12092655. [PMID: 32957652 PMCID: PMC7563319 DOI: 10.3390/cancers12092655] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 09/03/2020] [Accepted: 09/09/2020] [Indexed: 01/06/2023] Open
Abstract
Simple Summary Chemotherapy of solid tumors has made very slow progress over many decades. A major problem has been that solid tumors very often contain non-dividing cells due to lack of oxygen deep in the tumor and these non-dividing cells resist most currently-used chemotherapy which usually only targets dividing cells. The present review demonstrates how a unique imaging system, FUCCI, which color codes cells depending on whether they are in a dividing or non-dividing phase, is being used to design very novel therapy that targets non-dividing cancer cells which can greatly improve the efficacy of cancer chemotherapy. Abstract Progress in chemotherapy of solid cancer has been tragically slow due, in large part, to the chemoresistance of quiescent cancer cells in tumors. The fluorescence ubiquitination cell-cycle indicator (FUCCI) was developed in 2008 by Miyawaki et al., which color-codes the phases of the cell cycle in real-time. FUCCI utilizes genes linked to different color fluorescent reporters that are only expressed in specific phases of the cell cycle and can, thereby, image the phases of the cell cycle in real-time. Intravital real-time FUCCI imaging within tumors has demonstrated that an established tumor comprises a majority of quiescent cancer cells and a minor population of cycling cancer cells located at the tumor surface or in proximity to tumor blood vessels. In contrast to most cycling cancer cells, quiescent cancer cells are resistant to cytotoxic chemotherapy, most of which target cells in S/G2/M phases. The quiescent cancer cells can re-enter the cell cycle after surviving treatment, which suggests the reason why most cytotoxic chemotherapy is often ineffective for solid cancers. Thus, quiescent cancer cells are a major impediment to effective cancer therapy. FUCCI imaging can be used to effectively target quiescent cancer cells within tumors. For example, we review how FUCCI imaging can help to identify cell-cycle-specific therapeutics that comprise decoy of quiescent cancer cells from G1 phase to cycling phases, trapping the cancer cells in S/G2 phase where cancer cells are mostly sensitive to cytotoxic chemotherapy and eradicating the cancer cells with cytotoxic chemotherapy most active against S/G2 phase cells. FUCCI can readily image cell-cycle dynamics at the single cell level in real-time in vitro and in vivo. Therefore, visualizing cell cycle dynamics within tumors with FUCCI can provide a guide for many strategies to improve cell-cycle targeting therapy for solid cancers.
Collapse
Affiliation(s)
- Shuya Yano
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (H.T.); (S.K.); (T.F.)
- Center for Graduate Medical Education, Okayama University Hospital, Okayama 700-8558, Japan
- Correspondence: ; Tel.: +81-86-235-7257; Fax: +81-86-221-8775
| | - Hiroshi Tazawa
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (H.T.); (S.K.); (T.F.)
- Center of Innovative Clinical Medicine, Okayama University Hospital, Okayama 700-8558, Japan
| | - Shunsuke Kagawa
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (H.T.); (S.K.); (T.F.)
- Minimally Invasive Therapy Center, Okayama University Hospital, Okayama 700-8558, Japan
| | - Toshiyoshi Fujiwara
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (H.T.); (S.K.); (T.F.)
| | - Robert M. Hoffman
- AntiCancer, Inc., San Diego, CA 92111, USA;
- Department of Surgery, University of California, San Diego, CA 92093, USA
| |
Collapse
|
46
|
Li R, Ng TS, Garlin MA, Weissleder R, Miller MA. Understanding the in vivo Fate of Advanced Materials by Imaging. ADVANCED FUNCTIONAL MATERIALS 2020; 30:1910369. [PMID: 38545084 PMCID: PMC10972611 DOI: 10.1002/adfm.201910369] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 02/09/2020] [Indexed: 11/13/2024]
Abstract
Engineered materials are ubiquitous in biomedical applications ranging from systemic drug delivery systems to orthopedic implants, and their actions unfold across multiple time- and length-scales. The efficacy and safety of biologics, nanomaterials, and macroscopic implants are all dictated by the same general principles of pharmacology as apply to small molecule drugs, comprising how the body affects materials (pharmacokinetics, PK) and conversely how materials affect the body (pharmacodynamics, PD). Imaging technologies play an increasingly insightful role in monitoring both of these processes, often simultaneously: translational macroscopic imaging modalities such as MRI and PET/CT offer whole-body quantitation of biodistribution and structural or molecular response, while ex vivo approaches and optical imaging via in vivo (intravital) microscopy reveal behaviors at subcellular resolution. In this review, the authors survey developments in imaging the in situ behavior of systemically and locally administered materials, with a particular focus on using microscopy to understand transport, target engagement, and downstream host responses at a single-cell level. The themes of microenvironmental influence, controlled drug release, on-target molecular action, and immune response, especially as mediated by macrophages and other myeloid cells are examined. Finally, the future directions of how new imaging technologies may propel efficient clinical translation of next-generation therapeutics and medical devices are proposed.
Collapse
Affiliation(s)
- Ran Li
- Center for Systems Biology, Massachusetts General Hospital Research Institute
| | - Thomas S.C. Ng
- Center for Systems Biology, Massachusetts General Hospital Research Institute
| | - Michelle A. Garlin
- Center for Systems Biology, Massachusetts General Hospital Research Institute
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital Research Institute
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School
- Department of Systems Biology, Harvard Medical School
| | - Miles A. Miller
- Center for Systems Biology, Massachusetts General Hospital Research Institute
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School
| |
Collapse
|
47
|
Demétrio de Souza França P, Guru N, Roberts S, Kossatz S, Mason C, Abrahão M, Ghossein RA, Patel SG, Reiner T. Fluorescence-guided resection of tumors in mouse models of oral cancer. Sci Rep 2020; 10:11175. [PMID: 32636416 PMCID: PMC7341853 DOI: 10.1038/s41598-020-67958-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 05/18/2020] [Indexed: 12/18/2022] Open
Abstract
Complete removal and negative margins are the goal of any surgical resection of primary oral cavity carcinoma. Current approaches to determine tumor boundaries rely heavily on surgeons' expertise, and final histopathological reports are usually only available days after surgery, precluding contemporaneous re-assessment of positive margins. Intraoperative optical imaging could address this unmet clinical need. Using mouse models of oral cavity carcinoma, we demonstrated that PARPi-FL, a fluorescent PARP inhibitor targeting the enzyme PARP1/2, can delineate oral cancer and accurately identify positive margins, both macroscopically and at cellular resolution. PARPi-FL also allowed identification of compromised margins based on fluorescence hotspots, which were not seen in margin-negative resections and control tongues. PARPi-FL was further able to differentiate tumor from low-grade dysplasia. Intravenous injection of PARPi-FL has significant potential for clinical translation and could aid surgeons in assessing oral cancer margins in vivo.
Collapse
Affiliation(s)
- Paula Demétrio de Souza França
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
- Department of Otorhinolaryngology and Head and Neck Surgery, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Navjot Guru
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Sheryl Roberts
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Susanne Kossatz
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
- Department of Nuclear Medicine, School of Medicine, Technische Universität München, Munich, Germany
| | - Christian Mason
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Marcio Abrahão
- Department of Otorhinolaryngology and Head and Neck Surgery, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Ronald A Ghossein
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Snehal G Patel
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Otorhinolaryngology, Weill Cornell Medical College, New York, NY, USA
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
- Department of Otorhinolaryngology, Weill Cornell Medical College, New York, NY, USA.
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
48
|
Whole-body tracking of single cells via positron emission tomography. Nat Biomed Eng 2020; 4:835-844. [PMID: 32541917 PMCID: PMC7423763 DOI: 10.1038/s41551-020-0570-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 05/15/2020] [Indexed: 01/14/2023]
Abstract
In vivo molecular imaging can measure the average kinetics and movement routes of injected cells through the body. Yet owing to the non-specific accumulation of the contrast agent and its efflux from the cells, most such imaging methods suffer from inaccurate estimations of the distribution of the cells. Here, we show that single human breast cancer cells loaded with mesoporous silica nanoparticles concentrating the 68Ga radioisotope and injected in immunodeficient mice can be tracked in real time from the pattern of annihilation photons detected by positron emission tomography, with respect to anatomical landmarks derived from X-ray computed tomography. We show that the cells travelled at an average velocity of 50 mm/s and arrested in the lungs two-to-three seconds after tail-vein injection in the mice, which is consistent with the blood-flow rate. Single-cell tracking could be used to determine the kinetics of cell trafficking and arrest during the earliest phase of the metastatic cascade, the trafficking of immune cells during cancer immunotherapy, and the distribution of cells after transplantation. One-sentence editorial summary: The travelling kinetics of single cells loaded with mesoporous silica nanoparticles concentrating the 68Ga radioisotope and injected in mice can be tracked in real time from the pattern of coincident gamma-rays detected by positron emission tomography.
Collapse
|
49
|
Gonda K, Negishi H, Takano-Kasuya M, Kitamura N, Furusawa N, Nakano Y, Hamada Y, Tokunaga M, Higuchi H, Tada H, Ishida T. Heterogeneous Drug Efficacy of an Antibody-Drug Conjugate Visualized Using Simultaneous Imaging of Its Delivery and Intracellular Damage in Living Tumor Tissues. Transl Oncol 2020; 13:100764. [PMID: 32403030 PMCID: PMC7218300 DOI: 10.1016/j.tranon.2020.100764] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 03/16/2020] [Indexed: 11/30/2022] Open
Abstract
Anticancer drug efficacy varies because the delivery of drugs within tumors and tumor responses are heterogeneous; however, these features are often more homogenous in vitro. This difference makes it difficult to accurately determine drug efficacy. Therefore, it is important to use living tumor tissues in preclinical trials to observe the heterogeneity in drug distribution and cell characteristics in tumors. In the present study, to accurately evaluate the efficacy of an antibody-drug conjugate (ADC) containing a microtubule inhibitor, we established a cell line that expresses a fusion of end-binding protein 1 and enhanced green fluorescent protein that serves as a microtubule plus-end-tracking protein allowing the visualization of microtubule dynamics. This cell line was xenografted into mice to create a model of living tumor tissue. The tumor cells possessed a greater number of microtubules with plus-ends, a greater number of meandering microtubules, and a slower rate of microtubule polymerization than the in vitro cells. In tumor tissues treated with fluorescent dye-labeled ADCs, heterogeneity was observed in the delivery of the drug to tumor cells, and microtubule dynamics were inhibited in a concentration-dependent manner. Moreover, a difference in drug sensitivity was observed between in vitro cells and tumor cells; compared with in vitro cells, tumor cells were more sensitive to changes in the concentration of the ADC. This study is the first to simultaneously evaluate the delivery and intracellular efficacy of ADCs in living tumor tissue. Accurate evaluation of the efficacy of ADCs is important for the development of effective anticancer drugs.
Collapse
Affiliation(s)
- Kohsuke Gonda
- Department of Medical Physics, Graduate School of Medicine, Tohoku University, Sendai, Miyagi 980-8575, Japan.
| | - Hiroshi Negishi
- Bio Systems Development Group, Bio Advanced Technology Division, Corporate R&D Headquarters, KONICAMINOLTA. INC., Hino, Tokyo, 191-8511, Japan
| | - Mayumi Takano-Kasuya
- Department of Medical Physics, Graduate School of Medicine, Tohoku University, Sendai, Miyagi 980-8575, Japan
| | - Narufumi Kitamura
- Department of Medical Physics, Graduate School of Medicine, Tohoku University, Sendai, Miyagi 980-8575, Japan
| | - Naoko Furusawa
- Bio Systems Development Group, Bio Advanced Technology Division, Corporate R&D Headquarters, KONICAMINOLTA. INC., Hino, Tokyo, 191-8511, Japan
| | - Yasushi Nakano
- Bio Systems Development Group, Bio Advanced Technology Division, Corporate R&D Headquarters, KONICAMINOLTA. INC., Hino, Tokyo, 191-8511, Japan
| | - Yoh Hamada
- Department of Gastroenterological Surgery, Graduate School of Medicine, Tohoku University, Sendai, Miyagi 980-8574, Japan
| | - Masayuki Tokunaga
- Department of Medical Physics, Graduate School of Medicine, Tohoku University, Sendai, Miyagi 980-8575, Japan
| | - Hideo Higuchi
- Department of Physics, Graduate School of Science, University of Tokyo, Tokyo 113-0033, Japan
| | - Hiroshi Tada
- Department of Breast and Endocrine Surgical Oncology, Graduate School of Medicine, Tohoku University, Sendai, Miyagi 980-8574, Japan
| | - Takanori Ishida
- Department of Breast and Endocrine Surgical Oncology, Graduate School of Medicine, Tohoku University, Sendai, Miyagi 980-8574, Japan
| |
Collapse
|
50
|
Jannetti SA, Zeglis BM, Zalutsky MR, Reiner T. Poly(ADP-Ribose)Polymerase (PARP) Inhibitors and Radiation Therapy. Front Pharmacol 2020; 11:170. [PMID: 32194409 PMCID: PMC7062869 DOI: 10.3389/fphar.2020.00170] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 02/07/2020] [Indexed: 12/15/2022] Open
Abstract
Poly(ADP-ribose)polymerase-1 (PARP1) is a DNA repair enzyme highly expressed in the nuclei of mammalian cells, with a structure and function that have attracted interest since its discovery. PARP inhibitors, moreover, can be used to induce synthetic lethality in cells where the homologous recombination (HR) pathway is deficient. Several small molecule PARP inhibitors have been approved by the FDA for multiple cancers bearing this deficiency These PARP inhibitors also act as radiosensitizing agents by delaying single strand break (SSB) repair and causing subsequent double strand break (DSB) generation, a concept that has been leveraged in various preclinical models of combination therapy with PARP inhibitors and ionizing radiation. Researchers have determined the efficacy of various PARP inhibitors at sub-cytotoxic concentrations in radiosensitizing multiple human cancer cell lines to ionizing radiation. Furthermore, several groups have begun evaluating combination therapy strategies in mouse models of cancer, and a fluorescent imaging agent that allows for subcellular imaging in real time has been developed from a PARP inhibitor scaffold. Other PARP inhibitor scaffolds have been radiolabeled to create PET imaging agents, some of which have also entered clinical trials. Most recently, these highly targeted small molecules have been radiolabeled with therapeutic isotopes to create radiotherapeutics and radiotheranostics in cancers whose primary interventions are surgical resection and whole-body radiotherapy. In this review we discuss the utilization of these small molecules in combination therapies and in scaffolds for imaging agents, radiotherapeutics, and radiotheranostics. Development of these radiolabeled PARP inhibitors has presented promising results for new interventions in the fight against some of the most intractable cancers.
Collapse
Affiliation(s)
- Stephen A. Jannetti
- Department of Biochemistry, Hunter College, New York, NY, United States
- Ph.D. Program in Biochemistry, CUNY Graduate Center, New York, NY, United States
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Brian M. Zeglis
- Department of Biochemistry, Hunter College, New York, NY, United States
- Ph.D. Program in Biochemistry, CUNY Graduate Center, New York, NY, United States
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Ph.D. Program in Chemistry, CUNY Graduate Center, New York, NY, United States
| | - Michael R. Zalutsky
- Department of Radiology, Duke University Medical Center, Durham, NC, United States
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Radiology, Weill Cornell Medical College, New York, NY, United States
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|