1
|
Gopal AA, Fernandez B, Delano J, Weissleder R, Dubach JM. PARP trapping is governed by the PARP inhibitor dissociation rate constant. Cell Chem Biol 2024; 31:1373-1382.e10. [PMID: 38262416 PMCID: PMC11259578 DOI: 10.1016/j.chembiol.2023.12.019] [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: 10/17/2022] [Revised: 09/13/2023] [Accepted: 12/22/2023] [Indexed: 01/25/2024]
Abstract
Poly(ADP-ribose) polymerase (PARP) inhibitors (PARPi) are a class of cancer drugs that enzymatically inhibit PARP activity at sites of DNA damage. Yet, PARPi function mainly by trapping PARP1 onto DNA with a wide range of potency among the clinically relevant inhibitors. How PARPi trap and why some are better trappers remain unknown. Here, we show trapping occurs primarily through a kinetic phenomenon at sites of DNA damage that correlates with PARPi koff. Our results suggest PARP trapping is not the physical stalling of PARP1 on DNA, rather the high probability of PARP re-binding damaged DNA in the absence of other DNA-binding protein recruitment. These results clarify how PARPi trap, shed new light on how PARPi function, and describe how PARPi properties correlate to trapping potency.
Collapse
Affiliation(s)
- Angelica A Gopal
- Institute for Innovation in Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Bianca Fernandez
- Institute for Innovation in Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Justin Delano
- Institute for Innovation in Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114; Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114; Department of Systems Biology, Harvard Medical School, Boston, MA 02115
| | - J Matthew Dubach
- Institute for Innovation in Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114; Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114.
| |
Collapse
|
2
|
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
|
3
|
Wang Q, Fu M, Gao L, Yuan X, Wang J. A Drug Repositioning Approach Reveals Ergotamine May Be a Potential Drug for the Treatment of Alzheimer's Disease. J Alzheimers Dis 2024; 101:1355-1366. [PMID: 39269834 DOI: 10.3233/jad-240235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
Background Alzheimer's disease (AD) is a neurodegenerative disorder that is the most common form of dementia in the elderly. The drugs currently used to treat AD only have limited effects and are not able to cure the disease. Drug repositioning has increasingly become a promising approach to find potential drugs for diseases like AD. Objective To screen potential drug candidates for AD based on the relationship between risk genes of AD and drugs. Methods We collected the risk genes of AD and retrieved the information of known drugs from DrugBank. Then, the AD-related genes and the targets of each drug were mapped to the human protein-protein interaction network (PPIN) to represent AD and the drugs on the network. The network distances between each drug and AD were calculated to screen the drugs proximal to AD-related genes on PPIN, and the screened drug candidates were further analyzed by molecular docking and molecular dynamics simulations. Results We compiled a list of 714 genes associated with AD. From 5,833 drugs used for human diseases, we identified 1,044 drugs that could be potentially used to treat AD. Then, amyloid-β (Aβ) protein, the key molecule involved in the pathogenesis of AD was selected as the target to further screen drugs that may inhibit Aβ aggregation by molecular docking. We found that ergotamine and RAF-265 could bind stably with Aβ. In further analysis by molecular dynamics simulations, both drugs exhibited reasonable stability. Conclusions Our work indicated that ergotamine and RAF-265 may be potential candidates for treating AD.
Collapse
Affiliation(s)
- Qiuchen Wang
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Mengjie Fu
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Lihui Gao
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Xin Yuan
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Ju Wang
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| |
Collapse
|
4
|
Pan S, Ding A, Li Y, Sun Y, Zhan Y, Ye Z, Song N, Peng B, Li L, Huang W, Shao H. Small-molecule probes from bench to bedside: advancing molecular analysis of drug-target interactions toward precision medicine. Chem Soc Rev 2023; 52:5706-5743. [PMID: 37525607 DOI: 10.1039/d3cs00056g] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
Over the past decade, remarkable advances have been witnessed in the development of small-molecule probes. These molecular tools have been widely applied for interrogating proteins, pathways and drug-target interactions in preclinical research. While novel structures and designs are commonly explored in probe development, the clinical translation of small-molecule probes remains limited, primarily due to safety and regulatory considerations. Recent synergistic developments - interfacing novel chemical probes with complementary analytical technologies - have introduced and expedited diverse biomedical opportunities to molecularly characterize targeted drug interactions directly in the human body or through accessible clinical specimens (e.g., blood and ascites fluid). These integrated developments thus offer unprecedented opportunities for drug development, disease diagnostics and treatment monitoring. In this review, we discuss recent advances in the structure and design of small-molecule probes with novel functionalities and the integrated development with imaging, proteomics and other emerging technologies. We further highlight recent applications of integrated small-molecule technologies for the molecular analysis of drug-target interactions, including translational applications and emerging opportunities for whole-body imaging, tissue-based measurement and blood-based analysis.
Collapse
Affiliation(s)
- Sijun Pan
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| | - Aixiang Ding
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| | - Yisi Li
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| | - Yaxin Sun
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| | - Yueqin Zhan
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| | - Zhenkun Ye
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| | - Ning Song
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| | - Bo Peng
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China
| | - Lin Li
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| | - Wei Huang
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China
| | - Huilin Shao
- Institute for Health Innovation & Technology, National University of Singapore, Singapore 117599, Singapore.
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore 117583, Singapore
| |
Collapse
|
5
|
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: 7] [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
|
6
|
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
|
7
|
Oh K, Jiang K. Synthesis of fluorescently labeled pyrazole derivative induceing a triple response in Arabidopsis seedlings. JOURNAL OF PESTICIDE SCIENCE 2022; 47:203-207. [PMID: 36514690 PMCID: PMC9716048 DOI: 10.1584/jpestics.d22-006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 09/14/2022] [Indexed: 06/17/2023]
Abstract
A fluorescent labeled pyrazole derivative with a dansyl moiety (EH-DF) was synthesized. Design of EH-DF was carried out by using a dansyl moiety to substitute the naphthalene moiety of the parent compound (EH-1). At a concentration of 30 µM, EH-DF displayed biological activity on inducing a triple response in Arabidopsis seedlings. Compared with the non-chemical treated control, the hypocotyl length of EH-DF-treated Arabidopsis seedlings was reduced from approximately 9.2±0.7 mm to 2.4±0.2 mm. The length of the roots was reduced from 1.7±0.1 mm to 1.0±0.1 mm, and the curvature of the hook of Arabidopsis seedlings increased from 60±16 degrees to 245±35 degrees. The maxim excitation wavelength and emission wavelength of EH-DF were 350 and 535 nm, respectively. Data obtained via fluorescent microscope analysis indicated that intensive fluorescent signals of EH-DF were observed in the shoot of Arabidopsis seedlings.
Collapse
Affiliation(s)
- Keimei Oh
- Department of Biotechnology, Faculty of Bioresource Sciences, Akita Prefectural University
| | - Kai Jiang
- Institute of Plant and Food Science, Department of Biology, School of Life Sciences, Southern University of Science and Technology (SUSTech)
| |
Collapse
|
8
|
Al-Amin RA, Johansson L, Abdurakhmanov E, Landegren N, Löf L, Arngården L, Blokzijl A, Svensson R, Hammond M, Lönn P, Haybaeck J, Kamali-Moghaddam M, Jensen A, Danielson U, Artursson P, Lundbäck T, Landegren U. Monitoring drug-target interactions through target engagement-mediated amplification on arrays and in situ. Nucleic Acids Res 2022; 50:e129. [PMID: 36189884 PMCID: PMC9825164 DOI: 10.1093/nar/gkac842] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 08/24/2022] [Accepted: 09/20/2022] [Indexed: 01/29/2023] Open
Abstract
Drugs are designed to bind their target proteins in physiologically relevant tissues and organs to modulate biological functions and elicit desirable clinical outcomes. Information about target engagement at cellular and subcellular resolution is therefore critical for guiding compound optimization in drug discovery, and for probing resistance mechanisms to targeted therapies in clinical samples. We describe a target engagement-mediated amplification (TEMA) technology, where oligonucleotide-conjugated drugs are used to visualize and measure target engagement in situ, amplified via rolling-circle replication of circularized oligonucleotide probes. We illustrate the TEMA technique using dasatinib and gefitinib, two kinase inhibitors with distinct selectivity profiles. In vitro binding by the dasatinib probe to arrays of displayed proteins accurately reproduced known selectivity profiles, while their differential binding to fixed adherent cells agreed with expectations from expression profiles of the cells. We also introduce a proximity ligation variant of TEMA to selectively investigate binding to specific target proteins of interest. This form of the assay serves to improve resolution of binding to on- and off-target proteins. In conclusion, TEMA has the potential to aid in drug development and clinical routine by conferring valuable insights in drug-target interactions at spatial resolution in protein arrays, cells and in tissues.
Collapse
Affiliation(s)
- Rasel A Al-Amin
- To whom correspondence should be addressed. Tel: +46 70 0535324;
| | - Lars Johansson
- Department of Medical Biochemistry and Biophysics, Chemical Biology Consortium Sweden (CBCS), Science for Life Laboratory, Karolinska Institutet, Solna, Sweden
| | - Eldar Abdurakhmanov
- Department of Chemistry-BMC, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Nils Landegren
- Center for Molecular Medicine, Department of Medicine (Solna), Science for Life Laboratory, Karolinska Institutet, Solna, Sweden
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Liza Löf
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Linda Arngården
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Andries Blokzijl
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Richard Svensson
- Department of Pharmacy, Uppsala University Drug Optimization and Pharmaceutical Profiling (UDOPP), Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Maria Hammond
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Peter Lönn
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Johannes Haybaeck
- Institute of Pathology, Neuropathology and Molecular Pathology, Medical University of Innsbruck, Innsbruck, Austria
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Masood Kamali-Moghaddam
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Annika Jenmalm Jensen
- Department of Medical Biochemistry and Biophysics, Chemical Biology Consortium Sweden (CBCS), Science for Life Laboratory, Karolinska Institutet, Solna, Sweden
| | - U Helena Danielson
- Department of Chemistry-BMC, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Per Artursson
- Department of Pharmacy, Uppsala University Drug Optimization and Pharmaceutical Profiling (UDOPP), Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Thomas Lundbäck
- Department of Medical Biochemistry and Biophysics, Chemical Biology Consortium Sweden (CBCS), Science for Life Laboratory, Karolinska Institutet, Solna, Sweden
| | - Ulf Landegren
- Correspondence may also be addressed to Ulf Landegren. Tel: +46 18 4714910; Fax: +46 18 4714808;
| |
Collapse
|
9
|
Spanning BODIPY fluorescence with self-assembled micellar clusters. Colloids Surf B Biointerfaces 2022; 216:112532. [PMID: 35525227 DOI: 10.1016/j.colsurfb.2022.112532] [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: 12/22/2021] [Revised: 04/10/2022] [Accepted: 04/28/2022] [Indexed: 11/23/2022]
Abstract
BODIPY dyes possess favorable optical properties for a variety of applications including in vivo and in vitro diagnostics. However, their utilization might be limited by their water insolubility and incompatibility with chemical modifications, resulting in low aggregation stability. Here, we outline the route for addressing this issue. We have demonstrated two approaches, based on dye entrapment in micellar coordination clusters (MCCs); this provides a general solution for water solubility as well as aggregation stability of the seven BODIPY derivatives. These derivatives have various bulky aromatic substituents in the 2,3,5,6- and meso-positions and can rotate relative to a dipyrrin core, which also provides molecular rotor properties. The molecular structural features and the presence of aromatic groups allows BODIPY dyes to be used as "supporting molecules", thus promoting micelle-micelle interaction and micellar network stabilization. In the second approach, self-micellization, following BODIPY use, leads to MCC formation without the use of any mediators, including chelators and/or metal ions. In both approaches, BODIPY exhibits an excellent optical response, at a concentration beyond its solubilization limit in aqueous media and without undesired crystallization. The suggested approaches represent systems used to encapsulate BODIPY in a capsule-based surfactant environment, enabling one to track the aggregation of BODIPY; these approaches represent an alternative system to study and apply BODIPY's molecular rotor properties. The stabilized compounds, i.e., the BODIPY-loaded MCCs, provide a unique feature of permeability to hydrophilic ligand-switching proteins such as BSA; they exhibit a bright "turn-on" fluorescence signal within the clusters via macromolecular complexation, thus expanding the possibilities of water-soluble BODIPY-loaded MCCs utilization for functional indicators.
Collapse
|
10
|
Zheng J, Xiao X, Qiu WR. DTI-BERT: Identifying Drug-Target Interactions in Cellular Networking Based on BERT and Deep Learning Method. Front Genet 2022; 13:859188. [PMID: 35754843 PMCID: PMC9213727 DOI: 10.3389/fgene.2022.859188] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/25/2022] [Indexed: 11/20/2022] Open
Abstract
Drug–target interactions (DTIs) are regarded as an essential part of genomic drug discovery, and computational prediction of DTIs can accelerate to find the lead drug for the target, which can make up for the lack of time-consuming and expensive wet-lab techniques. Currently, many computational methods predict DTIs based on sequential composition or physicochemical properties of drug and target, but further efforts are needed to improve them. In this article, we proposed a new sequence-based method for accurately identifying DTIs. For target protein, we explore using pre-trained Bidirectional Encoder Representations from Transformers (BERT) to extract sequence features, which can provide unique and valuable pattern information. For drug molecules, Discrete Wavelet Transform (DWT) is employed to generate information from drug molecular fingerprints. Then we concatenate the feature vectors of the DTIs, and input them into a feature extraction module consisting of a batch-norm layer, rectified linear activation layer and linear layer, called BRL block and a Convolutional Neural Networks module to extract DTIs features further. Subsequently, a BRL block is used as the prediction engine. After optimizing the model based on contrastive loss and cross-entropy loss, it gave prediction accuracies of the target families of G Protein-coupled receptors, ion channels, enzymes, and nuclear receptors up to 90.1, 94.7, 94.9, and 89%, which indicated that the proposed method can outperform the existing predictors. To make it as convenient as possible for researchers, the web server for the new predictor is freely accessible at: https://bioinfo.jcu.edu.cn/dtibert or http://121.36.221.79/dtibert/. The proposed method may also be a potential option for other DITs.
Collapse
Affiliation(s)
- Jie Zheng
- Computer Department, Jing-De-Zhen Ceramic Institute, Jing-De-Zhen, China
| | - Xuan Xiao
- Computer Department, Jing-De-Zhen Ceramic Institute, Jing-De-Zhen, China
| | - Wang-Ren Qiu
- Computer Department, Jing-De-Zhen Ceramic Institute, Jing-De-Zhen, China
| |
Collapse
|
11
|
Hann E, Malagu K, Stott A, Vater H. The importance of plasma protein and tissue binding in a drug discovery program to successfully deliver a preclinical candidate. PROGRESS IN MEDICINAL CHEMISTRY 2022; 61:163-214. [PMID: 35753715 DOI: 10.1016/bs.pmch.2022.04.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Plasma protein binding and tissue binding are arguably two of the most critical parameters that are measured as part of a drug discovery program since, according to the free drug hypothesis, it is the free drug that is responsible for both efficacy and toxicity. This chapter aims to deconstruct the role of plasma protein and tissue binding in drug discovery programs, and to consider the conclusion made by Pfizer and Genentech/Depomed a decade ago that optimising plasma protein binding as an independent parameter does not significantly influence efficacy. This chapter will also examine how binding metrics are applied in drug discovery programs and explore circumstances where optimising plasma protein or tissue binding can be an effective strategy to deliver a candidate molecule for preclinical development with an early indication of sufficient therapeutic index.
Collapse
Affiliation(s)
- Elizabeth Hann
- Charles River Laboratories, Robinson Building, Chesterford Research Park, Saffron Walden, United Kingdom.
| | - Karine Malagu
- Charles River Laboratories, Robinson Building, Chesterford Research Park, Saffron Walden, United Kingdom
| | - Andrew Stott
- Charles River Laboratories, Robinson Building, Chesterford Research Park, Saffron Walden, United Kingdom
| | - Huw Vater
- Charles River Laboratories, Robinson Building, Chesterford Research Park, Saffron Walden, United Kingdom
| |
Collapse
|
12
|
Payne NC, Kalyakina AS, Singh K, Tye MA, Mazitschek R. Bright and stable luminescent probes for target engagement profiling in live cells. Nat Chem Biol 2021; 17:1168-1177. [PMID: 34675420 PMCID: PMC8555866 DOI: 10.1038/s41589-021-00877-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 08/11/2021] [Indexed: 01/18/2023]
Abstract
The pace of progress in biomedical research directly depends on techniques that enable the quantitative interrogation of interactions between proteins and other biopolymers, or with their small-molecule ligands. Time-resolved Förster resonance energy transfer (TR-FRET) assay platforms offer high sensitivity and specificity. However, the paucity of accessible and biocompatible luminescent lanthanide complexes, which are essential reagents for TR-FRET-based approaches, and their poor cellular permeability have limited broader adaptation of TR-FRET beyond homogeneous and extracellular assay applications. Here, we report the development of CoraFluors, a new class of macrotricyclic terbium complexes, which are synthetically readily accessible, stable in biological media and exhibit photophysical and physicochemical properties that are desirable for biological studies. We validate the performance of CoraFluors in cell-free systems, identify cell-permeable analogs and demonstrate their utility in the quantitative domain-selective characterization of Keap1 ligands, as well as in isoform-selective target engagement profiling of HDAC1 inhibitors in live cells.
Collapse
Affiliation(s)
- N Connor Payne
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Chemistry & Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Alena S Kalyakina
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, USA
- Karlsruhe Institute of Technology, Institute of Organic Chemistry, Karlsruhe, Germany
| | - Kritika Singh
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Mark A Tye
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Graduate School of Arts and Sciences, Cambridge, MA, USA
| | - Ralph Mazitschek
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, USA.
- Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
13
|
Huang Q, Chen B, Shen J, Liu L, Li J, Shi J, Li Q, Zuo X, Wang L, Fan C, Li J. Encoding Fluorescence Anisotropic Barcodes with DNA Fameworks. J Am Chem Soc 2021; 143:10735-10742. [PMID: 34242004 DOI: 10.1021/jacs.1c04942] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Fluorescence anisotropy (FA) holds great potential for multiplexed analysis and imaging of biomolecules since it can effectively discriminate fluorophores with overlapping emission spectra. Nevertheless, its susceptibility to environmental variation hampers its widespread applications in biology and biotechnology. In this study, we design FA DNA frameworks (FAFs) by scaffolding fluorophores in a fluorescent protein-like microenvironment. We find that the FA stability of the fluorophores is remarkably improved due to the sequestration effects of FAFs. The FA level of the fluorophores can be finely tuned when placed at different locations on an FAF, analogous to spectral shifts of protein-bound fluorophores. The high programmability of FAFs further enables the design of a spectrum of encoded FA barcodes for multiplexed sensing of nucleic acids and multiplexed labeling of live cells. This FAF system thus establishes a new paradigm for designing multiplexing FA probes for cellular imaging and other biological applications.
Collapse
Affiliation(s)
- Qiuling Huang
- Division of Physical Biology, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bin Chen
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acids Chemistry and Nanomedicine State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Jianlei Shen
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Lei Liu
- Division of Physical Biology, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
| | - Jiajun Li
- Division of Physical Biology, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
| | - Jiye Shi
- Division of Physical Biology, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
| | - Qian Li
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiaolei Zuo
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acids Chemistry and Nanomedicine State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Lihua Wang
- The Interdisciplinary Research Center, Shanghai Synchrotron Radiation Facility, Zhangjiang Laboratory, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.,Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai 200127, China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jiang Li
- Division of Physical Biology, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China.,The Interdisciplinary Research Center, Shanghai Synchrotron Radiation Facility, Zhangjiang Laboratory, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| |
Collapse
|
14
|
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: 1] [Impact Index Per Article: 0.3] [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
|
15
|
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: 1.0] [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
|
16
|
Gopal AA, Kazarine A, Dubach JM, Wiseman PW. Recent advances in nonlinear microscopy: Deep insights and polarized revelations. Int J Biochem Cell Biol 2020; 130:105896. [PMID: 33253831 DOI: 10.1016/j.biocel.2020.105896] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/12/2020] [Accepted: 11/19/2020] [Indexed: 11/16/2022]
Abstract
Nonlinear microscopy is a technique that utilizes nonlinear interactions between light and matter to image fluorescence and scattering phenomena in biological tissues. Very high peak intensities from focused short pulsed lasers are required for nonlinear excitation due to the extremely low probability of the simultaneous arrival of multiple photons of lower energy to excite fluorophores or interact with selective structures for harmonic generation. Combined with reduced scattering from the utilization of longer wavelengths, the inherent spatial confinement associated with achieving simultaneous arrival of photons within the focal volume enables deep imaging with low out-of-focus background for nonlinear imaging. This review provides an introduction to the different contrast mechanisms available with nonlinear imaging and instrumentation commonly used in nonlinear microscopy. Furthermore, we discuss some recent advances in nonlinear microscopy to extend the imaging penetration depth, conduct histopathological investigations on fresh tissues and examine the molecular order and orientation of molecules using polarization nonlinear microscopy.
Collapse
Affiliation(s)
- A A Gopal
- Center for Systems Biology and Institute for Innovation in Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA; Department of Chemistry, McGill University, Montreal, Quebec, Canada
| | - A Kazarine
- Department of Chemistry, McGill University, Montreal, Quebec, Canada
| | - J M Dubach
- Center for Systems Biology and Institute for Innovation in Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - P W Wiseman
- Department of Chemistry, McGill University, Montreal, Quebec, Canada; Department of Physics, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
17
|
Meng Q, Ma X, Xie B, Deng X, Huang J, Zhou HB, Dong C. Establishment of evaluation criteria for the development of high quality ERα-targeted fluorescent probes. Analyst 2020; 145:5989-5995. [PMID: 32856648 DOI: 10.1039/d0an01172j] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
ERα-targeted fluorescent probes are important tools for ERα study. In order to develop high quality ERα-targeted probes, a sound and complete evaluation system is essential but has not been established yet. Herein, we set up a series of evaluation criteria for ERα-targeted fluorescent probes including ERα binding affinity, fluorescence quantum yield, cytotoxicity, ERα tracking capacity, ERα selectivity and ERα labeling ability. To verify the practicability of the evaluation criteria, we designed and synthesized two ERα-targeted fluorescent probes and fully characterized their properties based on the proposed evaluation criteria. It showed that the probes exhibited better performance. Moreover, we applied the probes in MCF-7 cells to study the ERα motion characteristics for the first time. We hope that our evaluation criteria could be helpful for the establishment of a complete evaluation system for ERα-targeted fluorescent probes.
Collapse
Affiliation(s)
- Qiuyu Meng
- Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Hubei Provincial Key Laboratory of Developmentally Originated Disease, State Key Laboratory of Virology, Wuhan University School of Pharmaceutical Sciences, Wuhan 430071, China.
| | | | | | | | | | | | | |
Collapse
|
18
|
Li Z, Xiao Y, Peng J, Locke D, Holmes D, Li L, Hamilton S, Cook E, Myer L, Vanderwall D, Cloutier N, Siddiqui AM, Whitehead P, Bishop R, Zhao L, Cvijic ME. Quantifying drug tissue biodistribution by integrating high content screening with deep-learning analysis. Sci Rep 2020; 10:14408. [PMID: 32873881 PMCID: PMC7463244 DOI: 10.1038/s41598-020-71347-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 08/09/2020] [Indexed: 01/23/2023] Open
Abstract
Quantitatively determining in vivo achievable drug concentrations in targeted organs of animal models and subsequent target engagement confirmation is a challenge to drug discovery and translation due to lack of bioassay technologies that can discriminate drug binding with different mechanisms. We have developed a multiplexed and high-throughput method to quantify drug distribution in tissues by integrating high content screening (HCS) with U-Net based deep learning (DL) image analysis models. This technology combination allowed direct visualization and quantification of biologics drug binding in targeted tissues with cellular resolution, thus enabling biologists to objectively determine drug binding kinetics.
Collapse
Affiliation(s)
- Zhuyin Li
- Lead Discovery and Optimization, Bristol-Myers Squibb, 3551 Lawrenceville Road, Princeton, NJ, 08540, USA.
| | - Youping Xiao
- Information Technology for R&D, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Jia Peng
- Lead Discovery and Optimization, Bristol-Myers Squibb, 3551 Lawrenceville Road, Princeton, NJ, 08540, USA
| | - Darren Locke
- Translational Medicine, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Derek Holmes
- Immunoscience Biology Discovery, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Lei Li
- Lead Discovery and Optimization, Bristol-Myers Squibb, 3551 Lawrenceville Road, Princeton, NJ, 08540, USA
| | - Shannon Hamilton
- Lead Discovery and Optimization, Bristol-Myers Squibb, 3551 Lawrenceville Road, Princeton, NJ, 08540, USA
| | - Erica Cook
- Lead Discovery and Optimization, Bristol-Myers Squibb, 3551 Lawrenceville Road, Princeton, NJ, 08540, USA
| | - Larnie Myer
- Lead Discovery and Optimization, Bristol-Myers Squibb, 3551 Lawrenceville Road, Princeton, NJ, 08540, USA
| | - Dana Vanderwall
- Information Technology for R&D, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Normand Cloutier
- Information Technology for R&D, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Akbar M Siddiqui
- Information Technology for R&D, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Paul Whitehead
- Information Technology for R&D, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Richard Bishop
- Information Technology for R&D, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Lei Zhao
- Cardiovascular Translational Research, Bristol-Myers Squibb, Hopewell, NJ, USA
| | - Mary Ellen Cvijic
- Lead Discovery and Optimization, Bristol-Myers Squibb, 3551 Lawrenceville Road, Princeton, NJ, 08540, USA
| |
Collapse
|
19
|
Feruglio PF, Vinegoni C, Weissleder R. Extended dynamic range imaging for noise mitigation in fluorescence anisotropy imaging. JOURNAL OF BIOMEDICAL OPTICS 2020; 25:JBO-200159R. [PMID: 32820624 PMCID: PMC7439791 DOI: 10.1117/1.jbo.25.8.086003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/27/2020] [Indexed: 06/11/2023]
Abstract
SIGNIFICANCE Fluorescence polarization (FP) and fluorescence anisotropy (FA) microscopy are powerful imaging techniques that allow to translate the common FP assay capabilities into the in vitro and in vivo cellular domain. As a result, they have found potential for mapping drug-protein or protein-protein interactions. Unfortunately, these imaging modalities are ratiometric in nature and as such they suffer from excessive noise even under regular imaging conditions, preventing accurate image-feature analysis of fluorescent molecules behaviors. AIM We present a high dynamic range (HDR)-based FA imaging modality for improving image quality in FA microscopy. APPROACH The method exploits ad hoc acquisition schemes to extend the dynamic range of individual FP channels, allowing to obtain FA images with increased signal-to-noise ratio. RESULTS A direct comparison between FA images obtained with our method and the standard, clearly indicates how an HDR-based FA imaging approach allows to obtain high-quality images, with the ability to correctly resolve image features at different values of FA and over a substantially higher range of fluorescence intensities. CONCLUSION The method presented is shown to outperform standard FA imaging microscopy narrowing the spread of the propagated error and yielding higher quality images. The method can be effectively and routinely used on any commercial imaging system and could be also translated to other microscopy ratiometric imaging modalities.
Collapse
Affiliation(s)
- Paolo Fumene Feruglio
- Massachusetts General Hospital, Harvard Medical School, Center for Systems Biology, Boston, Massachusetts, United States
- University of Verona, Department of Neuroscience, Biomedicine, and Movement Sciences, Verona, Italy
- ITS Meccatronico Veneto, Vicenza, Italy
| | - Claudio Vinegoni
- Massachusetts General Hospital, Harvard Medical School, Center for Systems Biology, Boston, Massachusetts, United States
| | - Ralph Weissleder
- Massachusetts General Hospital, Harvard Medical School, Center for Systems Biology, Boston, Massachusetts, United States
- Harvard Medical School, Department of Systems Biology, Boston, Massachusetts, United States
| |
Collapse
|
20
|
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: 3.5] [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
|
21
|
Deng L, Huang X, Ren J. In Situ Study of the Drug–Target Protein Interaction in Single Living Cells by Combining Fluorescence Correlation Spectroscopy with Affinity Probes. Anal Chem 2020; 92:7020-7027. [DOI: 10.1021/acs.analchem.0c00263] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Liyun Deng
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, Shanghai 200240, People’s Republic of China
| | - Xiangyi Huang
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, Shanghai 200240, People’s Republic of China
| | - Jicun Ren
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, Shanghai 200240, People’s Republic of China
| |
Collapse
|
22
|
Stefaniak J, Huber KVM. Importance of Quantifying Drug-Target Engagement in Cells. ACS Med Chem Lett 2020; 11:403-406. [PMID: 32292539 DOI: 10.1021/acsmedchemlett.9b00570] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Measuring and quantifying the binding of a drug to a protein target inside living cells and thereby correlating biochemical or biophysical activity with target engagement in cells or tissue represents a key step in target validation and drug development. A prototypic target engagement assay should allow for unbiased determination of small molecule-protein interactions in order to confirm cellular mechanism-of-action (MoA) while avoiding major artificial perturbations of cellular homeostasis and integrity. Recently, several new additions to the chemical biology toolbox have expanded our ability to study drug action in intact cells and enabled surveying of intracellular residence time and binding kinetics, which are particularly important for potent receptor ligands and therapeutic moieties with limited therapeutic index.
Collapse
Affiliation(s)
- Jakub Stefaniak
- Structural Genomics Consortium, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, U.K
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, U.K
- Chemistry Research Laboratory, University of Oxford, Oxford OX1 3TA, U.K
| | - Kilian V. M. Huber
- Structural Genomics Consortium, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, U.K
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, U.K
| |
Collapse
|
23
|
Robers MB, Friedman-Ohana R, Huber KVM, Kilpatrick L, Vasta JD, Berger BT, Chaudhry C, Hill S, Müller S, Knapp S, Wood KV. Quantifying Target Occupancy of Small Molecules Within Living Cells. Annu Rev Biochem 2020; 89:557-581. [PMID: 32208767 DOI: 10.1146/annurev-biochem-011420-092302] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The binding affinity and kinetics of target engagement are fundamental to establishing structure-activity relationships (SARs) for prospective therapeutic agents. Enhancing these binding parameters for operative targets, while minimizing binding to off-target sites, can translate to improved drug efficacy and a widened therapeutic window. Compound activity is typically assessed through modulation of an observed phenotype in cultured cells. Quantifying the corresponding binding properties under common cellular conditions can provide more meaningful interpretation of the cellular SAR analysis. Consequently, methods for assessing drug binding in living cells have advanced and are now integral to medicinal chemistry workflows. In this review, we survey key technological advancements that support quantitative assessments of target occupancy in cultured cells, emphasizing generalizable methodologies able to deliver analytical precision that heretofore required reductionist biochemical approaches.
Collapse
Affiliation(s)
- M B Robers
- Promega Corporation, Madison, Wisconsin 53711, USA; , ,
| | | | - K V M Huber
- Target Discovery Institute and Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, United Kingdom; .,Structural Genomics Consortium, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - L Kilpatrick
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, United Kingdom; , .,Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands NG7 2UH, United Kingdom
| | - J D Vasta
- Promega Corporation, Madison, Wisconsin 53711, USA; , ,
| | - B-T Berger
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, 60438 Frankfurt, Germany; ,
| | - C Chaudhry
- Lead Discovery and Optimization, Bristol-Myers Squibb, Princeton, New Jersey 08648, USA;
| | - S Hill
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, United Kingdom; , .,Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands NG7 2UH, United Kingdom
| | - S Müller
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, 60438 Frankfurt, Germany; , .,Structural Genomics Consortium, Buchmann Institute for Life Sciences, Goethe University Frankfurt, 60438 Frankfurt, Germany;
| | - S Knapp
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, 60438 Frankfurt, Germany; , .,Structural Genomics Consortium, Buchmann Institute for Life Sciences, Goethe University Frankfurt, 60438 Frankfurt, Germany; .,German Cancer Network (DKTK), Frankfurt/Mainz, 60438 Frankfurt, Germany.,Frankfurt Cancer Institute (FCI), Goethe University, 60596 Frankfurt am Main, Germany
| | - K V Wood
- Promega Corporation, Madison, Wisconsin 53711, USA; , , .,Current affiliation: Light Bio, Inc., Mount Horeb, Wisconsin 53572, USA;
| |
Collapse
|
24
|
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: 13.3] [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
|
25
|
Perrin L, Bayarmagnai B, Gligorijevic B. Frontiers in Intravital Multiphoton Microscopy of Cancer. Cancer Rep (Hoboken) 2020; 3:e1192. [PMID: 32368722 PMCID: PMC7197974 DOI: 10.1002/cnr2.1192] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 03/12/2019] [Accepted: 03/21/2019] [Indexed: 12/23/2022] Open
Abstract
Background Cancer is a highly complex disease which involves the co-operation of tumor cells with multiple types of host cells and the extracellular matrix. Cancer studies which rely solely on static measurements of individual cell types are insufficient to dissect this complexity. In the last two decades, intravital microscopy has established itself as a powerful technique that can significantly improve our understanding of cancer by revealing the dynamic interactions governing cancer initiation, progression and treatment effects, in living animals. This review focuses on intravital multiphoton microscopy (IV-MPM) applications in mouse models of cancer. Recent Findings IV-MPM studies have already enabled a deeper understanding of the complex events occurring in cancer, at the molecular, cellular and tissue levels. Multiple cells types, present in different tissues, influence cancer cell behavior via activation of distinct signaling pathways. As a result, the boundaries in the field of IV-MPM are continuously being pushed to provide an integrated comprehension of cancer. We propose that optics, informatics and cancer (cell) biology are co-evolving as a new field. We have identified four emerging themes in this new field. First, new microscopy systems and image processing algorithms are enabling the simultaneous identification of multiple interactions between the tumor cells and the components of the tumor microenvironment. Second, techniques from molecular biology are being exploited to visualize subcellular structures and protein activities within individual cells of interest, and relate those to phenotypic decisions, opening the door for "in vivo cell biology". Third, combining IV-MPM with additional imaging modalities, or omics studies, holds promise for linking the cell phenotype to its genotype, metabolic state or tissue location. Finally, the clinical use of IV-MPM for analyzing efficacy of anti-cancer treatments is steadily growing, suggesting a future role of IV-MPM for personalized medicine. Conclusion IV-MPM has revolutionized visualization of tumor-microenvironment interactions in real time. Moving forward, incorporation of novel optics, automated image processing, and omics technologies, in the study of cancer biology, will not only advance our understanding of the underlying complexities but will also leverage the unique aspects of IV-MPM for clinical use.
Collapse
Affiliation(s)
- Louisiane Perrin
- Department of BioengineeringTemple UniversityPhiladelphiaPennsylvania
| | | | - Bojana Gligorijevic
- Department of BioengineeringTemple UniversityPhiladelphiaPennsylvania
- Fox Chase Cancer CenterCancer Biology ProgramPhiladelphiaPennsylvania
| |
Collapse
|
26
|
Petazzi RA, Aji AK, Chiantia S. Fluorescence microscopy methods for the study of protein oligomerization. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 169:1-41. [DOI: 10.1016/bs.pmbts.2019.12.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
27
|
Suzuki S, Ikuta M, Yoshii T, Nakamura A, Kuwata K, Tsukiji S. Golgi recruitment assay for visualizing small-molecule ligand–target engagement in cells. Chem Commun (Camb) 2020; 56:7961-7964. [DOI: 10.1039/d0cc02020f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A Golgi recruitment (G-REC) assay is developed as a new method for visualizing small-molecule ligand–target engagement in living cells.
Collapse
Affiliation(s)
- Sachio Suzuki
- Department of Nanopharmaceutical Sciences
- Nagoya Institute of Technology
- Nagoya 466-8555
- Japan
| | - Masahiro Ikuta
- Department of Life Science and Applied Chemistry
- Nagoya Institute of Technology
- Nagoya 466-8555
- Japan
| | - Tatsuyuki Yoshii
- Department of Life Science and Applied Chemistry
- Nagoya Institute of Technology
- Nagoya 466-8555
- Japan
- PRESTO, Japan Science and Technology Agency (JST)
| | - Akinobu Nakamura
- Department of Life Science and Applied Chemistry
- Nagoya Institute of Technology
- Nagoya 466-8555
- Japan
| | - Keiko Kuwata
- Institute of Transformative Bio-Molecules (ITbM)
- Nagoya University
- Nagoya 464-8602
- Japan
| | - Shinya Tsukiji
- Department of Nanopharmaceutical Sciences
- Nagoya Institute of Technology
- Nagoya 466-8555
- Japan
- Department of Life Science and Applied Chemistry
| |
Collapse
|
28
|
Ng TS, Garlin MA, Weissleder R, Miller MA. Improving nanotherapy delivery and action through image-guided systems pharmacology. Theranostics 2020; 10:968-997. [PMID: 31938046 PMCID: PMC6956809 DOI: 10.7150/thno.37215] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 08/04/2019] [Indexed: 12/12/2022] Open
Abstract
Despite recent advances in the translation of therapeutic nanoparticles (TNPs) into the clinic, the field continues to face challenges in predictably and selectively delivering nanomaterials for the treatment of solid cancers. The concept of enhanced permeability and retention (EPR) has been coined as a convenient but simplistic descriptor of high TNP accumulation in some tumors. However, in practice EPR represents a number of physiological variables rather than a single one (including dysfunctional vasculature, compromised lymphatics and recruited host cells, among other aspects of the tumor microenvironment) — each of which can be highly heterogenous within a given tumor, patient and across patients. Therefore, a clear need exists to dissect the specific biophysical factors underlying the EPR effect, to formulate better TNP designs, and to identify patients with high-EPR tumors who are likely to respond to TNP. The overall pharmacology of TNP is governed by an interconnected set of spatially defined and dynamic processes that benefit from a systems-level quantitative approach, and insights into the physiology have profited from the marriage between in vivo imaging and quantitative systems pharmacology (QSP) methodologies. In this article, we review recent developments pertinent to image-guided systems pharmacology of nanomedicines in oncology. We first discuss recent developments of quantitative imaging technologies that enable analysis of nanomaterial pharmacology at multiple spatiotemporal scales, and then examine reports that have adopted these imaging technologies to guide QSP approaches. In particular, we focus on studies that have integrated multi-scale imaging with computational modeling to derive insights about the EPR effect, as well as studies that have used modeling to guide the manipulation of the EPR effect and other aspects of the tumor microenvironment for improving TNP action. We anticipate that the synergistic combination of imaging with systems-level computational methods for effective clinical translation of TNPs will only grow in relevance as technologies increase in resolution, multiplexing capability, and in the ability to examine heterogeneous behaviors at the single-cell level.
Collapse
|
29
|
Vinegoni C, Feruglio PF, Gryczynski I, Mazitschek R, Weissleder R. Fluorescence anisotropy imaging in drug discovery. Adv Drug Deliv Rev 2019; 151-152:262-288. [PMID: 29410158 PMCID: PMC6072632 DOI: 10.1016/j.addr.2018.01.019] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 01/29/2018] [Accepted: 01/30/2018] [Indexed: 12/15/2022]
Abstract
Non-invasive measurement of drug-target engagement can provide critical insights in the molecular pharmacology of small molecule drugs. Fluorescence polarization/fluorescence anisotropy measurements are commonly employed in protein/cell screening assays. However, the expansion of such measurements to the in vivo setting has proven difficult until recently. With the advent of high-resolution fluorescence anisotropy microscopy it is now possible to perform kinetic measurements of intracellular drug distribution and target engagement in commonly used mouse models. In this review we discuss the background, current advances and future perspectives in intravital fluorescence anisotropy measurements to derive pharmacokinetic and pharmacodynamic measurements in single cells and whole organs.
Collapse
Affiliation(s)
- Claudio Vinegoni
- Center for System Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Paolo Fumene Feruglio
- Center for System Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Department of Neurological, Biomedical and Movement Sciences, University of Verona, Verona, Italy
| | - Ignacy Gryczynski
- University of North Texas Health Science Center, Institute for Molecular Medicine, Fort Worth, TX, United States
| | - Ralph Mazitschek
- Center for System Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ralph Weissleder
- Center for System Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
30
|
Richardson PL, Marin VL, Koeniger SL, Baranczak A, Wilsbacher JL, Kovar PJ, Bacon-Trusk PE, Cheng M, Hopkins TA, Haman ST, Vasudevan A. Controlling cellular distribution of drugs with permeability modifying moieties. MEDCHEMCOMM 2019; 10:974-984. [PMID: 31303996 PMCID: PMC6595965 DOI: 10.1039/c8md00412a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 04/16/2019] [Indexed: 12/31/2022]
Abstract
Phenotypic screening provides compounds with very limited target cellular localization data. In order to select the most appropriate target identification methods, determining if a compound acts at the cell-surface or intracellularly can be very valuable. In addition, controlling cell-permeability of targeted therapeutics such as antibody-drug conjugates (ADCs) and targeted nanoparticle formulations can reduce toxicity from extracellular release of drug in undesired tissues or direct activity in bystander cells. By incorporating highly polar, anionic moieties via short polyethylene glycol linkers into compounds with known intracellular, and cell-surface targets, we have been able to correlate the cellular activity of compounds with their subcellular site of action. For compounds with nuclear (Brd, PARP) or cytosolic (dasatinib, NAMPT) targets, addition of the permeability modifying group (small sulfonic acid, polycarboxylic acid, or a polysulfonated fluorescent dye) results in near complete loss of biological activity in cell-based assays. For cell-surface targets (H3, 5HT1A, β2AR) significant activity was maintained for all conjugates, but the results were more nuanced in that the modifiers impacted binding/activity of the resulting conjugates. Taken together, these results demonstrate that small anionic compounds can be used to control cell-permeability independent of on-target activity and should find utility in guiding target deconvolution studies and controlling drug distribution of targeted therapeutics.
Collapse
Affiliation(s)
- Paul L Richardson
- Drug Discovery Science and Technologies , 1 North Waukegan Rd , North Chicago , IL 60064 , USA .
| | - Violeta L Marin
- Drug Discovery Science and Technologies , 1 North Waukegan Rd , North Chicago , IL 60064 , USA .
| | - Stormy L Koeniger
- Drug Discovery Science and Technologies , 1 North Waukegan Rd , North Chicago , IL 60064 , USA .
| | - Aleksandra Baranczak
- Drug Discovery Science and Technologies , 1 North Waukegan Rd , North Chicago , IL 60064 , USA .
| | | | | | | | - Min Cheng
- Discovery Oncology , AbbVie Inc. , USA
| | | | | | - Anil Vasudevan
- Drug Discovery Science and Technologies , 1 North Waukegan Rd , North Chicago , IL 60064 , USA .
| |
Collapse
|
31
|
Ma X, Özliseli E, Zhang Y, Pan G, Wang D, Zhang H. Fabrication of redox-responsive doxorubicin and paclitaxel prodrug nanoparticles with microfluidics for selective cancer therapy. Biomater Sci 2019; 7:634-644. [PMID: 30534690 DOI: 10.1039/c8bm01333k] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Cancer is an exceptionally confounding disease that demands the development of powerful drug/drugs, without inducing heavy adverse side effects. Thus, different approaches have been applied to improve the targeted delivery of cancer drugs: for example by using nanocarriers. However, nanocarriers are foreign materials, which need further validation for their biocompatibility and biodegradability. In this study, we have chemically conjugated the hydrophilic anticancer drug doxorubicin (DOX) with the hydrophobic drug paclitaxel (PTX) through a redox-sensitive disulfide bond, abbreviated to DOX-S-S-PTX. Subsequently, due to its amphiphilic characterization, the prodrug can self-assemble into nanoparticles under microfluidic nanoprecipitation. These novel prodrug nanoparticles have a super-high drug loading degree of 89%, which is impossible to achieve by any nanocarrier systems, and can be tailored to 180 nm to deliver themselves to the target, and release DOX and PTX under redox conditions, which are often found in cancer cells. By evaluating cell viability in MDA-MB-231, MDA-MB-231/ADR and MEF cell lines, we observed that the prodrug nanoparticles effectively killed the cancer cells, and selectively conquered the MDA-MB-231/ADR. Meanwhile, MEF cells were spared due to their lack of a redox condition. The cell interaction results show that the reduced intermediate of the prodrug can also bind to parent drug biological targets. The hemolysis results show that the nanoparticles are biocompatible in blood. Computer modelling suggested that the prodrug is unlikely to bind to biological targets that parent drugs still strongly interact with. Finally, we confirm that the prodrug nanoparticles have no therapeutic effect in blood or healthy cells, but can selectively eliminate the cancer cells that meet the redox conditions to cleave the disulfide bond and release the drugs DOX and PTX.
Collapse
Affiliation(s)
- Xiaodong Ma
- Department of Radiology affiliated Hospital of Jiangsu University Jiangsu University, 212001 Zhenjiang, P.R. China
| | | | | | | | | | | |
Collapse
|
32
|
Vasaturo M, Cotugno R, Fiengo L, Vinegoni C, Dal Piaz F, De Tommasi N. The anti-tumor diterpene oridonin is a direct inhibitor of Nucleolin in cancer cells. Sci Rep 2018; 8:16735. [PMID: 30425290 PMCID: PMC6233161 DOI: 10.1038/s41598-018-35088-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 10/25/2018] [Indexed: 11/15/2022] Open
Abstract
The bioactive plant diterpene oridonin displays important pharmacological activities and is widely used in traditional Chinese medicine; however, its molecular mechanism of action is still incompletely described. In vitro and in vivo data have demonstrated anti-tumor activity of oridonin and its ability to interfere with several cell pathways; however, presently only the molecular chaperone HSP70 has been identified as a direct potential target of this compound. Here, using a combination of different proteomic approaches, innovative Cellular Thermal Shift Assay (CETSA) experiments, and classical biochemical methods, we demonstrate that oridonin interacts with Nucleolin, effectively modulating the activity of this multifunctional protein. The ability of oridonin to target Nucleolin and/or HSP70 could account for the bioactivity profile of this plant diterpene. Recently, Nucleolin has attracted attention as a druggable target, as its diverse functions are implicated in pathological processes such as cancer, inflammation, and viral infection. However, up to now, no small molecule as Nucleolin binders has been reported, thus our finding represents the first evidence of Nucleolin modulation by a small inhibitor.
Collapse
Affiliation(s)
- Michele Vasaturo
- Università degli Studi di Salerno, Department of Pharmacy, Via Giovanni Paolo II, 84084, Fisciano, (SA), Italy
- Università degli Studi di Salerno, Ph. D. School of Pharmacy, Via Giovanni Paolo II, 84084, Fisciano, (SA), Italy
| | - Roberta Cotugno
- Università degli Studi di Salerno, Department of Pharmacy, Via Giovanni Paolo II, 84084, Fisciano, (SA), Italy
| | - Lorenzo Fiengo
- Università degli Studi di Salerno, Department of Pharmacy, Via Giovanni Paolo II, 84084, Fisciano, (SA), Italy
- Università degli Studi di Salerno, Ph. D. School of Pharmacy, Via Giovanni Paolo II, 84084, Fisciano, (SA), Italy
| | - Claudio Vinegoni
- Harvard Medical School, MGH Center for Systems Biology, 185 Cambridge Steet, 02144, Boston, MA, USA
| | - Fabrizio Dal Piaz
- Università degli Studi di Salerno, Department of Medicine and Surgery, Via S. Allende, 84081, Baronissi, (SA), Italy.
| | - Nunziatina De Tommasi
- Università degli Studi di Salerno, Department of Pharmacy, Via Giovanni Paolo II, 84084, Fisciano, (SA), Italy
| |
Collapse
|
33
|
Xiao Y, An FF, Chen J, Xiong S, Zhang XH. The impact of light irradiation timing on the efficacy of nanoformula-based photo/chemo combination therapy. J Mater Chem B 2018; 6:3692-3702. [PMID: 32254832 DOI: 10.1039/c8tb00427g] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Photo/chemo combination therapy has been demonstrated to be a generally more powerful strategy for treating cancers than a single treatment modality. However, it is unknown whether the timing of light irradiation has any impact on therapeutic efficacy. We designed a carrier-free and self-monitoring nanodrug to monitor the entire dual-drug release profile and determined the impact of photodynamic therapy (PDT) at different time points. The designed nanodrug consists of the chemotherapeutic doxorubicin (DOX) and the photosensitizer pheophorbide A (PhA). The drugs form a fluorescence resonance energy transfer (FRET) pair (DOX transferring energy to PhA) when present at a precise ratio in the combination nanodrug. Due to the FRET effect, the DOX-PhA nanoparticles (NPs) show PhA fluorescence in a normal pH environment (such as cytoplasm). However, the FRET effect is lost when the NPs are disassembled in an acidic environment (such as lysosomes), and the DOX fluorescence is recovered. By real-time fluorescence variation monitoring, we determined the key time points when the drugs reached various subcellular locations, which helped us to determine the PDT-triggering time points and investigate the impact on the therapeutic effect in the combination therapy. Furthermore, the PDT was triggered at these established time points both in vitro and in vivo, which revealed that the best PDT-triggering time point in the combination therapy was achieved after nuclear entry of DOX. The study suggests that the optimization of combination therapy, not only photo/chemo but also chemo/chemo combination therapy, may require not only a controlled drug ratio but also a controlled drug release profile and target arrival time.
Collapse
Affiliation(s)
- Yafang Xiao
- Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano & Soft Materials (FUNSOM), Joint International Research Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, 199 Ren'ai Road, Suzhou, 215123, Jiangsu, P. R. China.
| | | | | | | | | |
Collapse
|
34
|
Comess KM, McLoughlin SM, Oyer JA, Richardson PL, Stöckmann H, Vasudevan A, Warder SE. Emerging Approaches for the Identification of Protein Targets of Small Molecules - A Practitioners’ Perspective. J Med Chem 2018; 61:8504-8535. [DOI: 10.1021/acs.jmedchem.7b01921] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Kenneth M. Comess
- AbbVie Inc., 1 Waukegan Road, North Chicago, Illinois 60064-1802, United States
| | - Shaun M. McLoughlin
- AbbVie Inc., 1 Waukegan Road, North Chicago, Illinois 60064-1802, United States
| | - Jon A. Oyer
- AbbVie Inc., 1 Waukegan Road, North Chicago, Illinois 60064-1802, United States
| | - Paul L. Richardson
- AbbVie Inc., 1 Waukegan Road, North Chicago, Illinois 60064-1802, United States
| | - Henning Stöckmann
- AbbVie Inc., 1 Waukegan Road, North Chicago, Illinois 60064-1802, United States
| | - Anil Vasudevan
- AbbVie Inc., 1 Waukegan Road, North Chicago, Illinois 60064-1802, United States
| | - Scott E. Warder
- AbbVie Inc., 1 Waukegan Road, North Chicago, Illinois 60064-1802, United States
| |
Collapse
|
35
|
Targeting Ligand Specificity Linked to Tumor Tissue Topological Heterogeneity via Single-Cell Micro-Pharmacological Modeling. Sci Rep 2018; 8:3638. [PMID: 29483578 PMCID: PMC5827036 DOI: 10.1038/s41598-018-21883-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 02/13/2018] [Indexed: 02/07/2023] Open
Abstract
Targeted therapy has held promise to be a successful anticancer treatment due to its specificity towards tumor cells that express the target receptors. However, not all targeting drugs used in the clinic are equally effective in tumor eradication. To examine which biochemical and biophysical properties of targeted agents are pivotal for their effective distribution inside the tumor and their efficient cellular uptake, we combine mathematical micro-pharmacological modeling with in vivo imaging of targeted human xenograft tumors in SCID mice. The mathematical model calibrated to experimental data was used to explore properties of the targeting ligand (diffusion and affinity) and ligand release schemes (rates and concentrations) with a goal to identify the properties of cells and ligands that enable high receptor saturation. By accounting for heterogeneities typical of in vivo tumors, our model was able to identify cell- and tissue-level barriers to efficient drug uptake. This work provides a base for utilizing experimentally measurable properties of a ligand-targeted agent and patient-specific attributes of the tumor tissue to support the development of novel targeted imaging agents and for improvement in their delivery to individual tumor cells.
Collapse
|
36
|
White MD, Zhao ZW, Plachta N. In Vivo Imaging of Single Mammalian Cells in Development and Disease. Trends Mol Med 2018; 24:278-293. [PMID: 29439932 DOI: 10.1016/j.molmed.2018.01.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 01/05/2018] [Accepted: 01/14/2018] [Indexed: 12/14/2022]
Abstract
Live imaging has transformed biomedical sciences by enabling visualization and analysis of dynamic cellular processes as they occur in their native contexts. Here, we review key recent efforts applying in vivo optical imaging with single-cell resolution to mammalian systems ranging from embryos to adult tissues and organs. We highlight insights into active processes regulating cell fate and morphogenesis during embryonic development, how neuronal circuitry and non-neuronal cell types contribute to neurological functions, and how novel imaging-based approaches enable the dissection of neurological disorders and cancer with high spatio-temporal resolution. The convergence of technical advancements in accessing, visualizing, and manipulating individual cells provides an unprecedented lens to probe mammalian cellular dynamics in vivo in both physiological and pathological states.
Collapse
Affiliation(s)
- Melanie D White
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore; These authors contributed equally to this work
| | - Ziqing W Zhao
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore; These authors contributed equally to this work
| | - Nicolas Plachta
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore.
| |
Collapse
|
37
|
Karolak A, Rejniak KA. Micropharmacology: An In Silico Approach for Assessing Drug Efficacy Within a Tumor Tissue. Bull Math Biol 2018; 81:3623-3641. [PMID: 29423880 DOI: 10.1007/s11538-018-0402-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 01/30/2018] [Indexed: 12/25/2022]
Abstract
Systemic chemotherapy is one of the main anticancer treatments used for most kinds of clinically diagnosed tumors. However, the efficacy of these drugs can be hampered by the physical attributes of the tumor tissue, such as tortuous vasculature, dense and fibrous extracellular matrix, irregular cellular architecture, tumor metabolic gradients, and non-uniform expression of the cell membrane receptors. This can impede the transport of therapeutic agents to tumor cells in sufficient quantities. In addition, tumor microenvironments undergo dynamic spatio-temporal changes during tumor progression and treatment, which can also obstruct drug efficacy. To examine ways to improve drug delivery on a cell-to-tissue scale (single-cell pharmacology), we developed the microscale pharmacokinetics/pharmacodynamics (microPKPD) modeling framework. Our model is modular and can be adjusted to include only the mathematical equations that are crucial for a biological problem under consideration. This modularity makes the model applicable to a broad range of pharmacological cases. As an illustration, we present two specific applications of the microPKPD methodology that help to identify optimal drug properties. The hypoxia-activated drugs example uses continuous drug concentrations, diffusive-advective transport through the tumor interstitium, and passive transmembrane drug uptake. The targeted therapy example represents drug molecules as discrete particles that move by diffusion and actively bind to cell receptors. The proposed modeling approach takes into account the explicit tumor tissue morphology, its metabolic landscape and/or specific receptor distribution. All these tumor attributes can be assessed from patients' diagnostic biopsies; thus, the proposed methodology can be developed into a tool suitable for personalized medicine, such as neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- Aleksandra Karolak
- Integrated Mathematical Oncology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Katarzyna A Rejniak
- Integrated Mathematical Oncology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA. .,Department of Oncologic Sciences, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
38
|
Abstract
The principles of engineering and physics have been applied to oncology for nearly 50 years. Engineers and physical scientists have made contributions to all aspects of cancer biology, from quantitative understanding of tumour growth and progression to improved detection and treatment of cancer. Many early efforts focused on experimental and computational modelling of drug distribution, cell cycle kinetics and tumour growth dynamics. In the past decade, we have witnessed exponential growth at the interface of engineering, physics and oncology that has been fuelled by advances in fields including materials science, microfabrication, nanomedicine, microfluidics, imaging, and catalysed by new programmes at the National Institutes of Health (NIH), including the National Institute of Biomedical Imaging and Bioengineering (NIBIB), Physical Sciences in Oncology, and the National Cancer Institute (NCI) Alliance for Nanotechnology. Here, we review the advances made at the interface of engineering and physical sciences and oncology in four important areas: the physical microenvironment of the tumour and technological advances in drug delivery; cellular and molecular imaging; and microfluidics and microfabrication. We discussthe research advances, opportunities and challenges for integrating engineering and physical sciences with oncology to develop new methods to study, detect and treat cancer, and we also describe the future outlook for these emerging areas.
Collapse
Affiliation(s)
- Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, 240 Skirkanich Hall, 210 S. 33rd Street, Philadelphia, Pennsylvania 19104, USA
- Department of Chemical Engineering, David H. Koch Institute for Integrated Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, USA
| | - Rakesh K Jain
- Edwin L. Steele Laboratories of Tumour Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, 100 Blossom Street, Cox 7, Boston, Massachusetts 02114, USA
| | - Robert Langer
- Department of Chemical Engineering, David H. Koch Institute for Integrated Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, USA
| |
Collapse
|
39
|
Prediction of intracellular exposure bridges the gap between target- and cell-based drug discovery. Proc Natl Acad Sci U S A 2017; 114:E6231-E6239. [PMID: 28701380 DOI: 10.1073/pnas.1701848114] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Inadequate target exposure is a major cause of high attrition in drug discovery. Here, we show that a label-free method for quantifying the intracellular bioavailability (Fic) of drug molecules predicts drug access to intracellular targets and hence, pharmacological effect. We determined Fic in multiple cellular assays and cell types representing different targets from a number of therapeutic areas, including cancer, inflammation, and dementia. Both cytosolic targets and targets localized in subcellular compartments were investigated. Fic gives insights on membrane-permeable compounds in terms of cellular potency and intracellular target engagement, compared with biochemical potency measurements alone. Knowledge of the amount of drug that is locally available to bind intracellular targets provides a powerful tool for compound selection in early drug discovery.
Collapse
|
40
|
Yang L, Hu Z, Luo J, Tang C, Zhang S, Ning W, Dong C, Huang J, Liu X, Zhou HB. Dual functional small molecule fluorescent probes for image-guided estrogen receptor-specific targeting coupled potent antiproliferative potency for breast cancer therapy. Bioorg Med Chem 2017; 25:3531-3539. [DOI: 10.1016/j.bmc.2017.05.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 04/21/2017] [Accepted: 05/02/2017] [Indexed: 12/21/2022]
|
41
|
Vinegoni C, Fumene Feruglio P, Brand C, Lee S, Nibbs AE, Stapleton S, Shah S, Gryczynski I, Reiner T, Mazitschek R, Weissleder R. Measurement of drug-target engagement in live cells by two-photon fluorescence anisotropy imaging. Nat Protoc 2017; 12:1472-1497. [PMID: 28686582 PMCID: PMC5928516 DOI: 10.1038/nprot.2017.043] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The ability to directly image and quantify drug-target engagement and drug distribution with subcellular resolution in live cells and whole organisms is a prerequisite to establishing accurate models of the kinetics and dynamics of drug action. Such methods would thus have far-reaching applications in drug development and molecular pharmacology. We recently presented one such technique based on fluorescence anisotropy, a spectroscopic method based on polarization light analysis and capable of measuring the binding interaction between molecules. Our technique allows the direct characterization of target engagement of fluorescently labeled drugs, using fluorophores with a fluorescence lifetime larger than the rotational correlation of the bound complex. Here we describe an optimized protocol for simultaneous dual-channel two-photon fluorescence anisotropy microscopy acquisition to perform drug-target measurements. We also provide the necessary software to implement stream processing to visualize images and to calculate quantitative parameters. The assembly and characterization part of the protocol can be implemented in 1 d. Sample preparation, characterization and imaging of drug binding can be completed in 2 d. Although currently adapted to an Olympus FV1000MPE microscope, the protocol can be extended to other commercial or custom-built microscopes.
Collapse
Affiliation(s)
- Claudio Vinegoni
- Center for System Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Paolo Fumene Feruglio
- Center for System Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Neurosciences, Biomedicine, and Movement Sciences, University of Verona, Verona, Italy
| | - Christian Brand
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Sungon Lee
- Center for System Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- School of Electrical Engineering, Hanyang University, Ansan, Republic of Korea
| | - Antoinette E Nibbs
- Center for System Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Shawn Stapleton
- Center for System Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Sunil Shah
- Institute for Molecular Medicine, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Ignacy Gryczynski
- Institute for Molecular Medicine, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Ralph Mazitschek
- Center for System Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Ralph Weissleder
- Center for System Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
42
|
Abstract
Imaging is widely used in anticancer drug development, typically for whole-body tracking of labelled drugs to different organs or to assess drug efficacy through volumetric measurements. However, increasing attention has been drawn to pharmacology at the single-cell level. Diverse cell types, including cancer-associated immune cells, physicochemical features of the tumour microenvironment and heterogeneous cell behaviour all affect drug delivery, response and resistance. This Review summarizes developments in the imaging of in vivo anticancer drug action, with a focus on microscopy approaches at the single-cell level and translational lessons for the clinic.
Collapse
Affiliation(s)
- Miles A. Miller
- Center for Systems Biology, 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
| |
Collapse
|
43
|
Carney B, Kossatz S, Reiner T. Molecular Imaging of PARP. J Nucl Med 2017; 58:1025-1030. [PMID: 28473593 DOI: 10.2967/jnumed.117.189936] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 04/26/2017] [Indexed: 11/16/2022] Open
Abstract
The poly(adenosine diphosphate-ribose)polymerase (PARP) family of enzymes is an important factor in the cellular DNA damage response and has gained much attention for its role in many diseases, particularly cancer. Targeted molecular imaging of PARP using fluorescent or radiolabeled tags has followed on the success of therapeutic inhibitors and gained momentum over the past few years. This review covers PARP imaging from the very first imaging agents up to the current state of the technology, with a focus on the clinical applications made possible by these agents.
Collapse
Affiliation(s)
- Brandon Carney
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Chemistry, Hunter College, and PhD Program in Chemistry, Graduate Center of City University of New York, New York, New York; and
| | - Susanne Kossatz
- Department of Radiology, 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
| |
Collapse
|
44
|
Miller MA, Weissleder R. Imaging the pharmacology of nanomaterials by intravital microscopy: Toward understanding their biological behavior. Adv Drug Deliv Rev 2017; 113:61-86. [PMID: 27266447 PMCID: PMC5136524 DOI: 10.1016/j.addr.2016.05.023] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 05/25/2016] [Indexed: 12/15/2022]
Abstract
Therapeutic nanoparticles (NPs) can deliver cytotoxic chemotherapeutics and other drugs more safely and efficiently to patients; furthermore, selective delivery to target tissues can theoretically be accomplished actively through coating NPs with molecular ligands, and passively through exploiting physiological "enhanced permeability and retention" features. However, clinical trial results have been mixed in showing improved efficacy with drug nanoencapsulation, largely due to heterogeneous NP accumulation at target sites across patients. Thus, a clear need exists to better understand why many NP strategies fail in vivo and not result in significantly improved tumor uptake or therapeutic response. Multicolor in vivo confocal fluorescence imaging (intravital microscopy; IVM) enables integrated pharmacokinetic and pharmacodynamic (PK/PD) measurement at the single-cell level, and has helped answer key questions regarding the biological mechanisms of in vivo NP behavior. This review summarizes progress to date and also describes useful technical strategies for successful IVM experimentation.
Collapse
Affiliation(s)
- Miles A Miller
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, Boston, MA 02114, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, Boston, MA 02114, USA; Department of Systems Biology, Harvard Medical School, 200 Longwood Ave, Boston, MA 02115, USA.
| |
Collapse
|
45
|
Mikula H, Stapleton S, Kohler RH, Vinegoni C, Weissleder R. Design and Development of Fluorescent Vemurafenib Analogs for In Vivo Imaging. Am J Cancer Res 2017; 7:1257-1265. [PMID: 28435463 PMCID: PMC5399591 DOI: 10.7150/thno.18238] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 12/17/2016] [Indexed: 12/30/2022] Open
Abstract
Herein we describe fluorescent derivatives of vemurafenib to probe therapeutic BRAF inhibition in live cells and in vivo. The compounds were evaluated and compared by determining target binding, inhibition of mutant BRAF melanoma cell lines and live cell imaging. We show that vemurafenib-BODIPY is a superior imaging drug to visualize the targets of vemurafenib in live cells and in vivo in non-resistant and resistant melanoma tumors.
Collapse
|
46
|
Abstract
The intrinsic limits of conventional cancer therapies prompted the development and application of various nanotechnologies for more effective and safer cancer treatment, herein referred to as cancer nanomedicine. Considerable technological success has been achieved in this field, but the main obstacles to nanomedicine becoming a new paradigm in cancer therapy stem from the complexities and heterogeneity of tumour biology, an incomplete understanding of nano-bio interactions and the challenges regarding chemistry, manufacturing and controls required for clinical translation and commercialization. This Review highlights the progress, challenges and opportunities in cancer nanomedicine and discusses novel engineering approaches that capitalize on our growing understanding of tumour biology and nano-bio interactions to develop more effective nanotherapeutics for cancer patients.
Collapse
Affiliation(s)
- Jinjun Shi
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Philip W Kantoff
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | | | - Omid C Farokhzad
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
47
|
Abstract
The intrinsic limits of conventional cancer therapies prompted the development and application of various nanotechnologies for more effective and safer cancer treatment, herein referred to as cancer nanomedicine. Considerable technological success has been achieved in this field, but the main obstacles to nanomedicine becoming a new paradigm in cancer therapy stem from the complexities and heterogeneity of tumour biology, an incomplete understanding of nano-bio interactions and the challenges regarding chemistry, manufacturing and controls required for clinical translation and commercialization. This Review highlights the progress, challenges and opportunities in cancer nanomedicine and discusses novel engineering approaches that capitalize on our growing understanding of tumour biology and nano-bio interactions to develop more effective nanotherapeutics for cancer patients.
Collapse
Affiliation(s)
- Jinjun Shi
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Philip W Kantoff
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | | | - Omid C Farokhzad
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
48
|
Dubach JM, Kim E, Yang K, Cuccarese M, Giedt RJ, Meimetis LG, Vinegoni C, Weissleder R. Quantitating drug-target engagement in single cells in vitro and in vivo. Nat Chem Biol 2016; 13:168-173. [PMID: 27918558 DOI: 10.1038/nchembio.2248] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 09/22/2016] [Indexed: 12/24/2022]
Abstract
Quantitation of drug target engagement in single cells has proven to be difficult, often leaving unanswered questions in the drug development process. We found that intracellular target engagement of unlabeled new therapeutics can be quantitated using polarized microscopy combined with competitive binding of matched fluorescent companion imaging probes. We quantitated the dynamics of target engagement of covalent BTK inhibitors, as well as reversible PARP inhibitors, in populations of single cells using a single companion imaging probe for each target. We then determined average in vivo tumor concentrations and found marked population heterogeneity following systemic delivery, revealing single cells with low target occupancy at high average target engagement in vivo.
Collapse
Affiliation(s)
- J Matthew Dubach
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Eunha Kim
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Katherine Yang
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael Cuccarese
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Randy J Giedt
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Labros G Meimetis
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Claudio Vinegoni
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Systems Biology, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
49
|
Vinegoni C, Dubach JM, Feruglio PF, Weissleder R. Two-photon Fluorescence Anisotropy Microscopy for Imaging and Direct Measurement of Intracellular Drug Target Engagement. IEEE JOURNAL OF SELECTED TOPICS IN QUANTUM ELECTRONICS : A PUBLICATION OF THE IEEE LASERS AND ELECTRO-OPTICS SOCIETY 2016; 22:6801607. [PMID: 27440991 PMCID: PMC4946648 DOI: 10.1109/jstqe.2015.2501384] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Small molecule therapeutic drugs must reach their intended cellular targets (pharmacokinetics) and engage them to modulate therapeutic effects (pharmacodynamics). These processes are often difficult to measure in vivo due to their complexities and occurrence within single cells. It has been particularly difficult to directly measure cellular drug target binding. Fluorescence polarization is commonly used in pharmacological screening assays to measure drug-protein or protein-protein interactions. We hypothesized that fluorescence polarization imaging could be adapted and used with fluorescently labeled drugs to measure drug target engagement in vivo. Here we summarize recent results using two photon fluorescence anisotropy microscopy. Our imaging technique offers quantitative pharmacological binding information of diverse molecular interactions at the microscopic level, differentiating between bound and unbound states. Used in combination with other recent advances in the development of novel fluorescently labeled drugs, we expect that the described imaging modality will provide a window into the distribution and efficacy of drugs in real time and in vivo at the cellular and subcellular level.
Collapse
Affiliation(s)
- Claudio Vinegoni
- Center for System Biology, Massachusetts General Hospital and Harvard Medical School, Richard B. Simches Research Center, 185 Cambridge Street, Boston 02114, USA
| | - John M. Dubach
- Center for System Biology, Massachusetts General Hospital and Harvard Medical School, Richard B. Simches Research Center, 185 Cambridge Street, Boston 02114, USA
| | - Paolo Fumene Feruglio
- Center for System Biology, Massachusetts General Hospital and Harvard Medical School, Richard B. Simches Research Center, 185 Cambridge Street, Boston 02114, USA and with the Department of Neurological and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy
| | - Ralph Weissleder
- Center for System Biology, Massachusetts General Hospital and Harvard Medical School, Richard B. Simches Research Center, 185 Cambridge Street, Boston 02114, USA
| |
Collapse
|
50
|
Vinegoni C, Leon Swisher C, Fumene Feruglio P, Giedt RJ, Rousso DL, Stapleton S, Weissleder R. Real-time high dynamic range laser scanning microscopy. Nat Commun 2016; 7:11077. [PMID: 27032979 PMCID: PMC4821995 DOI: 10.1038/ncomms11077] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 02/19/2016] [Indexed: 01/21/2023] Open
Abstract
In conventional confocal/multiphoton fluorescence microscopy, images are typically acquired under ideal settings and after extensive optimization of parameters for a given structure or feature, often resulting in information loss from other image attributes. To overcome the problem of selective data display, we developed a new method that extends the imaging dynamic range in optical microscopy and improves the signal-to-noise ratio. Here we demonstrate how real-time and sequential high dynamic range microscopy facilitates automated three-dimensional neural segmentation. We address reconstruction and segmentation performance on samples with different size, anatomy and complexity. Finally, in vivo real-time high dynamic range imaging is also demonstrated, making the technique particularly relevant for longitudinal imaging in the presence of physiological motion and/or for quantification of in vivo fast tracer kinetics during functional imaging. Confocal and multiphoton fluorescence microscopy often suffers from low dynamic range. Here the authors develop a high dynamic range, laser scanning fluorescence technique by simultaneously recording different light intensity ranges. The method can be adapted to commercial systems.
Collapse
Affiliation(s)
- C Vinegoni
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Richard B. Simches Research Center, 185 Cambridge Street, Boston, Massachusetts 02114, USA
| | - C Leon Swisher
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Richard B. Simches Research Center, 185 Cambridge Street, Boston, Massachusetts 02114, USA
| | - P Fumene Feruglio
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Richard B. Simches Research Center, 185 Cambridge Street, Boston, Massachusetts 02114, USA.,Department of Neurological, Biomedical and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy
| | - R J Giedt
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Richard B. Simches Research Center, 185 Cambridge Street, Boston, Massachusetts 02114, USA
| | - D L Rousso
- Center for Brain Science, Department of Molecular and Cell Biology, Harvard University, 52 Oxford Street, Cambridge, Massachusetts 02138, USA
| | - S Stapleton
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Richard B. Simches Research Center, 185 Cambridge Street, Boston, Massachusetts 02114, USA
| | - R Weissleder
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Richard B. Simches Research Center, 185 Cambridge Street, Boston, Massachusetts 02114, USA
| |
Collapse
|