1
|
Liu H, Zhao J, Xue Y, Zhang J, Bai H, Pan S, Peng B, Li L, Voelcker NH. X-Ray-Induced Drug Release for Cancer Therapy. Angew Chem Int Ed Engl 2023; 62:e202306100. [PMID: 37278399 DOI: 10.1002/anie.202306100] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 06/05/2023] [Accepted: 06/06/2023] [Indexed: 06/07/2023]
Abstract
Drug delivery systems (DDSs) are designed to deliver therapeutic agents to specific target sites while minimizing systemic toxicity. Recent developments in drug-loaded DDSs have demonstrated promising characteristics and paved new pathways for cancer treatment. Light, a prevalent external stimulus, is widely utilized to trigger drug release. However, conventional light sources primarily concentrate on the ultraviolet (UV) and visible light regions, which suffer from limited biological tissue penetration. This limitation hinders applications for deep-tissue tumor drug release. Given their deep tissue penetration and well-established application technology, X-rays have recently received attention for the pursuit of controlled drug release. With precise spatiotemporal and dosage controllability, X-rays stand as an ideal stimulus for achieving controlled drug release in deep-tissue cancer therapy. This article explores the recent advancements in using X-rays for stimulus-triggered drug release in DDSs and delves into their action mechanisms.
Collapse
Affiliation(s)
- Hui Liu
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials & Engineering (IBME) and Ningbo Institute of Northwestern Polytechnical University, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Jun Zhao
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials & Engineering (IBME) and Ningbo Institute of Northwestern Polytechnical University, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Yufei Xue
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials & Engineering (IBME) and Ningbo Institute of Northwestern Polytechnical University, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Jiaxin Zhang
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials & Engineering (IBME) and Ningbo Institute of Northwestern Polytechnical University, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Hua Bai
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials & Engineering (IBME) and Ningbo Institute of Northwestern Polytechnical University, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Sijun Pan
- The Institute of Flexible Electronics, IFE, Future Technologies), Xiamen University, Xiamen, 361005, Fujian, China
| | - Bo Peng
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials & Engineering (IBME) and Ningbo Institute of Northwestern Polytechnical University, Northwestern Polytechnical University, Xi'an, 710072, China
- Monash Institute of Pharmaceutical Sciences (MIPS), Monash University, 399 Royal Parade, Parkville, Victoria, 3052, Australia
- Wuhan National Laboratory for Optoelectronics, Advanced Biomedical Imaging Facility, 13 Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Lin Li
- The Institute of Flexible Electronics, IFE, Future Technologies), Xiamen University, Xiamen, 361005, Fujian, China
| | - Nicolas H Voelcker
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials & Engineering (IBME) and Ningbo Institute of Northwestern Polytechnical University, Northwestern Polytechnical University, Xi'an, 710072, China
- Monash Institute of Pharmaceutical Sciences (MIPS), Monash University, 399 Royal Parade, Parkville, Victoria, 3052, Australia
| |
Collapse
|
2
|
Amin H, Amin MA, Osman SK, Mohammed AM, Zayed G. Chitosan nanoparticles as a smart nanocarrier for gefitinib for tackling lung cancer: Design of experiment and in vitro cytotoxicity study. Int J Biol Macromol 2023; 246:125638. [PMID: 37392910 DOI: 10.1016/j.ijbiomac.2023.125638] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 06/25/2023] [Accepted: 06/28/2023] [Indexed: 07/03/2023]
Abstract
Due to its poor solubility and systemic side effects, gefitinib (Gef) has limited application in treatment of lung cancer. In this study, we used design of experiment (DOE) tools to gain the necessary knowledge for the synthesis of high-quality gefitinib loaded chitosan nanoparticles (Gef-CSNPs) capable of delivering and concentrating Gef at A549 cells, thereby increasing therapeutic effectiveness while decreasing adverse effects. The optimized Gef-CSNPs were characterized by SEM, TEM, DSC, XRD, and FTIR analyses. The optimized Gef-CSNPs had a particle size of 158±3.6 nm, an entrapment efficiency of 93±1.2 %, and a release of 97±0.6 % after 8 h. The in vitro cytotoxicity of the optimized Gef-CSNPs was found to be significantly higher than pure Gef (IC50 = 10.08 ± 0.76 μg/mL and IC50 = 21.65 ± 0.32 μg/mL), respectively. In the A549 human cell line, the optimized Gef-CSNPs formula outperformed pure Gef in terms of cellular uptake (3.286 ± 0.12 μg/mL and 1.777 ± 0.1 μg/mL) and apoptotic population (64.82 ± 1.25 % and 29.38 ± 1.11 %), respectively. These findings explain why researchers are so interested in using natural biopolymers to combat lung cancer, and they paint an optimistic picture of their potential as a promising tool in the fight against lung cancer.
Collapse
Affiliation(s)
- Haitham Amin
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt.
| | - Mohammed A Amin
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt; Department of Pharmaceutics, College of Pharmacy, Qassim University, Qassim 51452, Saudi Arabia.
| | - Shaaban K Osman
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt.
| | - Ahmed M Mohammed
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt.
| | - Gamal Zayed
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt; Al-Azhar Centre of Nanosciences and Applications (ACNA), Al-Azhar University, Assiut 71524, Egypt.
| |
Collapse
|
3
|
Amin H, Osman SK, Mohammed AM, Zayed G. Gefitinib-loaded starch nanoparticles for battling lung cancer: Optimization by full factorial design and in vitro cytotoxicity evaluation. Saudi Pharm J 2023; 31:29-54. [PMID: 36685309 PMCID: PMC9845129 DOI: 10.1016/j.jsps.2022.11.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022] Open
Abstract
Lung cancer is the number one killer among all cancer types. For decades, clinicians have been using conventional chemotherapeutics, but they can't rely on them alone anymore, because they poison bad cells and good cells as well. Researchers exploited nanotechnology as a potential tool to develop a platform for drug delivery to improve therapeutic efficiency. A quality by design synthesis of gefitinib-loaded starch nanoparticles (Gef-StNPs) has emerged as an essential tool to study and optimize the factors included in their synthesis. Therefore, we applied design of experiment (DOE) tools to attain the essential knowledge for the synthesis of high-quality Gef-StNPs that can deliver and concentrate the gefitinib (Gef) at A549 cells, thereby improving therapeutic efficacy and minimizing adverse effects. The in vitro cytotoxicity after exposing the A549 human lung cancer cells to the optimized Gef-StNPs was found to be much higher than that of the pure Gef (IC50 = 6.037 ± 0.24 and 21.65 ± 0.32 μg/mL, respectively). The optimized Gef-StNPs formula showed superiority over the pure Gef regarding the cellular uptake in A549 human cell line (3.976 ± 0.14 and 1.777 ± 0.1 μg/mL) and apoptotic population (77.14 ± 1.43 and 29.38 ± 1.11 %), respectively. The results elucidate why researchers have a voracious appetite for using natural biopolymers to combat lung cancer and paint an optimistic picture of their potential to be a promising tool in battling lung cancer.
Collapse
Affiliation(s)
- Haitham Amin
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt
| | - Shaaban K. Osman
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt
| | - Ahmed M. Mohammed
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt
| | - Gamal Zayed
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt
- Al-Azhar Centre of Nanosciences and Applications (ACNA), Al-Azhar University, Assiut 71524, Egypt
| |
Collapse
|
4
|
Barth ND, Mendive‐Tapia L, Subiros‐Funosas R, Ghashghaei O, Lavilla R, Maiorino L, He X, Dransfield I, Egeblad M, Vendrell M. A Bivalent Activatable Fluorescent Probe for Screening and Intravital Imaging of Chemotherapy-Induced Cancer Cell Death. ANGEWANDTE CHEMIE (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 134:e202113020. [PMID: 38505298 PMCID: PMC10947113 DOI: 10.1002/ange.202113020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Indexed: 11/11/2022]
Abstract
The detection and quantification of apoptotic cells is a key process in cancer research, particularly during the screening of anticancer therapeutics and in mechanistic studies using preclinical models. Intravital optical imaging enables high-resolution visualisation of cellular events in live organisms; however, there are few fluorescent probes that can reliably provide functional readouts in situ without interference from tissue autofluorescence. We report the design and optimisation of the fluorogenic probe Apotracker Red for real-time detection of cancer cell death. The strong fluorogenic behaviour, high selectivity, and excellent stability of Apotracker Red make it a reliable optical reporter for the characterisation of the effects of anticancer drugs in cells in vitro and for direct imaging of chemotherapy-induced apoptosis in vivo in mouse models of breast cancer.
Collapse
Affiliation(s)
- Nicole D. Barth
- Centre for Inflammation ResearchThe University of EdinburghUK
| | | | | | - Ouldouz Ghashghaei
- Laboratory of Medicinal ChemistryFaculty of Pharmacy and Institute of Biomedicine (IBUB)University of BarcelonaSpain
| | - Rodolfo Lavilla
- Laboratory of Medicinal ChemistryFaculty of Pharmacy and Institute of Biomedicine (IBUB)University of BarcelonaSpain
| | - Laura Maiorino
- Cold Spring Harbor LaboratoryCold Spring HarborNY11724USA
| | - Xue‐Yan He
- Cold Spring Harbor LaboratoryCold Spring HarborNY11724USA
| | - Ian Dransfield
- Centre for Inflammation ResearchThe University of EdinburghUK
| | - Mikala Egeblad
- Cold Spring Harbor LaboratoryCold Spring HarborNY11724USA
| | - Marc Vendrell
- Centre for Inflammation ResearchThe University of EdinburghUK
| |
Collapse
|
5
|
Barth ND, Mendive‐Tapia L, Subiros‐Funosas R, Ghashghaei O, Lavilla R, Maiorino L, He X, Dransfield I, Egeblad M, Vendrell M. A Bivalent Activatable Fluorescent Probe for Screening and Intravital Imaging of Chemotherapy-Induced Cancer Cell Death. Angew Chem Int Ed Engl 2022; 61:e202113020. [PMID: 34762762 PMCID: PMC8991960 DOI: 10.1002/anie.202113020] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Indexed: 11/21/2022]
Abstract
The detection and quantification of apoptotic cells is a key process in cancer research, particularly during the screening of anticancer therapeutics and in mechanistic studies using preclinical models. Intravital optical imaging enables high-resolution visualisation of cellular events in live organisms; however, there are few fluorescent probes that can reliably provide functional readouts in situ without interference from tissue autofluorescence. We report the design and optimisation of the fluorogenic probe Apotracker Red for real-time detection of cancer cell death. The strong fluorogenic behaviour, high selectivity, and excellent stability of Apotracker Red make it a reliable optical reporter for the characterisation of the effects of anticancer drugs in cells in vitro and for direct imaging of chemotherapy-induced apoptosis in vivo in mouse models of breast cancer.
Collapse
Affiliation(s)
- Nicole D. Barth
- Centre for Inflammation ResearchThe University of EdinburghUK
| | | | | | - Ouldouz Ghashghaei
- Laboratory of Medicinal ChemistryFaculty of Pharmacy and Institute of Biomedicine (IBUB)University of BarcelonaSpain
| | - Rodolfo Lavilla
- Laboratory of Medicinal ChemistryFaculty of Pharmacy and Institute of Biomedicine (IBUB)University of BarcelonaSpain
| | - Laura Maiorino
- Cold Spring Harbor LaboratoryCold Spring HarborNY11724USA
| | - Xue‐Yan He
- Cold Spring Harbor LaboratoryCold Spring HarborNY11724USA
| | - Ian Dransfield
- Centre for Inflammation ResearchThe University of EdinburghUK
| | - Mikala Egeblad
- Cold Spring Harbor LaboratoryCold Spring HarborNY11724USA
| | - Marc Vendrell
- Centre for Inflammation ResearchThe University of EdinburghUK
| |
Collapse
|
6
|
Tsuboi S, Jin T. In Vitro and In Vivo Fluorescence Imaging of Antibody-Drug Conjugate-Induced Tumor Apoptosis Using Annexin V-EGFP Conjugated Quantum Dots. ACS OMEGA 2022; 7:2105-2113. [PMID: 35071899 PMCID: PMC8772308 DOI: 10.1021/acsomega.1c05636] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 12/14/2021] [Indexed: 05/24/2023]
Abstract
Antibody-drug conjugates (ADCs) are conjugates of a monoclonal antibody and a cytotoxic drug that induce tumor apoptosis. The evaluation of ADC-induced tumor apoptosis is crucial for the development of ADCs for cancer therapy. To evaluate the efficacy of ADCs, we present in vitro and in vivo fluorescence imaging techniques for ADC-induced tumor apoptosis using annexin V-EGFP (EGFP: enhanced green fluorescent protein) conjugated quantum dots (annexin V-EGFP-QDs). This probe emits visible (VIS) and near-infrared (NIR) dual fluorescence at 515 nm (EGFP emission) and 850 nm (QD emission), which can be used for the detection of tumor apoptosis at the cellular and whole-body levels. By using annexin V-EGFP-QDs, we achieved VIS and NIR fluorescence imaging of human epidermal growth factor receptor 2-positive breast tumor apoptosis induced by an ADC, Kadcyla (trastuzumab emtansine). The results show that the in vitro and in vivo fluorescence imaging of ADC-induced tumor apoptosis using annexin V-EGFP-QDs is a useful tool to evaluate the efficacy of ADCs for cancer therapy.
Collapse
Affiliation(s)
- Setsuko Tsuboi
- RIKEN Center for Biosystems Dynamics
Research, RIKEN, Furuedai 6-2-3, Suita, Osaka 565-0874, Japan
| | - Takashi Jin
- RIKEN Center for Biosystems Dynamics
Research, RIKEN, Furuedai 6-2-3, Suita, Osaka 565-0874, Japan
| |
Collapse
|
7
|
Jouberton E, Schmitt S, Maisonial-Besset A, Chautard E, Penault-Llorca F, Cachin F. Interest and Limits of [18F]ML-10 PET Imaging for Early Detection of Response to Conventional Chemotherapy. Front Oncol 2021; 11:789769. [PMID: 34988022 PMCID: PMC8722713 DOI: 10.3389/fonc.2021.789769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 11/29/2021] [Indexed: 11/25/2022] Open
Abstract
One of the current challenges in oncology is to develop imaging tools to early detect the response to conventional chemotherapy and adjust treatment strategies when necessary. Several studies evaluating PET imaging with 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) as a predictive tool of therapeutic response highlighted its insufficient specificity and sensitivity. The [18F]FDG uptake reflects only tumor metabolic activity and not treatment-induced cell death, which seems to be relevant for therapeutic evaluation. Therefore, to evaluate this parameter in vivo, several cell death radiotracers have been developed in the last years. However, few of them have reached the clinical trials. This systematic review focuses on the use of [18F]ML-10 (2-(5-[18F]fluoropentyl)-2-methylmalonic acid) as radiotracer of apoptosis and especially as a measure of tumor response to treatment. A comprehensive literature review concerning the preclinical and clinical investigations conducted with [18F]ML-10 was performed. The abilities and applications of this radiotracer as well as its clinical relevance and limitations were discussed. Most studies highlighted a good ability of the radiotracer to target apoptotic cells. However, the increase in apoptosis during treatment did not correlate with the radiotracer tumoral uptake, even using more advanced image analysis (voxel-based analysis). [18F]ML-10 PET imaging does not meet current clinical expectations for early detection of the therapeutic response to conventional chemotherapy. This review has pointed out the challenges of applying various apoptosis imaging strategies in clinical trials, the current methodologies available for image analysis and the future of molecular imaging to assess this therapeutic response.
Collapse
Affiliation(s)
- Elodie Jouberton
- Service de Médecine Nucléaire, Centre Jean PERRIN, Clermont-Ferrand, France
- Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Université Clermont Auvergne, INSERM, Clermont-Ferrand, France
- *Correspondence: Elodie Jouberton,
| | - Sébastien Schmitt
- Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Université Clermont Auvergne, INSERM, Clermont-Ferrand, France
| | - Aurélie Maisonial-Besset
- Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Université Clermont Auvergne, INSERM, Clermont-Ferrand, France
| | - Emmanuel Chautard
- Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Université Clermont Auvergne, INSERM, Clermont-Ferrand, France
- Service de Pathologie, Centre Jean PERRIN, Clermont-Ferrand, France
| | - Frédérique Penault-Llorca
- Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Université Clermont Auvergne, INSERM, Clermont-Ferrand, France
- Service de Pathologie, Centre Jean PERRIN, Clermont-Ferrand, France
| | - Florent Cachin
- Service de Médecine Nucléaire, Centre Jean PERRIN, Clermont-Ferrand, France
- Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Université Clermont Auvergne, INSERM, Clermont-Ferrand, France
| |
Collapse
|
8
|
Malviya R, Verma S, Sundram S. Advancement and Strategies for the Development of Peptide-Drug Conjugates: Pharmacokinetic Modulation, Role and Clinical Evidence Against Cancer Management. Curr Cancer Drug Targets 2021; 22:286-311. [PMID: 34792003 DOI: 10.2174/1568009621666211118111506] [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: 05/11/2021] [Revised: 07/29/2021] [Accepted: 09/09/2021] [Indexed: 12/24/2022]
Abstract
Currently, many new treatment strategies are being used for the management of cancer. Among them, chemotherapy based on peptides has been of great interest due to the unique features of peptides. This review discusses the role of peptide and peptides analogues in the treatment of cancer, with special emphasis on their pharmacokinetic modulation and research progress. Low molecular weight, targeted drug delivery, enhanced permeability, etc., of the peptide-linked drug conjugates, lead to an increase in the effectiveness of cancer therapy. Various peptides have recently been developed as drugs and vaccines with an altered pharmacokinetic parameter which has subsequently been assessed in different phases of the clinical study. Peptides have made a great impact in the area of cancer therapy and diagnosis. Targeted chemotherapy and drug delivery techniques using peptides are emerging as excellent tools in minimizing problems with conventional chemotherapy. It can be concluded that new advances in using peptides to treat different types of cancer have been shown by different clinical studies indicating that peptides could be used as an ideal therapeutic method in treating cancer due to the novel advantages of peptides. The development of identifying and synthesizing novel peptides could provide a promising choice to patients with cancer.
Collapse
Affiliation(s)
- Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida. India
| | - Swati Verma
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida. India
| | - Sonali Sundram
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida. India
| |
Collapse
|
9
|
Oh K, Chi DY. Direct Fluorination Strategy for the Synthesis of Fluorine‐18 Labeled Oligopeptide—[
18
F
]
ApoPep
‐7. B KOREAN CHEM SOC 2021. [DOI: 10.1002/bkcs.12350] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Keumrok Oh
- Department of Chemistry Sogang University 35 Baekbeomro Mapogu, Seoul 04107 Korea
| | - Dae Yoon Chi
- Department of Chemistry Sogang University 35 Baekbeomro Mapogu, Seoul 04107 Korea
| |
Collapse
|
10
|
Co CM, Izuagbe S, Zhou J, Zhou N, Sun X, Borrelli J, Tang L. Click chemistry-based pre-targeting cell delivery for cartilage regeneration. Regen Biomater 2021; 8:rbab018. [PMID: 34211730 DOI: 10.1093/rb/rbab018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/25/2021] [Accepted: 04/05/2021] [Indexed: 12/12/2022] Open
Abstract
A fraction of the OA patient population is affected by post-traumatic osteoarthritis (PTOA) following acute joint injuries. Stopping or reversing the progression of PTOA following joint injury could improve long-term functional outcomes, reduced disability, and medical costs. To more effectively treat articular cartilage injury, we have developed a novel cell-based therapy that involves the pre-targeting of apoptotic chondrocytes and the delivery of healthy, metabolically active chondrocytes using click chemistry. Specifically, a pre-targeting agent was prepared via conjugating apoptotic binding peptide (ApoPep-1) and trans-cyclooctene (TCO) onto polyethylene glycol (PEG) polymer carrier. The pre-targeting agent would be introduced to injured areas of articular cartilage, leading to the accumulation of TCO groups on the injured areas from actively binding to apoptotic chondrocytes. Subsequently, methyltetrazine (Tz)-bearing chondrocytes would be immobilized on the surface of TCO-coated injured cartilage via Tz-TCO click chemistry reaction. Using an ex vivo human cartilage explant PTOA model, the effectiveness of this new approach was evaluated. Our studies show that this novel approach (Tz-TCO click chemistry) significantly enhanced the immobilization of healthy and metabolically active chondrocytes to the areas of apoptotic chondrocytes. Histological analyses demonstrated that this treatment regimen would significantly reduce the area of cartilage degeneration and enhance ECM regeneration. The results support that Tz-TCO click chemistry-mediated cell delivery approach has great potential in clinical applications for targeting and treatment of cartilage injury.
Collapse
Affiliation(s)
- Cynthia M Co
- Department of Bioengineering, University of Texas at Arlington, PO Box 19138, Arlington, TX 76019, USA
| | - Samira Izuagbe
- Department of Bioengineering, University of Texas at Arlington, PO Box 19138, Arlington, TX 76019, USA
| | - Jun Zhou
- Department of Bioengineering, University of Texas at Arlington, PO Box 19138, Arlington, TX 76019, USA
| | - Ning Zhou
- Department of Radiology, University of Texas Southwestern Medical, Dallas, TX 75390, USA
| | - Xiankai Sun
- Department of Radiology, University of Texas Southwestern Medical, Dallas, TX 75390, USA
| | - Joseph Borrelli
- Department of Bioengineering, University of Texas at Arlington, PO Box 19138, Arlington, TX 76019, USA
| | - Liping Tang
- Department of Bioengineering, University of Texas at Arlington, PO Box 19138, Arlington, TX 76019, USA
| |
Collapse
|
11
|
Vargas-Sanchez K, Losada-Barragán M, Mogilevskaya M, Novoa-Herrán S, Medina Y, Buendía-Atencio C, Lorett-Velásquez V, Martínez-Bernal J, Gonzalez-Reyes RE, Ramírez D, Petry KG. Screening for Interacting Proteins with Peptide Biomarker of Blood-Brain Barrier Alteration under Inflammatory Conditions. Int J Mol Sci 2021; 22:ijms22094725. [PMID: 33946948 PMCID: PMC8124558 DOI: 10.3390/ijms22094725] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/25/2021] [Accepted: 04/26/2021] [Indexed: 02/06/2023] Open
Abstract
Neurodegenerative diseases are characterized by increased permeability of the blood-brain barrier (BBB) due to alterations in cellular and structural components of the neurovascular unit, particularly in association with neuroinflammation. A previous screening study of peptide ligands to identify molecular alterations of the BBB in neuroinflammation by phage-display, revealed that phage clone 88 presented specific binding affinity to endothelial cells under inflammatory conditions in vivo and in vitro. Here, we aimed to identify the possible target receptor of the peptide ligand 88 expressed under inflammatory conditions. A cross-link test between phage-peptide-88 with IL-1β-stimulated human hCMEC cells, followed by mass spectrometry analysis, was used to identify the target of peptide-88. We modeled the epitope-receptor molecular interaction between peptide-88 and its target by using docking simulations. Three proteins were selected as potential target candidates and tested in enzyme-linked immunosorbent assays with peptide-88: fibronectin, laminin subunit α5 and laminin subunit β-1. Among them, only laminin subunit β-1 presented measurable interaction with peptide-88. Peptide-88 showed specific interaction with laminin subunit β-1, highlighting its importance as a potential biomarker of the laminin changes that may occur at the BBB endothelial cells under pathological inflammation conditions.
Collapse
Affiliation(s)
- Karina Vargas-Sanchez
- Grupo de Neurociencia Translacional, Facultad de Medicina, Universidad de los Andes, Bogotá 111711, Colombia
- Correspondence: ; Tel.: +57-13102405706
| | - Monica Losada-Barragán
- Grupo de Biología Celular y Funcional e Ingeniería de Moléculas, Departamento de Biología, Universidad Antonio Nariño, Bogotá 110231, Colombia; (M.L.-B.); (Y.M.)
| | - Maria Mogilevskaya
- Grupo de Investigación GINIC-HUS, Universidad ECCI, Bogotá 111311, Colombia;
| | - Susana Novoa-Herrán
- Grupo de Investigación en Hormonas (Hormone Research Laboratory), Departamento de Química, Universidad Nacional de Colombia, Bogotá 111321, Colombia; or
- Grupo de Fisiología Molecular, Subdirección de Investigación Científica y Tecnológica, Instituto Nacional de Salud, Bogotá 111321, Colombia
| | - Yehidi Medina
- Grupo de Biología Celular y Funcional e Ingeniería de Moléculas, Departamento de Biología, Universidad Antonio Nariño, Bogotá 110231, Colombia; (M.L.-B.); (Y.M.)
| | - Cristian Buendía-Atencio
- Grupo de Investigación en Modelado y Computación Científica, Departamento de Química, Universidad Antonio Nariño, Bogotá 110231, Colombia;
| | - Vaneza Lorett-Velásquez
- Facultad de Medicina y Ciencias de la Salud, Universidad Militar Nueva Granada, Bogotá 110231, Colombia; (V.L.-V.); (J.M.-B.)
| | - Jessica Martínez-Bernal
- Facultad de Medicina y Ciencias de la Salud, Universidad Militar Nueva Granada, Bogotá 110231, Colombia; (V.L.-V.); (J.M.-B.)
| | - Rodrigo E. Gonzalez-Reyes
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencia Neurovitae-UR, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá 111711, Colombia;
| | - David Ramírez
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, El llano Subercaseaux 2801, Santiago 8900000, Chile;
| | - Klaus G. Petry
- INSERM U1049 and U1029 Neuroinflammation and Angiogenesis Group, Bordeaux University, F33000 Bordeaux, France;
| |
Collapse
|
12
|
So IS, Kang JH, Hong JW, Sung S, Hasan AF, Sa KH, Han SW, Kim IS, Kang YM. A novel apoptosis probe, cyclic ApoPep-1, for in vivo imaging with multimodal applications in chronic inflammatory arthritis. Apoptosis 2021; 26:209-218. [PMID: 33655467 DOI: 10.1007/s10495-021-01659-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2021] [Indexed: 11/26/2022]
Abstract
Apoptosis plays an essential role in the pathophysiologic processes of rheumatoid arthritis. A molecular probe that allows spatiotemporal observation of apoptosis in vitro, in vivo, and ex vivo concomitantly would be useful to monitoring or predicting pathophysiologic stages. In this study we investigated whether cyclic apoptosis-targeting peptide-1 (CApoPep-1) can be used as an apoptosis imaging probe in inflammatory arthritis. We tested the utility of CApoPep-1 for detecting apoptotic immune cells in vitro and ex vivo using flow cytometry and immunofluorescence. The feasibility of visualizing and quantifying apoptosis using this probe was evaluated in a murine collagen-induced arthritis (CIA) model, especially after treatment. CApoPep-1 peptide may successfully replace Annexin V for in vitro and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay for ex vivo in the measurement of apoptotic cells, thus function as a sensitive probe enough to be used clinically. In vivo imaging in CIA mice revealed that CApoPep-1 had 42.9 times higher fluorescence intensity than Annexin V for apoptosis quantification. Furthermore, it may be used as an imaging probe for early detection of apoptotic response in situ after treatment. The CApoPep-1 signal was mostly co-localized with the TUNEL signal (69.6% of TUNEL+ cells) in defined cell populations in joint tissues of CIA mice. These results demonstrate that CApoPep-1 is sufficiently sensitive to be used as an apoptosis imaging probe for multipurpose applications which could detect the same target across in vitro, in vivo, to ex vivo in inflammatory arthritis.
Collapse
Affiliation(s)
- In-Seop So
- Department of Internal Medicine (Rheumatology), Kyungpook National University School of Medicine, 680 Gukchaebosang-ro, Junggu, Daegu, 41944, Republic of Korea
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Jin Hee Kang
- Department of Internal Medicine (Rheumatology), Kyungpook National University School of Medicine, 680 Gukchaebosang-ro, Junggu, Daegu, 41944, Republic of Korea
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea
- Department of Biochemistry and Cell Biology, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Jung Wan Hong
- Department of Internal Medicine (Rheumatology), Kyungpook National University School of Medicine, 680 Gukchaebosang-ro, Junggu, Daegu, 41944, Republic of Korea
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Shijin Sung
- Department of Internal Medicine (Rheumatology), Kyungpook National University School of Medicine, 680 Gukchaebosang-ro, Junggu, Daegu, 41944, Republic of Korea
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea
- Department of Biochemistry and Cell Biology, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Al Faruque Hasan
- Department of Internal Medicine (Rheumatology), Kyungpook National University School of Medicine, 680 Gukchaebosang-ro, Junggu, Daegu, 41944, Republic of Korea
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea
- Department of Biochemistry and Cell Biology, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Keum Hee Sa
- Department of Internal Medicine (Rheumatology), Kyungpook National University School of Medicine, 680 Gukchaebosang-ro, Junggu, Daegu, 41944, Republic of Korea
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea
- Department of Biochemistry and Cell Biology, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Seung Woo Han
- Department of Internal Medicine (Rheumatology), Kyungpook National University School of Medicine, 680 Gukchaebosang-ro, Junggu, Daegu, 41944, Republic of Korea
| | - In San Kim
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Young Mo Kang
- Department of Internal Medicine (Rheumatology), Kyungpook National University School of Medicine, 680 Gukchaebosang-ro, Junggu, Daegu, 41944, Republic of Korea.
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea.
- Department of Biochemistry and Cell Biology, Kyungpook National University School of Medicine, Daegu, Republic of Korea.
| |
Collapse
|
13
|
Tsuboi S, Jin T. Dual-colour (near-infrared/visible) emitting annexin V for fluorescence imaging of tumour cell apoptosis in vitro and in vivo. RSC Adv 2020; 10:38244-38250. [PMID: 35517522 PMCID: PMC9057337 DOI: 10.1039/d0ra06495e] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/03/2020] [Indexed: 12/03/2022] Open
Abstract
Indocyanine green (ICG) labelled recombinant annexin V proteins (ICG-EGFP-Annexin V and ICG-mPlum-Annexin V) were synthesized for dual-colour fluorescence imaging of tumour cell apoptosis in vitro and in vivo. The ICG-labelled fluorescent annexin V proteins showed dual (near-infrared and visible) fluorescence emissions with binding ability to phosphatidylserines on the plasma membranes of apoptotic cells. Although several types of fluorescence labelled annexin V (e.g. FITC-annexin V, Cy3- and Cy5-annexin V) have been reported, there are no dual-colour (near-infrared/visible) emitting apoptosis-detection probes which can be used in vitro and in vivo. In this paper, the utilities of the dual-colour fluorescent annexin V are demonstrated for in vitro and in vivo fluorescence imaging of the apoptosis of human breast tumour cells induced by an antibody-drug conjugate, Kadcyla. The results suggest that the present annexin V probes will be useful to visualize the action of anti-cancer drugs against tumours both at the cellular and whole-body level.
Collapse
Affiliation(s)
- Setsuko Tsuboi
- RIKEN Center for Biosystems Dynamics Research (BDR) RIKEN Furuedai 6-2-3 Suita Osaka 565-0874 Japan
| | - Takashi Jin
- RIKEN Center for Biosystems Dynamics Research (BDR) RIKEN Furuedai 6-2-3 Suita Osaka 565-0874 Japan
- Graduate School of Frontier Biosciences, Osaka University Yamada-oka 1-3 Suita Osaka 565-0871 Japan
| |
Collapse
|
14
|
Zhang D, Jin Q, Jiang C, Gao M, Ni Y, Zhang J. Imaging Cell Death: Focus on Early Evaluation of Tumor Response to Therapy. Bioconjug Chem 2020; 31:1025-1051. [PMID: 32150392 DOI: 10.1021/acs.bioconjchem.0c00119] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Cell death plays a prominent role in the treatment of cancer, because most anticancer therapies act by the induction of cell death including apoptosis, necrosis, and other pathways of cell death. Imaging cell death helps to identify treatment responders from nonresponders and thus enables patient-tailored therapy, which will increase the likelihood of treatment response and ultimately lead to improved patient survival. By taking advantage of molecular probes that specifically target the biomarkers/biochemical processes of cell death, cell death imaging can be successfully achieved. In recent years, with the increased understanding of the molecular mechanism of cell death, a variety of well-defined biomarkers/biochemical processes of cell death have been identified. By targeting these established cell death biomarkers/biochemical processes, a set of molecular imaging probes have been developed and evaluated for early monitoring treatment response in tumors. In this review, we mainly present the recent advances in identifying useful biomarkers/biochemical processes for both apoptosis and necrosis imaging and in developing molecular imaging probes targeting these biomarkers/biochemical processes, with a focus on their application in early evaluation of tumor response to therapy.
Collapse
Affiliation(s)
- Dongjian Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, P.R. China.,Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, P.R. China
| | - Qiaomei Jin
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, P.R. China.,Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, P.R. China
| | - Cuihua Jiang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, P.R. China.,Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, P.R. China
| | - Meng Gao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, P.R. China.,Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, P.R. China
| | - Yicheng Ni
- Theragnostic Laboratory, Campus Gasthuisberg, KU Leuven, Leuven 3000, Belgium
| | - Jian Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, P.R. China.,Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, P.R. China
| |
Collapse
|
15
|
Xu H, Cao B, Li Y, Mao C. Phage nanofibers in nanomedicine: Biopanning for early diagnosis, targeted therapy, and proteomics analysis. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 12:e1623. [PMID: 32147974 DOI: 10.1002/wnan.1623] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 01/02/2020] [Accepted: 02/04/2020] [Indexed: 12/16/2022]
Abstract
Display of a peptide or protein of interest on the filamentous phage (also known as bacteriophage), a biological nanofiber, has opened a new route for disease diagnosis and therapy as well as proteomics. Earlier phage display was widely used in protein-protein or antigen-antibody studies. In recent years, its application in nanomedicine is becoming increasingly popular and encouraging. We aim to review the current status in this research direction. For better understanding, we start with a brief introduction of basic biology and structure of the filamentous phage. We present the principle of phage display and library construction method on the basis of the filamentous phage. We summarize the use of the phage displayed peptide library for selecting peptides with high affinity against cells or tissues. We then review the recent applications of the selected cell or tissue targeting peptides in developing new targeting probes and therapeutics to advance the early diagnosis and targeted therapy of different diseases in nanomedicine. We also discuss the integration of antibody phage display and modern proteomics in discovering new biomarkers or target proteins for disease diagnosis and therapy. Finally, we propose an outlook for further advancing the potential impact of phage display on future nanomedicine. This article is categorized under: Biology-Inspired Nanomaterials > Protein and Virus-Based Structures.
Collapse
Affiliation(s)
- Hong Xu
- Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma, USA
| | - Binrui Cao
- Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma, USA
| | - Yan Li
- Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma, USA
| | - Chuanbin Mao
- Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma, USA
| |
Collapse
|
16
|
Jung H, Park S, Gunassekaran GR, Jeon M, Cho YE, Baek MC, Park JY, Shim G, Oh YK, Kim IS, Kim C, Lee B. A Peptide Probe Enables Photoacoustic-Guided Imaging and Drug Delivery to Lung Tumors in K-rasLA2 Mutant Mice. Cancer Res 2019; 79:4271-4282. [PMID: 31243044 DOI: 10.1158/0008-5472.can-18-3089] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 05/12/2019] [Accepted: 06/18/2019] [Indexed: 11/16/2022]
Abstract
The lack of molecular targets and targeting probes remains a major drawback for targeted imaging and drug delivery in lung cancer. In this study, we exploited in vivo phage display to identify a novel targeting probe that homes to the tumor in a K-rasLA2 mutant mouse lung cancer model. Compared with other candidate peptides selected from 5 rounds of phage display, the CRQTKN peptide homed to tumor nodules in the lung of mutant mice at higher levels. Photoacoustic tomography of mutant mice detected lung tumors via tumor homing of the near-infrared fluorescence dye-labeled CRQTKN peptide. Ex vivo photoacoustic images of isolated organs further demonstrated tumor homing of the CRQTKN peptide, whereas minimal accumulation was observed in control organs, such as the liver. Compared with untargeted liposomes and doxorubicin, doxorubicin-loaded liposomes whose surface was modified with the CRQTKN peptide more efficiently delivered doxorubicin and reduced the number or size of tumor lesions in K-rasLA2 mutant mice. Analysis of hematologic parameters and liver and kidney function showed no significant systemic side effects by the treatments. Affinity-based identification was used to detect TNF receptor superfamily member 19L (TNFRSF19L), which was upregulated in lung tumors of mutant mice, as the receptor for the CRQTKN peptide. In conclusion, these results suggest that the CRQTKN peptide is a promising targeting probe for photoacoustic-guided detection and drug delivery to lung cancer, and acts by binding to TNFRSF19L. SIGNIFICANCE: These findings present a new tumor-targeting probe for photoacoustic-guided detection and drug delivery.
Collapse
Affiliation(s)
- Hyunkyung Jung
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,CMRI, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Sungjo Park
- Department of Creative IT Engineering, Pohang University of Science and Technology, Gyeongbuk, Republic of Korea.,Department of Electrical Engineering, Pohang University of Science and Technology, Gyeongbuk, Republic of Korea
| | - Gowri Rangaswamy Gunassekaran
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,CMRI, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Mansik Jeon
- School of Electronics Engineering, College of IT Engineering, Kyungpook National University, Daegu, Republic of Korea
| | - Young-Eun Cho
- Department of Molecular Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Moon-Chang Baek
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,CMRI, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,Department of Molecular Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Jae Yong Park
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,CMRI, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Gayong Shim
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Yu-Kyoung Oh
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - In-San Kim
- Biomedical Center, Korea Institute of Science and Technology, Seoul, Republic of Korea.,KU-KIST School, Korea University, Seoul, Republic of Korea
| | - Chulhong Kim
- Department of Creative IT Engineering, Pohang University of Science and Technology, Gyeongbuk, Republic of Korea. .,Department of Electrical Engineering, Pohang University of Science and Technology, Gyeongbuk, Republic of Korea
| | - Byungheon Lee
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea. .,BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,CMRI, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
17
|
The Continuing Evolution of Molecular Functional Imaging in Clinical Oncology: The Road to Precision Medicine and Radiogenomics (Part I). Mol Diagn Ther 2019; 23:1-26. [PMID: 30411216 DOI: 10.1007/s40291-018-0366-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The present era of precision medicine sees 'cancer' as a consequence of molecular derangements occurring at the commencement of the disease process, with morphologic changes happening much later in the process of tumorigenesis. Conventional imaging techniques, such as computed tomography (CT), ultrasound, and magnetic resonance imaging (MRI), play an integral role in the detection of disease at a macroscopic level. However, molecular functional imaging (MFI) techniques entail the visualisation and quantification of biochemical and physiological processes occurring during tumorigenesis, and thus has the potential to play a key role in heralding the transition from the concept of 'one size fits all' to 'precision medicine'. Integration of MFI with other fields of tumour biology such as genomics has spawned a novel concept called 'radiogenomics', which could serve as an indispensable tool in translational cancer research. With recent advances in medical image processing, such as texture analysis, deep learning, and artificial intelligence (AI), the future seems promising; however, their clinical utility remains unproven at present. Despite the emergence of novel imaging biomarkers, a majority of these require validation before clinical translation is possible. In this two-part review, we discuss the systematic collaboration across structural, anatomical, and molecular imaging techniques that constitute MFI. Part I reviews positron emission tomography, radiogenomics, AI, and optical imaging, while part II reviews MRI, CT and ultrasound, their current status, and recent advances in the field of precision oncology.
Collapse
|
18
|
Oliveira MC, Correia JDG. Biomedical applications of radioiodinated peptides. Eur J Med Chem 2019; 179:56-77. [PMID: 31238251 DOI: 10.1016/j.ejmech.2019.06.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/04/2019] [Accepted: 06/04/2019] [Indexed: 02/08/2023]
Abstract
The overexpression of peptide receptors in certain tumors as compared to endogeneous expression levels represents the molecular basis for the design of peptide-based tools for targeted nuclear imaging and therapy. Receptor targeting with radiolabelled peptides became a very important imaging and/or therapeutic approach in nuclear medicine and oncology. A great variety of peptides has been radiolabelled with clinical relevant radionuclides, such as radiometals and radiohalogens. However, to the best of our knowledge concise and updated reviews providing information about the biomedical application of radioiodinated peptides are still missing. This review outlines the synthetic efforts in the preparation of radioiodinated peptides highlighting the importance of radioiodine in nuclear medicine, giving an overview of the most relevant radioiodination strategies that have been employed and describes relevant examples of their use in the biomedical field.
Collapse
Affiliation(s)
- Maria Cristina Oliveira
- Centro de Ciências e Tecnologias Nucleares, Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, Estrada Nacional 10 (km 139,7), 2695-066, Bobadela LRS, Portugal.
| | - João D G Correia
- Centro de Ciências e Tecnologias Nucleares, Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, Estrada Nacional 10 (km 139,7), 2695-066, Bobadela LRS, Portugal.
| |
Collapse
|
19
|
Vadevoo SMP, Gurung S, Khan F, Haque ME, Gunassekaran GR, Chi L, Permpoon U, Lee B. Peptide-based targeted therapeutics and apoptosis imaging probes for cancer therapy. Arch Pharm Res 2019; 42:150-158. [DOI: 10.1007/s12272-019-01125-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 01/28/2019] [Indexed: 12/22/2022]
|
20
|
Bumbaca B, Li Z, Shah DK. Pharmacokinetics of protein and peptide conjugates. Drug Metab Pharmacokinet 2019; 34:42-54. [PMID: 30573392 PMCID: PMC6378135 DOI: 10.1016/j.dmpk.2018.11.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 10/29/2018] [Accepted: 11/19/2018] [Indexed: 12/20/2022]
Abstract
Protein and peptide conjugates have become an important component of therapeutic and diagnostic medicine. These conjugates are primarily designed to improve pharmacokinetics (PK) of those therapeutic or imaging agents, which do not possess optimal disposition characteristics. In this review we have summarized preclinical and clinical PK of diverse protein and peptide conjugates, and have showcased how different conjugation approaches are used to obtain the desired PK. We have classified the conjugates into peptide conjugates, non-targeted protein conjugates, and targeted protein conjugates, and have highlighted diagnostic and therapeutic applications of these conjugates. In general, peptide conjugates demonstrate very short half-life and rapid renal elimination, and they are mainly designed to achieve high contrast ratio for imaging agents or to deliver therapeutic agents at sites not reachable by bulky or non-targeted proteins. Conjugates made from non-targeted proteins like albumin are designed to increase the half-life of rapidly eliminating therapeutic or imaging agents, and improve their delivery to tissues like solid tumors and inflamed joints. Targeted protein conjugates are mainly developed from antibodies, antibody derivatives, or endogenous proteins, and they are designed to improve the contrast ratio of imaging agents or therapeutic index of therapeutic agents, by enhancing their delivery to the site-of-action.
Collapse
Affiliation(s)
- Brandon Bumbaca
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, USA
| | - Zhe Li
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, USA
| | - Dhaval K Shah
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, USA.
| |
Collapse
|
21
|
Cho YS, Chung SW, Kim HR, Won TH, Choi JU, Kim IS, Kim SY, Byun Y. The novel strategy for concurrent chemoradiotherapy by conjugating the apoptotic cell-binding moiety to caspase-3 activated doxorubicin prodrug. J Control Release 2019; 296:241-249. [PMID: 30659905 DOI: 10.1016/j.jconrel.2019.01.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 12/26/2018] [Accepted: 01/16/2019] [Indexed: 11/17/2022]
Abstract
The selective targeting of cytotoxic agents to a tumor has shown limited success by difficulties in identifying the appropriate target molecules, and more importantly, by the phenotypically dynamic nature of the tumor cells and intratumoral heterogeneity. In an attempt to overcome these issues and efficiently deliver cytotoxic drugs to the tumor, we previously reported a strategy termed radiation-induced apoptosis-targeted chemotherapy (RIATC), which utilizes the radiotherapy for intentionally triggering the caspase-3 and in situ amplification of tumor apoptosis by caspase-3 activated prodrug. Herein, we propose an advanced form of RIATC prodrug, AP1-DEVD-S-DOX, that could more actively target to the ligands of radiation-induced tumor cells, which could accumulate more prodrugs, thereby allowing more effective in situ activation and amplification of tumor apoptosis, comparing to RIATC. Indeed, AP1-DEVD-S-DOX was able to exert improved doxorubicin (DOX) delivery to the tumor and anticancer effect than the RIATC prodrug that lacks apoptotic cell-binding property but having a similar degree of off-target distribution in the other organs. Accordingly, AP1-DEVD-S-DOX could be an efficient prodrug for concurrent chemoradiotherapy by selectively delivering doxorubicin to the tumor with less systemic cytotoxicity.
Collapse
Affiliation(s)
- Young Seok Cho
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergent Science and Technology, Seoul National University, Seoul 08826, South Korea
| | - Seung Woo Chung
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, South Korea; Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Ha Rin Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Tae Hyung Won
- Boyce Thompson Institute and Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Jeong Uk Choi
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - In-San Kim
- Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, South Korea; KU-KIST School, Korea University, Seoul 02841, South Korea
| | - Sang Yoon Kim
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea.
| | - Youngro Byun
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergent Science and Technology, Seoul National University, Seoul 08826, South Korea; Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, South Korea.
| |
Collapse
|
22
|
SPECT Imaging of Treatment-Related Tumor Necrosis Using Technetium-99m-Labeled Rhein. Mol Imaging Biol 2018; 21:660-668. [DOI: 10.1007/s11307-018-1285-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
23
|
Haque ME, Khan F, Chi L, Gurung S, Vadevoo SMP, Park RW, Kim DK, Kim SK, Lee B. A Phage Display-Identified Peptide Selectively Binds to Kidney Injury Molecule-1 (KIM-1) and Detects KIM-1-Overexpressing Tumors in vivo. Cancer Res Treat 2018; 51:861-875. [PMID: 30282451 PMCID: PMC6639206 DOI: 10.4143/crt.2018.214] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 09/26/2018] [Indexed: 01/05/2023] Open
Abstract
PURPOSE This study was carried out to identify a peptide that selectively binds to kidney injury molecule-1 (KIM-1) by screening a phage-displayed peptide library and to use the peptide for the detection of KIM-1overexpressing tumors in vivo. MATERIALS AND METHODS Biopanning of a phage-displayed peptide library was performed on KIM-1-coated plates. The binding of phage clones, peptides, and a peptide multimer to the KIM-1 protein and KIM-1-overexpressing and KIM-1-low expressing cells was examined by enzyme-linked immunosorbent assay, fluorometry, and flow cytometry. A biotin-peptide multimer was generated using NeutrAvidin. In vivo homing of the peptide to KIM-1-overexpressing and KIM1-low expressing tumors in mice was examined by whole-body fluorescence imaging. RESULTS A phage clone displaying the CNWMINKEC peptide showed higher binding affinity to KIM-1 and KIM-1-overexpressing 769-P renal tumor cells compared to other phage clones selected after biopanning. The CNWMINKEC peptide and a NeutrAvidin/biotin-CNWMINKEC multimer selectively bound to KIM-1 over albumin and to KIM-1-overexpressing 769-P cells and A549 lung tumor cells compared to KIM-1-low expressing HEK293 normal cells. Co-localization and competition assays using an anti-KIM-1 antibody demonstrated that the binding of the CNWMINKEC peptide to 769-P cells was specifically mediated by KIM-1. The CNWMINKEC peptide was not cytotoxic to cells and was stable for up to 24 hours in the presence of serum. Whole-body fluorescence imaging demonstrated selective homing of the CNWM-INKEC peptide to KIM-1-overexpressing A498 renal tumor compared to KIM1-low expressing HepG2 liver tumor in mice. CONCLUSION The CNWMINKEC peptide is a promising probe for in vivo imaging and detection of KIM-1‒overexpressing tumors.
Collapse
Affiliation(s)
- Md Enamul Haque
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Korea.,BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu, Korea.,CMRI, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Fatima Khan
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Lianhua Chi
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Smriti Gurung
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Korea
| | | | - Rang-Woon Park
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Dong-Kyu Kim
- Laboratory Animal Center, Daegu Gyeongbuk Medical Innovation Foundation, Daegu, Korea
| | - Sang Kyoon Kim
- Laboratory Animal Center, Daegu Gyeongbuk Medical Innovation Foundation, Daegu, Korea
| | - Byungheon Lee
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Korea.,BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu, Korea.,CMRI, School of Medicine, Kyungpook National University, Daegu, Korea
| |
Collapse
|
24
|
Shim HE, Mushtaq S, Song L, Lee CH, Lee H, Jeon J. Development of a new thiol-reactive prosthetic group for site-specific labeling of biomolecules with radioactive iodine. Bioorg Med Chem Lett 2018; 28:2875-2878. [PMID: 30049578 DOI: 10.1016/j.bmcl.2018.07.028] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 06/21/2018] [Accepted: 07/17/2018] [Indexed: 12/11/2022]
Abstract
In this report, we describe the radiosynthesis of a new thiol-targeting prosthetic group for efficient radioactive iodine labeling of biomolecules. Radioiodination using the precursor 3 was performed to obtain 125I-labeled tetrazole 4b with high radiochemical yield (73%) and radiochemical purity. Using the radiolabeled 4b, a single free cysteine containing peptide and human serum albumin were labeled with 125I in modest-to-good radiochemical yields (65-99%) under mildly reactive conditions. A biodistribution study of [125I]7 in normal ICR mice exhibited lower thyroid uptake values than those of 125I-labeled human serum albumin prepared via a traditional radiolabeling method. Thus, [125I]7 could be employed as an effective radiotracer for molecular imaging and biodistribution studies. The results clearly demonstrate that 4b has the potential to be effectively implemented as a prosthetic group in the preparation of radiolabeled biomolecules.
Collapse
Affiliation(s)
- Ha Eun Shim
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup 56212, Republic of Korea; Department of Chemistry, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Sajid Mushtaq
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup 56212, Republic of Korea; Radiation Biotechnology and Applied Radioisotope Science, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Lee Song
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup 56212, Republic of Korea
| | - Chang Heon Lee
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup 56212, Republic of Korea
| | - Hyosun Lee
- Department of Chemistry, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Jongho Jeon
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup 56212, Republic of Korea; Radiation Biotechnology and Applied Radioisotope Science, University of Science and Technology, Daejeon 34113, Republic of Korea.
| |
Collapse
|
25
|
Bae SM, Park SJ, Choi M, Song M, Cho YE, Do EJ, Ryu YM, Park S, Lee BH, Lee SW, Hwang SW, Park SH, Yang DH, Ye BD, Byeon JS, Yang SK, Joo J, Kim SY, Myung SJ. PSP1, a Phosphatidylserine-Recognizing Peptide, Is Useful for Visualizing Radiation-Induced Apoptosis in Colorectal Cancer In Vitro and In Vivo. Transl Oncol 2018; 11:1044-1052. [PMID: 29982102 PMCID: PMC6034579 DOI: 10.1016/j.tranon.2018.06.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 06/12/2018] [Accepted: 06/15/2018] [Indexed: 02/07/2023] Open
Abstract
Accurate and timely visualization of apoptotic status in response to radiation is necessary for deciding whether to continue radiation or change to another mode of treatment. This is especially critical in patients with colorectal cancer, which requires a delicate combination of surgery, radiation, and chemotherapy in order to achieve optimal outcome. In this study, we investigated the potential of phosphatidylserine-recognizing peptide 1 (PSP1) as an apoptosis-targeting probe, which identifies phosphatidylserine on cell surfaces. We first screened colon cancer cell lines for their sensitivity to radiation and selected two cell lines: HCT116 and HT29. Cell binding assay using fluorescence-activated cell sorting and optical imaging showed that HCT116 cells had better binding to PSP1 than HT29 cells. Thus, mouse xenograft model using HCT116 cells was generated and was topically irradiated with either single or fractionated dose of radiation followed by systemic administration of PSP1 for subsequent molecular optical imaging. We confirmed that the PSP1 probe was selectively bound to apoptosis-induced tumor in a radiation dose-dependent manner. We also observed that fractionated radiation regimen, which is recently being used in clinical situation, was more effective in inducing tumor apoptosis than corresponding single-dose radiation treatment. We then evaluated the correlation between tumor targeting of PSP1 and suppression effect of tumor development and found that tumor volume and fluorescence intensity were correlated before (correlation coefficient r2 = 0.534) and after (r2 = 0.848) radiation therapy. Our study shows that PSP1 peptide is an efficient index probe for deciding "go or no-go" for radiation therapy in colorectal cancer.
Collapse
Affiliation(s)
- Sang Mun Bae
- Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of medicine, Seoul, Republic of Korea
| | - Soo Jung Park
- Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of medicine, Seoul, Republic of Korea; Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of medicine, Seoul, Republic of Korea
| | - Myoungeun Choi
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Miyeoun Song
- Department of Life and Nanopharmaceutical Science, Kyung Hee University, Seoul, Republic of Korea
| | - Young Eun Cho
- Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of medicine, Seoul, Republic of Korea
| | - Eun-Ju Do
- Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of medicine, Seoul, Republic of Korea
| | - Yeon-Mi Ryu
- Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of medicine, Seoul, Republic of Korea
| | - Sunha Park
- Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of medicine, Seoul, Republic of Korea
| | - Byung-Heon Lee
- Department of Biochemistry and Cell Biology, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Sang-Wook Lee
- Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sung Wook Hwang
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sang Hyoung Park
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Dong-Hoon Yang
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Byong Duk Ye
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jeong-Sik Byeon
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Suk-Kyun Yang
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jinmyoung Joo
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sang-Yeob Kim
- Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of medicine, Seoul, Republic of Korea; Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Seung-Jae Myung
- Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of medicine, Seoul, Republic of Korea; Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
26
|
Choi H, Han JH, Lim SY, Lee I, Cho YS, Chun EJ, Lee WW. Imaging of Myocardial Ischemia-Reperfusion Injury Using Sodium [ 18F]Fluoride Positron Emission Tomography/Computed Tomography in Rats and Humans. Mol Imaging 2018; 16:1536012117704767. [PMID: 28654382 PMCID: PMC5470131 DOI: 10.1177/1536012117704767] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Positron emission tomography (PET)/computed tomography (CT) using sodium [18F]fluoride (Na[18F]F) has been proven to be a promising hot-spot imaging modality for myocardial infarction (MI). We investigated Na[18F]F uptake in ischemia–reperfusion injury (IRI) of rats and humans. Sodium [18F]fluoride PET/CT was performed in Sprague-Dawley rats that had IRI surgery, and it readily demonstrated prominent Na[18F]F uptake in the infarct area post-IRI. Sodium [18F]fluoride uptake was matched with negative 2,3,5-triphenyl-2H-tetrazolium chloride staining results, accompanied by myocardial apoptosis and associated with positive calcium staining results. Furthermore, area at risk was negative for Na[18F]F uptake. Cyclosporine A (CysA) treatment reduced standardized uptake value of 18F over the infarct area, and a significant decrease in infarct size was also observed by the CysA treatment. In humans, Na[18F]F PET/CT readily demonstrated increased Na[18F]F uptake in the 2 patients with MI post-percutaneous coronary intervention. In conclusion, this study sheds light on the potential utility of Na[18F]F PET/CT as a hot-spot imaging modality for myocardial IRI.
Collapse
Affiliation(s)
- Hongyoon Choi
- 1 Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Jeong Hee Han
- 1 Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Sue Yeon Lim
- 1 Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Inki Lee
- 1 Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Young-Seok Cho
- 2 Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Eun Ju Chun
- 3 Department of Radiology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Won Woo Lee
- 1 Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seoul, Korea.,4 Institute of Radiation Medicine, Medical Research Center, Seoul National University, Seoul, Korea
| |
Collapse
|
27
|
Rybczynska AA, Boersma HH, de Jong S, Gietema JA, Noordzij W, Dierckx RAJO, Elsinga PH, van Waarde A. Avenues to molecular imaging of dying cells: Focus on cancer. Med Res Rev 2018. [PMID: 29528513 PMCID: PMC6220832 DOI: 10.1002/med.21495] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Successful treatment of cancer patients requires balancing of the dose, timing, and type of therapeutic regimen. Detection of increased cell death may serve as a predictor of the eventual therapeutic success. Imaging of cell death may thus lead to early identification of treatment responders and nonresponders, and to “patient‐tailored therapy.” Cell death in organs and tissues of the human body can be visualized, using positron emission tomography or single‐photon emission computed tomography, although unsolved problems remain concerning target selection, tracer pharmacokinetics, target‐to‐nontarget ratio, and spatial and temporal resolution of the scans. Phosphatidylserine exposure by dying cells has been the most extensively studied imaging target. However, visualization of this process with radiolabeled Annexin A5 has not become routine in the clinical setting. Classification of death modes is no longer based only on cell morphology but also on biochemistry, and apoptosis is no longer found to be the preponderant mechanism of cell death after antitumor therapy, as was earlier believed. These conceptual changes have affected radiochemical efforts. Novel probes targeting changes in membrane permeability, cytoplasmic pH, mitochondrial membrane potential, or caspase activation have recently been explored. In this review, we discuss molecular changes in tumors which can be targeted to visualize cell death and we propose promising biomarkers for future exploration.
Collapse
Affiliation(s)
- Anna A Rybczynska
- Molecular Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.,Department of Genetics, University of Groningen, Groningen, the Netherlands
| | - Hendrikus H Boersma
- Molecular Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.,Department of Clinical Pharmacy & Pharmacology, University of Groningen, Groningen, the Netherlands
| | - Steven de Jong
- Department of Medical Oncology, University of Groningen, Groningen, the Netherlands
| | - Jourik A Gietema
- Department of Medical Oncology, University of Groningen, Groningen, the Netherlands
| | - Walter Noordzij
- Molecular Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Rudi A J O Dierckx
- Molecular Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.,Department of Nuclear Medicine, Ghent University, Ghent, Belgium
| | - Philip H Elsinga
- Molecular Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Aren van Waarde
- Molecular Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
28
|
Shekhar A, Heeger P, Reutelingsperger C, Arbustini E, Narula N, Hofstra L, Bax JJ, Narula J. Targeted Imaging for Cell Death in Cardiovascular Disorders. JACC Cardiovasc Imaging 2018; 11:476-493. [DOI: 10.1016/j.jcmg.2017.11.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 11/19/2017] [Accepted: 11/27/2017] [Indexed: 01/30/2023]
|
29
|
Preclinical Evaluation of Radioiodinated Hoechst 33258 for Early Prediction of Tumor Response to Treatment of Vascular-Disrupting Agents. CONTRAST MEDIA & MOLECULAR IMAGING 2018; 2018:5237950. [PMID: 29681781 PMCID: PMC5846351 DOI: 10.1155/2018/5237950] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 10/17/2017] [Accepted: 12/04/2017] [Indexed: 11/17/2022]
Abstract
This study aimed to explore the use of 131I-Hoechst 33258 (131I-H33258) for early prediction of tumor response to vascular-disrupting agents (VDAs) with combretastatin-A4 phosphate (CA4P) as a representative. Necrosis avidity of 131I-H33258 was evaluated in mouse models with muscle necrosis and blocking was used to confirm the tracer specificity. Therapy response was evaluated by 131I-H33258 SPECT/CT imaging 24 h after CA4P therapy in W256 tumor-bearing rats. Radiotracer uptake in tumors was validated ex vivo using γ-counting, autoradiography, and histopathological staining. Results showed that 131I-H33258 had predominant necrosis avidity and could specifically bind to necrotic tissue. SPECT/CT imaging demonstrated that an obvious “hot spot” could be observed in the CA4P-treated tumor. Ex vivo γ-counting revealed 131I-H33258 uptake in tumors was increased 2.8-fold in rats treated with CA4P relative to rats treated with vehicle. Autoradiography and corresponding H&E staining suggested that 131I-H33258 was mainly localized in necrotic tumor area and the higher overall uptake in the treated tumors was attributed to the increased necrosis. These results suggest that 131I-H33258 can be used to image induction of cell necrosis 24 h after CA4P therapy, which support further molecular design of probes based on scaffold H33258 for monitoring of tumor response to VDAs treatment.
Collapse
|
30
|
Wei Y, Gao L, Wang L, Shi L, Wei E, Zhou B, Zhou L, Ge B. Polydopamine and peptide decorated doxorubicin-loaded mesoporous silica nanoparticles as a targeted drug delivery system for bladder cancer therapy. Drug Deliv 2017; 24:681-691. [PMID: 28414557 PMCID: PMC8241003 DOI: 10.1080/10717544.2017.1309475] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
We reported a simple polydopamine (PDA)-based surface modification method to prepare novel targeted doxorubicin-loaded mesoporous silica nanoparticles and peptide CSNRDARRC conjugation (DOX-loaded MSNs@PDA-PEP) for enhancing the therapeutic effects on bladder cancer. Drug-loaded NPs were characterized in terms of size, size distribution, zeta potential, transmission electron microscopy (TEM), Brunauer-Emmett-Teller (BET) surface area and drug loading content. In vitro drug release indicated that DOX-loaded MSNs@PDA and MSNs@PDA-PEP had similar release kinetic profiles of DOX. The PDA coating well controlled DOX release and was highly sensitive to pH value. Confocal laser scanning microscopy (CLSM) showed that drug-loaded MSNs could be internalized by human bladder cancer cell line HT-1376, and DOX-loaded MSNs@PDA-PEP had the highest cellular uptake efficiency due to ligand-receptor recognition. The antitumor effects of DOX-loaded nanoparticles were evaluated by the MTT assay in vitro and by a xenograft tumor model in vivo, demonstrating that targeted nanocarriers DOX-loaded MSNs@PDA-PEP were significantly superior to free DOX and DOX-loaded MSNs@PDA. The novel DOX-loaded MSNs@PDA-PEP, which specifically recognized HT-1376 cells, can be used as a potential targeted drug delivery system for bladder cancer therapy.
Collapse
Affiliation(s)
- Yi Wei
- a Department of Urology , Affiliated Hospital of Guilin Medical University , Guilin , P.R. China
| | - Li Gao
- a Department of Urology , Affiliated Hospital of Guilin Medical University , Guilin , P.R. China
| | - Lu Wang
- b College of Biotechnology , and
| | - Lin Shi
- c Pharmaceutical College, Guilin Medical University , Guilin , P.R. China
| | - Erdong Wei
- a Department of Urology , Affiliated Hospital of Guilin Medical University , Guilin , P.R. China
| | - Baotong Zhou
- a Department of Urology , Affiliated Hospital of Guilin Medical University , Guilin , P.R. China
| | - Li Zhou
- a Department of Urology , Affiliated Hospital of Guilin Medical University , Guilin , P.R. China
| | - Bo Ge
- a Department of Urology , Affiliated Hospital of Guilin Medical University , Guilin , P.R. China
| |
Collapse
|
31
|
Xie B, Tomaszewski MR, Neves AA, Ros S, Hu DE, McGuire S, Mullins SR, Tice D, Sainson RCA, Bohndiek SE, Wilkinson RW, Brindle KM. Optoacoustic Detection of Early Therapy-Induced Tumor Cell Death Using a Targeted Imaging Agent. Clin Cancer Res 2017; 23:6893-6903. [PMID: 28821560 DOI: 10.1158/1078-0432.ccr-17-1029] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 07/01/2017] [Accepted: 08/11/2017] [Indexed: 11/16/2022]
Abstract
Purpose: The development of new treatments and their deployment in the clinic may be assisted by imaging methods that allow an early assessment of treatment response in individual patients. The C2A domain of Synaptotagmin-I (C2Am), which binds to the phosphatidylserine (PS) exposed by apoptotic and necrotic cells, has been developed as an imaging probe for detecting cell death. Multispectral optoacoustic tomography (MSOT) is a real-time and clinically applicable imaging modality that was used here with a near infrared (NIR) fluorophore-labeled C2Am to image tumor cell death in mice treated with a TNF-related apoptosis-inducing ligand receptor 2 (TRAILR2) agonist and with 5-fluorouracil (5-FU).Experimental Design: C2Am was labeled with a NIR fluorophore and injected intravenously into mice bearing human colorectal TRAIL-sensitive Colo205 and TRAIL-resistant HT-29 xenografts that had been treated with a potent agonist of TRAILR2 and in Colo205 tumors treated with 5-FU.Results: Three-dimensional (3D) MSOT images of probe distribution showed development of tumor contrast within 3 hours of probe administration and a signal-to-background ratio in regions containing dead cells of >10 after 24 hours. A site-directed mutant of C2Am that is inactive in PS binding showed negligible binding. Tumor retention of the active probe was strongly correlated (R2 = 0.97, P value < 0.01) with a marker of apoptotic cell death measured in histologic sections obtained post mortem.Conclusions: The rapid development of relatively high levels of contrast suggests that NIR fluorophore-labeled C2Am could be a useful optoacoustic imaging probe for detecting early therapy-induced tumor cell death in the clinic. Clin Cancer Res; 23(22); 6893-903. ©2017 AACR.
Collapse
Affiliation(s)
- Bangwen Xie
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - Michal R Tomaszewski
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - André A Neves
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - Susana Ros
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - De-En Hu
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - Sarah McGuire
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | | | | | | | - Sarah E Bohndiek
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | | | - Kevin M Brindle
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
32
|
An optical probe for detecting chondrocyte apoptosis in response to mechanical injury. Sci Rep 2017; 7:10906. [PMID: 28883614 PMCID: PMC5589871 DOI: 10.1038/s41598-017-10653-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 08/10/2017] [Indexed: 11/08/2022] Open
Abstract
Cartilage injury induced by acute excessive contact stress is common and mostly affects young adult. Although early detection of cartilage injury may prevent serious and lifelong arthritic complications, early detection and treatment is not possible due to the lack of a reliable detection method. Since chondrocyte injury and subsequent cell death are the early signs of cartilage injury, it is likely that cartilage cell apoptosis can be used to predict the extent of injury. To test this hypothesis, a near infrared probe was fabricated to have high affinity to apoptotic cells. In vitro tests show that this apoptosis probe has low toxicity, high specificity, and high affinity to apoptotic cells. In addition, there is a positive relationship between apoptotic cell numbers and fluorescence intensities. Using a mouse xiphoid injury model, we found significant accumulation of the apoptosis probes at the injured xiphoid cartilage site. There was also a positive correlation between probe accumulation and the number of apoptotic chondrocytes within the injured xiphoid cartilage, which was confirmed by TUNEL assay. The results support that the apoptosis probes may serve as a powerful tool to monitor the extent of mechanical force-induced cartilage injury in vivo.
Collapse
|
33
|
Savla R, Minko T. Nanoparticle design considerations for molecular imaging of apoptosis: Diagnostic, prognostic, and therapeutic value. Adv Drug Deliv Rev 2017; 113:122-140. [PMID: 27374457 DOI: 10.1016/j.addr.2016.06.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 06/21/2016] [Indexed: 12/13/2022]
Abstract
The present review analyzes various approaches for the design and synthesis of different nanoparticles for imaging and therapy. Nanoparticles for computed tomography (CT), magnetic resonance imaging (MRI), positron emission tomography (PET) and optical imaging are discussed. The influence of nanoparticle size, shape, surface charge, composition, surface functionalization, active targeting and other factors on imaging and therapeutic efficacy is analyzed. Cyto- and genotoxicity of nanoparticles are also discussed. Special attention in the review is paid to the imaging of apoptotic tissues and cells in different diseases.
Collapse
Affiliation(s)
- Ronak Savla
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, United States
| | - Tamara Minko
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, United States; Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, United States; Environmental and Occupational Health Sciences Institute, Piscataway, NJ 08854, United States.
| |
Collapse
|
34
|
Soudy R, Etayash H, Bahadorani K, Lavasanifar A, Kaur K. Breast Cancer Targeting Peptide Binds Keratin 1: A New Molecular Marker for Targeted Drug Delivery to Breast Cancer. Mol Pharm 2017; 14:593-604. [PMID: 28157321 DOI: 10.1021/acs.molpharmaceut.6b00652] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The biomarkers or receptors expressed on cancer cells and the targeting ligands with high binding affinity for biomarkers play a key role in early detection and treatment of breast cancer. The breast cancer targeting peptide p160 (12-mer) and its enzymatically stable analogue 18-4 (10-mer) showed marked potential for breast cancer drug delivery using cell studies and animal models. Herein, we used affinity purification, liquid chromatography-tandem mass spectrometry, and proteomics to identify keratin 1 (KRT1) as the target receptor highly expressed on breast cancer cells for p160 peptide(s). Western blot and immunocytochemistry in MCF-7 breast cancer cells confirmed the identity of KRT1. We demonstrate that the p160 or 18-4 binding to MCF-7 breast cancer cells is dependent on the expression of KRT1, and we confirm peptide-KRT1 binding specificity using SPR experiments (Kd ∼ 1.1 μM and 0.98 μM for p160 and 18-4, respectively). Furthermore, we assessed the ability of peptide 18-4 to improve the cellular uptake and anticancer activity of a pro-apoptotic antimicrobial peptide, microcin J25 (MccJ25), in breast cancer cells. A covalent conjugate of peptide 18-4 with MccJ25 showed preferential cytotoxicity toward breast cancer cells with minimal cytotoxicity against normal HUVEC cells. The conjugate inhibited the growth of MDA-MB-435 MDR multidrug-resistant cells with an IC50 comparable to that of nonresistant cells. Conjugation improved selective cellular uptake of MccJ25, and the conjugate triggered cancer cell death by apoptosis. Our findings establish KRT1 as a new marker for breast cancer targeting. Additionally, it pinpoints the potential use of antimicrobial lasso peptides as a novel class of anticancer therapeutics.
Collapse
Affiliation(s)
- Rania Soudy
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta , Edmonton, Alberta T6G 2E1, Canada.,Department of Medicine, University of Alberta , Edmonton, Alberta T6G 2B7, Canada.,Faculty of Pharmacy, Cairo University , Giza, Egypt
| | - Hashem Etayash
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta , Edmonton, Alberta T6G 2E1, Canada
| | - Kamran Bahadorani
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta , Edmonton, Alberta T6G 2E1, Canada
| | - Afsaneh Lavasanifar
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta , Edmonton, Alberta T6G 2E1, Canada.,Department of Chemical and Material Engineering, University of Alberta , Edmonton, Alberta T6G 2V4, Canada
| | - Kamaljit Kaur
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta , Edmonton, Alberta T6G 2E1, Canada.,Chapman University School of Pharmacy (CUSP), Harry and Diane Rinker Health Science Campus, Chapman University , Irvine, California 92618-1908, United States
| |
Collapse
|
35
|
Lee Y, Kim S, Kim D, Jon S. A histone H1-binding-aptide-based apoptosis imaging probe for monitoring tumor responses to cancer therapy. MEDCHEMCOMM 2017; 8:390-393. [PMID: 30108755 DOI: 10.1039/c6md00696e] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 12/16/2016] [Indexed: 11/21/2022]
Abstract
Here we report the feasibility of using a high-affinity histone H1-binding peptide as a potential molecular apoptosis imaging probe. The Cy5.5-labeled peptide showed high selectivity for drug-treated apoptotic tumors compared with non-apoptotic tumors in various in vivo settings, enabling detection of tumor responses to both docetaxel chemotherapy and Herceptin antibody therapy as early as 12 hours after treatment with a high signal-to-background ratio, even at extremely low doses.
Collapse
Affiliation(s)
- Yonghyun Lee
- KAIST Institute for the BioCentury , Department of Biological Sciences , Korea Advanced Institute of Science and Technology , 291 Daehak-ro , Daejeon 34141 , Republic of Korea
| | - Sunghyun Kim
- Korea Institute of Ceramic Engineering and Technology , Jinju 660-031 , Korea
| | - Daejin Kim
- KAIST Institute for the BioCentury , Department of Biological Sciences , Korea Advanced Institute of Science and Technology , 291 Daehak-ro , Daejeon 34141 , Republic of Korea
| | - Sangyong Jon
- KAIST Institute for the BioCentury , Department of Biological Sciences , Korea Advanced Institute of Science and Technology , 291 Daehak-ro , Daejeon 34141 , Republic of Korea
| |
Collapse
|
36
|
Perreault A, Richter S, Bergman C, Wuest M, Wuest F. Targeting Phosphatidylserine with a 64Cu-Labeled Peptide for Molecular Imaging of Apoptosis. Mol Pharm 2016; 13:3564-3577. [DOI: 10.1021/acs.molpharmaceut.6b00666] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Amanda Perreault
- Department of Oncology, Cross
Cancer Institute, University of Alberta, 11560 University Avenue, Edmonton, Alberta T6G 2X4, Canada
| | - Susan Richter
- Department of Oncology, Cross
Cancer Institute, University of Alberta, 11560 University Avenue, Edmonton, Alberta T6G 2X4, Canada
| | - Cody Bergman
- Department of Oncology, Cross
Cancer Institute, University of Alberta, 11560 University Avenue, Edmonton, Alberta T6G 2X4, Canada
| | - Melinda Wuest
- Department of Oncology, Cross
Cancer Institute, University of Alberta, 11560 University Avenue, Edmonton, Alberta T6G 2X4, Canada
| | - Frank Wuest
- Department of Oncology, Cross
Cancer Institute, University of Alberta, 11560 University Avenue, Edmonton, Alberta T6G 2X4, Canada
| |
Collapse
|
37
|
Probing amyloid beta-induced cell death using a fluorescence-peptide conjugate in Alzheimer's disease mouse model. Brain Res 2016; 1646:514-521. [DOI: 10.1016/j.brainres.2016.06.041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 05/03/2016] [Accepted: 06/28/2016] [Indexed: 12/18/2022]
|
38
|
Jung HK, Kim S, Park RW, Park JY, Kim IS, Lee B. Bladder tumor-targeted delivery of pro-apoptotic peptide for cancer therapy. J Control Release 2016; 235:259-267. [PMID: 27282414 DOI: 10.1016/j.jconrel.2016.06.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 05/09/2016] [Accepted: 06/03/2016] [Indexed: 11/25/2022]
Abstract
The overall prognosis of conventional chemotherapy for the treatment of bladder cancer is poor and reduction of its systemic side effects remains an unsolved issue. Targeted therapy for bladder cancer could improve therapeutic efficacy and reduce side effects. This study investigated a hybrid peptide (named Bld-1-KLA) composed of the CSNRDARRC peptide (Bld-1), which binds to bladder tumor cells, and the d-KLAKLAKKLAKLAK (KLA) peptide, which disrupts mitochondrial membrane and induces apoptotic cell death, as a bladder cancer-targeted therapeutic agent. Bld-1-KLA selectively bound to HT1376 bladder tumor cells and efficiently internalized into the cells but not to other types of tumor and normal cell lines. Bld-1-KLA exerted cytotoxic effects selectively to HT1376 cells (LC50=41.5μM), but not to other types of cells. Pretreatment of cells with Bld-1 inhibited the binding and cytotoxicity by Bld-1-KLA in HT1376 cells. It induced apoptosis of bladder tumor cells, while Bld-1 or KLA alone showed much lesser effect on apoptosis, and was co-localized in mitochondria. Bld-1-KLA was stable up to 24h in serum. In vivo fluorescence imaging showed that homing of Bld-1-KLA in the tumor in HT1376 tumor-bearing nude mice was greater than that of the control peptide-KLA after intravenous injection. Treatment of tumor-bearing mice with Bld-1-KLA, compared to the control peptide-KLA, induced apoptosis of tumor cells and inhibited tumor growth more efficiently. No significant side effects on body weight and the liver and myeloid function were observed in mice treated with Bld-1-KLA. These results suggest that Bld-1-KLA is a promising therapeutic agent for targeted therapy of bladder cancer.
Collapse
Affiliation(s)
- Hyun-Kyung Jung
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea; CMRI, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Soyoun Kim
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea; CMRI, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Rang-Woon Park
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea; CMRI, School of Medicine, Kyungpook National University, Daegu, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Jae-Yong Park
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, Republic of Korea; Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - In-San Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea; KU-KIST School, Korea University, Seoul, Republic of Korea
| | - Byungheon Lee
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea; CMRI, School of Medicine, Kyungpook National University, Daegu, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.
| |
Collapse
|
39
|
Abstract
PURPOSE Sodium [(18)F]fluoride (Na[(18)F]F) positron emission tomography with integrated computed tomography (PET/CT) has not been used for imaging myocardial infarction (MI). Here, we aimed to investigate the Na[(18)F]F PET/CT features of MI in a rat model. PROCEDURES MI was induced by coronary artery ligation in 8-week-old male Spraque-Dawley rats (300 ± 10 g) and confirmed by triphenyl tetrazolium chloride (TTC) staining. Na[(18)F]F PET/CT images were obtained using an animal-dedicated PET/CT scanner (NanoPET/CT, Mediso) in vivo and ex vivo. Uptake of Na[(18)F]F was quantitated using the standardized uptake value (SUV). Myocardial apoptosis was evaluated using histone-1 targeted peptide (ApoPep-1) and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining, while calcium accumulation was investigated using von Kossa's staining. Na[(18)F]F PET/CT was compared with (99m)Tc-methoxyisobutylisonitrile (MIBI) or (99m)Tc-hydroxymethylenediphosphonate (HMDP) single photon emission computed tomography/computed tomography (SPECT/CT) in rats with day 1 MI. RESULTS The rats showed strong Na[(18)F]F uptake both in vivo and ex vivo; the maximal uptake occurred 1 day after MI (SUV ratio of infarct to lung = 4.56 ± 0.74, n = 7, P = 0.0183 vs the control). The Na[(18)F]F uptake area perfectly matched the apoptotic area, determined by ApoPep-1 uptake and TUNEL assay. However, calcification, assessed by von Kossa's staining, was absent in the infarct. Na[(18)F]F PET/CT showed an increased uptake at the perfusion deficit area in [(99m)Tc]MIBI SPECT/CT and an equivalent signal to [(99m)Tc]HMDP SPECT/CT in rats with day 1 MI. CONCLUSIONS Na[(18)F]F PET/CT is a promising hot-spot imaging modality for MI.
Collapse
Affiliation(s)
- Jeong Hee Han
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, 173-82 Gumi-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 463-707, Republic of Korea
| | | | | | | |
Collapse
|
40
|
Che X, Chi L, Park CY, Cho GH, Park N, Kim SG, Lee BH, Choi JY. A novel method to detect articular chondrocyte death during early stages of osteoarthritis using a non-invasive ApoPep-1 probe. Arthritis Res Ther 2015; 17:309. [PMID: 26530111 PMCID: PMC4632461 DOI: 10.1186/s13075-015-0832-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 10/21/2015] [Indexed: 11/19/2022] Open
Abstract
Introduction Current methods for early diagnosis of osteoarthritis (OA) are limited. We assessed whether in vivo detection of chondrocyte death by ApoPep-1 (CQRPPR), a peptide that binds to histone H1 of apoptotic and necrotic cells, could be used to detect the initiation of OA. Methods Apoptosis-induced ATDC5 cells were labeled with Annexin V and ApoPep-1. Surgical destabilization of the medial meniscus (DMM) was performed on both knees of 12-week-old male mice and severity of OA was determined by histological analysis according to the Osteoarthritis Research Society International (OARSI) guidelines. At 1, 2, 4, and 8 weeks post-surgery, mice were intravenously injected with fluorescence-labeled ApoPep-1 or control peptide and in vivo imaging was performed within 30 minutes of injection by near-infrared fluorescence (NIRF). Binding of ApoPep-1 to OA joints was demonstrated by ex vivo imaging and immunofluorescent staining using TUNEL and histone H1 and type II collagen antibodies. Results Strong signals of ApoPep-1 were observed on the apoptotic ATDC5 cells. Knees corresponded to grade II, III, and V OA at 2, 4, and 8 weeks after DMM, respectively. Between 2 and 8 weeks after surgery, the in vivo NIRF signal at OA-ApoPep1-injected joints was consistently stronger than sham-operated or OA-control peptide-injected joints. ApoPep-1, TUNEL, and histone H1 signals were stronger in grade II OA cartilage than sham-operated cartilage when detected by immunofluorescent staining. Type II collagen expression was similar between grade II OA and sham group. Conclusion ApoPep-1 can be used to detect OA in vivo by binding to apoptotic chondrocytes. This is a novel, sensitive, and rapid method which can detect apoptotic cells in OA rodent models soon after its onset.
Collapse
Affiliation(s)
- Xiangguo Che
- Department of Biochemistry and Cell Biology, Kyungpook National University School of Medicine, Daegu, 700-422, Republic of Korea.
| | - Lianhua Chi
- Department of Biochemistry and Cell Biology, Kyungpook National University School of Medicine, Daegu, 700-422, Republic of Korea.
| | - Clara Yongjoo Park
- Department of Biochemistry and Cell Biology, Kyungpook National University School of Medicine, Daegu, 700-422, Republic of Korea.
| | - Gyoung-Ho Cho
- Department of Biochemistry and Cell Biology, Kyungpook National University School of Medicine, Daegu, 700-422, Republic of Korea.
| | - Narae Park
- Department of Biochemistry and Cell Biology, Kyungpook National University School of Medicine, Daegu, 700-422, Republic of Korea.
| | - Seong-Gon Kim
- Department of Oral and Maxillofacial Surgery, College of Dentistry, Gangneung-Wonju National University, Gangneung, 210-702, Republic of Korea.
| | - Byung-Heon Lee
- Department of Biochemistry and Cell Biology, Kyungpook National University School of Medicine, Daegu, 700-422, Republic of Korea.
| | - Je-Yong Choi
- Department of Biochemistry and Cell Biology, Kyungpook National University School of Medicine, Daegu, 700-422, Republic of Korea.
| |
Collapse
|
41
|
Kwak W, Ha YS, Soni N, Lee W, Park SI, Ahn H, An GI, Kim IS, Lee BH, Yoo J. Apoptosis imaging studies in various animal models using radio-iodinated peptide. Apoptosis 2015; 20:110-21. [PMID: 25430587 DOI: 10.1007/s10495-014-1059-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Apoptosis has a role in many medical disorders and treatments; hence, its non-invasive evaluation is one of the most riveting research topics. Currently annexin V is used as gold standard for imaging apoptosis. However, several drawbacks, including high background, slow body clearance, make it a suboptimum marker for apoptosis imaging. In this study, we radiolabeled the recently identified histone H1 targeting peptide (ApoPep-1) and evaluated its potential as a new apoptosis imaging agent in various animal models. ApoPep-1 (CQRPPR) was synthesized, and an extra tyrosine residue was added to its N-terminal end for radiolabeling. This peptide was radiolabeled with (124)I and (131)I and was tested for its serum stability. Surgery- and drug-induced apoptotic rat models were prepared for apoptosis evaluation, and PET imaging was performed. Doxorubicin was used for xenograft tumor treatment in mice, and the induced apoptosis was studied. Tumor metabolism and proliferation were assessed by [(18)F]FDG and [(18)F]FLT PET imaging and compared with ApoPep-1 after doxorubicin treatment. The peptide was radiolabeled at high purity, and it showed reasonably good stability in serum. Cell death was easily imaged by radiolabeled ApoPep-1 in an ischemia surgery model. And, liver apoptosis was more clearly identified by ApoPep-1 rather than [(124)I]annexin V in cycloheximide-treated models. Three doxorubicin doses inhibited tumor growth, which was evaluated by 30-40% decreases of [(18)F]FDG and [(18)F]FLT PET uptake in the tumor area. However, ApoPep-1 demonstrated more than 200% increase in tumor uptake after chemotherapy, while annexin V did not show any meaningful uptake in the tumor compared with the background. Biodistribution data were also in good agreement with the microPET imaging results. All of the experimental data clearly demonstrated high potential of the radiolabeled ApoPep-1 for in vivo apoptosis imaging.
Collapse
Affiliation(s)
- Wonjung Kwak
- Department of Molecular Medicine, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Zeng W, Wang X, Xu P, Liu G, Eden HS, Chen X. Molecular imaging of apoptosis: from micro to macro. Theranostics 2015; 5:559-82. [PMID: 25825597 PMCID: PMC4377726 DOI: 10.7150/thno.11548] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 02/18/2015] [Indexed: 12/21/2022] Open
Abstract
Apoptosis, or programmed cell death, is involved in numerous human conditions including neurodegenerative diseases, ischemic damage, autoimmune disorders and many types of cancer, and is often confused with other types of cell death. Therefore strategies that enable visualized detection of apoptosis would be of enormous benefit in the clinic for diagnosis, patient management, and development of new therapies. In recent years, improved understanding of the apoptotic machinery and progress in imaging modalities have provided opportunities for researchers to formulate microscopic and macroscopic imaging strategies based on well-defined molecular markers and/or physiological features. Correspondingly, a large collection of apoptosis imaging probes and approaches have been documented in preclinical and clinical studies. In this review, we mainly discuss microscopic imaging assays and macroscopic imaging probes, ranging in complexity from simple attachments of reporter moieties to proteins that interact with apoptotic biomarkers, to rationally designed probes that target biochemical changes. Their clinical translation will also be our focus.
Collapse
|
43
|
Morales DP, Braun GB, Pallaoro A, Chen R, Huang X, Zasadzinski JA, Reich NO. Targeted intracellular delivery of proteins with spatial and temporal control. Mol Pharm 2014; 12:600-9. [PMID: 25490248 PMCID: PMC4319691 DOI: 10.1021/mp500675p] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
![]()
While
a host of methods exist to deliver genetic materials or small
molecules to cells, very few are available for protein delivery to
the cytosol. We describe a modular, light-activated nanocarrier that
transports proteins into cells by receptor-mediated endocytosis and
delivers the cargo to the cytosol by light triggered endosomal escape.
The platform is based on hollow gold nanoshells (HGN) with polyhistidine
tagged proteins attached through an avidity-enhanced, nickel chelation
linking layer; here, we used green fluorescent protein (GFP) as a
model deliverable cargo. Endosomal uptake of the GFP loaded nanocarrier
was mediated by a C-end Rule (CendR) internalizing peptide fused to
the GFP. Focused femtosecond pulsed-laser excitation triggered protein
release from the nanocarrier and endosome disruption, and the released
protein was capable of targeting the nucleoli, a model intracellular
organelle. We further demonstrate the generality of the approach by
loading and releasing Sox2 and p53. This method for targeting of individual
cells, with resolution similar to microinjection, provides spatial
and temporal control over protein delivery.
Collapse
Affiliation(s)
- Demosthenes P Morales
- Department of Chemistry and Biochemistry, University of California , Santa Barbara, California 93106, United States
| | | | | | | | | | | | | |
Collapse
|
44
|
Jung KO, Youn H, Kim YH, Kim S, Na J, Kim YI, Park JW, Kang KW, Lee DS, Chung JK. Relationship between apoptosis imaging and radioiodine therapy in tumor cells with different sodium iodide symporter gene expression. Mol Imaging 2014; 13. [PMID: 25431214 DOI: 10.2310/7290.2014.00050] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The therapeutic efficacy of radioiodine (¹³¹I) therapy has been reported to be variable among cancer patients and even between metastatic regions in the same patients. Because the expression level of sodium iodide symporter (NIS) cannot reflect the efficacy of therapy, other strategies are required to predict the precise therapeutic effect of ¹³¹I therapy. In this research, we investigated the correlation between iodine (I) uptake, apoptosis imaging, and therapeutic efficacy. Two HT29 cell lines, cytomegalovirus (CMV)-NIS (or NIS+++) and TERT-NIS (or NIS+), were established by retroviral transfection. I uptake was estimated by I-uptake assay and gamma camera imaging. Apoptosis was evaluated by confocal microscopy and a Maestro fluorescence imaging system (CRi Inc., Woburn, MA) using ApoFlamma (BioACTs, Seoul, Korea), a fluorescent dye-conjugated apoptosis-targeting peptide 1 (ApoPep-1). Therapeutic efficacy was determined by tumor size. The CMV-NIS showed higher I uptake and ApoFlamma signals than TERT-NIS. In xenograft models, CMV-NIS also showed high 99m technetium signals and ApoFlamma signals. Tumor reduction had a stronger correlation with apoptosis imaging signals than with gamma camera imaging signals, which reflect I uptake. Higher NIS-expressing tumors showed increased apoptosis and I uptake, resulting in a significant tumor reduction. Moreover, tumor reduction showed a strong correlation with ApoFlamma imaging compared to I-uptake imaging.
Collapse
|
45
|
Kim S, Kim D, Lee Y, Jeon H, Lee BH, Jon S. Conversion of Low-Affinity Peptides to High-Affinity Peptide Binders by Using a β-Hairpin Scaffold-Assisted Approach. Chembiochem 2014; 16:43-6. [DOI: 10.1002/cbic.201402450] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2014] [Indexed: 12/27/2022]
|
46
|
Lee SK, Mortensen LJ, Lin CP, Tung CH. An authentic imaging probe to track cell fate from beginning to end. Nat Commun 2014; 5:5216. [PMID: 25323442 PMCID: PMC4852472 DOI: 10.1038/ncomms6216] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 09/09/2014] [Indexed: 12/18/2022] Open
Abstract
Accurate tracing of cell viability is critical for optimizing delivery methods and evaluating the efficacy and safety of cell therapeutics. A nanoparticle-based cell tracker is developed to image cell fate from live to dead. The particle is fabricated from two types of optically quenched polyelectrolytes, a life indicator and a death indicator, through electrostatic interactions. On incubation with cells, the fabricated bifunctional nanoprobes are taken up efficiently and the first colour is produced by normal intracellular proteolysis, reflecting the healthy status of the cells. Depending on the number of coated layers, the signal can persist for several replication cycles. However, as the cells begin dying, the second colour appears quickly to reflect the new cell status. Using this chameleon-like cell tracker, live cells can be distinguished from apoptotic and necrotic cells instantly and definitively.
Collapse
Affiliation(s)
- Seung Koo Lee
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medical College, 413 East 69th Street, Box 290, New York, New York 10021, USA
| | - Luke J Mortensen
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Charles P Lin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Ching-Hsuan Tung
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medical College, 413 East 69th Street, Box 290, New York, New York 10021, USA
| |
Collapse
|
47
|
Padmanaban G, Park H, Choi JS, Cho YW, Kang WC, Moon CI, Kim IS, Lee BH. Identification of peptides that selectively bind to myoglobin by biopanning of phage displayed-peptide library. J Biotechnol 2014; 187:43-50. [DOI: 10.1016/j.jbiotec.2014.07.435] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2014] [Revised: 06/28/2014] [Accepted: 07/18/2014] [Indexed: 11/26/2022]
|
48
|
Jung HK, Wang K, Jung MK, Kim IS, Lee BH. In vivo near-infrared fluorescence imaging of apoptosis using histone H1-targeting peptide probe after anti-cancer treatment with cisplatin and cetuximab for early decision on tumor response. PLoS One 2014; 9:e100341. [PMID: 24949860 PMCID: PMC4065102 DOI: 10.1371/journal.pone.0100341] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 05/23/2014] [Indexed: 12/29/2022] Open
Abstract
Early decision on tumor response after anti-cancer treatment is still an unmet medical need. Here we investigated whether in vivo imaging of apoptosis using linear and cyclic (disulfide-bonded) form of ApoPep-1, a peptide that recognizes histone H1 exposed on apoptotic cells, at an early stage after treatment could predict tumor response to the treatment later. Treatment of stomach tumor cells with cistplatin or cetuximab alone induced apoptosis, while combination of cisplatin plus cetuximab more efficiently induced apoptosis, as detected by binding with linear and cyclic form of ApoPep-1. However, the differences between the single agent and combination treatment were more remarkable as detected with the cyclic form compared to the linear form. In tumor-bearing mice, apoptosis imaging was performed 1 week and 2 weeks after the initiation of treatment, while tumor volumes and weights were measured 3 weeks after the treatment. In vivo fluorescence imaging signals obtained by the uptake of ApoPep-1 to tumor was most remarkable in the group injected with cyclic form of ApoPep-1 at 1 week after combined treatment with cisplatin plus cetuximab. Correlation analysis revealed that imaging signals by cyclic ApoPep-1 at 1 week after treatment with cisplatin plus cetuximab in combination were most closely related with tumor volume changes (r2 = 0.934). These results demonstrate that in vivo apoptosis imaging using Apopep-1, especially cyclic ApoPep-1, is a sensitive and predictive tool for early decision on stomach tumor response after anti-cancer treatment.
Collapse
Affiliation(s)
- Hyun-Kyung Jung
- Department of Biochemistry and Cell Biology and School of Medicine, Kyungpook National University, Daegu, Korea
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu, Korea
| | - Kai Wang
- Department of Plastic Surgery, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Min Kyu Jung
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Korea
| | - In-San Kim
- Department of Biochemistry and Cell Biology and School of Medicine, Kyungpook National University, Daegu, Korea
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu, Korea
| | - Byung-Heon Lee
- Department of Biochemistry and Cell Biology and School of Medicine, Kyungpook National University, Daegu, Korea
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu, Korea
- * E-mail:
| |
Collapse
|
49
|
Chen H, Chen Y, Yang H, Xu W, Zhang M, Ma Y, Achilefu S, Gu Y. A dual-targeting nanocarrier based on modified chitosan micelles for tumor imaging and therapy. Polym Chem 2014. [DOI: 10.1039/c4py00495g] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
50
|
Ye D, Shuhendler AJ, Cui L, Tong L, Tee SS, Tikhomirov G, Felsher DW, Rao J. Bioorthogonal cyclization-mediated in situ self-assembly of small-molecule probes for imaging caspase activity in vivo. Nat Chem 2014; 6:519-26. [PMID: 24848238 PMCID: PMC4031611 DOI: 10.1038/nchem.1920] [Citation(s) in RCA: 352] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 03/13/2014] [Indexed: 02/06/2023]
Abstract
Directed self-assembly of small molecules in living systems could enable a myriad of applications in biology and medicine, and it has been widely used to synthesize supramolecules and nano/microstructures in solution and in living cells. However, controlling self-assembly of synthetic small molecules in living animals is challenging because of the complex and dynamic in vivo physiological environment. Here we employed an optimized first-order bioorthogonal cyclization reaction to control self-assembly of a fluorescent small molecule, and demonstrated its in vivo applicability by imaging of casapae-3/7 activity in human tumor xenograft mouse models of chemotherapy. The in situ assembled fluorescent nanoparticles have been successfully imaged in both apoptotic cells and tumor tissues using three-dimensional structured illumination microscopy. This strategy combines the advantages offered by small molecules with those of nanomaterials and should find widespread use for non-invasive imaging of enzyme activity in vivo.
Collapse
Affiliation(s)
- Deju Ye
- 1] Molecular Imaging Program at Stanford, Departments of Radiology and Chemistry, Stanford University, Stanford, California 94305-5484, USA [2]
| | - Adam J Shuhendler
- 1] Molecular Imaging Program at Stanford, Departments of Radiology and Chemistry, Stanford University, Stanford, California 94305-5484, USA [2]
| | - Lina Cui
- Molecular Imaging Program at Stanford, Departments of Radiology and Chemistry, Stanford University, Stanford, California 94305-5484, USA
| | - Ling Tong
- Departments of Medicine and Pathology, Division of Oncology, School of Medicine Stanford University, Stanford, California, 94305-5151, USA
| | - Sui Seng Tee
- Molecular Imaging Program at Stanford, Departments of Radiology and Chemistry, Stanford University, Stanford, California 94305-5484, USA
| | - Grigory Tikhomirov
- Molecular Imaging Program at Stanford, Departments of Radiology and Chemistry, Stanford University, Stanford, California 94305-5484, USA
| | - Dean W Felsher
- Departments of Medicine and Pathology, Division of Oncology, School of Medicine Stanford University, Stanford, California, 94305-5151, USA
| | - Jianghong Rao
- Molecular Imaging Program at Stanford, Departments of Radiology and Chemistry, Stanford University, Stanford, California 94305-5484, USA
| |
Collapse
|