1
|
Jiang Y, Li Y, Fu X, Wu Y, Wang R, Zhao M, Mao C, Shi S. Interplay between G protein-coupled receptors and nanotechnology. Acta Biomater 2023; 169:1-18. [PMID: 37517621 DOI: 10.1016/j.actbio.2023.07.049] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 06/15/2023] [Accepted: 07/25/2023] [Indexed: 08/01/2023]
Abstract
G protein-coupled receptors (GPCRs), as the largest family of membrane receptors, actively modulate plasma membrane and endosomal signalling. Importantly, GPCRs are naturally nanosized, and spontaneously formed nanoaggregates of GPCRs (natural nano-GPCRs) may enhance GPCR-related signalling and functions. Although GPCRs are the molecular targets of the majority of marketed drugs, the poor pharmacokinetics and physicochemical properties of GPCR ligands greatly limit their clinical applicability. Nanotechnology, as versatile techniques, can encapsulate GPCR ligands to assemble synthetic nano-GPCRs to overcome their obstacles, robustly elevating drug efficacy and safety. Moreover, endosomal delivery of GPCR ligands by nanoparticles can precisely initiate sustained endosomal signal transduction, while nanotechnology has been widely utilized for isolation, diagnosis, and detection of GPCRs. In turn, due to overexpression of GPCRs on the surface of various types of cells, GPCR ligands can endow nanoparticles with active targeting capacity for specific cells via ligand-receptor binding and mediate receptor-dependent endocytosis of nanoparticles. This significantly enhances the potency of nanoparticle delivery systems. Therefore, emerging evidence has revealed the interplay between GPCRs and nanoparticles, although investigations into their relationship have been inadequate. This review aims to summarize the interaction between GPCRs and nanotechnology for understanding their mutual influences and utilizing their interplay for biomedical applications. It will provide a fundamental platform for developing powerful and safe GPCR-targeted drugs and nanoparticle systems. STATEMENT OF SIGNIFICANCE: GPCRs as molecular targets for the majority of marketed drugs are naturally nanosized, and even spontaneously form nano aggregations (nano-GPCRs). Nanotechnology has also been applied to construct synthetic nano-GPCRs or detect GPCRs, while endosomal delivery of GPCR ligands by nanoparticles can magnify endosomal signalling. Meanwhile, molecular engineering of nanoparticles with GPCRs or their ligands can modulate membrane binding and endocytosis, powerfully improving the efficacy of nanoparticle system. However, there are rare summaries on the interaction between GPCRs and nanoparticles. This review will not only provide a versatile platform for utilizing nanoparticles to modulate or detect GPCRs, but also facilitate better understanding of the designated value of GPCRs for molecular engineering of biomaterials with GPCRs in therapeutical application.
Collapse
Affiliation(s)
- Yuhong Jiang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China.
| | - Yuke Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xiujuan Fu
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Yue Wu
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Rujing Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Mengnan Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Canquan Mao
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Sanjun Shi
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
2
|
Pranav U, Malhotra M, Pathan S, Jayakannan M. Structural Engineering of Star Block Biodegradable Polymer Unimolecular Micelles for Drug Delivery in Cancer Cells. ACS Biomater Sci Eng 2023; 9:743-759. [PMID: 36579913 DOI: 10.1021/acsbiomaterials.2c01201] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The present investigation reports the structural engineering of biodegradable star block polycaprolactone (PCL) to tailor-make aggregated micelles and unimolecular micelles to study their effect on drug delivery aspects in cancer cell lines. Fully PCL-based star block copolymers were designed by varying the arm numbers from two to eight while keeping the arm length constant throughout. Multifunctional initiators were exploited for stepwise solvent-free melt ring-opening polymerization of ε-caprolactone and γ-substituted caprolactone to construct star block copolymers having a PCL hydrophobic core and a carboxylic PCL hydrophilic shell, respectively. A higher arm number and a higher degree of branching in star polymers facilitated the formation of unimolecular micelles as opposed to the formation of conventional multimicellar aggregates in lower arm analogues. The dense core of the unimolecular micelles enabled them to load high amounts of the anticancer drug doxorubicin (DOX, ∼12-15%) compared to the aggregated micelles (∼3-4%). The star unimolecular micelle completely degraded leading to 90% release of the loaded drug upon treatment with the lysosomal esterase enzyme in vitro. The anticancer efficacies of these DOX-loaded unimolecular micelles were tested in a breast cancer cell line (MCF-7), and their IC50 values were found to be much lower compared to those of aggregated micelles. Time-dependent cellular uptake studies by confocal microscopy revealed that unimolecular micelles were readily taken up by the cells, and enhancement of the drug concentration was observed at the intracellular level up to 36 h. The present work opens new synthetic strategies for building a next-generation biodegradable unimolecular micellar nanoplatform for drug delivery in cancer research.
Collapse
Affiliation(s)
- Upendiran Pranav
- Department of Chemistry, Indian Institute of Science Education and Research (IISER Pune), Dr. Homi Bhabha Road, Pune 411008 Maharashtra, India
| | - Mehak Malhotra
- Department of Chemistry, Indian Institute of Science Education and Research (IISER Pune), Dr. Homi Bhabha Road, Pune 411008 Maharashtra, India
| | - Shahidkhan Pathan
- Department of Chemistry, Indian Institute of Science Education and Research (IISER Pune), Dr. Homi Bhabha Road, Pune 411008 Maharashtra, India
| | - Manickam Jayakannan
- Department of Chemistry, Indian Institute of Science Education and Research (IISER Pune), Dr. Homi Bhabha Road, Pune 411008 Maharashtra, India
| |
Collapse
|
3
|
Bhattacharya S, Majumdar Nee Paul S. Application of conventional metallic nanoparticles on male reproductive system - challenges and countermeasures. Syst Biol Reprod Med 2023; 69:32-49. [PMID: 36427189 DOI: 10.1080/19396368.2022.2140087] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The application of nanotechnology in the present era has substantial impact on different industrial and medical fields. However, the advancement in nanotechnology for potential therapeutic and consumer benefits has been an anxious cause regarding the probable hazardous consequences of these molecules in biological systems and the environment. The toxic effects can perturb the physiologic system broadly and reproductive function and fertility specifically. Despite engineered nanomaterials (ENMs) having a wide range of applications, toxicological investigations of the probable ramifications of ENMs on the reproductive systems of mammals and fertility remains in its nascence. Complication in the male reproductive system is quite a pertinent issue in today's world which comprises of benign prostatic enlargement, prostate cancer, and unhealthy sperm production. The therapeutic drugs should not only be active in minimum dose but also site-specific in action, criteria being met by nanomedicines. Nanomedicine therapy is promising but encompasses the chances of adverse effects of being cytotoxic and generating oxidative stress. These hurdles can be overcome by creating coated nanoparticles with organic substances, modification of shape and size, and synthesizing biocompatible green nanoparticles. This review attempts to look into the applications of most widely used metals like zinc, titanium, silver, and gold nanoparticles in the therapy of the male reproductive system, their prospective harmful effects, and the way out to create a safe therapeutic system by specific modifications of these metal and metal oxide nanoparticles.
Collapse
Affiliation(s)
- Sonali Bhattacharya
- Department of Zoology (Post Graduate Studies), Rishi Bankim Chandra College, West Bengal State University, Naihati, West Bengal, India
| | - Sudipta Majumdar Nee Paul
- Department of Zoology (Post Graduate Studies), Rishi Bankim Chandra College, West Bengal State University, Naihati, West Bengal, India
| |
Collapse
|
4
|
Li X, Guo T, Feng Q, Bai T, Wu L, Liu Y, Zheng X, Jia J, Pei J, Wu S, Song Y, Zhang Y. Progress of thrombus formation and research on the structure-activity relationship for antithrombotic drugs. Eur J Med Chem 2022; 228:114035. [PMID: 34902735 DOI: 10.1016/j.ejmech.2021.114035] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 11/11/2021] [Accepted: 11/30/2021] [Indexed: 01/07/2023]
Abstract
Many populations suffer from thrombotic disorders such as stroke, myocardial infarction, unstable angina and thromboembolic disease. Thrombus is one of the major threatening factors to human health and the prevalence of cardio-cerebrovascular diseases induced by thrombus is growing worldwide, even some persons got rare and severe blood clots after receiving the AstraZeneca COVID vaccine unexpectedly. In terms of mechanism of thrombosis, antithrombotic drugs have been divided into three categories including anticoagulants, platelet inhibitors and fibrinolytics. Nowadays, a large number of new compounds possessing antithrombotic activities are emerging in an effort to remove the inevitable drawbacks of previously approved drugs such as the high risk of bleeding, a slow onset of action and a narrow therapeutic window. In this review, we describe the causes and mechanisms of thrombus formation firstly, and then summarize these reported active compounds as potential antithrombotic candidates based on their respective mechanism, hoping to promote the development of more effective bioactive molecules for treating thrombotic disorders.
Collapse
Affiliation(s)
- Xiaoan Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Biomedicine Key Laboratory of Shaanxi Province, Northwest University, Xi'an, 710069, China; Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Tiantian Guo
- College of Food Science and Technology, Northwest University, Xi'an, 710069, China
| | - Qian Feng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Biomedicine Key Laboratory of Shaanxi Province, Northwest University, Xi'an, 710069, China
| | - Tiantian Bai
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Biomedicine Key Laboratory of Shaanxi Province, Northwest University, Xi'an, 710069, China
| | - Lei Wu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Biomedicine Key Laboratory of Shaanxi Province, Northwest University, Xi'an, 710069, China
| | - Yubo Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Biomedicine Key Laboratory of Shaanxi Province, Northwest University, Xi'an, 710069, China
| | - Xu Zheng
- Shaanxi Institute for Food and Drug, Xi'an, 710000, China
| | - Jianzhong Jia
- Shaanxi Institute for Food and Drug, Xi'an, 710000, China
| | - Jin Pei
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Shaoping Wu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Biomedicine Key Laboratory of Shaanxi Province, Northwest University, Xi'an, 710069, China.
| | - Yiming Song
- School of Chemical Engineering, Northwest University, 229 Taibai Road, Xi'an, Shaanxi, 710069, China.
| | - Yongmin Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Biomedicine Key Laboratory of Shaanxi Province, Northwest University, Xi'an, 710069, China; Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, UMR 8232, 4 Place Jussieu, 75005, Paris, France
| |
Collapse
|
5
|
Mehrizi TZ, Kafiabad SA, Eshghi P. Effects and treatment applications of polymeric nanoparticles on improving platelets' storage time: a review of the literature from 2010 to 2020. Blood Res 2021; 56:215-228. [PMID: 34880140 PMCID: PMC8721452 DOI: 10.5045/br.2021.2021094] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 07/26/2021] [Accepted: 08/25/2021] [Indexed: 12/13/2022] Open
Abstract
Maintaining the quality of platelet products and increasing their storage time are priorities for treatment applications. The formation of platelet storage lesions that limit the storage period and preservation temperature, which can prepare a decent environment for bacterial growth, are the most important challenges that researchers are dealing with in platelet preservation. Nanotechnology is an emerging field of science that has introduced novel solutions to resolve these problems. Here, we reviewed the reported effects of polymeric nanoparticles-including chitosan, dendrimers, polyethylene glycol (PEG), and liposome-on platelets in articles from 2010 to 2020. As a result, we concluded that the presence of dendrimer nanoparticles with a smaller size, negative charge, low molecular weight, and low concentration along with PEGylation can increase the stability and survival of platelets during storage. In addition, PEGylation of platelets can also be a promising approach to improve the quality of platelet bags during storage.
Collapse
Affiliation(s)
- Tahereh Zadeh Mehrizi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Sedigheh Amini Kafiabad
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Peyman Eshghi
- Pediatric Congenital Hematologic Disorders Research Center, Shahid Beheshti University of Medical Sciences and Iran Blood Transfusion Organization, Tehran, Iran
| |
Collapse
|
6
|
Abstract
As basic research into GPCR signaling and its association with disease has come into fruition, greater clarity has emerged with regards to how these receptors may be amenable to therapeutic intervention. As a diverse group of receptor proteins, which regulate a variety of intracellular signaling pathways, research in this area has been slow to yield tangible therapeutic agents for the treatment of a number of diseases including cancer. However, recently such research has gained momentum based on a series of studies that have sought to define GPCR proteins dynamics through the elucidation of their crystal structures. In this chapter, we define the approaches that have been adopted in developing better therapeutics directed against the specific parts of the receptor proteins, such as the extracellular and the intracellular domains, including the ligands and auxiliary proteins that bind them. Finally, we also briefly outline how GPCR-derived signaling transduction pathways hold great potential as additional targets.
Collapse
Affiliation(s)
- Surinder M Soond
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russian Federation.
| | - Andrey A Zamyatnin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russian Federation; Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russian Federation.
| |
Collapse
|
7
|
Tambe P, Kumar P, Paknikar KM, Gajbhiye V. Smart triblock dendritic unimolecular micelles as pioneering nanomaterials: Advancement pertaining to architecture and biomedical applications. J Control Release 2019; 299:64-89. [PMID: 30797002 DOI: 10.1016/j.jconrel.2019.02.026] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 02/19/2019] [Accepted: 02/19/2019] [Indexed: 11/08/2022]
|
8
|
PEGylation: a promising strategy to overcome challenges to cancer-targeted nanomedicines: a review of challenges to clinical transition and promising resolution. Drug Deliv Transl Res 2019; 9:721-734. [DOI: 10.1007/s13346-019-00631-4] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
9
|
Ma X, Xiong Y, Lee LTO. Application of Nanoparticles for Targeting G Protein-Coupled Receptors. Int J Mol Sci 2018; 19:E2006. [PMID: 29996469 PMCID: PMC6073629 DOI: 10.3390/ijms19072006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 06/28/2018] [Accepted: 07/04/2018] [Indexed: 01/01/2023] Open
Abstract
Nanoparticles (NPs) have attracted unequivocal attention in recent years due to their potential applications in therapeutics, bio-imaging and material sciences. For drug delivery, NP-based carrier systems offer several advantages over conventional methods. When conjugated with ligands and drugs (or other therapeutic molecules), administrated NPs are able to deliver cargo to targeted sites through ligand-receptor recognition. Such targeted delivery is especially important in cancer therapy. Through this targeted cancer nanotherapy, cancer cells are killed with higher specificity, while the healthy cells are spared. Furthermore, NP drug delivery leads to improved drug load, enhanced drug solubility and stability, and controlled drug release. G protein-coupled receptors (GPCRs) are a superfamily of cell transmembrane receptors. They regulate a plethora of physiological processes through ligand-receptor-binding-induced signaling transduction. With recent evidence unveiling their roles in cancer, GPCR agonists and antagonists have quickly become new targets in cancer therapy. This review focuses on the application of some notable nanomaterials, such as dendrimers, quantum dots, gold nanoparticles, and magnetic nanoparticles, in GPCR-related cancers.
Collapse
Affiliation(s)
- Xin Ma
- Centre of Reproduction Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau, China.
| | - Yunfang Xiong
- Centre of Reproduction Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau, China.
| | - Leo Tsz On Lee
- Centre of Reproduction Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau, China.
| |
Collapse
|
10
|
Matus MF, Vilos C, Cisterna BA, Fuentes E, Palomo I. Nanotechnology and primary hemostasis: Differential effects of nanoparticles on platelet responses. Vascul Pharmacol 2018; 101:1-8. [DOI: 10.1016/j.vph.2017.11.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 10/12/2017] [Accepted: 11/14/2017] [Indexed: 12/19/2022]
|
11
|
Elkin I, Banquy X, Barrett CJ, Hildgen P. Non-covalent formulation of active principles with dendrimers: Current state-of-the-art and prospects for further development. J Control Release 2017; 264:288-305. [PMID: 28887136 DOI: 10.1016/j.jconrel.2017.09.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 08/28/2017] [Accepted: 09/01/2017] [Indexed: 12/18/2022]
|
12
|
Heo JY, Kang SH, Kim YH, You S, Jin KS, Kim SW, Jung HY, Jung KO, Lee CH, Kim MJ, Sung SE, Kim B, Choi IS, Youn H, Chung JK, Kim SK, Kim Y. Toward redesigning the PEG surface of nanocarriers for tumor targeting: impact of inner functionalities on size, charge, multivalent binding, and biodistribution. Chem Sci 2017; 8:5186-5195. [PMID: 28970905 PMCID: PMC5618790 DOI: 10.1039/c6sc05640g] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 04/17/2017] [Indexed: 01/15/2023] Open
Abstract
Achieving accurate and efficacious tumor targeting with minimal off-target effects is of paramount importance in designing diagnostic and therapeutic agents for cancer. In this respect, nanocarriers have gained enormous popularity because of their attainable multifunctional features, as well as tumor-targeting potential by extravasation. However, once administered into the bloodstream, nanocarriers face various in vivo obstacles that may significantly impair their performance needed for clinical translation. Herein, we demonstrate a strategy to enhance tumor-targeting efficiency by embedding functionalities in the interior region of partially PEGylated nanocarriers (ca. 10 nm in diameter), intended for active or passive targeting. The cooperative impact of these topologically inner functional groups (IFGs) was marked: enhancements of >100-fold in IC50in vitro (e.g., a high-avidity ligand with cationic IFGs) and >2-fold in tumor accumulation at 2 h post-injection in vivo (e.g., a high-avidity ligand with anionic IFGs), both against the fully PEGylated counterpart. Analogous to allosteric modulators, properly employed IFGs may substantially improve the process of effectively directing nanocarriers to tumors, which is otherwise solely dependent on avidity or extravasation.
Collapse
Affiliation(s)
- Ju Young Heo
- Korea Research Institute of Bioscience and Biotechnology , Daejeon , 34141 , Korea .
- Department of Chemistry , Korea Advanced Institute of Science and Technology , Daejeon , 34141 , Korea
| | - Se Hun Kang
- Molecular Imaging and Therapy Branch , National Cancer Center , Goyang , 10408 , Korea .
| | - Young-Hwa Kim
- Department of Biomedical Sciences , Seoul National University College of Medicine , Seoul , 03080 , Korea .
- Cancer Research Institute , Seoul National University College of Medicine , Seoul , 03080 , Korea
| | - Suyeon You
- Korea Research Institute of Bioscience and Biotechnology , Daejeon , 34141 , Korea .
| | - Kyeong Sik Jin
- Pohang Accelerator Laboratory , Pohang University of Science and Technology , Pohang , 37673 , Korea
| | - Seung Won Kim
- Molecular Imaging and Therapy Branch , National Cancer Center , Goyang , 10408 , Korea .
| | - Hye-Youn Jung
- Korea Research Institute of Bioscience and Biotechnology , Daejeon , 34141 , Korea .
| | - Kyung Oh Jung
- Department of Biomedical Sciences , Seoul National University College of Medicine , Seoul , 03080 , Korea .
- Cancer Research Institute , Seoul National University College of Medicine , Seoul , 03080 , Korea
| | - Chul-Hee Lee
- Department of Biomedical Sciences , Seoul National University College of Medicine , Seoul , 03080 , Korea .
- Cancer Research Institute , Seoul National University College of Medicine , Seoul , 03080 , Korea
| | - Mi Jung Kim
- Korea Research Institute of Bioscience and Biotechnology , Daejeon , 34141 , Korea .
| | - Soo-Eun Sung
- Korea Research Institute of Bioscience and Biotechnology , Daejeon , 34141 , Korea .
| | - Boram Kim
- Korea Research Institute of Bioscience and Biotechnology , Daejeon , 34141 , Korea .
| | - Insung S Choi
- Department of Chemistry , Korea Advanced Institute of Science and Technology , Daejeon , 34141 , Korea
| | - Hyewon Youn
- Department of Biomedical Sciences , Seoul National University College of Medicine , Seoul , 03080 , Korea .
- Cancer Research Institute , Seoul National University College of Medicine , Seoul , 03080 , Korea
- Department of Nuclear Medicine , Seoul National University Hospital , Seoul , 03080 , Korea
| | - June-Key Chung
- Department of Biomedical Sciences , Seoul National University College of Medicine , Seoul , 03080 , Korea .
- Cancer Research Institute , Seoul National University College of Medicine , Seoul , 03080 , Korea
- Department of Nuclear Medicine , Seoul National University Hospital , Seoul , 03080 , Korea
| | - Seok-Ki Kim
- Molecular Imaging and Therapy Branch , National Cancer Center , Goyang , 10408 , Korea .
| | - Yoonkyung Kim
- Korea Research Institute of Bioscience and Biotechnology , Daejeon , 34141 , Korea .
- Korea University of Science and Technology (UST) , Daejeon , 34113 , Korea
| |
Collapse
|
13
|
Suk JS, Xu Q, Kim N, Hanes J, Ensign LM. PEGylation as a strategy for improving nanoparticle-based drug and gene delivery. Adv Drug Deliv Rev 2016. [DOI: '10.1016/j.addr.2015.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2023]
|
14
|
PEGylation as a strategy for improving nanoparticle-based drug and gene delivery. Adv Drug Deliv Rev 2016; 99:28-51. [PMID: 26456916 DOI: 10.1016/j.addr.2015.09.012] [Citation(s) in RCA: 2642] [Impact Index Per Article: 293.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Revised: 09/21/2015] [Accepted: 09/26/2015] [Indexed: 12/12/2022]
Abstract
Coating the surface of nanoparticles with polyethylene glycol (PEG), or "PEGylation", is a commonly used approach for improving the efficiency of drug and gene delivery to target cells and tissues. Building from the success of PEGylating proteins to improve systemic circulation time and decrease immunogenicity, the impact of PEG coatings on the fate of systemically administered nanoparticle formulations has, and continues to be, widely studied. PEG coatings on nanoparticles shield the surface from aggregation, opsonization, and phagocytosis, prolonging systemic circulation time. Here, we briefly describe the history of the development of PEGylated nanoparticle formulations for systemic administration, including how factors such as PEG molecular weight, PEG surface density, nanoparticle core properties, and repeated administration impact circulation time. A less frequently discussed topic, we then describe how PEG coatings on nanoparticles have also been utilized for overcoming various biological barriers to efficient drug and gene delivery associated with other modes of administration, ranging from gastrointestinal to ocular. Finally, we describe both methods for PEGylating nanoparticles and methods for characterizing PEG surface density, a key factor in the effectiveness of the PEG surface coating for improving drug and gene delivery.
Collapse
|
15
|
You S, Jung HY, Lee C, Choe YH, Heo JY, Gang GT, Byun SK, Kim WK, Lee CH, Kim DE, Kim YI, Kim Y. High-performance dendritic contrast agents for X-ray computed tomography imaging using potent tetraiodobenzene derivatives. J Control Release 2016; 226:258-67. [PMID: 26812006 DOI: 10.1016/j.jconrel.2016.01.036] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 01/13/2016] [Accepted: 01/21/2016] [Indexed: 10/22/2022]
Abstract
The use of computed tomography (CT) for vascular imaging is critical in medical emergencies requiring urgent diagnostic decisions, such as cerebral ischemia and many cardiovascular diseases. Small-molecule iodinated contrast media are often injected intravenously as radiopaque agents during CT imaging to achieve high contrast enhancement of vascular systems. The rapid excretion rate of these agents is overcome by injecting a significantly high dose of iodine, which can have serious side effects. Here we report a simple method to prepare blood-pool contrast agents for CT based on dendrimers for the first time using tetraiodobenzene derivatives as potent radiopaque moieties. Excellent in vivo safety has been demonstrated for these small (13-22nm) unimolecular water-soluble dendritic contrast agents, which exhibit high contrast enhancement in the blood-pool and effectively extend their blood half-lives. Our method is applicable to virtually any scaffold with suitable surface groups and may fulfill the current need for safer, next-generation iodinated CT contrast agents.
Collapse
Affiliation(s)
- Suyeon You
- Biomedical Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea
| | - Hye-Youn Jung
- Biomedical Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea
| | - Chaewoon Lee
- Biomedical Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea
| | - Yun Hui Choe
- Biomedical Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea
| | - Ju Young Heo
- Biomedical Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea; Department of Chemistry, Korea Advanced Institute of Science and Technology, Daejeon 305-701, Republic of Korea
| | - Gil-Tae Gang
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea
| | - Sang-Kyung Byun
- Functional Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea; Korea University of Science and Technology (UST), Daejeon 305-350, Republic of Korea
| | - Won Kon Kim
- Functional Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea; Korea University of Science and Technology (UST), Daejeon 305-350, Republic of Korea
| | - Chul-Ho Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea; Korea University of Science and Technology (UST), Daejeon 305-350, Republic of Korea
| | - Dong-Eog Kim
- Department of Neurology, Dongguk University Ilsan Hospital, Goyang 410-773, Republic of Korea
| | - Young Il Kim
- Department of Radiology, Seoul National University Hospital, Seoul 110-744, Republic of Korea
| | - Yoonkyung Kim
- Biomedical Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea; Korea University of Science and Technology (UST), Daejeon 305-350, Republic of Korea.
| |
Collapse
|
16
|
Zheng S, Bai YY, Liu Y, Gao X, Li Y, Changyi Y, Wang Y, Chang D, Ju S, Li C. Salvaging brain ischemia by increasing neuroprotectant uptake via nanoagonist mediated blood brain barrier permeability enhancement. Biomaterials 2015; 66:9-20. [PMID: 26188608 DOI: 10.1016/j.biomaterials.2015.07.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Revised: 07/03/2015] [Accepted: 07/09/2015] [Indexed: 12/28/2022]
Abstract
Ischemic stroke is a leading cause of adult disability and cognitive impairment worldwide. Neuroprotective therapy aims to save neurons by impeding the deleterious ischemic insults. However, the low efficiency of the neuroprotectants crossing blood brain barrier (BBB) prevents their clinical translation. In this work, a nanoagonist (NA) was developed to enhance neuroprotectant uptake by specifically increasing BBB permeability in brain ischemia. This NA first targeted ischemic brain vasculatures, temporarily opened local BBB by activating adenosine 2A receptors, and up-regulated the neuroprotectant uptake in brain ischemia. This NA significantly increased the delivery of superoxide dismutase (SOD), a free radical scavenger, into mouse brain ischemia. The combined treatment of NA/SOD achieved a five-fold ischemic volume reduction rate compared to the animal models treated with SOD alone. Non-invasive magnetic resonance imaging (MRI) confirmed the ischemia targeted BBB opening, increased brain drug delivery efficiency and up-regulated therapeutic response during the combined NA/SOD treatment. Since the inefficient brain drug delivery is a general problem for the treatment of central nervous system (CNS) diseases, this work provides a novel strategy to deliver therapeutics by crossing BBB with high efficiency and targeting specificity.
Collapse
Affiliation(s)
- Shuyan Zheng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, 826 Zhangheng Rd., Shanghai 201203, China
| | - Ying-Ying Bai
- Jiangsu Key Laboratory of Molecular and Functional Imaging Department of Radiology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Rd., Nanjing 210009, China
| | - Yikang Liu
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Xihui Gao
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, 826 Zhangheng Rd., Shanghai 201203, China
| | - Yan Li
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Rd., Shenyang 110016, China
| | - Yinzhi Changyi
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, 826 Zhangheng Rd., Shanghai 201203, China
| | - Yuancheng Wang
- Jiangsu Key Laboratory of Molecular and Functional Imaging Department of Radiology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Rd., Nanjing 210009, China
| | - Di Chang
- Jiangsu Key Laboratory of Molecular and Functional Imaging Department of Radiology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Rd., Nanjing 210009, China
| | - Shenghong Ju
- Jiangsu Key Laboratory of Molecular and Functional Imaging Department of Radiology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Rd., Nanjing 210009, China.
| | - Cong Li
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, 826 Zhangheng Rd., Shanghai 201203, China.
| |
Collapse
|
17
|
Ilinskaya AN, Dobrovolskaia MA. Nanoparticles and the blood coagulation system. Part I: benefits of nanotechnology. Nanomedicine (Lond) 2013; 8:773-84. [PMID: 23656264 DOI: 10.2217/nnm.13.48] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Nanotechnology is proven to provide certain benefits in drug delivery by improving solubility, increasing uptake to target sites and changing pharmacokinetics profiles of traditional drugs. Since properties of many materials change tremendously at the nanoscale levels, nanotechnology is also being explored in various industrial applications. As such, nanoparticles are rapidly entering various areas of industry, biology and medicine. The benefits of using nanotechnology for industrial and biomedical applications are often tempered by concerns about the safety of these new materials. One such area of concern includes their effect on the immune system. While nanoparticle interactions with various constituents of the immune system have been reviewed before, little attention was given to nanoparticle effects on the blood coagulation system. Nanoparticle interface with the blood coagulation system may lead to either benefits to the host or adverse reactions. This article reviews recent advances in our understanding of nanoparticle interactions with plasma coagulation factors, platelets, endothelial cells and leukocytes. Part I is focused on desirable interactions between nanoparticles and the coagulation system, and discusses benefits of using nanotechnology to intervene in coagulation disorders. Undesirable interactions posing safety concerns are covered in part II, which will be published in the June issue of Nanomedicine.
Collapse
Affiliation(s)
- Anna N Ilinskaya
- Nanotechnology Characterization Laboratory, Advanced Technology Program, SAIC-Frederick Inc., NCI-Frederick, 1050 Boyles Street, Building 469, Frederick, MD 21702, USA
| | | |
Collapse
|
18
|
Multivalent ligand: design principle for targeted therapeutic delivery approach. Ther Deliv 2012; 3:1171-87. [DOI: 10.4155/tde.12.99] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Multivalent interactions of biological molecules play an important role in many biochemical events. A multivalent ligand comprises of multiple copies of ligands conjugated to scaffolds, allowing the simultaneous binding of multivalent ligands to multiple binding sites or receptors. Many research groups have successfully designed and synthesized multivalent ligands to increase the binding affinity, avidity and specificity of the ligand to the receptor. A multimeric ligand is a promising option for the specific treatment of diseases. In this review, the factors affecting multivalent interactions, including the size and shape of the ligand, geometry and an arrangement of ligands on the scaffold, linker length, thermodynamic, and kinetics of the interactions are discussed. Examples of the multivalent ligand applications for therapeutic delivery are also summarized.
Collapse
|
19
|
Chanyshev B, Shainberg A, Isak A, Litinsky A, Chepurko Y, Tosh DK, Phan K, Gao ZG, Hochhauser E, Jacobson KA. Anti-ischemic effects of multivalent dendrimeric A₃ adenosine receptor agonists in cultured cardiomyocytes and in the isolated rat heart. Pharmacol Res 2011; 65:338-46. [PMID: 22154845 DOI: 10.1016/j.phrs.2011.11.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Revised: 11/21/2011] [Accepted: 11/22/2011] [Indexed: 12/28/2022]
Abstract
Adenosine released during myocardial ischemia mediates cardioprotective preconditioning. Multivalent drugs covalently bound to nanocarriers may differ greatly in chemical and biological properties from the corresponding monomeric agents. Here, we conjugated chemically functionalized nucleosides to poly(amidoamine) (PAMAM) dendrimeric polymers and investigated their effects in rat primary cardiac cell cultures and in the isolated heart. Three conjugates of A₃ adenosine receptor (AR) agonists, chain-functionalized at the C2 or N⁶ position, were cardioprotective, with greater potency than monomeric agonist Cl-IB-MECA. Multivalent amide-linked MRS5216 was selective for A₁ and A₃ARs, and triazole-linked MRS5246 and MRS5539 (optionally containing fluorescent label) were A₃AR-selective. The conjugates protected ischemic rat cardiomyocytes, an effect blocked by an A₃AR antagonist MRS1523, and isolated hearts with significantly improved infarct size, rate of pressure product, and rate of contraction and relaxation. Thus, strategically derivatized nucleosides tethered to biocompatible polymeric carriers display enhanced cardioprotective potency via activation of A₃AR on the cardiomyocyte surface.
Collapse
Affiliation(s)
- Bella Chanyshev
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Kecskés A, Tosh DK, Wei Q, Gao ZG, Jacobson KA. GPCR ligand dendrimer (GLiDe) conjugates: adenosine receptor interactions of a series of multivalent xanthine antagonists. Bioconjug Chem 2011; 22:1115-27. [PMID: 21539392 DOI: 10.1021/bc1005812] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Previously, G protein-coupled receptor (GPCR) agonists were tethered from polyamidoamine (PAMAM) dendrimers to provide high receptor affinity and selectivity. Here, we prepared GPCR ligand--dendrimer (GLiDe) conjugates from a potent adenosine receptor (AR) antagonist; such agents are of interest for treating Parkinson's disease, asthma, and other conditions. Xanthine amine congener (XAC) was appended with an alkyne group on an extended C8 substituent for coupling by Cu(I)-catalyzed click chemistry to azide-derivatized G4 (fourth-generation) PAMAM dendrimers to form triazoles. These conjugates also contained triazole-linked PEG groups (8 or 22 moieties per 64 terminal positions) for increasing water-solubility and optionally prosthetic groups for spectroscopic characterization and affinity labeling. Human AR binding affinity increased progressively with the degree of xanthine substitution to reach K(i) values in the nanomolar range. The order of affinity of each conjugate was hA(2A)AR > hA(3)AR > hA(1)AR, while the corresponding monomer was ranked hA(2A)AR > hA(1)AR ≥ hA(3)AR. The antagonist activity of the most potent conjugate 14 (34 xanthines per dendrimer) was examined at the G(i)-coupled A(1)AR. Conjugate 14 at 100 nM right-shifted the AR agonist concentration--response curve in a cyclic AMP functional assay in a parallel manner, but at 10 nM (lower than its K(i) value), it significantly suppressed the maximal agonist effect in calcium mobilization. This is the first systematic probing of a potent AR antagonist tethered on a dendrimer and its activity as a function of variable loading.
Collapse
Affiliation(s)
- Angela Kecskés
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
| | | | | | | | | |
Collapse
|
21
|
Müller CE, Jacobson KA. Recent developments in adenosine receptor ligands and their potential as novel drugs. BIOCHIMICA ET BIOPHYSICA ACTA 2011; 1808:1290-308. [PMID: 21185259 PMCID: PMC3437328 DOI: 10.1016/j.bbamem.2010.12.017] [Citation(s) in RCA: 337] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Revised: 12/14/2010] [Accepted: 12/15/2010] [Indexed: 01/16/2023]
Abstract
Medicinal chemical approaches have been applied to all four of the adenosine receptor (AR) subtypes (A(1), A(2A), A(2B), and A(3)) to create selective agonists and antagonists for each. The most recent class of selective AR ligands to be reported is the class of A(2B)AR agonists. The availability of these selective ligands has facilitated research on therapeutic applications of modulating the ARs and in some cases has provided clinical candidates. Prodrug approaches have been developed which improve the bioavailability of the drugs, reduce side-effects, and/or may lead to site-selective effects. The A(2A) agonist regadenoson (Lexiscan®), a diagnostic drug for myocardial perfusion imaging, is the first selective AR agonist to be approved. Other selective agonists and antagonists are or were undergoing clinical trials for a broad range of indications, including capadenoson and tecadenoson (A(1) agonists) for atrial fibrillation, or paroxysmal supraventricular tachycardia, respectively, apadenoson and binodenoson (A(2A) agonists) for myocardial perfusion imaging, preladenant (A(2A) antagonist) for the treatment of Parkinson's disease, and CF101 and CF102 (A(3) agonists) for inflammatory diseases and cancer, respectively.
Collapse
|
22
|
Chen B, van der Poll DG, Jerger K, Floyd WC, Fréchet JMJ, Szoka FC. Synthesis and properties of star-comb polymers and their doxorubicin conjugates. Bioconjug Chem 2011; 22:617-24. [PMID: 21375296 DOI: 10.1021/bc100400u] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We describe a six-step synthesis to water-soluble doxorubicin (DOX)-loaded biodegradable PEGylated star-comb polymers with favorable pharmaceutical properties by atom transfer radical polymerization (ATRP) starting with a commercially available tripentaerythritol carrying eight reactive sites. The low polydispersity polymers degrade in a stepwise manner into lower molecular weight (MW) fragments by 15 days at 37 °C at either pH 5.0 or pH 7.4. The half-life of the star-comb polymers in blood is dependent upon the molecular weight; the 44 kDa star-comb has a t(1/2, β) of 30.5 ± 2.1 h, which is not significantly changed (28.6 ± 2.7 h) when 6.6 wt % of DOX is attached to it via a pH-sensitive hydrazone linker. The star-comb polymers have low accumulation in organs but a high accumulation in C26 flank tumors implanted in Balb/C mice. The hydrodynamic diameter of polymer-DOX conjugates measured by dynamic light scattering increases from 8 to 35 to 41 nm as the loading is increased from 6.6 to 8.4 to 10.2 wt %. Although there is no significant difference in the t(1/2, β) or in the accumulation of polymer-DOX in C-26 tumors, the uptake of polymer in the spleen is significantly higher for polymers with DOX loadings greater than 6.6 wt %. Polymer accumulation in other vital organs is independent of the DOX loading. The facile synthesis, biodegradability, long circulation time, and high tumor accumulation of the attached drug suggests that the water-soluble star-comb polymers have promise in therapeutic applications.
Collapse
Affiliation(s)
- Bo Chen
- Department of Bioengineering, Therapeutic Sciences and Pharmaceutical Chemistry, University of California, San Francisco , California 94143-0912, United States
| | | | | | | | | | | |
Collapse
|
23
|
Pharmacochemistry of the platelet purinergic receptors. Purinergic Signal 2011; 7:305-24. [PMID: 21484092 DOI: 10.1007/s11302-011-9216-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2010] [Accepted: 01/11/2011] [Indexed: 10/18/2022] Open
Abstract
Platelets contain at least five purinergic G protein-coupled receptors, e.g., the pro-aggregatory P2Y(1) and P2Y(12) receptors, a P2Y(14) receptor (GPR105) of unknown function, and anti-aggregatory A(2A) and A(2B) adenosine receptor (ARs), in addition to the ligand-gated P2X1 ion channel. Probing the structure-activity relationships (SARs) of the P2X and P2Y receptors for extracellular nucleotides has resulted in numerous new agonist and antagonist ligands. Selective agents derived from known ligands and novel chemotypes can be used to help define the subtypes pharmacologically. Some of these agents have entered into clinical trials in spite of the challenges of drug development for these classes of receptors. The functional architecture of P2 receptors was extensively explored using mutagenesis and molecular modeling, which are useful tools in drug discovery. In general, novel drug delivery methods, prodrug approaches, allosteric modulation, and biased agonism would be desirable to overcome side effects that tend to occur even with receptor subtype-selective ligands. Detailed SAR analyses have been constructed for nucleotide and non-nucleotide ligands at the P2Y(1), P2Y(12), and P2Y(14) receptors. The thienopyridine antithrombotic drugs Clopidogrel and Prasugrel require enzymatic pre-activation in vivo and react irreversibly with the P2Y(12) receptor. There is much pharmaceutical development activity aimed at identifying reversible P2Y(12) receptor antagonists. The screening of chemically diverse compound libraries has identified novel chemotypes that act as competitive, non-nucleotide antagonists of the P2Y(1) receptor or the P2Y(12) receptor, and antithrombotic properties of the structurally optimized analogues were demonstrated. In silico screening at the A(2A) AR has identified antagonist molecules having novel chemotypes. Fluorescent and other reporter groups incorporated into ligands can enable new technology for receptor assays and imaging. The A(2A) agonist CGS21680 and the P2Y(1) receptor antagonist MRS2500 were derivatized for covalent attachment to polyamidoamine dendrimeric carriers of MW 20,000, and the resulting multivalent conjugates inhibited ADP-promoted platelet aggregation. In conclusion, a wide range of new pharmacological tools is available to control platelet function by interacting with cell surface purine receptors.
Collapse
|
24
|
de Castro S, Maruoka H, Hong K, Kilbey SM, Costanzi S, Hechler B, Brown GG, Gachet C, Harden TK, Jacobson KA. Functionalized congeners of P2Y1 receptor antagonists: 2-alkynyl (N)-methanocarba 2'-deoxyadenosine 3',5'-bisphosphate analogues and conjugation to a polyamidoamine (PAMAM) dendrimer carrier. Bioconjug Chem 2010; 21:1190-205. [PMID: 20565071 DOI: 10.1021/bc900569u] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The P2Y(1) receptor is a prothrombotic G protein-coupled receptor (GPCR) activated by ADP. Preference for the North (N) ring conformation of the ribose moiety of adenine nucleotide 3',5'-bisphosphate antagonists of the P2Y(1) receptor was established by using a ring-constrained methanocarba (a bicyclo[3.1.0]hexane) ring as a ribose substitute. A series of covalently linkable N(6)-methyl-(N)-methanocarba-2'-deoxyadenosine-3',5'-bisphosphates containing extended 2-alkynyl chains was designed, and binding affinity at the human (h) P2Y(1) receptor determined. The chain of these functionalized congeners contained hydrophilic moieties, a reactive substituent, or biotin, linked via an amide. Variation of the chain length and position of an intermediate amide group revealed high affinity of carboxylic congener 8 (K(i) 23 nM) and extended amine congener 15 (K(i) 132 nM), both having a 2-(1-pentynoyl) group. A biotin conjugate 18 containing an extended epsilon-aminocaproyl spacer chain exhibited higher affinity than a shorter biotinylated analogue. Alternatively, click coupling of terminal alkynes of homologous 2-dialkynyl nucleotide derivatives to alkyl azido groups produced triazole derivatives that bound to the P2Y(1) receptor following deprotection of the bisphosphate groups. The preservation of receptor affinity of the functionalized congeners was consistent with new P2Y(1) receptor modeling and ligand docking. Attempted P2Y(1) antagonist conjugation to PAMAM dendrimer carriers by amide formation or palladium-catalyzed reaction between an alkyne on the dendrimer and a 2-iodopurine-derivatized nucleotide was unsuccessful. A dialkynyl intermediate containing the chain length favored in receptor binding was conjugated to an azide-derivatized dendrimer, and the conjugate inhibited ADP-promoted human platelet aggregation. This is the first example of attaching a strategically functionalized P2Y receptor antagonist to a PAMAM dendrimer to produce a multivalent conjugate exhibiting a desired biological effect, i.e., antithrombotic action.
Collapse
Affiliation(s)
- Sonia de Castro
- Molecular Recognition Section and Laboratory of Biological Modeling, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, 20892-0810, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Nwe K, Bryant LH, Brechbiel MW. Poly(amidoamine) dendrimer based MRI contrast agents exhibiting enhanced relaxivities derived via metal preligation techniques. Bioconjug Chem 2010; 21:1014-7. [PMID: 20462240 DOI: 10.1021/bc1000802] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
This report presents the preparation and characterization of three [Gd-C-DOTA](-1)-dendrimer assemblies by way of analysis, NMRD spectroscopy, and photon correlation spectroscopy (PCS). The metal-ligand chelates were preformed in alcohol media prior to conjugation to generation 4, 5, and 6 PAMAM dendrimers. The dendrimer-based agents were purified by Sephadex G-25 column chromatography. The combustion analysis, SE-HPLC, and UV-vis data indicated chelate to dendrimer ratios of 28:1, 61:1 and 115:1, respectively. Molar relaxivity measured at pH 7.4, 22 degrees C, and 3 T (29.6, 49.8, and 89.1 mM(-1) s(-1)) indicated the viability of conjugates as MRI contrast agents. 1/T(1) NMRD profiles were measured at 23 degrees C and indicated that at 22 MHz the 1/T(1) reached a plateau at 60, 85, and 140 mM(-1) s(-1) for the generation 4, 5, and 6 dendrimer conjugates, respectively. The PCS data showed the respective sizes of 5.2, 6.5, and 7.8 nm for G-4, 5, and 6 conjugates.
Collapse
Affiliation(s)
- Kido Nwe
- Radioimmune & Inorganic Chemistry Section, Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland 20892, USA
| | | | | |
Collapse
|
26
|
Jacobson KA. GPCR ligand-dendrimer (GLiDe) conjugates: future smart drugs? Trends Pharmacol Sci 2010; 31:575-9. [PMID: 20961625 DOI: 10.1016/j.tips.2010.09.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2010] [Revised: 09/15/2010] [Accepted: 09/16/2010] [Indexed: 10/18/2022]
Abstract
Unlike nanocarriers that are intended to release their drug cargo at the site of action, biocompatibile polyamidoamine (PAMAM) conjugates are designed to act at cell surface G protein-coupled receptors (GPCRs) without drug release. These multivalent GPCR ligand-dendrimer (GLiDe) conjugates display qualitatively different pharmacological properties in comparison with monomeric drugs. They might be useful as novel tools to study GPCR homodimers and heterodimers as well as higher aggregates. The structure of the conjugate determines the profile of biological activity, receptor selectivity, and physical properties such as water solubility. Prosthetic groups for characterization and imaging of receptors can be introduced without loss of affinity. The feasibility of targeting multiple adenosine and P2Y receptors for synergistic effects has been shown. Testing in vivo will be needed to explore the effects on pharmacokinetics and tissue targeting.
Collapse
Affiliation(s)
- Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes, Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
27
|
Jiarpinitnun C, Kiessling LL. Unexpected enhancement in biological activity of a GPCR ligand induced by an oligoethylene glycol substituent. J Am Chem Soc 2010; 132:8844-5. [PMID: 20536128 DOI: 10.1021/ja102640c] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Polyethylene glycol (PEG) is widely used, and many biologically active molecules are modified with oligoethylene glycol substituents to enhance their half-lives in circulation. The pervasive use of PEG substituents is partly due to their presumed inertness. Our investigation of formyl peptide receptor (FPR)-mediated chemotaxis reveals that oligoethylene glycol substitution can enhance the ability of the peptide chemoattractant N-formyl-methionine-leucine-phenylalanine (fMLF) to activate signal transduction through FPR, a transmembrane G-protein-coupled receptor.
Collapse
Affiliation(s)
- Chutima Jiarpinitnun
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | | |
Collapse
|
28
|
Das A, Sanjayan GJ, Kecskés M, Yoo L, Gao ZG, Jacobson KA. Nucleoside conjugates of quantum dots for characterization of G protein-coupled receptors: strategies for immobilizing A2A adenosine receptor agonists. J Nanobiotechnology 2010; 8:11. [PMID: 20478037 PMCID: PMC2883535 DOI: 10.1186/1477-3155-8-11] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2010] [Accepted: 05/17/2010] [Indexed: 01/19/2023] Open
Abstract
Background Quantum dots (QDs) are crystalline nanoparticles that are compatible with biological systems to provide a chemically and photochemically stable fluorescent label. New ligand probes with fluorescent reporter groups are needed for detection and characterization of G protein-coupled receptors (GPCRs). Results Synthetic strategies for coupling the A2A adenosine receptor (AR) agonist CGS21680 (2-[4-(2-carboxyethyl)phenylethylamino]-5'-N-ethylcarboxamidoadenosine) to functionalized QDs were explored. Conjugates tethered through amide-linked chains and poly(ethyleneglycol) (PEG) displayed low solubility and lacked receptor affinity. The anchor to the dendron was either through two thiol groups of (R)-thioctic acid or through amide formation to a commercial carboxy-derivatized QD. The most effective approach was to use polyamidoamine (PAMAM) D5 dendrons as multivalent spacer groups, grafted on the QD surface through a thioctic acid moiety. In radioligand binding assays, dendron nucleoside conjugate 11 displayed a moderate affinity at the human A2AAR (Kiapp 1.02 ± 0.15 μM). The QD conjugate of increased water solubility 13, resulting from the anchoring of this dendron derivative, interacted with the receptor with Kiapp of 118 ± 54 nM. The fluorescence emission of 13 occurred at 565 nm, and the presence of the pendant nucleoside did not appreciably quench the fluorescence. Conclusions This is a feasibility study to demonstrate a means of conjugating to a QD a small molecular pharmacophore of a GPCR that is relatively hydrophobic. Further enhancement of affinity by altering the pharmacophore or the linking structures will be needed to make useful affinity probes.
Collapse
Affiliation(s)
- Arijit Das
- Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | | | | | | | | | |
Collapse
|