51
|
Jeevanandam J, Tan KX, Danquah MK, Guo H, Turgeson A. Advancing Aptamers as Molecular Probes for Cancer Theranostic Applications-The Role of Molecular Dynamics Simulation. Biotechnol J 2020; 15:e1900368. [PMID: 31840436 DOI: 10.1002/biot.201900368] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 12/06/2019] [Indexed: 12/24/2022]
Abstract
Theranostics cover emerging technologies for cell biomarking for disease diagnosis and targeted introduction of drug ingredients to specific malignant sites. Theranostics development has become a significant biomedical research endeavor for effective diagnosis and treatment of diseases, especially cancer. An efficient biomarking and targeted delivery strategy for theranostic applications requires effective molecular coupling of binding ligands with high affinities to specific receptors on the cancer cell surface. Bioaffinity offers a unique mechanism to bind specific target and receptor molecules from a range of non-targets. The binding efficacy depends on the specificity of the affinity ligand toward the target molecule even at low concentrations. Aptamers are fragments of genetic materials, peptides, or oligonucleotides which possess enhanced specificity in targeting desired cell surface receptor molecules. Aptamer-target binding results from several inter-molecular interactions including hydrogen bond formation, aromatic stacking of flat moieties, hydrophobic interaction, electrostatic, and van der Waals interactions. Advancements in Systematic Evolution of Ligands by Exponential Enrichment (SELEX) assay has created the opportunity to artificially generate aptamers that specifically bind to desired cancer and tumor surface receptors with high affinities. This article discusses the potential application of molecular dynamics (MD) simulation to advance aptamer-mediated receptor targeting in targeted cancer therapy. MD simulation offers real-time analysis of the molecular drivers of the aptamer-receptor binding and generate optimal receptor binding conditions for theranostic applications. The article also provides an overview of different cancer types with focus on receptor biomarking and targeted treatment approaches, conventional molecular probes, and aptamers that have been explored for cancer cells targeting.
Collapse
Affiliation(s)
- Jaison Jeevanandam
- Department of Chemical Engineering, Faculty of Engineering and Science, Curtin University, Miri, Sarawak, 98009, Malaysia
| | - Kei Xian Tan
- School of Materials Science & Engineering, Nanyang Technological University, Singapore, 639798
| | | | - Haobo Guo
- Department of Computer Science and Engineering, University of Tennessee, Chattanooga, TN, 37403, USA.,SimCenter, University of Tennessee, Chattanooga, TN, 37403, USA
| | - Andrew Turgeson
- Chemical Engineering Department, University of Tennessee, Chattanooga, TN, 37403, USA
| |
Collapse
|
52
|
Chen S, Xu Z, Yang W, Lin X, Li J, Li J, Yang H. Logic-Gate-Actuated DNA-Controlled Receptor Assembly for the Programmable Modulation of Cellular Signal Transduction. Angew Chem Int Ed Engl 2019; 58:18186-18190. [PMID: 31595614 DOI: 10.1002/anie.201908971] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/18/2019] [Indexed: 02/01/2023]
Abstract
Programming cells to sense multiple inputs and activate cellular signal transduction cascades is of great interest. Although this goal has been achieved through the engineering of genetic circuits using synthetic biology tools, a nongenetic and generic approach remains highly demanded. Herein, we present an aptamer-controlled logic receptor assembly for modulating cellular signal transduction. Aptamers were engineered as "robotic arms" to capture target receptors (c-Met and CD71) and a DNA logic assembly functioned as a computer processor to handle multiple inputs. As a result, the DNA assembly brings c-Met and CD71 into close proximity, thus interfering with the ligand-receptor interactions of c-Met and inhibiting its functions. Using this principle, a set of logic gates was created that respond to DNA strands or light irradiation, modulating the c-Met/HGF signal pathways. This simple modular design provides a robust chemical tool for modulating cellular signal transduction.
Collapse
Affiliation(s)
- Shan Chen
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, P. R. China.,Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Zhifei Xu
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, P. R. China
| | - Wen Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, P. R. China
| | - Xiahui Lin
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, P. R. China
| | - Jingying Li
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, P. R. China.,College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, P. R. China
| | - Juan Li
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, P. R. China.,Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Huanghao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, P. R. China
| |
Collapse
|
53
|
Chen S, Xu Z, Yang W, Lin X, Li J, Li J, Yang H. Logic‐Gate‐Actuated DNA‐Controlled Receptor Assembly for the Programmable Modulation of Cellular Signal Transduction. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201908971] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Shan Chen
- MOE Key Laboratory for Analytical Science of Food Safety and BiologyFujian Provincial Key Laboratory of Analysis and Detection Technology for Food SafetyState Key Laboratory of Photocatalysis on Energy and EnvironmentCollege of ChemistryFuzhou University Fuzhou 350108 P. R. China
- Institute of Molecular MedicineRenji HospitalShanghai Jiao Tong University School of MedicineShanghai Jiao Tong University Shanghai 200240 P. R. China
| | - Zhifei Xu
- MOE Key Laboratory for Analytical Science of Food Safety and BiologyFujian Provincial Key Laboratory of Analysis and Detection Technology for Food SafetyState Key Laboratory of Photocatalysis on Energy and EnvironmentCollege of ChemistryFuzhou University Fuzhou 350108 P. R. China
| | - Wen Yang
- MOE Key Laboratory for Analytical Science of Food Safety and BiologyFujian Provincial Key Laboratory of Analysis and Detection Technology for Food SafetyState Key Laboratory of Photocatalysis on Energy and EnvironmentCollege of ChemistryFuzhou University Fuzhou 350108 P. R. China
| | - Xiahui Lin
- MOE Key Laboratory for Analytical Science of Food Safety and BiologyFujian Provincial Key Laboratory of Analysis and Detection Technology for Food SafetyState Key Laboratory of Photocatalysis on Energy and EnvironmentCollege of ChemistryFuzhou University Fuzhou 350108 P. R. China
| | - Jingying Li
- MOE Key Laboratory for Analytical Science of Food Safety and BiologyFujian Provincial Key Laboratory of Analysis and Detection Technology for Food SafetyState Key Laboratory of Photocatalysis on Energy and EnvironmentCollege of ChemistryFuzhou University Fuzhou 350108 P. R. China
- College of Biological Science and EngineeringFuzhou University Fuzhou 350108 P. R. China
| | - Juan Li
- MOE Key Laboratory for Analytical Science of Food Safety and BiologyFujian Provincial Key Laboratory of Analysis and Detection Technology for Food SafetyState Key Laboratory of Photocatalysis on Energy and EnvironmentCollege of ChemistryFuzhou University Fuzhou 350108 P. R. China
- Institute of Molecular MedicineRenji HospitalShanghai Jiao Tong University School of MedicineShanghai Jiao Tong University Shanghai 200240 P. R. China
| | - Huanghao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and BiologyFujian Provincial Key Laboratory of Analysis and Detection Technology for Food SafetyState Key Laboratory of Photocatalysis on Energy and EnvironmentCollege of ChemistryFuzhou University Fuzhou 350108 P. R. China
| |
Collapse
|
54
|
Kuwahara M, Fujita H, Kataoka Y, Nakajima Y, Yamada M, Sugimoto N. In situ condensation of an anti-cancer drug into fibrin gel enabling effective inhibition of tumor cell growth. Chem Commun (Camb) 2019; 55:11679-11682. [PMID: 31524892 DOI: 10.1039/c9cc06418d] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
We show herein the highly effective inhibition of tumor cell growth using a gel consisting of a fibrin polymer formed with the in situ condensation of a camptothecin (CPT) derivative as an anti-cancer drug, which is efficiently conveyed with a carrier aptamer from a solution to the gel in a phenomenon, called selective oligonucleotide entrapment in fibrin polymers (SOEF).
Collapse
Affiliation(s)
- Masayasu Kuwahara
- Graduate School of Integrated Basic Sciences, Nihon University, 3-25-40 Sakurajosui, Setagaya-ku, Tokyo 156-8550, Japan.
| | - Hiroto Fujita
- Graduate School of Integrated Basic Sciences, Nihon University, 3-25-40 Sakurajosui, Setagaya-ku, Tokyo 156-8550, Japan.
| | - Yuka Kataoka
- Graduate School of Integrated Basic Sciences, Nihon University, 3-25-40 Sakurajosui, Setagaya-ku, Tokyo 156-8550, Japan.
| | - Yasuyo Nakajima
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Graduate School of Medicine, Gunma University, 3-39-15 Showa-machi, Maebashi, 371-8511, Japan
| | - Masanobu Yamada
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Graduate School of Medicine, Gunma University, 3-39-15 Showa-machi, Maebashi, 371-8511, Japan
| | - Naoki Sugimoto
- Frontier Institute for Biomolecular Engineering Research (FIBER), Konan University, 7-1-20 Minatojima-minamimachi, Kobe, 650-0047, Japan and Graduate School of Frontiers of Innovative Research in Science and Technology (FIRST), Konan University, 7-1-20 Minatojima-minamimachi, Kobe, 650-0047, Japan
| |
Collapse
|
55
|
Fechter P, Cruz Da Silva E, Mercier MC, Noulet F, Etienne-Seloum N, Guenot D, Lehmann M, Vauchelles R, Martin S, Lelong-Rebel I, Ray AM, Seguin C, Dontenwill M, Choulier L. RNA Aptamers Targeting Integrin α5β1 as Probes for Cyto- and Histofluorescence in Glioblastoma. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 17:63-77. [PMID: 31226519 PMCID: PMC6586995 DOI: 10.1016/j.omtn.2019.05.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 05/03/2019] [Accepted: 05/03/2019] [Indexed: 02/07/2023]
Abstract
Nucleic acid aptamers are often referred to as chemical antibodies. Because they possess several advantages, like their smaller size, temperature stability, ease of chemical modification, lack of immunogenicity and toxicity, and lower cost of production, aptamers are promising tools for clinical applications. Aptamers against cell surface protein biomarkers are of particular interest for cancer diagnosis and targeted therapy. In this study, we identified and characterized RNA aptamers targeting cells expressing integrin α5β1. This αβ heterodimeric cell surface receptor is implicated in tumor angiogenesis and solid tumor aggressiveness. In glioblastoma, integrin α5β1 expression is associated with an aggressive phenotype and a decrease in patient survival. We used a complex and original hybrid SELEX (selective evolution of ligands by exponential enrichment) strategy combining protein-SELEX cycles on the recombinant α5β1 protein, surrounded by cell-SELEX cycles using two different cell lines. We identified aptamer H02, able to differentiate, in cyto- and histofluorescence assays, glioblastoma cell lines, and tissues from patient-derived tumor xenografts according to their α5 expression levels. Aptamer H02 is therefore an interesting tool for glioblastoma tumor characterization.
Collapse
Affiliation(s)
- Pierre Fechter
- CNRS, UMR 7242, Biotechnologie et Signalisation Cellulaire, Institut de Recherche de l'Ecole de Biotechnologie de Strasbourg, Université de Strasbourg, 67400 Illkirch-Graffenstaden, France
| | - Elisabete Cruz Da Silva
- CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Tumoral Signaling and Therapeutic Targets, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France
| | - Marie-Cécile Mercier
- CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Tumoral Signaling and Therapeutic Targets, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France
| | - Fanny Noulet
- CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Tumoral Signaling and Therapeutic Targets, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France
| | - Nelly Etienne-Seloum
- CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Tumoral Signaling and Therapeutic Targets, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France; Département de Pharmacie, Centre de Lutte Contre le Cancer Paul Strauss, 67000 Strasbourg, France
| | - Dominique Guenot
- EA 3430, Progression Tumorale et Micro-environnement, Approches Translationnelles et Épidémiologie, Université de Strasbourg, 67000 Strasbourg, France
| | - Maxime Lehmann
- CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Tumoral Signaling and Therapeutic Targets, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France
| | - Romain Vauchelles
- CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Tumoral Signaling and Therapeutic Targets, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France
| | - Sophie Martin
- CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Tumoral Signaling and Therapeutic Targets, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France
| | - Isabelle Lelong-Rebel
- CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Tumoral Signaling and Therapeutic Targets, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France
| | - Anne-Marie Ray
- CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Tumoral Signaling and Therapeutic Targets, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France
| | - Cendrine Seguin
- CNRS, UMR 7199, Laboratoire de Conception et Application de Molécules Bioactives, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France
| | - Monique Dontenwill
- CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Tumoral Signaling and Therapeutic Targets, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France
| | - Laurence Choulier
- CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Tumoral Signaling and Therapeutic Targets, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France.
| |
Collapse
|
56
|
Vandghanooni S, Eskandani M, Barar J, Omidi Y. Aptamedicine: a new treatment modality in personalized cancer therapy. ACTA ACUST UNITED AC 2019; 9:67-70. [PMID: 31334037 PMCID: PMC6637218 DOI: 10.15171/bi.2019.09] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 11/15/2018] [Indexed: 11/21/2022]
Abstract
Aptamers ( Aps ) are short single-strand nucleic acids exhibiting unique 3D structure which facilitate their targeting potential against various cancer molecular markers ( CMMs ). Such features of Aps not only make them as suitable homing agents in targeted drug delivery systems (DDSs) but also candidate them as macromolecules that inhibit the interaction of the target ligand with other proteins. On the other hand, the conjugation of Aps with another therapeutic molecule such as antisense oligonucleotides (ASOs), siRNAs/miRNAs, Aps , toxins, chemotherapeutic agents, DNAzymes/Ribozymes provides hopeful strategy to eradicate the malignancies and overcome the off-target unwanted side effects. Such prominent features of Aps make them a promising treatment modality to overcome the tumor complexity and heterogeneity, which can be consequently applied for personalized therapy of cancer by using bispecific Ap-based therapeutics.
Collapse
Affiliation(s)
- Somayeh Vandghanooni
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Eskandani
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jaleh Barar
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Omidi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
57
|
Citartan M, Kaur H, Presela R, Tang TH. Aptamers as the chaperones (Aptachaperones) of drugs-from siRNAs to DNA nanorobots. Int J Pharm 2019; 567:118483. [PMID: 31260780 DOI: 10.1016/j.ijpharm.2019.118483] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/25/2019] [Accepted: 06/27/2019] [Indexed: 12/16/2022]
Abstract
Aptamers, nucleic acid ligands that are specific against their corresponding targets are increasingly employed in a variety of applications including diagnostics and therapeutics. The specificity of the aptamers against their targets is also used as the basis for the formulation of the aptamer-based drug delivery system. In this review, we aim to provide an overview on the chaperoning roles of aptamers in acting as the cargo or load carriers, delivering contents to the targeted sites via cell surface receptors. Internalization of the aptamer-biomolecule conjugates via receptor-mediated endocytosis and the strategies to augment the rate of endocytosis are underscored. The cargos chaperoned by aptamers, ranging from siRNAs to DNA origami are illuminated. Possible impediments to the aptamer-based drug deliveries such as susceptibility to nuclease resistance, potentiality for immunogenicity activation, tumor heterogeneity are speculated and the corresponding amendment strategies to address these shortcomings are discussed. We prophesy that the future of the aptamer-based drug delivery will take a trajectory towards DNA nanorobot-based assay.
Collapse
Affiliation(s)
- Marimuthu Citartan
- Advanced Medical & Dental Institute (AMDI), Universiti Sains Malaysia, Bertam, 13200 Kepala Batas, Penang, Malaysia.
| | - Harleen Kaur
- Aurobindo Biologics, Biologics R&D Center, Unit-17, Industrial Area, Survey No: 77 & 78, Indrakaran Village, Kandi(Mandal), Sangareddy (District), Hyderabad 502329, India
| | - Ravinderan Presela
- Advanced Medical & Dental Institute (AMDI), Universiti Sains Malaysia, Bertam, 13200 Kepala Batas, Penang, Malaysia
| | - Thean-Hock Tang
- Advanced Medical & Dental Institute (AMDI), Universiti Sains Malaysia, Bertam, 13200 Kepala Batas, Penang, Malaysia.
| |
Collapse
|
58
|
Hong S, Ding P, Luo Y, Gao T, Zhang Y, Pei R. Aptamer-integrated α-Gal liposomes as bispecific agents to trigger immune response for killing tumor cells. J Biomed Mater Res A 2019; 107:1176-1183. [PMID: 30650243 DOI: 10.1002/jbm.a.36609] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 12/03/2018] [Accepted: 01/09/2019] [Indexed: 12/15/2022]
Abstract
A novel bispecific α-Gal liposome was constructed by self-assembling AS1411 aptamers into the α-Gal containing liposomes. The α-Gal liposomes were prepared using cell membranes of red blood cells from rabbit, which are composed of cholesterol, phospholipids, and α-Gal glycolipids. AS1411 is a DNA aptamer with high specificity and affinity for nucleolin and could integrate into liposomes by the modification of cholesterol. The bispecific α-Gal liposomes surface-functionalized by α-Gal and AS1411 aptamer could recognize anti-Gal antibodies and nucleolin overexpressed by tumor cells simultaneously, followed by activating the immune system to attack the tumor cells, resulting in the lysis of the tumor cells by antibody dependent cell-mediated cytotoxicity. Under simulated tumor environment, the lysis rate of MCF-7 cells treated by the AS1411 modified α-Gal liposomes drastically increased compared to the liposomes without AS1411 aptamer. This study suggests that the AS1411 modified α-Gal liposomes can recognize nucleolin-overexpressing tumor cells selectively, subsequently improve the effect of the immunotherapy with high specificity. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 1176-1183, 2019.
Collapse
Affiliation(s)
- Shanni Hong
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei, China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China
| | - Pi Ding
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei, China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China
| | - Yu Luo
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China
| | - Tian Gao
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei, China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China
| | - Ye Zhang
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China
| | - Renjun Pei
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei, China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China
| |
Collapse
|
59
|
Ueki R, Atsuta S, Ueki A, Hoshiyama J, Li J, Hayashi Y, Sando S. DNA aptamer assemblies as fibroblast growth factor mimics and their application in stem cell culture. Chem Commun (Camb) 2019; 55:2672-2675. [PMID: 30746545 DOI: 10.1039/c8cc08080a] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Replacing expensive and thermally unstable growth factors with synthetic alternatives has been an important issue in stem cell-based regenerative medicines. Here we developed DNA aptamer-assemblies that act as functional mimics of basic fibroblast growth factor (bFGF), one of the essential factors for stem cell culture. The most potent aptamer assembly named TD0, composed solely of 76-mer single-stranded DNA, could support the self-renewal and pluripotency of induced pluripotent stem cells (iPSCs). This work presents the first application of DNA aptamer in the maintenance of iPSCs.
Collapse
Affiliation(s)
- Ryosuke Ueki
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, Tokyo 113-8656, Japan.
| | | | | | | | | | | | | |
Collapse
|
60
|
Li Z, Hu Y, An Y, Duan J, Li X, Yang XD. Novel Bispecific Aptamer Enhances Immune Cytotoxicity Against MUC1-Positive Tumor Cells by MUC1-CD16 Dual Targeting. Molecules 2019; 24:molecules24030478. [PMID: 30699986 PMCID: PMC6385031 DOI: 10.3390/molecules24030478] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 01/25/2019] [Accepted: 01/28/2019] [Indexed: 01/05/2023] Open
Abstract
A promising strategy in cancer immunotherapy is the employment of a bispecific agent that can bind with both tumor markers and immunocytes for recruitment of lymphocytes to tumor sites and enhancement of anticancer immune reactions. Mucin1 (MUC1) is a tumor marker overexpressed in almost all adenocarcinomas, making it a potentially important therapeutic target. CD16 is expressed in several types of immunocytes, including NK cells, γδ-T cells, monocytes, and macrophages. In this study, we constructed the first bispecific aptamer (BBiApt) targeting both MUC1 and CD16. This aptamer consisted of two MUC1 aptamers and two CD16 aptamers linked together by three 60 nt DNA spacers. Compared with monovalent MUC1 or CD16 aptamers, BBiApt showed more potent avidity to both MUC1-positive tumor cells and CD16-positive immunocytes. Competition experiments indicated that BBiApt and monovalent aptamers bound to the same sites on the target cells. Moreover, BBiApt recruited more CD16-positive immunocytes around MUC1-positive tumor cells and enhanced the immune cytotoxicity against the tumor cells in vitro. The results suggest that, apart from bispecific antibodies, bispecific aptamers may also potentially serve as a novel strategy for targeted enhancement of antitumor immune reactions against MUC1-expressing malignancies.
Collapse
Affiliation(s)
- Zhaoyi Li
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China.
| | - Yan Hu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China.
| | - Yacong An
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China.
| | - Jinhong Duan
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China.
| | - Xundou Li
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China.
| | - Xian-Da Yang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China.
| |
Collapse
|
61
|
Wang L, Li W, Sun J, Zhang SY, Yang S, Li J, Li J, Yang HH. Imaging of Receptor Dimers in Zebrafish and Living Cells via Aptamer Recognition and Proximity-Induced Hybridization Chain Reaction. Anal Chem 2018; 90:14433-14438. [PMID: 30444610 DOI: 10.1021/acs.analchem.8b04015] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
On cell-membrane surfaces, receptor-protein dimers play fundamental roles in many signaling pathways that are crucial for normal biological processes and cancer development. Efficient and sensitive analysis of receptor dimers in the native environment is highly desirable. Herein, we present a strategy for amplified imaging of receptor dimers in zebrafish and living cells that relies on aptamer recognition and proximity-induced hybridization chain reaction. Taking advantage of specific aptamer recognition and enzyme-free signal amplification, this strategy is successfully applied to the visualization of c-Met-receptor dimers in an HGF-independent or -dependent manner. Therefore, the developed imaging strategy paves the way for further investigation of the dimerization or oligomerization states of cell-surface receptors and their corresponding activation processes in zebrafish and living cells.
Collapse
Affiliation(s)
- Liping Wang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry , Fuzhou University , Fuzhou 350116 , People's Republic of China.,Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai Jiao Tong University , Shanghai 200240 , People's Republic of China
| | - Wei Li
- College of Biological Science and Engineering , Fuzhou University , Fuzhou 350116 , People's Republic of China
| | - Jin Sun
- College of Biological Science and Engineering , Fuzhou University , Fuzhou 350116 , People's Republic of China
| | - Su-Yun Zhang
- Department of Medical Oncology , Fujian Medical University Union Hospital , Fuzhou 350001 , People's Republic of China
| | - Sheng Yang
- Department of Medical Oncology , Fujian Medical University Union Hospital , Fuzhou 350001 , People's Republic of China
| | - Jingying Li
- College of Biological Science and Engineering , Fuzhou University , Fuzhou 350116 , People's Republic of China
| | - Juan Li
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry , Fuzhou University , Fuzhou 350116 , People's Republic of China.,Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai Jiao Tong University , Shanghai 200240 , People's Republic of China
| | - Huang-Hao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry , Fuzhou University , Fuzhou 350116 , People's Republic of China
| |
Collapse
|
62
|
Kim J, Jang D, Park H, Jung S, Kim DH, Kim WJ. Functional-DNA-Driven Dynamic Nanoconstructs for Biomolecule Capture and Drug Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1707351. [PMID: 30062803 DOI: 10.1002/adma.201707351] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 03/13/2018] [Indexed: 06/08/2023]
Abstract
The discovery of sequence-specific hybridization has allowed the development of DNA nanotechnology, which is divided into two categories: 1) structural DNA nanotechnology, which utilizes DNA as a biopolymer; and 2) dynamic DNA nanotechnology, which focuses on the catalytic reactions or displacement of DNA structures. Recently, numerous attempts have been made to combine DNA nanotechnologies with functional DNAs such as aptamers, DNAzymes, amplified DNA, polymer-conjugated DNA, and DNA loaded on functional nanoparticles for various applications; thus, the new interdisciplinary research field of "functional DNA nanotechnology" is initiated. In particular, a fine-tuned nanostructure composed of functional DNAs has shown immense potential as a programmable nanomachine by controlling DNA dynamics triggered by specific environments. Moreover, the programmability and predictability of functional DNA have enabled the use of DNA nanostructures as nanomedicines for various biomedical applications, such as cargo delivery and molecular drugs via stimuli-mediated dynamic structural changes of functional DNAs. Here, the concepts and recent case studies of functional DNA nanotechnology and nanostructures in nanomedicine are reviewed, and future prospects of functional DNA for nanomedicine are indicated.
Collapse
Affiliation(s)
- Jinhwan Kim
- Center for Self-Assembly and Complexity, Institute for Basic Science (IBS), Pohang, 37673, Korea
| | - Donghyun Jang
- Department of Chemistry, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Korea
| | - Hyeongmok Park
- Department of Chemistry, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Korea
| | - Sungjin Jung
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Korea
| | - Dae Heon Kim
- Department of Biology, Sunchon National University, Sunchon, 57922, Korea
| | - Won Jong Kim
- Center for Self-Assembly and Complexity, Institute for Basic Science (IBS), Pohang, 37673, Korea
- Department of Chemistry, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Korea
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Korea
| |
Collapse
|
63
|
Zhang Y, Gao H, Zhou W, Sun S, Zeng Y, Zhang H, Liang L, Xiao X, Song J, Ye M, Yang Y, Zhao J, Wang Z, Liu J. Targeting c-met receptor tyrosine kinase by the DNA aptamer SL1 as a potential novel therapeutic option for myeloma. J Cell Mol Med 2018; 22:5978-5990. [PMID: 30353654 PMCID: PMC6237600 DOI: 10.1111/jcmm.13870] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 08/03/2018] [Indexed: 12/13/2022] Open
Abstract
Hepatocyte growth factor (HGF)/c‐met pathway activation has been implicated in the pathogenesis of multiple myeloma (MM), and blocking this pathway has been considered a rational therapeutic strategy for treating MM. Aptamers are single‐stranded nucleic acid molecules that fold into complex 3D structures and bind to a variety of targets. Recently, it was reported that DNA aptamer SL1 exhibited high specificity and affinity for c‐met and inhibited HGF/c‐met signaling in SNU‐5 cells. However, as the first c‐met‐targeted DNA aptamer to be identified, application of SL1 to myeloma treatment requires further investigation. Here, we explore the potential application of SL1 in MM. Our results indicated that c‐met expression is gradually increased in MM patients and contributes to poor outcomes. SL1 selectively bound to c‐met‐positive MM cells but not to normal B cells and suppressed the growth, migration and adhesion of MM cells in vitro in a co‐culture model performed with HS5 cells, wherein SL1 inhibited HGF‐induced activation of c‐met signaling. In vivo and ex vivo fluorescence imaging showed that SL1 accumulated in the c‐met positive tumour areas. In addition, SL1 was active against CD138+ primary MM cells and displayed a synergistic inhibition effect with bortezomib. Collectively, our data suggested that SL1 could be beneficial as a c‐met targeted antagonist in MM.
Collapse
Affiliation(s)
- Yibin Zhang
- Key Laboratory of Nanobiological Technology of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, China.,Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China.,Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, China
| | - Hongmei Gao
- Nursing Department, Xiangya Hospital, Central South University, Changsha, China
| | - Weihua Zhou
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Sunming Sun
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Yayue Zeng
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Hui Zhang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, China
| | - Long Liang
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Xiaojuan Xiao
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Jianhui Song
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Mao Ye
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, China
| | - Yujia Yang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Jingfeng Zhao
- Key Laboratory of Nanobiological Technology of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, China
| | - Zi Wang
- Key Laboratory of Nanobiological Technology of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, China.,Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Jing Liu
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| |
Collapse
|
64
|
Soldevilla MM, Meraviglia-Crivelli de Caso D, Menon AP, Pastor F. Aptamer-iRNAs as Therapeutics for Cancer Treatment. Pharmaceuticals (Basel) 2018; 11:E108. [PMID: 30340426 PMCID: PMC6315413 DOI: 10.3390/ph11040108] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 10/04/2018] [Accepted: 10/10/2018] [Indexed: 02/07/2023] Open
Abstract
Aptamers are single-stranded oligonucleotides (ssDNA or ssRNA) that bind and recognize their targets with high affinity and specificity due to their complex tertiary structure. Aptamers are selected by a method called SELEX (Systematic Evolution of Ligands by EXponential enrichment). This method has allowed the selection of aptamers to different types of molecules. Since then, many aptamers have been described for the potential treatment of several diseases including cancer. It has been described over the last few years that aptamers represent a very useful tool as therapeutics, especially for cancer therapy. Aptamers, thanks to their intrinsic oligonucleotide nature, present inherent advantages over other molecules, such as cell-based products. Owing to their higher tissue penetrability, safer profile, and targeting capacity, aptamers are likely to become a novel platform for the delivery of many different types of therapeutic cargos. Here we focus the review on interfering RNAs (iRNAs) as aptamer-based targeting delivered agents. We have gathered the most reliable information on aptamers as targeting and carrier agents for the specific delivery of siRNAs, shRNA, microRNAs, and antisense oligonucleotides (ASOs) published in the last few years in the context of cancer therapy.
Collapse
Affiliation(s)
- Mario M Soldevilla
- Molecular Therapy Program, Aptamer Core, Center for the Applied Medical Research (CIMA), University of Navarra (UNAV), 31008 Pamplona, Spain.
- Navarre Health Research Institute (IdiSNA), 31008 Pamplona, Spain.
| | - Daniel Meraviglia-Crivelli de Caso
- Molecular Therapy Program, Aptamer Core, Center for the Applied Medical Research (CIMA), University of Navarra (UNAV), 31008 Pamplona, Spain.
- Navarre Health Research Institute (IdiSNA), 31008 Pamplona, Spain.
| | - Ashwathi P Menon
- Molecular Therapy Program, Aptamer Core, Center for the Applied Medical Research (CIMA), University of Navarra (UNAV), 31008 Pamplona, Spain.
- Navarre Health Research Institute (IdiSNA), 31008 Pamplona, Spain.
| | - Fernando Pastor
- Molecular Therapy Program, Aptamer Core, Center for the Applied Medical Research (CIMA), University of Navarra (UNAV), 31008 Pamplona, Spain.
- Navarre Health Research Institute (IdiSNA), 31008 Pamplona, Spain.
| |
Collapse
|
65
|
Batool S, Argyropoulos KV, Azad R, Okeoma P, Zumrut H, Bhandari S, Dekhang R, Mallikaratchy PR. Dimerization of an aptamer generated from Ligand-guided selection (LIGS) yields a high affinity scaffold against B-cells. Biochim Biophys Acta Gen Subj 2018; 1863:232-240. [PMID: 30342154 DOI: 10.1016/j.bbagen.2018.10.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 09/13/2018] [Accepted: 10/04/2018] [Indexed: 12/18/2022]
Abstract
Nucleic Acid Aptamers (NAAs) are a class of synthetic DNA or RNA molecules that bind specifically to their target. We recently introduced an aptamer termed R1.2 against membrane Immunoglobulin M (mIgM) expressing B-cell neoplasms using Ligand Guided Selection (LIGS). While LIGS-generated aptamers are highly specific, their lower affinity prevents aptamers from being used for translational applications. Highly specific aptamers with higher affinity can increase targetability, boosting the application of aptamers as diagnostic and therapeutic molecules. Herein, we report that dimerization of R1.2, an aptamer generated from LIGS, leads to high affinity variants without compromising the specificity. Three dimeric aptamer analogues with variable linker lengths were designed to evaluate the effect of linker length in affinity. The optimized dimeric R1.2 against cultured B-cell neoplasms, four donor B-cell samples and mIgM-positive Waldenström's Macroglobulinemia (WM) showed specificity. Furthermore, confocal imaging of dimeric aptamer and anti-IgM antibody in purified B-cells suggests co-localization. Binding assays against IgM knockout Burkitt's Lymphoma cells utilizing CRISPR/Cas9 further validated specificity of dimeric R1.2. Collectively, our findings show that LIGS-generated aptamers can be re-engineered into dimeric aptamers with high specificity and affinity, demonstrating wide-range of applicability of LIGS in developing clinically practical diagnostic and therapeutic aptamers.
Collapse
Affiliation(s)
- Sana Batool
- Department of Chemistry, Lehman College, The City University of New York, 250 Bedford Park Blvd, NY 10468, USA
| | - Kimon V Argyropoulos
- Immunology Program, Memorial Sloan Kettering Cancer Center, 408 E69th street, New York, NY, 10021, USA
| | - Roksana Azad
- Ph.D. Program in Chemistry and Biochemistry, CUNY Graduate Center, 365 Fifth Avenue, New York, NY 10016, USA
| | - Precious Okeoma
- Department of Chemistry, Lehman College, The City University of New York, 250 Bedford Park Blvd, NY 10468, USA
| | - Hasan Zumrut
- Ph.D. Program in Chemistry and Biochemistry, CUNY Graduate Center, 365 Fifth Avenue, New York, NY 10016, USA
| | - Sanam Bhandari
- Department of Chemistry, Lehman College, The City University of New York, 250 Bedford Park Blvd, NY 10468, USA
| | - Rigzin Dekhang
- Department of Chemistry, Lehman College, The City University of New York, 250 Bedford Park Blvd, NY 10468, USA
| | - Prabodhika R Mallikaratchy
- Department of Chemistry, Lehman College, The City University of New York, 250 Bedford Park Blvd, NY 10468, USA; Ph.D. Program in Chemistry and Biochemistry, CUNY Graduate Center, 365 Fifth Avenue, New York, NY 10016, USA; Ph.D. Program in Molecular, Cellular and Developmental Biology, CUNY Graduate Center, 365 Fifth Avenue, New York, NY 10016, USA.
| |
Collapse
|
66
|
Chen S, Li J, Liang H, Lin XH, Li J, Yang HH. Light-Induced Activation of c-Met Signalling by Photocontrolled DNA Assembly. Chemistry 2018; 24:15988-15992. [PMID: 30155946 DOI: 10.1002/chem.201803868] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Indexed: 12/19/2022]
Abstract
Optical manipulation appears to be a powerful tool for spatiotemporally controlling a variety of cellular functions. Herein, a photocontrolled DNA assembly approach is described which enables light-induced activation of cellular signal transduction by triggering protein dimerization (c-Met signalling in this case). Three kinds of DNA probes are designed, including a pair of receptor recognition probes with adaptors and a blocker probe with a photocleavable linker (PC-linker). By implementing PC-linkers in blocker probes, the designed DNA probes response to light irradiation, which then induces the assembly of receptor recognition probes through adaptor complementing. Consequently, light-mediated DNA assembly promotes the dimerization of c-Met receptors, resulting in activation of c-Met signalling. It is demonstrated that the proposed photocontrolled DNA assembly approach is effective for regulating c-Met signalling and modulating cellular behaviours, such as cell proliferation and migration. Therefore, this simple approach may offer a promising strategy to manipulate cell signalling pathways precisely in living cells.
Collapse
Affiliation(s)
- Shan Chen
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350116, P. R. China
| | - Jingying Li
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350116, P. R. China
| | - Hong Liang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350116, P. R. China
| | - Xia-Hui Lin
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350116, P. R. China
| | - Juan Li
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350116, P. R. China
| | - Huang-Hao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350116, P. R. China
| |
Collapse
|
67
|
Vandghanooni S, Eskandani M, Barar J, Omidi Y. Bispecific therapeutic aptamers for targeted therapy of cancer: a review on cellular perspective. J Mol Med (Berl) 2018; 96:885-902. [PMID: 30056527 DOI: 10.1007/s00109-018-1669-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 06/03/2018] [Accepted: 07/09/2018] [Indexed: 12/15/2022]
Abstract
Aptamers (Aps), as short single-strand nucleic acids, can bind to their corresponding molecular targets with the high affinity and specificity. In comparison with the monoclonal antibodies (mAbs) and peptides, unique physicochemical and biological characteristics of Aps make them excellent targeting agents for different types of cancer molecular markers (CMMs). Much attention has been paid to the Ap-based multifunctional chimeric and therapeutic systems, which provide promising outcomes in the targeted therapy of various formidable diseases, including malignancies. In the Ap-based chimeric systems, a targeting Ap is conjugated to another therapeutic molecule (e.g., siRNA/miRNA, Ap, toxins, chemotherapeutic agents, DNAzyme/ribozymes) with a capability of binding to a specific cell surface receptor at the desired target site. Having been engineered as multifunctional nanosystems (NSs), Ap-based hybrid scaffolds can be used to concurrently target multiple markers/pathways in cancerous cells, causing drastic inhibitory effects on the growth and the progression of tumor cells. Multi/bispecific Aps composed of two/more Aps provide a versatile tool for the optimal and active targeting of cell surface receptor(s) with markedly high affinity and avidity. Targeting the optimum activity of key receptors and dominant signaling pathways in the activation of immunity, the multi/bispecific Ap-based therapeutics can also be used to enhance the antitumor activity of the immune system. Further, the bispecific systems can be designed to induce cytotoxicity in a heterogeneous population of cancer cells with different CMMs. In this review, we provide some important insights into the construction and applications of the Ap-based chimeric NSs and discuss the multifunctional Ap chimera and their effects on the signaling pathways in cancer.
Collapse
Affiliation(s)
- Somayeh Vandghanooni
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Eskandani
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jaleh Barar
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Omidi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
68
|
Li H, Wang M, Shi T, Yang S, Zhang J, Wang HH, Nie Z. A DNA-Mediated Chemically Induced Dimerization (D-CID) Nanodevice for Nongenetic Receptor Engineering To Control Cell Behavior. Angew Chem Int Ed Engl 2018; 57:10226-10230. [PMID: 29944203 DOI: 10.1002/anie.201806155] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Indexed: 12/31/2022]
Abstract
Small-molecule regulation is a powerful switching tool to manipulate cell signal transduction for a desired function; however, most available methods usually require genetic engineering to endow cells with responsiveness to user-defined small molecules. Herein, we demonstrate a nongenetic approach for small-molecule-controlled receptor activation and consequent cell behavior manipulation that is based on DNA-mediated chemically induced dimerization (D-CID). D-CID uses a programmable chemical-responsive DNA nanodevice to trigger DNA strand displacement and induce the activation of c-Met, a tyrosine kinase receptor cognate for hepatocyte growth factor, through dimerization. Through the use of various functional nucleic acids, including aptamers and DNAzymes, as recognition modules, the versatility of D-CID in inducing c-Met signaling upon addition of various small-molecular or ionic cues, including ATP, histidine, and Zn2+ , is demonstrated. Moreover, owing its multi-input properties, D-CID can be used to manipulate the behaviors of multiple cell populations simultaneously in a selective and programmable fashion.
Collapse
Affiliation(s)
- Hao Li
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan University, Changsha, 410082, P. R. China
| | - Miao Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan University, Changsha, 410082, P. R. China
| | - Tianhui Shi
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan University, Changsha, 410082, P. R. China
| | - Sihui Yang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan University, Changsha, 410082, P. R. China
| | - Jinghui Zhang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan University, Changsha, 410082, P. R. China
| | - Hong-Hui Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan University, Changsha, 410082, P. R. China
| | - Zhou Nie
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan University, Changsha, 410082, P. R. China
| |
Collapse
|
69
|
Li H, Wang M, Shi T, Yang S, Zhang J, Wang HH, Nie Z. A DNA-Mediated Chemically Induced Dimerization (D-CID) Nanodevice for Nongenetic Receptor Engineering To Control Cell Behavior. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201806155] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Hao Li
- State Key Laboratory of Chemo/Biosensing and Chemometrics; College of Chemistry and Chemical Engineering; College of Biology; Hunan University; Changsha 410082 P. R. China
| | - Miao Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics; College of Chemistry and Chemical Engineering; College of Biology; Hunan University; Changsha 410082 P. R. China
| | - Tianhui Shi
- State Key Laboratory of Chemo/Biosensing and Chemometrics; College of Chemistry and Chemical Engineering; College of Biology; Hunan University; Changsha 410082 P. R. China
| | - Sihui Yang
- State Key Laboratory of Chemo/Biosensing and Chemometrics; College of Chemistry and Chemical Engineering; College of Biology; Hunan University; Changsha 410082 P. R. China
| | - Jinghui Zhang
- State Key Laboratory of Chemo/Biosensing and Chemometrics; College of Chemistry and Chemical Engineering; College of Biology; Hunan University; Changsha 410082 P. R. China
| | - Hong-Hui Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics; College of Chemistry and Chemical Engineering; College of Biology; Hunan University; Changsha 410082 P. R. China
| | - Zhou Nie
- State Key Laboratory of Chemo/Biosensing and Chemometrics; College of Chemistry and Chemical Engineering; College of Biology; Hunan University; Changsha 410082 P. R. China
| |
Collapse
|
70
|
Oligonucleotide aptamers against tyrosine kinase receptors: Prospect for anticancer applications. Biochim Biophys Acta Rev Cancer 2018; 1869:263-277. [PMID: 29574128 DOI: 10.1016/j.bbcan.2018.03.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 03/19/2018] [Accepted: 03/20/2018] [Indexed: 02/07/2023]
Abstract
Transmembrane receptor tyrosine kinases (RTKs) play crucial roles in cancer cell proliferation, survival, migration and differentiation. Area of intense research is searching for effective anticancer therapies targeting these receptors and, to date, several monoclonal antibodies and small-molecule tyrosine kinase inhibitors have entered the clinic. However, some of these drugs show limited efficacy and give rise to acquired resistance. Emerging highly selective compounds for anticancer therapy are oligonucleotide aptamers that interact with their targets by recognizing a specific three-dimensional structure. Because of their nucleic acid nature, the rational design of advanced strategies to manipulate aptamers for both diagnostic and therapeutic applications is greatly simplified over antibodies. In this manuscript, we will provide a comprehensive overview of oligonucleotide aptamers as next generation strategies to efficiently target RTKs in human cancers.
Collapse
|
71
|
Liang H, Chen S, Li P, Wang L, Li J, Li J, Yang HH, Tan W. Nongenetic Approach for Imaging Protein Dimerization by Aptamer Recognition and Proximity-Induced DNA Assembly. J Am Chem Soc 2018. [PMID: 29522674 DOI: 10.1021/jacs.7b11311] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Herein, we report a nongenetic and real-time approach for imaging protein dimerization on living cell surfaces by aptamer recognition and proximity-induced DNA assembly. We use the aptamer specific for the receptor monomer as a recognition probe. When receptor dimerization occurs, the dimeric receptors bring two aptamer probes into close proximity, thereby triggering dynamic DNA assembly. The proposed approach was successfully applied to visualize dimerization of Met receptor and transforming growth factor-β type II receptor. This approach allows us to image the two states (monomer/dimer) of a receptor protein on living cell surfaces in real time, opening a universal method for further investigation of protein dimerization and the corresponding activation processes in signal transduction.
Collapse
Affiliation(s)
- Hong Liang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry , Fuzhou University , Fuzhou 350116 , People's Republic of China
| | - Shan Chen
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry , Fuzhou University , Fuzhou 350116 , People's Republic of China
| | - Peipei Li
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry , Fuzhou University , Fuzhou 350116 , People's Republic of China
| | - Liping Wang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry , Fuzhou University , Fuzhou 350116 , People's Republic of China
| | - Jingying Li
- College of Biological Science and Engineering , Fuzhou University , Fuzhou 350116 , People's Republic of China
| | - Juan Li
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry , Fuzhou University , Fuzhou 350116 , People's Republic of China.,Institute of Molecular Medicine, Renji Hospital , Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University , Shanghai , 200240 , People's Republic of China
| | - Huang-Hao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry , Fuzhou University , Fuzhou 350116 , People's Republic of China.,College of Biological Science and Engineering , Fuzhou University , Fuzhou 350116 , People's Republic of China
| | - Weihong Tan
- Institute of Molecular Medicine, Renji Hospital , Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University , Shanghai , 200240 , People's Republic of China.,Department of Chemistry and Department of Physiology and Functional Genomics, Center for Research at the Bio/Nano Interface, UF Health Cancer Center , University of Florida , Gainesville , Florida 32611-7200 , United States
| |
Collapse
|
72
|
Delley CL, Liu L, Sarhan MF, Abate AR. Combined aptamer and transcriptome sequencing of single cells. Sci Rep 2018; 8:2919. [PMID: 29440771 PMCID: PMC5811598 DOI: 10.1038/s41598-018-21153-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 01/29/2018] [Indexed: 01/09/2023] Open
Abstract
The transcriptome and proteome encode distinct information that is important for characterizing heterogeneous biological systems. We demonstrate a method to simultaneously characterize the transcriptomes and proteomes of single cells at high throughput using aptamer probes and droplet-based single cell sequencing. With our method, we differentiate distinct cell types based on aptamer surface binding and gene expression patterns. Aptamers provide advantages over antibodies for single cell protein characterization, including rapid, in vitro, and high-purity generation via SELEX, and the ability to amplify and detect them with PCR and sequencing.
Collapse
Affiliation(s)
- Cyrille L Delley
- Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, 94158, California, USA
| | - Leqian Liu
- Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, 94158, California, USA
| | - Maen F Sarhan
- Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, 94158, California, USA
| | - Adam R Abate
- Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, 94158, California, USA. .,California Institute for Quantitative Biosciences, University of California San Francisco, San Francisco, 94158, California, USA. .,Chan Zuckerberg Biohub, San Francisco, 94158, California, USA.
| |
Collapse
|
73
|
Prusty DK, Adam V, Zadegan RM, Irsen S, Famulok M. Supramolecular aptamer nano-constructs for receptor-mediated targeting and light-triggered release of chemotherapeutics into cancer cells. Nat Commun 2018; 9:535. [PMID: 29416033 PMCID: PMC5803212 DOI: 10.1038/s41467-018-02929-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 01/09/2018] [Indexed: 12/01/2022] Open
Abstract
Platforms for targeted drug-delivery must simultaneously exhibit serum stability, efficient directed cell internalization, and triggered drug release. Here, using lipid-mediated self-assembly of aptamers, we combine multiple structural motifs into a single nanoconstruct that targets hepatocyte growth factor receptor (cMet). The nanocarrier consists of lipidated versions of a cMet-binding aptamer and a separate lipidated GC-rich DNA hairpin motif loaded with intercalated doxorubicin. Multiple 2',6'-dimethylazobenzene moieties are incorporated into the doxorubicin-binding motif to trigger the release of the chemotherapeutics by photoisomerization. The lipidated DNA scaffolds self-assemble into spherical hybrid-nanoconstructs that specifically bind cMet. The combined features of the nanocarriers increase serum nuclease resistance, favor their import into cells presumably mediated by endocytosis, and allow selective photo-release of the chemotherapeutic into the targeted cells. cMet-expressing H1838 tumor cells specifically internalize drug-loaded nanoconstructs, and subsequent UV exposure enhances cell mortality. This modular approach thus paves the way for novel classes of powerful aptamer-based therapeutics.
Collapse
Affiliation(s)
- Deepak K Prusty
- Life and Medical Sciences (LIMES) Institute, Chemical Biology & Medicinal Chemistry Unit, c/o Kekulé Institute of Organic Chemistry and Biochemistry, Gerhard-Domagk-Strasse 1, 53121, Bonn, Germany
- Stiftung Caesar, Max-Planck-Fellowship Group Chemical Biology, Ludwig-Erhard-Allee 2, 53175, Bonn, Germany
| | - Volker Adam
- Life and Medical Sciences (LIMES) Institute, Chemical Biology & Medicinal Chemistry Unit, c/o Kekulé Institute of Organic Chemistry and Biochemistry, Gerhard-Domagk-Strasse 1, 53121, Bonn, Germany
| | - Reza M Zadegan
- Nanoscale Materials & Device Group, Micron School of Materials Science and Engineering, Boise State University, Boise, USA
| | - Stephan Irsen
- Stiftung Caesar, Elektronenmikroskopie und Analytik, Ludwig-Erhard-Allee 2, 53175, Bonn, Germany
| | - Michael Famulok
- Life and Medical Sciences (LIMES) Institute, Chemical Biology & Medicinal Chemistry Unit, c/o Kekulé Institute of Organic Chemistry and Biochemistry, Gerhard-Domagk-Strasse 1, 53121, Bonn, Germany.
- Stiftung Caesar, Max-Planck-Fellowship Group Chemical Biology, Ludwig-Erhard-Allee 2, 53175, Bonn, Germany.
| |
Collapse
|
74
|
Hori SI, Herrera A, Rossi JJ, Zhou J. Current Advances in Aptamers for Cancer Diagnosis and Therapy. Cancers (Basel) 2018; 10:cancers10010009. [PMID: 29301363 PMCID: PMC5789359 DOI: 10.3390/cancers10010009] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 12/22/2017] [Accepted: 12/26/2017] [Indexed: 12/24/2022] Open
Abstract
Nucleic acid aptamers are single-stranded oligonucleotides that interact with target molecules with high affinity and specificity in unique three-dimensional structures. Aptamers are generally isolated by a simple selection process called systematic evolution of ligands by exponential enrichment (SELEX) and then can be chemically synthesized and modified. Because of their high affinity and specificity, aptamers are promising agents for biomarker discovery, as well as cancer diagnosis and therapy. In this review, we present recent progress and challenges in aptamer and SELEX technology and highlight some representative applications of aptamers in cancer therapy.
Collapse
Affiliation(s)
- Shin-Ichiro Hori
- Department of Molecular and Cellular Biology, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd, Duarte, CA 91010, USA.
- Drug Discovery & Disease Research Laboratory, Shionogi & Co., Ltd., 3-1-1, Futaba-cho, Toyonaka, Osaka 561-0825, Japan.
| | - Alberto Herrera
- Department of Molecular and Cellular Biology, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd, Duarte, CA 91010, USA.
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd, Duarte, CA 91010, USA.
| | - John J Rossi
- Department of Molecular and Cellular Biology, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd, Duarte, CA 91010, USA.
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd, Duarte, CA 91010, USA.
| | - Jiehua Zhou
- Department of Molecular and Cellular Biology, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd, Duarte, CA 91010, USA.
| |
Collapse
|
75
|
Chen C, Zhou S, Cai Y, Tang F. Nucleic acid aptamer application in diagnosis and therapy of colorectal cancer based on cell-SELEX technology. NPJ Precis Oncol 2017; 1:37. [PMID: 29872716 PMCID: PMC5871892 DOI: 10.1038/s41698-017-0041-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 10/17/2017] [Accepted: 10/19/2017] [Indexed: 12/20/2022] Open
Abstract
Nucleic acid aptamers are a class of high-affinity nucleic acid ligands. They serve as “chemical antibodies” since their high affinity and specificity. Nucleic acid aptamers are generated from nucleic acid random-sequence using a systematic evolution of ligands by exponential enrichment (SELEX) technology. SELEX is a process of effectively selecting aptamers from different targets. A newly developed cell-based SELEX technique has been widely used in biomarker discovery, early diagnosis and targeted cancer therapy, particular at colorectal cancer (CRC). Combined with nanostructures, nano-aptamer-drug delivery system was constructed for drug delivery. Various nanostructures functionalized with aptamers are highly efficient and has been used in CRC therapeutic applications. In the present, we introduce a cell- SELEX technique, and summarize the potential application of aptamers as biomarkers in CRC diagnosis and therapy. And some characteristics of aptamer-targeted nanocarriers in CRC have been expatiated. The challenges and perspectives for cell-SELEX are also discussed.
Collapse
Affiliation(s)
- Chan Chen
- 1Clinical Laboratory and Medical Research Center, Zhuhai Hospital of Jinan University, Zhuhai People's Hospital, 519000 Zhuhai, Guangdong China
| | - Shan Zhou
- 1Clinical Laboratory and Medical Research Center, Zhuhai Hospital of Jinan University, Zhuhai People's Hospital, 519000 Zhuhai, Guangdong China
| | - Yongqiang Cai
- 1Clinical Laboratory and Medical Research Center, Zhuhai Hospital of Jinan University, Zhuhai People's Hospital, 519000 Zhuhai, Guangdong China
| | - Faqing Tang
- 1Clinical Laboratory and Medical Research Center, Zhuhai Hospital of Jinan University, Zhuhai People's Hospital, 519000 Zhuhai, Guangdong China.,2Clinical Laboratory, Hunan Cancer Hospital & The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 410006 Changsha, China
| |
Collapse
|
76
|
Batool S, Bhandari S, George S, Okeoma P, Van N, Zümrüt HE, Mallikaratchy P. Engineered Aptamers to Probe Molecular Interactions on the Cell Surface. Biomedicines 2017; 5:biomedicines5030054. [PMID: 28850067 PMCID: PMC5618312 DOI: 10.3390/biomedicines5030054] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 08/01/2017] [Accepted: 08/08/2017] [Indexed: 01/08/2023] Open
Abstract
Significant progress has been made in understanding the nature of molecular interactions on the cell membrane. To decipher such interactions, molecular scaffolds can be engineered as a tool to modulate these events as they occur on the cell membrane. To guarantee reliability, scaffolds that function as modulators of cell membrane events must be coupled to a targeting moiety with superior chemical versatility. In this regard, nucleic acid aptamers are a suitable class of targeting moieties. Aptamers are inherently chemical in nature, allowing extensive site-specific chemical modification to engineer sensing molecules. Aptamers can be easily selected using a simple laboratory-based in vitro evolution method enabling the design and development of aptamer-based functional molecular scaffolds against wide range of cell surface molecules. This article reviews the application of aptamers as monitors and modulators of molecular interactions on the mammalian cell surface with the aim of increasing our understanding of cell-surface receptor response to external stimuli. The information gained from these types of studies could eventually prove useful in engineering improved medical diagnostics and therapeutics.
Collapse
Affiliation(s)
- Sana Batool
- Department of Chemistry, Lehman College, The City University of New York, 250 Bedford Park Blvd. West, Bronx, New York, NY 10468, USA.
| | - Sanam Bhandari
- Department of Chemistry, Lehman College, The City University of New York, 250 Bedford Park Blvd. West, Bronx, New York, NY 10468, USA.
| | - Shanell George
- Department of Chemistry, Lehman College, The City University of New York, 250 Bedford Park Blvd. West, Bronx, New York, NY 10468, USA.
| | - Precious Okeoma
- Department of Chemistry, Lehman College, The City University of New York, 250 Bedford Park Blvd. West, Bronx, New York, NY 10468, USA.
| | - Nabeela Van
- Department of Chemistry, Lehman College, The City University of New York, 250 Bedford Park Blvd. West, Bronx, New York, NY 10468, USA.
| | - Hazan E Zümrüt
- Ph.D. Programs in Chemistry and Biochemistry, CUNY Graduate Center, 365 Fifth Avenue, New York, NY 10016, USA.
| | - Prabodhika Mallikaratchy
- Department of Chemistry, Lehman College, The City University of New York, 250 Bedford Park Blvd. West, Bronx, New York, NY 10468, USA.
- Ph.D. Programs in Chemistry and Biochemistry, CUNY Graduate Center, 365 Fifth Avenue, New York, NY 10016, USA.
- Ph.D. Program in Molecular, Cellular and Developmental Biology, CUNY Graduate Center, 365 Fifth Avenue, New York, NY 10016, USA.
| |
Collapse
|
77
|
Yokoyama T, Tsukakoshi K, Yoshida W, Saito T, Teramoto K, Savory N, Abe K, Ikebukuro K. Development of HGF-binding aptamers with the combination of G4 promoter-derived aptamer selection and in silico maturation. Biotechnol Bioeng 2017. [PMID: 28627727 DOI: 10.1002/bit.26354] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
We describe the selection of aptamers based on bioinformatics-based approaches without Systematic Evolution of Ligands by EXponential enrichment (SELEX). SELEX is a potent method; however, it is time intensive and the PCR-amplification step, which is essential step for SELEX, leads to the loss of good aptamers. We have developed an aptamer-screening method, G4 promoter-derived aptamer selection (G4PAS), and an aptamer-improving method, in silico maturation (ISM). They are based on in silico sequence selection and computer assisted directed evolution, respectively. In this study, we succeeded in identifying new aptamers against hepatocyte growth factor (HGF) by G4PAS as well as improving the specificity of the HGF aptamers by ISM. Using ISM improved the specificity of the aptamer for HGF by up to 45-fold in comparison with the original aptamer. These methods enable easy and efficient identification of good aptamers, and the combination of G4PAS with ISM can thus serve as a potent approach for aptamer identification. Biotechnol. Bioeng. 2017;114: 2196-2203. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Tomomi Yokoyama
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| | - Kaori Tsukakoshi
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| | - Wataru Yoshida
- School of Biotechnology and Bioscience, Tokyo University of Technology, Hachioji, Tokyo, Japan
| | - Taiki Saito
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| | - Kentaro Teramoto
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| | - Nasa Savory
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| | - Koichi Abe
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| | - Kazunori Ikebukuro
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| |
Collapse
|
78
|
Abstract
Aptamers are nucleic acids referred to as chemical antibodies as they bind to their specific targets with high affinity and selectivity. They are selected via an iterative process known as ‘selective evolution of ligands by exponential enrichment’ (SELEX). Aptamers have been developed against numerous cancer targets and among them, many tumor cell-membrane protein biomarkers. The identification of aptamers targeting cell-surface proteins has mainly been performed by two different strategies: protein- and cell-based SELEX, when the targets used for selection were proteins and cells, respectively. This review aims to update the literature on aptamers targeting tumor cell surface protein biomarkers, highlighting potentials, pitfalls of protein- and cell-based selection processes and applications of such selected molecules. Aptamers as promising agents for diagnosis and therapeutic approaches in oncology are documented, as well as aptamers in clinical development.
Collapse
|
79
|
Guillerey C, Huntington ND, Smyth MJ. Targeting natural killer cells in cancer immunotherapy. Nat Immunol 2017; 17:1025-36. [PMID: 27540992 DOI: 10.1038/ni.3518] [Citation(s) in RCA: 767] [Impact Index Per Article: 109.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Accepted: 06/22/2016] [Indexed: 12/14/2022]
Abstract
Alteration in the expression of cell-surface proteins is a common consequence of malignant transformation. Natural killer (NK) cells use an array of germline-encoded activating and inhibitory receptors that scan for altered protein-expression patterns, but tumor evasion of detection by the immune system is now recognized as one of the hallmarks of cancer. NK cells display rapid and potent immunity to metastasis or hematological cancers, and major efforts are now being undertaken to fully exploit NK cell anti-tumor properties in the clinic. Diverse approaches encompass the development of large-scale NK cell-expansion protocols for adoptive transfer, the establishment of a microenvironment favorable to NK cell activity, the redirection of NK cell activity against tumor cells and the release of inhibitory signals that limit NK cell function. In this Review we detail recent advances in NK cell-based immunotherapies and discuss the advantages and limitations of these strategies.
Collapse
Affiliation(s)
- Camille Guillerey
- Immunology of Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Australia.,School of Medicine, University of Queensland, Herston, Australia
| | - Nicholas D Huntington
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, The University of Melbourne, Melbourne, Australia
| | - Mark J Smyth
- Immunology of Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Australia.,School of Medicine, University of Queensland, Herston, Australia
| |
Collapse
|
80
|
Ueki R, Atsuta S, Ueki A, Sando S. Nongenetic Reprogramming of the Ligand Specificity of Growth Factor Receptors by Bispecific DNA Aptamers. J Am Chem Soc 2017; 139:6554-6557. [PMID: 28459560 DOI: 10.1021/jacs.7b02411] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The reprogramming of receptor-ligand interactions affords an opportunity to direct cells to respond to user-defined external cues. Although this has often been achieved via the genetic engineering of receptors, an alternative, nongenetic approach is highly demanded. In this article, we propose the design of oligonucleotide-based synthetic switches that feature the ability to reprogram the ligand specificity of the growth factor receptor. We demonstrated that our synthetic switches induced growth factor signaling via the formation of the dynamic complex with specific external cues that would otherwise not induce the signaling. This chemical approach may be applied to designing a new class of chemical tools that can control the activities of native cells and represent smart and safer regenerative drugs.
Collapse
Affiliation(s)
- Ryosuke Ueki
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo , 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Saki Atsuta
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo , 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Ayaka Ueki
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo , 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Shinsuke Sando
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo , 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| |
Collapse
|
81
|
Jasinski D, Haque F, Binzel DW, Guo P. Advancement of the Emerging Field of RNA Nanotechnology. ACS NANO 2017; 11:1142-1164. [PMID: 28045501 PMCID: PMC5333189 DOI: 10.1021/acsnano.6b05737] [Citation(s) in RCA: 222] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 01/03/2017] [Indexed: 05/14/2023]
Abstract
The field of RNA nanotechnology has advanced rapidly during the past decade. A variety of programmable RNA nanoparticles with defined shape, size, and stoichiometry have been developed for diverse applications in nanobiotechnology. The rising popularity of RNA nanoparticles is due to a number of factors: (1) removing the concern of RNA degradation in vitro and in vivo by introducing chemical modification into nucleotides without significant alteration of the RNA property in folding and self-assembly; (2) confirming the concept that RNA displays very high thermodynamic stability and is suitable for in vivo trafficking and other applications; (3) obtaining the knowledge to tune the immunogenic properties of synthetic RNA constructs for in vivo applications; (4) increased understanding of the 4D structure and intermolecular interaction of RNA molecules; (5) developing methods to control shape, size, and stoichiometry of RNA nanoparticles; (6) increasing knowledge of regulation and processing functions of RNA in cells; (7) decreasing cost of RNA production by biological and chemical synthesis; and (8) proving the concept that RNA is a safe and specific therapeutic modality for cancer and other diseases with little or no accumulation in vital organs. Other applications of RNA nanotechnology, such as adapting them to construct 2D, 3D, and 4D structures for use in tissue engineering, biosensing, resistive biomemory, and potential computer logic gate modules, have stimulated the interest of the scientific community. This review aims to outline the current state of the art of RNA nanoparticles as programmable smart complexes and offers perspectives on the promising avenues of research in this fast-growing field.
Collapse
Affiliation(s)
| | | | - Daniel W Binzel
- College of Pharmacy, Division
of Pharmaceutics and Pharmaceutical Chemistry; College of Medicine,
Department of Physiology & Cell Biology; and Dorothy M. Davis
Heart and Lung Research Institute, The Ohio
State University, Columbus, Ohio 43210, United States
| | - Peixuan Guo
- College of Pharmacy, Division
of Pharmaceutics and Pharmaceutical Chemistry; College of Medicine,
Department of Physiology & Cell Biology; and Dorothy M. Davis
Heart and Lung Research Institute, The Ohio
State University, Columbus, Ohio 43210, United States
| |
Collapse
|
82
|
Chen M, Yu Y, Jiang F, Zhou J, Li Y, Liang C, Dang L, Lu A, Zhang G. Development of Cell-SELEX Technology and Its Application in Cancer Diagnosis and Therapy. Int J Mol Sci 2016; 17:ijms17122079. [PMID: 27973403 PMCID: PMC5187879 DOI: 10.3390/ijms17122079] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 11/28/2016] [Accepted: 12/02/2016] [Indexed: 12/20/2022] Open
Abstract
SELEX (systematic evolution of ligands by exponential enrichment) is a process involving the progressive isolation of high selective ssDNA/RNA from a combinatorial single-stranded oligonucleotide library through repeated rounds of binding, partitioning and amplification. SELEX-derived single-stranded DNA/RNA molecules, called aptamers, are selected against a wide range of targets, including purified proteins, live cells, tissues, microorganisms, small molecules and so on. With the development of SELEX technology over the last two decades, various modified SELEX processes have been arisen. A majority of aptamers are selected against purified proteins through traditional SELEX. Unfortunately, more and more evidence showed aptamers selected against purified membrane proteins failed to recognize their targets in live cells. Cell-SELEX could develop aptamers against a particular target cell line to discriminate this cell line from others. Therefore, cell-SELEX has been widely used to select aptamers for the application of both diagnosis and therapy of various diseases, especially for cancer. In this review, the advantages and limitations of cell-SELEX and SELEX against purified protein will be compared. Various modified cell-SELEX techniques will be summarized, and application of cell-SELEX in cancer diagnosis and therapy will be discussed.
Collapse
Affiliation(s)
- Man Chen
- Institute of Integrated Bioinformedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong 999077, China.
- Institute of Precision Medicine and Innovative Drug Discovery, HKBU (Haimen) Institute of Science and Technology, Haimen 226100, China.
| | - Yuanyuan Yu
- Institute of Integrated Bioinformedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong 999077, China.
- Shenzhen Lab of Comninatorial Compounds and Targeted Drug Delivery, HKBU Institute of Research and Continuing Education, Shenzhen 518000, China.
| | - Feng Jiang
- Institute of Integrated Bioinformedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong 999077, China.
- Institute of Precision Medicine and Innovative Drug Discovery, HKBU (Haimen) Institute of Science and Technology, Haimen 226100, China.
- The State Key Laboratory Base of Novel Functional Materials and Preparation Science, Faculty of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, China.
| | - Junwei Zhou
- Institute of Integrated Bioinformedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong 999077, China.
| | - Yongshu Li
- Institute of Integrated Bioinformedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong 999077, China.
| | - Chao Liang
- Institute of Integrated Bioinformedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong 999077, China.
| | - Lei Dang
- Institute of Integrated Bioinformedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong 999077, China.
| | - Aiping Lu
- Institute of Integrated Bioinformedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong 999077, China.
- Institute of Precision Medicine and Innovative Drug Discovery, HKBU (Haimen) Institute of Science and Technology, Haimen 226100, China.
- Shenzhen Lab of Comninatorial Compounds and Targeted Drug Delivery, HKBU Institute of Research and Continuing Education, Shenzhen 518000, China.
| | - Ge Zhang
- Institute of Integrated Bioinformedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong 999077, China.
- Institute of Precision Medicine and Innovative Drug Discovery, HKBU (Haimen) Institute of Science and Technology, Haimen 226100, China.
- Shenzhen Lab of Comninatorial Compounds and Targeted Drug Delivery, HKBU Institute of Research and Continuing Education, Shenzhen 518000, China.
| |
Collapse
|
83
|
Aptamers for CD Antigens: From Cell Profiling to Activity Modulation. MOLECULAR THERAPY-NUCLEIC ACIDS 2016; 6:29-44. [PMID: 28325295 PMCID: PMC5363458 DOI: 10.1016/j.omtn.2016.12.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 12/02/2016] [Accepted: 12/02/2016] [Indexed: 01/01/2023]
Abstract
Nucleic acid-based aptamers are considered to be a promising alternative to antibodies because of their strong and specific binding to diverse targets, fast and inexpensive chemical synthesis, and easy labeling with a fluorescent dye or therapeutic agent. Cluster of differentiation (CD) proteins are among the most popular antigens for aptamers on the cell surface. These anti-CD aptamers could be used in cell biology and biomedicine, from simple cell phenotyping by flow cytometry or fluorescent microscopy to diagnosis and treatment of HIV/AIDS to cancer and immune therapies. The unique feature of aptamers is that they can act simultaneously as an agonist and antagonist of CD receptors depending on a degree of aptamer oligomerization. Aptamers can also deliver small interfering RNA to silence vital genes in CD-positive cells. In this review, we summarize nucleic acid sequences of anti-CD aptamers and their use, which have been validated in multiple studies.
Collapse
|
84
|
Vorobyeva M, Vorobjev P, Venyaminova A. Multivalent Aptamers: Versatile Tools for Diagnostic and Therapeutic Applications. Molecules 2016; 21:molecules21121613. [PMID: 27898020 PMCID: PMC6274531 DOI: 10.3390/molecules21121613] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 11/11/2016] [Accepted: 11/18/2016] [Indexed: 11/24/2022] Open
Abstract
Nucleic acid aptamers generated through an in vitro selection are currently extensively applied as very valuable biomolecular tools thanks to their prominent advantages. Diversity of spatial structures, ease of production through chemical synthesis and a large variety of chemical modifications make aptamers convenient building blocks for the generation of multifunctional constructs. An opportunity to combine different aptamer functionalities with other molecules of interest such as reporter groups, nanoparticles, chemotherapeutic agents, siRNA or antisense oligonucleotides provides a widest range of applications of multivalent aptamers. The present review summarizes approaches to the design of multivalent aptamers, various examples of multifunctional constructs and the prospects of employing them as components of biosensors, probes for affinity capture, tools for cell research and potential therapeutic candidates.
Collapse
Affiliation(s)
- Mariya Vorobyeva
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Lavrentiev Ave., 8, 630090 Novosibirsk, Russia.
| | - Pavel Vorobjev
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Lavrentiev Ave., 8, 630090 Novosibirsk, Russia.
| | - Alya Venyaminova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Lavrentiev Ave., 8, 630090 Novosibirsk, Russia.
| |
Collapse
|
85
|
Zumrut HE, Ara MN, Maio GE, Van NA, Batool S, Mallikaratchy PR. Ligand-guided selection of aptamers against T-cell Receptor-cluster of differentiation 3 (TCR-CD3) expressed on Jurkat.E6 cells. Anal Biochem 2016; 512:1-7. [PMID: 27519622 DOI: 10.1016/j.ab.2016.08.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 08/06/2016] [Accepted: 08/08/2016] [Indexed: 10/21/2022]
Abstract
We recently introduced a screening technology termed ligand-guided selection, (LIGS), to selectively identify target-specific aptamers from an evolved cell-SELEX library. Cell-SELEX utilizes a large combinatorial single-stranded oligonucleotide library and progressively selects DNA ligands against whole cells with variable DNA-binding affinities and specificities by repeated rounds of partition and amplification. LIGS exploits the partition step and introduces a secondary, pre-existing high-affinity monoclonal antibody (mAb) ligand to outcompete and elute specific aptamers towards the binding target of the antibody, not the cell. Here, using anti-CD3ε mAb against the cluster of differentiation 3 (CD3ε), as the guiding ligand against one of the domains of the T-cell Receptor (TCR) complex expressed on Jurkat.E6 cells, we discovered three specific aptamers against TCR complex expressed on an immortalized line of human T lymphocyte cells. In sum, we demonstrate that specific aptamers can be identified utilizing an antibody against a single domain of a multidomain protein complex in their endogenous state with neither post- nor pre-SELEX protein manipulation.
Collapse
Affiliation(s)
- Hasan E Zumrut
- Department of Chemistry, Lehman College, City University of New York, 250 Bedford Park Blvd. West, Bronx, NY 10468, USA; Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY 10016, USA
| | - Mst N Ara
- Department of Chemistry, Lehman College, City University of New York, 250 Bedford Park Blvd. West, Bronx, NY 10468, USA
| | - George E Maio
- Department of Chemistry, Lehman College, City University of New York, 250 Bedford Park Blvd. West, Bronx, NY 10468, USA
| | - Nabeela A Van
- Department of Chemistry, Lehman College, City University of New York, 250 Bedford Park Blvd. West, Bronx, NY 10468, USA
| | - Sana Batool
- Department of Chemistry, Lehman College, City University of New York, 250 Bedford Park Blvd. West, Bronx, NY 10468, USA
| | - Prabodhika R Mallikaratchy
- Department of Chemistry, Lehman College, City University of New York, 250 Bedford Park Blvd. West, Bronx, NY 10468, USA; Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY 10016, USA; Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, NY 10016, USA.
| |
Collapse
|
86
|
Merkle T, Holder IT, Hartig JS. The dual aptamer approach: rational design of a high-affinity FAD aptamer. Org Biomol Chem 2016; 14:447-450. [PMID: 26586417 DOI: 10.1039/c5ob02026c] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
A design strategy for high-affinity aptamers of complex biomolecules is presented. We developed an RNA with FAD-binding properties by combining known ATP- and FMN-aptamers. Cooperative binding of FAD was shown by SPR spectroscopy and fluorescence assays. The strategy should be transferable to several other biomolecules.
Collapse
Affiliation(s)
- T Merkle
- Department of Chemistry and Konstanz Research School Chemical Biology (KoRS-CB), University of Konstanz, Universitätsstr. 10, 78457 Konstanz, Germany.
| | | | | |
Collapse
|
87
|
Aptamers: A Feasible Technology in Cancer Immunotherapy. J Immunol Res 2016; 2016:1083738. [PMID: 27413756 PMCID: PMC4931050 DOI: 10.1155/2016/1083738] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 05/22/2016] [Indexed: 12/21/2022] Open
Abstract
Aptamers are single-chained RNA or DNA oligonucleotides (ODNs) with three-dimensional folding structures which allow them to bind to their targets with high specificity. Aptamers normally show affinities comparable to or higher than that of antibodies. They are chemically synthesized and therefore less expensive to manufacture and produce. A variety of aptamers described to date have been shown to be reliable in modulating immune responses against cancer by either blocking or activating immune receptors. Some of them have been conjugated to other molecules to target the immune system and reduce off-target side effects. Despite the success of first-line treatments against cancer, the elevated number of relapsing cases and the tremendous side effects shown by the commonly used agents hinder conventional treatments against cancer. The advantages provided by aptamers could enhance the therapeutic index of a given strategy and therefore enhance the antitumor effect. Here we recapitulate the provided benefits of aptamers with immunomodulatory activity described to date in cancer therapy and the benefits that aptamer-based immunotherapy could provide either alone or combined with first-line treatments in cancer therapy.
Collapse
|
88
|
Rudchenko MN, Zamyatnin AA. Prospects for using self-assembled nucleic acid structures. BIOCHEMISTRY (MOSCOW) 2016; 80:391-9. [PMID: 25869355 DOI: 10.1134/s000629791504001x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
According to the central dogma in molecular biology, nucleic acids are assigned with key functions on storing and executing genetic information in any living cell. However, features of nucleic acids are not limited only with properties providing template-dependent biosynthetic processes. Studies of DNA and RNA unveiled unique features of these polymers able to make various self-assembled three-dimensional structures that, among other things, use the complementarity principle. Here, we review various self-assembled nucleic acid structures as well as application of DNA and RNA to develop nanomaterials, molecular automata, and nanodevices. It can be expected that in the near future results of these developments will allow designing novel next-generation diagnostic systems and medicinal drugs.
Collapse
Affiliation(s)
- M N Rudchenko
- Research Division, Hospital for Special Surgery, New York, NY 10021, USA.
| | | |
Collapse
|
89
|
Aptamers Binding to c-Met Inhibiting Tumor Cell Migration. PLoS One 2015; 10:e0142412. [PMID: 26658271 PMCID: PMC4676636 DOI: 10.1371/journal.pone.0142412] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 10/21/2015] [Indexed: 01/04/2023] Open
Abstract
The human receptor tyrosine kinase c-Met plays an important role in the control of critical cellular processes. Since c-Met is frequently over expressed or deregulated in human malignancies, blocking its activation is of special interest for therapy. In normal conditions, the c-Met receptor is activated by its bivalent ligand hepatocyte growth factor (HGF). Also bivalent antibodies can activate the receptor by cross linking, limiting therapeutic applications. We report the generation of the RNA aptamer CLN64 containing 2'-fluoro pyrimidine modifications by systematic evolution of ligands by exponential enrichment (SELEX). CLN64 and a previously described single-stranded DNA (ssDNA) aptamer CLN3 exhibited high specificities and affinities to recombinant and cellular expressed c-Met. Both aptamers effectively inhibited HGF-dependent c-Met activation, signaling and cell migration. We showed that these aptamers did not induce c-Met activation, revealing an advantage over bivalent therapeutic molecules. Both aptamers were shown to bind overlapping epitopes but only CLN3 competed with HGF binding to cMet. In addition to their therapeutic and diagnostic potential, CLN3 and CLN64 aptamers exhibit valuable tools to further understand the structural and functional basis for c-Met activation or inhibition by synthetic ligands and their interplay with HGF binding.
Collapse
|
90
|
Ueki R, Ueki A, Kanda N, Sando S. Oligonucleotide-Based Mimetics of Hepatocyte Growth Factor. Angew Chem Int Ed Engl 2015; 55:579-82. [PMID: 26592704 DOI: 10.1002/anie.201508572] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Revised: 10/21/2015] [Indexed: 11/10/2022]
Abstract
Oligonucleotide-based hepatocyte growth factor (HGF) mimetics are described. A DNA aptamer to Met, a cognate receptor for HGF, was shown to induce Met activation when used in dimer form. The most potent aptamer dimer, ss-0, which was composed solely of 100-mer single-stranded DNA, exhibited nanomolar potency. Aptamer ss-0 reproduced HGF-induced cellular behaviors, including migration and proliferation. The present work sheds light on oligonucleotides as a novel chemical entity for the design of growth factor mimetics.
Collapse
Affiliation(s)
- Ryosuke Ueki
- Department of Chemistry and Biotechnology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656 (Japan)
| | - Ayaka Ueki
- Department of Chemistry and Biotechnology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656 (Japan)
| | - Naoto Kanda
- Department of Chemistry and Biotechnology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656 (Japan)
| | - Shinsuke Sando
- Department of Chemistry and Biotechnology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656 (Japan).
| |
Collapse
|
91
|
Ueki R, Ueki A, Kanda N, Sando S. Oligonucleotide‐Based Mimetics of Hepatocyte Growth Factor. Angew Chem Int Ed Engl 2015. [DOI: 10.1002/ange.201508572] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Ryosuke Ueki
- Department of Chemistry and Biotechnology The University of Tokyo 7-3-1 Hongo Bunkyo-ku, Tokyo 113-8656 Japan
| | - Ayaka Ueki
- Department of Chemistry and Biotechnology The University of Tokyo 7-3-1 Hongo Bunkyo-ku, Tokyo 113-8656 Japan
| | - Naoto Kanda
- Department of Chemistry and Biotechnology The University of Tokyo 7-3-1 Hongo Bunkyo-ku, Tokyo 113-8656 Japan
| | - Shinsuke Sando
- Department of Chemistry and Biotechnology The University of Tokyo 7-3-1 Hongo Bunkyo-ku, Tokyo 113-8656 Japan
| |
Collapse
|
92
|
Khedri M, Rafatpanah H, Abnous K, Ramezani P, Ramezani M. Cancer immunotherapy via nucleic acid aptamers. Int Immunopharmacol 2015; 29:926-936. [PMID: 26603636 DOI: 10.1016/j.intimp.2015.10.013] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Revised: 10/09/2015] [Accepted: 10/13/2015] [Indexed: 02/07/2023]
Abstract
Over the past decade, immune therapy has become a standard treatment for a variety of cancers. Monoclonal antibodies, immune adjuvants and vaccines against oncogenic viruses are now well-established cancer therapies. Immune modulation is a principal element of supportive care for many high-dose chemotherapy regimens. Aptamers are short nucleic acids that bind to defined targets with high affinity and specificity. The first aptamers have been selected around two decades ago by an in vitro process named SELEX (systematic evolution of ligands by exponential enrichment). Since then, numerous aptamers with specificities for a variety of targets from small molecules to proteins or even whole cells have been selected. Targeting immunomodulatory ligands in the progressive tumor lesions of the patients would be prophylactic or therapeutic and may reduce drug-associated toxicities. A new class of inhibitory and agonistic ligands composed of short oligonucleotide (ODN) aptamers was developed recently that exhibited bioactivities comparable or superior to that of antibodies. This paper addressed progress in cancer immunotherapy with nucleic acid aptamers and highlighted recent developments either in immune system targeting or in immunotherapy methods involved aptamers. We discussed aptamer limitations when used as therapeutic agents for cancer treatment and suggested ways to overcome those limitations.
Collapse
Affiliation(s)
- Mostafa Khedri
- Department of Immunology, Immunology Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Houshang Rafatpanah
- Department of Immunology, Immunology Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Pouria Ramezani
- Pharmaceutical Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
93
|
Dickgiesser S, Rasche N, Nasu D, Middel S, Hörner S, Avrutina O, Diederichsen U, Kolmar H. Self-Assembled Hybrid Aptamer-Fc Conjugates for Targeted Delivery: A Modular Chemoenzymatic Approach. ACS Chem Biol 2015; 10:2158-65. [PMID: 26131766 DOI: 10.1021/acschembio.5b00315] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Over the past decade, DNA and RNA aptamers have attracted keen research interest due to their ability to specifically bind targets of therapeutic relevance. However, their application is often hampered by a short serum half-life and missing effector functions. Conjugation of aptamers to antibody Fc fragments could improve pharmacokinetics, enable immune effector mechanisms, and provide an option for the introduction of desired payloads (e.g., toxins or fluorescent dyes). We developed a modular scaffold-supported system based on human IgG1 Fc fragments, which allows for its dual functionalization with moieties of interest. In our approach, two bioorthogonal, enzyme-mediated reactions were used in combination with oxime ligation and self-assembly based on PNA-DNA base pairing. Thus, an engineered synthetic peptide nucleic acid (PNA) oligomer was coupled to the C-termini of the Fc dimer upon sequence-specific sortase A-mediated transpeptidation. Hybridization of the resulting Fc-PNA conjugate with a tailored DNA aptamer that binds cancer-related hepatocyte growth factor receptor (c-MET) led to a hybrid construct which showed strong and specific binding to c-MET and was readily internalized by c-MET-overexpressing cells. To install an additional orthogonally addressable site, aldehyde tag technology was applied followed by oxime ligation with an aminooxy-bearing fluorescent dye as model cargo. Delivery of fluorescent probe specifically to c-MET-overexpressing cells was confirmed by flow cytometry. Our approach can provide access to engineered aptamer-Fc conjugates with desired target specificity and cytotoxic payloads.
Collapse
Affiliation(s)
- Stephan Dickgiesser
- Institute
for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Darmstadt, Germany
| | - Nicolas Rasche
- Protein Engineering
and Antibody Technologies, Merck-Serono, Merck KGaA, Darmstadt, Germany
| | - Daichi Nasu
- Institute
for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Darmstadt, Germany
| | - Stephen Middel
- Institute
for Organic and Biomolecular Chemistry, Georg-August University Göttingen, Göttingen, Germany
| | - Sebastian Hörner
- Institute
for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Darmstadt, Germany
| | - Olga Avrutina
- Institute
for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Darmstadt, Germany
| | - Ulf Diederichsen
- Institute
for Organic and Biomolecular Chemistry, Georg-August University Göttingen, Göttingen, Germany
| | - Harald Kolmar
- Institute
for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Darmstadt, Germany
| |
Collapse
|
94
|
Ueki R, Sando S. A DNA aptamer to c-Met inhibits cancer cell migration. Chem Commun (Camb) 2015; 50:13131-4. [PMID: 25223895 DOI: 10.1039/c4cc06016d] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The HGF/c-Met pathway is known to play a key role in cancer metastasis. CLN0003_SL1, a 50-mer DNA aptamer that binds to the c-Met protein, was found to inhibit HGF-induced c-Met activation. This aptamer could suppress cancer cell motility in vitro.
Collapse
Affiliation(s)
- R Ueki
- INAMORI Frontier Research Center, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | | |
Collapse
|
95
|
Barman J. Targeting cancer cells using aptamers: cell-SELEX approach and recent advancements. RSC Adv 2015. [DOI: 10.1039/c4ra12407c] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aptamers are short single stranded nucleic acid based therapeutic and diagnostic molecules which can be isolated from a random pool of oligonucleotides by Systematic Evolution of Ligands by EXponential Enrichment (SELEX).
Collapse
Affiliation(s)
- Jharna Barman
- Agricultural and Ecological Research Unit
- Biological Science Division
- Indian Statistical Institute
- Kolkata
- India
| |
Collapse
|
96
|
Zhu H, Li J, Zhang XB, Ye M, Tan W. Nucleic acid aptamer-mediated drug delivery for targeted cancer therapy. ChemMedChem 2014; 10:39-45. [PMID: 25277749 DOI: 10.1002/cmdc.201402312] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Indexed: 12/21/2022]
Abstract
Aptamers are emerging as promising therapeutic agents and recognition elements. In particular, cell-SELEX (systematic evolution of ligands by exponential enrichment) allows in vitro selection of aptamers selective to whole cells without prior knowledge of the molecular signatures on the cell surface. The advantage of aptamers is their high affinitiy and binding specificity towards the target. This Minireview focuses on single-stranded (ss) oligonucleotide (DNA or RNA)-based aptamers as cancer therapeutics/theranostics. Specifically, aptamer-nanomaterial conjugates, aptamer-drug conjugates, targeted phototherapy and targeted biotherapy are covered in detail. In reviewing the literature, the potential of aptamers as delivery systems for therapeutic and imaging applications in cancer is clear, however, major challenges remain to be resolved, such as the poorly understood pharmacokinetics, toxicity and off-target effects, before they can be fully exploited in a clinical setting.
Collapse
Affiliation(s)
- Huijie Zhu
- Molecular Science & Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing & Chemometrics, College of Chemistry & Chemical Engineering, and College of Biology, Collaborative Innovation Center for Molecular Engineering for Theranostics, Hunan University, Changsha, 410082 (China)
| | | | | | | | | |
Collapse
|
97
|
Sun H, Zhu X, Lu PY, Rosato RR, Tan W, Zu Y. Oligonucleotide aptamers: new tools for targeted cancer therapy. MOLECULAR THERAPY. NUCLEIC ACIDS 2014; 3:e182. [PMID: 25093706 PMCID: PMC4221593 DOI: 10.1038/mtna.2014.32] [Citation(s) in RCA: 352] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2014] [Accepted: 05/30/2014] [Indexed: 02/07/2023]
Abstract
Aptamers are a class of small nucleic acid ligands that are composed of RNA or single-stranded DNA oligonucleotides and have high specificity and affinity for their targets. Similar to antibodies, aptamers interact with their targets by recognizing a specific three-dimensional structure and are thus termed “chemical antibodies.” In contrast to protein antibodies, aptamers offer unique chemical and biological characteristics based on their oligonucleotide properties. Hence, they are more suitable for the development of novel clinical applications. Aptamer technology has been widely investigated in various biomedical fields for biomarker discovery, in vitro diagnosis, in vivo imaging, and targeted therapy. This review will discuss the potential applications of aptamer technology as a new tool for targeted cancer therapy with emphasis on the development of aptamers that are able to specifically target cell surface biomarkers. Additionally, we will describe several approaches for the use of aptamers in targeted therapeutics, including aptamer-drug conjugation, aptamer-nanoparticle conjugation, aptamer-mediated targeted gene therapy, aptamer-mediated immunotherapy, and aptamer-mediated biotherapy.
Collapse
Affiliation(s)
- Hongguang Sun
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas, USA
| | - Xun Zhu
- Department of Immunology, Norman Bethune College of Medicine, Jilin University, Jilin, China
| | | | - Roberto R Rosato
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas, USA
| | - Wen Tan
- School of Biosciences and Bioengineering, South China University of Technology, Guangzhou, China
| | - Youli Zu
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas, USA
| |
Collapse
|
98
|
Baier C, Fino A, Sanchez C, Farnault L, Rihet P, Kahn-Perlès B, Costello RT. Natural killer cells modulation in hematological malignancies. Front Immunol 2013; 4:459. [PMID: 24391641 PMCID: PMC3867693 DOI: 10.3389/fimmu.2013.00459] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 12/02/2013] [Indexed: 12/26/2022] Open
Abstract
Hematological malignancies (HM) treatment improved over the last years resulting in increased achievement of complete or partial remission, but unfortunately high relapse rates are still observed, due to remaining minimal residual disease. Therefore, sustainment of long-term remission is crucial, using either drug maintenance treatment or by boosting or prolonging an immune response. Immune system has a key role in tumor surveillance. Nonetheless, tumor-cells evade the specific T-lymphocyte mediated immune surveillance using many mechanisms but especially by the down-regulation of the expression of HLA class I antigens. In theory, these tumor-cells lacking normal expression of HLA class I molecules should be destroyed by natural killer (NK) cells, according to the missing-self hypothesis. NK cells, at the frontier of innate and adaptive immune system, have a central role in tumor-cells surveillance as demonstrated in the setting of allogenic stem cell transplantation. Nevertheless, tumors develop various mechanisms to escape from NK innate immune pressure. Abnormal NK cytolytic functions have been described in many HM. We present here various mechanisms involved in the escape of HM from NK-cell surveillance, i.e., NK-cells quantitative and qualitative abnormalities.
Collapse
Affiliation(s)
- Céline Baier
- UMR1090 TAGC, INSERM , Marseille , France ; UMR1090 TAGC, Aix-Marseille Université , Marseille , France
| | - Aurore Fino
- UMR1090 TAGC, INSERM , Marseille , France ; UMR1090 TAGC, Aix-Marseille Université , Marseille , France
| | | | - Laure Farnault
- UMR1090 TAGC, INSERM , Marseille , France ; Service d'hématologie, APHM, Hôpital de la Conception , Marseille , France
| | - Pascal Rihet
- UMR1090 TAGC, INSERM , Marseille , France ; UMR1090 TAGC, Aix-Marseille Université , Marseille , France
| | - Brigitte Kahn-Perlès
- UMR1090 TAGC, INSERM , Marseille , France ; UMR1090 TAGC, Aix-Marseille Université , Marseille , France
| | - Régis T Costello
- UMR1090 TAGC, INSERM , Marseille , France ; UMR1090 TAGC, Aix-Marseille Université , Marseille , France ; Service d'hématologie, APHM, Hôpital de la Conception , Marseille , France
| |
Collapse
|
99
|
Chen H, Zheng X, Di B, Wang D, Zhang Y, Xia H, Mao Q. Aptamer modification improves the adenoviral transduction of malignant glioma cells. J Biotechnol 2013. [DOI: 10.1016/j.jbiotec.2013.10.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
100
|
Meyer M, Scheper T, Walter JG. Aptamers: versatile probes for flow cytometry. Appl Microbiol Biotechnol 2013; 97:7097-109. [PMID: 23838792 DOI: 10.1007/s00253-013-5070-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Revised: 06/17/2013] [Accepted: 06/17/2013] [Indexed: 12/21/2022]
Abstract
Aptamers are nucleic acid oligomers with distinct conformational shapes that allow them to bind targets with high affinity and specificity. Aptamers are selected from a random oligonucleotide library by their capability to bind a certain molecular target. A variety of targets ranging from small molecules like amino acids to complex targets and whole cells have been used to select aptamers. These characteristics and the ability to create specific aptamers against virtually any cell type in a process termed "systematic evolution by exponential enrichment" make them interesting tools for flow cytometry. In this contribution, we review the application of aptamers as probes for flow cytometry, especially cell-phenotyping and detection of various cancer cell lines and virus-infected cells and pathogens. We also discuss the potential of aptamers combined with nanoparticles such as quantum dots for the generation of new multivalent detector molecules with enhanced affinity and sensitivity. With regard to recent advancements in aptamer selection and the decreasing costs for oligonucleotide synthesis, aptamers may rise as potent competitors for antibodies as molecular probes in flow cytometry.
Collapse
Affiliation(s)
- Michael Meyer
- Institut für Technische Chemie, Leibniz Universität Hannover, Callinstr. 5, 30167 Hannover, Germany
| | | | | |
Collapse
|