Identification and Engineering of Aptamers for Theranostic Application in Human Health and Disorders
Abstract
:1. Introduction
2. Applications
2.1. Bioimaging
2.1.1. Magnetic Resonance Imaging
2.1.2. Positron Emission Tomography
2.2. Diagnostics
2.2.1. Fluorescent Detection
2.2.2. Colorimetric Detection
2.2.3. Pathogen Detection
2.3. Therapeutics
2.3.1. Therapeutic Drug Cargo
2.3.2. Multiagent Cargo
2.3.3. Inhibitory Agent
2.3.4. Immune Modulators
2.3.5. Aptamers in Clinical Trials
3. Chemical Modifications of Aptamer
3.1. End Modifications of the Backbone
3.2. Modifications of Sugar
3.3. Modifications of Phosphodiester Linkage
3.4. Modifications of the Base
4. Role of Aptamer in COVID-19 Control
4.1. Detection of Nucleic Acid
4.2. Detection of Protein
4.3. Therapeutics for COVID-19
4.4. Recent Advancements
5. Future Prospect: Aptamers in Stem Cell Research, Single Cell Proteomics and Immunotherapy
6. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Galas, D.J.; Schmitz, A. DNAse footprinting: A simple method for the detection of protein-DNA binding specificity. Nucleic Acids Res. 1978, 5, 3157–3170. [Google Scholar] [CrossRef]
- Van Dyke, M.M.; Dervan, P.B. Echinomycin binding sites on DNA. Science 1984, 225, 1122–1127. [Google Scholar] [CrossRef]
- Garner, M.M.; Revzin, A. A gel electrophoresis method for quantifying the binding of proteins to specific DNA regions: Application to components of the Escherichia coli lactose operon regulatory system. Nucleic Acids Res. 1981, 9, 3047–3060. [Google Scholar] [CrossRef] [Green Version]
- Tuerk, C.; Gold, L. Systematic evolution of ligands by exponential enrichment: RNA ligands to bacteriophage T4 DNA polymerase. Science 1990, 249, 505–510. [Google Scholar] [CrossRef]
- Ellington, A.D.; Szostak, J.W. In vitro selection of RNA molecules that bind specific ligands. Nature 1990, 346, 818–822. [Google Scholar] [CrossRef]
- Hermann, T.; Patel, D.J. Adaptive recognition by nucleic acid aptamers. Science 2000, 287, 820–825. [Google Scholar] [CrossRef] [Green Version]
- Bock, L.C.; Griffin, L.C.; Latham, J.A.; Vermaas, E.H.; Toole, J.J. Selection of single-stranded DNA molecules that bind and inhibit human thrombin. Nature 1992, 355, 564–566. [Google Scholar] [CrossRef] [PubMed]
- Pleiko, K.; Saulite, L.; Parfejevs, V.; Miculis, K.; Vjaters, E.; Riekstina, U. Differential binding cell-SELEX method to identify cell-specific aptamers using high-throughput sequencing. Sci. Rep. 2019, 9, 8142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ali, M.H.; Elsherbiny, M.E.; Emara, M. Updates on Aptamer Research. Int. J. Mol. Sci. 2019, 20, 2511. [Google Scholar] [CrossRef] [Green Version]
- Xiang, D.; Zheng, C.; Zhou, S.F.; Qiao, S.; Tran, P.H.; Pu, C.; Li, Y.; Kong, L.; Kouzani, A.Z.; Lin, J.; et al. Superior Performance of Aptamer in Tumor Penetration over Antibody: Implication of Aptamer-Based Theranostics in Solid Tumors. Theranostics 2015, 5, 1083–1097. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xiang, D.; Shigdar, S.; Qiao, G.; Wang, T.; Kouzani, A.Z.; Zhou, S.F.; Kong, L.; Li, Y.; Pu, C.; Duan, W. Nucleic acid aptamer-guided cancer therapeutics and diagnostics: The next generation of cancer medicine. Theranostics 2015, 5, 23–42. [Google Scholar] [CrossRef] [Green Version]
- Huang, B.T.; Lai, W.Y.; Chang, Y.C.; Wang, J.W.; Yeh, S.D.; Lin, E.P.; Yang, P.C. A CTLA-4 Antagonizing DNA Aptamer with Antitumor Effect. Mol. Nucleic Acids 2017, 8, 520–528. [Google Scholar] [CrossRef] [Green Version]
- Chopra, A.; Shukla, R.; Sharma, T.K. Aptamers as an Emerging Player in Biology. Aptamers Synth. Antibodies 2014, 1, 1–16. [Google Scholar]
- Grover, V.P.; Tognarelli, J.M.; Crossey, M.M.; Cox, I.J.; Taylor-Robinson, S.D.; McPhail, M.J. Magnetic Resonance Imaging: Principles and Techniques: Lessons for Clinicians. J. Clin. Exp. Hepatol. 2015, 5, 246–255. [Google Scholar] [CrossRef] [Green Version]
- Xu, W.; Lu, Y. A smart magnetic resonance imaging contrast agent responsive to adenosine based on a DNA aptamer-conjugated gadolinium complex. Chem. Commun. (Camb) 2011, 47, 4998–5000. [Google Scholar] [CrossRef]
- Yigit, M.V.; Mazumdar, D.; Kim, H.K.; Lee, J.H.; Odintsov, B.; Lu, Y. Smart "turn-on" magnetic resonance contrast agents based on aptamer-functionalized superparamagnetic iron oxide nanoparticles. Chembiochem 2007, 8, 1675–1678. [Google Scholar] [CrossRef] [PubMed]
- Berger, A. How does it work? Positron emission tomography. BMJ 2003, 326, 1449. [Google Scholar] [CrossRef] [PubMed]
- Jacobson, O.; Weiss, I.D.; Wang, L.; Wang, Z.; Yang, X.; Dewhurst, A.; Ma, Y.; Zhu, G.; Niu, G.; Kiesewetter, D.O.; et al. 18F-Labeled Single-Stranded DNA Aptamer for PET Imaging of Protein Tyrosine Kinase-7 Expression. J. Nucl. Med. 2015, 56, 1780–1785. [Google Scholar] [CrossRef] [Green Version]
- Dougherty, C.A.; Cai, W.; Hong, H. Applications of aptamers in targeted imaging: State of the art. Curr. Top. Med. Chem. 2015, 15, 1138–1152. [Google Scholar] [CrossRef] [PubMed]
- Yoon, S.; Rossi, J.J. Targeted Molecular Imaging Using Aptamers in Cancer. Pharmaceuticals 2018, 11, 71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Song, K.M.; Lee, S.; Ban, C. Aptamers and their biological applications. Sensors 2012, 12, 612–631. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, Y.; Sun, L.; Zhao, Q. Development of aptamer fluorescent switch assay for aflatoxin B1 by using fluorescein-labeled aptamer and black hole quencher 1-labeled complementary DNA. Anal. Bioanal. Chem. 2018, 410, 6269–6277. [Google Scholar] [CrossRef]
- Wu, X.; Zhao, Z.; Bai, H.; Fu, T.; Yang, C.; Hu, X.; Liu, Q.; Champanhac, C.; Teng, I.T.; Ye, M.; et al. DNA Aptamer Selected against Pancreatic Ductal Adenocarcinoma for in vivo Imaging and Clinical Tissue Recognition. Theranostics 2015, 5, 985–994. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wu, X.; Liu, H.; Han, D.; Peng, B.; Zhang, H.; Zhang, L.; Li, J.; Liu, J.; Cui, C.; Fang, S.; et al. Elucidation and Structural Modeling of CD71 as a Molecular Target for Cell-Specific Aptamer Binding. J. Am. Chem. Soc. 2019, 141, 10760–10769. [Google Scholar] [CrossRef] [PubMed]
- Dhiman, A.; Kalra, P.; Bansal, V.; Bruno, J.G.; Sharma, T.K. Aptamer-based point-of-care diagnostic platforms. Sens. Acutators B 2017, 246, 535–553. [Google Scholar] [CrossRef]
- Hwang, S.H.J.; Jeong, S.; Choi, H.J.; Eun, H.; Jo, M.G.; Kwon, W.Y.; Kim, S.; Kim, Y.; Lee, M.; Park, K.S. Target-Induced Aggregation of Gold Nanoparticles for Colorimetric Detection of Bisphenol A. J. Nanomater. 2019, 2019, 3676384. [Google Scholar] [CrossRef] [Green Version]
- Quesada-Gonzalez, D.; Merkoci, A. Nanomaterial-based devices for point-of-care diagnostic applications. Chem. Soc. Rev. 2018, 47, 4697–4709. [Google Scholar] [CrossRef]
- Reid, R.; Chatterjee, B.; Das, S.J.; Ghosh, S.; Sharma, T.K. Application of aptamers as molecular recognition elements in lateral flow assays. Anal. Biochem. 2020, 593, 113574. [Google Scholar] [CrossRef] [PubMed]
- Xu, H.; Mao, X.; Zeng, Q.; Wang, S.; Kawde, A.N.; Liu, G. Aptamer-functionalized gold nanoparticles as probes in a dry-reagent strip biosensor for protein analysis. Anal. Chem. 2009, 81, 669–675. [Google Scholar] [CrossRef] [PubMed]
- Lee, K.H.; Zeng, H. Aptamer-Based ELISA Assay for Highly Specific and Sensitive Detection of Zika NS1 Protein. Anal. Chem. 2017, 89, 12743–12748. [Google Scholar] [CrossRef]
- Dhiman, A.; Haldar, S.; Mishra, S.K.; Sharma, N.; Bansal, A.; Ahmad, Y.; Kumar, A.; Sharma, T.K.; Tyagi, J.S. Generation and application of DNA aptamers against HspX for accurate diagnosis of tuberculous meningitis. Tuberc. (Edinb) 2018, 112, 27–36. [Google Scholar] [CrossRef] [PubMed]
- Kumari, P.; Lavania, S.; Tyagi, S.; Dhiman, A.; Rath, D.; Anthwal, D.; Gupta, R.K.; Sharma, N.; Gadpayle, A.K.; Taneja, R.S.; et al. A novel aptamer-based test for the rapid and accurate diagnosis of pleural tuberculosis. Anal. Biochem. 2019, 564–565, 80–87. [Google Scholar] [CrossRef] [PubMed]
- Lavania, S.; Das, R.; Dhiman, A.; Myneedu, V.P.; Verma, A.; Singh, N.; Sharma, T.K.; Tyagi, J.S. Aptamer-Based TB Antigen Tests for the Rapid Diagnosis of Pulmonary Tuberculosis: Potential Utility in Screening for Tuberculosis. ACS Infect. Dis. 2018, 4, 1718–1726. [Google Scholar] [CrossRef] [PubMed]
- Wang, R.; Zhao, J.; Jiang, T.; Kwon, Y.M.; Lu, H.; Jiao, P.; Liao, M.; Li, Y. Selection and characterization of DNA aptamers for use in detection of avian influenza virus H5N1. J. Virol. Methods 2013, 189, 362–369. [Google Scholar] [CrossRef]
- Das, R.; Chaterjee, B.; Kapil, A.; Sharma, T.K. Aptamer-NanoZyme mediated sensing platform for the rapid detection of Escherichia coli in fruit juice. Sens. Bio-Sens. Res. 2020, 27. [Google Scholar] [CrossRef]
- Das, R.; Dhiman, A.; Kapil, A.; Bansal, V.; Sharma, T.K. Aptamer-mediated colorimetric and electrochemical detection of Pseudomonas aeruginosa utilizing peroxidase-mimic activity of gold NanoZyme. Anal. Bioanal. Chem. Vol. 2019, 1229–1238. [Google Scholar] [CrossRef]
- Das, R.; Dhiman, A.; Mishra, S.K.; Haldar, S.; Sharma, N.; Bansal, A.; Ahmad, Y.; Kumar, A.; Tyagi, J.S.; Sharma, T.K. Structural switching electrochemical DNA aptasensor for the rapid diagnosis of tuberculous meningitis. Int. J. Nanomed. 2019, 14, 2103–2113. [Google Scholar] [CrossRef] [Green Version]
- Kaur, H.; Bruno, J.G.; Kumar, A.; Sharma, T.K. Aptamers in the Therapeutics and Diagnostics Pipelines. Theranostics 2018, 8, 4016–4032. [Google Scholar] [CrossRef]
- Bruno, J.G.; Phillips, T.; Montez, T.; Garcia, A.; Sivils, J.C.; Mayo, M.W.; Greis, A.; Metrix, L. Development of a fluorescent enzyme-linked DNA aptamer-magnetic bead sandwich assay and portable fluorometer for sensitive and rapid listeria detection. J. Fluoresc. 2015, 25, 173–183. [Google Scholar] [CrossRef]
- Muller, J.; Friedrich, M.; Becher, T.; Braunstein, J.; Kupper, T.; Berdel, P.; Gravius, S.; Rohrbach, F.; Oldenburg, J.; Mayer, G.; et al. Monitoring of plasma levels of activated protein C using a clinically applicable oligonucleotide-based enzyme capture assay. J. Thromb. Haemost. 2012, 10, 390–398. [Google Scholar] [CrossRef]
- De Groote, M.A.; Nahid, P.; Jarlsberg, L.; Johnson, J.L.; Weiner, M.; Muzanyi, G.; Janjic, N.; Sterling, D.G.; Ochsner, U.A. Elucidating novel serum biomarkers associated with pulmonary tuberculosis treatment. PLoS ONE 2013, 8, e61002. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kraemer, S.; Vaught, J.D.; Bock, C.; Gold, L.; Katilius, E.; Keeney, T.R.; Kim, N.; Saccomano, N.A.; Wilcox, S.K.; Zichi, D.; et al. From SOMAmer-based biomarker discovery to diagnostic and clinical applications: A SOMAmer-based, streamlined multiplex proteomic assay. PLoS ONE 2011, 6, e26332. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lollo, B.; Steele, F.; Gold, L. Beyond antibodies: New affinity reagents to unlock the proteome. Proteomics 2014, 14, 638–644. [Google Scholar] [CrossRef] [PubMed]
- Eid, C.; Palko, J.W.; Katilius, E.; Santiago, J.G. Rapid Slow Off-Rate Modified Aptamer (SOMAmer)-Based Detection of C-Reactive Protein Using Isotachophoresis and an Ionic Spacer. Anal. Chem. 2015, 87, 6736–6743. [Google Scholar] [CrossRef] [PubMed]
- Viraka Nellore, B.P.; Kanchanapally, R.; Pramanik, A.; Sinha, S.S.; Chavva, S.R.; Hamme, A., II; Ray, P.C. Aptamer-conjugated graphene oxide membranes for highly efficient capture and accurate identification of multiple types of circulating tumor cells. Bioconjug. Chem. 2015, 26, 235–242. [Google Scholar] [CrossRef] [PubMed]
- Rothlisberger, P.; Gasse, C.; Hollenstein, M. Nucleic Acid Aptamers: Emerging Applications in Medical Imaging, Nanotechnology, Neurosciences, and Drug Delivery. Int. J. Mol. Sci. 2017, 18, 2430. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huwyler, J.; Wu, D.; Pardridge, W.M. Brain drug delivery of small molecules using immunoliposomes. Proc. Natl. Acad. Sci. USA 1996, 93, 14164–14169. [Google Scholar] [CrossRef] [Green Version]
- Alshaer, W.; Hillaireau, H.; Vergnaud, J.; Ismail, S.; Fattal, E. Functionalizing Liposomes with anti-CD44 Aptamer for Selective Targeting of Cancer Cells. Bioconjug. Chem. 2015, 26, 1307–1313. [Google Scholar] [CrossRef]
- Kang, H.; O’Donoghue, M.B.; Liu, H.; Tan, W. A liposome-based nanostructure for aptamer directed delivery. Chem. Commun. (Camb) 2010, 46, 249–251. [Google Scholar] [CrossRef] [PubMed]
- Plourde, K.; Derbali, R.M.; Desrosiers, A.; Dubath, C.; Vallee-Belisle, A.; Leblond, J. Aptamer-based liposomes improve specific drug loading and release. J. Control. Release 2017, 251, 82–91. [Google Scholar] [CrossRef] [Green Version]
- Farokhzad, O.C.; Cheng, J.; Teply, B.A.; Sherifi, I.; Jon, S.; Kantoff, P.W.; Richie, J.P.; Langer, R. Targeted nanoparticle-aptamer bioconjugates for cancer chemotherapy in vivo. Proc. Natl. Acad. Sci. USA 2006, 103, 6315–6320. [Google Scholar] [CrossRef] [Green Version]
- Huang, Y.F.; Shangguan, D.; Liu, H.; Phillips, J.A.; Zhang, X.; Chen, Y.; Tan, W. Molecular assembly of an aptamer-drug conjugate for targeted drug delivery to tumor cells. Chembiochem 2009, 10, 862–868. [Google Scholar] [CrossRef] [Green Version]
- McNamara, J.O., II; Andrechek, E.R.; Wang, Y.; Viles, K.D.; Rempel, R.E.; Gilboa, E.; Sullenger, B.A.; Giangrande, P.H. Cell type-specific delivery of siRNAs with aptamer-siRNA chimeras. Nat. Biotechnol. 2006, 24, 1005–1015. [Google Scholar] [CrossRef]
- Jeong, H.; Lee, S.H.; Hwang, Y.; Yoo, H.; Jung, H.; Kim, S.H.; Mok, H. Multivalent Aptamer-RNA Conjugates for Simple and Efficient Delivery of Doxorubicin/siRNA into Multidrug-Resistant Cells. Macromol. Biosci. 2017, 17. [Google Scholar] [CrossRef]
- Yin, H.; Xiong, G.; Guo, S.; Xu, C.; Xu, R.; Guo, P.; Shu, D. Delivery of Anti-miRNA for Triple-Negative Breast Cancer Therapy Using RNA Nanoparticles Targeting Stem Cell Marker CD133. Mol. Ther. 2019, 27, 1252–1261. [Google Scholar] [CrossRef] [Green Version]
- Guo, S.; Piao, X.; Li, H.; Guo, P. Methods for construction and characterization of simple or special multifunctional RNA nanoparticles based on the 3WJ of phi29 DNA packaging motor. Methods 2018, 143, 121–133. [Google Scholar] [CrossRef]
- Guo, S.; Vieweger, M.; Zhang, K.; Yin, H.; Wang, H.; Li, X.; Li, S.; Hu, S.; Sparreboom, A.; Evers, B.M.; et al. Ultra-thermostable RNA nanoparticles for solubilizing and high-yield loading of paclitaxel for breast cancer therapy. Nat. Commun. 2020, 11, 972. [Google Scholar] [CrossRef] [Green Version]
- Yang, D.; Meng, X.; Yu, Q.; Xu, L.; Long, Y.; Liu, B.; Fang, X.; Zhu, H. Inhibition of hepatitis C virus infection by DNA aptamer against envelope protein. Antimicrob. Agents Chemother. 2013, 57, 4937–4944. [Google Scholar] [CrossRef] [Green Version]
- Susek, K.H.; Karvouni, M.; Alici, E.; Lundqvist, A. The Role of CXC Chemokine Receptors 1–4 on Immune Cells in the Tumor Microenvironment. Front. Immunol. 2018, 9, 2159. [Google Scholar] [CrossRef]
- Pastor, F. Aptamers: A New Technological Platform in Cancer Immunotherapy. Pharmaceuticals 2016, 9, 64. [Google Scholar] [CrossRef] [Green Version]
- Fu, Z.; Xiang, J. Aptamers, the Nucleic Acid Antibodies, in Cancer Therapy. Int. J. Mol. Sci. 2020, 21, 2793. [Google Scholar] [CrossRef]
- Santulli-Marotto, S.; Nair, S.K.; Rusconi, C.; Sullenger, B.; Gilboa, E. Multivalent RNA aptamers that inhibit CTLA-4 and enhance tumor immunity. Cancer Res. 2003, 63, 7483–7489. [Google Scholar]
- Prodeus, A.; Abdul-Wahid, A.; Fischer, N.W.; Huang, E.H.; Cydzik, M.; Gariepy, J. Targeting the PD-1/PD-L1 Immune Evasion Axis With DNA Aptamers as a Novel Therapeutic Strategy for the Treatment of Disseminated Cancers. Mol. Nucleic Acids 2015, 4, e237. [Google Scholar] [CrossRef]
- Lai, W.Y.; Huang, B.T.; Wang, J.W.; Lin, P.Y.; Yang, P.C. A Novel PD-L1-targeting Antagonistic DNA Aptamer With Antitumor Effects. Mol. Nucleic Acids 2016, 5, e397. [Google Scholar] [CrossRef] [Green Version]
- McNamara, J.O.; Kolonias, D.; Pastor, F.; Mittler, R.S.; Chen, L.; Giangrande, P.H.; Sullenger, B.; Gilboa, E. Multivalent 4-1BB binding aptamers costimulate CD8+ T cells and inhibit tumor growth in mice. J. Clin. Investig. 2008, 118, 376–386. [Google Scholar] [CrossRef]
- Dollins, C.M.; Nair, S.; Boczkowski, D.; Lee, J.; Layzer, J.M.; Gilboa, E.; Sullenger, B.A. Assembling OX40 aptamers on a molecular scaffold to create a receptor-activating aptamer. Chem. Biol. 2008, 15, 675–682. [Google Scholar] [CrossRef] [Green Version]
- Ng, E.W.; Shima, D.T.; Calias, P.; Cunningham, E.T., Jr.; Guyer, D.R.; Adamis, A.P. Pegaptanib, a targeted anti-VEGF aptamer for ocular vascular disease. Nat. Rev. Drug Discov. 2006, 5, 123–132. [Google Scholar] [CrossRef]
- Ferrara, N.; Gerber, H.P.; LeCouter, J. The biology of VEGF and its receptors. Nat. Med. 2003, 9, 669–676. [Google Scholar] [CrossRef]
- Lee, J.H.; Canny, M.D.; De Erkenez, A.; Krilleke, D.; Ng, Y.S.; Shima, D.T.; Pardi, A.; Jucker, F. A therapeutic aptamer inhibits angiogenesis by specifically targeting the heparin binding domain of VEGF165. Proc. Natl. Acad. Sci. USA 2005, 102, 18902–18907. [Google Scholar] [CrossRef] [Green Version]
- Ruckman, J.; Green, L.S.; Beeson, J.; Waugh, S.; Gillette, W.L.; Henninger, D.D.; Claesson-Welsh, L.; Janjic, N. 2′-Fluoropyrimidine RNA-based aptamers to the 165-amino acid form of vascular endothelial growth factor (VEGF165). Inhibition of receptor binding and VEGF-induced vascular permeability through interactions requiring the exon 7-encoded domain. J. Biol. Chem. 1998, 273, 20556–20567. [Google Scholar] [CrossRef] [Green Version]
- Chen, Y.; Wiesmann, C.; Fuh, G.; Li, B.; Christinger, H.W.; McKay, P.; de Vos, A.M.; Lowman, H.B. Selection and analysis of an optimized anti-VEGF antibody: Crystal structure of an affinity-matured Fab in complex with antigen. J. Mol. Biol. 1999, 293, 865–881. [Google Scholar] [CrossRef] [Green Version]
- Nonaka, Y.; Sode, K.; Ikebukuro, K. Screening and improvement of an anti-VEGF DNA aptamer. Molecules 2010, 15, 215–225. [Google Scholar] [CrossRef]
- Hasegawa, H.; Taira, K.I.; Sode, K.; Ikebukuro, K. Improvement of Aptamer Affinity by Dimerization. Sensors 2008, 8, 1090–1098. [Google Scholar] [CrossRef] [Green Version]
- Gilbert, J.C.; DeFeo-Fraulini, T.; Hutabarat, R.M.; Horvath, C.J.; Merlino, P.G.; Marsh, H.N.; Healy, J.M.; Boufakhreddine, S.; Holohan, T.V.; Schaub, R.G. First-in-human evaluation of anti von Willebrand factor therapeutic aptamer ARC1779 in healthy volunteers. Circulation 2007, 116, 2678–2686. [Google Scholar] [CrossRef] [PubMed]
- Spiel, A.O.; Mayr, F.B.; Ladani, N.; Wagner, P.G.; Schaub, R.G.; Gilbert, J.C.; Jilma, B. The aptamer ARC1779 is a potent and specific inhibitor of von Willebrand Factor mediated ex vivo platelet function in acute myocardial infarction. Platelets 2009, 20, 334–340. [Google Scholar] [CrossRef] [PubMed]
- Markus, H.S.; McCollum, C.; Imray, C.; Goulder, M.A.; Gilbert, J.; King, A. The von Willebrand inhibitor ARC1779 reduces cerebral embolization after carotid endarterectomy: A randomized trial. Stroke 2011, 42, 2149–2153. [Google Scholar] [CrossRef] [Green Version]
- Biesecker, G.; Dihel, L.; Enney, K.; Bendele, R.A. Derivation of RNA aptamer inhibitors of human complement C5. Immunopharmacology 1999, 42, 219–230. [Google Scholar] [CrossRef]
- Waters, E.K.; Genga, R.M.; Schwartz, M.C.; Nelson, J.A.; Schaub, R.G.; Olson, K.A.; Kurz, J.C.; McGinness, K.E. Aptamer ARC19499 mediates a procoagulant hemostatic effect by inhibiting tissue factor pathway inhibitor. Blood 2011, 117, 5514–5522. [Google Scholar] [CrossRef]
- Rosenberg, J.E.; Bambury, R.M.; Van Allen, E.M.; Drabkin, H.A.; Lara, P.N., Jr.; Harzstark, A.L.; Wagle, N.; Figlin, R.A.; Smith, G.W.; Garraway, L.A.; et al. A phase II trial of AS1411 (a novel nucleolin-targeted DNA aptamer) in metastatic renal cell carcinoma. Investig. New Drugs 2014, 32, 178–187. [Google Scholar] [CrossRef]
- Reyes-Reyes, E.M.; Salipur, F.R.; Shams, M.; Forsthoefel, M.K.; Bates, P.J. Mechanistic studies of anticancer aptamer AS1411 reveal a novel role for nucleolin in regulating Rac1 activation. Mol. Oncol. 2015, 9, 1392–1405. [Google Scholar] [CrossRef] [Green Version]
- Jaffe, G.J.; Eliott, D.; Wells, J.A.; Prenner, J.L.; Papp, A.; Patel, S. A Phase 1 Study of Intravitreous E10030 in Combination with Ranibizumab in Neovascular Age-Related Macular Degeneration. Ophthalmology 2016, 123, 78–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jaffe, G.J.; Ciulla, T.A.; Ciardella, A.P.; Devin, F.; Dugel, P.U.; Eandi, C.M.; Masonson, H.; Mones, J.; Pearlman, J.A.; Quaranta-El Maftouhi, M.; et al. Dual Antagonism of PDGF and VEGF in Neovascular Age-Related Macular Degeneration: A Phase IIb, Multicenter, Randomized Controlled Trial. Ophthalmology 2017, 124, 224–234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Carrasquillo, K.G.; Ricker, J.A.; Rigas, I.K.; Miller, J.W.; Gragoudas, E.S.; Adamis, A.P. Controlled delivery of the anti-VEGF aptamer EYE001 with poly(lactic-co-glycolic)acid microspheres. Invest. Ophthalmol. Vis. Sci. 2003, 44, 290–299. [Google Scholar] [CrossRef] [Green Version]
- Eyetech Study, G. Preclinical and phase 1A clinical evaluation of an anti-VEGF pegylated aptamer (EYE001) for the treatment of exudative age-related macular degeneration. Retina 2002, 22, 143–152. [Google Scholar] [CrossRef]
- Roccaro, A.M.; Sacco, A.; Purschke, W.G.; Moschetta, M.; Buchner, K.; Maasch, C.; Zboralski, D.; Zollner, S.; Vonhoff, S.; Mishima, Y.; et al. SDF-1 inhibition targets the bone marrow niche for cancer therapy. Cell Rep. 2014, 9, 118–128. [Google Scholar] [CrossRef] [Green Version]
- Hoellenriegel, J.; Zboralski, D.; Maasch, C.; Rosin, N.Y.; Wierda, W.G.; Keating, M.J.; Kruschinski, A.; Burger, J.A. The Spiegelmer NOX-A12, a novel CXCL12 inhibitor, interferes with chronic lymphocytic leukemia cell motility and causes chemosensitization. Blood 2014, 123, 1032–1039. [Google Scholar] [CrossRef]
- Oberthur, D.; Achenbach, J.; Gabdulkhakov, A.; Buchner, K.; Maasch, C.; Falke, S.; Rehders, D.; Klussmann, S.; Betzel, C. Crystal structure of a mirror-image L-RNA aptamer (Spiegelmer) in complex with the natural L-protein target CCL2. Nat. Commun. 2015, 6, 6923. [Google Scholar] [CrossRef] [Green Version]
- Menne, J.; Eulberg, D.; Beyer, D.; Baumann, M.; Saudek, F.; Valkusz, Z.; Wiecek, A.; Haller, H.; Emapticap Study, G. C-C motif-ligand 2 inhibition with emapticap pegol (NOX-E36) in type 2 diabetic patients with albuminuria. Nephrol. Dial. Transpl. 2017, 32, 307–315. [Google Scholar] [CrossRef] [Green Version]
- Schwoebel, F.; van Eijk, L.T.; Zboralski, D.; Sell, S.; Buchner, K.; Maasch, C.; Purschke, W.G.; Humphrey, M.; Zollner, S.; Eulberg, D.; et al. The effects of the anti-hepcidin Spiegelmer NOX-H94 on inflammation-induced anemia in cynomolgus monkeys. Blood 2013, 121, 2311–2315. [Google Scholar] [CrossRef] [PubMed]
- Troisi, R.; Napolitano, V.; Spiridonova, V.; Russo Krauss, I.; Sica, F. Several structural motifs cooperate in determining the highly effective anti-thrombin activity of NU172 aptamer. Nucleic Acids Res. 2018, 46, 12177–12185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gragoudas, E.S.; Adamis, A.P.; Cunningham, E.T., Jr.; Feinsod, M.; Guyer, D.R.; VEGF Inhibition Study in Ocular Neovascularization Clinical Trial Group. Pegaptanib for neovascular age-related macular degeneration. N. Engl. J. Med. 2004, 351, 2805–2816. [Google Scholar] [CrossRef] [Green Version]
- Manresa, N.; Mulero, J.; Losada, M.; Zafrilla, P. Effect of Pegaptanib and Ranibizumab on Plasma and Vitreous Homocysteine in Patients with Exudative Age-Related Macular Degeneration. Retina 2015, 35, 1765–1771. [Google Scholar] [CrossRef] [PubMed]
- Povsic, T.J.; Wargin, W.A.; Alexander, J.H.; Krasnow, J.; Krolick, M.; Cohen, M.G.; Mehran, R.; Buller, C.E.; Bode, C.; Zelenkofske, S.L.; et al. Pegnivacogin results in near complete FIX inhibition in acute coronary syndrome patients: RADAR pharmacokinetic and pharmacodynamic substudy. Eur. Heart J. 2011, 32, 2412–2419. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Staudacher, D.L.; Putz, V.; Heger, L.; Reinohl, J.; Hortmann, M.; Zelenkofske, S.L.; Becker, R.C.; Rusconi, C.P.; Bode, C.; Ahrens, I. Direct factor IXa inhibition with the RNA-aptamer pegnivacogin reduces platelet reactivity in vitro and residual platelet aggregation in patients with acute coronary syndromes. Eur. Heart J. Acute Cardiovasc. Care 2017, 2048872617703065. [Google Scholar] [CrossRef]
- Cohen, M.G.; Purdy, D.A.; Rossi, J.S.; Grinfeld, L.R.; Myles, S.K.; Aberle, L.H.; Greenbaum, A.B.; Fry, E.; Chan, M.Y.; Tonkens, R.M.; et al. First clinical application of an actively reversible direct factor IXa inhibitor as an anticoagulation strategy in patients undergoing percutaneous coronary intervention. Circulation 2010, 122, 614–622. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Povsic, T.J.; Cohen, M.G.; Chan, M.Y.; Zelenkofske, S.L.; Wargin, W.A.; Harrington, R.A.; Alexander, J.H.; Rusconi, C.P.; Becker, R.C. Dose selection for a direct and selective factor IXa inhibitor and its complementary reversal agent: Translating pharmacokinetic and pharmacodynamic properties of the REG1 system to clinical trial design. J. Thromb. Thrombolysis 2011, 32, 21–31. [Google Scholar] [CrossRef] [PubMed]
- Tanaka, K.A.; Szlam, F.; Rusconi, C.P.; Levy, J.H. In-vitro evaluation of anti-factor IXa aptamer on thrombin generation, clotting time, and viscoelastometry. Thromb. Haemost. 2009, 101, 827–833. [Google Scholar] [CrossRef] [PubMed]
- Shangguan, D.; Li, Y.; Tang, Z.; Cao, Z.C.; Chen, H.W.; Mallikaratchy, P.; Sefah, K.; Yang, C.J.; Tan, W. Aptamers evolved from live cells as effective molecular probes for cancer study. Proc. Natl. Acad. Sci. USA 2006, 103, 11838–11843. [Google Scholar] [CrossRef] [Green Version]
- Shangguan, D.; Cao, Z.; Meng, L.; Mallikaratchy, P.; Sefah, K.; Wang, H.; Li, Y.; Tan, W. Cell-specific aptamer probes for membrane protein elucidation in cancer cells. J. Proteome Res. 2008, 7, 2133–2139. [Google Scholar] [CrossRef] [Green Version]
- Lakhin, A.V.; Tarantul, V.Z.; Gening, L.V. Aptamers: Problems, solutions and prospects. Acta Nat. 2013, 5, 34–43. [Google Scholar] [CrossRef] [Green Version]
- Ni, S.; Zhuo, Z.; Pan, Y.; Yu, Y.; Li, F.; Liu, J.; Wang, L.; Wu, X.; Li, D.; Wan, Y.; et al. Recent Progress in Aptamer Discoveries and Modifications for Therapeutic Applications. ACS Appl. Mater. Interfaces 2021, 13, 9500–9519. [Google Scholar] [CrossRef]
- Maier, K.E.; Levy, M. From selection hits to clinical leads: Progress in aptamer discovery. Mol. Methods Clin. Dev. 2016, 5, 16014. [Google Scholar] [CrossRef] [Green Version]
- Dougan, H.; Lyster, D.M.; Vo, C.V.; Stafford, A.; Weitz, J.I.; Hobbs, J.B. Extending the lifetime of anticoagulant oligodeoxynucleotide aptamers in blood. Nucl. Med. Biol. 2000, 27, 289–297. [Google Scholar] [CrossRef]
- de Smidt, P.C.; Le Doan, T.; de Falco, S.; van Berkel, T.J. Association of antisense oligonucleotides with lipoproteins prolongs the plasma half-life and modifies the tissue distribution. Nucleic Acids Res. 1991, 19, 4695–4700. [Google Scholar] [CrossRef] [Green Version]
- Mallikaratchy, P.R.; Ruggiero, A.; Gardner, J.R.; Kuryavyi, V.; Maguire, W.F.; Heaney, M.L.; McDevitt, M.R.; Patel, D.J.; Scheinberg, D.A. A multivalent DNA aptamer specific for the B-cell receptor on human lymphoma and leukemia. Nucleic Acids Res. 2011, 39, 2458–2469. [Google Scholar] [CrossRef]
- Pasternak, A.; Hernandez, F.J.; Rasmussen, L.M.; Vester, B.; Wengel, J. Improved thrombin binding aptamer by incorporation of a single unlocked nucleic acid monomer. Nucleic Acids Res. 2011, 39, 1155–1164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Padilla, R.; Sousa, R. Efficient synthesis of nucleic acids heavily modified with non-canonical ribose 2’-groups using a mutantT7 RNA polymerase (RNAP). Nucleic Acids Res. 1999, 27, 1561–1563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Klussmann, S.; Nolte, A.; Bald, R.; Erdmann, V.A.; Furste, J.P. Mirror-image RNA that binds D-adenosine. Nat. Biotechnol. 1996, 14, 1112–1115. [Google Scholar] [CrossRef] [PubMed]
- Eulberg, D.; Klussmann, S. Spiegelmers: Biostable aptamers. Chembiochem 2003, 4, 979–983. [Google Scholar] [CrossRef]
- Williams, K.P.; Liu, X.H.; Schumacher, T.N.; Lin, H.Y.; Ausiello, D.A.; Kim, P.S.; Bartel, D.P. Bioactive and nuclease-resistant L-DNA ligand of vasopressin. Proc. Natl. Acad. Sci. USA 1997, 94, 11285–11290. [Google Scholar] [CrossRef] [Green Version]
- Vater, A.; Klussmann, S. Turning mirror-image oligonucleotides into drugs: The evolution of Spiegelmer(®) therapeutics. Drug Discov. Today 2015, 20, 147–155. [Google Scholar] [CrossRef] [Green Version]
- Sacca, B.; Lacroix, L.; Mergny, J.L. The effect of chemical modifications on the thermal stability of different G-quadruplex-forming oligonucleotides. Nucleic Acids Res. 2005, 33, 1182–1192. [Google Scholar] [CrossRef] [Green Version]
- Lee, K.Y.; Kang, H.; Ryu, S.H.; Lee, D.S.; Lee, J.H.; Kim, S. Bioimaging of nucleolin aptamer-containing 5-(N-benzylcarboxyamide)-2’-deoxyuridine more capable of specific binding to targets in cancer cells. J. Biomed. Biotechnol. 2010, 2010, 168306. [Google Scholar] [CrossRef] [Green Version]
- Yan, Y.; Chang, L.; Wang, L. Laboratory testing of SARS-CoV, MERS-CoV, and SARS-CoV-2 (2019-nCoV): Current status, challenges, and countermeasures. Rev. Med. Virol. 2020, 30, e2106. [Google Scholar] [CrossRef]
- Wang, M.Y.; Zhao, R.; Gao, L.J.; Gao, X.F.; Wang, D.P.; Cao, J.M. SARS-CoV-2: Structure, Biology, and Structure-Based Therapeutics Development. Front. Cell Infect. Microbiol. 2020, 10, 587269. [Google Scholar] [CrossRef]
- Dolgosheina, E.V.; Jeng, S.C.; Panchapakesan, S.S.; Cojocaru, R.; Chen, P.S.; Wilson, P.D.; Hawkins, N.; Wiggins, P.A.; Unrau, P.J. RNA mango aptamer-fluorophore: A bright, high-affinity complex for RNA labeling and tracking. ACS Chem. Biol. 2014, 9, 2412–2420. [Google Scholar] [CrossRef] [PubMed]
- Compton, J. Nucleic acid sequence-based amplification. Nature 1991, 350, 91–92. [Google Scholar] [CrossRef] [PubMed]
- Abdolahzadeh, A.; Dolgosheina, E.V.; Unrau, P.J. RNA detection with high specificity and sensitivity using nested fluorogenic Mango NASBA. RNA 2019, 25, 1806–1813. [Google Scholar] [CrossRef] [PubMed]
- Ahn, D.G.; Jeon, I.J.; Kim, J.D.; Song, M.S.; Han, S.R.; Lee, S.W.; Jung, H.; Oh, J.W. RNA aptamer-based sensitive detection of SARS coronavirus nucleocapsid protein. Analyst 2009, 134, 1896–1901. [Google Scholar] [CrossRef]
- Le, T.T.; Chang, P.; Benton, D.J.; McCauley, J.W.; Iqbal, M.; Cass, A.E.G. Dual Recognition Element Lateral Flow Assay Toward Multiplex Strain Specific Influenza Virus Detection. Anal. Chem. 2017, 89, 6781–6786. [Google Scholar] [CrossRef] [Green Version]
- Yamamoto, R.; Baba, T.; Kumar, P.K. Molecular beacon aptamer fluoresces in the presence of Tat protein of HIV-1. Genes Cells 2000, 5, 389–396. [Google Scholar] [CrossRef]
- Jung, J.I.; Han, S.R.; Lee, S.W. Development of RNA aptamer that inhibits methyltransferase activity of dengue virus. Biotechnol. Lett. 2018, 40, 315–324. [Google Scholar] [CrossRef]
- Lee, C.H.; Lee, Y.J.; Kim, J.H.; Lim, J.H.; Kim, J.H.; Han, W.; Lee, S.H.; Noh, G.J.; Lee, S.W. Inhibition of hepatitis C virus (HCV) replication by specific RNA aptamers against HCV NS5B RNA replicase. J. Virol. 2013, 87, 7064–7074. [Google Scholar] [CrossRef] [Green Version]
- Zhou, J.; Li, H.; Li, S.; Zaia, J.; Rossi, J.J. Novel dual inhibitory function aptamer-siRNA delivery system for HIV-1 therapy. Mol. Ther. 2008, 16, 1481–1489. [Google Scholar] [CrossRef]
- Schmitz, A.; Weber, A.; Bayin, M.; Breuers, S.; Fieberg, V.; Famulok, M.; Mayer, G. A SARS-CoV-2 Spike Binding DNA Aptamer that Inhibits Pseudovirus Infection by an RBD-Independent Mechanism. Angew. Chem. 2021, 60, 10279–10285. [Google Scholar] [CrossRef] [PubMed]
- Zhang, L.; Fang, X.; Liu, X.; Ou, H.; Zhang, H.; Wang, J.; Li, Q.; Cheng, H.; Zhang, W.; Luo, Z. Discovery of sandwich type COVID-19 nucleocapsid protein DNA aptamers. Chem. Commun. (Camb) 2020, 56, 10235–10238. [Google Scholar] [CrossRef] [PubMed]
- Gupta, A.; Anand, A.; Jain, N.; Goswami, S.; Ananthraj, A.; Patil, S.; Singh, R.; Kumar, A.; Shrivastava, T.; Bhatnagar, S.; et al. A novel G-quadruplex aptamer-based spike trimeric antigen test for the detection of SARS-CoV-2. Mol. Nucleic Acids 2021. [Google Scholar] [CrossRef] [PubMed]
- Biospectrum. VIT to Develop Aptamer-Based POC Diagnostic Kit for COVID-19. Available online: https://www.biospectrumindia.com/news/58/18389/vit-to-develop-aptamer-based-poc-diagnostic-kit-for-covid-19.html (accessed on 28 April 2021).
- World Health Organisation. WHO Coronavirus (COVID-19) Dashboard. Available online: https://covid19.who.int/ (accessed on 26 May 2021).
- Iwagawa, T.; Ohuchi, S.P.; Watanabe, S.; Nakamura, Y. Selection of RNA aptamers against mouse embryonic stem cells. Biochimie 2012, 94, 250–257. [Google Scholar] [CrossRef] [PubMed]
- Yoon, J.W.; Jang, I.H.; Heo, S.C.; Kwon, Y.W.; Choi, E.J.; Bae, K.H.; Suh, D.S.; Kim, S.C.; Han, S.; Haam, S.; et al. Isolation of Foreign Material-Free Endothelial Progenitor Cells Using CD31 Aptamer and Therapeutic Application for Ischemic Injury. PLoS ONE 2015, 10, e0131785. [Google Scholar] [CrossRef] [PubMed]
- Kim, Y.; Wu, Q.; Hamerlik, P.; Hitomi, M.; Sloan, A.E.; Barnett, G.H.; Weil, R.J.; Leahy, P.; Hjelmeland, A.B.; Rich, J.N. Aptamer identification of brain tumor-initiating cells. Cancer Res. 2013, 73, 4923–4936. [Google Scholar] [CrossRef] [Green Version]
- Song, Y.; Zhu, Z.; An, Y.; Zhang, W.; Zhang, H.; Liu, D.; Yu, C.; Duan, W.; Yang, C.J. Selection of DNA aptamers against epithelial cell adhesion molecule for cancer cell imaging and circulating tumor cell capture. Anal. Chem. 2013, 85, 4141–4149. [Google Scholar] [CrossRef]
- Minakshi, P.; Kumar, R.; Ghosh, M.; Saini, H.M.; Ranjan, K.; Brar, B.; Prasad, G. Single-Cell Proteomics: Technology and Applications. In Single-Cell Omics; Academic Press: Cambridge, MA, USA, 2019; pp. 283–318. [Google Scholar]
- Su, Y.; Shi, Q.; Wei, W. Single cell proteomics in biomedicine: High-dimensional data acquisition, visualization, and analysis. Proteomics 2017, 17. [Google Scholar] [CrossRef] [PubMed]
- Sedykh, S.E.; Prinz, V.V.; Buneva, V.N.; Nevinsky, G.A. Bispecific antibodies: Design, therapy, perspectives. Drug Des. Devel. 2018, 12, 195–208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Labrijn, A.F.; Janmaat, M.L.; Reichert, J.M.; Parren, P. Bispecific antibodies: A mechanistic review of the pipeline. Nat. Rev. Drug Discov. 2019, 18, 585–608. [Google Scholar] [CrossRef]
- Kalra, P.; Dhiman, A.; Cho, W.C.; Bruno, J.G.; Sharma, T.K. Simple Methods and Rational Design for Enhancing Aptamer Sensitivity and Specificity. Front. Mol. Biosci. 2018, 5, 41. [Google Scholar] [CrossRef] [PubMed]
Product Name | Developer | Applications | Detection Mode | References |
---|---|---|---|---|
Aptocyto | Aptamer Science Inc. | Isolation of biomarker positive cells from a heterogeneous population using a magnetic bead | Flowcytometry-based detection | [38] |
Aptoprep | Aptamer Science Inc. | Aptamer-based protein (biomarker) pull down from sample using magnetic beads | Fluorescence-based detection | [38] |
AflaSense | Neoventerus Biotechnology Inc. | Fungal aflatoxin detection in food industry | Fluorescence-based detection | [38] |
CibusDx | CibusDx | Food-borne and water-borne pathogen detection | Electrochemical-based detection | [39] |
OLIGOBIND | Sekisui Diagnostics | Active Thrombin level detection in plasma sample | Fluorogenic activity-based detection | [40] |
OTASense | Neoventerus Biotechnology Inc. | Fungal Ochratoxin A detection in food industry | Fluorescence-based detection | [38] |
SOMAscan | SomaLogic | Novel biomarker detection associated with different diseases | SOMAmer-based detection | [41,42,43,44] |
Aptamer | Target | Medical Condition | Clinical Status | References | Trial Identifier (Clinical Trials.gov Identifier) |
---|---|---|---|---|---|
ARC1779 | von Willebrand factor | von Willebrand’s disease | Phase III (Awaiting) | [74,75,76] | NCT00432770 NCT00507338 NCT00632242 NCT00694785 NCT00726544 NCT00742612 |
ARC1905 | Complement Factor 5 (C5) | Neovascular age-related macular degeneration | Phase I | [77] | NCT00709527 NCT00950638 NCT02686658 NCT03362190 NCT03364153 NCT03374670 |
ARC19499 | Tissue Factor Pathway Inhibitor (TFPI) | Haemophilia | Terminated | [78] | NCT01191372 |
AS1411 | Nucleolin | Advanced solid tumors, Renal cell carcinoma, Acute myeloid leukaemia (AML) | Phase III (Awaiting) | [79,80] | NCT00512083 NCT00740441 NCT00881244 NCT01034410 |
E10030 | PDGF | Von Hippel Lindau disease, Age-related macular degeneration | Phase III (Awaiting) | [81,82] | NCT00569140 NCT01089517 NCT01940887 NCT01940900 NCT01944839 NCT02591914 NCT02214628 NCT02859441 |
EYE001 | VEGF | Wet age-related macular degeneration | Phase I (Completed) | [83,84] | NCT00021736 NCT00040313 NCT00056199 NCT00150202 NCT00239928 NCT00321997 NCT00736307 |
NOX-A12 | CXCL12 | Chronic lymphocytic Leukaemia, Multiple myelomas, Metastatic pancreatic and colorectal cancer | Phase II | [85,86] | NCT00976378 NCT01194934 NCT01486797 NCT01521533 NCT01947712 NCT03168139 |
NOX-E36 | CCL2 | Type II diabetes mellitus | Phase II | [87,88] | NCT00976729 NCT01085292 NCT01372124 NCT01547897 |
NOX-H94 | Hepcidin | Anaemia of chronic inflammation | Phase I (Completed) | [89] | NCT01372137 NCT01522794 NCT01691040 NCT02079896 |
NU172 | Thrombin | Coronary artery disease | Phase II | [90] | NCT00808964 |
Pegaptanib (Macugen) | VEGF165 | Age-related Macular Degeneration (AMD), Diabetic macular oedema, Uveitis, Diabetic cystoid oedema, Proliferative Diabetic Retinopathy (PDR) | In market | [70,91,92] | NCT00406107 NCT00549055 NCT00788177 NCT00790803 NCT01175070 NCT01486771 NCT01487070 NCT01573572 |
RB006 | Factor IX | Coronary artery disease | Phase III (Awaiting) | [93,94] | NCT00715455 NCT00932100 NCT01872572 |
REG1 System | Factor IX | Coronary artery disease, Acute coronary syndrome | Phase III (Terminated) | [95,96,97] | NCT00113997 NCT00715455 NCT00932100 NCT01848106 NCT02435082 NCT02797535 etc. |
Sgc8 | PTK7 | Colorectal cancer | Recruiting | [98,99] | NCT03385148 |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Basu, D.; Chakraborty, S.; Pal, R.; Sharma, T.K.; Sarkar, S. Identification and Engineering of Aptamers for Theranostic Application in Human Health and Disorders. Int. J. Mol. Sci. 2021, 22, 9661. https://doi.org/10.3390/ijms22189661
Basu D, Chakraborty S, Pal R, Sharma TK, Sarkar S. Identification and Engineering of Aptamers for Theranostic Application in Human Health and Disorders. International Journal of Molecular Sciences. 2021; 22(18):9661. https://doi.org/10.3390/ijms22189661
Chicago/Turabian StyleBasu, Debleena, Sourabrata Chakraborty, Riddhi Pal, Tarun Kumar Sharma, and Siddik Sarkar. 2021. "Identification and Engineering of Aptamers for Theranostic Application in Human Health and Disorders" International Journal of Molecular Sciences 22, no. 18: 9661. https://doi.org/10.3390/ijms22189661
APA StyleBasu, D., Chakraborty, S., Pal, R., Sharma, T. K., & Sarkar, S. (2021). Identification and Engineering of Aptamers for Theranostic Application in Human Health and Disorders. International Journal of Molecular Sciences, 22(18), 9661. https://doi.org/10.3390/ijms22189661