1,145
Views
57
CrossRef citations to date
0
Altmetric
Reviews

Aptamers: current challenges and future prospects

, , &
Pages 127-135 | Received 22 Oct 2015, Accepted 20 Nov 2015, Published online: 18 Dec 2015

References

  • Papers of special note have been highlighted as either of interest (•) or of considerable interest (••) to readers.
  • Tuerk C, Gold L. Systematic evolution of ligands by exponential enrichment: RNA ligands to bacteriophage T4 DNA polymerase. Science. 1990;249(4968):505–510.
  • Gordon EM, Barrett RW, Dower WJ, et al. Applications of combinatorial technologies to drug discovery. 2. Combinatorial organic synthesis, library screening strategies, and future directions. J Med Chem 1994 May 13;37(10):1385–1401
  • Smith GP. Filamentous fusion phage: novel expression vectors that display cloned antigens on the virion surface. Science. 1985;228(4705):1315–1317.
  • He M, Taussig MJ. Eukaryotic ribosome display with in situ DNA recovery. Nat Methods. 2007 Mar;4(3):281–288.
  • Stoltenburg R, Reinemann C, Strehlitz B. SELEX–a (r)evolutionary method to generate high-affinity nucleic acid ligands. Biomol Eng. 2007;24(4):381–403.

•• The authors describe the systematic evolution of ligands by exponential enrichment process for aptamer discovery and generated a table of aptamers developed until 2007. This review should be a starting point for those who are new in the aptamer field.

  • Seok Kim Y, Ahmad Raston NH, Bock Gu M. Aptamer-based nanobiosensors. Biosens Bioelectron. 2015. DOI: http://dx.doi.org/10.1016/j.bios.2015.06.040i

• This review offers a comprehensive lecture about the great potential of aptamers as biosensors and should be read to understand how aptamers can influence in the nanotechnology field.

•• The authors demonstrated experimentally the by-products formation after polymerase chain reaction (PCR) of a heterogeneous template and tried to understand the variables that influence the production of PCR artifacts.

  • Tolle F, Wilke J, Wengel J, et al. By-product formation in repetitive PCR amplification of DNA libraries during SELEX. PLoS ONE. 2014;9(12):e114693.
  • Schutze T, Wilhelm B, Greiner N, et al. Probing the SELEX process with next-generation sequencing. PLoS ONE. 2011;6(12):e29604.
  • Nimjee SM, Rusconi CP, Sullenger BA. Aptamers: an emerging class of therapeutics. Annu Rev Med. 2005;56:555–583.
  • Jayasena SD. Aptamers: an emerging class of molecules that rival antibodies in diagnostics. Clin Chem. 1999 Sep;45(9):1628–1650.
  • Hall B, Micheletti JM, Satya P, et al. Design, synthesis, and amplification of DNA pools for in vitro selection. Current Protocols in Molecular Biology. John Wiley & Sons, Inc; 2001.
  • Glokler J, Schutze T, Konthur Z. Automation in the high-throughput selection of random combinatorial libraries–different approaches for select applications. Molecules. 2010 Apr;15(4):2478–2490.
  • Rusconi CP, Yeh A, Lyerly HK, et al. Blocking the initiation of coagulation by RNA aptamers to factor VIIa. Thromb Haemost. 2000;84(5):841–848.
  • 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 Apr;22(2):143–152.
  • Oney S, Lam RT, Bompiani KM, et al. Development of universal antidotes to control aptamer activity. Nat Med. 2009 Oct;15(10):1224–1228.

• The authors described an approach to generate universal antidotes to control the activity aptamers targeting coagulation factor IXa and von Willebrand factor.

  • Rusconi CP, Scardino E, Layzer J, et al. RNA aptamers as reversible antagonists of coagulation factor IXa. Nature 2002 Sep 5;419(6902):90–94
  • Ramaswamy G, Slack FJ. siRNA. A guide for RNA silencing. Chem Biol. 2002 Oct;9(10):1053–1055.
  • McNamara JO 2nd, Andrechek ER, Wang Y, et al. Cell type-specific delivery of siRNAs with aptamer-siRNA chimeras. Nat Biotechnol. 2006;24(8):1005–1015.
  • Dassie JP, Liu XY, Thomas GS, et al. Systemic administration of optimized aptamer-siRNA chimeras promotes regression of PSMA-expressing tumors. Nat Biotechnol. 2009;27(9):839–849.
  • Thiel KW, Hernandez LI, Dassie JP, et al. Delivery of chemo-sensitizing siRNAs to HER2+-breast cancer cells using RNA aptamers. Nucl Acid Res. 2012 Jul;40(13):6319–6337.
  • Gourronc FA, Rockey WM, Thiel WH, et al. Identification of RNA aptamers that internalize into HPV-16 E6/E7 transformed tonsillar epithelial cells. Virology. 2013;446(1–2):325–333.
  • Zhou J, Bobbin ML, Burnett JC, et al. Current progress of RNA aptamer-based therapeutics. Front Genet. 2012;3:234.
  • Zhou J, Li H, Li S, et al. Novel dual inhibitory function aptamer-siRNA delivery system for HIV-1 therapy. Mol Ther. 2008;16(8):1481–1489.
  • Seeman NC. DNA in a material world. Nature. 2003;421(6921):427–431.
  • Goodman RP, Berry RM, Turberfield AJ. The single-step synthesis of a DNA tetrahedron. Chem Commun. 2004 Jun;21(12):1372–1373.
  • Liu Y, Lin C, Li H, et al. Aptamer-directed self-assembly of protein arrays on a DNA nanostructure. Angewandte Chemie (International Ed 2005 Jul 11;44(28):4333–4338
  • Shu Y, Cinier M, Shu D, et al. Assembly of multifunctional phi29 pRNA nanoparticles for specific delivery of siRNA and other therapeutics to targeted cells. Methods. 2011;54(2):204–214.
  • Zhou J, Shu Y, Guo P, et al. Dual functional RNA nanoparticles containing phi29 motor pRNA and anti-gp120 aptamer for cell-type specific delivery and HIV-1 inhibition. Methods. 2011;54(2):284–294.
  • Khaled A, Guo S, Li F, et al. Controllable self-assembly of nanoparticles for specific delivery of multiple therapeutic molecules to cancer cells using RNA nanotechnology. Nano Letters. 2005;5(9):1797–1808.
  • Niemeyer CM, Adler M. Nanomechanical devices based on DNA. Angewandte Chemie (International Ed). 2002;41(20):3779–3783.
  • Dittmer WU, Reuter A, Simmel FC. A DNA-based machine that can cyclically bind and release thrombin. Angewandte Chemie (International Ed). 2004;43(27):3550–3553.
  • Douglas SM, Bachelet I, Church GM. A logic-gated nanorobot for targeted transport of molecular payloads. Science. 2012;335(6070):831–834.
  • Liao WC, Lu CH, Hartmann R, et al. Adenosine triphosphate-triggered release of macromolecular and nanoparticle loads from aptamer/DNA-cross-linked microcapsules. ACS Nano; 2015 Sep 22;9(9):9078–9086.
  • Kanagawa T. Bias and artifacts in multitemplate polymerase chain reactions (PCR). J Biosci Bioeng. 2003;96(4):317–323.
  • Dutton CM, Paynton C, Sommer SS. General method for amplifying regions of very high G+C content. Nucleic Acids Res. 1993;21(12):2953–2954.
  • Margulies M, Egholm M, Altman WE, et al. Genome sequencing in microfabricated high-density picolitre reactors. Nature 2005 Sep 15;437(7057):376–380
  • Fedurco M, Romieu A, Williams S, et al. BTA, a novel reagent for DNA attachment on glass and efficient generation of solid-phase amplified DNA colonies. Nucleic Acids Res. 2006;34(3):e22.
  • Hoinka J, Berezhnoy A, Sauna ZE, et al. AptaCluster - a method to cluster HT-SELEX aptamer pools and lessons from its application. Res Comput Mol Biol. 2014;8394:115–128.
  • Hoinka J, Berezhnoy A, Dao P, et al. Large scale analysis of the mutational landscape in HT-SELEX improves aptamer discovery. Nucleic Acids Res 2015 Jul 13;43(12):5699–5707
  • Shao K, Ding W, Wang F, et al. Emulsion PCR: a high efficient way of PCR amplification of random DNA libraries in aptamer selection. PLoS ONE. 2011;6(9):e24910.
  • Szeitner Z, Andras J, Gyurcsanyi RE, et al. Is less more? Lessons from aptamer selection strategies. J Pharm Biomed Anal. 2014 Dec;101:58–65.
  • Gopinath SC. Methods developed for SELEX. Anal Bioanal Chem. 2007 Jan;387(1):171–182.
  • Cox JC, Ellington AD. Automated selection of anti-protein aptamers. Bioorg Med Chem. 2001 Oct;9(10):2525–2531.
  • Mendonsa SD, Bowser MT. In vitro evolution of functional DNA using capillary electrophoresis. J Am Chem Soc. 2004;126(1):20–21.
  • Berezovski M, Drabovich A, Krylova SM, et al. Nonequilibrium capillary electrophoresis of equilibrium mixtures: a universal tool for development of aptamers. J Am Chem Soc 2005 Mar 9;127(9):3165–3171
  • Lou X, Qian J, Xiao Y, et al. Micromagnetic selection of aptamers in microfluidic channels. Proc Natl Acad Sci U S A. 2009;106(9):2989–2994.
  • Cho M, Xiao Y, Nie J, et al. Quantitative selection of DNA aptamers through microfluidic selection and high-throughput sequencing. Proc Natl Acad Sci U S A 2010 Aug 31;107(35):15373–15378
  • Qian J, Lou X, Zhang Y, et al. Generation of highly specific aptamers via micromagnetic selection. Anal Chem 2009 Jul 1;81(13):5490–5495
  • Oh SS, Ahmad KM, Cho M, et al. Improving aptamer selection efficiency through volume dilution, magnetic concentration, and continuous washing in microfluidic channels. Anal Chem 2011 Sep 1;83(17):6883–6889
  • Park JW, Tatavarty R, Kim DW, et al. Immobilization-free screening of aptamers assisted by graphene oxide. Chem Commun 2012 Feb 18;48(15):2071–2073
  • Riley KR, Saito S, Gagliano J, et al. Facilitating aptamer selection and collection by capillary transient isotachophoresis with laser-induced fluorescence detection. J Chromatogr. 2014 Nov 14;1368:183–189
  • Jing M, Bowser MT. Isolation of DNA aptamers using micro free flow electrophoresis. Lab on a Chip. 2011;11(21):3703–3709.
  • Gold L, Ayers D, Bertino J, et al. Aptamer-based multiplexed proteomic technology for biomarker discovery. PLoS ONE. 2010;5(12):e15004.

• The first author was the first to describe the systematic evolution of ligands by exponential enrichment process and he is the founder of Somalogic. In this article, the authors applied their slow off-rate modified aptamer technology for the development of a proteomic assay to identify clinically relevant biomarkers.

  • Fujita S, Arinaga K, Fujihara T, et al. Novel protein detection system using DNA as constituent material. Fujitsu Sci Tech J. 2012 April;48(2):237–243.

• This article describes the technology developed by Apta Biosciences and shows how modified nucleic acids can result in the development of a solid biomarker discovery platform.

  • Williams KP, Liu XH, Schumacher TN, et al. Bioactive and nuclease-resistant L-DNA ligand of vasopressin. Proc Natl Acad Sci U S A 1997 Oct 14;94(21):11285–11290
  • Klussmann S, Nolte A, Bald R, et al. Mirror-image RNA that binds D-adenosine. Nat Biotechnol. 1996;14(9):1112–1115.
  • Green LS, Jellinek D, Bell C, et al. Nuclease-resistant nucleic acid ligands to vascular permeability factor/vascular endothelial growth factor. Chem Biol. 1995;2(10):683–695.
  • Diafa S, Hollenstein M. Generation of aptamers with an expanded chemical repertoire. Molecules. 2015;20(9):16643–16671.

•• This review describes the usage of modified nucleotides in the context of aptamers. It is a recommended lecture for those who are new in the field of aptamers and wants to understand how modified nucleotides can lead to novel aptamer-based technologies.

  • Eaton BE. The joys of in vitro selection: chemically dressing oligonucleotides to satiate protein targets. Curr Opin Chem Biol. 1997 Jun;1(1):10–16.
  • Loakes D, Holliger P. Polymerase engineering: towards the encoded synthesis of unnatural biopolymers. Chem Commun. 2009 Aug;21(31):4619–4631.
  • Pinheiro VB, Taylor AI, Cozens C, et al. Synthetic genetic polymers capable of heredity and evolution. Science 2012 Apr 20;336(6079):341–344
  • Chushak Y, Stone MO. In silico selection of RNA aptamers. Nucleic Acids Res. 2009 Jul;37(12):e87.
  • Asai R, Nishimura SI, Takahashi K. DNA aptamers that recognize fluorophore using on-chip screening in combination with an in silico evolution. Nucleic Acids Res. 2003;3:321–322.
  • Maurer K, Yazvenko N, Wilmoth J, et al. Use of a multiplexed CMOS microarray to optimize and compare oligonucleotide binding to DNA probes synthesized or immobilized on individual electrodes. Sensors. 2010;10(8):7371–7385.
  • Cho M, Soo Oh S, Nie J, et al. Quantitative selection and parallel characterization of aptamers. Proc Natl Acad Sci U S A 2013 Nov 12;110(46):18460–18465
  • Cagnin S, Caraballo M, Guiducci C, et al. Overview of electrochemical DNA biosensors: new approaches to detect the expression of life. Sensors. 2009;9(4):3122–3148.

•• This review is highly suggested to understand one of the technologies that can lead to a solid high-throughput and automated aptamer discovery platform. The authors describe current technologies for the generation of DNA microarrays and those that can be adapted for aptamer discovery are mentioned.

•• This review is highly suggested to understand the technology behind next-generation sequencing systems. The authors explain in a comprehensive fashion Illumina’s and Life Technologies system among others.

  • Merriman B, Ion Torrent R, Team D, et al. Progress in ion torrent semiconductor chip based sequencing. Electrophoresis. 2012;33(23):3397–3417.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.