<<

International Journal of Molecular Sciences

Review Chemical Modifications of Aptamers for Therapeutic Purposes

Shuaijian Ni 1,2,† ID , Houzong Yao 1,2,†, Lili Wang 1,2,†, Jun Lu 1,2, Feng Jiang 1,2,3,*, Aiping Lu 1,2,* and Ge Zhang 1,2,* ID

1 Institute of Precision Medicine and Innovative Drug Discovery, School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong 999077, China; [email protected] (S.N.); [email protected] (H.Y.); [email protected] (L.W.); [email protected] (J.L.) 2 Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong 999077, China 3 Faculty of Materials Science and Chemical Engineering, the State Key Laboratory Base of Novel Functional Materials and Preparation Science, Ningbo University, Ningbo 315211, Zhejiang, China * Correspondence: [email protected] (F.J.); [email protected] (A.L.); [email protected] (G.Z.); Tel.: +852-3411-2456 (A.L.); +852-3411-2958 (G.Z.) † These authors contributed equally to this work.

Received: 10 July 2017; Accepted: 1 August 2017; Published: 2 August 2017

Abstract: Nucleic acid aptamers have minimal immunogenicity, high chemical synthesis production, low cost and high chemical stability when compared with . However, the susceptibility to degradation, rapid excretion through renal filtration and insufficient binding affinity hindered their development as drug candidates for therapeutic applications. In this review, we will discuss methods to conquer these challenges and highlight recent developments of chemical modifications and technological advances that may enable early aptamers to be translated into clinical therapeutics.

Keywords: nucleic acid aptamer; nuclease degradation; rapid excretion; binding affinity; chemical modification

1. Introduction In 1990, several groups isolated the first nucleic acid aptamers by “SELEX” (Systematic Evolution of Ligands by Exponential Enrichment) or “in vitro selection” (a demarcation resulting from whether the technique was learned from Tuerk and Gold [1] or Ellington and Szostak [2], respectively). Through 3D conformational complementarities, aptamers bind to a wide range of targets, including small metal ions and organic molecules, , , viruses, , whole cells and even targets within live animals [3]. Being similar to the binding of antibodies and antigens, the binding between aptamer and its target has comparable binding affinity and specificity, which makes aptamers a promising class of therapeutic alternatives to antibodies [4]. In addition, nucleic acid aptamers have minimal immunogenicity, high chemical synthesis production, low cost and high chemical stability, drawing extensive attention of researchers to the development of aptamer therapeutics [5]. However, the susceptibility to nuclease degradation and rapid excretion through renal filtration severely limit the practical usage of aptamers [6,7]. Many aptamers with potent activities have unacceptable short half-lives in vivo [8,9]. Besides, the binding affinity and specificity of unmodified nucleic acid aptamers are sometimes insufficient for successful implementation as therapeutic agent [10]. The generation of high quality aptamers from conventional SELEX is generally below

Int. J. Mol. Sci. 2017, 18, 1683; doi:10.3390/ijms18081683 www.mdpi.com/journal/ijms Int. J. Mol. Sci. 2017, 18, 1683 2 of 21

30% [11]. Therefore, many attempts of post-SELEX chemical modifications should be done in order to solveInt. J. Mol. these Sci. challenges2017, 18, 1683(Figure 1). 2 of 20

Figure 1. TheThe common common strategies strategies in inthethe chemical chemical modifi modificationscations of nucleic of nucleic acid acidaptamers aptamers and their and theirpurposes. purposes. Among Among the modifications, the modifications, such such as modi as modificationsfications on onthe the terminals terminals of of nucleic nucleic acids, modificationsmodifications on the phosphodiester linkage, modifi modificationscations on the sugar ring and modificationsmodifications on the bases, the 3′0 end capping with inverted thymidine [6,12] [6,12] and PEGylation [13] [13] have been the common strategies in the chemical modificationsmodifications of nucleic acid aptamers for developmentdevelopment clinical therapeutics [[14–17].14–17].

In this review, the standard synthetic method of solid phase phosphoramidite chemistry for In this review, the standard synthetic method of solid phase phosphoramidite chemistry for nucleic nucleic acid aptamers preparation will be introduced firstly [18,19]. Then, the chemical modification acid aptamers preparation will be introduced firstly [18,19]. Then, the chemical modification strategies strategies of aptamers for resisting nuclease degradation [12,20–26], improving target binding of aptamers for resisting nuclease degradation [12,20–26], improving target binding affinities [10,27–31] affinities [10,27–31] and resisting renal clearance [32–36] will be summarized, sequentially. Among and resisting renal clearance [32–36] will be summarized, sequentially. Among the modifications, the modifications, such as modifications on the terminals of nucleic acids, modifications on the such as modifications on the terminals of nucleic acids, modifications on the phosphodiester linkage, phosphodiester linkage, modifications on the sugar ring and modifications on the bases, the 3′ end modifications on the sugar ring and modifications on the bases, the 30 end capping with inverted capping with inverted thymidine [6,12] and PEGylation [13] have been the common strategies in thymidine [6,12] and PEGylation [13] have been the common strategies in the chemical modifications the chemical modifications of nucleic acid aptamers for development clinical therapeutics (e.g., of nucleic acid aptamers for development clinical therapeutics (e.g., [14–17], etc.). pegaptanib [14–17], etc.). More excitingly, aptamers with improved binding affinities are being More excitingly, aptamers with improved binding affinities are being generated with modifications on generated with modifications on the bases [29] or substitutions of two non-bridging phosphate the bases [29] or substitutions of two non-bridging phosphate oxygen atoms in nucleic acids by sulfur oxygen atoms in nucleic acids by sulfur replacement [10] (see “SOMAmers” and “PS2 walk” replacement [10] (see “SOMAmers” and “PS2 walk” below). below). 2. Chemical Synthesis of Nucleic Acid Aptamers 2. Chemical Synthesis of Nucleic Acid Aptamers 2.1. Synthesis of DNA Aptamers 2.1. Synthesis of DNA Aptamers DNA aptamers can be synthesized through the classic solid phase phosphoramidite four-step processDNA on aptamers the automated can be DNA synthesized synthesizer through [18]. the classic solid phase phosphoramidite four-step processThe on four-step the automated method DNA is shown synthesizer in Figure [18].2. First, the 4,4 0-Dimethoxytriphenylmethyl (DMT) groupThe is removedfour-step frommethod the deoxynucleosideis shown in Figure (50 -end)2. First, which the is4,4 linked′-Dimethoxytriphenylmethyl to the control pore glass (DMT) (CPG) columns.group is removed Large excess from of the acid deoxynucleoside solution (trichloroacetic (5′-end) acidwhich (TCA)) is linked could to be the used control for the pore deprotection glass (CPG) of DMT.columns. In the Large second excess step ofof the acid cycle, solution an internucleotide (trichloroacetic bond calledacid phosphite(TCA)) could trimester be isused synthesized. for the Then,deprotection in the thirdof DMT. step, In the the reaction second product step of fromthe cycle, Step 2an should internucleotide be treated bond with cappingcalled phosphite agent to trimester is synthesized. Then, in the third step, the reaction product from Step 2 should be treated with capping agent to cap the unreacted free 5′-OH group. In the last step (Step 4), the new phosphite is oxidized to the corresponding phosphotriester by iodine. The cycle is repeated, once for each base, to produce the required . Finally, the nucleic acid aptamers could be Int. J. Mol. Sci. 2017, 18, 1683 3 of 21 cap the unreacted free 50-OH group. In the last step (Step 4), the new phosphite is oxidized to the correspondingInt. J. Mol. Sci. 2017, phosphotriester 18, 1683 by iodine. The cycle is repeated, once for each base, to produce3 of 20 the required oligonucleotide. Finally, the nucleic acid aptamers could be cleaved from the CPG by concentratedcleaved from ammonium the CPG hydroxide.by concentrated The protecting ammoni groupsum hydroxide. for phosphates The protecting and heterocyclic groups bases for couldphosphates be removed and heterocyclic at the same bases time could [18,37 ,be38 ].removed at the same time [18,37,38].

Figure 2. Four-stepFour-step phosphoramidite phosphoramidite oligodeoxynucleotid oligodeoxynucleotidee synthesis synthesis cycle cycle (adapted (adapted from from [18]). [ 18The]). Thephosphoramidite phosphoramidite method, method, pioneered pioneered by Marvin by Marvin Caruthers Caruthers in the inea therly 1980s, early 1980s,and enhanced and enhanced by the byapplication the application of solid-phase of solid-phase technology technology and automation, and automation, is now is nowfirmly firmly established established as the as themethod method of ofchoice. choice. Phosphoramidite Phosphoramidite oligonucleot oligonucleotideide synthesis synthesis proceeds proceeds in the in the3′ to 3 05to′ direction 50 direction (opposite (opposite to the to the5′ to 5 03to′ direction 30 direction of ofDNA DNA biosynthesis biosynthesis in inDNA DNA replication). replication). One One nucleotide nucleotide is is added added per synthesis cycle. The phosphoramidite DNA synthesissynthesis cyclecycle consistsconsists ofof aa seriesseries ofof stepssteps outlinedoutlined inin thethe figure.figure.

At present, the application of four-step method is very common. For the most part, progress in At present, the application of four-step method is very common. For the most part, progress in the solid phase nucleic acid synthesis field has not changed this fundamental approach. For R&D the solid phase nucleic acid synthesis field has not changed this fundamental approach. For R&D purposes, shortened aptamers with 20 to 50 nucleotides in length can be generated in individual purposes, shortened aptamers with 20 to 50 nucleotides in length can be generated in individual labs labs using “lab scale” DNA or RNA synthesizers [39] (e.g., Expedite 8909, ABI394). using “lab scale” DNA or RNA synthesizers [39] (e.g., Expedite 8909, ABI394).

2.2. Synthesis of RNA Aptamers Several ofof synthetic synthetic strategies strategies for thefor solid-phasethe solid-phase synthesis synthesis of RNA of had RNA been had reported been[ 19reported,40–42]. Among[19,40–42]. the Among combinations the combinations of different of coupling/activation different coupling/activation chemistries chemistries and protecting and groupsprotecting for thegroups 20-hydroxyl for the 2′-hydroxyl and exocyclic and amineexocyclic groups, amine thegroups, tert-butyldimethylsilyl the tert-butyldimethylsilyl protection protection of the riboseof the 2ribose0-hydroxyl 2′-hydroxyl group combinedgroup combined with the with standard the standard protecting protecting groups groups for the exocyclicfor the exocyclic amine groupsamine (benzoylgroups (benzoyl for adenosine, for adenosine, acetyl ac foretyl cytidine, for cytidine, and and isobutyryl isobutyryl for for guanosine) guanosine) were were mostmost widely used [[40,43].40,43]. Phosphoramidite Phosphoramidite monomers monomers were were usually usually activated with 4,5-dicyanoimidazole,4,5-dicyanoimidazole, 5-ethylthio-1H-tetrazole (ETT) (ETT) or or 5-benzylthio-1 5-benzylthio-1HH-tetrazole-tetrazole (BTT) (BTT) (Figure (Figure 3).3). The The solid solid supports supports for forRNA RNA synthesis synthesis were were polymeric polymeric supports supports or orCPGs CPGs with with different different linkers linkers and and pore pore sizes. The finalfinal productproduct couldcould be be cleaved cleaved from from the the CPG CPG by concentrated by concentrated ammonium ammonium hydroxide. hydroxide. The protection The protection groups cangroups also can be removedalso be removed at the same at the time same [19 time,40,44 [19,40,44–46].–46]. Int. J. Mol. Sci. 2017, 18, 1683 4 of 21 Int.Int. J.J. Mol.Mol. Sci.Sci. 2017,, 18,, 16831683 4 of 20

Figure 3. Solid-phaseSolid-phase RNA RNA synthesis synthesis viavia the the phosphoram phosphoramiditephosphoramiditeidite methodmethod (adapted(adapted fromfrom [19]).[19]). [19]). InIn RNARNA synthesis,synthesis, thethe 2220′′-hydroxy-hydroxy groupgroupgroup is isis protected protectedprotected with withwith TBDMS TBDMSTBDMS (t -butyldimethylsilyl)((tt-butyldimethylsilyl)-butyldimethylsilyl) group, group,group, which whichwhich can cancan be beremoved removed by by treatment treatment with with fluoride fluoride ion. ion.

3. Modifications Modifications of Nucleic Acid AptamersAptamers

3.1. Aptamer Aptamer Derivatives Derivatives for for Resisting Resisting Nuclease Degradation

3.1.1. Terminal Terminal 3 ′0′–3′′0 andand 55 ′′0–5–5′′0 InternucleotideInternucleotideInternucleotide LinkageLinkage Linkage The 3 ′0′–3–3′′0 andand 55 ′′0–5–5′′ 0 inversionsinversionsinversions werewere were testedtested tested inin in 19911991 1991 byby SeligerSeliger etet al. al. [12]. [[12].12]. The The 3 3′0′-end-end cappingcapping with with invertedinverted thymidinethymidinethymidine has hashas also alsoalso been beenbeen a common aa commoncommon strategy strastrategytegy among amongamong aptamers aptamersaptamers for diseases forfor diseasesdiseases therapy therapytherapy in ongoing inin ongoingor completed or completed clinical trials clinical [15,47 trials]. Research [15,47]. suggested Research that suggested 30-inverted that dT 3′′ modification-inverted-inverted dTdT could modificationmodification increase couldthe stability increase and the resistance stability and of aptamers resistance to of 30 -exonucleaseaptamers to 3 in′′-exonuclease-exonuclease human serum. inin humanhuman Synthesis serum.serum. of 3 0SynthesisSynthesis-inverted ofdT 3 modified′′-inverted-invertedaptamers dTdT modifiedmodified (Figure aptamersaptamers4) needed (Figur(Figur modifiede 4) needed CPG withmodified the 5 CPG0-hydroxyl with the of the5′′-hydroxyl-hydroxyl first nucleoside ofof thethe firstfirstattached, nucleosidenucleoside followed attached,attached, by chain followedfollowed elongation byby chainchain in standard elongationelongation 30→ inin50 standardfashionstandard [12 33′′,→205,21′′ fashionfashion]. [12,20,21].[12,20,21].

Figure 4. Solid-phaseSolid-phase synthesissynthesis of of 3′0′-inverted-inverted dT dT modifiedmodifiedmodified aptamers.aptamers. SynthesisSynthesis ofof 3 3′0′-inverted-inverted dTdT modifiedmodified aptamersaptamers needs needs modified modified CPG CPG with with the 5the0-hydroxyl 5′′-hydroxyl-hydroxyl of the ofof first thethe nucleoside firstfirst nucleoside attached, attached, followed followedfollowedby chain elongationbyby chainchain elongationelongation in standard inin standard 3standard0→50 fashion. 33′′→5′′ fashion.fashion. Int. J. Mol. Sci. 2017, 18, 1683 5 of 20 Int. J. Mol. Sci. Sci. 20172017,, 1818,, 1683 1683 5 of 20 21

3.1.2. 3′- Conjugates 3.1.2. 30-Biotin Conjugates In some ways, 3′-biotin (Figure 5) could resist the activity of 3′-exonuclease, which was similar 0 0 to 3′-invertedIn some ways, dT modification. 3 -biotin (Figure Dougan5) could et al. resist [36] the investigated activity of 3the-exonuclease, 3′-biotin-streptavidin which was conjugates similar to 0 0 of3 -inverted the dT modification. aptamer to Douganfind that et the al. [336′-biotin] investigated rendered the resistance 3 -biotin-streptavidin to the 3′-exonuclease conjugates in of the 0 0 bloodthrombin of mouse aptamer or to rabbits. find that In the addition, 3 -biotin the rendered 3′-biotin-streptavidin resistance to the conjugates 3 -exonuclease slowed in the down blood the of 0 clearancemouse or rate rabbits. of aptamers In addition, in blood the 3 circulation-biotin-streptavidin system in conjugatesvivo [36]. A slowed similar down 3′-biotin the approach clearance was rate 0 alsoof aptamers used to protect in blood the circulation DNA aptamer system targetingin vivo the[36 ].SARS A similar coronavirus 3 -biotin helicase approach for up was to also31 and used 16 toh protectin 5% and the 10% DNA fetal aptamer bovine targeting serum, the whereas SARS coronavirusthe original helicaseaptamer for can up only to 31 sustain and 16 half h in of 5% that and time 10% [20].fetal bovine serum, whereas the original aptamer can only sustain half of that time [20].

Figure 5. Structure of the 3′-biotin conjugate. 3′-Biotin could inhibit the activity of 3′-exonuclease, Figure 5. Structure of the 330′-biotin conjugate.conjugate. 3 3′0-Biotin-Biotin could could inhibit inhibit the the activity of 30′-exonuclease, which was similar to 3′-inverted dT modification. In addition, the 3′-biotin conjugates slowed down which was similar to 330′-inverted dT modification.modification. In addition, the 330′-biotin conjugates slowed down the clearance rate in blood circulation system in vivo. the clearance rate in blood circulation systemsystem inin vivo.vivo.

3.1.3. Modifications on the Sugar Ring 3.1.3. Modifications Modifications on the Sugar Ring 2′-Substitutions 2′0-Substitutions 0 0 0 0 ModificationsModifications to the sugars such as 2 ′-fluoro-fluoro (2 ′-F) or 2 ′-amino-amino (2 (2′-NH-NH22)) ribose groups groups (Figure (Figure 66)) on the residues have been available fo forr incorporation into enzymatically derived nucleic 0 acids for some years. Although both are effective at improving serum half-life, 22′-F modificationsmodifications 0 quickly garnered favor over 2′-NH2 duedue to the increased coupling efficiencyefficiency during solid-phase 0 synthesis, and elimination of extraextra deprotectiondeprotection stepssteps duringduring 22′-NH-NH2 purification.purification. The The more bulky 0 0 2′--O-methylO-methyl (2′-OMe) modificationsmodifications havehave been been previously previously used used as as a a post-selection post-selection modification modification due due to totheir their increased increased nuclease nuclease resistance resistance and and high high duplex duplex melting melting temperature temperature which which could could be seenbe seen in the in theclinical clinical examples examples [48, 49[48,49].].

Figure 6. 22′0-substitutions-substitutions utilized utilized to to enhance enhance the the stability stability of of aptamers aptamers inin vivo vivo (adapted(adapted from from [39]). [39]). 2′0-Substitutions-Substitutions can can easily easily be be incorporated incorporated into into ap aptamerstamers during during chemical chemical synthesis synthesis and and include: include: (i) 0 0 0 0 0 2(i′)-H; 2 -H; (ii) (2ii′-OH;) 2 -OH; (iii) ( iii2′-NH) 2 -NH2; (iv2;() 2iv′-F;) 2 and-F; and (v) 2 (v′-OMe.) 2 -OMe.

LNA, UNA, 2 ′0-F-F ANA Locked nucleic acid (LNA) (Figure 77)) isis anan analoganalog ofof ribonucleotide ribonucleotide withwith aa methylenemethylene linkagelinkage between 2 ′0-O-O and 4 ′0-C-C of the sugar ring. This modificationmodification showed great resistance to and increased thermostability thus thus could could be be used used to to generate the the most stable pairs [50,51]. [50,51]. Darfeuille Darfeuille et et al. al. also found that thethe LNA/DNALNA/DNA chimera chimera LNA5, LNA5, a stablestable complex that against HIV-1 trans-activatingtrans-activating response (TAR) (TAR) RNA, RNA, was was able able to tomaintain maintain the the intact intact structure structure within within 20 h 20in bovine h in bovine serum serum [52]. Shi [52 et]. al. developed a new LNA/DNA chimeric aptamer probe through proper LNA incorporation and Int. J. Mol. Sci. 2017, 18, 1683 6 of 20

Int. J. Mol. Sci. 2017, 18, 1683 6 of 21 3′-3′-thymidine (3′-3′-T) capping. The serum stability of original aptamer was gradually enhanced while its specificity and affinity were perfectly maintained. Especially TD05.6 aptamer which had a 7-baseShi pair-LNA et al. developed substitution a new LNA/DNA exhibited chimerica ten-fold aptamer elevated probe stability through in proper serum LNA and incorporation a much slower 0 0 0 0 clearanceand 3 rate-3 -thymidine in mice [53]. (3 -3 -T) capping. The serum stability of original aptamer was gradually enhanced whileUnlike its specificityLNA, a andstructurally affinity were rigid perfectly modificati maintained.on that Especially increases TD05.6 the aptamerthermostability which had of a a 7-base pair-LNA substitution exhibited a ten-fold elevated stability in serum and a much slower modified-oligonucleotide thus protects it from nucleases degradation in cells, unlocked nucleic acid clearance rate in mice [53]. (UNA) (Figure 7) in which a bond between C2′ and C3′ of the sugar ring was absent makes aptamers Unlike LNA, a structurally rigid modification that increases the thermostability of morea modified-oligonucleotideflexible [54]. Due to its nature thus protects of flexibility, it from nucleases UNA could degradation alleviate in strain cells, unlocked in tight nucleicloop structures. acid Pasternak(UNA) et (Figure al. found7) in which that UNA a bond modifications between C2 0 and on C3the0 of loop the sugarregions ring of was a 15-mer absent makes thrombin aptamers targeted DNAmore aptamer flexible increased [54]. Due its to itsthermo naturedynamic of flexibility, stability. UNA However, could alleviate modifications strain in tight within loop structures.the G-quartet structuresPasternak were et al.unfavorable found that UNAfor quadruplex modifications format on theion loop [55]. regions They of also a 15-mer demonstrated thrombin targeted that UNA DNA could be placedaptamer in increased many itspositions thermodynamic without stability. affecting However, the thrombin-binding modifications within affi thenity G-quartet and structuresanticoagulant efficiencywere unfavorableof the aptamer for quadruplex [55]. formation [55]. They also demonstrated that UNA could be placed inIt manyhas been positions found without that affectingmodifications the thrombin-binding at the 2′-position affinity of andthe anticoagulantsugar ring would efficiency bring of the about aptamer [55]. different effects on thermostability based on th0 e molecularity of G-quadruplex. Peng et al. discoveredIt hasthat, been in found both thatanti-HIV modifications phosphorot at thehioate 2 -position aptamer of the and sugar thrombin-binding ring would bring aboutaptamer, different effects on thermostability based on the molecularity of G-quadruplex. Peng et al. discovered substitution of guanines (G) that adopted anti-conformation with 2′-F-G could maintain the that, in both anti-HIV phosphorothioate aptamer and thrombin-binding aptamer, substitution of quadruplexguanines conformation, (G) that adopted while anti-conformation substituting with guanin 20-F-Ges couldwith maintainsyn-conformation the quadruplex was conformation,not favored [25]. Morewhile importantly, substituting two guanines 2′-F-modified with syn-conformation thrombin-binding was notaptamers favored (PG13 [25]. Moreand importantly,PG14) showed approximatelytwo 20-F-modified four-fold thrombin-binding increased binding aptamers affinity (PG13 andto thrombin PG14) showed and approximatelyup to seven-fold four-fold higher nucleaseincreased resistance. binding As affinity a result, to thrombin the 2′-deoxy-2 and up to′-fluoro- seven-foldD-arabinonucleic higher nuclease acid resistance. (2′-F ANA) As a result,(Figure 7) 0 0 0 modificationthe 2 -deoxy-2 was-fluoro- very suitableD-arabinonucleic for improving acid (2 -F the ANA) biological (Figure7) modificationand physicochemical was very suitable properties for of DNAimproving G-quartets the [25]. biological and physicochemical properties of DNA G-quartets [25].

FigureFigure 7. 7.StructuresStructures of ofLocked Locked nucleic nucleic acid acid (LNA), unlockedunlocked nucleicnucleic acid acid (UNA) (UNA) and and 0 0 0 2′-deoxy-22 -deoxy-2′-fluoro--fluoro-D-arabinonucleicD-arabinonucleic acid acid (2 (2′-F-F ANA). ANA). LNA LNA is an analoganalog ofof ribonucleotideribonucleotide with with a 0 0 0 methylenea methylene linkage linkage between between 2′-O 2and-O 4 and′-C 4of-C the of sugar the sugar ring. ring. UNA UNA misses misses a bond a bond between between C2′ C2and C3′ 0 0 0 of theand sugar C3 of ring. the sugar2′-F ANA ring. adopts 2 -F ANA anti-conformation adopts anti-conformation with 2′-F-G. with 2 -F-G.

3.1.4.3.1.4. Modifications Modifications on onthe the Phosphodiester Phosphodiester Linkage Linkage Methylphosphonate or Phosphorothioate Methylphosphonate or Phosphorothioate Replaced phosphodiester linkage of DNA with methylphosphonate or phosphorothioate analog isReplaced commonly phosphodiester used for aptamer linkage modification. of DNA Thermodynamic with methylphosphonate studies revealed or that phosphorothioate loss of the negative analog is commonlycharge of theused phosphate for aptamer backbone, modification. as the methylphosphonate Thermodynamic analog studies (Figure revealed8), destabilized that loss the of the negativeG-quadruplex charge of structure the phosphate [56]. The backbone, ionic radii ofas thethe oligonucleotide methylphosphonat backbonee analog atoms (Figure also have 8), an destabilized impact the G-quadruplexin the stabilization structure of G-quadruplex [56]. The ionic structures. radii Saccaof the et oligonucleotide al. found that substitution backbone of atoms the phosphate also have an impactbackbone in the atom stabilization O with S (phosphorothioateof G-quadruplex analog, structures. Figure Sacca8) might et influenceal. found the that thermal substitution stability ofof the phosphatethe G-quadruplex backbone structureatom O with in a molecularity-dependentS (phosphorothioate analog, manner Figure [56]. 8) might influence the thermal stability ofThe the thermodynamic G-quadruplex stability structure of in the a phosphodiestermolecularity-dependent linkage of manner the thrombin-binding [56]. aptamer d(GGTTGGTGTGGTTGG) with thiophosphoryl substitutions at different internucleotide sites were The thermodynamic stability of the phosphodiester linkage of the thrombin-binding aptamer studied [23,24]. Complete substitution by thiophosphorylated was limited as their d(GGTTGGTGTGGTTGG) with thiophosphoryl substitutions at different internucleotide sites were studied [23,24]. Complete substitution by thiophosphorylated oligonucleotides was limited as their high toxicities, so partial substitutions with the maximum thermal stability were selected for evaluating their stabilities under conditions of nuclease RQ1 DNAse hydrolysis and their antithrombin activities in blood plasma [24]. Aptamer d(GGSTSTSGGTGTGGSTSTSGG) with Int. J. Mol. Sci. 2017, 18, 1683 7 of 21

Int. J. Mol. Sci. 2017, 18, 1683 7 of 20 high toxicities, so partial substitutions with the maximum thermal stability were selected for evaluating thio-substitutionsInt. J. theirMol. Sci. stabilities 2017, 18 , underin 1683 both conditions TT loops of nucleaseexhibited RQ1 similar DNAse antithrombin hydrolysis and efficiency their antithrombin to the activitiesunmodified7 of 20 aptamerin blood but plasmabetter resistance [24]. Aptamer to the d(GGSTSTSGGTGTGGSTSTSGG) degradation of DNA nuclease with in blood thio-substitutions serum [23]. in both TT thio-substitutionsloops exhibited similarin both antithrombin TT loops exhibited efficiency tosimilar the unmodified antithrombin aptamer efficiency but better resistanceto the unmodified to the aptamerdegradation but better of DNA resistance nuclease to inthe blood degradat serumion [23 of]. DNA nuclease in blood serum [23].

Figure 8. Structures of methylphosphonate and phosphorothioate.

More recently,Figure phosphorodiothioateFigure 8. Structures 8. Structures of methylphosph of methylphosphonatelinkages (PS2)onate wereand and phosphorothioate. phosphorothioate.employed to stabilize phosphate backbone. The substitution of both non-bridging oxygen atoms with sulfur could give rise to a phosphorodithioateMoreMore recently, recently, phosphorodiothioatelinkage, phosphorodiothioate which, similar linkages linkages to natural (PS2) (PS2) wereDNA, were employed employedis achiral to to stabilizeat stabilize phosphorus. phosphate phosphate In addition,backbone.backbone. it The was substitution The reported substitution that of bothPS2 of bothsubstitutions non-bridging non-bridging dramatically oxygen oxygen atoms atoms improved with with sulfur sulfurtarget could couldbinding give give riseaffinity rise to toa byphosphorodithioate ~1000-folda phosphorodithioate (see PS2 linkage, walk linkage, below) wh which,ich, [10]. similar similar toto naturalnatural DNA, DNA, is achiral is achiral at phosphorus. at phosphorus. In addition, In addition,it was it reported was reported that PS2 that substitutions PS2 substitutions dramatically dramatically improved targetimproved binding target affinity binding by ~1000-fold affinity Replacedby ~1000-fold(see PS2 by walkTriazole (see below) PS2 walk [10]. below) [10]. ReplacedReplacement by Triazole of the oligonucleotide phosphodiester linkage with triazole linkages has shown Replaced by Triazole great promiseReplacement [57–60]. of These the oligonucleotide triazole analogs phosphodiester can be obtained linkage withthrough triazole automated linkages has phosphoramidite shown great synthesispromiseReplacement with [57– 60modified]. of These the triazoleoligonucleotide dinucleoside analogs can blocksphosphodiester be obtained [61] throughor linkagethe automatedclick with reaction phosphoramiditetriazole between linkages synthesis azide-has shown and alkyne-bearinggreatwith promise modified [57–60]. nucleosides dinucleoside These [62,63]. triazole blocks Figure analogs [61] or9 shows the can click be three obtained reaction types between throughof promising azide-automated triazole and alkyne-bearing phosphoramidite internucleotide modificationssynthesisnucleosides with [64]. [62modified,63 ]. Figure dinucleoside9 shows three typesblocks of promising[61] or triazolethe click internucleotide reaction modificationsbetween azide- [ 64]. and alkyne-bearing nucleosides [62,63]. Figure 9 shows three types of promising triazole internucleotide modifications [64].

FigureFigure 9. Fragments 9. Fragments of oligonucleotide of oligonucleotide analogs analogs with with different typestypes of of triazole triazole internucleotide internucleotide modificationsmodifications (adapted (adapted from from[64]). [A,64 ]).B, C A, represen B, Ct represent three different three types different of triazole types internucleotide of triazole modifications.Figureinternucleotide 9. Fragments modifications. of oligonucleotide analogs with different types of triazole internucleotide modifications (adapted from [64]). A, B, C represent three different types of triazole internucleotide Varizhukmodifications.Varizhuk et al. et al.synthesized synthesized several several new new oligonucleotide analogs analogs with with triazole triazole internucleotide internucleotide linkageslinkages through through the the click click reaction reaction as as shown shown in in FigureFigure 10 10.. These These analogs analogs bore bore DNA DNA hybridization hybridization affinitiesaffinitiesVarizhuk similar similar et toal. tothose synthesized those of of original original several oligonucleotidesoligonucleotides new oligonucleotide and and increased increased analogs resistance resistance with to nuclease triazole to nuclease cleavage internucleotide cleavage [64]. [64].linkages through the click reaction as shown in Figure 10. These analogs bore DNA hybridization affinities similar to those of original oligonucleotides and increased resistance to nuclease cleavage [64].

Figure 10. Synthesis of the triazole internucleoside linked oligonucleotide analogs with increased resistance to DNAses and polymerases (adapted from [64]). Figure 10. Synthesis of the triazole internucleoside linked oligonucleotide analogs with increased resistance to DNAses and polymerases (adapted from [64]). Int. J. Mol. Sci. 2017, 18, 1683 7 of 20 thio-substitutions in both TT loops exhibited similar antithrombin efficiency to the unmodified aptamer but better resistance to the degradation of DNA nuclease in blood serum [23].

Figure 8. Structures of methylphosphonate and phosphorothioate.

More recently, phosphorodiothioate linkages (PS2) were employed to stabilize phosphate backbone. The substitution of both non-bridging oxygen atoms with sulfur could give rise to a phosphorodithioate linkage, which, similar to natural DNA, is achiral at phosphorus. In addition, it was reported that PS2 substitutions dramatically improved target binding affinity by ~1000-fold (see PS2 walk below) [10].

Replaced by Triazole Replacement of the oligonucleotide phosphodiester linkage with triazole linkages has shown great promise [57–60]. These triazole analogs can be obtained through automated phosphoramidite synthesis with modified dinucleoside blocks [61] or the click reaction between azide- and alkyne-bearing nucleosides [62,63]. Figure 9 shows three types of promising triazole internucleotide modifications [64].

Figure 9. Fragments of oligonucleotide analogs with different types of triazole internucleotide modifications (adapted from [64]). A, B, C represent three different types of triazole internucleotide modifications.

Varizhuk et al. synthesized several new oligonucleotide analogs with triazole internucleotide linkages through the click reaction as shown in Figure 10. These analogs bore DNA hybridization affinitiesInt. J.similar Mol. Sci. 2017to those, 18, 1683 of original oligonucleotides and increased resistance to nuclease8 of cleavage 21 [64].

FigureFigure 10. Synthesis 10. Synthesis of the of thetriazole triazole internucleoside internucleoside linkedlinked oligonucleotide oligonucleotide analogs analogs with increasedwith increased resistanceresistance to DNAses to DNAses and and polymerases polymerases (adapted (adapted fromfrom [ 64[64]).]).

Later in 2013, Varizhuk et al. synthesized a series of triazole-modified DNA aptamers with structure similar to thrombin-inhibiting G-quadruplexes TBA15 (Thrombin-Binding Aptamer) and TBA31, then tested their secondary structure stabilities, binding affinities for thrombin and anticoagulant effects [65]. A modification in the central loop of the aptamer quadruplex resulted in an anticoagulant activity similar to that of TBA15. Although the modification failed to enhance thrombin binding affinity, it protected aptamers from nuclease hydrolysis thus increased their stabilities. The novel aptamers were potent thrombin inhibitors and could be an alternative to the known anticoagulant drugs [59].

3.1.5. The Mirror Image L-DNA

Natural are all in D-form. A chiral transition could result in the mirror image L-DNA (Figure 11) that may display high resistance to the degradation of nucleases and retain the affinity to targets. Based on the sequences of D-form aptamers, the L-enantiomeric oligonucleotide aptamers (also called as Spiegelmers) were then chemically synthesized [66]. Based on the domain approach, Purschke et al. found a 65-mer Spiegelmer that bound to a stable 25-amino acids length domain of bacterial staphylococcal enterotoxin B [64]. The L-DNA Spiegelmer showed comparable binding affinity to the L- domain and slightly reduced affinity to the whole bacterial staphylococcal enterotoxin B . Through an in vitro-selection process, which was started from a random pool of oligonucleotides, a 67-mer Spiegelmer with a dissociation constant (Kd) of 20 nM for gonadotropin-releasing hormone (GnRH) was reported by Wlotzka et al. [67]. This Spiegelmer was an effective antagonist to GnRH in Chinese hamster and castrated rat models. Besides, the PEGylated Spiegelmer showed more pronounced inhibition activity and longer plasma half-life [67]. Towards the same target, other Spiegelmers with high specificity and affinity were identified through the usage of Spiegelmer technology by Leva et al. [68]. Firstly, aptamers that bind to D-GnRH with Kd of 50–100 nM were isolated, and then their enantiomers were synthesized. The resulting Spiegelmers had similar affinities to that of D-aptamers [68]. Many clinical evaluated aptamers such as NOX-A12, NOX-H94 and NOX-E36 are all L-aptamers [69,70]. A number of different strategies and chemical modifications are now available to enhance the stability of aptamers to nuclease (Table1). Among these modifications, 2 0-fluoro or 20-O-methyl-substitutions and 30 end capping with inverted thymidine have been the common strategies in the chemical modifications of nucleic acid aptamers for resisting nuclease degradation. Int. J. Mol. Sci. 2017, 18, 1683 8 of 20

Later in 2013, Varizhuk et al. synthesized a series of triazole-modified DNA aptamers with structure similar to thrombin-inhibiting G-quadruplexes TBA15 (Thrombin-Binding Aptamer) and TBA31, then tested their secondary structure stabilities, binding affinities for thrombin and anticoagulant effects [65]. A modification in the central loop of the aptamer quadruplex resulted in an anticoagulant activity similar to that of TBA15. Although the modification failed to enhance thrombin binding affinity, it protected aptamers from nuclease hydrolysis thus increased their stabilities. The novel aptamers were potent thrombin inhibitors and could be an alternative to the known anticoagulant drugs [59].

3.1.5. The Mirror Image L-DNA

Natural DNAs are all in D-form. A chiral transition could result in the mirror image L-DNA (Figure 11) that may display high resistance to the degradation of nucleases and retain the affinity to targets. Based on the sequences of D-form aptamers, the L-enantiomeric oligonucleotide aptamers (also called as Spiegelmers) were then chemically synthesized [66]. Based on the domain approach, Purschke et al. found a 65-mer Spiegelmer that bound to a stable 25-amino acids length domain of bacterial staphylococcal enterotoxin B [64]. The L-DNA Spiegelmer showed comparable binding affinity to the L-peptide domain and slightly reduced affinity to the whole bacterial staphylococcal enterotoxin B protein. Through an in vitro-selection process, which was started from a random pool of oligonucleotides, a 67-mer Spiegelmer with a dissociation constant (Kd) of 20 nM for gonadotropin-releasing hormone (GnRH) was reported by Wlotzka et al. [67]. This Spiegelmer was an effective antagonist to GnRH in Chinese hamster and castrated rat models. Besides, the PEGylated Spiegelmer showed more pronounced inhibition activity and longer plasma half-life [67]. Towards the same target, other Spiegelmers with high specificity and affinity were identified through the usage of Spiegelmer technology by Leva et al. [68]. Firstly, aptamers that bind to D-GnRH with Kd of 50–100 nM were isolated, and then their enantiomers were synthesized. The resulting Spiegelmers had similar affinitiesInt. J. Mol. Sci.to that2017, of18, D 1683-aptamers [68]. Many clinical evaluated aptamers such as NOX-A12, NOX-H949 of 21 and NOX-E36 are all L-aptamers [69,70].

Figure 11. Structures of L-deoxyoligonucleotide (L-DNA). Mirror image aptamers are composed of Figure 11. Structures of L-deoxyoligonucleotide (L-DNA). Mirror image aptamers are composed of non-natural L-ribose nucleotides. The molecules are initially selected from natural D-ribose aptamer non-natural L-ribose nucleotides. The molecules are initially selected from natural D-ribose aptamer libraries against a non-natural target, for example a D-peptide. Once optimized as a D-aptamer, the libraries against a non-natural target, for example a D-peptide. Once optimized as a D-aptamer, the L mirror imageimageL -aptamer-aptamer (Spiegelmer) (Spiegelmer) is synthesizedis synthesized chemically chemically and intrinsicallyand intrinsically bound bound to the naturalto the naturalL-target, L-target, such as such a naturally as a naturally occurring occurring protein. protein.

A numberTable 1. ofChemical different modifications strategies and of nucleic chemical acid aptamersmodifications for resisting are now nuclease available degradation. to enhance the stability of aptamers to nuclease (Table 1). Among these modifications, 2′-fluoro or 2′-O-methyl-substitutionsModification Sites and 3′ end capping with Strategy inverted thymidine have been Applications the common strategies in the chemical modificaterminaltions of 30–3 nucleic0or 50–50 internucleotideacid aptamers linkage for resisting1, 30-biotin nuclease degradation. ends of nucleic acid chain [12,15,36,47] conjugates; 20-fluoro, 20-O-methyl and 20-amino-substitutions 1, locked sugar ring of nucleoside nucleic acid (LNA), unlocked nucleic acid (UNA) and [25,48,49,52–55] 0 0 0 2 -deoxy-2 -fluoro-D-arabinonucleic acid (2 -F ANA); methylphosphonate or phosphorothioate, phosphodiester linkage [23,24,56–59] replaced by triazole;

mirror image L-enantiomeric oligonucleotide aptamers (Spiegelmers) [66–70] 1 20-fluoro or 20-O-methyl-substitutions and 30 end capping with inverted thymidine have been the common strategies in the chemical modifications of nucleic acid aptamers for resisting nuclease degradation.

3.2. Aptamer Derivatives for Resisting Renal Clearance

3.2.1. 50-End with Cholesterol Even with stabilizing backbone modification, small aptamers are subjected to rapid excretion through renal clearance mainly through glomerular filtration. Formulation with bulky moiety enlarges the size of aptamers, overcoming the renal filtration and extending circulation time, evidently [32,33]. Cholesterol can be derivatized to the 50-end of an aptamer to form a cholesterol-oligonucleotide (cholODN) conjugate. Smidt et al. added cholesterol at the 50-end of a 16-mer oligonucleotide (ODN) through a phosphate spacer (Figure 12), the half-time of the resulting cholODN (9–11 min) in plasma was considerably longer than the unmodified ODN (<1 min) [71]. The resulting cholODN can be further linked with low-density lipoprotein (LDL) to form cholODN-LDL complex that turned out to be stable against degradation by rat serum nucleases. The cholODN had a roughly 10-fold longer plasma half-life than the unmodified ODN [71]. Lee and coworkers modified a 29 nucleotide-long 20-F pyrimidine modified RNA aptamer with cholesterol to form a cholesterol-conjugated aptamer (chol-aptamer) (Figure 12) which can be efficiently absorbed into the cell and inhibits Hepatitis C virus RNA replication [71]. The chol-aptamer had no toxicity in vitro or in vivo. It did not induce any notable alteration in the gene expression profile, including innate immune-related genes. Moreover, administration of the chol-aptamer was well Int. J. Mol. Sci. 2017, 18, 1683 9 of 20

Table 1. Chemical modifications of nucleic acid aptamers for resisting nuclease degradation.

Modification Sites Strategy Applications ends of nucleic acid chain terminal 3′–3′or 5′–5′internucleotide linkage1, 3′-biotin conjugates; [12,15,36,47] 2′-fluoro, 2′-O-methyl and 2′-amino-substitutions 1, locked nucleic sugar ring of nucleoside acid (LNA), unlocked nucleic acid (UNA) and [25,48,49,52–55] 2′-deoxy-2′-fluoro-D-arabinonucleic acid (2′-F ANA); phosphodiester linkage methylphosphonate or phosphorothioate, replaced by triazole; [23,24,56–59] mirror image L-enantiomeric oligonucleotide aptamers (Spiegelmers) [66–70] 1 2′-fluoro or 2′-O-methyl-substitutions and 3′ end capping with inverted thymidine have been the common strategies in the chemical modifications of nucleic acid aptamers for resisting nuclease degradation.

3.2. Aptamer Derivatives for Resisting Renal Clearance

3.2.1. 5′-End with Cholesterol Even with stabilizing backbone modification, small aptamers are subjected to rapid excretion through renal clearance mainly through glomerular filtration. Formulation with bulky moiety enlarges the size of aptamers, overcoming the renal filtration and extending circulation time, evidently [32,33]. Cholesterol can be derivatized to the 5′-end of an aptamer to form a cholesterol-oligonucleotide (cholODN) conjugate. Smidt et al. added cholesterol at the 5′-end of a 16-mer oligonucleotide (ODN) through a phosphate spacer (Figure 12), the half-time of the resulting cholODN (9–11 min) in plasma was considerably longer than the unmodified ODN (<1 min) [71]. The resulting cholODN can be further linked with low-density lipoprotein (LDL) to form cholODN-LDL complex that turned out to be stable against degradation by rat serum nucleases. The cholODN had a roughly 10-fold longer plasma half-life than the unmodified ODN [71]. Lee and coworkers modified a 29 nucleotide-long 2′-F pyrimidine modified RNA aptamer with cholesterol to form a cholesterol-conjugated aptamer (chol-aptamer) (Figure 12) which can be efficiently absorbed into the cell and inhibits Hepatitis C virus RNA replication [71]. The chol-aptamerInt. J. Mol. Sci. had2017 no, 18 ,toxicity 1683 in vitro or in vivo. It did not induce any notable alteration in10 the of 21 gene expression profile, including innate immune-related genes. Moreover, administration of the chol-aptamertolerated inwas mice well without tolerated any abnormalities in mice observed.without Noticeably,any abnormalities cholesterol conjugationobserved. showedNoticeably, cholesterollonger half-lifeconjugation with approximatelyshowed longer nine half-life times lowerwith approximately of clearance rate nine in plasma. times Inlower other of words, clearance rate itin extended plasma. the In durationother words, time that it extended the aptamer the stayed duration in plasma, time thus that enhanced the aptamer the stability stayed when in plasma, the thus aptamerenhanced was the exposed stability to bodywhen [32 the]. aptamer was exposed to body [32].

FigureFigure 12. Structures 12. Structures of ofcholesterol-oligonucleotide cholesterol-oligonucleotide conjugates conjugates (adapted (adapted from from [71]). [71]). Cholesterol Cholesterol can be can be derivatizedderivatized toto the the 50 -end5′-end of an of aptamer an aptamer to form to a cholesterol-oligonucleotide form a cholesterol-oligonucleotide (cholODN) conjugate. (cholODN)

Int.conjugate. J. Mol.The Sci. half-time The2017 , 18half-time of, 1683 the resulting of the cholODN resulting in plasmacholOD wasN in considerably plasma was longer considerably than the control longer ODN. than10 ofthe 20 control ODN. 0 3.2.2. 5 ′-End-End with with Dialkyl Dialkyl Lipids Lipids Willis et al. reported the preparation and functionalfunctional properties of a nuclease-resistantnuclease-resistant vascular endothelial (VEGF) aptamer which was attached to liposome bilayers through a lipid group. The The resulting resulting liposome-anchored liposome-anchored aptamer aptamer maintained maintained the the high high binding binding affinity affinity to to VEGF. VEGF. Moreover, thethe residenceresidence timetime in in plasma plasma was was considerably considerably improved improved when when compared compared with with that that of the of theoriginal original aptamer aptamer [72]. They[72]. used They the used solid the phase solid phosphoramidite phase phosphoramidite method to prepare method a dialkylglycerolto prepare a (DAG)dialkylglycerol modified (DAG) VEGF aptamermodified in VEGF which aptamer two 18-carbon in which saturated two 18-carbon unbranched saturated hydrocarbon unbranched chains werehydrocarbon attached chains via a were tetraethylene attached via glycol a tetraethylene linker. The glycol DAG linker. phosphoramidite The DAG phosphoramidite was synthesized was in 0 synthesizedseven steps andin seven then steps introduced and then to introduced the 5 -end ofto thethe VEGF5′-end aptamerof the VEGF (Figure aptamer 13)[73 (Figure]. Afterwards, 13) [73]. Afterwards,the DAG-modified the DAG-modified VEGF aptamer VEGF was incorporatedaptamer was intoincorporated the bilayers into of the liposomes, bilayers which of liposomes, resulted whichin aptamers resulted with in improvedaptamers inhibitorywith improved activity inhibitory toward VEGF-inducedactivity toward endothelial VEGF-induced cell proliferation endothelial cellin vitro proliferation and increased in vitro vascular and increased permeability vascular in vivo permeability [73]. in vivo [73].

Figure 13. SynthesisSynthesis of of the the dialkylglycerol dialkylglycerol (DAG) (DAG) modi modifiedfied VEGF VEGF aptamer aptamer (adapted (adapted from from [72]). [72]). Liposome-anchored aptamer maintained maintained the the high high bi bindingnding affinity affinity to to VEGF. Moreover, the plasma residence time was considerably improved whenwhen comparedcompared withwith thatthat ofof thethe originaloriginal aptamer.aptamer.

3.2.3. 5′-End PEGylation In 2011, Hoffmann et al. described the PEGylation of amino-modified NOX-E36 oligonucleotide by using N-hydroxysuccinimide (NHS)-ester-activated polyethylene glycol (PEG), which was most widely used, especially for manufacturing large quantities of PEGylated oligonucleotides. Following synthesis and two-step deprotection, the resulting intermediate amino-modified oligonucleotide reacted with NHS-ester-activated PEG to form oligonucleotide-PEG conjugate (Figure 14). Other coupling methods such as activation by p-nitrophenyl carbonate or thiol-maleimide coupling could also be used [74]. The choice of coupling strategies should be made under consideration of the following factors: (1) compatibility with the oligonucleotide; (2) accessibility of the modified oligonucleotide; and (3) reactivity of the activated PEG, which should only react at the functionalization site of the oligonucleotide. MP7 is one of the DNA aptamers that bind specifically to the murine extracellular domain of PD-1 (Programmed death protein 1) and block the PD-1:PD-L1 (Programmed death- 1) interaction. However, the unmodified DNA aptamer exhibited very short in vivo half-time (<1 h) owing to the rapid renal filtration of such small molecule [75]. It has been reported that conjugation of aptamers with high molecular weight PEG could limit the rate of filtration and extended half-life up to 24–48 h [32,75]. Thus, MP7 was modified at its 5′-termini with a 40 kDa PEG (Figure 15). The PEGylated form of MP7 retained the ability to block PD-1 binding to PD-L1, and significantly suppressed the growth of PD-L1 positive colon carcinoma in vivo [76,77] (Table 2). Int. J. Mol. Sci. 2017, 18, 1683 11 of 21

3.2.3. 50-End PEGylation In 2011, Hoffmann et al. described the PEGylation of amino-modified NOX-E36 oligonucleotide by using N-hydroxysuccinimide (NHS)-ester-activated polyethylene glycol (PEG), which was most widely used, especially for manufacturing large quantities of PEGylated oligonucleotides. Following synthesis and two-step deprotection, the resulting intermediate amino-modified oligonucleotide reacted with NHS-ester-activated PEG to form oligonucleotide-PEG conjugate (Figure 14). Other coupling methods such as activation by p-nitrophenyl carbonate or thiol-maleimide coupling could also be used [74]. The choice of coupling strategies should be made under consideration of the following factors: (1) compatibility with the oligonucleotide; (2) accessibility of the modified oligonucleotide; and (3) reactivity of the activated PEG, which should only react at the functionalization site of the oligonucleotide. MP7 is one of the DNA aptamers that bind specifically to the murine extracellular domain of PD-1 (Programmed death protein 1) and block the PD-1:PD-L1 (Programmed death-ligand 1) interaction. However, the unmodified DNA aptamer exhibited very short in vivo half-time (<1 h) owing to the rapid renal filtration of such small molecule [75]. It has been reported that conjugation of aptamers with high molecular weight PEG could limit the rate of filtration and extended half-life up to 24–48 h [32,75]. Thus, MP7 was modified at its 50-termini with a 40 kDa PEG (Figure 15). The PEGylated form of MP7 retained the ability to block PD-1 binding to PD-L1, and significantly suppressed the growth of PD-L1 positiveInt. J. Mol. colonSci. 2017 carcinoma, 18, 1683 in vivo [76,77] (Table2). 11 of 20

NH2

Base protected group O DMTO O MMTHN O P O Base O O O H NC P O OTBDMS O N H Base protected group O OH

O O Base phosphoramidite cycle O O O OTBDMS O OH solid phase support solid phase support O H O O PEG N N O PEG O Base N N PEG O O P O 1) PEG O O O O H 2) deprotection O OH Base pegylated oligonucleotide O O

OH OH

Figure 14. AdditionAddition of of the the aminolinker aminolinker to to 5 5′0-end-end of of the the oligonucleotide and and PEGylation of amino-modifiedamino-modified oligonucleotide with with 40 40 kDa Y-shaped PEG ( n = ~450) (adapted (adapted from from [74]). [74]). Amino-modifiedAmino-modified oligonucleotide could be reacted with NHS-ester-activated PEG to form oligonucleotide-PEG conjugate. Conjugation Conjugation of of aptamers aptamers with with high high molecular weight PEG could limit the rate of filtrationfiltration andand extended half-life up toto 24–4824–48 h.h.

Figure 15. Reaction scheme of aptamer conjugating to a 40 kDa polyethylene glycol (PEG) at the 5′-termini (adapted from [76]).

Table 2. Aptamer derivatives for resisting renal clearance.

Modification Sites Strategy Applications 5′-end with cholesterol; ends of nucleic acid chain 5′-end with dialkyl lipids; [32,33,71–77] 5′-end PEGylation 1 1 Terminal PEGylation has been the common strategy in the chemical modifications of nucleic acid aptamers for resisting renal clearance.

3.3. Aptamer Derivatives for Improving Binding Affinity and Target Selectivity

3.3.1. Modifications on the Bases; SOMAmers Aptamers with improved binding affinities are being generated with modifications on the base. AS1411 aptamer is a 26-mer single strand DNA 5-d(GGTGGTGGTGGTTGTGGTG GTGGTGG)-3′ which binds to the nucleolin protein expressed on the surfaces of cancer cells [78–80]. Recent research has shown that 5-BzdU (5-(N-benzylcarboxyamide)-2-deoxyuridine) modification (Figure 16) of the AS1411 aptamer might selectively increase its targeting affinity to cancer cells while the normal healthy cells have no significant influence [26]. Int. J. Mol. Sci. 2017, 18, 1683 11 of 20

NH2

Base protected group O DMTO O MMTHN O P O Base O O O H NC P O OTBDMS O N H Base protected group O OH

O O Base phosphoramidite cycle O O O OTBDMS O OH solid phase support solid phase support O H O O PEG N N O PEG O Base N N PEG O O P O 1) PEG O O O O H 2) deprotection O OH Base pegylated oligonucleotide O O

OH OH Figure 14. Addition of the aminolinker to 5′-end of the oligonucleotide and PEGylation of amino-modified oligonucleotide with 40 kDa Y-shaped PEG (n = ~450) (adapted from [74]). Amino-modified oligonucleotide could be reacted with NHS-ester-activated PEG to form Int. J. Mol.oligonucleotide-PEG Sci. 2017, 18, 1683 conjugate. Conjugation of aptamers with high molecular weight PEG could12 of 21 limit the rate of filtration and extended half-life up to 24–48 h.

Figure 15. Reaction scheme scheme of of aptamer conjugating to to a 40 kDa polyethylene glycol (PEG) at the 5′0-termini-termini (adapted from [76]). [76]).

Table 2. Aptamer derivatives for resistingresisting renal clearance. Modification Sites Strategy Applications Modification Sites Strategy Applications 5′-end with cholesterol; 0 ends of nucleic acid chain 55′-end-end with with cholesterol; dialkyl lipids; [32,33,71–77] ends of nucleic acid chain 50-end with dialkyl lipids; [32,33,71–77] 1 50-end5′-end PEGylation PEGylation1 1 1 Terminal PEGylation PEGylation has has been been the commonthe common strategy strategy in thechemical in the chemical modifications modifications of nucleic acid of nucleic aptamers acid for aptamersresisting renal for clearance.resisting renal clearance.

3.3.Int. J. Aptamer Mol. Sci. 2017 Derivatives , 18, 1683 for Improving Binding AffinityAffinity and Target SelectivitySelectivity 12 of 20

3.3.1. Modifications Modifications on the Bases; SOMAmers Aptamers withwith improvedimproved binding binding affinities affinities are ar beinge being generated generated with with modifications modifications on the on base. the 0 AS1411base. AS1411 aptamer aptamer is a 26-mer is singlea 26-mer strand single DNA strand 5-d(GGTGGTGGTGGTTGTGGTGGTGGTGG)-3 DNA 5-d(GGTGGTGGTGGTTGTGGTG whichGTGGTGG)-3 binds to′ thewhich nucleolin bindsprotein to the expressednucleolin onprotein the surfaces expressed of cancer on the cells surfaces [78–80]. Recentof cancer research cells [78–80].has shown Recent that5-BzdU research (5-( Nhas-benzylcarboxyamide)-2-deoxyuridine) shown that 5-BzdU (5-(N-benzylcarboxyamide)-2-deoxyuridine) modification (Figure 16) of the AS1411modification aptamer (Figure might 16) selectively of the AS1411 increase aptamer its targeting migh affinityt selectively to cancer increase cells while its targeting the normal affinity healthy to Int.cancercells J. Mol. have cells Sci. no 2017while significant, 18 ,the 1683 normal influence healthy [26]. cells have no significant influence [26]. 12 of 20

Figure 16. Structure of 5-BzdU (5-(N-benzylcarboxyamide)-2′-deoxyuridine).

The benzyl could be replaced by the other functional groups such as naphtyl, triptamino, isobutyl and so on (Figure 17). These additional groups might increase the affinities of aptamers to their targets [81–84] (Table 3).

Table 3. Aptamer derivatives for improving binding affinity and specificity.

Modification Strategy Applications Sites Figure 16. Structure of 5-BzdU (5-(N-benzylcarboxyamide)-20′-deoxyuridine). Figure 16.5-(StructureN-benzylcarboxyamide)-2 of 5-BzdU (5-(N-benzylcarboxyamide)-2′-deoxyuridine modification-deoxyuridine). 1, base of nucleoside [78–84] Slow Off-rate Modified Aptamers (SOMAmers) The benzyl could be replaced by the other functional groups such as naphtyl, triptamino, phosphodiesterThe benzyl could be replaced by the other functional groups such as naphtyl, triptamino, isobutyl isobutyl and so on (Figure 17). Thphosphorodithioateese additional groups (PS2) might substitution increase the affinities of[10,85,86] aptamers to and solinkage on (Figure 17). These additional groups might increase the affinities of aptamers to their their targets [81–84] (Table 3). targets1 The benzyl [81–84 could] (Table be replaced3). by the other functional groups such as naphtyl, triptamino, isobutyl and so on. Table 3. Aptamer derivatives for improving binding affinity and specificity.

Modification Strategy Applications Sites 5-(N-benzylcarboxyamide)-2′-deoxyuridine modification 1, base of nucleoside [78–84] Slow Off-rate Modified Aptamers (SOMAmers) phosphodiester Figure 17.phosphorodithioate Structures of naphtyl, (PS2)triptamino substitution and isobutyl. [10,85,86] linkage Figure 17. Structures of naphtyl, triptamino and isobutyl. 1 TheThe benzyl base could modifications be replaced by have the other also functional made sigroupsgnificant such advancementsas naphtyl, triptamino, to give isobutyl aptamers and so on. protein-like functionality [11,87]. The SOMAmers (Slow Off-rate Modified Aptamers) not only display improved binding affinities and binding kinetics (in particular, slow off-rates) when compared to traditional aptamers, but also the inclusion of these modifications in their libraries significantly increased the selection “hit rate” [88]. The power of this kind of base modifications has been further demonstrated through the discovery of a 32 nucleotide SOMAmer, SL1025, which binds IL-6 with 200 pmol/L binding affinity and exhibits very little Figure 17. Structures of naphtyl, triptamino and isobutyl. nuclease degradation over a 48-hour incubation in human serum [47,89]. The base modifications have also made significant advancements to give aptamers 3.3.2. Crystal Structure Based Modifications protein-like functionality [11,87]. The SOMAmers (Slow Off-rate Modified Aptamers) not only displayNucleic improved acid aptamersbinding affiniti are muches and smaller binding than kinetics antibodies. (in part Inicular, recent slow years, off-rates) there whenare many comparedstudies on tothe traditional crystallization aptamers, and X-ray but also diffraction the inclusion analysis of these of aptamers modifications or the complexin their libraries of aptamers significantlyand [90–93].increased It isthe an selectioneffective method“hit rate” to develop[88]. The modified power aptamers of this withkind higherof base affinity modificationsand selectivity hasaccording been furtheto the rcrystal demonstrated structures. through Autotaxin the (ATX) discovery is a plasma of a 32lysophospholipase nucleotide SOMAmer,D which can SL1025, hydrolyze which lysophosphatidylcholine binds IL-6 with 200 pmol/L (LPC) binding and generate affinity lysophosphatidic and exhibits very acid little (LPA) nuclease[94,95]. DNA degradation aptamer over RB011 a 48-hour is an inhibitor incubation against in human ATX. serum Nureki [47,89]. and coworkers had investigated the crystal structure of ATX in complex with RB011 [96]. The results showed that RB011 inhibited 3.3.2.the activity Crystal of Structure ATX by Basedpreventing Modifications its binding to LPC substrates. The hydrophobic pocket of ATX could be occupied by some inhibitors such as HA155 or 3BoA [97,98], but RB011 did not occlude the Nucleic acid aptamers are much smaller than antibodies. In recent years, there are many hydrophobic pocket. Thus, the researchers introduced some hydrophobic groups such as p-methyl studies on the crystallization and X-ray diffraction analysis of aptamers or the complex of aptamers and p-isopropyl into the backbone phosphate of RB011, resulting in RB012 and RB013, respectively. and enzyme [90–93]. It is an effective method to develop modified aptamers with higher affinity and selectivity according to the crystal structures. Autotaxin (ATX) is a plasma lysophospholipase D which can hydrolyze lysophosphatidylcholine (LPC) and generate lysophosphatidic acid (LPA) [94,95]. DNA aptamer RB011 is an inhibitor against ATX. Nureki and coworkers had investigated the crystal structure of ATX in complex with RB011 [96]. The results showed that RB011 inhibited the activity of ATX by preventing its binding to LPC substrates. The hydrophobic pocket of ATX could be occupied by some inhibitors such as HA155 or 3BoA [97,98], but RB011 did not occlude the hydrophobic pocket. Thus, the researchers introduced some hydrophobic groups such as p-methyl and p-isopropyl into the backbone phosphate of RB011, resulting in RB012 and RB013, respectively. Int. J. Mol. Sci. 2017, 18, 1683 13 of 21

Table 3. Aptamer derivatives for improving binding affinity and specificity.

Modification Sites Strategy Applications 5-(N-benzylcarboxyamide)-20-deoxyuridine base of nucleoside modification 1, Slow Off-rate Modified [78–84] Aptamers (SOMAmers) phosphodiester linkage phosphorodithioate (PS2) substitution [10,85,86] 1 The benzyl could be replaced by the other functional groups such as naphtyl, triptamino, isobutyl and so on.

The base modifications have also made significant advancements to give aptamers protein-like functionality [11,87]. The SOMAmers (Slow Off-rate Modified Aptamers) not only display improved binding affinities and binding kinetics (in particular, slow off-rates) when compared to traditional aptamers, but also the inclusion of these modifications in their libraries significantly increased the selection “hit rate” [88]. The power of this kind of base modifications has been further demonstrated through the discovery of a 32 nucleotide SOMAmer, SL1025, which binds IL-6 with 200 pmol/L binding affinity and exhibits very little nuclease degradation over a 48-hour incubation in human serum [47,89].

3.3.2. Crystal Structure Based Modifications Nucleic acid aptamers are much smaller than antibodies. In recent years, there are many studies on the crystallization and X-ray diffraction analysis of aptamers or the complex of aptamers and enzyme [90–93]. It is an effective method to develop modified aptamers with higher affinity and selectivity according to the crystal structures. Autotaxin (ATX) is a plasma lysophospholipase D which can hydrolyze lysophosphatidylcholine (LPC) and generate lysophosphatidic acid (LPA) [94,95]. DNA aptamer RB011 is an inhibitor against ATX. Nureki and coworkers had investigated the crystal structure of ATX in complex with RB011 [96]. The results showed that RB011 inhibited the activity of ATX by preventing its binding to LPC substrates. The hydrophobic pocket of ATX could be occupied by some inhibitors such as HA155 or 3BoA [97,98], but RB011 did not occlude the hydrophobic pocket. Thus, the researchers introduced some hydrophobic groups such as p-methyl and p-isopropyl into the backbone phosphate of RB011, resulting in RB012 and RB013, respectively. The activities of both RB012 and RB013 (IC50 values for LPC were 1.8 and 0.85 nM, respectively) were more potent than that of RB011 (4.4 nM) [96]. These results suggested that modifications aimed to occlude the hydrophobic pocket could significantly increase inhibitory activity. This is a successful modification based on the crystal structural information.

3.3.3. NMR Spectroscopy Guided Aptamer Optimization Nucleic acid aptamers are widely used for biotechnological or biomedical purpose. High resolution structure information of aptamer–ligand complexes could help reveal the fundamental aspects of nucleic acid folding and nucleic acid-small molecule interactions. Structure information of aptamers and aptamer–ligand complexes constitute the starting point for rational function directed chemical modifications. Duchardt-Ferner E et al. reported the NMR resonance assignment of an RNA aptamer binding to the fluorescent ligand tetramethylrhodamine (TMR) in complex with the ligand 5-carboxy-tetramethylrhodamine (5-TAMRA) as a starting point for a high-resolution structure determination using NMR spectroscopy in solution [99]. This and other reports indicated that NMR guided aptamer optimization could be an optional strategy for aptamer improving binding affinity [100–102].

3.3.4. PS2 Walk The binding affinity and specificity of unmodified nucleic acid aptamers are sometimes insufficient for successful implementation as therapeutic agents, compared with monoclonal . Post-SELEX Int. J. Mol. Sci. 2017, 18, 1683 13 of 20

The activities of both RB012 and RB013 (IC50 values for LPC were 1.8 and 0.85 nM, respectively) were more potent than that of RB011 (4.4 nM) [96]. These results suggested that modifications aimed to occlude the hydrophobic pocket could significantly increase inhibitory activity. This is a successful modification based on the crystal structural information.

3.3.3. NMR Spectroscopy Guided Aptamer Optimization Nucleic acid aptamers are widely used for biotechnological or biomedical purpose. High resolution structure information of aptamer–ligand complexes could help reveal the fundamental aspects of nucleic acid folding and nucleic acid-small molecule interactions. Structure information of aptamers and aptamer–ligand complexes constitute the starting point for rational function directed chemical modifications. Duchardt-Ferner E et al. reported the NMR resonance assignment of an RNA aptamer binding to the fluorescent ligand tetramethylrhodamine (TMR) in complex with the ligand 5-carboxy-tetramethylrhodamine (5-TAMRA) as a starting point for a high-resolution structure determination using NMR spectroscopy in solution [99]. This and other reports indicated that NMR guided aptamer optimization could be an optional strategy for aptamer improving binding affinity [100–102].

3.3.4. PS2 Walk Int. J. Mol. Sci. 2017, 18, 1683 14 of 21 The binding affinity and specificity of unmodified nucleic acid aptamers are sometimes insufficient for successful implementation as therapeutic agents, compared with monoclonal optimization of one Bn-dU and one Nap-dU SOMAmer led to improvements in IL-6 binding (10-fold) antibody. Post-SELEX optimization of one Bn-dU and one Nap-dU SOMAmer led to improvements and inhibition activity (greater than 20-fold), resulting in lead SOMAmers with sub-nanomolar affinity in IL-6 binding (10-fold) and inhibition activity (greater than 20-fold), resulting in lead SOMAmers (Kd = 0.2 nM) and potency (IC50 = 0.2 nM) [92]. The PS2 (phosphorodithioate) walk strategy is another with sub-nanomolar affinity (Kd = 0.2 nM) and potency (IC50 = 0.2 nM) [92]. The PS2 option [85,86] (Figure 18). It was reported that the application of the PS2 substitution on a single (phosphorodithioate) walk strategy is another option [85,86] (Figure 18). It was reported that the nucleotide of nucleic acid aptamers could significantly improve target binding affinity by ~1000-fold application of the PS2 substitution on a single nucleotide of nucleic acid aptamers could (from nanomolar to picomolar). An X-ray co-crystal structure of the α-thrombin-PS2-aptamer complex significantly improve target binding affinity by ~1000-fold (from nanomolar to picomolar). An revealed a localized induced-fit folding of the PS2-containing aptamer which leads to increased target X-ray co-crystal structure of the α-thrombin-PS2-aptamer complex revealed a localized induced-fit interaction [10]. folding of the PS2-containing aptamer which leads to increased target interaction [10].

Figure 18. SchematicSchematic of of the the PS2-walk PS2-walk library library of sequence variants each containing a single PS2 modification.modification. ModificationModification hot hot spots spots along along the phosphatethe phosphate backbone backbone of the aptamerof the aptamer could be identifiedcould be byidentified phosphorodithioate by phosphorodithioate (PS2) substitution (PS2) substitution on a single on nucleotide a single nucleotide of nucleic acidof nucleic sequences. acid sequences.

It is worth noting that the effect of PSO substitution (see Section 3.1.4. above) cannot be It is worth noting that the effect of PSO substitution (see Section 3.1.4. above) cannot be predicted predicted since the PSO backbone modification is chiral and the chemical synthesis of PSO since the PSO backbone modification is chiral and the chemical synthesis of PSO using phosphoramidite methodology typically results in a mixture of diastereoisomers with a fairly limited influence on the affinity improvement. The promising PS2 derivatives are achiral, representing a class of closely related mimics of natural nucleic acids.

4. Conclusions In this review, we introduced the general solid phase synthesis method of nucleic acid aptamers. In addition, a number of chemical modifications of both DNA and RNA aptamers are summarized here. Among all the modifications shown in the Figure 19, 50-end PEGylation (for resisting renal clearance) and 30-end capping strategy (for resisting nuclease degradation) with inverted thymidine are the most commonly used strategy in recent studies. These two methods have been used in the aptamers for disease therapy in ongoing or completed clinical trials [15,47]. Int. J. Mol. Sci. 2017, 18, 1683 14 of 20 using phosphoramidite methodology typically results in a mixture of diastereoisomers with a fairly limited influence on the affinity improvement. The promising PS2 derivatives are achiral, representing a class of closely related mimics of natural nucleic acids.

4. Conclusions In this review, we introduced the general solid phase synthesis method of nucleic acid aptamers. In addition, a number of chemical modifications of both DNA and RNA aptamers are summarized here. Among all the modifications shown in the Figure 19, 5′-end PEGylation (for resisting renal clearance) and 3′-end capping strategy (for resisting nuclease degradation) with Int.inverted J. Mol. Sci.thymidine2017, 18, 1683 are the most commonly used strategy in recent studies. These two methods15 of 21 have been used in the aptamers for disease therapy in ongoing or completed clinical trials [15,47].

Figure 19. Summary of the chemical modificationsmodifications of nucleic acid aptamers.

The nucleobase and phosphodiester linkage modifications (for improving target binding The nucleobase and phosphodiester linkage modifications (for improving target binding affinity) can also optimize the properties of aptamers. Excitingly, the established technologies affinity) can also optimize the properties of aptamers. Excitingly, the established technologies provide an opportunity to generate nucleic acid aptamers of substantially improved affinity with a provide an opportunity to generate nucleic acid aptamers of substantially improved affinity with SOMAmer strategy or a single PS2-moiety substitution and without negatively affecting specificity. a SOMAmer strategy or a single PS2-moiety substitution and without negatively affecting specificity. These technologies also provide crucial insights that could significantly accelerate the development These technologies also provide crucial insights that could significantly accelerate the development of of nucleic acid aptamer-based therapeutics for clinical applications. With the development of nucleic acid aptamer-based therapeutics for clinical applications. With the development of post-SELEX post-SELEX modifications of nucleic acid aptamers, the inherent physicochemical characteristics modifications of nucleic acid aptamers, the inherent physicochemical characteristics (metabolic (metabolic instability, insufficient binding affinity and rapid renal filtration) of nucleic acid instability, insufficient binding affinity and rapid renal filtration) of nucleic acid aptamers have aptamers have been improved constantly, which provide a strong impetus of developing nucleic been improved constantly, which provide a strong impetus of developing nucleic acid aptamers acid aptamers for therapeutic purposes (Table 4). for therapeutic purposes (Table4). Table 4. Chemical modifications of nucleic acid aptamers for different purposes. Table 4. Chemical modifications of nucleic acid aptamers for different purposes. Improving Binding Resistance Nuclease Strategy ImprovingAffinity Binding and to Renal Resistance Resistance to Renal Strategy Nuclease Resistance Affinity and Target Target Selectivity ClearanceClearance 3′-3′inversion/ 3′-T capping [12,20,21] Selectivity 0 0 0 3 -3 inversion/ 3 5-T′-5 capping′inversion [12,20,21] [12] 0 0 5 -5 inversion3′-biotin conjugates [12] [20,36] 30-biotin conjugates [20,36] 20-fluoro,2′-fluoro, 20-O-methyl 2′-O and-methyl 20-amino-substitutions1 and 2′-amino-substitutions1 [39 ,48,49][39,48,49] locked nucleiclocked acid nucleic (LNA) acid (LNA) [52,53] [52,53] unlockedunlocked nucleic acid nucleic (UNA) acid (UNA) [54,55] [54,55] 0 0 0 2 -deoxy-22′-deoxy-2-fluoro-D′-fluoro--arabinonucleicD-arabinonucleic acid (2 -F ANA) acid (2′-F ANA) [25] [25] methylphosphonate [56] phosphorothioatemethylphosphonate [23,24] [56] replaced byphosphorothioate triazole [57–60] [23,24] L-enantiomeric oligonucleotidereplaced aptamers by triazole (Spiegelmers) [66–70] [57–60] 0 L-enantiomeric5 -end with oligonucleotide cholesterol aptamers (Spiegelmers) [66–70] [32,33,71 ] 50-end with dialkyl lipids [72,73] 5′-end with cholesterol [32,33,71] 50-end PEGylation [32,74–77] 5-(N-benzylcarboxyamide)-2-deoxyuridine modification1, Slow Off-rate Modified Aptamers [78–84] (SOMAmers) phosphorodithioate (PS2) substitution [10,85,86]

Acknowledgments: We thank the other academic staff members in Aiping Lu and Ge Zhang’s group at Hong Kong Baptist University (Hong Kong, China). We also thank Hong Kong Baptist University (Hong Kong, China) and the State Key Laboratory of Bioorganic and Natural Products Chemistry (Shanghai, China) for providing critical comments and technical support. This study was supported by the Hong Kong General Research Fund (HKBU12102914 to Ge Zhang), the Faculty Research Grant of Hong Kong Baptist University (FRG2/12-13/027 to Ge Zhang) and Open Project of Shanghai Institute of Organic Chemistry (SKLBNPC17344 to Feng Jiang). Int. J. Mol. Sci. 2017, 18, 1683 16 of 21

Author Contributions: Shuaijian Ni, Houzong Yao, Lili Wang and Jun Lu wrote the manuscript. Feng Jiang, Aiping Lu and Ge Zhang revised and approved the manuscript. Conflicts of Interest: The authors declare no conflict of interest.

References

1. Tuerk, C.; Gold, L. Systematic evolution of ligands by exponential enrichment: RNA ligands to bacteriophage T4 DNA polymerase. Science 1990, 249, 505–510. [CrossRef][PubMed] 2. Ellington, A.D.; Szostak, J.W. In vitro selection of RNA molecules that bind specific ligands. Nature 1990, 346, 818. [CrossRef][PubMed] 3. Zhou, J.; Rossi, J. Aptamers as targeted therapeutics: Current potential and challenges. Nat. Rev. Drug Discov. 2017, 16, 181–202. [CrossRef][PubMed] 4. Gelinas, A.D.; Davies, D.R.; Janjic, N. Embracing proteins: Structural themes in aptamer-protein complexes. Curr. Opin. Struct. Biol. 2016, 36, 122–132. [CrossRef][PubMed] 5. Ashrafuzzaman, M. Aptamers as both drugs and drug-carriers. BioMed Res. Int. 2014, 2014.[CrossRef] [PubMed] 6. Dass, C.R.; Saravolac, E.G.; Li, Y.; Sun, L.Q. Cellular uptake, distribution, and stability of 10–23 . Antisense Nucleic Acid Drug Dev. 2002, 12, 289–299. [CrossRef][PubMed] 7. Morrissey, D.V.; Blanchard, K.; Shaw, L.; Jensen, K.; Lockridge, J.A.; Dickinson, B.; McSwiggen, J.A.; Vargeese, C.; Bowman, K.; Shaffer, C.S.; et al. Activity of stabilized short interfering RNA in a mouse model of hepatitis B virus replication. Hepatology 2005, 41, 1349–1356. [CrossRef][PubMed] 8. Griffin, L.C.; Tidmarsh, G.F.; Bock, L.C.; Toole, J.J.; Leung, L.L. In vivo anticoagulant properties of a novel nucleotide-based thrombin inhibitor and demonstration of regional anticoagulation in extracorporeal circuits. Blood 1993, 81, 3271–3276. [PubMed] 9. Pagratis, N.C.; Bell, C.; Chang, Y.F.; Jennings, S.; Fitzwater, T.; Jellinek, D.; Dang, C. Potent 20-amino-, and 20-fluoro-20-deoxyribonucleotide RNA inhibitors of keratinocyte growth factor. Nat. Biotechnol. 1997, 15, 68–73. [CrossRef][PubMed] 10. Abeydeera, N.D.; Egli, M.; Cox, N.; Mercier, K.; Conde, J.N.; Pallan, P.S.; Mizurini, D.M.; Sierant, M.; Hibti, F.E.; Hassell, T.; et al. Evoking picomolar binding in RNA by a single phosphorodithioate linkage. Nucleic Acids Res. 2016, 44, 8052–8064. [CrossRef][PubMed] 11. Gold, L.; Ayers, D.; Bertino, J.; Bock, C.; Bock, A.; Brody, E.N.; Carter, J.; Dalby, A.B.; Eaton, B.E.; Fitzwater, T.; et al. Aptamer-based multiplexed proteomic technology for discovery. PLoS ONE 2010, 5, e15004. [CrossRef][PubMed] 12. Ortigao, J.R.; Rosch, H.; Montenarh, M.; Frohlich, A.; Seliger, H. Oligonucleotide analogs with terminal 30, 30-and 50, 50-internucleotidic linkages as antisense inhibitors of viral replication. Antisense Res. Dev. 1991, 1, 380. [CrossRef] 13. 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. [CrossRef][PubMed] 14. Doggrell, S.A. Pegaptanib: The first antiangiogenic agent approved for neovascular macular degeneration. Expert Opin. Pharmacother. 2005, 6, 1421–1423. [CrossRef][PubMed] 15. Fine, S.L.; Martin, D.F.; Kirkpatrick, P. Pegaptanib sodium. Nat. Rev. Drug Discov. 2005, 4, 187–188. [CrossRef] [PubMed] 16. Kiire, C.A.; Morjaria, R.; Rudenko, A.; Fantato, A.; Smith, L.; Smith, A.; Chong, V. Intravitreal pegaptanib for the treatment of ischemic diabetic macular edema. Clin. Ophthalmol. 2015, 9, 2305–2311. [CrossRef] [PubMed] 17. Vinores, S.A. Technology evaluation: Pegaptanib, Eyetech/Pfizer. Curr. Opin. Mol. Ther. 2003, 5, 673–679. [PubMed] 18. Caruthers, M.H.; Barone, A.D.; Beaucage, S.L.; Dodds, D.R.; Fisher, E.F.; McBride, L.J.; Matteucci, M.; Stabinsky, Z.; Tang, J.Y. Chemical synthesis of deoxyoligonucleotides by the phosphoramidite method. Methods Enzymol. 1987, 154, 287–313. [PubMed] 19. Sproat, B.S. RNA synthesis using 20-O-(tert-butyldimethylsilyl) protection. Oligonucleotide Synth. 2005, 17–31. 20. Shum, K.T.; Tanner, J.A. Differential inhibitory activities and stabilisation of DNA aptamers against the SARS coronavirus helicase. Chembiochem 2008, 9, 3037–3045. [CrossRef][PubMed] Int. J. Mol. Sci. 2017, 18, 1683 17 of 21

21. Shaw, J.P.; Kent, K.; Bird, J.; Fishback, J.; Froehler, B. Modified deoxyoligonucleotides stable to exonuclease degradation in serum. Nucleic Acids Res. 1991, 19, 747–750. [CrossRef][PubMed] 22. 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. [CrossRef][PubMed] 23. Zaitseva, M.; Kaluzhny, D.; Shchyolkina, A.; Borisova, O.; Smirnov, I.; Pozmogova, G. Conformation and thermostability of oligonucleotide d(GGTTGGTGTGGTTGG) containing thiophosphoryl internucleotide bonds at different positions. Biophys. Chem. 2010, 146, 1–6. [CrossRef][PubMed] 24. Pozmogova, G.; Zaitseva, M.; Smirnov, I.; Shvachko, A.; Murina, M.; Sergeenko, V. Anticoagulant effects of thioanalogs of thrombin-binding DNA-aptamer and their stability in the plasma. Bull. Exp. Biol. Med. 2010, 150, 180–184. [CrossRef][PubMed] 0 0 25. Peng, C.G.; Damha, M.J. G-quadruplex induced stabilization by 2 -deoxy-2 -fluoro-D-arabinonucleic acids (20F-ANA). Nucleic Acids Res. 2007, 35, 4977–4988. [CrossRef][PubMed] 26. Lee, K.Y.; Kang, H.; Ryu, S.H.; Lee, D.S.; Lee, J.H.; Kim, S. Bioimaging of nucleolin aptamer-containing 5-(N-benzylcarboxyamide)-20-deoxyuridine more capable of specific binding to targets in cancer cells. J. Biomed. Biotechnol. 2010, 2010, 168306. [CrossRef][PubMed] 27. Pallan, P.S.; Yang, X.; Sierant, M.; Abeydeera, N.D.; Hassell, T.; Martinez, C.; Janicka, M.; Nawrot, B.; Egli, M. Crystal structure, stability and Ago2 affinity of phosphorodithioate-modified . Rsc. Adv. 2014, 4, 64901–64904. [CrossRef] 28. Ashley, S.L.; Xia, M.; Murray, S.; O’Dwyer, D.N.; Grant, E.; White, E.S.; Flaherty, K.R.; Martinez, F.J.; Moore, B.B. Six-SOMAmer Index relating to immune, protease and angiogenic functions predicts progression in IPF. PLoS ONE 2016, 11, e0159878. [CrossRef][PubMed] 29. 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. [CrossRef][PubMed] 30. 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. [CrossRef][PubMed] 31. Park, N.J.; Wang, X.; Diaz, A.; Goos-Root, D.M.; Bock, C.; Vaught, J.D.; Sun, W.; Strom, C.M. Measurement of cetuximab and panitumumab-unbound serum EGFR extracellular domain using an assay based on slow off-rate modified aptamer (SOMAmer) reagents. PLoS ONE 2013, 8, e71703. [CrossRef][PubMed] 32. Healy, J.M.; Lewis, S.D.; Kurz, M.; Boomer, R.M.; Thompson, K.M.; Wilson, C.; McCauley, T.G. Pharmacokinetics and biodistribution of novel aptamer compositions. Pharm. Res. 2004, 21, 2234–2246. [CrossRef][PubMed] 33. Watson, S.R.; Chang, Y.F.; O’Connell, D.; Weigand, L.; Ringquist, S.; Parma, D.H. Anti-L-selectin aptamers: Binding characteristics, pharmacokinetic parameters, and activity against an intravascular target in vivo. Antisense Nucleic Acid Drug Dev. 2000, 10, 63–75. [CrossRef][PubMed] 34. 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. [CrossRef][PubMed] 35. Van Eijk, L.; Swinkels, D.; John, A.; Schwoebel, F.; Fliegert, F.; Summo, L.; Vauleon, S.; Laarakkers, J.; Riecke, K.; Pickkers, P. Randomized double-blind placebo-controlled PK/PD study on the effects of a single intravenous dose of the anti-hepcidin Spiegelmer NOX-H94 on serum iron during experimental human endotoxemia. Crit. Care 2013, 17, P352. 36. 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. [CrossRef] 37. Dellinger, D.J.; Betley, J.R.; Wyrzykiewicz, T.K.; Caruthers, M.H. Synthesis of DNA using a new two-step cycle. Oligonucleotide Synth. 2005, 288, 1–16. 38. Beaucage, S.L.; Caruthers, M.H. Synthetic strategies and parameters involved in the synthesis of oligodeoxyribonucleotides according to the phosphoramidite method. Curr. Protoc. Nucleic Acid Chem. 2001. [CrossRef] 39. Maier, K.E.; Levy, M. From selection hits to clinical leads: Progress in aptamer discovery. Mol. Ther. Methods Clin. Dev. 2016, 5, 16014. [CrossRef][PubMed] Int. J. Mol. Sci. 2017, 18, 1683 18 of 21

40. Usman, N.; Ogilvie, K.; Jiang, M.; Cedergren, R. The automated chemical synthesis of long oligoribuncleotides using 20-O-silylated ribonucleoside 30-O-phosphoramidites on a controlled-pore glass support: Synthesis of a 43-nucleotide sequence similar to the 30-half molecule of an formylmethionine tRNA. J. Am. Chem. Soc. 1987, 109, 7845–7854. 41. Shiba, Y.; Masuda, H.; Watanabe, N.; Ego, T.; Takagaki, K.; Ishiyama, K.; Ohgi, T.; Yano, J. Chemical synthesis of a very long oligoribonucleotide with 2-cyanoethoxymethyl (CEM) as the 20-O-protecting group: Structural identification and biological activity of a synthetic 110mer precursor-microRNA candidate. Nucleic Acids Res. 2007, 35, 3287–3296. [CrossRef][PubMed] 42. Ohgi, T.; Masutomi, Y.; Ishiyama, K.; Kitagawa, H.; Shiba, Y.; Yano, J. A new RNA synthetic method with a 20-O-(2-cyanoethoxymethyl) protecting group. Org. Lett. 2005, 7, 3477–3480. [CrossRef][PubMed] 43. Usman, N.; Pon, R.T.; Ogilvie, K.K. Preparation of ribonucleoside 30-O-phosphoramidites and their application to the automated solid phase synthesis of oligonucleotides. Tetrahedron Lett. 1985, 26, 4567–4570. [CrossRef] 44. Sinha, N.; Davis, P.; Usman, N.; Perez, J.; Hodge, R.; Kremsky, J.; Casale, R. Labile exocyclic amine protection of nucleosides in DNA, RNA and oligonucleotide analog synthesis facililating N-deacylation, minimizing depurination and chain degradation. Biochimie 1993, 75, 13–23. [CrossRef] 45. Welz, R.; Müller, S. 5-(Benzylmercapto)-1H-tetrazole as activator for 20-O-TBDMS phosphoramidite building blocks in RNA synthesis. Tetrahedron Lett. 2002, 43, 795–797. [CrossRef] 46. Westman, E.; Stromberg, R. Removal of t-butyldimethylsilyl protection in RNA-synthesis. Triethylamine trihydrofluoride (TEA, 3HF) is a more reliable alternative to tetrabutylammonium fluoride (TBAF). Nucleic Acids Res. 1994, 22, 2430–2431. [CrossRef][PubMed] 47. Gupta, S.; Hirota, M.; Waugh, S.M.; Murakami, I.; Suzuki, T.; Muraguchi, M.; Shibamori, M.; Ishikawa, Y.; Jarvis, T.C.; Carter, J.D.; et al. Chemically modified DNA aptamers bind interleukin-6 with high affinity and inhibit signaling by blocking its interaction with interleukin-6 receptor. J. Biol. Chem. 2014, 289, 8706–8719. [CrossRef][PubMed] 48. Padilla, R.; Sousa, R. Efficient synthesis of nucleic acids heavily modified with non-canonical ribose 20-groups using a mutantT7 RNA polymerase (RNAP). Nucleic Acids Res. 1999, 27, 1561–1563. [CrossRef][PubMed] 49. Ruckman, J.; Green, L.S.; Beeson, J.; Waugh, S.; Gillette, W.L.; Henninger, D.D.; Claesson-Welsh, L.; Janjic, N. 20-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. [CrossRef][PubMed] 50. Obika, S.; Nanbu, D.; Hari, Y.; Morio, K.-I.; In, Y.; Ishida, T.; Imanishi, T. Synthesis of 20-O,40-C-methyleneuridine and -cytidine. Novel bicyclic nucleosides having a fixed C3, -endo sugar puckering. Tetrahedron Lett. 1997, 38, 8735–8738. [CrossRef] 51. Koshkin, A.A.; Singh, S.K.; Nielsen, P.; Rajwanshi, V.K.; Kumar, R.; Meldgaard, M.; Olsen, C.E.; Wengel, J. LNA (Locked Nucleic Acids): Synthesis of the adenine, cytosine, guanine, 5-methylcytosine, thymine and uracil bicyclonucleoside monomers, oligomerisation, and unprecedented nucleic acid recognition. Tetrahedron 1998, 54, 3607–3630. [CrossRef] 52. Darfeuille, F.; Hansen, J.B.; Orum, H.; Di Primo, C.; Toulme, J.J. LNA/DNA chimeric oligomers mimic RNA aptamers targeted to the TAR RNA element of HIV-1. Nucleic Acids Res. 2004, 32, 3101–3107. [CrossRef] [PubMed] 53. Shi, H.; He, X.; Cui, W.; Wang, K.; Deng, K.; Li, D.; Xu, F. Locked nucleic acid/DNA chimeric aptamer probe for tumor diagnosis with improved serum stability and extended imaging window in vivo. Anal. Chim. Acta 2014, 812, 138–144. [CrossRef][PubMed] 54. Campbell, M.A.; Wengel, J. Locked vs. unlocked nucleic acids (LNA vs. UNA): Contrasting structures work towards common therapeutic goals. Chem. Soc. Rev. 2011, 40, 5680–5689. [CrossRef][PubMed] 55. 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. [CrossRef][PubMed] 56. 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. [CrossRef][PubMed] 57. El-Sagheer, A.H.; Brown, T. Click chemistry with DNA. Chem. Soc. Rev. 2010, 39, 1388–1405. [CrossRef] [PubMed] Int. J. Mol. Sci. 2017, 18, 1683 19 of 21

58. Mutisya, D.; Selvam, C.; Kennedy, S.D.; Rozners, E. Synthesis and properties of triazole-linked RNA. Bioorg. Med. Chem. Lett. 2011, 21, 3420–3422. [CrossRef][PubMed] 59. Sau, S.P.; Hrdlicka, P.J. C20-pyrene-functionalized triazole-linked DNA: Universal DNA/RNA hybridization probes. J. Org. Chem. 2012, 77, 5–16. [CrossRef][PubMed] 60. El-Sagheer, A.H.; Brown, T. Click nucleic acid ligation: Applications in biology and nanotechnology. Acc. Chem. Res. 2012, 45, 1258–1267. [CrossRef][PubMed] 61. Chandrasekhar, S.; Srihari, P.; Nagesh, C.; Kiranmai, N.; Nagesh, N.; Idris, M.M. Synthesis of readily accessible triazole-linked dimer deoxynucleoside phosphoramidite for solid-phase oligonucleotide synthesis. Synthesis 2010, 2010, 3710–3714. [CrossRef] 62. Nuzzi, A.; Massi, A.; Dondoni, A. Model studies toward the synthesis of thymidine oligonucleotides with triazole internucleosidic linkages via iterative Cu(I)-promoted azide–alkyne ligation chemistry. Mol. Inform. 2007, 26, 1191–1199. 63. Lucas, R.; Zerrouki, R.; Granet, R.; Krausz, P.; Champavier, Y. A rapid efficient microwave-assisted synthesis of a 30,50-pentathymidine by copper (I)-catalyzed [3 + 2] cycloaddition. Tetrahedron 2008, 64, 5467–5471. [CrossRef] 64. Varizhuk, A.M.; Kaluzhny, D.N.; Novikov, R.A.; Chizhov, A.O.; Smirnov, I.P.; Chuvilin, A.N.; Tatarinova, O.N.; Fisunov, G.Y.; Pozmogova, G.E.; Florentiev, V.L. Synthesis of triazole-linked oligonucleotides with high affinity to DNA complements and an analysis of their compatibility with biosystems. J. Org. Chem. 2013, 78, 5964–5969. [CrossRef][PubMed] 65. Varizhuk, A.M.; Tsvetkov, V.B.; Tatarinova, O.N.; Kaluzhny, D.N.; Florentiev, V.L.; Timofeev, E.N.; Shchyolkina, A.K.; Borisova, O.F.; Smirnov, I.P.; Grokhovsky, S.L.; et al. Synthesis, characterization and in vitro activity of thrombin-binding DNA aptamers with triazole internucleotide linkages. Eur. J. Med. Chem. 2013, 67, 90–97. [CrossRef][PubMed] 66. 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. [CrossRef][PubMed] 67. Wlotzka, B.; Leva, S.; Eschgfaller, B.; Burmeister, J.; Kleinjung, F.; Kaduk, C.; Muhn, P.; Hess-Stumpp, H.; Klussmann, S. In vivo properties of an anti-GnRH Spiegelmer: An example of an oligonucleotide-based therapeutic substance class. Proc. Natl. Acad. Sci. USA 2002, 99, 8898–8902. [CrossRef][PubMed] 68. Leva, S.; Lichte, A.; Burmeister, J.; Muhn, P.; Jahnke, B.; Fesser, D.; Erfurth, J.; Burgstaller, P.; Klussmann, S. GnRH binding RNA and DNA Spiegelmers: A novel approach toward GnRH antagonism. Chem. Biol. 2002, 9, 351–359. [CrossRef] 69. Yu, Y.; Liang, C.; Lv, Q.; Li, D.; Xu, X.; Liu, B.; Lu, A.; Zhang, G. Molecular selection, modification and development of therapeutic oligonucleotide aptamers. Int. J. Mol. Sci. 2016, 17, 358. [CrossRef][PubMed] 70. Purschke, W.G.; Radtke, F.; Kleinjung, F.; Klussmann, S. A DNA Spiegelmer to staphylococcal enterotoxin B. Nucleic Acids Res. 2003, 31, 3027–3032. [CrossRef][PubMed] 71. Lee, C.H.; Lee, S.H.; Kim, J.H.; Noh, Y.H.; Noh, G.J.; Lee, S.W. Pharmacokinetics of a cholesterol-conjugated aptamer against the Hepatitis C Virus (HCV) NS5B Protein. Mol. Ther. Nucleic Acids 2015, 4, e254. [CrossRef] [PubMed] 72. Willis, M.C.; Collins, B.D.; Zhang, T.; Green, L.S.; Sebesta, D.P.; Bell, C.; Kellogg, E.; Gill, S.C.; Magallanez, A.; Knauer, S.; et al. Liposome-anchored vascular endothelial growth factor aptamers. Bioconjug. Chem. 1998, 9, 573–582. [CrossRef][PubMed] 73. Green, L.S.; Jellinek, D.; Bell, C.; Beebe, L.A.; Feistner, B.D.; Gill, S.C.; Jucker, F.M.; Janjic, N. Nuclease-resistant nucleic acid ligands to vascular permeability factor/vascular endothelial growth factor. Chem. Biol. 1995, 2, 683–695. [CrossRef] 74. Hoffmann, S.; Hoos, J.; Klussmann, S.; Vonhoff, S. RNA aptamers and spiegelmers: Synthesis, purification, and post-synthetic PEG conjugation. Curr. Protoc. Nucleic Acid Chem. 2011, 46, 1–30. 75. Da Pieve, C.; Blackshaw, E.; Missailidis, S.; Perkins, A.C. PEGylation and biodistribution of an anti-MUC1 aptamer in MCF-7 tumor-bearing mice. Bioconjug. Chem. 2012, 23, 1377–1381. [CrossRef][PubMed] 76. Prodeus, A.; Abdul-Wahid, A.; Fischer, N.W.; Huang, E.H.; Cydzik, M.; Gariépy, 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. Ther. Nucleic Acids 2015, 4, e237. [CrossRef][PubMed] Int. J. Mol. Sci. 2017, 18, 1683 20 of 21

77. Tan, L.; Neoh, K.G.; Kang, E.T.; Choe, W.S.; Su, X. PEGylated anti-MUC1 aptamer-doxorubicin complex for targeted drug delivery to MCF7 breast cancer cells. Macromol. Biosci. 2011, 11, 1331–1335. [CrossRef] [PubMed] 78. Bates, P.J.; Laber, D.A.; Miller, D.M.; Thomas, S.D.; Trent, J.O. Discovery and development of the G-rich oligonucleotide AS1411 as a novel treatment for cancer. Exp. Mol. Pathol. 2009, 86, 151–164. [CrossRef] [PubMed] 79. Reyes-Reyes, E.M.; Teng, Y.; Bates, P.J. A new paradigm for aptamer therapeutic AS1411 action: Uptake by macropinocytosis and its stimulation by a nucleolin-dependent mechanism. Cancer Res. 2010, 70, 8617–8629. [CrossRef][PubMed] 80. Trinh, T.L.; Zhu, G.; Xiao, X.; Puszyk, W.; Sefah, K.; Wu, Q.; Tan, W.; Liu, C. A synthetic aptamer-drug adduct for targeted liver cancer therapy. PLoS ONE 2015, 10, e0136673. [CrossRef][PubMed] 81. Kimoto, M.; Yamashige, R.; Matsunaga, K.I.; Yokoyama, S.; Hirao, I. Generation of high-affinity DNA aptamers using an expanded genetic alphabet. Nat. Biotechnol. 2013, 31, 453–457. [CrossRef][PubMed] 82. Sefah, K.; Yang, Z.; Bradley, K.M.; Hoshika, S.; Jimenez, E.; Zhang, L.; Zhu, G.; Shanker, S.; Yu, F.; Turek, D.; et al. In vitro selection with artificial expanded genetic information systems. Proc. Natl. Acad. Sci. USA 2014, 111, 1449–1454. [CrossRef][PubMed] 83. Yang, Z.; Durante, M.; Glushakova, L.G.; Sharma, N.; Leal, N.A.; Bradley, K.M.; Chen, F.; Benner, S.A. Conversion strategy using an expanded genetic alphabet to assay nucleic acids. Anal. Chem. 2013, 85, 4705–4712. [CrossRef][PubMed] 84. Chumakov, A.; Yuhina, E.; Frolova, E.; Kravchenko, J.; Chumakov, S. Expanding the application potential of DNA aptamers by their functionalization. Russ. J. Bioorg. Chem. 2016, 42, 1–13. [CrossRef] 85. Tonkinson, J.L.; Guvakova, M.; Khaled, Z.; Lee, J.; Yakubov, L.; Marshall, W.S.; Caruthers, M.H.; Stein, C. Cellular pharmacology and protein binding of phosphoromonothioate and phosphorodithioate oligodeoxynucleotides: A comparative study. Antisense Res. Dev. 1994, 4, 269–278. [CrossRef][PubMed] 86. Zandarashvili, L.; Nguyen, D.; Anderson, K.M.; White, M.A.; Gorenstein, D.G.; Iwahara, J. Entropic enhancement of protein-DNA affinity by oxygen-to-sulfur substitution in DNA phosphate. Biophys. J. 2015, 109, 1026–1037. [CrossRef][PubMed] 87. Vaught, J.D.; Bock, C.; Carter, J.; Fitzwater, T.; Otis, M.; Schneider, D.; Rolando, J.; Waugh, S.; Wilcox, S.K.; Eaton, B.E. Expanding the chemistry of DNA for in vitro selection. J. Am. Chem. Soc. 2010, 132, 4141–4151. [CrossRef][PubMed] 88. Davies, D.R.; Gelinas, A.D.; Zhang, C.; Rohloff, J.C.; Carter, J.D.; O’Connell, D.; Waugh, S.M.; Wolk, S.K.; Mayfield, W.S.; Burgin, A.B.; et al. Unique motifs and hydrophobic interactions shape the binding of modified DNA ligands to protein targets. Proc. Natl. Acad. Sci. USA 2012, 109, 19971–19976. [CrossRef] [PubMed] 89. Rohloff, J.C.; Gelinas, A.D.; Jarvis, T.C.; Ochsner, U.A.; Schneider, D.J.; Gold, L.; Janjic, N. Nucleic acid ligands with protein-like side chains: Modified aptamers and their use as diagnostic and therapeutic agents. Mol. Ther. Nucleic Acids 2014, 3, e201. [CrossRef][PubMed] 90. Forster, C.; Brauer, A.B.; Brode, S.; Schmidt, K.S.; Perbandt, M.; Meyer, A.; Rypniewski, W.; Betzel, C.; Kurreck, J.; Furste, J.P.; et al. Comparative crystallization and preliminary X-ray diffraction studies of locked nucleic acid and RNA stems of a tenascin C-binding aptamer. Acta Crystallogr. Sect. F Struct. Biol. Cryst. Commun. 2006, 62 Pt 7, 665–668. [CrossRef][PubMed] 91. Forster, C.; Oberthuer, D.; Gao, J.; Eichert, A.; Quast, F.G.; Betzel, C.; Nitsche, A.; Erdmann, V.A.; Furste, J.P. Crystallization and preliminary X-ray diffraction data of an LNA 7-mer duplex derived from a ricin aptamer. Acta Crystallogr. Sect. F Struct. Biol. Cryst. Commun. 2009, 65 Pt 9, 881–885. [CrossRef][PubMed] 92. Gelinas, A.D.; Davies, D.R.; Edwards, T.E.; Rohloff, J.C.; Carter, J.D.; Zhang, C.; Gupta, S.; Ishikawa, Y.; Hirota, M.; Nakaishi, Y.; et al. Crystal structure of interleukin-6 in complex with a modified nucleic acid ligand. J. Biol. Chem. 2014, 289, 8720–8734. [CrossRef][PubMed] 93. Hottin, A.; Marx, A. Structural insights into the processing of nucleobase-modified nucleotides by DNA polymerases. Acc. Chem. Res. 2016, 49, 418–427. [CrossRef][PubMed] 94. Umezu-Goto, M.; Kishi, Y.; Taira, A.; Hama, K.; Dohmae, N.; Takio, K.; Yamori, T.; Mills, G.B.; Inoue, K.; Aoki, J. Autotaxin has lysophospholipase D activity leading to tumor cell growth and motility by lysophosphatidic acid production. J. Cell Biol. 2002, 158, 227–233. [CrossRef][PubMed] Int. J. Mol. Sci. 2017, 18, 1683 21 of 21

95. Tokumura, A.; Majima, E.; Kariya, Y.; Tominaga, K.; Kogure, K.; Yasuda, K.; Fukuzawa, K. Identification of human plasma lysophospholipase D, a lysophosphatidic acid-producing enzyme, as autotaxin, a multifunctional phosphodiesterase. J. Biol. Chem. 2002, 277, 39436–39442. [CrossRef][PubMed] 96. Kato, K.; Ikeda, H.; Miyakawa, S.; Futakawa, S.; Nonaka, Y.; Fujiwara, M.; Okudaira, S.; Kano, K.; Aoki, J.; Morita, J.; et al. Structural basis for specific inhibition of autotaxin by a DNA aptamer. Nat. Struct. Mol. Biol. 2016, 23, 395–401. [CrossRef][PubMed] 97. Hausmann, J.; Kamtekar, S.; Christodoulou, E.; Day, J.E.; Wu, T.; Fulkerson, Z.; Albers, H.M.; van Meeteren, L.A.; Houben, A.J.; van Zeijl, L.; et al. Structural basis of substrate discrimination and integrin binding by autotaxin. Nat. Struct. Mol. Biol. 2011, 18, 198–204. [CrossRef][PubMed] 98. Kawaguchi, M.; Okabe, T.; Okudaira, S.; Nishimasu, H.; Ishitani, R.; Kojima, H.; Nureki, O.; Aoki, J.; Nagano, T. Screening and X-ray crystal structure-based optimization of autotaxin (ENPP2) inhibitors, using a newly developed fluorescence probe. ACS Chem. Biol. 2013, 8, 1713–1721. [CrossRef][PubMed] 99. Duchardt-Ferner, E.; Juen, M.; Kreutz, C.; Wöhnert, J. NMR resonance assignments for the tetramethylrhodamine binding RNA aptamer 3 in complex with the ligand 5-carboxy-tetramethylrhodamine. Biomol. NMR Assign. 2017, 11, 29–34. [CrossRef][PubMed] 100. Amano, R.; Aoki, K.; Miyakawa, S.; Nakamura, Y.; Kozu, T.; Kawai, G.; Sakamoto, T. NMR monitoring of the SELEX process to confirm enrichment of structured RNA. Sci. Rep. 2017, 7, 283. [CrossRef][PubMed] 101. Minagawa, H.; Onodera, K.; Fujita, H.; Sakamoto, T.; Akitomi, J.; Kaneko, N.; Shiratori, I.; Kuwahara, M.; Horii, K.; Waga, I. Selection characterization and application of artificial DNA aptamer containing appended bases with sub-nanomolar affinity for a salivary biomarker. Sci. Rep. 2017, 7, 42716. [CrossRef][PubMed] 102. Thirunavukarasu, D.; Chen, T.; Liu, Z.; Hongdilokkul, N.; Romesberg, F.E. Selection of 20-fluoro-modified aptamers with optimized properties. J. Am. Chem. Soc. 2017, 139, 2892–2895. [CrossRef][PubMed]

© 2017 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 (http://creativecommons.org/licenses/by/4.0/).