<<

Nucleobase Protection of Deoxyribo- and UNIT 2.1

SALIENT FEATURES OF of synthesis. For example, the exocyclic amino SYNTHESIS groups of , the 2′-hydroxy groups DNA and RNA are biopolymers comprised of sugars (UNIT 2.2), and the internucleotidic of units. The synthesis of DNA and phosphodiester functions, are protected using RNA as segments consisting of several nucleo- persistent protecting groups. tides each, referred to as , is The present commentary addresses issues quite a challenging endeavor, requiring the related to the persistent protection of nucleo- rapid and quantitative coupling of nucleosidic bases. The commentary is not a comprehensive units to form internucleotidic linkages. In order review of published literature but is intended to to achieve high efficiency in the coupling reac- highlight the salient features of pro- tions, it is necessary to direct the reaction to the tection. Only selected references are cited, and desired site in the . This is achieved readers should refer to recent reviews (Beau- by using appropriate protecting group strate- cage and Iyer, 1992; Iyer and Beaucage, 1999) gies. and primary literature for more comprehensive To date, five methods of oligonucleotide citations. synthesis have been reported, all of which use protected : (1) The H-phosphonate GENERAL ASPECTS OF approach (Michelson and Todd, 1955; Hall et NUCLEOBASE PROTECTION al., 1957; Froehler et al., 1986, and references The choice of nucleobase protecting groups therein; Garegg et al., 1986, and references and deprotection protocols is of paramount therein; UNIT 3.4); (2) the phosphodiester ap- importance in oligonucleotide synthesis, and is proach, pioneered by Khorana and co-workers dictated by many considerations. The protect- (Gilham and Khorana, 1958; Khorana, 1979); ing groups for nucleosides should be designed (3) the phosphotriester approach, which her- on the basis of several criteria, which have been alded a new era in synthesis by summarized by Reese (1978). (1) It should be enabling more rapid synthesis and purification possible to introduce the protecting group using (Letsinger and Ogilvie, 1969, and references a stable reagent that is readily obtainable. (2) therein; Reese, 1978); (4) the phosphite strat- The group should be achiral. (3) The nuclear egy (Letsinger and Lunsford, 1976, and refer- magnetic resonance (NMR) spectrum of the ences therein; Matteucci and Caruthers, 1980); protected nucleoside should be simple to inter- and (5) the phosphoramidite approach, pres- pret. (4) The protecting group should be readily ently the most popular method for oligonu- installed on the nucleoside. (5) The group cleotide synthesis (Beaucage and Caruthers, should enhance the solubility of the nucleoside 1981; UNIT 3.3). Over the years, improvements in organic solvents so that it can be adapted for in coupling chemistries, along with advances coupling reactions. (6) The group should be in methods of solid-phase assembly, have revo- stable to reagents employed in oligonucleotide lutionized the art of oligonucleotide synthesis. synthesis. (7) The group should not cause other In the synthesis of oligonucleotides, persist- structural changes in the nucleoside during its ent as well as transient protection of amino, installation or removal, or during oligonu- imido, and hydroxy groups of nucleosides are cleotide assembly. employed. Transient protecting groups are used A number of nucleobase protecting groups to temporarily block one of the hydroxy groups. have been reported. Improved procedures for For example, the 4,4′-dimethoxy trityl group their installation and removal have been pub- (DMTr) is used for transient protection of the lished. Nevertheless, unforeseen problems 5′ hydroxyl of a nucleoside (UNIT 2.3). The tran- have been reported when certain protecting sient protecting group is removed at the begin- groups are employed in oligonucleotide syn- ning of each coupling cycle. This enables the thesis. For example, N-acylated nucleo- coupling reaction to be directed to the desired sides, particularly N-acylated , hydroxy group during the synthesis cycle. On are more predisposed to depurination than their the other hand, persistent protecting groups are unprotected counterparts, making the growing Protection of those that remain on the nucleoside throughout oligonucleotide chain susceptible to cleavage Nucleosides for Oligonucleotide chain assembly and are only removed at the end during the synthesis cycle. In fact, each pro- Synthesis Contributed by Radhakrishnan P. Iyer 2.1.1 Current Protocols in Nucleic Acid Chemistry (2000) 2.1.1-2.1.17 Copyright © 2000 by John Wiley & Sons, Inc. tected purine and nucleobase exhib- Table 2.1.1 pKa Values of Nucleoside its different patterns of reactivity compared Nucleobases with the unprotected nucleoside. Certain pro- Nucleobase (site of protonation/ pK tecting groups are also sensitive to reagents deprotonation)a a used in oligonucleotide synthesis, resulting ′ either in their premature removal or in produc- 2 -Deoxythymidine (N3) 9.93 (N1) 9.42 tion of base-modified products (reviewed by (N3) 9.38 Beaucage and Iyer, 1992). (N3) 4.17 The sections that follow attempt to highlight (N1) 3.52 the structural basis for the nucleophilicity and aFor numbering of nucleoside positions, see Figure 2.1.1. reactivity of nucleosides. An overview of the For additional pKa values, see Clauwaert and Stockx strategies employed for nucleobase protection (1986); Dunn and Hall (1975). is also provided.

NUCLEOSIDE TAUTOMERISM tions, the tautomeric equilibrium could shift to AND pK VALUES where both imino and enol forms could exist a (Saenger, 1984). In turn, these factors influence The nucleophilicity of nucleobases (Fig. the reactivity of the nucleosides. 2.1.1) is dictated by the pKa of the amino and amido functions and their tautomeric forms. Table 2.1.1 lists the pKa values of nucleobases. REACTIVITY OF NUCLEOSIDES The amide-like nitrogens (N3 of uridine and Nucleosides participate in electrophilic and N1 of guanosine) are acidic in character, nucleophilic substitution reactions as well as whereas the ring nitrogens are basic. Therefore, addition reactions (Shabarova and Bogdanov, at strongly alkaline pH, the proton at N3 of 1994). Quite clearly, protecting groups and the uridine and and that at N1 of guanos- protocols for their installation and removal ine are removed. Under acidic conditions (at should be designed to avoid various side reac- pH ∼3), the sites of protonation are N1 of tions. adenosine and N3 of cytidine. At more acidic Nucleobases undergo substitution reactions pH, the N7 of guanosine and adenosine and O4 with electrophilic reagents. For example, both of uridine are protonated. Thus, all the bases N- and O-alkylation of the imide and lactam remain mostly uncharged in the physiological groups occur with alkylating agents. The N7 range of pH 5 to 9 (Saenger, 1984). position of is also a potential site for It is noteworthy that each of the nucleosides electrophilic attack (Fig. 2.1.5). Because of A, C, and G becomes protonated at one of the these competing reactions, simple alkylation of ring nitrogens rather than on the exocyclic exocyclic amino function is not a viable pro- amino group. Thus, the electron pair of the tection strategy for nucleobases. On the other amino group can delocalize into the heteroaro- hand, it is possible to chemoselectively acylate matic ring. Indeed the C–NH2 bonds of A, C, the exocyclic amino group. Thus, acyl-type and G are ∼1.34 Å long and have 40% to 50% protecting groups are widely used for the pro- double-bond character (Saenger, 1984). The tection of the exocyclic amino groups of nu- charge on a nucleobase, its tautomeric struc- cleosides (Fig. 2.1.7). tures, and its ability to form and accept hydro- The imide/lactam NH of thymidine, uridine gen bonds are also determined by its pKa values (pKa, 9.38), and guanosine (pKa, 9.42) is as well as by the pH of the medium. At physi- weakly acidic and can deprotonate under basic ological pH, the major nucleobases exist almost conditions. The resulting nucleophilic anion exclusively in the amino and keto tautomeric can react with a variety of reagents such as forms. However, under appropriate pH condi- activated phosphates, dicyclohexylcarbodi-

O NH O NH2 O 2 4 7 6 7 H C 5 3 3 N 5 1 N 5 1 3 4 NH 5 3 5 4 N 6 NH 5'1 5'2 N 5'1 5'2 NH 8 8 2 2 2 6 1 H H 1 2 H H 1 9 2 9 6 6 HO N 4 HO N HO N O HO N O N O N 4 N NH2 HO 5' O 3 O O 5' O O 3 1' 1' 4' 3' 2' 4' 3' 2'

HO HO HO HO H HO OH Nucleobase 2'1 Protection of deoxyadenosine deoxythymidine deoxycytosine uridine Deoxyribo- and Ribonucleosides Figure 2.1.1 Structures and numbering of nucleosides. 2.1.2

Current Protocols in Nucleic Acid Chemistry imide (DCC), mesitylene sulfonyl chloride, 1- Tri-O-acetyluridine (S.1) reacts with MSNT (mesitylene-2-sulfonyl)-3-nitro-1,2,4-triazole to produce the triazolo derivative S.2 (Fig. (MSNT), acid chlorides, phosphitylating re- 2.1.2; Reese and Ubasawa, 1980). During de- agents, and electrophilic reagents that are em- protection with ammonium hydroxide, S.2 ployed during coupling reactions. These side gives cytidine (S.3). Interestingly, during de- reactions result in nucleobase-derived N- and protection with either the tetramethyl- O-products. guanidinium salt of syn-4-nitrobenzaldoxime Nucleosides also react with a variety of or tetrabutylammonium fluoride (TBAF), S.2 nucleophilic reagents. For example, reverts to uridine (den Hartog et al., 1982). reacts with hydroxylamine at neutral and acidic The triazolo derivative S.2 is also known to pH to give the corresponding hydroxylamine react with other nucleophiles. For example, in derivative. It reacts with hydrazine at neutral the presence of methanol and 1,8-diazabicy- clo[3.4.0]undecene-7 (DBU), S.2 gives the cor- pH to give the corresponding hydrazide. In- responding 4-OMe derivative (Li et al., 1987). deed, nucleophilic substitution reaction of the S.1 reacts with phosphorylating reagents at exocyclic amino groups of cytosine proceeds O4 and undergoes 4-triazolation in the presence rapidly in the presence of alkali and amines. of the phosphorylating reagent 2- or 4-chlo- Under oxidative conditions, certain nucleo- rophenyl phosphorodi-1,2,4-triazolide. The bases (for example, and cytosine) can triazolo derivative is converted to the fluores- form N-oxides. Also, the C8 position of guanos- cent pyridinium salt S.4 in the presence of ine is vulnerable to hydrolytic attack under pyridine (Divakar and Reese, 1982; Sung, either strongly acidic or strongly alkaline con- 1982; Huynh-Dinh et al., 1985). Further reac- ditions. The 5,6 double bond of pyrimidine tion with triazole gives S.5 (Fig. 2.1.3). nucleosides also reacts with halogens and ha- The purine nucleosides also react with elec- lohydrins to give the corresponding addition trophilic reagents. For example, nu- products (Shabarova and Bogdanov, 1994). Se- cleosides react with MSNT to form the corre- lected examples of the side reactions that occur sponding triazolo derivatives. Reaction of the during oligonucleotide synthesis are given be- N2-protected guanosine nucleoside S.6 with low. mesitylene sulfonyl chloride gives the crystal-

NO2 N N O N NH2

NH N N MSNT NH3 AcO N O AcO N O HO N O O O O (PhO)2P(O)OH/C5H5N

AcO OAc AcO OAc HO OH 1 2 3

MSNT, 1-(mesitylene-2-sulfonyl)-3-nitro-1,2,4-triazole

Figure 2.1.2 Conversion of tri-O-acetyluridine to cytidine via a nitrotriazolide intermediate. Re- printed from Iyer and Beaucage (1999) with permission from Elsevier Science Publishing.

N N O N N

NH N N MSNT 1,2,4-triazole AcO N O AcO N O AcO N O O O O 4-ClPhOP(O)Cl2/C5H5N

AcO OAc AcO OAc AcO OAc 1 4 5 Protection of Figure 2.1.3 Formation of the triazolo derivative S.5 via the pyridinium intermediate S.4. MSNT, Nucleosides for 1-(mesitylene-2-sulfonyl)-3-nitro-1,2,4-triazole. Reprinted from Iyer and Beaucage (1999) with Oligonucleotide Synthesis permission from Elsevier Science Publishing. 2.1.3

Current Protocols in Nucleic Acid Chemistry R OOS O O N N NH N R-SO2-Cl AcO N AcO N O N NHTr O N NHTr DMAP

AcO OAc AcO OAc 6 7

DMAP, 4-dimethylaminopyridine; Tr, triphenylmethyl

6 Figure 2.1.4 Formation of an O -sulfonated derivative of guanosine. R⋅SO2⋅Cl, mesitylene sulfonyl chloride. Reprinted from Iyer and Beaucage (1999) with permission from Elsevier Science Publish- ing. R, 2,4,6-trimethylphenyl.

line O6-sulfonated derivative S.7 (Fig. 2.1.4; 1987). During oxidation of S.8 with iodine, O- Bridson et al., 1977; Francois et al., 1985). In to N-phosphoryl migration occurs (S.9 → S.10; the presence of pyridine, S.7 is converted to the Pon et al., 1985b). Once formed, S.10 induces corresponding C6 pyridinium compound. depurination resulting in chain-cleaved prod- The side reactions that are predominant in ucts upon treatment with ammonium hydroxide phosphodiester and phosphotriester chemistry during the final step of deprotection. have been summarized (Reese and Ubasawa, It is pertinent that base modifications can 1980). These side reactions are of concern when also occur during oligonucleotide deprotection. the H-phosphonate method is employed in oli- For example, residues are significantly gonucleotide synthesis in conjunction with modified as N3-methylthymine during depro- phosphorochloridates as coupling reagents. tection of oligonucleoside methyl phosphotri- The imide and lactam functions of nucleo- esters with aqueous ammonium hydroxide (Ur- sides also react with phosphitylating reagents. dea et al., 1986). However, when deprotection For example, the 2′-deoxyguanosine derivative of the methyl phosphate backbone of oligonu- S.20 (Fig. 2.1.5) reacts with methyl phos- cleotides is accomplished by treatment with phoramidites to give the O6-phosphitylated thiophenol or 2-mercaptobenzothiazole, fol- product S.8 (Pon et al., 1985a; Nielsen et al., lowed by aqueous ammonium hydroxide to

OR′ O RO P O N N N NH RO P N OR′ DMTrO N DMTrO N O N NHi-Bu O N NHi-Bu

AcO AcO 20 8

[31P NMR, d 133.95, 133.79 ppm]

I2

OR′ I OR′ O RO P O RO P O N N NH N

DMTrO N DMTrO N O N NHi-Bu O N NHi-Bu

AcO AcO 10 9

Nucleobase DMTr, 4,4'-dimethoxytriphenylmethyl; i-Bu, isobutyryl Protection of Deoxyribo- and Figure 2.1.5 Reaction of an N2-isobutyryl-2′-deoxyguanosine derivative with phosphitylating Ribonucleosides reagents. R, Me; R′, nucleoside. 2.1.4

Current Protocols in Nucleic Acid Chemistry remove nucleobase protecting groups, the for- the imide/lactam function of nucleosides can mation of N3-methylthymine is considerably potentially avert the side reactions. This has reduced (McBride et al., 1987; Andrus and been borne out by success in the synthesis of a Beaucage, 1988). number of (Reese, 1978). The reactivity of nucleosides and the atten- dant risk of base modification suggest an ap- PROTECTION OF IMIDE AND parent need to protect imide and lactam groups LACTAM FUNCTIONS of nucleosides in oligonucleotide synthesis. It A number of groups have been proposed for is expected that the use of protecting groups at the protection of thymine and and N3 and

Nucleobase Deprotection Protecting group References (protection site) conditions

U (O4) syn-4-Nitrobenzaldoxime/ Nylias et al., 1987 tetramethylguanidine MacMillan and Verdine, 1991

2,4,6-trimethylphenyl

NO2 G (O6) syn-4-Nitrobenzaldoxime/ Jones et al., 1981 U (O4) tetramethylguanidine Zhou and Chattopadhyaya, 1986 Zhou et al., 1986 Reese and Skone, 1984

2-nitrophenyl R T (O4) syn-4-Nitrobenzaldoxime/ Jones et al., 1981 U (O4) tetramethylguanidine Reese and Skone, 1984 R Scalfi-Happ et al., 1987 Rao et al., 1987 R = H or Me

Cl

G (O6) syn-4-Nitrobenzaldoxime/ Reese and Skone, 1984 tetramethylguanidine Rao et al., 1987 Hagen and Chladek, 1989 R R = H or Cl

O O S G (N1, O6) NH4OH, heat Mag and Engels, 1988 T/U (N3) (4-NO2)Ph

2-(4-nitrophenylsulfonyl)ethyl

Ph O T/U (N3) 10% Pd/H2 Krecmerová et al., 1990 benzyloxymethyl

R G (O6) Pyridine-2-aldoxime/ Pfister et al., 1988 T (O4) tetramethylguanidine Van Aerschot et al., 1988 0.5 M DBU/pyr R = NO2 or CN

R G (O6) R=Me3Si: n-Bu4NF/THF Hagen and Chladek, 1989 R=PhS: 4-NO2PhS: NaIO4/ Gaffney and Jones, 1982 Engels and Mag, 1982 R = Me3Si; PhS; NH4OH, heat; CN; 4-NO2PhS R=CN; DBU, or NH4OH

+ - G (O6) Pd(PPh3)4/PPh3, n-BuNH3 HCO2 Hayakawa et al., 1993 T (O4) allyl

Protection of Figure 2.1.6 Protecting groups for the imide/lactam functions of guanine, uracil, and thymine. Nucleosides for DBU, 1,8-diazabicyclo[3.4.0]undecene-7; NalO , sodium periodate; pyr, pyridine; THF, tetrahydro- Oligonucleotide 4 Synthesis furan. 2.1.5

Current Protocols in Nucleic Acid Chemistry Protecting group Nucleobase Deprotection References (protection site) conditions

Ph3CS U (N3) 0.1 M I2/THF/collidine/H2O Takaku et al., 1988 triphenylmethyl sulfenyl

O G (O6) NH4OH Kamimura et al., 1983b Ph2N diphenylcarbamoyl O OO S O U (N3) DBU/morpholine/pyr Nyilas et al., 1988 R R = H; Me O

Sekine, 1989, U (N3, O4) NH4OH Kamimura et al., 1983a Kamaike et al., 1988 R R = H; Me; OMe; Cl

NO2

U (N3) Pyr/H2O or 0.2 M n-Bu4NF/ Sekine, 1989 R S THF Welch et al., 1985

R = H; Me; NO2

O G (N2, O6); NH4OH/MeOH Fujii et al., 1987 U (N3) n-BuS butylthiocarbonyl O Pd[PPh ] /PPh , U (N3, O4) 3 4 3 Hayakawa et al., 1986 n-BuNH2/THF O allyloxycarbonyl

U (N3) NH4OH, heat, or Ito et al., 1986 MeO + - O Ph3C BF4 /MeCN/H2O (methoxyethoxy)methyl

Figure 2.1.6 (Continued)

O4, and of guanine at O6. A representative list when using the phosphotriester method in oli- of these groups is shown in Figure 2.1.6. For gonucleotide synthesis, O6-modified guanine the benefit of the readers, references that in- and O4-modified uracil revert to guanine and clude experimental details are cited. uracil residues upon “oximate” treatment that The imide- and lactam-protecting groups follows chain assembly. (2) When the phos- can be installed in two steps: sulfonylation or phoramidite method is used, O6-phosphitylated triazolation, followed by displacement of the deoxyguanosine reverts to deoxyguanosine on resulting O-sulfonate or triazolide by the nu- contact with water or acetate ions (Mag and cleophilic protecting group. The protecting Engels, 1988). Thus, during the synthesis cycle, groups can also be installed directly—for ex- if capping is performed (using acetic anhy- ample, via Mitsunobu alkylation (UNIT 1.2) or via dride) after coupling, any O6-modified deoxy- the transient protection approach. The prepara- tion of these protected nucleosides appears to guanosine can potentially revert back to deoxy- be straightforward (Jones et al., 1981; Gaffney guanosine. (3) Base modifications generated and Jones, 1982; Trichtinger et al., 1983; Nyilas during oligonucleotide chain assembly depend et al., 1987; Kamaike et al., 1988). on the reagents employed and the contact time. Solid-phase oligonucleotide synthesis uses Nucleobase However, it is still a matter of debate whether Protection of imide and lactam protection is necessary in automated pulsed delivery of reagents, result- Deoxyribo- and oligonucleotide synthesis. There are a number ing in shorter reaction times compared with Ribonucleosides of factors to be considered. (1) As noted before, solution-phase synthesis. Consequently, side 2.1.6

Current Protocols in Nucleic Acid Chemistry reactions during oligonucleotide chain assem- can be installed by peracylation of the nucleo- bly appear to be minimal. side followed by chemoselective O-deacyla- Thus, in principle, appropriate adjustment tion (Schaller et al., 1963). (2) The recent trend of synthesis protocols may obviate the neces- has been to use the “transient” protection ap- sity for imide and lactam protection. Neverthe- proach (Ti et al., 1982). In this procedure, the less, β-cyanoethyl protection is generally rec- nucleoside is persilylated using a silylating ommended for protection of guanosine at O6 agent, and the acyl function is installed on the or uridine at O4, and anisoyl for uridine at N3. amino group using the corresponding acid chloride or acid anhydride. (3) Some acyl func- PROTECTION OF EXOCYCLIC tions such as benzoyl, α-phenylcinnamoyl, and AMINO GROUPS naphthaloyl are directly incorporated on the On the basis of a number of criteria for nucleobase using the corresponding anhydride protecting groups, as outlined previously, N- (Watanabe and Fox, 1966; Bhat et al., 1989). acyl-type protection for the exocyclic amino (4) The exocyclic amino group of cytidine and groups has emerged as a logical choice. Figure can be directly acylated using 2.1.7 shows selected examples of N-acyl pro- activated esters (Igolen and Morin, 1980), acid tecting groups and the reagents used to effect chlorides (Mishra and Misra, 1986, and refer- their deprotection. ences therein), or alkyloxycarbonylbenzotria- The N-acyl groups are introduced into nu- zoles (Himmelsbach et al., 1984). (5) Recent cleosides by a number of procedures. (1) They reports suggest that site-selective incorporation

Protecting group Nucleobase Deprotection References (protection site) conditions O

7-Deaza-6-methyl-G (N2) NH4OH Seela and Driller, 1989 H formyl O C (N4) NH3/EtOH (1:1) Köster et al., 1981 Chaix et al., 1989 acetyl O G (N2); C (N4) NH4OH, heat Köster et al., 1981 Büchi and Khorana, 1972 Schulhof et al. 1987 isobutyryl O G (N2) NH4OH Schulhof et al., 1987 MeO

methoxyacetyl O G (N2); A (N6); C (N4) Pd[PPh3]4/HCO2H/Et2NH Hayakawa et al., 1986 O Hayakawa et al., 1990 allyloxycarbonyl

O i-PrO G (N2); A (N6); C (N4) NH4OH Uznanski et al., 1989

isopropoxyacetyl

O

G (N2); A (N6); C (N4) 0.5 M NH2NH2·H2O/ Ogilvie et al., 1982 pyr/AcOH (4:1) O levulinyl O G (N2); A (N6); C (N4) Multiple protocols Iyer et al., 1997

4-pentenoyl Protection of Nucleosides for Figure 2.1.7 Protecting groups for the exocyclic amino function of nucleobases. RT, room Oligonucleotide Synthesis temperature; pyr, pyridine. 2.1.7

Current Protocols in Nucleic Acid Chemistry Protecting group Nucleobase Deprotection References (protection site) conditions O G (N2); A (N6); C (N4) DBU or DBU/pyr Himmelsbach et al., 1984 Pfleiderer et al., 1985 (4-NO2)Ph O Pfister and Pfleiderer, 1989 4-nitrophenylethyloxycarbonyl R O G (N2) 5 M NH3/ MeOH Jones et al., 1981 Rao et al., 1987 Köster et al., 1981 Balgobin et al., 1981 R = H, C(CH3)3

O

O G (N2); A (N6); C (N4) NH4OH/pyr Scalfi-Happ et al., 1987 Et3N/pyr Hagen and Chládek, 1989 Heikkilä and Chattopadhyaya, 1983

9-fluorenylmethoxycarbonyl O Ph G (N2); A (N6); C (N4) NH4OH, heat Nagaich and Misra, 1989

Ph H α-phenylcinnamoyl R O O G (N2); A (N6) NH4OH Chaix et al., 1989 Schulhof et al., 1987 Singh and Misra, 1988

R = H, Cl R O Köster et al., 1981 O G (N2); A (N6); C (N4) 0.2 M NaOH/MeOH Sinha et al., 1993 or NH OH or NH gas 4 3 Boal et al., 1996

CH3C R = H, Cl

4-(tert-butyl)phenoxyacetyl O

G (N2); A (N6); C (N4) NH4OH Brown et al., 1989 Rao et al., 1987 Tanimura et al., 1988 R Schaller et al., 1963 Balgobin et al., 1981 R = H, OMe, Cl, NO2, NMe2, CMe3

O O O S G (N2); A (N6); C (N4) Tetramethylguani- Nyilas et al., 1988 O dine/morpholine/pyr; DBU/morpholine/pyr; R NH4OH/morpholine/pyr. R =H; Cl; NO2 O Ph G (N2) NH4OH, heat Marugg et al., 1984 Ph diphenylacetyl O Cl G (N2) 0.2 M NaOH/MeOH Koster et al., 1981

Cl 3,4-dichlorobenzoyl O MeO G (N2); A (N6); C (N4) NH4OH, heat Mishra and Misra, 1986

PhO 3-methoxy-4-phenoxybenzoyl Nucleobase Protection of Deoxyribo- and Ribonucleosides Figure 2.1.7 (Continued) 2.1.8

Current Protocols in Nucleic Acid Chemistry Protecting group Nucleobase Deprotection References (protection site) conditions O

C (N4) 0.2 M NaOH/MeOH (1:1) Köster et al., 1981

PhN N 4-(phenylazo)benzoyl

O

G (N2); A (N6); C (N4) 0.05 M K2CO3/MeOH; Kuijpers et al., 1990 NH4OH, RT OR

O R = Ac, Ph

G (N2); A (N6); C (N4) NH4OH, heat Dikshit et al., 1988

O O

1,8-naphthaloyl O

G (N2); A (N6); C (N4) n-Bu4NF/pyr/H2O Dreef-Tromp et al., 1990 O Ph Si C(CH ) Ph 3 3 2-(tert-butyldiphenylsilyloxymethyl)benzoyl

Figure 2.1.7 (Continued) and removal of N-acyl protecting groups can deoxyadenosine or deoxyguanosine. The hy- also be achieved by enzymatic methods (Prasad drolytic lability is also determined by inductive, and Wengel, 1996). resonance, and steric effects. For example, in a Overall, most N-acyl protecting groups are series of N-acyl nucleosides, N-benzoyl nucleo- stable in neutral or acidic medium and moder- sides are hydrolyzed sixteen times faster than ately stable at high pH (pH >13). However, N-(2,4-dimethyl)benzoyl nucleosides, presum- many N-acyl protecting groups are readily re- ably because of steric effects. Similarly, N-(2,4- moved by ammonolysis. This observation dimethoxy)benzoyl nucleoside is hydrolyzed forms the basis for the widespread use of 28% eight times faster than N-(4-dimethyl- NH4OH as a deprotection reagent when N-acyl- amino)benzoyl nucleoside, perhaps due to a com- protected nucleosides are employed in oligonu- bination of inductive and resonance effects. cleotide synthesis. The choice of a particular N-acyl protecting A salient feature of N-acyl protecting groups group also depends on the type of coupling is that their stability in alkaline pH can be modu- chemistry that is employed. For example, when lated by the steric and electronic characteristics phosphodiester and phosphotriester chemis- of specific acyl groups. A study comparing the tries are used in oligonucleotide synthesis, it is stability of various N-acyl nucleosides in alkaline necessary to select sturdy N-acyl protecting medium (0.2 N NaOH/MeOH) has been reported groups that can withstand the harsh reagents (Köster et al., 1981). Importantly, the stability of and conditions employed during synthesis. the acyl function towards alkaline hydrolysis is This requirement is met by the benzoyl group Protection of determined by the of the heterocyclic base. for adenine and cytosine, and the isobutyryl Nucleosides for For example, the rate of deacylation of N-acyl group for guanine. However, removal of these Oligonucleotide Synthesis derivatives of deoxycytidine is faster than that of protecting groups requires prolonged heating 2.1.9

Current Protocols in Nucleic Acid Chemistry (12 to 14 hr) with 28% NH4OH at 55°C. In spite mechanism for depurination of deoxyadenos- of this limitation, these protecting groups have ine (Fig. 2.1.8) involves initial protonation of remained popular even with the advent of auto- the nucleoside to produce the N1-protonated mated solid-phase oligonucleotide synthesis form S.11, and then equilibration (prototropic using phosphoramidite and H-phosphonate shift) to the N7-protonated species S.12 or S.13, chemistries. followed by cleavage of the to give S.15 via the oxonium S.14 (Zoltewicz et PROTECTION OF PURINE al., 1970; Zoltewicz and Clark, 1972). It is also NUCLEOBASES: THE PROBLEM conceivable that, at lower pH, depurination can OF DEPURINATION occur via protonation of the purine nucleobase ′ The development of suitable protecting at both N1 and N7. Presence of the 2 -OH has a groups for purine nucleobases has been an area significant effect on the nucleoside’s susceptibil- of considerable interest because purine nucleo- ity to depurination. For example, guanosine and sides rapidly depurinate under acidic condi- adenosine are more resistant to depurination tions. The problem is compounded by the acid- compared with deoxyadenosine and deoxy- labile DMTr group used for protection of the guanosine. Deoxyadenosine itself depurinates 5′-OH in solid-phase oligonucleotide synthe- 1200 times faster than adenosine (York, 1981). sis. Prior to each coupling step in the synthesis Interestingly, N-acyl-protected purine nu- cycle, the DMTr group is removed by exposure cleosides (particularly deoxyadenosine) are to a strong acid such as 2% dichloroacetic acid more prone to depurination than unprotected in dichloromethane. Consequently, the grow- nucleosides. Among N-acyl-protected de- ing oligonucleotide chain is repeatedly exposed oxyadenosines, protection at N6 with α- to strongly acidic conditions, potentially result- phenylcinnamoyl, naphthaloyl, 3-methoxy-4- ing in depurination and reduced yield of the phenoxybenzoyl, 9-fluorenylmethoxycar- desired “full-length” product. bonyl (FMOC), and tert-butylphenoxyacetyl The kinetics and mechanisms of nucleoside (t-PAC) groups (Fig. 2.1.7) provides greater depurination have been investigated by several resistance to depurination than with N6-benzoyl research groups (Romero et al., 1978; Oivanen (reviewed in Beaucage and Iyer, 1992). It is et al., 1987; Suzuki et al., 1994). The presumed believed that in the case of acyl-protected

NH2 NH2 7 6 N 1 N N NH 9 H HO N N HO N N O 3 O

HO HO 11

NH H 2 N N

HO HO HO N O O O N OH

HO HO HO oxonium ion 12 15 14

O H N NH

HO N O N NH2

HO 13

Nucleobase Protection of Figure 2.1.8 Scheme showing the proposed depurination mechanism for 2′-deoxyguanosine and Deoxyribo- and 2′-deoxyadenosine catalyzed by protic acids. Modified from Iyer and Beaucage (1999) with Ribonucleosides permission from Elsevier Science Publishing. 2.1.10

Current Protocols in Nucleic Acid Chemistry Cl Cl OCOPh

Cl Cl OCOPh Ph Ph O C O N O N O O N O HN N N N N N N N N OCOPh N N RO N RO N N RO N RO N N O O O O

RO RO RO RO 16 17 18 19

Figure 2.1.9 Examples of N6-protecting groups for 2′-deoxyadenosine derivatives that reduce depurination. Reprinted from Iyer and Beaucage (1999) with permission from Elsevier Science Publishing. R, DMTr. purine nucleosides under acidic conditions, the solvent used during the oxidation step in solid- initial site of protonation is N7, rendering the phase oligonucleotide synthesis. Thus, alter- protonated species more prone to glycosidic nate oxidants have to be employed for the cleavage. Naturally, caution should be exer- oxidation step. cised in the synthesis of oligonucleotides Since N-acyl-protected purine nucleosides whose sequence contains at are sensitive to deacylation, alternate protecting the 3′ terminus. groups have been investigated. Prominent Bis-acylation has also been studied as a among these are the amidine protecting groups, strategy to reduce depurination. Imide protect- which are introduced using an exchange reac- ing groups S.16 and S.17, as well as the diamide tion with appropriate amidine acetals. Interest- protecting group S.18, have been investigated ingly, N-amidine-protected nucleosides (Fig. (Fig. 2.1.9; Kume et al., 1982, 1984). However, 2.1.10) resist depurination 20-fold better than these groups were labile to aqueous pyridine, a the corresponding N-benzoyl nucleosides

Protecting group Nucleobase Deprotection References (protection site) conditions

H G (N2), A (N6) NH4OH, heat Smrt and Sorm, 1967 Vu et al., 1990 Sproat et al., 1991 Caruthers et al., 1985 Me2N McBride et al., 1986 (dimethylamino)methylene H G (N2), A (N6) NH4OH, heat; 0.5 M McBride et al., 1986 NH2NH2·H2O/pyr/AcOH Froehler and Matteucci, 1983 n-Bu2N (di-n-butylamino)methylene H A (N6) NH4OH/10% NH4OAc/heat; Froehler and Matteucci, 1983 0.5 M NH2NH2·H2O/pyr/AcOH i-Pr2N (diisopropylamino)methylene

G (N2), A (N6) NH4OH, heat McBride et al., 1986 Me2N 1-(dimethylamino)ethylidene

H A (N6) NH4OH/10% NH4OAc/heat; Froehler and Matteucci, 1983 0.5 M NH2NH2·H2O/pyr/AcOH i-Bu2N (diisobutylamino)methylene

C (N4) NH4OH, heat McBride et al., 1986 N Me N-methylpyrrolidin-2-ylidene Protection of Nucleosides for Oligonucleotide Synthesis Figure 2.1.10 Amidine protecting groups for exocyclic amino functions. Pyr, pyridine. 2.1.11

Current Protocols in Nucleic Acid Chemistry (Smrt and Sorm, 1967; Holy and Zemlicka, member of the family of N-acyl protecting 1969, and references therein; Froehler and Mat- groups, the N4-acetyl of cytosine has been used teucci, 1983; Caruthers et al., 1985; Vu et al., for “ultrafast” DNA synthesis using the phos- 1990; Sproat et al., 1991). phoramidite approach. Rapid deprotection It is presumed that the protonation sites of rates were achieved using methylamine/ammo- amidine-protected nucleosides are N1 and N6 nia (Reddy et al., 1994). This group is unsuit- instead of N1 and N7, respectively, resulting in able for use in phosphodiester and phosphotri- a slower rate of depurination. Indeed, amidine- ester chemistries, however. protected nucleoside phosphoramidites are fre- The synthesis of oligonucleotide analogs quently employed in solid-phase oligonu- carrying sensitive backbones requires protect- cleotide synthesis. However, changes are re- ing groups that (1) withstand the synthetic rig- quired in the oxidation step to avoid nucleobase ors of chain assembly, and (2) can be removed modifications (Mullah et al., 1995). chemoselectively under mild conditions. Nu- Nucleosides protected with O-nitrophenyl- cleobases protected by phenoxyacetyl (Singh sulfonyl and tris(benzoyloxy)trityl (S.19) and Misra, 1988; Chaix et al., 1989; Sproat et groups also appear to be more resistant to depu- al., 1991), and their derivatives such as t-PAC, rination (Shimidzu and Letsinger, 1968; Honda show accelerated deacylation (Köster et al., et al., 1984, and references therein). However, 1981; Sinha et al., 1993) under mildly basic the derived nucleoside phosphoramidites cou- conditions. It is presumed that the inductive ple less efficiently than the N-acyl-protected effect of the phenoxy group renders the amide nucleoside phosphoramidites (Sekine et al., carbonyl group more susceptible to nucleo- 1985). philic attack, facilitating rapid base-catalyzed Depurination is also influenced by other hydrolysis. Thus, PAC- and t-PAC-phos- factors such as the nature of the solid support phoramidites have been employed for rapid (controlled-pore glass versus polystyrene; see synthesis of oligonucleotides and certain ana- UNIT 3.1), the composition of the deblocking logs. As a rule, following oligonucleotide as- solution and deblocking time, and the washing sembly on solid support, the PAC and t-PAC solvent and washing time that are employed in groups are removed under milder conditions solid-phase synthesis of oligonucleotides (Paul (28% NH4OH, room temperature; Sinha et al., and Royappa, 1996). Depurination is faster at 1993) or, according to a recent report, using terminal sites than at internal sites in an oli- gaseous amines under pressure (Boal et al., gonucleotide chain (Suzuki et al., 1994). Inter- 1996). However, the use of t-PAC-protected estingly, a solution of 15% dichloroacetic acid nucleoside phosphoramidites results in trans (DCA) in methylene chloride was ideal as a acylation of the t-PAC protecting groups during detritylating reagent that induced minimal the capping step when acetic anhydride is em- depurination compared to the traditionally used ployed as a capping reagent. Thus, tert-butyl- 2% DCA/methylene chloride. phenoxyacetic anhydride should be used for the It is pertinent that with modern DNA syn- capping step in solid-phase oligonucleotide thesizers the pulsed delivery of reagents to the synthesis (Sinha et al., 1993). synthesis columns, in conjunction with opti- The concept of neighboring-group partici- mized synthesis programs, results in short con- pation has also been used to design acyl pro- tact times and has greatly minimized the depu- tecting groups and to accelerate the deprotec- rination problem. Thus, N-acyl-protected nu- tion step (Dreef-Tromp et al., 1990; Kuijpers et cleoside (benzoyl for dA and dC, and isobutyryl al., 1990). For example, removal of the 2-(ace- for dG) phosphoramidites and H-phosphonates toxymethyl)benzoyl group from nucleosides can be used for the efficient synthesis of oli- under basic conditions is accelerated by intra- gonucleotides. molecular participation of the deacylated hy- droxymethyl group. RECENT TRENDS IN New protecting groups have also been intro- NUCLEOBASE PROTECTION duced that can be chemoselectively removed Over the past few years, new applications of under neutral or mildly basic conditions. In- oligonucleotides in diagnostics and therapeu- deed, the allyloxycarbonyl protecting group tics have emerged, necessitating the expedi- (Hayakawa et al., 1986, 1990) is chemoselec- Nucleobase tious synthesis of large numbers of oligonu- tively removed using Pd(0), whereas the (p-ni- Protection of cleotides. In order to speed the synthesis proc- trophenyl)ethoxycarbonyl group (Trichtinger Deoxyribo- and ess, more “labile” N-acyl protecting groups for et al., 1983; Pfleiderer et al., 1985; Pfister et al., Ribonucleosides nucleosides have been sought. As the simplest 1988) and the 2-dansylethoxy cabonyl group 2.1.12

Current Protocols in Nucleic Acid Chemistry (Wagner and Pfleiderer, 1997) are selectively and for the synthesis and manufacture of novel removed using DBU. Consequently, support- analogs. It is hoped that the present commen- bound oligonucleotides can be prepared using tary will serve as a framework for developing building blocks that carry these protecting groups. new protecting group strategies for oligonu- Recently, the N-pent-4-enoyl (PNT) group has cleotide synthesis that could meet these chal- been introduced as a new acyl protecting group lenges. for situations where multiple deprotection pro- tocols are used (Iyer et al., 1997; see also ACKNOWLEDGMENT references therein). PNT-protected nucleoside The author wishes to thank Dr. WenQiang phosphoramidites can potentially be used for Zhou for his help with the artwork in this the rapid synthesis of oligonucleotides and oli- manuscript. gonucleotide analogs, as well as for the prepa- ration of support-bound oligonucleotides. LITERATURE CITED Several groups have been reported for nu- Andrus, A. and Beaucage, S.L. 1988. 2-Mercap- cleobase protection and may be particularly tobenzothiazole—an improved reagent for the valuable in the synthesis of oligonucleotides removal of methylphosphate protecting groups from oligodeoxynucleotide phosphotriesters. intended for specific applications. Figure 2.1.7 Tetrahedron Lett. 29:5479-5482. shows a partial list of protecting groups. A Balgobin, N., Josephson, S., and Chattopadhyaya, complete list of such groups is covered in other J.B. 1981. A general approach to the chemical reviews (Sonveaux, 1986; Beaucage and Iyer, synthesis of oligodeoxyribonucleotides. Acta. 1992; Iyer and Beaucage, 1999). Nonetheless, Chem. Scand. B35:201-212. it should be noted that many of these still need Beaucage, S.L. and Caruthers, M.H. 1981. De- to be evaluated in routine synthesis. oxynucleoside phosphoramidites—A new class Because of various issues associated with of key intermediates for deoxypolynucleotide nucleobase protection, oligonucleotide synthe- synthesis. Tetrahedron Lett. 22:1859-1862. sis has been evaluated using building blocks Beaucage, S.L. and Iyer, R.P. 1992. Advances in the bearing unprotected nucleobases. However, synthesis of oligonucleotides by the phos- phoramidite approach. Tetrahedron 48:2223- these efforts have met with only limited success 2311. (Narang et al., 1972; Fourrey and Varenne, Beaucage, S.L. and Iyer, R.P. 1993a. The function- 1985; Gryaznov and Letsinger, 1991; Uchi- alization of oligonucleotides via phos- yama et al., 1993, and references therein). phoramidite derivatives. Tetrahedron 49:1925- Clearly, more work is necessary in this area. 1963. Beaucage, S.L. and Iyer, R.P. 1993b. The synthesis CONCLUSION of modified oligonucleotides by the phos- phoramidite approach and their applications. Evidently, a nucleobase protecting group Tetrahedron 49:6123-6194. should meet several criteria before it can be Beaucage, S.L. and Iyer, R.P. 1993c. The synthesis adapted for routine oligonucleotide synthesis. of specific and unrelated phos- It is imperative, therefore, that the potential for phorylated by the phosphoramidite side reactions be closely examined when de- method. Tetrahedron 49:10441-10488. signing new reagents, evaluating new protect- Bhat, V., Ugarkar, B.G., Sayeed, V.A., Grimm, K., ing groups, and implementing modifications of Kosora, N., and Domenico, P. 1989. A simple established protocols during oligonucleotide and convenient method for the selective N-acy- synthesis, as well as during the manufacture of lations of cytosine nucleosides. Nucleosides Nu- cleotides 8:179-183. oligonucleotides and their analogs. The development of nucleobase protecting Boal, J.H., Wilk, A., Harindranath, N., Max, E.E., Kempe, T., and Beaucage, S.L. 1996. Cleavage groups and deprotection protocols has been of oligodeoxyribonucleotides from controlled- crucial for the successful synthesis of oligonu- pore glass supports and their rapid deprotection cleotides, functionalized oligonucleotides, oli- by gaseous amines. Nucl. Acids Res. 24:3115- gonucleotide analogs, ribonucleotides, and 3117. phosphorylated biomolecules (reviewed in Bridson, P.K., Markiewicz, W., and Reese, C.B. Beaucage and Iyer, 1992, 1993a,b,c). As in the 1977. Acylation of 2′,3′,5′-tri-O-acetylguanosine. past, oligonucleotides are expected to play a J. Chem. Soc., Chem. Commun. 791-792. dominant role in fostering advances in func- Brown, J.M., Christodoulou, C., Modak, A.S., tional genomics, proteomics, diagnostics, and Reese, C.B., and Serafinowska, H.T. 1989. Syn- thesis of the 3′-terminal half of yeast Protection of therapeutics. Consequently, continued demand transfer ribonucleic acid (tRNAala) by the phos- Nucleosides for exists for simultaneous synthesis of large num- photriester approach in solution. Part 2. J. Chem. Oligonucleotide Synthesis bers of oligonucleotides in miniature formats, Soc. Perkin Trans. 1:1751-1767. 2.1.13

Current Protocols in Nucleic Acid Chemistry Büchi, H. and Khorana, H.G. 1972. CV. Total syn- Froehler, B.C., Ng, P.G., and Matteucci, M.D. 1986. thesis of the structural for an alanine trans- Synthesis of DNA via deoxynucleoside H-phos- fer ribonucleic acid from yeast. Chemical syn- phonate intermediates. Nucl. Acids Res. thesis of an icosadeoxyribonucleotide corre- 14:5399-5407. sponding to the nucleotide sequence 31 to 50. J. Mol. Biol. 72:251-288. Fujii, M., Yamakage, S., Takaku, H., and Hata, T. 1987. (Butylthio)carbonyl group: A new protect- Caruthers, M.H., McBride, L.J., Bracco, L.P., and ing group for the guanine residue in oligoribonu- Dubendorff, J.W. 1985. Studies on nucleotide cleotide synthesis. Tetrahedron Lett. 28:5713- chemistry 15. Synthesis of oligodeoxynu- 5716. cleotides using amidine protected nucleosides. Nucleosides 4:95-105. Gaffney, B.L. and Jones, R.A. 1982. A new strategy Chaix, C., Molko, D., and Téoule, R. 1989. The use for the protection of deoxyguanosine during oli- of labile base protecting groups in oligoribonu- gonucleotide synthesis. Tetrahedron Lett. cleotide synthesis. Tetrahedron Lett. 30:71-74. 23:2257-2260. Clauwaert, J. and Stockx, J. 1986. Interactions of Garegg, P.J., Lindh, I., Regberg, T., Stawinski, J., polynucleotides and their components. I. Disso- Strömberg, R., and Henrichson, C. 1986. Nu- ciation constants of the bases and their deriva- cleoside H-phosphonates. IV. Automated solid tives. Z. Naturforsch. B. 23:25-30. phase synthesis of oligoribonucleotides by the hydrogenphosphonate approach. Tetrahedron den Hartog, J.A.J., Willie, G., Scheublin, R.A., and Lett. 27:4055-4058. van Boom, J.H. 1982. Chemical synthesis of a messenger ribonucleic acid fragment: AUGUU- Gilham, P.T. and Khorana, H.G. 1958. Studies on CUUCUUCUUCUUC. 21:1009- polynucleotides. I. A new and general method for 1018. the chemical synthesis of the C5′-C3′ internu- Dikshit, A., Chaddha, M., Singh, R.K., and Misra, cleotidic linkage. Syntheses of deoxyribo-dinu- K. 1988. Naphthaloyl group: A new selective cleotides. J. Am. Chem. Soc. 80:6212-6222. amino protecting group for deoxynucleosides in oligonucleotide synthesis. Can. J. Chem. Gryaznov, S.M. and Letsinger, R.L. 1991. Synthesis 66:2989-2994. of oligonucleotides via monomers with unpro- tected bases. J. Am. Chem. Soc. 113:5876-5877. Divakar, K.J. and Reese, C.B. 1982. 4-(1,2,4-Tria- zol-1-yl)- and 4-(3-nitro-1,2,4-triazol-1-yl)-1- Hagen, M.D. and Chládek, S. 1989. General synthe- (β-D-2,3,5-tri-O-acetylarabinofuranosyl)pyrim sis of 2′(3′)-O-aminoacyl oligoribonucleotides. idin-2(1H)-ones. Valuable intermediates in the The protecion of the guanine moiety. J. Org. synthesis of derivatives of 1-(β-D-arabinofura- Chem. 54:3189-3195. nosyl)cytosine (Ara-C). J. Chem. Soc. Perkin Trans. 1:1171-1176. Hall, R.H., Todd, A.R., and Webb, R.F. 1957. Nu- cleotides. Part XLI. Mixed anhydrides as inter- Dreef-Tromp, C.M., van Dam, E.M.A., van den Elst, mediates in the synthesis of dinucleoside phos- H., van der Marel, G.A., and van Boom, J.H. phates. J. Chem. Soc. 3291-3296. 1990. Solid-phase synthesis of H-Phe-Tyr- (pATAT)-NH2: A nucleopeptide fragment from Hayakawa, Y., Kato, H., Uchiyama, M., Kajino, H., the nucleoprotein of bacteriophage φX174. Nucl. and Noyori, R. 1986. Allyloxycarbonyl group: A Acids Res. 18:6491-6495. versatile blocking group for nucleotide synthe- Dunn, D.B., and Hall, R.H. 1975. Purines, pyrimid- sis. J. Org. Chem. 51:2400-2402. ines, nucleosides and nucleotides: Physical con- Hayakawa, Y., Wakabayashi, S., Kato, H., and Noy- stants and spectral properties. In Handbook of ori, R. 1990. The allylic protection method in Biochemistry and Molecular , 3rd ed., solid-phase oligonucleotide synthesis. An effi- Vol. 1: Nucleic Acids (G.D. Fasman, ed.) pp. cient preparation of solid-anchored DNA oli- 65-125. CRC Press, Boca Raton, Fla. gomers. J. Am. Chem. Soc. 112:1691-1696. Engels, J.W. and Mag, M. 1982. Amide protection in oligodeoxynucleotide synthesis. Nucleosides Hayakawa, Y., Hirose, M., and Noyori, R. 1993. Nucleotides 6:473-475. O-Allyl protection of guanine and thymine resi- dues in oligodeoxyribonucleotides. J. Org. Fourrey, J.-L. and Varenne, J. 1985. Preparation and Chem. 58:5551-5555. phosphorylation reactivity of N-nonacylated nu- cleoside phosphoramidites. Tetrahedron Lett. Heikkilä, J. and Chattopadhyaya, J. 1983. The 9- 26:2663-2666. fluorenylmethoxycarbonyl (Fmoc) group for the Francois, P., Hamoir, G., Sonveaux, E., Vermeersch, protection of amino functions of cytidine, adeno- ′ H., and Ma, Y. 1985. On the phosphorylation of sine, guanosine and their 2 -deoxysugar deriva- and the protection of de- tives. Acta Chem. Scand. B37:263-265. oxyguanosine. Bull. Soc. Chim. Belg. 94:821- Himmelsbach, F., Schulz, B.S., Trichtinger, T., 823. Charubala, R., and Pfleiderer, W. 1984. The p- Nucleobase Froehler, B.C. and Matteucci, M.D. 1983. Dialkyl- nitrophenylethyl (NPE) group. A versatile new Protection of formamidines: Depurination resistant N6-pro- blocking group for phosphate and aglycone pro- Deoxyribo- and tecting group for deoxyadenosine. Nucl. Acids tection in nucleosides and nucleotides. Tetrahe- Ribonucleosides Res. 11:8031-8036. dron 40:59-72. 2.1.14

Current Protocols in Nucleic Acid Chemistry Holy, A. and Zemlicka, J. 1969. Oligonucleotidic Krecmerová, M., Hrebabecky, H., and Holy, A. compounds. XXXIII. A study on hydrolysis of 1990. Synthesis of 5′-O-phosphonomethyl de- N-dimethylaminomethylenecytidine, -adeno- rivatives of pyridine 2′-deoxynucleosides. Col- sine, -guanosine, and related 2′-deoxy com- lect. Czech. Chem. Commun. 55:2521-2536. pounds. Collect. Czech. Chem. Commun. Kuijpers, W.H.A., Huskens, J., and van Boeckel, 34:2449-2458. C.A.A. 1990. The 2-(acetoxymethyl)benzoyl Honda, S., Urakami, K., Koura, K., Terada, K., Sato, (AMB) group as a new base-protecting group, Y., Kohno, K., Sekine, M., and Hata, T. 1984. designed for the protection of phosphate modi- Synthesis of oligoribonucleotides by use of S,S- fied oligonucleotides. Tetrahedron Lett. diphenyl N-monomethoxytrityl 31:6729-6732. 3′-phosphorodithioates. Tetrahedron 40:153- Kume, A., Sekine, M., and Hata, T. 1982. Phthaloyl 163. group: A new amino protecting group of de- oxyadenosine in oligonucleotide synthesis. Tet- Huynh-Dinh, T., Langlois d’Estaintot, B., Allard, P., rahedron Lett. 23:4365-4368. and Igolen, J. 1985. Synthèse simplifiée de son- des mixtes avec des triazolo-nucléosides. Tetra- Kume, A., Iwase, R., Sekine, M., and Hata, T. 1984. hedron Lett. 26:431-434. Cyclic diacyl groups for protection of the N6- amino group of deoxyadenosine in oligode- Igolen, J. and Morin, C. 1980. Rapid synthesis of oxynucleotide synthesis. Nucl. Acids Res. ′ protected 2 -deoxycytidine derivatives. J. Org. 12:8525-8538. Chem. 45:4802-4804. Letsinger, R.L. and Ogilvie, K.K. 1969. Synthesis Ito, T., Ueda, S., and Takaku, H. 1986. of oligothymidylates via phosphotriester inter- (Methoxyethoxy)methyl group: New amide and mediates. J. Am. Chem. Soc. 91:3350-3355. hydroxyl protecting groups of uridine in oli- Letsinger, R.L. and Lunsford, W.B. 1976. Synthesis gonucleotide synthesis. J. Org. Chem. 51:931- of thymidine oligonucleotides by phosphite tri- 933. ester intermediates. J. Am. Chem. Soc. 98:3655- Iyer, R.P. and Beaucage, S.L. 1999. Oligonucleotide 3661. synthesis. In Comprehensive Natural Products Li, B.F.L., Reese, C.B., and Swann, P.F. 1987. Syn- Chemistry, Vol. 7: DNA and Aspects of Molecu- thesis and characterization of oligodeoxynu- lar Biology (E.T. Kool, ed.) pp. 105-152. El- cleotides containing 4-O-methylthymine. Bio- sevier Science Publishing, New York. chemistry 26:1086-1093. Iyer, R.P., Yu, D., Habus, I., Ho, N.H., Johnson, S., MacMillan, A.M. and Verdine, G.L. 1991. Engineer- Devlin, T., Jiang, Z., Zhou, W., Xie, J., and ing tethered DNA molecules by the convertible Agrawal, S. 1997. N-Pent-4-enoyl (PNT) group nucleoside approach. Tetrahedron 47:2603- as a universal nucleobase protector: Applications 2616. in the rapid and facile synthesis of oligonu- Mag, M. and Engels, J.W. 1988. Synthesis and struc- cleotides, analogs and conjugates. Tetrahedron ture assignments of amide protected nucleosides 53:2731-2750. and their use as phosphoramidites in deoxyoli- Jones, S.S., Reese, C.B., Sibanda, S., and Ubasawa, gonucleotide synthesis. Nucl. Acids Res. A. 1981. The protection of uracil and guanine 16:3525-3543. residues in oligonucleotide synthesis. Tetrahe- Marugg, J.E., Tromp, M., Jhurani, P., Hoyng, C.F., dron Lett. 22:4755-4758. van der Marel, G.A., and van Boom, J.H. 1984. Synthesis of DNA fragments by the hydroxyben- Kamaike, K., Hasegawa, Y., and Ishido, Y. 1988. A 3 zotriazole phosphodiester approach. Tetrahe- simple, preparative procedure for N -anisoylu- dron 40:73-78. ridine and O6-diphenylcarbamoylguanosine 2′- O-(tetrahydropyran-2-yl) derivatives via the cor- Matteucci, M.D. and Caruthers, M.H. 1980. The responding 3′,5′-dibenzoates. Nucleosides Nu- synthesis of oligodeoxypyrimidines on a poly- cleotides 7:37-43. mer support. Tetrahedron Lett. 21:719-722. Kamimura, T., Masegi, T., Urakami, K., Honda, S., McBride, L.J., Kierzek, R., Beaucage, S.L., and Sekine, M., and Hata, T. 1983a. A new protecting Caruthers, M.H. 1986. Amidine protecting tactics for the uracil residue in oligoribonu- groups for oligonucleotide synthesis. J. Am. cleotide synthesis. Chem. Lett. 1051-1054. Chem. Soc. 108:2040-2048. McBride, L.J., Eadie, J.S., Efcavitch, J.W., and An- Kamimura, T., Tsuchiya, M., Koura, K., Sekine, M., drus, A. 1987. Base modification and the phos- and Hata, T. 1983b. Diphenylcarbamoyl and phoramidite approach. Nucleosides Nucleotides propionyl groups: A new combination of protect- 6:297-300. ing groups on the guanine residue. Tetrahedron Lett. 24:2775-2778. Michelson, A.M. and Todd, A.R. 1955. Nucleotides. Part XXXII. Synthesis of a dithymidine dinu- Khorana, H.G. 1979. Total synthesis of a gene. cleotide containing a 3′:5′-internucleotidic link- Science 203:614-625. age. J. Chem. Soc. 2632-2638. Köster, H., Kulikowski, K., Liese, T., Heikens, W., Mishra, R.K. and Misra, K. 1986. Improved synthe- Protection of and Kohli, V. 1981. N-acyl protecting groups for sis of oligodeoxyribonucleotide using 3- Nucleosides for deoxynucleosides. A quantitative and compara- methoxy-4-phenoxybenzoyl group for amino Oligonucleotide Synthesis tive study. Tetrahedron 37:363-369. protection. Nucl. Acids Res. 14:6197-6213. 2.1.15

Current Protocols in Nucleic Acid Chemistry Mullah, B., Andrus, A., Zhao, H., and Jones, R.A. Prasad, A.K. and Wengel, J. 1996. Enzyme-medi- 1995. Oxidative conversion of N-dimethylfor- ated protecting group chemistry on the hydroxyl mamidine nucleosides to N-cyano nucleosides. groups of nucleosides. Nucleosides Nucleotides Tetrahedron Lett. 36:4373-4376. 15:1347-1359. Nagaich, A.K. and Misra, K. 1989. Highly efficient Rao, T.S., Reese, C.B., Serafinowska, H.T., Takaku, synthesis of oligodeoxyribonucleotides using α- H., and Zappia, G. 1987. Solid-phase synthesis phenyl cinnamoyl group for selective amino pro- of the 3′-terminal nonadecaribonucleoside oc- tection. Nucl. Acids Res. 17:5125-5134. tadecaphosphate sequence of yeast alanine trans- Narang, S.A., Itakura, K., and Wightman, R.H. fer ribonucleic acid. Tetrahedron Lett. 28:4897- 1972. A simplification in the synthesis of de- 4900. oxyribooligonucleotides. Can. J. Chem. 50:769- Reddy, M.P., Hanna, N.B., and Farooqui, F. 1994. 770. Fast cleavage and deprotection of oligonu- Nielsen, J., Dahl, O., Remaud, G., and Chattopad- cleotides. Tetrahedron Lett. 35:4311-4314. hyaya, J. 1987. Phosphitylation of guanine or Reese, C.B. 1978. The chemical synthesis of oligo- bases during the preparation of nucleo- and poly-nucleotides by the phosphotriester ap- side phosphoramidites. Isolation of model prod- proach. Tetrahedron 34:3143-3179. ucts as thiophosphoric amide derivatives and structure elucidation by 15N NMR spectroscopy. Reese, C.B. and Ubasawa, A. 1980. Reaction be- Acta. Chem. Scand. B41:633-639. tween 1-arenesulphonyl-3-nitro-1,2,4-triazoles Nyilas, A., Zhou, X.-X., Welch, C.J., and Chattopad- and nucleoside residues. Elucidation of the na- hyaya, J. 1987. A versatile strategy for the O4- ture of side-reactions during oligonucleotide protection and modification of the lactam func- synthesis. Tetrahedron Lett. 21:2265-2268. tion of uridine and uridylic acid. Nucl. Acids Res. Reese, C.B. and Skone, P.A. 1984. The protection Symp. Ser. 18:157-160. of thymine and guanine residues in oligode- Nyilas, A., Földesi, A., and Chattopadhyaya, J. oxyribonucleotide synthesis. J. Chem. Soc. Per- 1988. Arenesulfonylethoxycarbonyl—A set of kin Trans. 1:1263. amino protecting groups for DNA and RNA Romero, R., Stein, R., Bull, H.G., and Cordes, E.H. synthesis. Nucleosides Nucleotides 7:787-793. 1978. Secondary deuterium isotope effects for Ogilvie, K.K., Nemer, M.J., Hakimelahi, G.H., acid-catalyzed hydrolysis of inosine and adeno- Proba, Z.A., and Lucas, M. 1982. N-Levulina- sine. J. Am. Chem. Soc. 100:7620-7624. tion of nucleosides. Tetrahedron Lett. 23:2615- Saenger, W. 1984. Principles of Nucleic Acids Struc- 2618. ture. Springer-Verlag, New York. Oivanen, M., Lönnberg, H., Zhou, X.X., and Chat- topadhyaya, J. 1987. Acidic hydrolysis of 6-sub- Scalfi-Happ, C., Happ, E., and Chládek, S. 1987. ′ ′ stituted 9-(2-deoxy-β-D-erythro-pentofurano- New approach to the synthesis of 2 (3 )-O-ami- ′ syl)purines and their 9-(1-alkoxyethyl) counter- noacyl-oligoribonucleotides related to the 3 -ter- parts: Kinetics and mechanism. Tetrahedron minus of aminoacyl transfer ribonucleic acid. 43:1133-1140. Nucleosides Nucleotides 6:345-348. Paul, C.H. and Royappa, A.T. 1996. Acid binding Schaller, H., Weimann, G., Lerch, W.B., and and detritylation during oligonucleotide synthe- Khorana, H.G. 1963. Studies on polynu- sis. Nucl. Acids Res. 24:3048-3052. cleotides. XXIV. The stepwise synthesis of spe- cific deoxyribopolynucleotides (4). Protected Pfister, M. and Pfleiderer, W. 1989. New results in derivatives of deoxyribonucleosides and new oligoribonucleotide synthesis. Nucleosides Nu- syntheses of -3′ phos- cleotides 8:1001-1006. phates. J. Am. Chem. Soc. 85:3821-3827. Pfister, M., Farkas, S., Charubala, R., and Pfleiderer, Schulhof, J.C., Molko, D., and Teoule, R. 1987. W. 1988. Recent progress in oligoribonucleotide Facile removal of new base protecting groups synthesis. Nucleosides Nucleotides 7:595-600. useful in oligonucleotide synthesis. Tetrahedron Pfleiderer, W., Himmelsbach, F., Charubala, R., Lett. 28:51-54. Schirmeister, H., Beiter, A., Schulz, B., and Seela, F. and Driller, H. 1989. 7-Deaza-2′-deoxy- Trichtinger, T. 1985. The p-nitrophenylethyl 6 2 group—An universal blocking group in nucleo- O -methylguanosine: Selective N -formylation side and nucleotide chemistry. Nucleosides Nu- via a formamidine, phosphoramidite. Synthesis cleotides 4:81-94. and properties of oligonucleotides. Nucleosides Nucleotides 8:1-21. Pon, R.T., Damha, M.J., and Ogilvie, K.K. 1985a. Necessary protection of the O6-position of Sekine, M. 1989. General method for the prepara- 3 4 guanosine during the solid phase synthesis of tion of N - and O -substituted uridine derivatives oligonucleotides by the phosphoramidite ap- by phase-transfer reactions. J. Org. Chem. proach. Tetrahedron Lett. 26:2525-2528. 54:2321-2326. Pon, R.T., Damha, M.J., and Ogilvie, K.K. 1985b. Sekine, M., Masuda, N., and Hata, T. 1985. Intro- Nucleobase Modification of guanine bases by nucleoside duction of the 4,4′,4′′-tris(benzoyloxy)trityl Protection of phosphoramidite reagents during the solid phase group into the exo amino groups of deoxyribonu- Deoxyribo- and synthesis of oligonucleotides. Nucl. Acids Res. cleosides and its properties. Tetrahedron Ribonucleosides 13:6447-6465. 41:5445-5453. 2.1.16

Current Protocols in Nucleic Acid Chemistry Shabarova, Z. and Bogdanov, A. 1994. Advanced Urdea, M.S., Ku, L., Horn, T., Gee, Y.G., and organic chemistry of nucleic acids. VCH Pub- Warner, B.D. 1986. Base modification and clon- lishers, New York. ing efficiency of oligodeoxyribonucleotides syn- thesized by the phosphoramidite method: Shimidzu, T. and Letsinger, R.L. 1968. Synthesis of Methyl versus cyanoethyl phosphorous protec- deoxyguanylyl-deoxyguanosine on an insoluble tion. Nucl. Acids Res. Symp. Ser. 16:257-260. polymer support. J. Org. Chem. 33:708-711. Uznanski, B., Grajkowski, A., and Wilk, A. 1989. Singh, R.K. and Misra, K. 1988. Improvements in The isopropoxyacetic group for convenient base oligodeoxyribonucleotide synthesis using phen- protection during solid-support synthesis of oli- oxyacetyl as amino protecting group. Indian J. godeoxyribonucleotides and their triester ana- Chem. 27B:409-417. logs. Nucl. Acids Res. 17:4863-4871. Van Aerschot, A., Herdewijn, P., Janssen, G., and Sinha, N.D., Davis, P., Usman, N., Pérez, J., Hodge, Vanderhaeghe, H. 1988. Protection of the lactam R., Kremsky, J., and Casale, R. 1993. Labile function of 2′-deoxyinosine with a 2-(4-ni- exocyclic amine protection of nucleosides in trophenyl)-ethyl moiety. Nucleosides Nucleo- DNA, RNA and oligonucleotide analog synthe- tides 7:519-536. sis facilitating N-deacylation, minimizing depu- rination and chain degradation. Biochimie 75:13- Vu, H., McCollum, C., Jacobson, K., Theisen, P., 23. Vinayak, R., Spiess, E., and Andrus, A. 1990. Fast oligonucleotide deprotection phos- Smrt, J. and Sorm, F. 1967. Oligonucleotidic com- phoramidite chemistry for DNA synthesis. Tet- pounds. XVIII. Synthesis of guanylyl-(3′-5′)- rahedron Lett. 31:7269-7272. uridylyl-(3′-5′)-arabinofuranosyluracil and guanylyl-(3′-5′)-uridylyl-(3′-5′)-arabinofurano Wagner, T. and Pfleiderer, W. 1997. Aglycone pro- -sylcytosine. Collect. Czech. Chem. Commun. tection by the (2-dansylethoxy)carbonyl (= {2- 32:3169-3176. {[5-(dimethylamino)naphthalen-1-yl] sul- fonyl}ethoxy}carbonyl; dnseoc) group—A new Sonveaux, E. 1986. The organic chemistry underly- variation in oligodeoxyribonucleotide synthesis. ing DNA synthesis. Bioorganic Chem. 14:274- Helv. Chim. Acta 80:200-212. 325. Watanabe, K.A. and Fox, J.J. 1966. A simple method for selective acylation of cytidine on the 4-amino Sproat, B.S., Iribarren, A.M., Guimil Garcia, R., and group. Angew. Chem. Intl. Ed. Engl. 5:579-580. Beijer, B. 1991. New synthetic routes to syn- thons suitable for 2′-O-allyloligoribonucleotide Welch, C.J., Bazin, H., Heikkilä, J., and Chattopad- assembly. Nucl. Acids Res. 19:733-738. hyaya, J. 1985. Synthesis of C-5 and N-3 arene- sulfenyl . Preparation and properties of Sung, W.L. 1982. Synthesis of 4-(1,2,4-triazol-1- a new class of uracil protecting group. Acta yl)pyrimidin-2(1H)-one- and its Chem. Scand. B39:203-212. application in synthesis of oligoribonucleotides. J. Org. Chem. 47:3623-3628. York, J.L. 1981. Effect of structure of the aglycon on the acid-catalyzed hydrolysis of adenine nu- Suzuki, T., Ohsumi, S., and Makino, K. 1994. cleosides. J. Org. Chem. 46:2171-2173. Mechanistic studies on depurination and Zhou, X.-X. and Chattopadhyaya, J. 1986. Site-spe- apurinic site chain breakage in oligodeoxyri- cific modification of the pyrimidine residue dur- bonucletides. Nucl. Acids Res. 22:4997-5003. ing the deprotection of the fully-protected di- Tetrahedron Takaku, H., Imai, K., and Nagai, M. 1988. uridylic acid. 42:5149-5156. Triphenylmethanesulfenyl group. A new pro- Zhou, X.X., Sandström, A., and Chattopadhyaya, J. tecting group for the uracil residue in oligori- 1986. A convenient preparation of 2-N-(4-t- bonucleotide synthesis. Chem. Lett. 857-860. butylbenzoyl)-6-O-(2-nitrophenyl)guanosine and its application in the synthesis of Tanimura, H., Fukazawa, T., Sekine, M., Hata, T., 5′(GpGpGpU)3′ constituting the 3′-anticodon Efcavitch, J.W., and Zon, G. 1988. The practical stem of E.coli tRNAIle. Chem. Scr. 26:241-249. synthesis of RNA fragments in the solid phase approach. Tetrahedron Lett. 29:577-578. Zoltewicz, J.A. and Clark, D.F. 1972. Kinetics and mechanism of the hydrolysis of guanosine and Ti, G.S., Gaffney, B.L., and Jones, R.A. 1982. Tran- 7-methylguanosine nucleosides in perchloric sient protection: Efficient one-flask synthesis of acid. J. Org. Chem. 37:1193-1197. protected deoxynucleosides. J. Am. Chem. Soc. Zoltewicz, J.A., Clark, D.F., Sharpless, T.W., and 104:1316-1319. Grahe, G. 1970. Kinetics and mechanism of the Trichtinger, T., Charubala, R., and Pfleiderer, W. hydrolysis of some purine nucleosides. J. Am. 1983. Synthesis of O6-p-nitrophenylethyl Chem. Soc. 92:1741-1750. guanosine and 2′-deoxyguanosine derivatives. Tetrahedron Lett. 24:711-714.

Uchiyama, M., Aso, Y., and Noyori, R. 1993. O-Se- Contributed by Radhakrishnan P. Iyer Protection of lective phosphorylation of nucleosides without OriGenix Technologies Nucleosides for N-protection. J. Org. Chem. 58:373-379. Laval, Quebec, Canada Oligonucleotide Synthesis 2.1.17

Current Protocols in Nucleic Acid Chemistry