Abstract
In the last decade, the field of oligonucleotide therapeutics has matured, with the regulatory approval of several single-stranded and double-stranded RNA drugs. In this Perspective, I discuss enabling developments and likely future directions in the field from the perspective of oligonucleotide chemistry.
INTRODUCTION
Most researchers in the pharmaceutical industry in the early 1990s believed that oligonucleotides stood little chance of success as a new class of drug. Textbook knowledge stated that large polyanionic structures do not enter cells (Gennis 1989) and thus, oligonucleotide drugs should not function in vivo. Furthermore, a manufacturing process for oligonucleotides seemed improbable, and therefore, there was no credible business case for such a class of therapeutics. In spite of this, today 13 oligonucleotide drugs have been approved by regulatory authorities (comprehensively reviewed in reference, Crooke et al. 2021). It is perhaps time to rewrite some chapters of the textbooks.
Today, oligonucleotide drugs are approved for use as medicines in the liver, the central nervous system (CNS), the skeletal muscle and the eye, and there are good reasons to believe that they will soon be validated in the lung, the kidney, and the bone marrow. Thus, oligonucleotides are now established as a major class of therapeutics, behind small-molecule drugs and therapeutic proteins.
At the annual Oligonucleotide Therapeutics Society meeting in Phoenix, I was asked by a young chemist where I thought that medicinal chemistry could play a role in the future of oligonucleotide drugs.
Medicinal chemistry milestones in oligonucleotide therapeutics
Looking back over 30 yr, a handful of milestones in the chemistry of oligonucleotides stand out. Oligonucleotide drugs are large, chemically synthesized structures, and therefore optimization of their pharmacodynamics (PD) and pharmacokinetics (PK) properties fell under the responsibility of medicinal chemists. Pioneering work was carried out by chemists through the 1980s and the 1990s, during which the ribonucleotide structure was systematically modified in efforts: (i) to protect single-stranded antisense oligonucleotides (ASOs) against metabolic degradation, while retaining their ability to hybridize with their targets and to recruit cellular effector enzymes; and (ii) to remain accessible via solid-phase synthesis. The experience gained in these areas streamlined efforts a decade later with a second emerging class of oligonucleotide drugs, the double-stranded small interfering RNAs (siRNAs) (Elbashir et al. 2001). In parallel with this work, major advances were made with oligonucleotide synthesizers, both in terms of synthesis throughput and synthesis scale. The introduction of 96-well machines, such as the Mermade 192, allowed researchers to synthesize oligonucleotides in “high-throughput.” This meant that instead of struggling to predict possible binding sites for potent oligonucleotides on a target mRNA with the help of RNA folding programs, or by assessing GC-content, it became routine in industry to synthesize and screen hundreds of reagents in a brute-force approach to identify experimentally and unambiguously the “best” oligonucleotide. In turn, access to large screening datasets powered the use of machine learning methods that revealed some of the sequence-dependent properties of potent oligonucleotides, as described in 2005 with siRNAs (Huesken et al. 2005). Meanwhile, at the opposite end of the synthesis spectrum, large capacity synthesizers were introduced, providing gram quantities of oligonucleotide reagents for routine testing in animal disease models, including nonhuman primates. Today, the OligoProcess synthesizer produces up to 15 kg of oligonucleotide in single batches. With these developments ongoing, the field had momentum.
Ribose modifications in single-stranded RNA drugs
The phosphodiesters of a native DNA or RNA oligonucleotide are quickly degraded by ubiquitous nucleases in vivo. Hence, medicinal chemists were tasked with modifying oligonucleotide structures to render them resistant to metabolism. However, researchers were alarmed to find that even minor modifications to the ribonucleotide unit of an ASO could severely reduce its affinity for a complementary RNA. Hence, over a period of two decades, hundreds of nucleoside modifications were designed, synthesized and tested in academia and industry, in search of the “perfect” modification (Wan and Seth 2016). The synthetic chemistry was resource-intensive, monotonous, and demanding. In most cases, it necessitated the synthesis of the four nucleosides as stable but reactive phosphoramidites (Fig. 1A), with protecting groups on the exocyclic amino groups of the nucleobases, and good solubility in acetonitrile solvent. These building blocks were subjected to solid phase synthesis, then harsh ammonia treatment, followed by purification and characterization. The resultant oligonucleotide was then evaluated for its binding affinity and selectivity toward a complementary RNA in in vitro assays. Not surprisingly, the rate of attrition was high and most of these modifications fell by the wayside; very few reached clinical evaluation and drug approval.
FIGURE 1.
Ribonucleoside modifications present in advanced oligonucleotide drug candidates. (A) Conventional cytidine phosphoramidite building block that is activated by tetrazole for coupling to the growing oligonucleotide on solid support; the reactive P (III) group and protecting groups in red. (B) Morpholino group. (C) Modifications on the 2′-O position of the ribose groups. (D) MOE group. (E) LNA and cEt groups. (F) Tricyclo DNA group. (G) 2′-F groups. (H) Phosphoryl guanidine (PN) group. (I) Mesyl phosphoramidite (MsPA) group. Modifications (C–E) are usually PS-modified in oligonucleotide drugs; 2′-F groups are either PS or PO-modified in siRNAs; groups (B–E,G–I) contain chiral internucleotide linkages. Blue illustrates the modification compared to native DNA and RNA structure.
Among the successful modifications, one of the most unusual was the phosphorodiamidate morpholino oligonucleotide (PMO) (Fig. 1B). Its elegant synthesis involves oxidative-mediated ring opening of the ribonucleoside, followed by ring closure with reductive amination, to produce a nucleobase-substituted morpholine cycle. The morpholines are linked by a phosphorodiamidate backbone (Iversen 2001). This chemistry was tested in the clinic with the splice-switching oligonucleotide eteplirsen. The target of eteplirsen is the pre-mRNA of dystrophin in skeletal muscle cells, to which it binds and alters splicing so as to exclude a deleterious exon. The approval of eteplirsen (2016) for the treatment of Duchenne muscular dystrophy (DMD) was controversial, due to the low level of correction that the drug reportedly achieves in the skeletal muscles of patients (Kesselheim and Avorn 2016). Nevertheless, its approval paved the way for three subsequent PMO drugs (golodirsen, vitolarsen; Aartsma-Rus and Corey 2020, casimersen) to address other disease-causing mutations in two other exons of dystrophin for DMD treatment (Crooke et al. 2021). The morpholino drugs were notable as one of the earliest demonstrations that an antisense drug could rescue a genetically derived, loss-of-function phenotype by altering the splicing of an mRNA.
Without doubt, the most successful means to modify DNA and RNA for therapeutic applications comprised two concomitant changes to the structure: exchange of the phosphodiester (PO) for the phosphorothioate (PS) group, as well as substitution of the ribose 2′-O-position (Fig. 1C; Manoharan 1999; Wan and Seth 2016). The pioneering work of F. Eckstein had shown that incorporation of PS linkages into the backbone of an oligonucleotide greatly improves its hydrophobicity and nuclease stability (Eckstein 2014). Fortunately, the PS group was easily adapted to solid-phase synthesis protocols and the modification was found—unexpectedly—to facilitate entry of PS oligonucleotides into cells (Loke et al. 1989; Beltinger et al. 1995; Stein et al. 2010; Miller et al. 2016). Furthermore, PS linkages in an ASO result in its weak binding to serum proteins, such as human albumin that retards its renal clearance (Gaus et al. 2019) and permits a wide distribution of a drug in vivo (Geary et al. 2015).
Substitution of the hydroxyl group at the 2′-position of the ribose was an obvious avenue of investigation for chemists (Manoharan 1999). A variety of different substituents were studied, ranging from small alkyl groups to alkyl chains containing aromatic, halogenated and amino groups. The most significant breakthrough came with the introduction of the 2′-O-methoxyethyl (MOE) group, described in a 1995 Helvetic publication by P. Martin (Fig. 1D; Martin 1995). The MOE group imposes a C3′-endo conformation on the riboses of an oligonucleotide, which enhances hybridization affinity and selectivity for target RNAs (Teplova et al. 1999). Furthermore, in combination with the PS linkage, an MOE substituent renders an oligonucleotide highly stable to endo- and exo-nucleases.
The MOE modification is today the most widely used chemical modification of single-stranded oligonucleotide drugs (for review, see Hall and Hill 2023). The modification was clinically validated with the approval of mipomersen, a 20-mer “gapmer” PS oligonucleotide bearing five MOE-modified riboses flanking a 10-mer DNA “window.” The DNA segment recruits RNase H1 to the target mRNA, thereby mediating its cleavage and terminating synthesis of the target protein (Monia et al. 1993). Mipomersen targets the liver as a treatment for familial hypercholesterolaemia (FH), a rare disorder of low-density lipoprotein cholesterol (LDL-C) metabolism (Raal et al. 2010). Despite mipomersen not being a commercial success, it generated spectacular data and was celebrated by the field as the first of the new-generation oligonucleotide drugs, able to suppress selectively the expression of a deleterious protein (Kastelein et al. 2006; Parham and Goldberg 2019).
The approval of mipomersen in the USA (2013) was quickly followed by that of nusinersen (2016), a breakthrough treatment for spinal muscular atrophy (SMA). Nusinersen is a fully PS-MOE-modified, 18-mer ASO that binds to SMN2 pre-mRNA and alters its splicing, to switch on production of a functional SMN protein (Hua et al. 2007; Corey 2017). It was the first oligonucleotide drug to work in the nervous system, confirming findings in the late 1990s that intrathecal delivery into the cerebral spinal fluid was a viable means to administer MOE oligonucleotides into the CNS (Barclay et al. 2002; Dogrul et al. 2002). Also, it is the only oligonucleotide to date to achieve “blockbuster drug” status.
A number of alternative ribose modifications for single-stranded RNA drugs are also worthy of mention. They include the structurally complex bicyclic “locked” nucleic acid (LNA, cEt) modifications (Koshkin et al. 1998; Seth et al. 2008) and tricyclic deoxyribose (TCA) derivatives (Renneberg and Leumann 2002) that endow oligonucleotides with very high RNA-binding affinities (Fig. 1E,F). However, for a variety of reasons, they have either fallen at (e.g., miravirsen; Janssen et al. 2013), or not yet cleared (e.g., danvatirsen; Hong et al. 2015), the last hurdles before regulatory approval. Intuitively, it seems likely that some of these structures will eventually achieve success in the clinic.
SiRNAs and oligonucleotide conjugates
The gapmer design of ASOs provided a workable solution for chemists aiming for a compromise between stability, affinity, and RNase H-compatibility. For siRNAs, the main difficulty with the PD properties was to achieve nuclease stability of the double-stranded RNA (dsRNA) in view of the sensitivity of the RNAi mechanism to structural modifications in the two strands (passenger and guide) (Bramsen et al. 2009). Furthermore, the mainstay substituents of antisense oligonucleotides, such as MOE, are poorly accepted by the RISC (RNA-induced silencing complex) machinery in many (but not all; Dorn et al. 2004) positions of the siRNA duplex. Eventually researchers from siRNA Therapeutics and Alnylam Pharmaceuticals converged on the replacement of all ribonucleotides in an siRNA with intricate arrangements of 2′-O-methyl (OMe) and 2′-fluoro (F) nucleotides (Fig. 1G; Foster et al. 2018). These fully modified siRNAs are then capped with a few terminal PS groups to top-up nuclease stability. This structural format was not effective in in vivo applications, since in contrast to single-stranded oligonucleotides, dsRNAs do not bind serum proteins and are quickly excreted from the body (Biscans et al. 2018). Furthermore, they do not undergo gymnosis—unaided uptake into cells—in contrast to their single-stranded counterparts (Dowdy 2017). This hurdle was countered by their formulation with multicomponent lipid nanoparticles (LNPs) (Bost et al. 2021), which were used for the first siRNA drug patisiran in the treatment of hereditary transthyretin-mediated amyloidosis (Adams et al. 2018). However, LNPs have mostly fallen out of favor for siRNA formulations, because of the complexity of their composition (Akinc et al. 2008; Jayaraman et al. 2012; Dowdy 2017) and their perceived potential for long term toxicity. Instead, the RNA field adopted a different strategy for siRNA delivery in vivo—the oligonucleotide conjugate.
The idea of conjugating functional groups to oligonucleotides to improve their PD and PK properties is decades old (for an excellent early review see, Manoharan 2002). A variety of innovative functional groups have been explored by chemists, ranging from intercalators (Asseline et al. 1984), peptides (Turner et al. 2005), stable metal complexes (Hall et al. 1996), and polyamines (Menzi et al. 2017). Today, for improved siRNA (and ASO) delivery, oligonucleotide conjugates can be grouped into those that improve systemic circulation, those that aid cellular uptake, or those that do both (Juliano 2016).
Hydrophobic groups such as cholesterol and other lipids were seen to alter the distribution of siRNAs from liver (Soutschek et al. 2004; Osborn et al. 2018), producing measurable target suppression in kidney, heart, lung, fat, and muscle (Biscans et al. 2018). Higher hydrophobicity of the conjugate group led to greater tissue retention of the siRNA, although higher siRNA accumulation in a tissue does not correlate with higher gene silencing in cells of the tissue, as reported by several groups (Halloy et al. 2020).
The conjugation of receptor-targeting ligands to ASOs and siRNAs has been championed by researchers at Ionis Therapeutics (Prakash et al. 2014) and Alnylam Pharmaceuticals (Nair et al. 2014). The conjugation of a targeting ligand composed of three N-acetylgalactosamine (GalNAc) moieties to the 3′-end of the siRNA passenger strand stands alone as a breakthrough for the RNAi field (Fig. 2A; Nair et al. 2014). GalNAc ligands show high affinity for asialoglycoprotein receptors (ASGPRs), expressed in hepatic cells. Upon ligand binding, ASGPRs undergo endocytosis, transporting the conjugated siRNA into cells (Spiess 1990). There, the conjugate is metabolically cleaved releasing its siRNA into the cytosol. The GalNAc group is so effective for hepatocyte delivery, that drug potency is improved by 10- to 30-fold, compared to nonconjugated ASOs (Crooke et al. 2019; Debacker et al. 2020). This conjugation strategy was clinically validated for siRNAs with the approval of givosiran targeting delta-aminolevulinic acid synthase 1 (ALAS1), for treating the rare metabolic disorder acute hepatic porphyria, as well as inclisiran, the first RNA drug to treat a common disease—atherosclerotic cardiovascular disease (Santulli et al. 2021).
FIGURE 2.

Oligonucleotide conjugates. (A) The GalNAc conjugate used for hepatic delivery of ASOs and siRNAs (Nair et al. 2014). (B) Mannose ligands used to deliver siRNAs to macrophages and dendritic cells via the CD-206 receptor (Uehara et al. 2022). (C) Peptide conjugate used to deliver Malat1-targeting gapmer oligonucleotides to pancreatic beta-cells (Knerr et al. 2021). (D) Neurotensin peptide used to deliver Malat1-targeting gapmer oligonucleotides to various regions of the brain (Nikan et al. 2020). Targeting ligands shown in blue; linker groups shown in pink.
The consequences of GalNAc-targeting for the oligonucleotide therapeutics field cannot be over-stated, and leading siRNA and antisense companies have stacked their clinical pipelines with GalNAc conjugates (Debacker et al. 2020). Researchers are now racing to identify the “next GalNAc” (Dowdy 2017). On paper, the approach appears daunting (Juliano 2016). For a given target cell type, one needs to identify a highly expressed surface receptor that is internalized upon ligand binding, for which a ligand is available and can be attached at an appropriate position of the oligonucleotide or the siRNA. In addition, the site of conjugation and the composition of the linker should not attenuate the ability of the ligand to interact with its receptor or prevent the receptor from internalizing. Three main classes of conjugate ligands have been investigated for targeting through specific receptors: carbohydrates, peptides, and antibodies.
Following the GalNAc example, a tetra-valent mannose ligand was conjugated to siRNAs for selective delivery to CD206-expressing macrophages and dendritic cells in vitro and in vivo (Fig. 2B; Uehara et al. 2022). Similar to the ASGPR, the CD-206 receptor is expressed selectively on the cell surface and undergoes fast recycling. Also, as for the GalNAc group, the multivalent ligand showed superior potency over a monovalent ligand, demanding the design and synthesis of a long, structurally complex linker group (Fig. 2B). In mice, these conjugates accumulated and elicited gene silencing in CD-206-expressing cells.
The most advanced example of receptor mediated targeting is that of the Glucagon-Like-Peptide-1 agonist (GLP-1), which was developed to target specifically pancreatic beta cells, where GLP1R expression is restricted (Ämmälä et al. 2018). Gapmer ASOs conjugated to the 37-amino acid peptide GLP-1 inhibited their targets in the pancreatic cells (Fig. 2C). Astonishingly, these ASOs are devoid of effects at low doses in liver, after systemic administration to ob/ob mice. An analogous approach is being pursued for targets in the brain, using ASOs or siRNA conjugated to a short 13-amino acid neurotensin peptide that binds with high affinity at the sortilin receptor (Fig. 2D; Nikan et al. 2020). The neuropeptide was conjugated to morpholino oligonucleotides. However, the reagents have exhibited relatively modest improvements in splice-modulating activity in the cortex and striatum of mice after intracerebroventricular injection.
New ways to use oligonucleotide conjugation as a means to improve drug trafficking are underway, as our understanding of how oligonucleotides trafficking in cells and in vivo increases (Juliano 2016). For example, ancillary groups that aid endosomal escape of the oligonucleotides in cells or that help traffic an oligonucleotide to the nucleus would be of potentially high value (Dowdy 2017). Such initiatives are supported by the development of new highly sensitive hybridization-based analytical techniques that can quantify oligonucleotides in individual protein complexes (Brunschweiger et al. 2016) or compartments of the cell (Godinho et al. 2017) or in distinct tissues/organs of the body (Boos et al. 2013).
Phosphorothioate linkages—the Dr. Jekyll and Mr. Hyde of oligonucleotide therapeutics
The PS-linkage is an indispensable part of many oligonucleotide drugs and is likely to remain so for the foreseeable future. In the early phases of the field, it powered advances in the technology, thanks to its favorable PK properties, its metabolic stability, its ease of synthesis and its compatibility with the RNase H mechanism. However, the PS-linkage is often maligned for its toxicity (Shen et al. 2019; Crooke et al. 2021), its metabolic instability in some sequence contexts and the hidden secrets of its isomeric composition. For some applications in vivo, efforts have been made to reduce the number of PS groups in an oligonucleotide, for example, by substituting selected linkages with stable PO-groups (Wahlestedt et al. 2000; Barclay et al. 2002), with alkyl phosphonates (Migawa et al. 2019), with phosphoryl guanidine (PN) groups (Fig. 1H; Kandasamy et al. 2022a,b) or with mesyl phosphoramidite (MsPA) groups (Fig. 1I; Miroshnichenko et al. 2019; Hammond et al. 2021). The PN and MsPA groups represent relatively new chemistries that are highly resistant to nucleases and are easily incorporated into the solid-phase synthesis cycle by substituting an azide synthon for sulfur during P (III) to P (V) conversion (Zhukov et al. 2021).
During conventional solid phase PS-oligonucleotide synthesis, the coupling of phosphoramidite building blocks (Fig. 1A) occurs with epimerization, mediated by nucleophilic tetrazole activators. Thus, PS stereochemistry is not controlled, and therefore each linkage in the oligonucleotide exists as an approximate 1:1 ratio of Rp and Sp diastereoisomers (Fig. 3A). Thus, the siRNA inclisiran with six PS groups comprises up to 64 (26) isomers, whereas the 20-mer ASO pelacarsen (Tsimikas et al. 2020) has 524,288 (219) possible diastereoisomers (Fig. 3B). Ravikumar and Cole studied the influence of various parameters on the Rp/Sp ratios produced during the coupling of conventional MOE phosphoramidites (Fig. 1A), including synthesis scale, solid supports, machines, reagent concentrations, tetrazole activators, and phosphodiester protecting groups. They concluded that activators and the phosphate protecting groups had the greatest influence during solid phase synthesis (Ravikumar and Cole 2003). These findings were consistent with later work by T. Wada on RNAs (Oka and Wada 2011) and on (si)RNAs by Jahns et al. (2015) who demonstrated that subtle changes in the Rp/Sp composition of PS RNAs in siRNAs significantly affects their properties in cells.
FIGURE 3.
Stereopure synthesis of PS-oligonucleotides. (A) The Rp and Sp diastereoisomers of a PS linkage. (B) Selected stereorandom and stereopure oligonucleotides tested in humans (5′–3′). (C) Phosphoramidite building block used to synthesize stereopure PS oligonucleotides (R is H, alkyl or silyl groups). (D) P (V) building blocks used to synthesize stereopure oligonucleotides. (-: Rp/Sp mixed PS linkage; -: PO linkage; v: Sp PS linkage; ^: Rp PS linkage; NM: 2′-OMe; NF: 2′-F; n: deoxyribonucleotide).
The pharmacological properties of an oligonucleotide are the sum activity of its component isomers. Each diastereoisomer exhibits its own distinct PD and PK properties (Jahns et al. 2015, 2021). In modern conventional drug development, the use of diastereosiomeric mixtures of drugs is avoided wherever possible. However, due to the strict requirement for quantitative coupling reactions during oligonucleotide synthesis, the field of oligonucleotide therapeutics has been exempt from this condition. Recent developments, however, suggest this aspect should be reexamined, since: (a) it is now possible (though challenging) to synthesize antisense PS-oligonucleotides stereospecifically (Iwamoto et al. 2017); (b) a loss of stereochemical reproducibility during manufacturing may have contributed to the failure of the first generation antisense drug mongersen (Arrico et al. 2022); and (c) innovative new P (III) (Fig. 3C; Li et al. 2017) and P (V) chemistry (Fig. 3D; Stec et al. 1991; Knouse et al. 2018) has stirred chemists to revisit methods of oligonucleotide synthesis.
The main premise of stereopure PS-oligonucleotides—besides the obvious benefits of working with a single molecular entity—is that one may be able to influence (improve) distinct PD and PK properties via control of PS stereochemistry, if methods are available to test/synthesize all possible diastereoisomers. For example, it has been demonstrated that some PS diastereoisomers in a stereorandom population of a PS gapmer show exaggerated toxicity, from the chiral interaction of selected PS groups with proteins (Hyjek-Składanowska et al. 2020). This toxicity was attenuated by a switch in the stereochemistry at specific PS centers. Moreover, there is the tantalizing prospect that through interactions with certain proteins, a distinct PS stereochemistry may for example, improve potency, aid target cell uptake or mediate allele-specific targeting etc. Indeed, it is long-known that a DNA segment composed of Rp centers in a PS gapmer oligonucleotide is RNase H-compatible, but is quickly degraded by nucleases; whereas the Sp diastereoisomers have better stability but show poor RNase H-compatibility (Koziolkiewicz et al. 1995).
The major breakthrough in the chemistry of stereopure PS-oligonucleotides was the introduction by the Wada group of new P (III) nucleoside building blocks and activators that enable stereospecific coupling on solid phase (Oka and Wada 2011; Nukaga et al. 2012). Initially, coupling yields with this chemistry were not sufficient to produce 20-mer oligonucleotides. However, by tinkering with the substituents on the chiral ancillary and the reaction conditions, chemists from Wave Life Sciences prevailed with the first chemical synthesis of “full-length” stereopure PS oligonucleotides (Iwamoto et al. 2017).
A second outcome of this seminal work was the discovery that a trivalent stereo-motif 3′-SpSpRp-5′ in the DNA segment of a stereopure PS gapmer provides both nuclease stability and RNase H compatibility (Iwamoto et al. 2017), circumventing the longstanding challenge (Wan et al. 2014) of how to exploit stereopurity in the DNA window of a gapmer. The authors demonstrated that the motif functions in gapmer oligonucleotides with different chemistries in the wings, although its benefit was not observed in some oligonucleotides with stereorandom PS-wings (Østergaard et al. 2020).
To date, the properties and applications of fully stereopure PS oligonucleotides have been described in a handful of prominent papers (Iwamoto et al. 2017; Li et al. 2017; Byrne et al. 2021; Liu et al. 2021; Kandasamy et al. 2022a). Critical analysis of the data in these works confirms that a stereodefined PS-backbone shows superior potency and duration of action to its stereorandom counterpart, in vitro and in vivo. Nevertheless, it cannot be forgotten that many factors other than potency play roles on the road to regulatory approval.
Pleasingly, the phosphoryl guanidine and mesyl phosphoramidate groups (Fig. 1H,I) can also be combined with Wada phosphoramidites to yield stereopure amidate linkages (Anderson et al. 2021; Kandasamy et al. 2022a), thereby reducing the PS content of an oligonucleotide without compromising either metabolic stability or stereopurity. The uncharged stereopure PN modification was incorporated into the wings of gapmer oligonucleotides, as well as splice switching oligonucleotides, that showed superior activity to their PS counterparts in the CNS (Kandasamy et al. 2022a). The authors suggested that these enhanced effects occurred through improved oligonucleotide delivery. From the first data with this chemistry, it seems likely that the stereopure PN modification has a bright future in the field.
Stereopure PS-gapmer oligonucleotides were “validated” in clinical trials of suvodirsen (a splice-switching oligonucleotide comprising 2′-OMe and 2′-F ribose modifications to treat DMD), as well as rovanersen and lexanersen (for Huntington`s disease). However, all three front-runner drugs failed to progress in these challenging disease indications, possibly for reasons of insufficient target exposure (DMD) or mechanism-related toxicity (Huntington's disease). However, the next wave of stereopure PS-oligonucleotides is already in clinical trials and therefore it seems likely that the approval of the first such drug is only a matter of time.
OUTLOOK
The examples of medicinal chemistry discussed in this Perspective—chemical modifications, oligonucleotide conjugates, PS stereochemistry—were selected to highlight three areas of future challenges for medicinal chemists in the oligonucleotide therapeutics field.
Arguably, the need for new ribonucleoside modifications in the field has receded in recent times. This is due to the ready accessibility of MOE and LNA chemistries. When combined with “routine” high-throughput synthesis/screening methods, potent oligonucleotides can be produced against any target for which the sequence is known, as originally envisioned by Zamecnik and Stephenson (Stephenson and Zamecnik 1978; Zamecnik and Stephenson 1978). Furthermore, barriers of intellectual property related to these ribose chemistries have mostly ebbed away, leaving freedom to operate in the field. Reassured by the success of nusinersen that the technology can deliver, many large pharma companies have initiated oligonucleotide (antisense or siRNA) programs. For example, dozens of MOE-oligonucleotides are at various stages of clinical testing (Crooke et al. 2021), sponsored by a variety of companies. Many of these clinical candidates are intended for use in rare diseases, where targets are clinically validated and competition with conventional drug classes is sparse. However, a growing number of programs are directed to the treatment of common diseases, with large patient populations. If only a small fraction of these new programs is clinically successful, it will likely create a strain on contract research organizations for oligonucleotide manufacture. On the other hand, it will also motivate chemists to seek out new methods of oligonucleotide synthesis that are better scalable and “greener” than current methods. Such initiatives may range from the development of new solid supports with higher loadings (similar to peptide solid supports), through solution-phase synthesis (Zhou et al. 2022) to even enzymatic synthesis (Freund et al. 2022).
Recently, many research groups have turned to the area of oligonucleotide conjugates, for enhanced oligonucleotide delivery. Juliano (2016) has described two parts to the delivery problem: first, how to transport the oligonucleotide to the target organ of interest, and then, how to deliver it into the right cellular compartments. Oligonucleotide conjugates offer excellent possibilities to address both objectives, possibly with dedicated conjugate groups for each. However, current oligonucleotide conjugates have high structural complexity for chemical synthesis (see structures drawn in full in Fig. 2). This complicates their development in the areas of synthesis/manufacture, companion analytics, as well as their metabolism and toxicity. These factors can be underestimated by chemists engaged in exploratory research. However, process chemists responsible for preclinical and clinical development of the drugs are sensitive to their large structures, where the conjugated group represents a significant part of the overall structure. Indeed, the manufacturing of inclisiran (Fig. 2A), containing the tri-antennary GalNAc ligand is a formidable achievement. One means to simplify these structures would be to replace carbohydrate- and peptide-targeting ligands with small-molecule ligands that are equally capable of binding selectively and potently to internalizing cell surface receptors. A few reports describe targeting with small-molecule ligands, for example, anisamide (Nakagawa et al. 2010) or anandamide (Willibald et al. 2012), but as yet this appears to be a largely unexplored area.
Over the years, oligonucleotide chemists have reveled in the “which is the best” arguments: Is an LNA superior to an MOE modification? Is the siRNA better than an ASO? Is a lipid nanoparticle formulation better than a conjugate? This banter extends to the merits of stereopure PS-oligonucleotides (Hyjek-Składanowska et al. 2020; Østergaard et al. 2020), and discussions between those with opposing views will continue, at least until the approval of the first stereopure PS drug settles the question. Based on emerging work, it appears that PS stereochemistry has much to offer in terms of improving the PK and PD properties of oligonucleotides. The challenge here is to design experiments that can link a particular fingerprint of PS stereochemistry to the desired property of interest.
In conclusion, young chemists rest assured: there is still a need for innovation in the oligonucleotide therapeutics field.
ACKNOWLEDGMENTS
I thank Tobias Haab for help with the structures, Januscz Laski and Artur Laski for useful suggestions and Alyssa Hill for input on the manuscript. This work was supported by the NCCR RNA and Disease and the Swiss National Science Foundation (number: 182880). J.H. is an inventor on a patent related to the structure in Figure 3C. We apologize to those many authors that have contributed important works to areas discussed in this article that we did not cite for reasons of space.
Footnotes
Article is online at http://www.rnajournal.org/cgi/doi/10.1261/rna.079511.122.
Freely available online through the RNA Open Access option.
REFERENCES
- Aartsma-Rus A, Corey DR. 2020. The 10th oligonucleotide therapy approved: golodirsen for Duchenne muscular dystrophy. Nucleic Acid Ther 30: 67–70. 10.1089/nat.2020.0845 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Adams D, Gonzalez-Duarte A, O'Riordan WD, Yang CC, Ueda M, Kristen AV, Tournev I, Schmidt HH, Coelho T, Berk JL, et al. 2018. Patisiran, an RNAi therapeutic, for hereditary transthyretin amyloidosis. N Engl J Med 379: 11–21. 10.1056/NEJMoa1716153 [DOI] [PubMed] [Google Scholar]
- Akinc A, Zumbuehl A, Goldberg M, Leshchiner ES, Busini V, Hossain N, Bacallado SA, Nguyen DN, Fuller J, Alvarez R, et al. 2008. A combinatorial library of lipid-like materials for delivery of RNAi therapeutics. Nat Biotechnol 26: 561–569. 10.1038/nbt1402 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ämmälä C, Drury WJ, Knerr L, Ahlstedt I, Stillemark-Billton P, Wennberg-Huldt C, Andersson EM, Valeur E, Jansson-Löfmark R, Janzén D, et al. 2018. Targeted delivery of antisense oligonucleotides to pancreatic β-cells. Sci Adv 4: eaat3386. 10.1126/sciadv.aat3386 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Anderson BA, Freestone GC, Low A, De-Hoyos CL, William JD, Østergaard ME, Migawa MT, Fazio M, Wan WB, Berdeja A, et al. 2021. Towards next generation antisense oligonucleotides: mesylphosphoramidate modification improves therapeutic index and duration of effect of gapmer antisense oligonucleotides. Nucleic Acids Res 49: 9026–9041. 10.1093/nar/gkab718 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arrico L, Stolfi C, Marafini I, Monteleone G, Demartis S, Bellinvia S, Viti F, McNulty M, Cabani I, Falezza A, et al. 2022. Inhomogeneous diastereomeric composition of Mongersen antisense phosphorothioate oligonucleotide preparations and related pharmacological activity impairment. Nucleic Acid Ther 32: 312–320. 10.1089/nat.2021.0089 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Asseline U, Toulme F, Thuong NT, Delarue M, Montenay-Garestier T, Hélène C. 1984. Oligodeoxynucleotides covalently linked to intercalating dyes as base sequence-specific ligands. Influence of dye attachment site. EMBO J 3: 795–800. 10.1002/j.1460-2075.1984.tb01887.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barclay J, Patel S, Dorn G, Wotherspoon G, Moffatt S, Eunson L, Abdel'al S, Natt F, Hall J, Winter J, et al. 2002. Functional downregulation of P2X3 receptor subunit in rat sensory neurons reveals a significant role in chronic neuropathic and inflammatory pain. J Neurosci 22: 8139–8147. 10.1523/JNEUROSCI.22-18-08139.2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beltinger C, Saragovi HU, Smith RM, LeSauteur L, Shah N, DeDionisio L, Christensen L, Raible A, Jarett L, Gewirtz AM. 1995. Binding, uptake, and intracellular trafficking of phosphorothioate-modified oligodeoxynucleotides. J Clin Invest 95: 1814–1823. 10.1172/JCI117860 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Biscans A, Coles A, Haraszti R, Echeverria D, Hassler M, Osborn M, Khvorova A. 2018. Diverse lipid conjugates for functional extra-hepatic siRNA delivery in vivo. Nucleic Acids Res 47: 1082–1096. 10.1093/nar/gky1239 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boos JA, Kirk DW, Piccolotto M-L, Zuercher W, Gfeller S, Neuner P, Dattler A, Wishart WL, Von Arx F, Beverly M, et al. 2013. Whole-body scanning PCR; a highly sensitive method to study the biodistribution of mRNAs, noncoding RNAs and therapeutic oligonucleotides. Nucleic Acids Res 41: e145. 10.1093/nar/gkt515 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bost JP, Barriga H, Holme MN, Gallud A, Maugeri M, Gupta D, Lehto T, Valadi H, Esbjorner EK, Stevens MM, et al. 2021. Delivery of oligonucleotide therapeutics: chemical modifications, lipid nanoparticles, and extracellular vesicles. ACS Nano 15: 13993–14021. 10.1021/acsnano.1c05099 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bramsen JB, Laursen MB, Nielsen AF, Hansen TB, Bus C, Langkjær N, Babu BR, Højland T, Abramov M, Van Aerschot A, et al. 2009. A large-scale chemical modification screen identifies design rules to generate siRNAs with high activity, high stability and low toxicity. Nucleic Acids Res 37: 2867–2881. 10.1093/nar/gkp106 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brunschweiger A, Gebert LFR, Lucic M, Pradère U, Jahns H, Berk C, Hunziker J, Hall J. 2016. Site-specific conjugation of drug-like fragments to an antimiR scaffold as a strategy to target miRNAs inside RISC. Chem Commun 52: 156–159. 10.1039/C5CC07478A [DOI] [PubMed] [Google Scholar]
- Byrne M, Vathipadiekal V, Apponi L, Iwamoto N, Kandasamy P, Longo K, Liu F, Looby R, Norwood L, Shah A, et al. 2021. Stereochemistry enhances potency, efficacy, and durability of Malat1 antisense oligonucleotides in vitro and in vivo in multiple species. Transl Vis Sci Technol 10: 23. 10.1167/tvst.10.1.23 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Corey DR. 2017. Nusinersen, an antisense oligonucleotide drug for spinal muscular atrophy. Nat Neurosci 20: 497. 10.1038/nn.4508 [DOI] [PubMed] [Google Scholar]
- Crooke ST, Baker BF, Xia S, Yu RZ, Viney NJ, Wang Y, Tsimikas S, Geary RS. 2019. Integrated assessment of the clinical performance of GalNAc3-conjugated 2′-O-methoxyethyl chimeric antisense oligonucleotides: I. Human volunteer experience. Nucleic Acid Ther 29: 16–32. 10.1089/nat.2018.0753 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Crooke ST, Baker BF, Crooke RM, Liang X. 2021. Antisense technology: an overview and prospectus. Nat Rev Drug Discov 20: 427–453. 10.1038/s41573-021-00162-z [DOI] [PubMed] [Google Scholar]
- Debacker AJ, Voutila J, Catley M, Blakey D, Habib N. 2020. Delivery of oligonucleotides to the liver with GalNAc: from research to registered therapeutic drug. Mol Ther 28: 1759–1771. 10.1016/j.ymthe.2020.06.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dogrul A, Gardell LR, Ma S, Ossipov MH, Porreca F, Lai J. 2002. ‘Knock-down’ of spinal CB1 receptors produces abnormal pain and elevates spinal dynorphin content in mice. Pain 100: 203–209. 10.1016/S0304-3959(02)00302-0 [DOI] [PubMed] [Google Scholar]
- Dorn G, Patel S, Wotherspoon G, Hemmings-Mieszczak M, Barclay J, Natt FJ, Martin P, Bevan S, Fox A, Ganju P, et al. 2004. siRNA relieves chronic neuropathic pain. Nucleic Acids Res 32: e49. 10.1093/nar/gnh044 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dowdy SF. 2017. Overcoming cellular barriers for RNA therapeutics. Nat Biotechnol 35: 222–229. 10.1038/nbt.3802 [DOI] [PubMed] [Google Scholar]
- Eckstein F. 2014. Phosphorothioates, essential components of therapeutic oligonucleotides. Nucleic Acid Ther 24: 374–387. 10.1089/nat.2014.0506 [DOI] [PubMed] [Google Scholar]
- Elbashir SM, Harborth J, Lendeckel W, Yalcin A, Weber K, Tuschl T. 2001. Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature 411: 494–498. 10.1038/35078107 [DOI] [PubMed] [Google Scholar]
- Foster DJ, Brown CR, Shaikh S, Trapp C, Schlegel MK, Qian K, Sehgal A, Rajeev KG, Jadhav V, Manoharan M, et al. 2018. Advanced siRNA designs further improve in vivo performance of GalNAc-siRNA conjugates. Mol Ther 26: 708–717. 10.1016/j.ymthe.2017.12.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Freund N, Taylor AI, Arangundy-Franklin S, Subramanian N, Peak-Chew S-Y, Whitaker AM, Freudenthal BD, Abramov M, Herdewijn P, Holliger P. 2022. A two-residue nascent-strand steric gate controls synthesis of 2′-O-methyl- and 2′-O-(2-methoxyethyl)-RNA. Nat Chem 15: 91–100. 10.1038/s41557-022-01050-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gaus HJ, Gupta R, Chappell AE, Østergaard ME, Swayze EE, Seth PP. 2019. Characterization of the interactions of chemically-modified therapeutic nucleic acids with plasma proteins using a fluorescence polarization assay. Nucleic Acids Res 47: 1110–1122. 10.1093/nar/gky1260 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Geary RS, Norris D, Yu R, Bennett CF. 2015. Pharmacokinetics, biodistribution and cell uptake of antisense oligonucleotides. Adv Drug Deliv Rev 87: 46–51. 10.1016/j.addr.2015.01.008 [DOI] [PubMed] [Google Scholar]
- Gennis RB. 1989. Biomembranes. Springer Verlag, NY. [Google Scholar]
- Godinho BM, Gilbert JW, Haraszti RA, Coles AH, Biscans A, Roux L, Nikan M, Echeverria D, Hassler M, Khvorova A. 2017. . Pharmacokinetic profiling of conjugated therapeutic oligonucleotides: a high-throughput method based upon serial blood microsampling coupled to peptide nucleic acid hybridization assay. Nucleic Acid Ther 27: 323–334. 10.1089/nat.2017.0690 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hall J, Hill AC. 2023. The MOE modification of RNA: origins and widescale impact on the oligonucleotide therapeutics field. Helv Chim Acta 10.1002/hlca.202200169 [DOI] [Google Scholar]
- Hall J, Hüsken D, Häner R. 1996. Towards artificial ribonucleases: the sequence-specific cleavage of RNA in a duplex. Nucleic Acids Res 24: 3522–3526. 10.1093/nar/24.18.3522 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Halloy F, Iyer PS, Ćwiek P, Ghidini A, Barman-Aksözen J, Wildner-Verhey van Wijk N, Theocharides APA, Minder EI, Schneider-Yin X, Schümperli D, et al. 2020. Delivery of oligonucleotides to bone marrow to modulate ferrochelatase splicing in a mouse model of erythropoietic protoporphyria. Nucleic Acids Res 48: 4658–4671. 10.1093/nar/gkaa229 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hammond SM, Sergeeva OV, Melnikov PA, Goli L, Stoodley J, Zatsepin TS, Stetsenko DA, Wood MJA. 2021. Mesyl phosphoramidate oligonucleotides as potential splice-switching agents: impact of backbone structure on activity and intracellular localization. Nucleic Acid Ther 31: 190–200. 10.1089/nat.2020.0860 [DOI] [PubMed] [Google Scholar]
- Hong D, Kurzrock R, Kim Y, Woessner R, Younes A, Nemunaitis J, Fowler N, Zhou T, Schmidt J, Jo M, et al. 2015. AZD9150, a next-generation antisense oligonucleotide inhibitor of STAT3 with early evidence of clinical activity in lymphoma and lung cancer. Sci Transl Med 7: 314ra185. 10.1126/scitranslmed.aac5272 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hua Y, Vickers TA, Baker BF, Bennett CF, Krainer AR. 2007. Enhancement of SMN2 exon 7 inclusion by antisense oligonucleotides targeting the exon. PLoS Biol 5: e73. 10.1371/journal.pbio.0050073 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huesken D, Lange J, Mickanin C, Weiler J, Asselbergs F, Warner J, Meloon B, Engel S, Rosenberg A, Cohen D, et al. 2005. Design of a genome-wide siRNA library using an artificial neural network. Nat Biotechnol 23: 995–1001. 10.1038/nbt1118 [DOI] [PubMed] [Google Scholar]
- Hyjek-Składanowska M, Vickers TA, Napiórkowska A, Anderson BA, Tanowitz M, Crooke ST, Liang XH, Seth PP, Nowotny M. 2020. Origins of the increased affinity of phosphorothioate-modified therapeutic nucleic acids for proteins. J Am Chem Soc 142: 7456–7468. 10.1021/jacs.9b13524 [DOI] [PubMed] [Google Scholar]
- Iversen PL. 2001. Phosphorodiamidate morpholino oligomers: favorable properties for sequence-specific gene inactivation. Curr Opin Mol Ther 3: 235–238. [PubMed] [Google Scholar]
- Iwamoto N, Butler DCD, Svrzikapa N, Mohapatra S, Zlatev I, Sah DWY, Meena, Standley SM, Lu G, Apponi LH, et al. 2017. Control of phosphorothioate stereochemistry substantially increases the efficacy of antisense oligonucleotides. Nat Biotechnol 35: 845–851. 10.1038/nbt.3948 [DOI] [PubMed] [Google Scholar]
- Jahns H, Roos M, Imig J, Baumann F, Wang Y, Gilmour R, Hall J. 2015. Stereochemical bias introduced during RNA synthesis modulates the activity of phosphorothioate siRNAs. Nat Commun 6: 6317. 10.1038/ncomms7317 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jahns H, Taneja N, Willoughby JLS, Akabane-Nakata M, Brown CR, Nguyen T, Bisbe A, Matsuda S, Hettinger M, Manoharan RM, et al. 2021. Chirality matters: stereo-defined phosphorothioate linkages at the termini of small interfering RNAs improve pharmacology in vivo. Nucleic Acids Res 50: 1221–1240. 10.1093/nar/gkab544 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Janssen HLA, Reesink HW, Lawitz EJ, Zeuzem S, Rodriguez-Torres M, Patel K, van der Meer AJ, Patick AK, Chen A, Zhou Y, et al. 2013. Treatment of HCV infection by targeting microRNA. N Engl J Med 368: 1685–1694. 10.1056/NEJMoa1209026 [DOI] [PubMed] [Google Scholar]
- Jayaraman M, Ansell SM, Mui BL, Tam YK, Chen J, Du X, Butler D, Eltepu L, Matsuda S, Narayanannair JK, et al. 2012. Maximizing the potency of siRNA lipid nanoparticles for hepatic gene silencing in vivo. Angew Chem Int Ed Engl 51: 8529–8533. 10.1002/anie.201203263 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Juliano RL. 2016. The delivery of therapeutic oligonucleotides. Nucleic Acids Res 44: 6518–6548. 10.1093/nar/gkw236 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kandasamy P, Liu Y, Aduda V, Akare S, Alam R, Andreucci A, Boulay D, Bowman K, Byrne M, Cannon M, et al. 2022a. Impact of guanidine-containing backbone linkages on stereopure antisense oligonucleotides in the CNS. Nucleic Acids Res 50: 5401–5423. 10.1093/nar/gkac037 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kandasamy P, McClorey G, Shimizu M, Kothari N, Alam R, Iwamoto N, Kumarasamy J, Bommineni GR, Bezigian A, Chivatakarn O, et al. 2022b. Control of backbone chemistry and chirality boost oligonucleotide splice switching activity. Nucleic Acids Res 50: 5443–5466. 10.1093/nar/gkac018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kastelein JJ, Wedel MK, Baker BF, Su J, Bradley JD, Yu RZ, Chuang E, Graham MJ, Crooke RM. 2006. Potent reduction of apolipoprotein B and low-density lipoprotein cholesterol by short-term administration of an antisense inhibitor of apolipoprotein B. Circulation 114: 1729–1735. 10.1161/CIRCULATIONAHA.105.606442 [DOI] [PubMed] [Google Scholar]
- Kesselheim AS, Avorn J. 2016. Approving a problematic muscular dystrophy drug. JAMA 316: 2357–2358. 10.1001/jama.2016.16437 [DOI] [PubMed] [Google Scholar]
- Knerr L, Prakash TP, Lee R, Drury WJ III, Nikan M, Fu W, Pirie E, Maria L, Valeur E, Hayen A, et al. 2021. Glucagon like peptide 1 receptor agonists for targeted delivery of antisense oligonucleotides to pancreatic beta cell. J Am Chem Soc 143: 3416–3429. 10.1021/jacs.0c12043 [DOI] [PubMed] [Google Scholar]
- Knouse KW, deGruyter JN, Schmidt MA, Zheng B, Vantourout JC, Kingston C, Mercer SE, McDonald IM, Olson RE, Zhu Y, et al. 2018. Unlocking P(V): reagents for chiral phosphorothioate synthesis. Science 361: 1234. 10.1126/science.aau3369 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koshkin AA, Singh SK, Nielsen P, Rajwanshi VK, Kumar R, Meldgaard M, Olsen CE, Wengel J. 1998. LNA (Locked Nucleic Acids): synthesis of the adenine, cytosine, guanine, 5-methylcytosine, thymine and uracil bicyclonucleoside monomers, oligomerisation, and unprecedented nucleic acid recognition. Tetrahedron 54: 3607–3630. 10.1016/S0040-4020(98)00094-5 [DOI] [Google Scholar]
- Koziolkiewicz M, Krakowiak A, Kwinkowski M, Boczkowska M, Stec WJ. 1995. Stereodifferentiation–the effect of P chirality of oligo (nucleoside phosphorothioates) on the activity of bacterial RNase H. Nucleic Acids Res 23: 5000–5005. 10.1093/nar/23.24.5000 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li M, Lightfoot HL, Halloy F, Malinowska AL, Berk C, Behera A, Schümperli D, Hall J. 2017. Synthesis and cellular activity of stereochemically-pure 2′-O-(2-methoxyethyl)-phosphorothioate oligonucleotides. Chem Commun 53: 541–544. 10.1039/C6CC08473G [DOI] [PubMed] [Google Scholar]
- Liu Y, Dodart JC, Tran H, Berkovitch S, Braun M, Byrne M, Durbin AF, Hu XS, Iwamoto N, Jang HG, et al. 2021. Variant-selective stereopure oligonucleotides protect against pathologies associated with C9orf72-repeat expansion in preclinical models. Nat Commun 12: 847. 10.1038/s41467-021-21112-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Loke SL, Stein CA, Zhang XH, Mori K, Nakanishi M, Subasinghe C, Cohen JS, Neckers LM. 1989. Characterization of oligonucleotide transport into living cells. Proc Natl Acad Sci 86: 3474–3478. 10.1073/pnas.86.10.3474 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Manoharan M. 1999. 2′-carbohydrate modifications in antisense oligonucleotide therapy: importance of conformation, configuration and conjugation. Biochim Biophys Acta 1489: 117–130. 10.1016/S0167-4781(99)00138-4 [DOI] [PubMed] [Google Scholar]
- Manoharan M. 2002. Oligonucleotide conjugates as potential antisense drugs with improved uptake, biodistribution, targeted delivery, and mechanism of action. Antisense Nucleic Acid Drug Dev 12: 103–128. 10.1089/108729002760070849 [DOI] [PubMed] [Google Scholar]
- Martin P. 1995. Ein neuer Zugang zu 2′-O-Alkylribonucleosiden und Eigenschaften deren Oligonucleotide. Helv Chim Acta 78: 486–504. 10.1002/hlca.19950780219 [DOI] [Google Scholar]
- Menzi M, Wild B, Pradère U, Malinowska AL, Brunschweiger A, Lightfoot HL, Hall J. 2017. Towards improved oligonucleotide therapeutics through faster target binding kinetics. Chemistry 23: 14221–14230. 10.1002/chem.201701670 [DOI] [PubMed] [Google Scholar]
- Migawa MT, Shen W, Wan WB, Vasquez G, Oestergaard ME, Low A, De Hoyos CL, Gupta R, Murray S, Tanowitz M, et al. 2019. Site-specific replacement of phosphorothioate with alkyl phosphonate linkages enhances the therapeutic profile of gapmer ASOs by modulating interactions with cellular proteins. Nucleic Acids Res 47: 5465–5479. 10.1093/nar/gkz247 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miller CM, Donner AJ, Blank EE, Egger AW, Kellar BM, Østergaard ME, Seth PP, Harris EN. 2016. Stabilin-1 and Stabilin-2 are specific receptors for the cellular internalization of phosphorothioate-modified antisense oligonucleotides (ASOs) in the liver. Nucleic Acids Res 44: 2782–2794. 10.1093/nar/gkw112 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miroshnichenko SK, Patutina OA, Burakova EA, Chelobanov BP, Fokina AA, Vlassov VV, Altman S, Zenkova MA, Stetsenko DA. 2019. Mesyl phosphoramidate antisense oligonucleotides as an alternative to phosphorothioates with improved biochemical and biological properties. Proc Natl Acad Sci 116: 1229–1234. 10.1073/pnas.1813376116 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Monia BP, Lesnik EA, Gonzalez C, Lima WF, McGee D, Guinosso CJ, Kawasaki AM, Cook PD, Freier SM. 1993. Evaluation of 2′-modified oligonucleotides containing 2′-deoxy gaps as antisense inhibitors of gene expression. J Biol Chem 268: 14514–14522. 10.1016/S0021-9258(19)85268-7 [DOI] [PubMed] [Google Scholar]
- Nair JK, Willoughby JLS, Chan A, Charisse K, Alam MR, Wang Q, Hoekstra M, Kandasamy P, Kelin AV, Milstein S, et al. 2014. Multivalent N-acetylgalactosamine-conjugated siRNA localizes in hepatocytes and elicits robust RNAi-mediated gene silencing. J Am Chem Soc 136: 16958–16961. 10.1021/ja505986a [DOI] [PubMed] [Google Scholar]
- Nakagawa O, Ming X, Huang L, Juliano RL. 2010. Targeted intracellular delivery of antisense oligonucleotides via conjugation with small-molecule ligands. J Am Chem Soc 132: 8848–8849. 10.1021/ja102635c [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nikan M, Tanowitz M, Dwyer CA, Jackson M, Gaus HJ, Swayze EE, Rigo F, Seth PP, Prakash TP. 2020. Targeted delivery of antisense oligonucleotides using neurotensin peptides. J Med Chem 63: 8471–8484. 10.1021/acs.jmedchem.0c00840 [DOI] [PubMed] [Google Scholar]
- Nukaga Y, Yamada K, Ogata T, Oka N, Wada T. 2012. Stereocontrolled solid-phase synthesis of phosphorothioate oligoribonucleotides using 2′-O-(2-cyanoethoxymethyl)-nucleoside 3'-O-oxazaphospholidine monomers. J Org Chem 77: 7913–7922. 10.1021/jo301052v [DOI] [PubMed] [Google Scholar]
- Oka N, Wada T. 2011. Stereocontrolled synthesis of oligonucleotide analogs containing chiral internucleotidic phosphorus atoms. Chem Soc Rev 40: 5829–5843. 10.1039/c1cs15102a [DOI] [PubMed] [Google Scholar]
- Osborn MF, Coles AH, Biscans A, Haraszti RA, Roux L, Davis S, Ly S, Echeverria D, Hassler MR, Godinho BMDC, et al. 2018. Hydrophobicity drives the systemic distribution of lipid-conjugated siRNAs via lipid transport pathways. Nucleic Acids Res 47: 1070–1081. 10.1093/nar/gky1232 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Østergaard ME, De Hoyos CL, Wan WB, Shen W, Low A, Berdeja A, Vasquez G, Murray S, Migawa MT, Liang XH, et al. 2020. Understanding the effect of controlling phosphorothioate chirality in the DNA gap on the potency and safety of gapmer antisense oligonucleotides. Nucleic Acids Res 48: 1691–1700. 10.1093/nar/gkaa031 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Parham JS, Goldberg AC. 2019. Mipomersen and its use in familial hypercholesterolemia. Expert Opin Pharmacother 20: 127–131. 10.1080/14656566.2018.1550071 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Prakash TP, Graham MJ, Yu J, Carty R, Low A, Chappell A, Schmidt K, Zhao C, Aghajan M, Murray HF, et al. 2014. Targeted delivery of antisense oligonucleotides to hepatocytes using triantennary N-acetyl galactosamine improves potency 10-fold in mice. Nucleic Acids Res 42: 8796–8807. 10.1093/nar/gku531 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Raal FJ, Santos RD, Blom DJ, Marais AD, Charng M-J, Cromwell WC, Lachmann RH, Gaudet D, Tan JL, Chasan-Taber S, et al. 2010. Mipomersen, an apolipoprotein B synthesis inhibitor, for lowering of LDL cholesterol concentrations in patients with homozygous familial hypercholesterolaemia: a randomised, double-blind, placebo-controlled trial. Lancet 375: 998–1006. 10.1016/S0140-6736(10)60284-X [DOI] [PubMed] [Google Scholar]
- Ravikumar VT, Cole DL. 2003. Diastereomeric process control in the synthesis of 2′-O-(2-methoxyethyl) oligoribonucleotide phosphorothioates as antisense drugs. Nucleosides Nucleotides Nucleic Acids 22: 1639–1645. 10.1081/NCN-120023089 [DOI] [PubMed] [Google Scholar]
- Renneberg D, Leumann CJ. 2002. Watson−Crick base-pairing properties of tricyclo-DNA. J Am Chem Soc 124: 5993–6002. 10.1021/ja025569+ [DOI] [PubMed] [Google Scholar]
- Santulli G, Jankauskas SS, Gambardella J. 2021. Inclisiran: a new milestone on the PCSK9 road to tackle cardiovascular risk. Eur Heart J Cardiovasc Pharmacother 7: e11–e12. 10.1093/ehjcvp/pvab014 [DOI] [PubMed] [Google Scholar]
- Seth PP, Siwkowski A, Allerson CR, Vasquez G, Lee S, Prakash TP, Kinberger G, Migawa MT, Gaus H, Bhat B, et al. 2008. Design, synthesis and evaluation of constrained methoxyethyl (cMOE) and constrained ethyl (cEt) nucleoside analogs. Nucleic Acids Symp Ser (Oxf) 52: 553–554. 10.1093/nass/nrn280 [DOI] [PubMed] [Google Scholar]
- Shen W, De Hoyos CL, Migawa MT, Vickers TA, Sun H, Low A, Bell TA, Rahdar M, Mukhopadhyay S, Hart CE, et al. 2019. Chemical modification of PS-ASO therapeutics reduces cellular protein-binding and improves the therapeutic index. Nat Biotechnol 37: 640–650. 10.1038/s41587-019-0106-2 [DOI] [PubMed] [Google Scholar]
- Soutschek J, Akinc A, Bramlage B, Charisse K, Constien R, Donoghue M, Elbashir S, Geick A, Hadwiger P, Harborth J, et al. 2004. Therapeutic silencing of an endogenous gene by systemic administration of modified siRNAs. Nature 432: 173–178. 10.1038/nature03121 [DOI] [PubMed] [Google Scholar]
- Spiess M. 1990. The asialoglycoprotein receptor: a model for endocytic transport receptors. Biochemistry 29: 10009–10018. 10.1021/bi00495a001 [DOI] [PubMed] [Google Scholar]
- Stec WJ, Grajkowski A, Koziolkiewicz M, Uznanski B. 1991. Novel route to oligo (deoxyribonucleoside phosphorothioates). Stereocontrolled synthesis of P-chiral oligo (deoxyribonucleoside phosphorothioates). Nucleic Acids Res 19: 5883–5888. 10.1093/nar/19.21.5883 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stein CA, Hansen JB, Lai J, Wu S, Voskresenskiy A, Høg A, Worm J, Hedtjärn M, Souleimanian N, Miller P, et al. 2010. Efficient gene silencing by delivery of locked nucleic acid antisense oligonucleotides, unassisted by transfection reagents. Nucleic Acids Res 38: e3. 10.1093/nar/gkp841 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stephenson ML, Zamecnik PC. 1978. Inhibition of Rous sarcoma viral RNA translation by a specific oligodeoxyribonucleotide. Proc Natl Acad Sci 75: 285–288. 10.1073/pnas.75.1.285 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Teplova M, Minasov G, Tereshko V, Inamati GB, Cook PD, Manoharan M, Egli M. 1999. Crystal structure and improved antisense properties of 2′-O-(2-methoxyethyl)-RNA. Nat Struct Biol 6: 535–539. 10.1038/9304 [DOI] [PubMed] [Google Scholar]
- Tsimikas S, Karwatowska-Prokopczuk E, Gouni-Berthold I, Tardif J-C, Baum SJ, Steinhagen-Thiessen E, Shapiro MD, Stroes ES, Moriarty PM, Nordestgaard BG, et al. 2020. Lipoprotein(a) reduction in persons with cardiovascular disease. N Engl J Med 382: 244–255. 10.1056/NEJMoa1905239 [DOI] [PubMed] [Google Scholar]
- Turner JJ, Arzumanov AA, Gait MJ. 2005. Synthesis, cellular uptake and HIV-1 Tat-dependent trans-activation inhibition activity of oligonucleotide analogues disulphide-conjugated to cell-penetrating peptides. Nucleic Acids Res 33: 27–42. 10.1093/nar/gki142 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Uehara K, Harumoto T, Makino A, Koda Y, Iwano J, Suzuki Y, Tanigawa M, Iwai H, Asano K, Kurihara K, et al. 2022. Targeted delivery to macrophages and dendritic cells by chemically modified mannose ligand-conjugated siRNA. Nucleic Acids Res 50: 4840–4859. 10.1093/nar/gkac308 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wahlestedt C, Salmi P, Good L, Kela J, Johnsson T, Hökfelt T, Broberger C, Porreca F, Lai J, Ren K, et al. 2000. Potent and nontoxic antisense oligonucleotides containing locked nucleic acids. Proc Natl Acad Sci 97: 5633–5638. 10.1073/pnas.97.10.5633 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wan WB, Seth PP. 2016. The medicinal chemistry of therapeutic oligonucleotides. J Med Chem 59: 9645–9667. 10.1021/acs.jmedchem.6b00551 [DOI] [PubMed] [Google Scholar]
- Wan WB, Migawa MT, Vasquez G, Murray HM, Nichols JG, Gaus H, Berdeja A, Lee S, Hart CE, Lima WF, et al. 2014. Synthesis, biophysical properties and biological activity of second generation antisense oligonucleotides containing chiral phosphorothioate linkages. Nucleic Acids Res 42: 13456–13468. 10.1093/nar/gku1115 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Willibald J, Harder J, Sparrer K, Conzelmann K-K, Carell T. 2012. Click-modified anandamide siRNA enables delivery and gene silencing in neuronal and immune cells. J Am Chem Soc 134: 12330–12333. 10.1021/ja303251f [DOI] [PubMed] [Google Scholar]
- Zamecnik PC, Stephenson ML. 1978. Inhibition of Rous sarcoma virus replication and cell transformation by a specific oligodeoxynucleotide. Proc Natl Acad Sci 75: 280–284. 10.1073/pnas.75.1.280 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou X, Kiesman WF, Yan W, Jiang H, Antia FD, Yang J, Fillon YA, Xiao L, Shi X. 2022. Development of kilogram-scale convergent liquid-phase synthesis of oligonucleotides. J Org Chem 87: 2087–2110. 10.1021/acs.joc.1c01756 [DOI] [PubMed] [Google Scholar]
- Zhukov SA, Pyshnyi DV, Kupryushkin MS. 2021. Synthesis of novel representatives of phosphoryl guanidine oligonucleotides. Russ J Bioorg Chem 47: 380–389. 10.1134/S1068162021020291 [DOI] [Google Scholar]


