Biomolecules & Therapeutics 2023; 31(3): 241-252  https://doi.org/10.4062/biomolther.2023.001
Drug Discovery Perspectives of Antisense Oligonucleotides
Yeonjoon Kim*
Qmine Co., Ltd., Seoul 03759, Republic of Korea
*E-mail: yeonjoonkim@gmail.com
Tel: +82-70-8670-8277, Fax: +82-2-6499-2888
Received: January 3, 2023; Revised: February 12, 2023; Accepted: February 13, 2023; Published online: March 2, 2023.
© The Korean Society of Applied Pharmacology. All rights reserved.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
Abstract
The era of innovative RNA therapies using antisense oligonucleotides (ASOs), siRNAs, and mRNAs is beginning. Since the emergence of the concept of ASOs in 1978, it took more than 20 years before they were developed into drugs for commercial use. Nine ASO drugs have been approved to date. However, they target only rare genetic diseases, and the number of chemistries and mechanisms of action of ASOs are limited. Nevertheless, ASOs are accepted as a powerful modality for next-generation medicines as they can theoretically target all disease-related RNAs, including (undruggable) protein-coding RNAs and non-coding RNAs. In addition, ASOs can not only downregulate but also upregulate gene expression through diverse mechanisms of action. This review summarizes the achievements in medicinal chemistry that enabled the translation of the ASO concept into real drugs, the molecular mechanisms of action of ASOs, the structure-activity relationship of ASO-protein binding, and the pharmacology, pharmacokinetics, and toxicology of ASOs. In addition, it discusses recent advances in medicinal chemistry in improving the therapeutic potential of ASOs by reducing their toxicity and enhancing their cellular uptake.
Keywords: Antisense oligonucleotide, ASO, Medicinal chemistry, Chemical modification, ASO-protein interaction, Structure-activity relationship
INTRODUCTION

Since the antisense oligonucleotide (ASO) concept (Zamecnik and Stephenson, 1978) and RNA interference mechanism (Fire et al., 1998) were disclosed, it took 20 years to introduce RNA therapeutics into the clinic. The first ASO drug, formivirsen, and the first small interfering (si)RNA drug, patisiran, were approved by the U.S. Food & Drug Administration in 1998 and 2018, respectively. The antisense concept could be translated into drugs owing to advances in many fields, including chemistry (medicinal, analytical, organic, and process), biology (RNA, genomics), and pharmacology. Specifically, innovations in medicinal chemistry, including endowing oligonucleotides with drug-likeness and sufficient binding affinity while maintaining target RNA degradation activity via natural enzymatic cleavage, were key. In addition to the several RNA-targeted drugs, including nine ASO drugs (Table 1) and five siRNA drugs, the two mRNA vaccines for COVID-19 approved in 2021 advanced the era of RNA therapy.

Table 1 Approved ASO drugs

DrugApproval yearTarget indicationTarget geneMode of actionChemistry
Formivirsen1998CMV retinitisCMVRNase H1PS-ODN
Mipomersen2013HoFHApoB-100RNase H1PS-MOE gapmer
Nusinersen2016SMASMN2 intron 7Exon inclusionPS-MOE
Eteplirsen2016DMDDystrophin exon 51Exon skippingPMO
Inotersen2018hATTRTTRRNase H1PS-MOE gapmer
Golodirsen2019DMDDystrophin exon 53Exon skippingPMO
Volanesorsen2019FCSApoC-IIIRNase H1PS-MOE gapmer
Viltolarsen2020DMDDystrophin exon 53Exon skippingPMO
Casimersen2021DMDDystrophin exon 45Exon skippingPMO

ApoB-100, apolipoprotein B-100; ApoC-III, apolipoprotein C-III; CMV, cytomegalovirus; DMD, Duchenne muscular dystrophy; hATTR, hereditary transthyretin-mediated amyloidosis; FCS, familial chylomicronemia syndrome; HoFH, homozygous familial hypercholesterolemia; MOE, 2′-methoxyethyl; ODN, oligodeoxynucleotide; PMO, phosphorodiamidate morpholino oligonucleotide; PS, phosphorothioate; SMA, spinal muscular atrophy; TTR, transthyretin. For details on chemistries, refer to the figures below.



RNA-targeted drugs bind to the complementary sequences of their target RNAs through Watson–Crick hybridization; therefore, they can modulate target RNA function very specifically, with few off-target effects, and they can be rationally designed based on sequence information. In addition, they are ideal for precision medicine or personalized medicine. For example, milasen was developed for a single patient named Mila (Kim et al., 2019). Most importantly, with RNA-targeted drugs, nearly all disease-related targets that are “undruggable” or “undrugged” by conventional medicinal approaches can theoretically be controlled. Only approximately 15% of all proteins are considered “druggable” (Makley and Gestwicki, 2013); therefore, 85% of proteins have not been targeted by small-molecule drugs or biologics and can be targeted by RNA therapeutics. Only 2% of human DNA is translated into proteins, while 90% of the genome is transcribed into RNAs, most of which are non-protein-coding (Djebali et al., 2012). Non-coding RNAs are also considered to be potential targets of RNA therapeutics.

RNA-targeted oligonucleotide drugs are either single-stranded or double-stranded. In contrast to double-stranded siRNAs, which can only degrade a target RNA to downregulate gene expression, ASOs possess more versatility as they can not only degrade RNA but also upregulate gene expression or manipulate mRNAs through diverse modes of action. Furthermore, ASOs can undergo more chemical modifications than siRNAs as the chemical modification of the latter is only limited to AGO2 enzyme substrates. Therefore, this review focuses on ASOs. It summarizes the past, present, and future of the medicinal chemistry of ASOs, including how to endow oligonucleotide derivatives with drug-likeness to allow them to overcome evolutionary defense systems that act against invading RNAs; such systems include lipid bilayers, RNases, the reticuloendothelial system, immunogenicity, and endocytosis (Dowdy, 2017). In addition, it discusses discoveries regarding the molecular mechanisms underlying the pharmacology, pharmacokinetics, and toxicology of ASOs, and perspectives for improving the therapeutic potential of ASOs by enhancing their therapeutic index and resolving the issue of tissue/cell-specific delivery.

CHEMICAL MODIFICATIONS AND DESIGNS TO ENDOW ASOS WITH DRUG-LIKENESS

Short DNA/RNA-like oligonucleotides with phosphodiester bonds are not drug-like because they are not able to cross the cell membrane, are rapidly degraded by nucleases and cleared by the kidneys through glomerular filtration, and activate innate immune responses. Even as they are taken up into the cells via endocytosis, they remain trapped inside the lipid bilayer of the endosome and cannot access their target RNAs in the cytosol or nucleus to exert pharmacological effects (Dowdy, 2017).

During the last decades, to endow ASOs with drug-likeness, various chemical modifications have been made and tested. Chemical modifications can be made in various sites of nucleotides, including the linkers, sugars, and nucleobases. Fig. 1 summarizes key chemical modifications that have already been applied in commercial products or are considered promising. Fig. 2 shows representative non-ribose sugar modifications, such as phosphorodiamidate morpholino oligomers (PMOs) and peptide nucleic acids (PNAs).

Figure 1. Chemical modifications in ASOs.

Figure 2. Non-ribose sugar modifications in ASOs.

Among them, however, only four chemical classes, including phosphorothioate (PS) oligodeoxynucleotides (ODNs), PS-2′-O-methoxyethyl (MOE) gapmers, and occupancy-only oligonucleotides such as PS-MOEs and PMOs, have been clinically accepted, as shown in Table 1 and Fig. 3. Therefore, in this section, the chemistries, designs, and pharmacological properties of these classes of ASOs will be described.

Figure 3. ASO designs. BNA, bridged nucleic acid; cEt, constrained ethyl; LNA, locked nucleic acid; MOE, methoxyethyl; PMO, phosphorodiamidate morpholino oligonucleotide; PNA, peptide nucleic acid; PS, phosphorothioate.

Nucleobase modifications

While numerous nucleobase modifications have been attempted to date, the only clinically accepted and commonly used modification to avoid CpG immune stimulation and enhance nuclease stability and binding affinity is 5-methyl cytosine (MeC, Fig. 1).

Linker modifications

The first breakthrough innovation for ASO drug-likeness was the introduction of a PS linkage (Fig. 1) (Stec et al., 1984). In this linker, one non-bridging oxygen of phosphodiester is replaced with a sulfur atom, which greatly alters the physicochemical and pharmacological properties of the resulting ASO (Sands et al., 1994; Eckstein, 2000). The PS modification greatly enhances nuclease resistance. Another key benefit of the PS linkage is that the negative charge in PS is more widely distributed than that in a phosphodiester (PO) bond, increasing lipophilicity. This prevents rapid renal clearance through stronger binding with plasma proteins, and enhances cellular uptake through stronger binding with cell surface proteins such as stabilin-1 and 2 (Miller et al., 2016). However, its stronger binding to immune receptors results in non-specific pro-inflammatory toxicity (Yanai et al., 2011), and the PS modification reduces ASO binding affinity to its complementary nucleic acids by –0.5°C per modification (Freier and Altmann, 1997). The lower potency and pro-inflammatory effect of PS-ODNs (Fig. 3) limit their clinical application and therefore, PS-ODNs require additional chemical modifications.

Sugar modifications

Sugar modification with 2′-alkyl substitution has proven to be a useful strategy to overcome the drawbacks of the PS modification. The most widely used modification, 2′-MOE (Fig. 1), has several advantages, including increased potency resulting from a higher binding affinity (Tm increase of 1.5°C per modification) (Freier and Altmann, 1997), enhanced nuclease resistance, decreased immune-stimulating effect (Henry et al., 2000), and prolonged tissue elimination half-life (Geary et al., 1997, 2001). The bulky alkoxy group at the 2′ position drives the conformational equilibrium of the furanose ring toward the RNA-like C3′-endo conformation (Plavec et al., 1993), which has a higher binding affinity for target RNA than the DNA-like C2′-endo conformation due to the gauche effect. Crystal structure analysis supported this conformation and revealed that hydration of MOE and phosphate oxygen plays a key role in overall stability (Teplova et al., 1999). As it does not support RNase H-mediated degradation, it has been used for occupancy-only oligonucleotides or as flanks or wings of RNase H-active MOE gapmers (Fig. 3).

As mentioned above, the C3′-endo conformation is preferred for strong binding with the target RNA. This conformation is fixed by creating a covalent tether between the 2′ and 4′ positions, leading to a 2′-O,4′-C-methylene-bridged nucleic acid (Fig. 1) or locked nucleic acid (LNA) (Obika et al., 1997; Koshkin et al., 1998) to afford unprecedentedly strong binding affinity for nucleic acids (Tm increase of ~5°C per modification). Owing to the high binding affinity, LNA gapmers (Fig. 3) show strongly improved potency, but are associated with an increased risk of hepatotoxicity, which may depend on ASO sequence and design (Swayze et al., 2007; Hagedorn et al., 2013). Interestingly, the addition of a methyl group to the LNA structure to form 2′,4′-constrained ethyl (cEt, Fig. 1) dramatically reduced hepatotoxicity without a loss in potency as evident in the cEt gapmer (Fig. 3) (Seth et al., 2009b). Although ASOs with such modifications (LNA/cEt gapmers) have not been approved for commercial use, numerous candidates are in clinical trials.

Non-ribose sugar modifications

PMOs (Fig. 2) are a neutral class of oligonucleotide analogs in which a morpholino ring replaces the ribofuranose and a phosphorodiamidate linker replaces the PO (Summerton and Weller, 1997). As they also do not support RNase H activity, they have been used as occupancy-only oligonucleotides for exon skipping and successfully marketed for the treatment of Duchenne muscular dystrophy (DMD). PMOs have the advantage of skeletal muscle-selective distribution, which makes them more efficacious for DMD treatment than other RNase H-inactive structures, such as 2′-MOE or cEt (Sheng et al., 2020).

PNAs (Fig. 2) are another neutral class of oligonucleotide analogs comprising a peptide scaffold and nucleobases for more efficient Watson–Crick hybridization with complementary nucleic acids. PNAs also do not support RNase H-mediated degradation and act as steric blockers or splicing modulators. However, they have low water solubility and poor cell permeability, limiting their application to molecular genetic diagnostics. There are no successful therapeutic PNAs as yet, despite several efforts to overcome the drawbacks, including structural modifications of the PNA backbone (Gupta et al., 2017), conjugation with cell-penetrating peptides (Taylor and Zahid, 2020) or N-acetylgalactosamine (GalNAc) (Bhingardeve et al., 2020), or lipid nanoparticle formulation (Gupta et al., 2016). The diverse efforts for PNA delivery have been reviewed elsewhere (Volpi et al., 2020).

ASO design and main mechanisms of action

All marketed ASO drugs can be classified in two groups based on their mechanisms of action: RNase H-mediated degradation for PS-ODN (formivirsen) and MOE gapmers (mipomersen, inotersen, and volanesorsen), or alternative splicing (exon inclusion or skipping) for occupancy-only oligonucleotides (PS-MOEs such as nusinersen and PMOs including eteplirsen, golodirsen, viltolarsen, and casimersen).

In RNase H-mediated degradation, the RNase H1 enzyme specifically recognizes and cleaves RNA-DNA-like heteroduplexes (Wu et al., 2004; Cerritelli and Crouch, 2009). For the ASO in an RNA-ASO duplex to be RNase H-active, at least five (Monia et al., 1993), but optimally, eight to ten contiguous deoxynucleotides are required. In addition, PS linkages can activate RNase H1 as PO linkages. Therefore, the central PS-ODN region of 8-10 nucleotides (gap) is flanked by various 3-5-nt 2′-modified nucleotides (wings) such as MOE, LNA, or cEt, which contribute to increased stability and affinity, to generate a gapmer structure (Fig. 3).

Since all 2′ modifications and non-ribose sugar modifications result in loss of RNase H activity, they result in occupancy-only oligonucleotides unless RNase H substrate ODNs are included (Fig. 3). The occupancy-only oligonucleotides bind to the target pre-mRNA and sterically block the spliceosome, leading to alternative splicing in the form of exon inclusion or skipping (nusinersen and four PMO drugs); as a result, they can be used in the treatment of spinal muscular atrophy (Garber, 2016) or DMD (Echevarría et al., 2018).

Spinal muscular atrophy is caused by homozygous loss of function of the survival of motor neuron 1 gene (SMN1). Although the redundant SMN2 gene produces limited amounts of full-length SMN protein, it frequently produces short and unstable protein because of exon 7 skipping during pre-mRNA splicing. Nusinersen is an 18-mer fully 2′-MOE-modified PS ASO that binds to and modifies the splicing of SMN2 pre-mRNA to include exon 7, thus restoring full-length SMN expression (Hua et al., 2007).

DMD is a rapidly progressing neuromuscular disorder. Mutations in the DMD gene that disrupt its reading frame introduce premature stop codons, halting gene expression. Eteplirsen repairs the reading frame by binding to exon 51 in the pre-mRNA, which is then skipped in translation, producing a shortened, but functional dystrophin (McDonald et al., 2021). Similarly, golodirsen (Servais et al., 2022) and viltolarsen (Clemens et al., 2020) adjust the splicing pattern of mutant DMD pre-mRNA by binding to exon 53. The absence of exon 53 in the mature mRNA restores dystrophin expression. Casimersen (Wagner et al., 2021) binds to and promotes the skipping of exon 45.

Additional molecular mechanisms for gene expression regulation by ASOs

In addition to the RNase H-mediated degradation or alternative splicing described above, ASOs have several other mechanisms to downregulate or upregulate gene expression, enhancing their versatility. Evolved RNA biology has identified diverse post-RNA-hybridization events of ASOs that are broadly classified into two mechanisms: enzymatic degradation and occupancy-only, as summarized in Table 2 (Crooke et al., 2021). Two enzymes degrade RNA-oligonucleotide duplexes. In addition to RNase H discussed above, ASO-RNA hybrids can recruit Argonaute 2 (AGO2) to degrade the target RNA. Occupancy-only can be further classified into alternative splicing and steric blocking. Alternative splicing of pre-mRNA leads to exon inclusion or skipping, as explained above, or it leads to nonsense-mediated decay (NMD) if the resulting mRNA contains a premature termination codon. The effects of steric blocking on the regulation of gene expression are known to vary based on the ASO binding site. The detailed mechanisms and structure-activity relationships in terms of the endpoint of gene regulation, i.e., downregulation or upregulation, are discussed below.

Table 2 Modes of action and relevant chemistry designs of ASOs

Enzymatic degradationOccupancy-only
Alternative splicingSteric blocking
DownregulationRNase H-mediatedNMDNGD
AGO2-mediatedTranslation arrest
5′-cap inhibition
Altering the polyadenylation site
UpregulationExon inclusionmiRNA function inhibition
Exon skippingModulation of uORF/TIE
Inhibition of NMD
ChemistryRNase H: DNA (w/wo PS linkage) or gapmerFully 2′-modified RNA
AGO2: dsRNA (siRNA w/wo 2′- F/OMe) or ssRNA (5′-phosphate)PMO
PNA

AGO2, Argonaute 2; NGD, no-go decay; NMD, nonsense-mediated decay; uORF, upstream open reading frame; TIE, translation inhibitory element.



Downregulation of gene expression: AGO2-mediated degradation is the main mechanism by which miRNA/siRNA cleaves its target RNA (Song et al., 2004). However, single-stranded siRNA stabilized with 2′-modification and a 5′-(E)-vinylphosphonate, which mimics the 5′-phosphate required for AGO2 activation, can be a substrate (guide strand) of AGO2 (Lima et al., 2012).

There are two RNA degradation mechanisms other than degradation by RNase H or AGO2: NMD and no-go decay (NGD). NMD of mRNA is initiated when ASO triggers splicing modulation of pre-mRNA to generate an mRNA that contains premature termination codons, which are NMD targets (Ward et al., 2014). Fully modified 2′-MOE ASOs that bind to the 3′ terminus of the open reading frame (ORF) degrade target RNA via NGD (Liang et al., 2019b). Fully modified cEt ASOs, which have a higher binding affinity to RNA are known to have a lower efficacy, which may be a result of the stronger binding to proteins. 2′-MOE ASOs with only one cEt have a similar efficacy, while less affined 2′-OMe ASOs have a lower efficacy.

A few reports have disclosed steric blocking as a minor mechanism of downregulation. ASOs can sterically block the translation of mature mRNAs (Melton, 1985; Iversen et al., 2003) and guide the cleavage of 5′-cap structures, inhibiting translation (Baker et al., 1999). Finally, ASOs can modulate the use of polyadenylation sites by binding to pre-mRNA sequences involved in cleavage and polyadenylation (Vickers et al., 2001).

Upregulation of gene expression: While miRNAs generally downregulate gene expression, ASOs can upregulate gene expression through the inhibition of miRNA function by binding to miRNAs or miRNA-binding sites (Ørom et al., 2006; De Santi et al., 2020)

Upstream ORFs in the 5′-UTR are sequences that are defined by an initiation codon in frame with a termination codon located upstream or downstream of the main AUG. They are present in approximately half of the human transcripts and inhibit protein expression (Calvo et al., 2009). A fully 2′-modified PO or PS-ASO targeting an upstream ORF dose-dependently increased protein expression by 30-150% (Liang et al., 2016b). Eighteen-mer PS-MOE, 12-16-mer PS-2′-OMe, and 16-mer PO-2′-OMe ASOs have been suggested as optimal lengths and chemistries for gene upregulation.

Translation was enhanced approximately 3-fold when the recruitment of translation initiation factors to the target mRNA was improved through the binding of fully 2′-modified 16-mer PS-cEt to translation inhibitory elements in 5′-UTRs (Liang et al., 2017).

As NMD regulates the normal expression of many genes, ASOs can upregulate gene expression by inhibiting NMD either through the degradation of NMD factors (Huang et al., 2018) or through targeting the binding site of the exon junction complex in an mRNA (Nomakuchi et al., 2016; Kim et al., 2022). Splicing modulation of an ASO preventing non-productive (NMD-inducing) alternative splicing upregulated gene expression (Lim et al., 2020).

ASO-PROTEIN INTERACTION: STRUCTURE-ACTIVITY RELATIONSHIP

Many years of research on the mechanisms underlying the potency, pharmacokinetics, and toxicity of ASOs have revealed that ASO-protein interaction is a key factor (Crooke et al., 2020a, 2020b). In general, the binding affinity of ASOs for several proteins depends on the PS linkage, gapmer design, and sequence (Vickers and Crooke, 2016). In addition, more hydrophobic 2′ modifications, such as fluorine (F), LNA, or cEt, tend to enhance protein binding compared to more hydrophilic 2′ modifications, such as MOE or OMe, leading to an approximate 10-fold difference in binding affinity for diverse proteins. Many proteins prefer binding to the 5′-wing and the ~6 nucleotides of the 5′-gap position, which contain more hydrophobic 2′ modifications (Shen et al., 2019). This section discusses the structure-activity relationship of ASO-protein interaction and its effects on ASO properties, such as nuclease resistance, pharmacokinetic profile, and cellular uptake and endosomal escape capabilities.

Nucleases

Ubiquitous nucleases cleave the PO bonds of unmodified DNA-type oligonucleotides, which therefore have a half-life of only minutes. PS linkages stabilize ASOs against nuclease activity, increasing their plasma half-life to up to 1 h. Chemical modification of the 2′ position with MOE, cEt, or LNA eliminates exonuclease activity, extending tissue elimination half-life to four weeks. MOE gapmers are highly resistant to exonucleases, but are slowly metabolized by endonucleases at the DNA gap site to yield short metabolites of DNA ends, which are degraded further by exonucleases and/or excreted in the urine due to weak plasma protein binding.

Plasma proteins

Plasma protein binding is the main factor for improving absorption and distribution by suppressing renal clearance (Gaus et al., 2019). As mentioned above, PS linkages are the key structure for protein binding. As effective binding requires at least 10-12 PS moieties, metabolites from endonucleases and exonucleases that have less than 10 PS groups are cleared from tissues and excreted in the urine via glomerular filtration (Geary et al., 2015). As albumin is the most abundant protein in plasma (600 µM) and its binding affinity to a PS-MOE gapmer has a Kd of 12.7 µM (Gaus et al., 2019), it plays a major role in the plasma protein binding of PS-ASOs. The binding affinity is sufficiently high to effectively inhibit renal clearance, but sufficiently low to facilitate tissue uptake. In addition, the binding affinity depends on the chemical structure and sequence of the ASO. For example, occupancy-only full PS-MOE has a Kd of 26.9 µM, and T20 PS-ODN has a Kd of 0.94 µM, whereas A20 PS-ODN has a Kd of 204.5 µM.

Cell surface proteins

ASOs are taken up into cells via endocytosis (Doherty and McMahon, 2009). Non-productive uptake results in the accumulation of ASOs in late endosomes and lysosomes, whereas productive uptake involves clathrin- and caveolin-independent endocytosis (Koller et al., 2011). Among the numerous cell surface proteins, stabilin-1 and 2 receptors are responsible for ASO cellular uptake and internalization (Miller et al., 2016). A binding affinity study (Gaus et al., 2018) revealed that PS-ODN had a similar affinity to full PS-MOE, PS-MOE gapmer (5-10-5), and PS-cEt gapmer (5-10-5), which implied that 2′ modifications do not strongly contribute to the binding affinity. The binding affinity decreased as the number of PS linkages decreased (15 PS has a Kd of 83.9 µM, whereas 10 PS and PO-MOE gapmer (5-10-5) have a Kd of >1,000 µM), which indicated that PS linkage is the major contributor to stabilin-2 binding and that at least 15 PS linkages are required for appropriate cellular uptake.

Asialoglycoprotein receptor is highly expressed on hepatocytes (Stockert, 1995). GalNAc is a ligand of this receptor and has been conjugated to an ASO (Prakash et al., 2014) as well as an siRNA (Zimmermann et al., 2017) for liver/hepatocyte-specific delivery. ASOs can be selectively delivered to pancreatic β-cells via conjugation with glucagon-like peptide 1 receptor agonist (Ämmälä et al., 2018; Knerr et al., 2021).

Intracellular proteins

After cellular internalization, ASOs are trafficked through multiple membrane-bound intracellular compartments. They then slowly escape endosomes into the cytoplasm and nucleus, where they bind to their target RNAs. The interaction between ASOs and diverse intracellular proteins impacts their intracellular location, including intracellular trafficking, endosomal escape, and transport to the nucleus, affecting their pharmacological effects, such as potency and toxicity.

PS-ASOs interact with more than 50 intracellular proteins, such as P54nrb, which have RNA-recognition motifs, as well as chaperone proteins, such as HSP90, which lack RNA- or DNA-binding domains, and other proteins (Liang et al., 2015, 2016a). Some proteins, including La/SSB, NPM1, ANXA2, VARS, and PC4, appear to enhance ASO activity, likely through mechanisms related to subcellular distribution. VARS and ANXA2, which are co-localized with ASOs in endocytic organelles, may facilitate ASO release from the endosomes. La and NPM1 may play a role in ASO nuclear accumulation. In contrast, Ku70 and Ku80 proteins inhibit ASO activity, most likely by competing with RNase H1 for ASO/RNA duplex binding. Thus, PS-ASOs bind a set of cellular proteins that affect ASO activity via different mechanisms.

Endosomal escape is the rate-limiting step for ASO action. Several studies have suggested that intermediate endosomal compartments, such as multivesicular bodies and late endosomes, are key sites for productive ASO release into the cytosol (Crooke et al., 2017b; Juliano, 2018). The detailed mechanism has not yet been fully unraveled, but recent studies have revealed that proteins, such as M6PR (Liang et al., 2019a), and COPII vesicles (Liang et al., 2018) may play key roles in this process. No chemical modifications or conjugates for improving endosomal escape of ASOs have been discovered yet, although some small-molecular additives (Yang et al., 2015; Wang et al., 2017; Juliano et al., 2018), conjugation with synthetic endosomal escape domains (Lönn et al., 2016), and diverse cell-penetrating peptides (Kondow-McConaghy et al., 2020) have been evaluated. As it is estimated that the endosomal escape rate is quite low (<1%), ASO potency would be dramatically improved if the endosomal escape issue were to be resolved.

TOXICITY OF ASOS: STRUCTURE-ACTIVITY RELATIONSHIP

The toxicity of ASOs can be classified into sequence-dependent and sequence-independent toxicity. Sequence-dependent toxicity stems from the hybridization of ASOs with unintended targets, such as long pre-mRNA transcripts (Burel et al., 2016) or long introns (Kamola et al., 2015); however, it occurs less frequently than once thought (Lima et al., 2014). Sequence-independent toxicity stems from the interaction of ASOs with several proteins, such as P54nrb and RNase H1 (Vickers et al., 2019), which have been suggested to be key proteins. Toxic ASOs bind to paraspeckle proteins and RNase H1 to mislocalize them to the nucleolus and inhibit nucleolar RNA transcription and processing, which leads to nucleolar stress and apoptosis (Shen et al., 2019). The hepatotoxicity of ASOs may originate from sequence motifs, such as TCC/TGC, that typically bind to hepatocellular proteins, increasing the activity of the P53 and nuclear factor erythroid-2-related factor 2 stress pathways (Burdick et al., 2014).

MOE gapmer ASOs

Integrated safety assessment of MOE gapmer ASOs for kidney and liver function, hematology, and complement activation in non-human primates and humans has revealed that this chemical class is well tolerated in humans compared to the toxicities, such as complement activation and effects on platelets, found in non-human primates (Crooke et al., 2016). Although systemically administered MOE gapmer ASOs are distributed mainly to the kidneys and metabolites are cleared in the urine, no marked renal toxicity has been observed in numerous clinical trials using up to >175 mg/week dose and 52-week treatment (Crooke et al., 2018). After 4.6 years of treatment with up to 624 mg/week, MOE gapmer ASOs had no negative impact on platelet count (Crooke et al., 2017a).

Compared to PS-ODNs, MOE gapmers are substantially less immunostimulatory. In addition, immunotoxicity is dose-dependent; therefore, more potent MOE or cEt gapmer ASOs for which the therapeutic dose is lower have a lower risk of immunotoxicity. The CpG motif stimulates the innate immune response; however, 5-methyl cytosine replacement reduced this side effect.

A few MOE gapmers, including ISIS 147420, has been known to induce severe inflammatory response in mice. The adverse effects are characterized by the induction of interferon-β, followed by acute transaminitis and extensive hepatocyte apoptosis and necrosis (Burel et al., 2012). A specific sequence motif in viral DNA or RNA may trigger this acute innate immune response; thus, a single-nucleotide change can abolish this toxicity.

LNA/cEt gapmer ASOs

As mentioned above, LNA gapmers have higher risk of hepatotoxicity than MOE or cEt gapmers. The hepatotoxicity depends on a number of parameters, such as their length, gapmer design, and sequence. Shorter LNA gapmers have lower hepatotoxicity (Seth et al., 2009a). Regardless of 2′ modifications, toxic ASOs are able to bind to paraspeckle proteins, such as P54nrb and PSF, as well as RNase H1, delocalizing them to the nucleoli, which leads to nucleolar stress, P53 activation, and apoptosis (Shen et al., 2019). It has been reported that more toxic ASOs bind more proteins than non-toxic ones, and 5′-MOE-DNA-3′-cEt gapmer (mixmer) is less toxic than cEt gapmer or 5′-cEt-DNA-3′-MOE gapmer (mixmer), which implies that the 5′-side and more hydrophobic residues are preferred for protein binding. The introduction of a 2′-OMe at gap position 2 (Fig. 4) mitigated cytotoxicity by suppressing protein binding. Several chemical modifications with the aim of reducing toxicity and improving the therapeutic potential of ASOs are discussed in more detail below.

Figure 4. Site-specific modifications to mitigate toxicity. MOP, methoxy propyl phosphonate; MsPA, mesyl phosphoramidate.
CHEMICAL MODIFICATIONS FOR IMPROVING THE THERAPEUTIC POTENTIAL OF ASO

As LNA and cEt gapmers with high affinity and potency have the issue of hepatotoxicity, diverse chemical modifications, including site-specific ones, have been developed, usually focused on the 5′ side of the gap, to mitigate toxicity (Fig. 4).

Nucleobase modifications

Recently, Yoshida et al. (2022) reported that some nucleobase modifications, such as 5-hydroxycytosine, 8-bromo, and amine guanine (Fig. 1), reduce the hepatotoxicity of LNA gapmers.

Linkage modifications

Alkylphosphonates: While the methyl phosphonate linkage (Fig. 1) was introduced long ago (Miller et al., 1979) and despite its advantages, such as having a neutral charge and nuclease resistance, it was not widely used due to RNase H inactivity, a relatively low affinity for RNA, and chemical instability under the basic condition required for the cleavage/deprotection step of oligonucleotide synthesis on a solid support. Recently, site-specific replacement of PS with alkyl phosphonates, such as methyl phosphonate or methoxy propyl phosphonate (Fig. 1), was reported to reduce toxicity by modulating the interactions with intracellular proteins (Migawa et al., 2019). For example, a 3-10-3 cEt gapmer (Fig. 3) in which one or two PS linkages at position 2 or 3 from the 5′-gap end were replaced with methyl phosphonate or methoxy propyl phosphonate (Fig. 4) showed comparable potency and substantially reduced hepatotoxicity, resulting in a higher therapeutic index than that of the parent ASO. The reduction in toxicity was accompanied by the absence of nucleolar mislocalization of paraspeckle protein P54nrb, ablation of P21 mRNA elevation, and caspase activation, which are thought to be key mechanisms of hepatotoxicity of PS-ASOs.

Mesyl phosphoramidate (MsPA): Oligonucleotide derivatives with an MsPA (Fig. 1) linkage, the synthetic method of which was recently developed (Chelobanov et al., 2017), have been demonstrated to have RNase H-activating ability, nuclease resistance, and anti-miRNA activity (Miroshnichenko et al., 2019). Their superior in vivo efficacy over PS (Patutina et al., 2020) and potential application in splice-switching (Hammond et al., 2021) have been demonstrated. A comprehensive study by (Anderson et al., 2021) on the impact of MsPA linkages on therapeutic properties showed that replacing up to 5 PS linkages in the gap with MsPA (Fig. 4) was well tolerated, and replacing specific PS linkages at appropriate locations greatly improved nuclease stability and potency and reduced both immune stimulation and cytotoxicity, leading to an improved therapeutic index, reduced pro-inflammatory effects, and extended effect duration.

Amides: Following the early attempts to replace PO with amide (1 in Fig. 5) (De Mesmaeker et al., 1994), the application of this neutral linkage to ASOs had not extensively been studied. However, recent systemic studies have investigated its impact on the pharmacological properties of an siRNA (Mutisya et al., 2017; Kotikam and Rozners, 2020) and an ASO (Epple et al., 2020). It was revealed that the amide linkage has no RNase H activity, but it could replace PO linkages at each wing of a gapmer to maintain RNA binding affinity and RNase H activity. The amide linkages combined with LNA (2 in Fig. 5) enhanced potency and cellular uptake, and reduced toxicity (Baker et al., 2022). The exon-skipping activity of an 18-mer ASO containing 4 LNA-amide linkages and 14 2′-OMe with 13 PS linkages (3 in Fig. 5) was higher than that of an 18-mer ASO with 17 PS linkages with all 2′-OMe or 8 LNA+10 2′-OMe. More importantly, the activity gap substantially increased when they were transfected without lipofectamine, which implied that the 18-mer ASO containing 4 LNA-amide linkages had substantially better cellular uptake ability.

Figure 5. Amide linkages and ASOs containing them. 1. Amide linkage 2. LNA-Amide linkage 3. Combination of LNA-amide linkage and 2-OMe-PS linkage.

Synergy by combination

The combination of 2′-OMe at the 5′-gap 2 position and two MsPA linkages at the 5′ end of the gap or in the 3′-wing improved the RNase H1 cleavage rate, considerably reduced the binding of proteins involved in cytotoxicity, and extended the elimination half-live (Zhang et al., 2022).

CONCLUSIONS AND PERSPECTIVES

During the last decades, advances in medicinal chemistry have led to the development of drug-like ASO drugs that are sufficiently potent to treat various rare diseases. Among many chemical modifications, PS linkage, 2′-modification with MOE or cEt, and PMO are the key achievements that have been successfully clinically applied. As numerous molecular mechanisms underlying the pharmacology, pharmacokinetics, and toxicology of ASOs, such as ASO-protein interactions, have been revealed, it is now possible to manipulate the chemical modification and design of ASOs to broaden their therapeutic margin by fine-tuning or combining available chemistries based on structure-activity relationships. For example, site-specific modifications with 2′-OMe, MsPA, or an amide linkage are efficient in reducing toxicity while maintaining potency.

For ASOs to be more widely used for common diseases as well, two key challenges remain to be resolved. The first is tissue/cell-specific delivery to organs other than the liver. Besides GalNAc ligand conjugates targeting the asialoglycoprotein receptor on hepatocytes, which have been approved for clinical uses, another specific and efficacious ligand-receptor system should be discovered for targeting another tissues or cells. The second is chemical modification or conjugation to enhance cellular uptake and endosomal escape for improved potency. Some neutral linkers, such as amides that reduce the negative charge of PS-ASOs, are promising for this purpose. They are important because they will enhance potency and safety and lower the therapeutic dose, and as a result, reduce the cost and the risk of dose-dependent toxicity.

CONFLICT OF INTEREST

The author is the founder and CEO of Qmine Co., Ltd.

References
  1. Ämmälä, C., Drury, W. J., Knerr, L., Ahlstedt, I., Stillemark-Billton, P., Wennberg-Huldt, C., Andersson, E. M., Valeur, E., Jansson-Löfmark, R., Janzén, D., Sundström, L., Meuller, J., Claesson, J., Andersson, P., Johansson, C., Lee, R. G., Prakash, T. P., Seth, P. P., Monia, B. P. and Andersson, S. (2018) Targeted delivery of antisense oligonucleotides to pancreatic β-cells. Sci. Adv. 4, eaat3386.
    Pubmed KoreaMed CrossRef
  2. Anderson, B. A., Freestone, G. C., Low, A., De-Hoyos, C. L., Iii, W. J. D., Ostergaard, M. E., Migawa, M. T., Fazio, M., Wan, W. B., Berdeja, A., Scandalis, E., Burel, S. A., Vickers, T. A., Crooke, S. T., Swayze, E. E., Liang, X. and Seth, P. P. (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.
    Pubmed KoreaMed CrossRef
  3. Baker, B. F., Lot, S. S., Kringel, J., Cheng-Flournoy, S., Villiet, P., Sasmor, H. M., Siwkowski, A. M., Chappell, L. L. and Morrow, J. R. (1999) Oligonucleotide-europium complex conjugate designed to cleave the 5' cap structure of the ICAM-1 transcript potentiates antisense activity in cells. Nucleic Acids Res. 27, 1547-1551.
    Pubmed KoreaMed CrossRef
  4. Baker, Y. R., Thorpe, C., Chen, J., Poller, L. M., Cox, L., Kumar, P., Lim, W. F., Lie, L., McClorey, G., Epple, S., Singleton, D., McDonough, M. A., Hardwick, J. S., Christensen, K. E., Wood, M. J. A., Hall, J. P., El-Sagheer, A. H. and Brown, T. (2022) An LNA-amide modification that enhances the cell uptake and activity of phosphorothioate exon-skipping oligonucleotides. Nat. Commun. 13, 4036.
    Pubmed KoreaMed CrossRef
  5. Bhingardeve, P., Madhanagopal, B. R., Naick, H., Jain, P., Manoharan, M. and Ganesh, K. (2020) Receptor-specific delivery of peptide nucleic acids conjugated to three sequentially linked N-acetyl galactosamine moieties into hepatocytes. J. Org. Chem. 85, 8812-8824.
    Pubmed CrossRef
  6. Burdick, A. D., Sciabola, S., Mantena, S. R., Hollingshead, B. D., Stanton, R., Warneke, J. A., Zeng, M., Martsen, E., Medvedev, A., Makarov, S. S., Reed, L. A., Davis, J. W. and Whiteley, L. O. (2014) Sequence motifs associated with hepatotoxicity of locked nucleic acid--modified antisense oligonucleotides. Nucleic Acids Res. 42, 4882-4891.
    Pubmed KoreaMed CrossRef
  7. Burel, S. A., Hart, C. E., Cauntay, P., Hsiao, J., Machemer, T., Katz, M., Watt, A., Bui, H. H., Younis, H., Sabripour, M., Freier, S. M., Hung, G., Dan, A., Prakash, T. P., Seth, P. P., Swayze, E. E., Bennett, C. F., Crooke, S. T. and Henry, S. P. (2016) Hepatotoxicity of high affinity gapmer antisense oligonucleotides is mediated by RNase H1 dependent promiscuous reduction of very long pre-mRNA transcripts. Nucleic Acids Res. 44, 2093-2109.
    Pubmed KoreaMed CrossRef
  8. Burel, S. A., Machemer, T., Ragone, F. L., Kato, H., Cauntay, P., Greenlee, S., Salim, A., Gaarde, W. A., Hung, G., Peralta, R., Freier, S. M. and Henry, S. P. (2012) Unique O-methoxyethyl ribose-DNA chimeric oligonucleotide induces an atypical melanoma differentiation-associated gene 5-dependent induction of type I interferon response. J. Pharmacol. Exp. Ther. 342, 150-162.
    Pubmed CrossRef
  9. Calvo, S. E., Pagliarini, D. J. and Mootha, V. K. (2009) Upstream open reading frames cause widespread reduction of protein expression and are polymorphic among humans. Proc. Natl. Acad. Sci. U. S. A. 106, 7507-7512.
    Pubmed KoreaMed CrossRef
  10. Cerritelli, S. M. and Crouch, R. J. (2009) Ribonuclease H: the enzymes in eukaryotes. FEBS J. 276, 1494-1505.
    Pubmed KoreaMed CrossRef
  11. Chelobanov, B. P., Burakova, E. A., Prokhorova, D. V., Fokina, A. A. and Stetsenko, D. A. (2017) New oligodeoxynucleotide derivatives containing N-(methanesulfonyl)-phosphoramidate (mesyl phosphoramidate) internucleotide group. Russ. J. Bioorg. Chem. 43, 664-668.
    CrossRef
  12. Clemens, P. R., Rao, V. K., Connolly, A. M., Harper, A. D., Mah, J. K., Smith, E. C., McDonald, C. M., Zaidman, C. M., Morgenroth, L. P., Osaki, H., Satou, Y., Yamashita, T. and Hoffman, E. P. (2020) Safety, tolerability, and efficacy of viltolarsen in boys with Duchenne muscular dystrophy amenable to exon 53 skipping: a phase 2 randomized clinical trial. JAMA Neurol. 77, 982-991.
    Pubmed KoreaMed CrossRef
  13. Crooke, S. T., Baker, B. F., Crooke, R. M. and Liang, X. H. (2021) Antisense technology: an overview and prospectus. Nat. Rev. Drug Discov. 20, 427-453.
    Pubmed CrossRef
  14. Crooke, S. T., Baker, B. F., Kwoh, T. J., Cheng, W., Schulz, D. J., Xia, S., Salgado, N., Bui, H. H., Hart, C. E., Burel, S. A., Younis, H. S., Geary, R. S., Henry, S. P. and Bhanot, S. (2016) Integrated safety assessment of 2'-O-methoxyethyl chimeric antisense oligonucleotides in nonhuman primates and healthy human volunteers. Mol. Ther. 24, 1771-1782.
    Pubmed KoreaMed CrossRef
  15. Crooke, S. T., Baker, B. F., Pham, N. C., Hughes, S. G., Kwoh, T. J., Cai, D., Tsimikas, S., Geary, R. S. and Bhanot, S. (2018) The effects of 2'-O-methoxyethyl oligonucleotides on renal function in humans. Nucleic Acid Ther. 28, 10-22.
    Pubmed KoreaMed CrossRef
  16. Crooke, S. T., Baker, B. F., Witztum, J. L., Kwoh, T. J., Pham, N. C., Salgado, N., McEvoy, B. W., Cheng, W., Hughes, S. G., Bhanot, S. and Geary, R. S. (2017a) The effects of 2'-O-methoxyethyl containing antisense oligonucleotides on platelets in human clinical trials. Nucleic Acid Ther. 27, 121-129.
    Pubmed KoreaMed CrossRef
  17. Crooke, S. T., Seth, P. P., Vickers, T. A. and Liang, X. H. (2020a) The interaction of phosphorothioate-containing RNA targeted drugs with proteins is a critical determinant of the therapeutic effects of these agents. J. Am. Chem. Soc. 142, 14754-14771.
    Pubmed CrossRef
  18. Crooke, S. T., Vickers, T. A. and Liang, X. H. (2020b) Phosphorothioate modified oligonucleotide-protein interactions. Nucleic Acids Res. 48, 5235-5253.
    Pubmed KoreaMed CrossRef
  19. Crooke, S. T., Wang, S., Vickers, T. A., Shen, W. and Liang, X. H. (2017b) Cellular uptake and trafficking of antisense oligonucleotides. Nat. Biotechnol. 35, 230-237.
    Pubmed CrossRef
  20. De Mesmaeker, A., Waldner, A., Lebreton, J., Hoffmann, P., Fritsch, V., Wolf, R. M. and Freier, S. M. (1994) Amides as a new type of backbone modification in oligonucleotides. Angew. Chem. Int. Ed. Engl. 33, 226-229.
    CrossRef
  21. De Santi, C., Fernández Fernández, E., Gaul, R., Vencken, S., Glasgow, A., Oglesby, I. K., Hurley, K., Hawkins, F., Mitash, N., Mu, F., Raoof, R., Henshall, D. C., Cutrona, M. B., Simpson, J. C., Harvey, B. J., Linnane, B., McNally, P., Cryan, S. A., MacLoughlin, R., Swiatecka-Urban, A. and Greene, C. M. (2020) Precise targeting of miRNA sites restores CFTR activity in CF bronchial epithelial cells. Mol. Ther. 28, 1190-1199.
    Pubmed KoreaMed CrossRef
  22. Djebali, S., Davis, C. A., Merkel, A., Dobin, A., Lassmann, T., Mortazavi, A., Tanzer, A., Lagarde, J., Lin, W., Schlesinger, F., Xue, C., Marinov, G. K., Khatun, J., Williams, B. A., Zaleski, C., Rozowsky, J., Röder, M., Kokocinski, F., Abdelhamid, R. F., Alioto, T., Antoshechkin, I., Baer, M. T., Bar, N. S., Batut, P., Bell, K., Bell, I., Chakrabortty, S., Chen, X., Chrast, J., Curado, J., Derrien, T., Drenkow, J., Dumais, E., Dumais, J., Duttagupta, R., Falconnet, E., Fastuca, M., Fejes-Toth, K., Ferreira, P., Foissac, S., Fullwood, M. J., Gao, H., Gonzalez, D., Gordon, A., Gunawardena, H., Howald, C., Jha, S., Johnson, R., Kapranov, P., King, B., Kingswood, C., Luo, O. J., Park, E., Persaud, K., Preall, J. B., Ribeca, P., Risk, B., Robyr, D., Sammeth, M., Schaffer, L., See, L.-H., Shahab, A., Skancke, J., Suzuki, A. M., Takahashi, H., Tilgner, H., Trout, D., Walters, N., Wang, H., Wrobel, J., Yu, Y., Ruan, X., Hayashizaki, Y., Harrow, J., Gerstein, M., Hubbard, T., Reymond, A., Antonarakis, S. E., Hannon, G., Giddings, M. C., Ruan, Y., Wold, B., Carninci, P., Guigó, R. and Gingeras, T. R. (2012) Landscape of transcription in human cells. Nature 489, 101-108.
    Pubmed KoreaMed CrossRef
  23. Doherty, G. J. and McMahon, H. T. (2009) Mechanisms of endocytosis. Annu. Rev. Biochem. 78, 857-902.
    Pubmed CrossRef
  24. Dowdy, S. F. (2017) Overcoming cellular barriers for RNA therapeutics. Nat. Biotechnol. 35, 222-229.
    Pubmed CrossRef
  25. Echevarría, L., Aupy, P. and Goyenvalle, A. (2018) Exon-skipping advances for Duchenne muscular dystrophy. Hum. Mol. Genet. 27, R163-R172.
    Pubmed CrossRef
  26. Eckstein, F. (2000) Phosphorothioate oligodeoxynucleotides: what is their origin and what is unique about them? Antisense Nucleic Acid Drug Dev. 10, 117-121.
    Pubmed CrossRef
  27. Epple, S., Thorpe, C., Baker, Y. R., El-Sagheer, A. H. and Brown, T. (2020) Consecutive 5′- and 3′-amide linkages stabilise antisense oligonucleotides and elicit an efficient RNase H response. Chem. Commun. 56, 5496-5499.
    Pubmed CrossRef
  28. Fire, A., Xu, S., Montgomery, M. K., Kostas, S. A., Driver, S. E. and Mello, C. C. (1998) Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature 391, 806-811.
    Pubmed CrossRef
  29. Freier, S. M. and Altmann, K. H. (1997) The ups and downs of nucleic acid duplex stability: structure-stability studies on chemically-modified DNA:RNA duplexes. Nucleic Acids Res. 25, 4429-4443.
    Pubmed KoreaMed CrossRef
  30. Garber, K. (2016) Big win possible for Ionis/Biogen antisense drug in muscular atrophy. Nat. Biotechnol. 34, 1002-1003.
    Pubmed CrossRef
  31. Gaus, H., Miller, C. M., Seth, P. P. and Harris, E. N. (2018) Structural determinants for the interactions of chemically modified nucleic acids with the stabilin-2 clearance receptor. Biochemistry 57, 2061-2064.
    Pubmed KoreaMed CrossRef
  32. Gaus, H. J., Gupta, R., Chappell, A. E., Østergaard, M. E., Swayze, E. E. and Seth, P. P. (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.
    Pubmed KoreaMed CrossRef
  33. Geary, R. S., Baker, B. F. and Crooke, S. T. (2015) Clinical and preclinical pharmacokinetics and pharmacodynamics of mipomersen (kynamro(®)): a second-generation antisense oligonucleotide inhibitor of apolipoprotein B. Clin. Pharmacokinet. 54, 133-146.
    Pubmed KoreaMed CrossRef
  34. Geary, R. S., Leeds, J. M., Fitchett, J., Burckin, T., Truong, L., Spainhour, C., Creek, M. and Levin, A. A. (1997) Pharmacokinetics and metabolism in mice of a phosphorothioate oligonucleotide antisense inhibitor of C-raf-1 kinase expression. Drug Metab. Dispos. 25, 1272-1281.
    Pubmed
  35. Geary, R. S., Watanabe, T. A., Truong, L., Freier, S., Lesnik, E. A., Sioufi, N. B., Sasmor, H., Manoharan, M. and Levin, A. A. (2001) Pharmacokinetic properties of 2'-O-(2-methoxyethyl)-modified oligonucleotide analogs in rats. J. Pharmacol. Exp. Ther. 296, 890-897.
    Pubmed
  36. Gupta, A., Bahal, R., Gupta, M., Glazer, P. M. and Saltzman, W. M. (2016) Nanotechnology for delivery of peptide nucleic acids (PNAs). J. Control. Release 240, 302-311.
    Pubmed KoreaMed CrossRef
  37. Gupta, A., Mishra, A. and Puri, N. (2017) Peptide nucleic acids: advanced tools for biomedical applications. J. Biotechnol. 259, 148-159.
    Pubmed KoreaMed CrossRef
  38. Hagedorn, P. H., Yakimov, V., Ottosen, S., Kammler, S., Nielsen, N. F., Hog, A. M., Hedtjarn, M., Meldgaard, M., Moller, M. R., Orum, H., Koch, T. and Lindow, M. (2013) Hepatotoxic potential of therapeutic oligonucleotides can be predicted from their sequence and modification pattern. Nucleic Acid Ther. 23, 302-310.
    Pubmed KoreaMed CrossRef
  39. Hammond, S. M., Sergeeva, O. V., Melnikov, P. A., Goli, L., Stoodley, J., Zatsepin, T. S., Stetsenko, D. A. and Wood, M. J. A. (2021) Mesyl phosphoramidate oligonucleotides as potential splice-switching agents: impact of backbone structure on activity and intracellular localization. Nucleic Acid Ther. 31, 190-200.
    Pubmed CrossRef
  40. Henry, S., Stecker, K., Brooks, D., Monteith, D., Conklin, B. and Bennett, C. F. (2000) Chemically modified oligonucleotides exhibit decreased immune stimulation in mice. J. Pharmacol. Exp. Ther. 292, 468-479.
    Pubmed
  41. Hua, Y., Vickers, T. A., Baker, B. F., Bennett, C. F. and Krainer, A. R. (2007) Enhancement of SMN2 exon 7 inclusion by antisense oligonucleotides targeting the exon. PLoS Biol. 5, e73.
    Pubmed KoreaMed CrossRef
  42. Huang, L., Low, A., Damle, S. S., Keenan, M. M., Kuntz, S., Murray, S. F., Monia, B. P. and Guo, S. (2018) Antisense suppression of the nonsense mediated decay factor Upf3b as a potential treatment for diseases caused by nonsense mutations. Genome Biol. 19, 4.
    Pubmed KoreaMed CrossRef
  43. Iversen, P. L., Arora, V., Acker, A. J., Mason, D. H. and Devi, G. R. (2003) Efficacy of antisense morpholino oligomer targeted to c-myc in prostate cancer xenograft murine model and a Phase I safety study in humans. Clin. Cancer Res. 9, 2510-2519.
  44. Juliano, R. L. (2018) Intracellular trafficking and endosomal release of oligonucleotides: what we know and what we don't. Nucleic Acid Ther. 28, 166-177.
    Pubmed KoreaMed CrossRef
  45. Juliano, R. L., Wang, L., Tavares, F., Brown, E. G., James, L., Ariyarathna, Y., Ming, X., Mao, C. and Suto, M. (2018) Structure-activity relationships and cellular mechanism of action of small molecules that enhance the delivery of oligonucleotides. Nucleic Acids Res. 46, 1601-1613.
    Pubmed KoreaMed CrossRef
  46. Kamola, P. J., Kitson, J. D., Turner, G., Maratou, K., Eriksson, S., Panjwani, A., Warnock, L. C., Douillard Guilloux, G. A., Moores, K., Koppe, E. L., Wixted, W. E., Wilson, P. A., Gooderham, N. J., Gant, T. W., Clark, K. L., Hughes, S. A., Edbrooke, M. R. and Parry, J. D. (2015) In silico and in vitro evaluation of exonic and intronic off-target effects form a critical element of therapeutic ASO gapmer optimization. Nucleic Acids Res. 43, 8638-8650.
    Pubmed KoreaMed CrossRef
  47. Kim, J., Hu, C., Moufawad El Achkar, C., Black, L. E., Douville, J., Larson, A., Pendergast, M. K., Goldkind, S. F., Lee, E. A., Kuniholm, A., Soucy, A., Vaze, J., Belur, N. R., Fredriksen, K., Stojkovska, I., Tsytsykova, A., Armant, M., DiDonato, R. L., Choi, J., Cornelissen, L., Pereira, L. M., Augustine, E. F., Genetti, C. A., Dies, K., Barton, B., Williams, L., Goodlett, B. D., Riley, B. L., Pasternak, A., Berry, E. R., Pflock, K. A., Chu, S., Reed, C., Tyndall, K., Agrawal, P. B., Beggs, A. H., Grant, P. E., Urion, D. K., Snyder, R. O., Waisbren, S. E., Poduri, A., Park, P. J., Patterson, A., Biffi, A., Mazzulli, J. R., Bodamer, O., Berde, C. B. and Yu, T. W. (2019) Patient-customized oligonucleotide therapy for a rare genetic disease. N. Engl. J. Med. 381, 1644-1652.
    Pubmed KoreaMed CrossRef
  48. Kim, Y. J., Nomakuchi, T., Papaleonidopoulou, F., Yang, L., Zhang, Q. and Krainer, A. R. (2022) Gene-specific nonsense-mediated mRNA decay targeting for cystic fibrosis therapy. Nat. Commun. 13, 2978.
    Pubmed KoreaMed CrossRef
  49. Knerr, L., Prakash, T. P., Lee, R., Drury Iii, W. J., Nikan, M., Fu, W., Pirie, E., Maria, L. D., Valeur, E., Hayen, A., Ölwegård-Halvarsson, M., Broddefalk, J., Ämmälä, C., Østergaard, M. E., Meuller, J., Sundström, L., Andersson, P., Janzén, D., Jansson-Löfmark, R., Seth, P. P. and Andersson, S. (2021) Glucagon Like Peptide 1 Receptor agonists for targeted delivery of antisense oligonucleotides to pancreatic beta cell. J. Am. Chem. Soc. 143, 3416-3429.
    Pubmed CrossRef
  50. Koller, E., Vincent, T. M., Chappell, A., De, S., Manoharan, M. and Bennett, C. F. (2011) Mechanisms of single-stranded phosphorothioate modified antisense oligonucleotide accumulation in hepatocytes. Nucleic Acids Res. 39, 4795-4807.
    Pubmed KoreaMed CrossRef
  51. Kondow-McConaghy, H. M., Muthukrishnan, N., Erazo-Oliveras, A., Najjar, K., Juliano, R. L. and Pellois, J.-P. (2020) Impact of the endosomal escape activity of cell-penetrating peptides on the endocytic pathway. ACS Chem. Biol. 15, 2355-2363.
    Pubmed KoreaMed CrossRef
  52. Koshkin, A. A., Singh, S. K., Nielsen, P., Rajwanshi, V. K., Kumar, R., Meldgaard, M., Olsen, C. E. and 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.
    CrossRef
  53. Kotikam, V. and Rozners, E. (2020) Amide-modified RNA: using protein backbone to modulate function of short interfering RNAs. Acc. Chem. Res. 53, 1782-1790.
    Pubmed KoreaMed CrossRef
  54. Liang, X., Sun, H., Hsu, C.-W., Nichols, J. G., Vickers, T. A., De Hoyos, C. L. and Crooke, S. T. (2019a) Golgi-endosome transport mediated by M6PR facilitates release of antisense oligonucleotides from endosomes. Nucleic Acids Res. 48, 1372-1391.
    Pubmed KoreaMed CrossRef
  55. Liang, X., Sun, H., Shen, W., Wang, S., Yao, J., Migawa, M. T., Bui, H.-H., Damle, S. S., Riney, S., Graham, M. J., Crooke, R. M. and Crooke, S. T. (2017) Antisense oligonucleotides targeting translation inhibitory elements in 5′ UTRs can selectively increase protein levels. Nucleic Acids Res. 45, 9528-9546.
    Pubmed KoreaMed CrossRef
  56. Liang, X. H., Nichols, J. G., Hsu, C. W., Vickers, T. A. and Crooke, S. T. (2019b) mRNA levels can be reduced by antisense oligonucleotides via no-go decay pathway. Nucleic Acids Res. 47, 6900-6916.
    Pubmed KoreaMed CrossRef
  57. Liang, X. H., Shen, W., Sun, H., Kinberger, G. A., Prakash, T. P., Nichols, J. G. and Crooke, S. T. (2016a) Hsp90 protein interacts with phosphorothioate oligonucleotides containing hydrophobic 2'-modifications and enhances antisense activity. Nucleic Acids Res. 44, 3892-3907.
    Pubmed KoreaMed CrossRef
  58. Liang, X. H., Shen, W., Sun, H., Migawa, M. T., Vickers, T. A. and Crooke, S. T. (2016b) Translation efficiency of mRNAs is increased by antisense oligonucleotides targeting upstream open reading frames. Nat. Biotechnol. 34, 875-880.
    Pubmed CrossRef
  59. Liang, X. H., Sun, H., Nichols, J. G., Allen, N., Wang, S., Vickers, T. A., Shen, W., Hsu, C. W. and Crooke, S. T. (2018) COPII vesicles can affect the activity of antisense oligonucleotides by facilitating the release of oligonucleotides from endocytic pathways. Nucleic Acids Res. 46, 10225-10245.
    Pubmed KoreaMed CrossRef
  60. Liang, X. H., Sun, H., Shen, W. and Crooke, S. T. (2015) Identification and characterization of intracellular proteins that bind oligonucleotides with phosphorothioate linkages. Nucleic Acids Res. 43, 2927-2945.
    Pubmed KoreaMed CrossRef
  61. Lim, K. H., Han, Z., Jeon, H. Y., Kach, J., Jing, E., Weyn-Vanhentenryck, S., Downs, M., Corrionero, A., Oh, R., Scharner, J., Venkatesh, A., Ji, S., Liau, G., Ticho, B., Nash, H. and Aznarez, I. (2020) Antisense oligonucleotide modulation of non-productive alternative splicing upregulates gene expression. Nat. Commun. 11, 3501.
    Pubmed KoreaMed CrossRef
  62. Lima, W. F., Prakash, T. P., Murray, H. M., Kinberger, G. A., Li, W., Chappell, A. E., Li, C. S., Murray, S. F., Gaus, H., Seth, P. P., Swayze, E. E. and Crooke, S. T. (2012) Single-stranded siRNAs activate RNAi in animals. Cell 150, 883-894.
    Pubmed CrossRef
  63. Lima, W. F., Vickers, T. A., Nichols, J., Li, C. and Crooke, S. T. (2014) Defining the factors that contribute to on-target specificity of antisense oligonucleotides. PLoS One 9, e101752.
    Pubmed KoreaMed CrossRef
  64. Lönn, P., Kacsinta, A. D., Cui, X.-S., Hamil, A. S., Kaulich, M., Gogoi, K. and Dowdy, S. F. (2016) Enhancing endosomal escape for intracellular delivery of macromolecular biologic therapeutics. Sci. Rep. 6, 32301.
    Pubmed KoreaMed CrossRef
  65. Makley, L. N. and Gestwicki, J. E. (2013) Expanding the number of 'druggable' targets: non-enzymes and protein-protein interactions. Chem. Biol. Drug Des. 81, 22-32.
    Pubmed KoreaMed CrossRef
  66. McDonald, C. M., Shieh, P. B., Abdel-Hamid, H. Z., Connolly, A. M., Ciafaloni, E., Wagner, K. R., Goemans, N., Mercuri, E., Khan, N., Koenig, E., Malhotra, J., Zhang, W., Han, B. and Mendell, J. R. (2021) Open-label evaluation of eteplirsen in patients with Duchenne muscular dystrophy amenable to exon 51 skipping: PROMOVI trial. J. Neuromuscul. Dis. 8, 989-1001.
    Pubmed KoreaMed CrossRef
  67. Melton, D. A. (1985) Injected anti-sense RNAs specifically block messenger RNA translation in vivo. Proc. Natl. Acad. Sci. U. S. A. 82, 144-148.
    Pubmed KoreaMed CrossRef
  68. Migawa, M. T., Shen, W., Wan, W. B., Vasquez, G., Oestergaard, M. E., Low, A., De Hoyos, C. L., Gupta, R., Murray, S., Tanowitz, M., Bell, M., Nichols, J. G., Gaus, H., Liang, X. H., Swayze, E. E., Crooke, S. T. and Seth, P. P. (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.
    Pubmed KoreaMed CrossRef
  69. Miller, C. M., Donner, A. J., Blank, E. E., Egger, A. W., Kellar, B. M., Østergaard, M. E., Seth, P. P. and Harris, E. N. (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.
    Pubmed KoreaMed CrossRef
  70. Miller, P. S., Yano, J., Yano, E., Carroll, C., Jayaraman, K. and Ts'o, P. O. (1979) Nonionic nucleic acid analogues. Synthesis and characterization of dideoxyribonucleoside methylphosphonates. Biochemistry 18, 5134-5143.
    Pubmed CrossRef
  71. Miroshnichenko, S. K., Patutina, O. A., Burakova, E. A., Chelobanov, B. P., Fokina, A. A., Vlassov, V. V., Altman, S., Zenkova, M. A. and Stetsenko, D. A. (2019) Mesyl phosphoramidate antisense oligonucleotides as an alternative to phosphorothioates with improved biochemical and biological properties. Proc. Natl. Acad. Sci. U. S. A. 116, 1229-1234.
    Pubmed KoreaMed CrossRef
  72. Monia, B. P., Lesnik, E. A., Gonzalez, C., Lima, W. F., McGee, D., Guinosso, C. J., Kawasaki, A. M., Cook, P. D. and Freier, S. M. (1993) Evaluation of 2'-modified oligonucleotides containing 2'-deoxy gaps as antisense inhibitors of gene expression. J. Biol. Chem. 268, 14514-14522.
    Pubmed CrossRef
  73. Mutisya, D., Hardcastle, T., Cheruiyot, S. K., Pallan, P. S., Kennedy, S. D., Egli, M., Kelley, M. L., Smith, A. V. B. and Rozners, E. (2017) Amide linkages mimic phosphates in RNA interactions with proteins and are well tolerated in the guide strand of short interfering RNAs. Nucleic Acids Res. 45, 8142-8155.
    Pubmed KoreaMed CrossRef
  74. Nomakuchi, T. T., Rigo, F., Aznarez, I. and Krainer, A. R. (2016) Antisense oligonucleotide-directed inhibition of nonsense-mediated mRNA decay. Nat. Biotechnol. 34, 164-166.
    Pubmed KoreaMed CrossRef
  75. Obika, S., Nanbu, D., Hari, Y., Morio, K., In, Y., Ishida, T. and Imanishi, T. (1997) Synthesis of 2′-O,4′-C-methyleneuridine and -cytidine. Novel bicyclic nucleosides having a fixed C3, -endo sugar puckering. Tetrahedron Lett. 38, 8735-8738.
    CrossRef
  76. Ørom, U. A., Kauppinen, S. and Lund, A. H. (2006) LNA-modified oligonucleotides mediate specific inhibition of microRNA function. Gene 372, 137-141.
    Pubmed CrossRef
  77. Patutina, O. A., Gaponova Miroshnichenko, S. K., Sen'kova, A. V., Savin, I. A., Gladkikh, D. V., Burakova, E. A., Fokina, A. A., Maslov, M. A., Shmendel, E. V., Wood, M. J. A., Vlassov, V. V., Altman, S., Stetsenko, D. A. and Zenkova, M. A. (2020) Mesyl phosphoramidate backbone modified antisense oligonucleotides targeting miR-21 with enhanced in vivo therapeutic potency. Proc. Natl. Acad. Sci. U. S. A. 117, 32370-32379.
    Pubmed KoreaMed CrossRef
  78. Plavec, J., Tong, W. and Chattopadhyaya, J. (1993) How do the gauche and anomeric effects drive the pseudorotational equilibrium of the pentofuranose moiety of nucleosides? J. Am. Chem. Soc. 115, 9734-9746.
    CrossRef
  79. Prakash, T. P., Graham, M. J., Yu, J., Carty, R., Low, A., Chappell, A., Schmidt, K., Zhao, C., Aghajan, M., Murray, H. F., Riney, S., Booten, S. L., Murray, S. F., Gaus, H., Crosby, J., Lima, W. F., Guo, S., Monia, B. P., Swayze, E. E. and Seth, P. P. (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.
    Pubmed KoreaMed CrossRef
  80. Sands, H., Gorey-Feret, L. J., Cocuzza, A. J., Hobbs, F. W., Chidester, D. and Trainor, G. L. (1994) Biodistribution and metabolism of internally 3H-labeled oligonucleotides. I. Comparison of a phosphodiester and a phosphorothioate. Mol. Pharmacol. 45, 932-943.
  81. Servais, L., Mercuri, E., Straub, V., Guglieri, M., Seferian, A. M., Scoto, M., Leone, D., Koenig, E., Khan, N., Dugar, A., Wang, X., Han, B., Wang, D. and Muntoni, F. (2022) Long-term safety and efficacy data of golodirsen in ambulatory patients with Duchenne muscular dystrophy amenable to exon 53 skipping: a first-in-human, multicenter, two-part, open-label, phase 1/2 trial. Nucleic Acid Ther. 32, 29-39.
    Pubmed KoreaMed CrossRef
  82. Seth, P. P., Siwkowski, A., Allerson, C. R., Vasquez, G., Lee, S., Prakash, T. P., Wancewicz, E. V., Witchell, D. and Swayze, E. E. (2009a) Short antisense oligonucleotides with novel 2'-4' conformationaly restricted nucleoside analogues show improved potency without increased toxicity in animals. J. Med. Chem. 52, 10-13.
    Pubmed CrossRef
  83. Seth, P. P., Siwkowski, A., Allerson, C. R., Vasquez, G., Lee, S., Prakash, T. P., Wancewicz, E. V., Witchell, D. and Swayze, E. E. (2009b) Short antisense oligonucleotides with novel 2'-4' conformationaly restricted nucleoside analogues show improved potency without increased toxicity in animals. J. Med. Chem. 52, 10-13.
    Pubmed CrossRef
  84. Shen, W., De Hoyos, C. L., Migawa, M. T., Vickers, T. A., Sun, H., Low, A., Bell, T. A., Rahdar, M., Mukhopadhyay, S., Hart, C. E., Bell, M., Riney, S., Murray, S. F., Greenlee, S., Crooke, R. M., Liang, X. H., Seth, P. P. and Crooke, S. T. (2019) Chemical modification of PS-ASO therapeutics reduces cellular protein-binding and improves the therapeutic index. Nat. Biotechnol. 37, 640-650.
    Pubmed CrossRef
  85. Sheng, L., Rigo, F., Bennett, C. F., Krainer, A. R. and Hua, Y. (2020) Comparison of the efficacy of MOE and PMO modifications of systemic antisense oligonucleotides in a severe SMA mouse model. Nucleic Acids Res. 48, 2853-2865.
    Pubmed KoreaMed CrossRef
  86. Song, J. J., Smith, S. K., Hannon, G. J. and Joshua-Tor, L. (2004) Crystal structure of Argonaute and its implications for RISC slicer activity. Science 305, 1434-1437.
    Pubmed CrossRef
  87. Stec, W. J., Zon, G. and Egan, W. (1984) Automated solid-phase synthesis, separation, and stereochemistry of phosphorothioate analogs of oligodeoxyribonucleotides. J. Am. Chem. Soc. 106, 6077-6079.
    CrossRef
  88. Stockert, R. J. (1995) The asialoglycoprotein receptor: relationships between structure, function, and expression. Physiol. Rev. 75, 591-609.
    Pubmed CrossRef
  89. Summerton, J. and Weller, D. (1997) Morpholino antisense oligomers: design, preparation, and properties. Antisense Nucleic Acid Drug Dev. 7, 187-195.
    Pubmed CrossRef
  90. Swayze, E. E., Siwkowski, A. M., Wancewicz, E. V., Migawa, M. T., Wyrzykiewicz, T. K., Hung, G., Monia, B. P. and Bennett, C. F. (2007) Antisense oligonucleotides containing locked nucleic acid improve potency but cause significant hepatotoxicity in animals. Nucleic Acids Res. 35, 687-700.
    Pubmed KoreaMed CrossRef
  91. Taylor, R. E. and Zahid, M. (2020) Cell penetrating peptides, novel vectors for gene therapy. Pharmaceutics 12, 225.
    Pubmed KoreaMed CrossRef
  92. Teplova, M., Minasov, G., Tereshko, V., Inamati, G. B., Cook, P. D., Manoharan, M. and Egli, M. (1999) Crystal structure and improved antisense properties of 2'-O-(2-methoxyethyl)-RNA. Nat. Struct. Biol. 6, 535-539.
    Pubmed KoreaMed CrossRef
  93. Vickers, T. A. and Crooke, S. T. (2016) Development of a quantitative BRET affinity assay for nucleic acid-protein interactions. PLoS One 11, e0161930.
    Pubmed KoreaMed CrossRef
  94. Vickers, T. A., Rahdar, M., Prakash, T. P. and Crooke, S. T. (2019) Kinetic and subcellular analysis of PS-ASO/protein interactions with P54nrb and RNase H1. Nucleic Acids Res. 47, 10865-10880.
    Pubmed KoreaMed CrossRef
  95. Vickers, T. A., Wyatt, J. R., Burckin, T., Bennett, C. F. and Freier, S. M. (2001) Fully modified 2' MOE oligonucleotides redirect polyadenylation. Nucleic Acids Res. 29, 1293-1299.
    Pubmed KoreaMed CrossRef
  96. Volpi, S., Cancelli, U., Neri, M. and Corradini, R. (2020) Multifunctional delivery systems for peptide nucleic acids. Pharmaceuticals (Basel) 14, 14.
    Pubmed KoreaMed CrossRef
  97. Wagner, K. R., Kuntz, N. L., Koenig, E., East, L., Upadhyay, S., Han, B. and Shieh, P. B. (2021) Safety, tolerability, and pharmacokinetics of casimersen in patients with Duchenne muscular dystrophy amenable to exon 45 skipping: a randomized, double-blind, placebo-controlled, dose-titration trial. Muscle Nerve 64, 285-292.
    Pubmed KoreaMed CrossRef
  98. Wang, L., Ariyarathna, Y., Ming, X., Yang, B., James, L. I., Kreda, S. M., Porter, M., Janzen, W. and Juliano, R. L. (2017) A novel family of small molecules that enhance the intracellular delivery and pharmacological effectiveness of antisense and splice switching oligonucleotides. ACS Chem. Biol. 12, 1999-2007.
    Pubmed KoreaMed CrossRef
  99. Ward, A. J., Norrbom, M., Chun, S., Bennett, C. F. and Rigo, F. (2014) Nonsense-mediated decay as a terminating mechanism for antisense oligonucleotides. Nucleic Acids Res. 42, 5871-5879.
    Pubmed KoreaMed CrossRef
  100. Wu, H., Lima, W. F., Zhang, H., Fan, A., Sun, H. and Crooke, S. T. (2004) Determination of the role of the human RNase H1 in the pharmacology of DNA-like antisense drugs. J. Biol. Chem. 279, 17181-17189.
    Pubmed CrossRef
  101. Yanai, H., Chiba, S., Ban, T., Nakaima, Y., Onoe, T., Honda, K., Ohdan, H. and Taniguchi, T. (2011) Suppression of immune responses by nonimmunogenic oligodeoxynucleotides with high affinity for high-mobility group box proteins (HMGBs). Proc. Natl. Acad. Sci. U. S. A. 108, 11542-11547.
    Pubmed KoreaMed CrossRef
  102. Yang, B., Ming, X., Cao, C., Laing, B., Yuan, A., Porter, M. A., Hull-Ryde, E. A., Maddry, J., Suto, M., Janzen, W. P. and Juliano, R. L. (2015) High-throughput screening identifies small molecules that enhance the pharmacological effects of oligonucleotides. Nucleic Acids Res. 43, 1987-1996.
    Pubmed KoreaMed CrossRef
  103. Yoshida, T., Morihiro, K., Naito, Y., Mikami, A., Kasahara, Y., Inoue, T. and Obika, S. (2022) Identification of nucleobase chemical modifications that reduce the hepatotoxicity of gapmer antisense oligonucleotides. Nucleic Acids Res. 50, 7224-7234.
    Pubmed KoreaMed CrossRef
  104. Zamecnik, P. C. and Stephenson, M. L. (1978) Inhibition of Rous sarcoma virus replication and cell transformation by a specific oligodeoxynucleotide. Proc. Natl. Acad. Sci. U. S. A. 75, 280-284.
    Pubmed KoreaMed CrossRef
  105. Zhang, L., Liang, X. H., De Hoyos, C. L., Migawa, M., Nichols, J. G., Freestone, G., Tian, J., Seth, P. P. and Crooke, S. T. (2022) The combination of mesyl-phosphoramidate inter-nucleotide linkages and 2'-O-methyl in selected positions in the antisense oligonucleotide enhances the performance of RNaseH1 active PS-ASOs. Nucleic Acid Ther. 32, 401-411.
    Pubmed KoreaMed CrossRef
  106. Zimmermann, T. S., Karsten, V., Chan, A., Chiesa, J., Boyce, M., Bettencourt, B. R., Hutabarat, R., Nochur, S., Vaishnaw, A. and Gollob, J. (2017) Clinical proof of concept for a novel hepatocyte-targeting GalNAc-siRNA conjugate. Mol. Ther. 25, 71-78.
    Pubmed KoreaMed CrossRef


This Article


Cited By Articles

Services
Social Network Service

e-submission

Archives