2023 Impact Factor
Since the antisense oligonucleotide (ASO) concept (Zamecnik and Stephenson, 1978) and RNA interference mechanism (Fire
Table 1 Approved ASO drugs
Drug | Approval year | Target indication | Target gene | Mode of action | Chemistry |
---|---|---|---|---|---|
Formivirsen | 1998 | CMV retinitis | CMV | RNase H1 | PS-ODN |
Mipomersen | 2013 | HoFH | ApoB-100 | RNase H1 | PS-MOE gapmer |
Nusinersen | 2016 | SMA | SMN2 intron 7 | Exon inclusion | PS-MOE |
Eteplirsen | 2016 | DMD | Dystrophin exon 51 | Exon skipping | PMO |
Inotersen | 2018 | hATTR | TTR | RNase H1 | PS-MOE gapmer |
Golodirsen | 2019 | DMD | Dystrophin exon 53 | Exon skipping | PMO |
Volanesorsen | 2019 | FCS | ApoC-III | RNase H1 | PS-MOE gapmer |
Viltolarsen | 2020 | DMD | Dystrophin exon 53 | Exon skipping | PMO |
Casimersen | 2021 | DMD | Dystrophin exon 45 | Exon skipping | PMO |
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
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.
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).
Among them, however, only four chemical classes, including phosphorothioate (PS) oligodeoxynucleotides (ODNs), PS-2′-
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).
The first breakthrough innovation for ASO drug-likeness was the introduction of a PS linkage (Fig. 1) (Stec
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
As mentioned above, the C3′-
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
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
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
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
Spinal muscular atrophy is caused by homozygous loss of function of the survival of motor neuron 1 gene (
DMD is a rapidly progressing neuromuscular disorder. Mutations in the
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
Table 2 Modes of action and relevant chemistry designs of ASOs
Enzymatic degradation | Occupancy-only | ||
---|---|---|---|
Alternative splicing | Steric blocking | ||
Downregulation | RNase H-mediated | NMD | NGD |
AGO2-mediated | Translation arrest | ||
5′-cap inhibition | |||
Altering the polyadenylation site | |||
Upregulation | Exon inclusion | miRNA function inhibition | |
Exon skipping | Modulation of uORF/TIE | ||
Inhibition of NMD | |||
Chemistry | RNase H: DNA (w/wo PS linkage) or gapmer | Fully 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
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
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
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
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
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
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
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
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 protein binding is the main factor for improving absorption and distribution by suppressing renal clearance (Gaus
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
Asialoglycoprotein receptor is highly expressed on hepatocytes (Stockert, 1995). GalNAc is a ligand of this receptor and has been conjugated to an ASO (Prakash
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
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
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
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
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
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
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).
Recently, Yoshida
Alkylphosphonates: While the methyl phosphonate linkage (Fig. 1) was introduced long ago (Miller
Mesyl phosphoramidate (MsPA): Oligonucleotide derivatives with an MsPA (Fig. 1) linkage, the synthetic method of which was recently developed (Chelobanov
Amides: Following the early attempts to replace PO with amide (1 in Fig. 5) (De Mesmaeker
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
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.
The author is the founder and CEO of Qmine Co., Ltd.