Fact-checked by Grok 2 weeks ago

Gapmer

A gapmer is a chimeric antisense oligonucleotide (ASO) consisting of a central DNA segment flanked by chemically modified RNA-like wings, engineered to hybridize with target messenger RNA (mRNA) and induce its degradation via RNase H-mediated cleavage for selective gene silencing. This design leverages the DNA gap—typically 7–12 deoxynucleotides long—to form a substrate for the RNase H enzyme, while the flanking wings (usually 2–7 nucleotides each) incorporate modifications such as 2'-O-methyl (2'-OMe), 2'-O-methoxyethyl (2'-MOE), or locked nucleic acids (LNAs) to confer resistance to nuclease degradation and enhance binding specificity and affinity. The gapmer approach originated in the 1980s as an evolution of early antisense technologies, with foundational work by researchers like Inoue et al. demonstrating improved efficacy through 2'-O-modified phosphorothioate ASOs. Gapmers function by binding complementary mRNA sequences in a sequence-specific manner, recruiting endogenous RNase H1 to cleave the RNA strand within the DNA-RNA , leading to mRNA and reduced protein expression; this mechanism is particularly effective for targeting gain-of-function in genetic disorders. Compared to other classes like steric blockers or modulators, gapmers offer potent and durable knockdown (often lasting weeks after a single dose) due to their reliance on cellular RNase H machinery, though they can pose risks of off-target effects from non-specific hybridization or immune activation if not optimized. Chemical backbone modifications, such as phosphorothioate linkages throughout the molecule, further improve by increasing and tissue distribution, particularly to the liver and . Therapeutically, gapmers have advanced from research tools for target validation to approved drugs, including mipomersen (Kynamro), approved in 2013 but discontinued in 2019 for , which targets mRNA, and inotersen (Tegsedi) for hereditary , both utilizing 2'-MOE gapmer designs. More recent approvals, such as tofersen (Qalsody) for SOD1-related (ALS) in 2023 and olezarsen (Tryngolza) for familial chylomicronemia in 2025, highlight their potential in monogenic diseases by reducing toxic protein levels. Ongoing innovations address design challenges like allele-specific silencing for heterozygous mutations, with mixmer variants (alternating RNA-DNA segments) showing up to 10-fold improved specificity over classical gapmers in models of . Despite successes, limitations including delivery barriers beyond the liver and potential necessitate continued refinements in chemistry and conjugation strategies.

Structure and Design

Core Architecture

A gapmer is a chimeric antisense oligonucleotide designed as a single-stranded molecule comprising a central DNA segment, known as the gap, typically consisting of 8-10 unmodified deoxynucleotides, flanked on both sides by shorter segments of modified nucleotides referred to as wings, usually 5-7 nucleotides each. This architecture allows the gapmer to hybridize to complementary target RNA while incorporating elements that enhance its therapeutic potential. The canonical gapmer motif is often represented in a 5-10-5 configuration, denoting five modified in the 5' wing, a ten-nucleotide DNA gap, and five modified in the 3' wing, though variations like 5-8-5 or 7-10-7 exist depending on the target and optimization needs. Throughout the entire structure, a phosphorothioate (PS) backbone modification replaces the non-bridging oxygen in the groups with , conferring resistance to degradation and improving pharmacokinetic properties such as cellular uptake and distribution. The basic structural notation is thus 5'-wing-DNA gap-wing-3', where the wings are chemically altered to boost hybridization stability without interfering with the gap's function. The central DNA gap provides a deoxyribonucleotide stretch that permits access by RNase H1 enzyme, while the flanking wings, often incorporating modifications such as locked nucleic acid (LNA) or 2'-O-methoxyethyl (2'-MOE) residues, increase the binding affinity to the target mRNA through enhanced thermodynamic stability of the duplex. This balanced design ensures effective target engagement in vivo, distinguishing gapmers from other antisense formats like fully modified oligonucleotides.

Chemical Modifications

Gapmers are chimeric antisense () featuring specific chemical modifications to enhance binding , nuclease resistance, and compatibility with RNase H1 enzymes. Early ASOs relied solely on phosphorothioate () backbone modifications for basic stability, but these uniform designs suffered from limited target and off-target effects. The to modern chimeric gapmers incorporated sugar and base modifications in the flanking regions, balancing high- binding with a central DNA-like gap for enzymatic cleavage, as exemplified in designs like the 5-10-5 where wings flank a 10-nucleotide DNA gap. The backbone of gapmers typically employs full PS linkages, where non-bridging oxygen atoms in the phosphodiester bonds are replaced by sulfur atoms. This modification imparts resistance to degradation by extracellular and intracellular nucleases, extending the of the ASO in biological fluids compared to unmodified phosphodiester backbones. PS linkages also facilitate cellular uptake via protein binding but can contribute to immune activation if not optimized. In the wing regions, sugar modifications such as (LNA), 2'-O-methoxyethyl (2'-MOE), or 2'-fluoro (2'-F) are introduced to increase the thermal stability of the RNA-DNA hybrid duplex. LNA constrains the sugar in a C3'-endo conformation via a between the 2'-O and 4'-C atoms, boosting the melting temperature (Tm) by 4-8°C per substitution in DNA-RNA duplexes and enabling shorter with high specificity. 2'-MOE replaces the 2'-OH with a methoxyethyl group, enhancing Tm by approximately 2°C per modification while improving resistance and reducing toxicity relative to unmodified . Similarly, 2'-F substitutions, where replaces the 2'-OH, provide a Tm increase of about 2.5°C per and confer substantial stability against exonucleases, though they require careful positioning to avoid . The central gap region consists of unmodified or minimally modified DNA nucleotides, typically with only PS backbone alterations, to maintain a deoxyribose-like conformation essential for recruitment and activation of RNase H1. This DNA segment, often 8-12 nucleotides long, ensures substrate recognition by the enzyme while avoiding steric hindrance from sugar modifications that could inhibit cleavage. Representative hybrid designs include LNA-DNA-LNA gapmers, where LNA wings flank a PS-DNA gap to achieve potent hybridization with minimal length, and MOE-DNA-MOE constructs, such as those in approved therapeutics, which combine moderate affinity gains with favorable pharmacokinetic profiles. These chimeric architectures represent a significant advancement over early PS-only ASOs, allowing tailored optimization of stability and target engagement.

Mechanism of Action

Target Recognition

Gapmers recognize their target mRNA through sequence-specific hybridization, primarily via Watson-Crick base pairing between the antisense oligonucleotide and complementary sequences. The central DNA gap region forms the core of this duplex, while the flanking modified wings provide high-affinity anchoring that stabilizes the overall structure and facilitates initial binding. This design ensures that the gapmer can effectively invade secondary structures in the target mRNA, positioning the DNA- hybrid for subsequent enzymatic recruitment. Sequence design principles for gapmers emphasize lengths of 18-25 nucleotides to balance potency, specificity, and manufacturability, typically comprising a central DNA gap of 8-10 nucleotides flanked by 3-5 modified nucleotides on each wing. Targeting strategies prioritize regions such as exon-intron junctions or the 3' untranslated region (3' UTR) to enhance specificity, as these sites often minimize cross-reactivity with alternative isoforms or non-target transcripts while maintaining accessibility for hybridization. For instance, designs avoiding highly conserved coding sequences in the 3' UTR can reduce unintended binding to homologous mRNAs. Chemical modifications, particularly (LNA) in the wings, play a crucial role in mitigating off-target binding by improving mismatch discrimination. LNA incorporation increases the thermal stability of perfectly matched duplexes while disproportionately destabilizing those with single mismatches, often by 3-4 kcal/ depending on the mismatch type and context (e.g., enhanced discrimination for C·C or A·G mismatches). This selective affinity allows gapmers to distinguish subtle sequence variations, such as single nucleotide polymorphisms, thereby reducing non-specific interactions across the . Thermodynamic considerations guide gapmer optimization through free energy (ΔG°) calculations for duplex formation, often using nearest-neighbor parameters adapted from : models to predict hybridization at physiological temperatures. The wings contribute significantly to the overall ΔG°, with LNA modifications raising the melting temperature (Tm) by approximately 3-8°C per residue, thereby lowering the of for matched while amplifying destabilization for mismatches (ΔΔG° up to 3.4 kcal/mol). This wing-driven ensures efficient capture without excessive off-target hybridization, as mismatches in wing regions are less tolerated than in the central .

RNase H-Mediated Degradation

Upon hybridization of the gapmer antisense oligonucleotide (ASO) to its complementary target mRNA, the central DNA gap region forms an RNA-DNA heteroduplex that serves as a substrate for endogenous RNase H enzymes, primarily recruiting RNase H1 to initiate degradation. This recruitment occurs in both the nucleus and cytoplasm, where RNase H1 binds to the hybrid with high specificity, recognizing the DNA-like phosphodiester backbone in the gap while the flanking modified wings protect the ASO from nucleases. The process is rate-limited by RNase H1 availability, typically taking around 40 minutes after hybridization to achieve cleavage. RNase H1 catalyzes endonucleolytic hydrolysis of the strand within the , cleaving the phosphodiester bonds via a divalent metal ion-dependent that activates a water . This results in RNA fragments bearing 5'- and 3'-hydroxyl termini, leaving the DNA strand of the gapmer intact for potential multiple rounds of activity or . The exhibits strict specificity for RNA-DNA hybrids, with cleavage efficiency influenced by hybrid length (optimal at 8-12 pairs in the gap) and minimal activity on RNA-RNA duplexes or single-stranded RNA. The cleaved mRNA fragments are rapidly processed and degraded by cellular exonucleases through intrinsic turnover pathways, leading to substantial reduction in target mRNA levels and subsequent protein knockdown, often achieving 70-90% in therapeutic contexts. This is 2- to 4-fold faster than the natural mRNA decay rate, ensuring potent . In contrast to RNase H2, which is chromatin-associated and primarily functions in and repair without participating in ASO-mediated , RNase H1 is the key for cytoplasmic and actions in gapmer therapeutics due to its localization and hybrid-cleavage proficiency.

Advantages

Potency and Specificity

Gapmers demonstrate high potency in silencing target genes, frequently achieving subnanomolar IC50 values in assays, such as 0.4 nM for (LNA)-modified gapmers targeting VR1 mRNA, owing to their enhanced binding affinity and efficient RNase H-mediated cleavage of the RNA-DNA hybrid. This allows for substantially lower dosing compared to first-generation antisense , as the catalytic nature of RNase H degradation enables multiple turnover events per gapmer . The specificity of gapmers is bolstered by their flanking wing regions, which incorporate modifications like 2'-O-methoxyethyl or LNA that stabilize the duplex with perfect-match targets while destabilizing mismatched hybrids, resulting in over 100-fold discrimination between fully complementary sequences and those with a single mismatch. For instance, rationally designed gapmers targeting single-nucleotide polymorphisms exhibit this pronounced selectivity, minimizing off-target effects in allele-specific applications. In comparison to phosphorothioate-only , which typically require higher concentrations for modest knockdown (e.g., IC50 values around 70 ), gapmers achieve 80-95% reduction in target protein expression at 10-50 doses in cellular models. This superior efficiency, coupled with low immune activation due to reduced protein binding from the modified wings, supports robust in diverse tissues including the liver and .

Stability Enhancements

The phosphorothioate (PS) backbone modification in gapmers replaces the non-bridging oxygen in the phosphate backbone with sulfur, conferring substantial resistance to degradation by exonucleases and endonucleases that rapidly break down unmodified DNA oligonucleotides. This enhancement significantly prolongs the stability of gapmers in biological environments, with intracellular and tissue half-lives typically ranging from 2 to 4 weeks, compared to hours for unmodified counterparts. The flanking wing regions of gapmers, often incorporating locked nucleic acid (LNA) or 2'-O-methoxyethyl (MOE) modifications, provide additional protection against serum nucleases, further bolstering overall durability. LNA-modified wings, in particular, create a rigid bicyclic structure that shields the oligonucleotide from enzymatic attack, resulting in up to a 10-fold increase in serum half-life relative to standard DNA oligonucleotides. MOE wings similarly contribute by sterically hindering nuclease access while maintaining compatibility with the central DNA gap for RNase H recruitment. Pharmacokinetic profiles of gapmers reflect these stability gains, with rapid distribution from to tissues leading to high accumulation in the liver and , where concentrations can persist for weeks. Clearance occurs slowly via urinary of short-chain metabolites and proteolytic/nucleolytic within tissues, minimizing rapid elimination and supporting sustained exposure. This pharmacokinetic behavior underpins the feasibility of infrequent dosing, such as weekly or monthly subcutaneous administrations, as demonstrated in approved gapmer therapeutics like and volanesorsen.

Therapeutics

Mipomersen (Kynamro)

, marketed as Kynamro, represents the first gapmer antisense oligonucleotide approved for clinical use, targeting (ApoB) mRNA to address severe . Developed by Pharmaceuticals (now ) in collaboration with , it received FDA approval on January 29, 2013, as an adjunct to maximally tolerated lipid-lowering therapy and for reducing cholesterol (LDL-C), ApoB, total , and non-HDL cholesterol in adults with homozygous (HoFH), a rare characterized by extremely high LDL-C levels due to mutations in the . Structurally, is a 20-nucleotide (20-mer) second-generation gapmer with a 5-10-5 design, featuring 2'-O-methoxyethyl (2'-MOE) modifications in the flanking wings for enhanced stability and a central gap to recruit RNase H for target degradation, all linked by a phosphorothioate backbone. It specifically binds to 20-21 of ApoB mRNA in the liver, inducing RNase H-mediated cleavage and reducing ApoB-100 protein synthesis, which in turn decreases hepatic production of (VLDL) and circulating LDL particles. This mechanism results in LDL-C reductions of 25-40% in HoFH patients, with the recommended dosing regimen being 200 mg administered subcutaneously once weekly. In pivotal phase 3 clinical trials, such as the randomized, double-blind, placebo-controlled study involving 34 HoFH patients, mipomersen demonstrated a mean 36% reduction in LDL-C from baseline after 26 weeks, compared to a 13% increase in the placebo group, alongside significant decreases in ApoB (38%) and non-HDL-C (35%). However, the therapy carries a black box warning for hepatotoxicity, with elevations in alanine aminotransferase (ALT) ≥3 times the upper limit of normal (ULN) occurring in up to 12% of patients and increases in hepatic fat content observed via magnetic resonance imaging. Due to these safety concerns, including injection-site reactions and flu-like symptoms leading to high discontinuation rates, mipomersen has seen limited clinical adoption; marketing was discontinued worldwide, with the FDA withdrawing approval in 2019 as the product was no longer marketed, and it is no longer available as of 2025.

Volanesorsen (Waylivra)

Volanesorsen, marketed as Waylivra, is a second-generation antisense oligonucleotide (ASO) developed by Ionis Pharmaceuticals and its affiliate Akcea Therapeutics as the first targeted therapy for familial chylomicronemia syndrome (FCS), a rare genetic disorder characterized by severe hypertriglyceridemia and recurrent pancreatitis due to impaired lipoprotein lipase activity. As a 20-nucleotide gapmer with 2'-O-methoxyethyl (2'-MOE) modifications on the wings and phosphorothioate backbone linkages, it specifically binds to apolipoprotein C-III (ApoC-III) mRNA in the liver, recruiting RNase H enzymes to cleave the target mRNA and thereby reducing ApoC-III protein production, which inhibits triglyceride clearance. Prior to volanesorsen, no approved pharmacologic treatments existed for FCS, leaving patients reliant on strict dietary management to mitigate life-threatening complications. The () granted conditional marketing authorization for volanesorsen on May 3, 2019, as an adjunct to diet for adult patients with genetically confirmed FCS at high risk of who are unresponsive to standard -lowering therapies. The recommended regimen involves 300 mg subcutaneous injections once weekly for the first , followed by every two weeks if levels are adequately reduced, with further adjustments at six and nine months based on response and safety monitoring. In the pivotal phase 3 APPROACH trial, involving 66 adults with FCS, volanesorsen achieved a mean 77% reduction in plasma levels from baseline at (from 2202 mg/dL to 485 mg/dL), compared to an 18% increase with , while also lowering ApoC-III by 84%. Additionally, 77% of treated patients reached levels below 750 mg/dL, versus 10% on , demonstrating substantial efficacy in this ultra-rare population with historically untreatable severe exceeding 1000 mg/dL. A key challenge with volanesorsen is its association with , observed in up to 48% of patients in clinical trials, where platelet counts fell below 100,000 per microliter, though severe cases (<25,000 per microliter) were rare and reversible upon discontinuation. This class effect of 2'-MOE ASOs necessitates mandatory baseline and ongoing platelet monitoring every two to four weeks, with dose interruptions or reductions if counts drop below 75,000 per microliter, and permanent discontinuation if below 50,000 per microliter, to prevent risks. Other common adverse effects include injection-site reactions in over 60% of patients, but the overall risk-benefit profile supports its use in this high-need population under a conditional approval requiring post-marketing studies to confirm long-term and efficacy.

Inotersen (Tegsedi)

Inotersen, marketed as Tegsedi, is a 20-mer antisense oligonucleotide gapmer modified with phosphorothioate linkages and 2'-O-methoxyethyl (2'-MOE) wings flanking a central DNA gap, designed to target transthyretin (TTR) mRNA in the liver. By binding to complementary sequences on TTR mRNA, inotersen recruits RNase H to cleave the target RNA, thereby reducing TTR protein production and limiting the formation of amyloid deposits associated with hereditary transthyretin-mediated (hATTR) amyloidosis. This approach addresses the polyneuropathy manifestation of hATTR, a progressive neurodegenerative condition caused by mutant TTR misfolding and extracellular deposition. The U.S. Food and Drug Administration (FDA) approved inotersen in October 2018 for the treatment of polyneuropathy in adults with hATTR amyloidosis, marking it as the second approved therapy for this indication following patisiran. Administered as a 300 mg subcutaneous injection once weekly, inotersen's formulation supports stable delivery via this route, enabling outpatient use. In the pivotal phase 3 NEURO-TTR study, a randomized, double-blind, placebo-controlled trial involving 172 patients with stage 1 or 2 hATTR polyneuropathy, inotersen demonstrated a 50% slower progression of neurologic impairment compared to placebo, as measured by the modified Neuropathy Impairment Score plus 7 (mNIS+7) over 66 weeks. The study also showed significant improvements in quality of life, with serum TTR levels reduced by approximately 80% from baseline, correlating with decreased amyloid deposition potential. Post-approval, inotersen is distributed exclusively through the Tegsedi Risk and (REMS) program to mitigate serious risks, including and . The REMS requires prescriber , patient , and mandatory —such as weekly platelet counts and monthly serum creatinine with —to detect early signs of these adverse events, which occurred in 20% and 3% of trial participants, respectively. This structured oversight ensures safe use while preserving access for eligible patients with hATTR .

Tofersen (Qalsody)

, marketed as Qalsody, is a 20-mer antisense with a 5-10-5 phosphorothioate backbone modification, designed as a gapmer to bind mRNA and induce its RNase H-mediated degradation in patients with (ALS) caused by 1 () gene mutations. The U.S. granted accelerated approval for on April 25, 2023, based on its reduction of neurofilament light chain () levels as a surrogate for neurodegeneration in SOD1-ALS adults. This approval marks the first for a specific genetic form of ALS, which accounts for about 2% of cases. Clinical data from the phase 1/2 trial demonstrated that of at the 100 mg dose resulted in a mean 37% reduction in (CSF) protein concentrations from baseline at day 85, compared to minimal change with . The subsequent phase 3 VALOR trial, involving 137 participants, showed further reductions, including up to 40% decrease in CSF in slower-progressing subgroups and a 60% reduction in plasma NfL levels versus 20% with over 28 weeks, supporting target engagement despite no significant improvement in primary clinical endpoints like ALS Functional Rating Scale scores. is dosed intrathecally at 100 mg (15 mL) with three loading doses every 14 days, followed by maintenance every 28 days, allowing direct delivery to motor neurons. As of 2025, confirmation of clinical benefit is under evaluation in ongoing phase 3 trials, including the ATLAS study assessing in presymptomatic mutation carriers to determine if early intervention delays onset. programs have been extended to provide to a broader cohort of early-stage -ALS patients outside clinical trials, enhancing availability in regions permitting such initiatives. By reducing toxic protein accumulation, offers a pioneering disease-modifying approach for this monogenic subset, potentially slowing neurodegeneration.

Safety and Toxicity

Common Adverse Effects

Gapmer antisense oligonucleotides, particularly those administered subcutaneously, frequently cause local injection site reactions including , pain, tenderness, pruritus, and swelling, affecting 20-84% of patients. These reactions are generally mild to moderate, dose-dependent, and resolve without intervention, though they contribute to treatment discontinuation in a small subset of cases. Flu-like symptoms, such as pyrexia, , , , and , arise early in therapy due to immune activation via (TLR) stimulation by the phosphorothioate backbone of gapmers, with incidences ranging from 10-30% across clinical evaluations. The phosphorothioate modifications enhance stability but can trigger innate immune responses contributing to these transient effects. Hematological adverse effects, notably , occur in TTR-targeting gapmers such as inotersen, where severe reductions in platelet count (grade 3 or higher) affect approximately 3% of patients and necessitate close . These events typically manifest within the first few months and may resolve with dose adjustment or supportive care. Hepatic transaminase elevations, including increases in (ALT) and aspartate aminotransferase (), are observed in 12-16% of patients receiving liver-targeted gapmers like , often correlating with hepatic fat accumulation but rarely progressing to severe injury. Routine liver function is recommended to manage these reversible changes.

Long-Term Risks and Mitigation

One of the primary long-term risks associated with gapmer antisense oligonucleotides stems from the accumulation of their phosphorothioate () backbones in the kidney and , which can lead to chronic , including and renal tubular degeneration. This buildup occurs due to the chemical stability of PS modifications, resulting in prolonged tissue retention and potential for sustained exposure. Clinical observations in patients receiving long-term gapmer have reported as a rare but serious , often linked to immune complex deposition in renal tissues. Off-target effects represent another concern in extended gapmer use, where unintended interactions may alter patterns or trigger immune modulation, such as activation of the response pathway. These effects arise from partial complementarity between the gapmer and non-target RNAs, potentially leading to widespread changes in . In central nervous system-targeted gapmers like , recent 2025 analyses have noted elevations in neuroinflammatory markers during prolonged treatment, suggesting possible late-onset contributions to , though direct causation remains under investigation. To mitigate these risks, strategies include dose adjustments to minimize cumulative exposure while preserving efficacy, as higher doses exacerbate PS accumulation. (GalNAc) conjugation enhances liver-specific targeting, reducing off-target renal uptake and attenuating in preclinical models by promoting clearance through productive endocytic pathways. Regular monitoring of renal function, platelet counts, and inflammatory markers is recommended for long-term users to enable early detection and intervention. Regulatory bodies have issued class-wide warnings for in gapmer-based therapies, with approximately half of FDA-approved carrying precautions or black-box alerts for liver enzyme elevations and due to prolonged exposure. Ongoing research focuses on developing safer backbone modifications, such as phosphorodithioate linkages or methylphosphonate alternatives, and on understanding sequence-chemistry interplay in (as of October 2025), to further reduce accumulation and off-target liabilities without compromising potency.

References

  1. [1]
    Possibilities and limitations of antisense oligonucleotide therapies ...
    Jan 5, 2024 · Gapmers are single-stranded ASOs consisting of a DNA core flanked by modified RNA-based structures resistant to RNase H. Gapmers can be designed ...
  2. [2]
    Strategies to improve the design of gapmer antisense ... - Cell Press
    Jun 4, 2024 · Gapmer antisense oligonucleotides (ASOs) hold therapeutic promise for allele-specific silencing, but face challenges in distinguishing ...
  3. [3]
    The powerful world of antisense oligonucleotides: From bench to ...
    Mar 31, 2020 · The resulting structure is known as a “gapmer.” The “gapmer” approach was first shown by Inoue for a 2′OME PO ASO (Inoue et al., 1987), followed ...
  4. [4]
    Invention and Early History of Gapmers - PubMed
    Gapmers are antisense oligonucleotides composed of a central DNA segment flanked by nucleotides of modified chemistry ... first emerged in the 1980s, much work h
  5. [5]
    Advances in oligonucleotide drug delivery - Nature
    Aug 11, 2020 · This Review will provide an overview of oligonucleotide-based ... a | Gapmer antisense oligonucleotides (ASOs), consisting of a DNA ...
  6. [6]
    Review Clinical Applications of Single-Stranded Oligonucleotides
    Feb 3, 2021 · This review highlights key applications of single-stranded oligonucleotides that function in a sequence-dependent manner when applied to modulate precursor ( ...
  7. [7]
    The chemical evolution of oligonucleotide therapies of clinical utility
    As an alternative to the gapmer approach, modifications that adopt a DNA-like conformation can also be used to improve affinity and stability of RNase H ...Rnase H-Dependent Asos · Conjugate Mediated Delivery · Table 1
  8. [8]
    Phosphorothioate modified oligonucleotide–protein interactions - NIH
    May 1, 2020 · Acute hepatotoxicity of 2′ fluoro-modified 5-10-5 gapmer phosphorothioate oligonucleotides in mice correlates with intracellular protein ...
  9. [9]
    Towards next generation antisense oligonucleotides ...
    Aug 20, 2021 · We show that all PS linkages in a gapmer ASO can be replaced with MsPA without compromising chemical stability and RNA binding affinity but ...Materials And Methods · Aso In Vitro And In Vivo... · Results
  10. [10]
    Locked nucleic acid: modality, diversity, and drug discovery
    It is widely recognized that gapmer potency is affected by the target nucleobase sequence. This is usually attributed to the structure of the RNA in the target ...
  11. [11]
    Design of antisense oligonucleotides stabilized by locked nucleic ...
    Compared to the 18mer DNA the oligonucleotides containing LNA have an increased melting temperature of 1.5–4°C per LNA depending on the positions of the ...
  12. [12]
    Antisense technology: A review - ScienceDirect.com
    Because it fixes the furanose in a favorable conformation in effect “pre-organizing the duplex,” 2ʹ-MOE increases the Tm per nucleotide by more than 2 degrees.
  13. [13]
    Chemistry, structure and function of approved oligonucleotide ...
    The gapmer ASO design provides a solution to this conundrum: the central PS-DNA window promotes cleavage by RNase H and 2′-O-modified wings afford stability (59 ...Oligonucleotide Therapeutics... · Splice Site Switching... · Rnai Therapeutics...
  14. [14]
    Fluorinated Nucleotide Modifications Modulate Allele Selectivity of ...
    We showed that replacing DNA nucleotides in the gap region of an ASO with other chemical modification can improve allele selectivity.Original Article · Results And Discussion · Human Rnase H1 Rna Cleavage...
  15. [15]
    Evaluation of off‐target effects of gapmer antisense oligonucleotides ...
    ASOs are synthetic DNA/RNA‐like single‐stranded oligonucleotides that bind to RNA through sequence‐specific Watson‐Crick base pairing. ... wings and DNA in ...
  16. [16]
  17. [17]
    Specificity of oligonucleotide gene therapy (OGT) agents
    ASO - antisense oligonucleotide;. dgapmer - ASO consisting of internal DNA gap flanked with LNA modified nucleotides;. ePMO - phosphorodiamidate morpholino ...
  18. [18]
    Stability and Mismatch Discrimination of Locked Nucleic Acid–DNA ...
    A single LNA modification confines the local conformation of nucleotides, causing a smaller, less unfavorable entropic loss when the single strand is restricted ...
  19. [19]
    Likelihood of Nonspecific Activity of Gapmer Antisense ... - NIH
    ... LNA gapmers binding to matched and mismatched targets. Although they did not directly measure duplex free energies for the LNA gapmers, our results support ...
  20. [20]
    Molecular Mechanisms of Antisense Oligonucleotides - PMC - NIH
    (C) An effective RNase H1 activating ASO approximately doubles the intrinsic rate of cellular RNA degradation. (D) The concentration of RNAse H1 is rate ...Phases Of Antisense Drug... · Fig. 1 · Prehybridization Events
  21. [21]
    DNA Heteroduplex Cleavage by Human RNase H1 - PMC - NIH
    Ribonuclease H1 is a conserved enzyme that cleaves the RNA strand of RNA•DNA heteroduplexes, and has important functions in the nuclear and mitochondrial ...
  22. [22]
    BNANC Gapmers Revert Splicing and Reduce RNA Foci with ... - NIH
    While neither DMPK gapmer showed significant knockdown at the lowest dose, they both induced over 70% knockdown at 30 nM. Interestingly, the DMPK gapmers ...
  23. [23]
    RNase H1-Dependent Antisense Oligonucleotides Are Robustly ...
    RNase H1-dependent antisense oligonucleotides (ASOs) are active in reducing levels of both cytoplasmic mRNAs and nuclear retained RNAs.
  24. [24]
    O-methyl RNA, phosphorothioates and small interfering ... - PubMed
    An LNA gapmer was found to be the most efficient single-stranded antisense oligonucleotide, with an IC50 of 0.4 nM being 175-fold lower than that of commonly ...
  25. [25]
    Rational design of antisense oligonucleotides targeting single ...
    Aug 19, 2013 · The resulting oligonucleotides demonstrate >100-fold discrimination ... gapmer ASOs targeting SNPs associated with expanded CAG repeats (22).
  26. [26]
    Gapmer Antisense Oligonucleotides Suppress the Mutant Allele of ...
    Gapmer AONs targeting mRNA sequences, with a length of 22-mer and a DNA gap size of 8–10 nt gave the most efficient allele-specific silencing effect. The ...Missing: UTR | Show results with:UTR
  27. [27]
    Metabolic Stability and Targeted Delivery of Oligonucleotides
    ... 2'-fluoro content that yield significantly improved potency and duration in preclin. ... A balance of greater stability supplied by the 2'-O-MOE ...
  28. [28]
    Antisense oligonucleotides: a novel Frontier in pharmacological ...
    The first systemically approved ASO therapy was mipomersen, an antisense 20-mer phosphorothioate 2′-methoxy-ethoxy (MOE) gapmer, developed by Genzyme for ...
  29. [29]
    Antisense locked nucleic acids efficiently suppress BCR/ABL and ...
    Aug 4, 2006 · In addition, full LNA and mixed LNA-DNA oligonucleotides show high serum stability, up to 10-fold increase of the half-life in human serum ...
  30. [30]
    2' MOE – An ASO modification | IDT - Integrated DNA Technologies
    Oct 4, 2023 · Increase stability and reduce toxicity with nuclease-resistant mods and flexible chimeric designs ... Chemical evolution of antisense oligos (ASOs).
  31. [31]
    Tissue pharmacokinetics of antisense oligonucleotides - ScienceDirect
    Here, we report concentration-time and RNA knockdown profiles for a gapmer ASO with locked nucleic acid ribose chemistry in mouse liver, kidney, heart, and lung ...
  32. [32]
    Nephrotoxicity of marketed antisense oligonucleotide drugs - PMC
    The mechanism of ASO metabolism is primarily via nucleolytic breakdown by endo- and exonucleases at the tissue site wherein they are accumulated. Kidney ...
  33. [33]
  34. [34]
    [PDF] 203568Orig1s000 - accessdata.fda.gov
    While the LDL-C reduction with mipomersen was modest and there were issues with high discontinuation rates in the open-label extension trial, ...
  35. [35]
    Mipomersen (Kynamro) Approved by FDA - National Lipid Association
    Jan 30, 2013 · On January 29, the US FDA approved mipomersen sodium injection as an adjunct to treat LDL-C, Apo B, total cholesterol, and non HDL-C in patients with ...
  36. [36]
    Kynamro wins FDA approval for homozygous familial ... - Healio
    Jan 30, 2013 · The FDA has approved mipomersen sodium as an adjunct to lipid-lowering medications and a healthy diet to reduce LDL, apolipoprotein B, total ...
  37. [37]
    Emerging LDL therapies: Mipomersen—antisense oligonucleotide ...
    Mipomersen was recently approved by the Food and Drug Administration for the treatment of homozygous familial FH. FH is caused by inherited genetic defects ...<|control11|><|separator|>
  38. [38]
    Mipomersen Dosage Guide + Max Dose, Adjustments - Drugs.com
    Jun 30, 2025 · Usual Adult Dose for Hyperlipidemia: 200 mg subcutaneously once weekly on the same day each week.Dose Adjustments · Precautions · Other Comments
  39. [39]
    Mipomersen in Familial Hypercholesterolemia: An Update on Health ...
    Feb 21, 2022 · Findings in the mipomersen arm demonstrated reductions in LDL-C of 36% from baseline compared to a 13% increase in LDL-C levels in the placebo ...
  40. [40]
    [PDF] This label may not be the latest approved by FDA. For current ...
    In a subcutaneous carcinogenicity study in mice, mipomersen sodium was administered for up to 104 weeks at doses of 5, 20, 60 mg/kg/week. There were ...
  41. [41]
    Kynamro (mipomersen) FDA Approval History - Drugs.com
    FDA approval history for Kynamro (mipomersen) used to treat High Cholesterol, Familial Homozygous. Supplied by Kastle Therapeutics.
  42. [42]
    Akcea and Ionis announce approval of WAYLIVRA® (volanesorsen ...
    May 7, 2019 · WAYLIVRA is an antisense oligonucleotide drug designed by Ionis and co-developed by Akcea and Ionis to reduce the production of ApoC-III, a ...
  43. [43]
    [PDF] Assessment report - WAYLIVRA - European Medicines Agency
    Feb 28, 2019 · ISIS 304801 (volanesorsen) was identified as the optimal 2′-MOE antisense inhibitor of human apoC-. III on the basis of its consistent and ...
  44. [44]
    Treatment with Volanesorsen, a 2 - PubMed Central
    Volanesorsen is a second-generation antisense oligonucleotide (ASO) drug targeted to human APOC3 mRNA. The hybridization of volanesorsen to the cognate mRNA ...Aso Volanesorsen · Results · Concentration/effect...
  45. [45]
    Waylivra | European Medicines Agency (EMA)
    Waylivra is indicated as an adjunct to diet in adult patients with genetically confirmed familial chylomicronemia syndrome (FCS) and at high risk for ...Missing: 2018 | Show results with:2018
  46. [46]
    Volanesorsen and Triglyceride Levels in Familial Chylomicronemia ...
    Aug 7, 2019 · Another side effect was thrombocytopenia, which was defined as a platelet count below 140,000 per microliter. A recently reported natural ...
  47. [47]
    Inotersen Treatment for Patients with Hereditary Transthyretin ...
    Jul 4, 2018 · Inotersen improved the course of neurologic disease and quality of life in patients with hereditary transthyretin amyloidosis.
  48. [48]
    [PDF] 1 FULL PRESCRIBING INFORMATION - accessdata.fda.gov
    A total of 112 adult patients with polyneuropathy caused by hereditary transthyretin-mediated amyloidosis (hATTR) received TEGSEDI in Study 1 and 60 patients ...
  49. [49]
    [PDF] SPINRAZA (nusinersen) injection, for intrathecal use
    See full prescribing information for. SPINRAZA. SPINRAZA (nusinersen) injection, for intrathecal use. Initial U.S. Approval: 2016. INDICATIONS AND USAGE.
  50. [50]
    Fluoroscopic guidance for intrathecal delivery of nusinersen in ...
    Because antisense oligonucleotides do not cross the intact blood-brain barrier, nusinersen is administered by intrathecal injection. Treatment is comprised ...
  51. [51]
    [PDF] Spinraza – Prescribing Information
    1 Loading doses followed by 12 mg (5 mL) once every 6 months. Post-lumbar puncture syndrome has also been observed after administration of SPINRAZA. 6.2.
  52. [52]
    Nusinersen versus Sham Control in Later-Onset Spinal Muscular ...
    Feb 14, 2018 · The ENDEAR trial also showed greater improvements in infants who received treatment with nusinersen earlier in their disease course than in ...
  53. [53]
    Results from a phase 1 study of nusinersen (ISIS-SMNRx) in ...
    In mouse models of SMA, nusinersen enhanced exon 7 inclusion, increased SMN protein production, and improved function. ... fold over a ∼3-fold increase in total ...
  54. [54]
    [PDF] This label may not be the latest approved by FDA. For current ...
    Tofersen, an antisense oligonucleotide, is a 20-base residue (20-mer) 5-10-5 MOE gapmer mixed backbone oligonucleotide. Of the nineteen internucleotide ...
  55. [55]
    FDA approves treatment of ALS associated with a mutation in the ...
    Apr 25, 2023 · FDA approved Qalsody (tofersen) to treat patients with amyotrophic lateral sclerosis (ALS) associated with a mutation in the superoxide dismutase 1 (SOD1) gene ...
  56. [56]
    Tofersen (Qalsody®) - Amyotrophic Lateral Sclerosis
    Jul 31, 2025 · In April 2023, tofersen, marketed in the U.S. as Qalsody® (Biogen), was approved by the FDA for the treatment of SOD1-linked ALS. The SOD1 gene ...
  57. [57]
    Phase 1–2 Trial of Antisense Oligonucleotide Tofersen for SOD1 ALS
    Jul 8, 2020 · A 25% reduction in the CSF SOD1 protein concentration was estimated to exceed both the SOD1 assay variability and the longitudinal ...
  58. [58]
    Trial of Antisense Oligonucleotide Tofersen for SOD1 ALS
    Sep 21, 2022 · Tofersen reduced concentrations of SOD1 in CSF and of neurofilament light chains in plasma over 28 weeks but did not improve clinical end points and was ...
  59. [59]
    NCT04856982 | A Study of BIIB067 (Tofersen) Initiated in Clinically ...
    The primary objective of this study is to evaluate the efficacy of tofersen in presymptomatic adult carriers of a superoxide dismutase 1 (SOD1) mutation ...
  60. [60]
    Tofersen/Qalsody - International Alliance of ALS/MND Associations
    Since 2021, Biogen has “expanded eligibility for its ongoing early access program to all people with SOD1-ALS, in countries where such programs are permitted by ...
  61. [61]
    FDA approves QALSODY™ (tofersen) as the first treatment targeting ...
    Accelerated approval marks a scientific advancement in treatment of superoxide dismutase 1 (SOD1)-amyotrophic lateral sclerosis (ALS); Approval based on ...
  62. [62]
    Injection site reactions after subcutaneous oligonucleotide therapy
    When administered subcutaneously (sc), ONs cause a specific local reaction originating around the injection site, such as erythema, itching, discomfort and pain ...
  63. [63]
    Adverse Drug Reactions and Toxicity of the Food and Drug ... - NIH
    The major safety concerns for the FDA-approved ASO drugs are severe hepatotoxicity, renal toxicity, and hypersensitivity reactions, which are commonly ...
  64. [64]
    Considerations in the Preclinical Assessment of the Safety of ...
    ... effects that can manifest in different ways in the clinic, including injection site reactions, flu-like symptoms, and thrombocytopenia [72–75]. Therefore ...
  65. [65]
    [PDF] lik - accessdata.fda.gov
    KYNAMRO can cause elevations in transaminases (5.1). • Measure alanine aminotransferase (ALT), aspartate aminotransferase.
  66. [66]
    Off-target effects of oligonucleotides and approaches of preclinical ...
    ... Flu-like symptoms, such as fatigue, flu-like illness, chills, fever, joint pain, and myalgia, Liver injury, liver steatosis. Inotersen, SC, Vacuolation and ...
  67. [67]
    Nephrotoxicity of marketed antisense oligonucleotide drugs
    It has also been observed that ONDs cause adverse effects on liver and kidney characterised by glomerulonephritis, tubular degeneration and increased protein ...
  68. [68]
    Block or degrade? Balancing on- and off-target effects of antisense ...
    RNA-seq reveals activation of the interferon response by all ASOs and additional off-target effects on gene expression by the repeat gapmer. After having ...
  69. [69]
    Neurodegenerative and neuroinflammatory changes in SOD1-ALS ...
    Apr 1, 2025 · We observed a significant decrease in neurofilament levels during Tofersen treatment, alongside an increase in neuroinflammatory markers.
  70. [70]
    Considerations in the Preclinical Assessment of the Safety of ...
    Importantly, gapmers using both moderate-affinity MOE and high-affinity LNA have caused unexpected renal toxicity in humans that had not been detected or ...
  71. [71]
    GalNAc Conjugation Attenuates the Cytotoxicity of Antisense ...
    These data further suggest that GalNAc conjugation can mitigate the nephrotoxic potential of AON sequences by direct protection of renal tubular cells. (A) ...Original Article · Results · Materials And Methods
  72. [72]
    Toxicity of Antisense Oligonucleotides is Determined by the ...
    Oct 13, 2025 · In fact, about half of approved ASO drugs carry warnings or precautions for toxicities such as hypersensitivity reactions, liver damage, or ...<|separator|>