What Oligonucleotide are being developed?

17 March 2025
Introduction to Oligonucleotides

Definition and Types
Oligonucleotides are short, chemically synthesized strands of nucleic acids composed of DNA, RNA, or their analogs. They typically range from 10 to 50 nucleotides in length and can be synthesized with various chemical modifications to enhance their stability, binding affinity, and pharmacokinetic properties. In modern drug development, several types of oligonucleotides are being developed:

Antisense oligonucleotides (ASOs) are single-stranded molecules that bind to specific RNA targets via Watson–Crick base pairing, triggering degradation of the target mRNA (often via RNase H) or modulating splicing events.
Small interfering RNAs (siRNAs) are double-stranded RNA molecules that leverage the RNA-induced silencing complex (RISC) to promote cleavage of complementary mRNA, leading to gene silencing.
Splice switching oligonucleotides (SSOs) or steric-blocking oligonucleotides are specifically designed to bind pre-mRNAs near splice sites and modify splicing events, which can restore normal protein expression or even upregulate certain gene products.
MicroRNA (miRNA) therapeutics include both inhibitors (anti-miRs) and mimics, which modulate the levels and function of endogenous miRNA to achieve a therapeutic modulation of gene expression.
Aptamers are structured single-stranded oligonucleotide ligands that fold into unique three-dimensional conformations, enabling them to bind proteins or other cellular targets with high specificity.
Decoys and oligonucleotide “sponges” are additional modalities that sequester target transcription factors or miRNAs away from their natural gene regulatory functions.

In addition to these, novel classes under preclinical or early clinical development involve chemically engineered forms such as locked nucleic acids (LNAs), phosphorodiamidate morpholino oligomers (PMOs), peptide nucleic acids (PNAs), and various conjugates with lipids or targeting ligands (e.g., GalNAc). These modifications serve to enhance tissue penetration, reduce immunogenicity, increase metabolic stability, and improve in vivo pharmacodynamics.

Historical Development
The conceptual basis for oligonucleotide-based therapeutics emerged over 30–40 years ago when the first demonstrations of targeted regulation of gene expression using short nucleic acid fragments were published. Early antisense oligonucleotides faced significant hurdles such as rapid nuclease degradation, poor cellular uptake, and insufficient target engagement due to their inherent high polarity and negative charge. Over subsequent decades, researchers focused on chemical modifications to the sugar-phosphate backbone (e.g., phosphorothioate modifications) and on alternate nucleotide chemistries that significantly improved both stability and target affinity.

The development trajectory mirrors that seen in other drug modalities such as monoclonal antibodies, with an extended period from initial proof-of-concept studies to clinical adoption. Milestones include the approval of the first ASO (fomivirsen) in the late 1990s, followed by clinical successes such as mipomersen for familial hypercholesterolemia and later the CNS-active antisense oligonucleotide nusinersen for spinal muscular atrophy. Each of these approvals signaled important advances in manufacturing, formulation, delivery strategies (e.g., intrathecal injections for CNS diseases), and regulatory acceptance of the modality.

Current Oligonucleotide Therapeutics

Types of Oligonucleotides in Development
A diverse range of oligonucleotides are currently being developed at various stages of research and clinical trials. They include:

Antisense Oligonucleotides (ASOs):
  – Gapmers, which are designed with a central region of DNA nucleotides flanked by modified RNA segments to induce RNase H-mediated degradation of target mRNA.
  – Splice-switching oligonucleotides (SSOs) that bind pre-mRNA to alter splicing events, thereby correcting aberrant splicing patterns or promoting exon inclusion or skipping.
  – Modified ASOs incorporating second- and third-generation chemistries such as 2′-O-methoxyethyl (2′-MOE) modifications, LNAs, and peptide conjugates to enhance drug-like properties and optimize tissue targeting.

siRNAs:
  – Molecules designed to trigger RNA-interference mechanisms through efficient incorporation into the RISC complex.
  – Recent developments have focused on siRNA modifications and conjugations, such as N-acetylgalactosamine (GalNAc) conjugation, that facilitate targeted delivery particularly to hepatocytes, enabling prolonged gene silencing through single subcutaneous administrations.

miRNA Therapeutics:
  – Anti-miR oligonucleotides developed to antagonize disease-causing miRNAs by binding to and inhibiting their function.
  – miRNA mimics are also investigated to restore the function of downregulated miRNAs in diseases such as cancer and cardiovascular disorders.

Aptamers and Other Nucleic Acid-Based Ligands:
  – Aptamers that can be used to target proteins which are undruggable by small molecules, providing specificity and selectivity through their unique tertiary structures.
  – Novel oligonucleotide decoys that interfere with transcription factor activity or sequester specific RNAs have also been proposed.

Next-Generation Conjugated and Modified Oligonucleotides:
  – These include formulations where oligonucleotides are covalently linked to cell-penetrating peptides, lipid nanoparticles, or other ligands to improve delivery and overcome endosomal entrapment.
  – Other technological innovations focus on enhancing the release of oligonucleotides from endosomal compartments to increase target engagement within the cytosol.

Importantly, the field is also exploring multifunctional and combination approaches. For instance, oligonucleotide therapeutics are being developed in combination with standard of care chemotherapies to target multiple cancer drivers at once, or to be used in tandem with immunotherapies to modulate the tumor microenvironment. Moreover, gene editing applications and programmable epigenomic mRNA candidates, which modulate gene expression at the transcriptional level before translation, are emerging as novel frontiers in oligonucleotide drug development.

Targeted Diseases
The spectrum of diseases targeted by these oligonucleotide therapeutics is broad and continues to expand as delivery technologies and chemical modifications improve. Key targeted disease areas include:

Neurological and Neurodegenerative Diseases:
  – Owing to the ability to bypass traditionally “undruggable” targets in the central nervous system, ASOs have been developed for conditions such as spinal muscular atrophy (nusinersen) and are also being explored for Alzheimer’s disease, Huntington's disease, and amyotrophic lateral sclerosis.
  – Local delivery techniques, such as intrathecal injections and direct brain administration, have been utilized to overcome challenges associated with the blood–brain barrier.

Rare and Genetic Diseases:
  – Many oligonucleotide therapeutics are being tailored for rare neuromuscular and genetic disorders, such as Duchenne muscular dystrophy (via exon skipping therapy) and familial amyloid polyneuropathy, by targeting specific genetic mutations and restoring normal gene function.

Cancer and Malignant Diseases:
  – Oligonucleotides are being developed to silence oncogenes (for example, targeting MYC or STAT3 via ASOs or siRNAs) and to target non-coding RNAs that contribute to tumorigenesis.
  – Combination approaches that use oligonucleotides in tandem with chemotherapy, radiation therapy, or immunotherapy are under active clinical investigation to extend therapeutic benefits with minimal toxicity.

Liver Diseases:
  – With the success of GalNAc conjugation technology, many siRNAs and ASOs are specifically developed to target hepatocytes for diseases such as TTR amyloidosis and hepatitis B, benefiting from enhanced liver uptake and prolonged effect.

Respiratory and Inflammatory Disorders:
  – Oligonucleotide therapeutics, particularly siRNAs and ASOs, are under development to treat chronic inflammatory respiratory diseases like asthma and chronic obstructive pulmonary disease (COPD), often using nanoparticle-based delivery systems to facilitate targeted pulmonary administration.

Cardiovascular Diseases and Other Conditions:
  – Emerging work in oligonucleotide therapeutics is also focusing on gene targets related to cardiovascular pathology, aiming to mitigate conditions through the modulation of gene expression patterns related to inflammation, remodeling, or metabolic dysregulation.

This wide therapeutic coverage reflects the underlying strength of oligonucleotide drugs—their sequence-based targeting ability allows them theoretically to address the root genetic causes of many diseases that are refractory to traditional small molecule or protein-based treatments.

Development Process and Challenges

Research and Development Stages
The advancement of oligonucleotide therapeutics from concept to clinical product involves several critical stages, each of which has evolved considerably over the past few decades:

Discovery and Preclinical Research:
  – Initial stages involve target identification and the design of oligonucleotide sequences that can specifically bind to disease-causing mRNAs or regulatory RNAs.
  – Early preclinical studies focus on evaluating binding affinity, catalytic mechanisms (such as RNase H activation in the case of ASOs), and the efficiency of target gene silencing in vitro.
  – In parallel, extensive medicinal chemistry efforts are undertaken to explore chemical modifications (e.g., 2′-MOE, LNA, phosphorothioate backbones) that enhance nuclease resistance, binding efficacy, and overall pharmacokinetics.
  – In vivo animal models are used to assess tissue distribution, pharmacodynamics, and potential toxicity. Recent developments include studies on quantifying endosomal escape, cellular uptake, and the impact of ligand conjugation on therapeutic index.

Manufacturing and Formulation Development:
  – The synthesis of oligonucleotides is primarily conducted via solid-phase synthesis methodologies which have been refined over the past 40 years to improve yield and purity.
  – Scale-up and process optimization remain an area of intense research, particularly as new chemical modifications are introduced that may affect the overall manufacturing process and sustainability.
  – Formulation research also focuses on novel delivery systems such as lipid nanoparticles, conjugates (e.g., GalNAc, peptides), and even exosomes to improve cellular uptake and tissue targeting.

Clinical Development:
  – Early phase clinical trials (Phase I/II) are conducted to establish safety, tolerability, and preliminary efficacy. Many oligonucleotide therapeutics have shown target engagement in patient tissues even at relatively low systemic doses owing to improved delivery chemistries.
  – Later stage trials further evaluate long-term efficacy, dosing frequency (for example, certain liver-targeting siRNAs show sustained activity for 6–9 months after a single administration), and combination therapy potential.
  – As oligonucleotide therapies target a wide range of diseases—from rare genetic disorders to common chronic diseases—the R&D process is tailored to address disease-specific challenges (for instance, intrathecal dosing for CNS diseases versus systemic dosing for liver diseases).

In summary, R&D for oligonucleotides has evolved from simple sequence-based gene silencing experiments to a sophisticated process that integrates chemical optimization, novel delivery platforms, large-scale manufacturing, and complex clinical design—all aimed at making these drugs both efficacious and safe.

Key Challenges in Development
Notwithstanding the substantial progress made, several challenges continue to impact the clinical translation of oligonucleotide therapeutics:

Delivery Efficiency and Cellular Uptake:
  – Due to the inherent polarity and high molecular weight of these molecules, crossing cellular membranes is a significant obstacle. Many oligonucleotides rely on endocytic pathways, leading to challenges with endosomal entrapment.
  – Although modifications such as ligand conjugation (e.g., GalNAc for liver targeting) have greatly improved uptake, efficient delivery to tissues such as the CNS or solid tumors remains an area of active investigation.
  – Strategies to improve endosomal escape—including nanoparticle formulations and chemical modifications—are under continuous development, as only a small fraction (often 1–2%) of the administered dose may ultimately reach the cytosol.

Stability and Nuclease Resistance:
  – Unmodified oligonucleotides are rapidly degraded in the bloodstream by nucleases. Chemical modifications such as phosphorothioate backbones, sugar modifications (2′-O modifications, MOE), and the development of locked nucleic acids (LNAs) help overcome these limitations.
  – Despite these modifications, balancing stability with target affinity and overall safety profile remains a complex endeavor since modifications may affect toxicity and biodistribution.

Off-target Effects and Immunogenicity:
  – The sequence-specific nature of oligonucleotides necessitates rigorous screening to minimize interactions with unintended targets, which could lead to off-target gene silencing or activation of immune responses.
  – Some modifications, while increasing stability, may also raise the risk of adverse effects such as complement activation or thrombocytopenia.
  – Developing highly specific sequences that avoid such adverse interactions while retaining potency is a delicate balance in the discovery phase.

Manufacturing and Process Scalability:
  – The synthesis of oligonucleotides by solid-phase methods is well established; however, as the demand grows and more complex modifications are employed, challenges related to scalability, cost efficiency, and sustainability (such as high process mass intensity and hazardous reagent use) arise.
  – Greener synthesis methods and process optimization strategies are being explored to mitigate environmental and economic burdens without compromising product quality.

Complexity in Combination Therapies:
  – As many oligonucleotide therapies are now being evaluated in combination with other treatments (chemotherapy, immunotherapy, radiotherapy), understanding drug–drug interactions and optimizing dosage regimens becomes increasingly complex.
  – The therapeutic index of combination strategies must be carefully calibrated to ensure synergistic effects without exacerbating toxicity.

Overall, each of these challenges requires a multidisciplinary approach that integrates advances in chemistry, formulation science, biology, and clinical trial design to fully realize the therapeutic potential of oligonucleotides.

Regulatory and Market Considerations

Regulatory Pathways
Oligonucleotide therapeutics have unique regulatory challenges arising from their molecular characteristics and mechanisms of action. Several key points are critical in understanding their regulatory landscape:

Quality and Safety Evaluation:
  – Regulatory agencies require detailed characterization of the oligonucleotide’s chemical composition, purity, and manufacturing process. This includes the analysis of batch-to-batch consistency and the demonstration of robust quality controls in the manufacturing process.
  – Safety assessments emphasize potential immunogenicity, off-target effects, and toxicities that could occur due to sequence-specific interactions or chemical modifications.
  – Nonclinical studies must be designed to address organ-specific toxicity, pharmacokinetic and pharmacodynamic profiles, and metabolite analysis, which are distinct from traditional small molecule studies.

Evaluation of Novel Delivery Mechanisms:
  – The regulatory submission must include data on the delivery methods and the use of novel excipients or carriers such as lipid nanoparticles, conjugated peptides, or nanocarriers. These are scrutinized closely to ensure that the formulation does not lead to unexpected adverse events.
  – Guidance documents from regulatory authorities increasingly acknowledge the uniqueness of oligonucleotide chemistry, and submissions are expected to follow both traditional IND requirements and newer guidelines specific to nucleic acid-based therapies.

Clinical Pharmacology and Drug Interaction Studies:
  – Given that oligonucleotides often rely on binding to plasma proteins for stability, understanding drug–drug interactions and the influence of endogenous factors on their pharmacokinetics is crucial.
  – Regulators evaluate the impact of modifications, formulation, and delivery on clinical outcomes, ensuring that the dosage regimen is both effective and safe over extended periods.

Patent and Intellectual Property Considerations:
  – The field is also characterized by numerous patents covering specific chemical modifications, formulations, and uses of oligonucleotides.
  – Regulatory submissions must navigate both clinical and intellectual property landscapes, ensuring that novel therapeutic agents are protected while meeting rigorous safety and efficacy requirements.

This robust regulatory framework is designed to support the translation of oligonucleotide therapeutics from preclinical research to the clinic while ensuring patient safety and product quality.

Market Trends and Potential
The market for oligonucleotide therapeutics has seen considerable growth over the past few years, driven by advancements in delivery technologies, improved chemical modifications, and successful clinical approvals that validate the modality. Key market considerations include:

Expanding Pipeline and Product Approvals:
  – The number of oligonucleotide therapeutics entering clinical trials has steadily increased, with recent approvals such as nusinersen and emerging siRNA formulations that target hepatocytes via GalNAc conjugation serving as proof-of-concept.
  – As new candidates advance into later-stage clinical trials and eventually gain market approval, the portfolio of therapeutic oligonucleotides is expected to expand, addressing diseases ranging from rare genetic disorders to cancer and inflammatory conditions.

Strategic Collaborations and Licensing:
  – Large pharmaceutical companies are actively acquiring or partnering with companies specializing in oligonucleotide therapies, which is a testament to the growing commercial potential of these modalities.
  – Licensing agreements (for instance, the transfer of technology from academic research to industrial development) have accelerated the pace of innovation and enabled rapid scalability of manufacturing processes.

Market Opportunities Driven by Unmet Medical Needs:
  – A significant advantage of oligonucleotide therapeutics is their ability to target “undruggable” genes that have been inaccessible to conventional approaches. This expands the potential market across multiple therapeutic areas such as CNS disorders, rare neuromuscular diseases, and cancers that are difficult to treat with small molecule drugs.
  – The versatility in design and the relatively rapid timeline for the development of new oligonucleotide sequences (once the target is known) allows for a personalized medicine approach in conditions with high genetic heterogeneity.

Challenges in Cost and Reimbursement:
  – Although progress in manufacturing has driven improvements in cost efficiency, many oligonucleotide therapeutics remain complex and expensive to produce. Cost considerations and reimbursement negotiations (as seen in gene therapy markets) are critical for widespread clinical adoption.
  – As manufacturing processes continue to mature and greener, more sustainable synthesis methods are adopted, costs are likely to decrease over time, further expanding market potential.

Future Growth Prospects:
  – With continuous improvements in delivery systems (e.g., nanoparticle-based carriers, ligand conjugation), more oligonucleotide therapeutics are expected to demonstrate significant clinical benefit, stimulating further investments in research and development.
  – Regulatory agencies are increasingly familiar with these modalities, suggesting that streamlined pathways for approval could further accelerate market growth.

Overall, the commercial landscape for oligonucleotide therapeutics is highly promising, with robust growth anticipated in the coming years as more products enter the market and address critical unmet needs across a spectrum of diseases.

Conclusion

In conclusion, a wide array of oligonucleotide therapeutics are currently being developed, each tailored for unique mechanisms of action and specific disease targets. Starting with the foundational concepts of antisense oligonucleotides, siRNAs, and splice-switching oligonucleotides, the field has rapidly evolved through extensive chemical modifications and innovative delivery strategies. This evolution—from early, minimally modified molecules exhibiting poor stability and cellular uptake to advanced, highly optimized agents incorporating diversified chemistries (e.g., ligand-conjugated, nanoparticle-formulated, and chemically stabilized oligonucleotides)—illustrates the transformative progress over the past decades.

The current research pipeline includes ASOs targeting genetic mutations in rare neuromuscular diseases, siRNAs harnessing GalNAc conjugation for liver-specific delivery, miRNA therapeutics aimed at rebalancing disease-associated gene regulatory networks, and further modalities that target oncogenes and transcription factors in various cancers. These therapeutics are being applied to an ever-widening spectrum of diseases including central nervous system disorders, rare genetic diseases, cancers, respiratory ailments, and cardiovascular diseases.

The development process involves rigorous preclinical research, innovatively improved manufacturing and formulation strategies, and well-designed clinical trials that collectively aim to maximize target engagement while minimizing off-target effects and toxicity. Challenges such as efficient cellular uptake, endosomal escape, off-target activities, and manufacturing scalability remain at the forefront of ongoing research efforts. Regulatory pathways are evolving in tandem to address these novel characteristics, ensuring thorough quality and safety assessments tailored specifically for nucleic acid-based modalities.

Market trends strongly indicate robust growth in this sector, driven by the clinical successes of early pioneering products and the continued interest from both academia and industry. As more oligonucleotide therapeutics demonstrate positive clinical outcomes, especially in areas with significant unmet medical needs, the commercial potential will further expand through strategic collaborations, improved cost efficiencies, and streamlined regulatory processes.

Ultimately, oligonucleotide therapeutics represent a paradigm shift in how we approach drug development. They enable personalized and precision medicine strategies by designing sequence-specific interventions that address the root cause of many incurable diseases. The path forward involves surmounting delivery and scalability challenges through ongoing innovation while leveraging their intrinsic ability to target almost any gene. Given the substantial progress across historical, current, and emerging therapeutic areas, the outlook for oligonucleotide development is both highly promising and transformative for modern medicine.

Curious to see how Eureka LS fits into your workflow? From reducing screening time to simplifying Markush drafting, our AI Agents are ready to deliver immediate value. Explore Eureka LS today and unlock powerful capabilities that help you innovate with confidence.