What are the different types of drugs available for lncRNA?

17 March 2025
Introduction to lncRNA

Long noncoding RNAs (lncRNAs) are a diverse and abundant class of RNA transcripts that exceed 200 nucleotides in length and do not encode proteins. They have been intensively characterized over the past decade and are now recognized as key regulators of gene expression. Their unique functions, expression patterns, and abilities to interact with DNA, RNA, and proteins have made them attractive both as biomarkers and as novel drug targets for many human diseases.

Definition and Functions of lncRNA

lncRNAs are defined as RNA molecules longer than 200 nucleotides that do not translate into proteins but are far from biologically inert. Instead, these transcripts serve pleiotropic roles in transcriptional regulation, chromatin remodeling, RNA splicing, translation modulation, and post‐transcriptional control. lncRNAs operate via multiple mechanisms that include:

- Scaffolding proteins and chromatin modifiers: lncRNAs can assemble ribonucleoprotein complexes by binding to chromatin modifiers (e.g., polycomb repressive complex 2) and transcription factors to modulate gene expression epigenetically.
- Acting as decoys or “sinks”: Some lncRNAs sequester microRNAs and proteins away from their intended targets, thus altering the normal regulatory pathways.
- Guiding regulatory complexes: They can help direct chromatin and transcriptional regulators to specific genomic loci by base pairing with DNA or by forming triple-helical structures.
- Modulating mRNA splicing and stability: lncRNAs may interact with splicing factors or directly affect mRNA stability, influencing protein production indirectly.

Together, these functions underscore the highly integrated roles of lncRNAs in controlling cellular homeostasis and dynamic responses to stimuli.

Role of lncRNA in Disease Pathogenesis

Dysregulation of lncRNAs is now widely implicated in the pathogenesis of numerous diseases ranging from cancer and cardiovascular conditions to neurological disorders and metabolic diseases. In cancer, for instance, lncRNAs can operate either as oncogenes or tumor suppressors by modulating processes such as cell proliferation, apoptosis, metastasis, and drug resistance. In inflammatory and cardiovascular diseases, aberrations in lncRNA expression have been shown to affect lipid metabolism, fibrosis, and immune regulation. Their tissue- and cell-specific expression renders them particularly suitable for such roles. This strong link between lncRNA aberrations and disease progression has directed considerable research toward harnessing these transcripts as both diagnostic markers and therapeutic targets.

Types of Drugs Targeting lncRNA

Therapeutic interventions against lncRNAs have rapidly evolved. While they initially emerged as novel biomarkers, the focus in recent years has shifted to directly targeting these transcripts to modulate their function. There are three main classes of drugs currently being developed for lncRNA targeting:

Small Molecule Inhibitors

Small molecule inhibitors are designed to bind specific structural motifs or “pockets” within the lncRNA molecule, thereby disrupting its secondary structure or interfering with its associations with RNA-binding proteins. This approach capitalizes on the unique three-dimensional folding characteristics of lncRNAs.

- Mechanism of Action:
Small molecules may interact directly with the lncRNA to block its ability to form essential complexes or alter its conformation so that it becomes unable to bind target proteins or genomic DNA. For instance, inhibitors have been identified that can target the triple helix structure found in certain lncRNAs, such as MALAT1, hence preventing its stabilization and function.
- Advantages:
One notable benefit of small molecule inhibitors is their generally favorable pharmacokinetic properties—including high tissue penetration—which can be particularly useful for reaching targets in difficult-to-access tissues. In addition, their small size facilitates chemical optimization, enabling structure–activity relationship studies.
- Recent Advances and Studies:
Several studies have reported the discovery of compounds that bind to oncogenic lncRNAs, disrupting their interactions with epigenetic complexes (e.g., HOTAIR binding to PRC2). Although most of these compounds are still at the preclinical stage, the approach shows promise as part of combination therapies with conventional small molecules.

Small molecule inhibitors continue to be refined by improving specificity to avoid off-target interactions and to optimize bioavailability. Their versatility makes them appealing for use in cancers, fibrosis, and other diseases where aberrant lncRNA activity is central to pathology.

Antisense Oligonucleotides

Antisense oligonucleotides (ASOs) are short, chemically synthesized single-stranded DNA molecules designed to bind complementary sequences on lncRNAs via Watson–Crick base pairing. Upon binding, several functional outcomes are possible:

- Degradation of the lncRNA:
Binding of an ASO may recruit RNase H, an endogenous enzyme that degrades the RNA strand of a DNA–RNA hybrid, thereby reducing the levels of the target lncRNA.
- Steric Blockade:
ASOs can interfere with the functional domains of the lncRNA without necessarily causing degradation. For instance, they may block binding sites for proteins or other RNAs, altering the function of the lncRNA.
- Advanced Chemistries:
Most recent efforts have focused on second- and third-generation ASOs such as gapmers, locked nucleic acids (LNAs), and peptide nucleic acids (PNAs). Their modified sugar backbones and chemical linkages confer increased nuclease resistance, higher binding affinity, and improved pharmacokinetic properties.
- Clinical Progress:
ASO-based therapeutics are among the most clinically advanced nucleic acid drugs. Notably, Andes-1537, an ASO targeting mitochondrial noncoding RNA, has undergone clinical testing for advanced solid tumors. In addition, several ASOs have been approved for genetic diseases, offering a proof-of-concept for their use in suppressing pathogenic lncRNAs in cancer and other disorders.

Given the ability of ASOs to modulate nuclear lncRNAs—often inaccessible to other techniques—they remain a cornerstone strategy in lncRNA-targeted therapeutics. Their modular design allows for rapid adaptation once a disease–associated lncRNA is identified.

RNA Interference (RNAi) Therapies

RNA interference (RNAi) therapies utilize short interfering RNAs (siRNAs) and short hairpin RNAs (shRNAs) to trigger sequence-specific degradation of the target lncRNA. Although RNAi is more commonly associated with targeting messenger RNAs, this mechanism has also been exploited to reduce the expression of cytoplasmic lncRNAs.

- Mechanism of Action:
siRNAs are typically 21–23 nucleotide double-stranded RNAs that are incorporated into the RNA-induced silencing complex (RISC). The guide strand then base pairs with the target lncRNA, leading to its cleavage and eventual degradation. Similarly, shRNAs are expressed within the cell and later processed into active siRNAs.
- Advantages:
RNAi strategies capitalize on the cell’s natural machinery for RNA degradation, often yielding potent knockdown effects. They can be designed rapidly using bioinformatic tools to ensure specificity for the target sequence.
- Challenges and Specificity:
Although RNAi is a powerful approach, certain lncRNAs that are primarily nuclear in location may be less amenable to RNAi because the RISC complex is largely cytoplasmic. Furthermore, off-target effects and the innate immune response to double-stranded RNA are important considerations that need to be addressed through chemical modifications and optimized delivery systems.
- Clinical Translation:
While most RNAi therapeutics have focused on mRNAs, several studies have demonstrated the feasibility of using RNAi to silence pathogenic lncRNAs, thereby modulating cancer cell behavior in preclinical models. Ongoing trials continue to assess the safety and efficacy of these approaches, and advances in nanoparticle-based delivery are improving their translational potential.

Taken together, RNAi therapies offer an alternative and complementary strategy to ASOs for targeting lncRNAs, particularly when the lncRNA is accessible in the cytoplasm. The ability to design multiple siRNA sequences to target different regions of the lncRNA may also help overcome challenges related to transcript variants and folding.

Development and Application of lncRNA-targeting Drugs

The development of lncRNA-targeting drugs involves sophisticated drug discovery processes, rigorous preclinical testing, and innovative clinical applications. Researchers are now converging multiple techniques, including chemical modification, advanced delivery systems, and genome-targeting tools, to bring these novel therapeutics closer to clinical reality.

Drug Discovery and Development Processes

The journey from target identification to clinical candidate for lncRNA-targeting drugs commences with understanding the aberrant expression and function of a particular lncRNA in disease. Key steps include:

- Target Identification and Validation:
High-throughput sequencing and bioinformatics are used to profile differentially expressed lncRNAs in disease versus normal tissues. Functional studies then validate whether a specific lncRNA contributes to pathogenesis through knockdown or overexpression experiments.
- Design of Therapeutic Molecules:
For ASOs, this involves selecting a sequence that is unique to the target lncRNA and incorporating chemical modifications (such as LNAs or gapmers) to enhance binding stability, bioavailability, and resistance against nucleases.
For small molecule inhibitors, drug discovery often begins with detailed structural analyses of the lncRNA via novel screening techniques, including small-molecule microarrays, nuclear magnetic resonance (NMR), and computational modeling.
RNAi therapeutics require the design of siRNAs that can be processed effectively by the cell’s RISC machinery and that have minimal off-target effects. Optimization of the duplex and extensive computational prediction of unintended targets are essential parts of the development process.
- Delivery and Pharmacokinetics:
A major focus in the development stage is the formulation and delivery system. Viral vectors (such as lentiviral or adenoviral systems) and nonviral carriers like lipid nanoparticles (LNPs) are under investigation to protect the nucleic acid and ensure it reaches the correct cellular compartment. Delivery challenges such as degradation by nucleases, immunogenicity, and inefficient cellular uptake have spurred innovations such as chemical modifications and conjugation to cell-specific ligands.
- Preclinical Evaluations:
Once a candidate molecule is synthesized, it is evaluated in vitro using disease-relevant cell lines to determine knockdown efficiency, off-target effects, and cell viability. Subsequent in vivo studies in animal models quantify pharmacokinetics, biodistribution, and therapeutic efficacy. For example, efficient ASO delivery in preclinical cancer models has been shown to reduce tumor growth significantly.
- Regulatory and Translational Considerations:
With several nucleic acid-based drugs already approved by regulatory bodies (e.g., patisiran and nusinersen), the regulatory framework is evolving for lncRNA-targeting drugs. Detailed toxicology studies, dose-escalation trials, and analyses of immunogenicity are performed before entry into clinical trials.

In summary, the development process for lncRNA therapeutics integrates target discovery, molecular engineering, innovative delivery systems, and rigorous preclinical and translational research.

Clinical Applications and Case Studies

Advances in lncRNA-targeting drug development have led to several promising clinical applications, particularly in oncology and genetic diseases:

- Cancer Therapies:
lncRNAs such as HOTAIR, MALAT1, and DANCR have been implicated in promoting tumor progression, metastasis, and chemoresistance. For example, preclinical studies using ASOs or RNAi to knock down oncogenic lncRNAs in breast cancer and hepatocellular carcinoma have reported significant reductions in tumor growth. Early-phase clinical trials are being initiated to test the safety and efficacy of these approaches in patients with advanced solid tumors.
- Cardiac Diseases:
Certain cardiac-specific lncRNAs have been associated with heart failure and myocardial fibrosis. Innovative companies such as HAYA Therapeutics are developing antisense oligonucleotides targeting cardiac lncRNAs like Wisper to treat heart failure conditions. Their lead candidate, HTX-001, exploits the tissue specificity of lncRNAs, allowing for a more focused and potentially safer intervention.
- Other Disease Indications:
In addition to oncology and cardiology, lncRNA-targeting strategies are being explored in neurological disorders, metabolic diseases, and even infectious diseases. The high specificity of lncRNAs in particular cell types underpins their potential as biomarkers and therapeutic targets in nearly every medical field.
- Case Example – Andes-1537:
Andes-1537 is an exemplary clinical candidate developed as an ASO targeting specific mitochondrial lncRNAs in cancer cells. In early clinical trials, Andes-1537 demonstrated tolerability in patients with advanced metastatic tumors, paving the way for further evaluation of lncRNA-targeting ASOs in clinical settings.
- Combination Therapies:
Researchers are also investigating the use of lncRNA-targeting drugs in combination with traditional chemotherapeutics or immunotherapies. For example, targeting lncRNAs that contribute to drug resistance could help sensitize tumors to existing therapies, thereby improving overall patient outcomes.

These clinical applications underscore the transformative potential of lncRNA-targeting drugs. They benefit from high specificity, modular design, and, in many cases, a more favorable dosage profile due to the often low endogenous levels of lncRNAs.

Challenges and Future Directions

Even as the promise of lncRNA-targeting therapeutics grows, significant challenges remain. Overcoming these limitations is central to enhancing the clinical translatability of these drugs.

Current Challenges in lncRNA-targeting Drug Development

The main challenges hindering the rapid clinical adoption of lncRNA-targeting drugs include:

- Delivery Issues:
Ensuring that therapeutic agents—whether ASOs, small molecules, or siRNAs—reach their target cells in adequate amounts without degradation or triggering an immune response is perhaps the most significant obstacle. Advances in nanoparticle and liposomal formulations are being made, but robust delivery vehicles for tissue-specific targeting remain an active area of research.
- Off-Target Effects and Specificity:
Because lncRNAs often share partially homologous sequences or have complex secondary structures, there is a substantial risk of off-target binding which can lead to unintended gene expression changes and toxicity. Both ASOs and RNAi therapeutics require careful design and bioinformatic analysis to predict and minimize these side effects.
- Stability and Pharmacokinetics:
RNA-based drugs are inherently labile and prone to rapid degradation by nucleases in the bloodstream. Although chemical modifications (e.g., LNAs, phosphorothioate bonds) have improved stability, ensuring long-term bioavailability without eliciting immunogenicity is still a challenge.
- Intracellular Localization:
The efficacy of RNAi is often limited by the predominantly cytoplasmic nature of the RISC machinery, while many lncRNAs function in the nucleus. This necessitates the use of different therapeutic strategies (ASOs versus RNAi) based on the subcellular localization of the target lncRNA, increasing the complexity of drug design.
- Understanding lncRNA Structure-Function Relationships:
Although a number of lncRNAs have been functionally characterized, many remain poorly understood. Their dynamic secondary and tertiary structures often complicate the process of rational drug design for small molecule inhibitors.
- Species-Specific Conservation:
Many lncRNAs are poorly conserved across species, posing challenges for preclinical animal studies and the extrapolation of efficacy and toxicity data to humans. This issue may be overcome by developing humanized models, but it remains a significant hurdle.

Future Research and Development Directions

In light of these challenges, future research will likely focus on several key areas:

- Innovative Delivery Systems:
There is a pressing need to develop new vectors and nanoparticle formulations that can safely and efficiently deliver RNA therapeutics to target tissues. Advances in LNP technology and conjugation with cell-specific ligands are promising directions for improving biodistribution and cellular uptake.
- Enhanced Chemical Modifications:
Continued exploration of novel chemical modifications (e.g., cEt nucleotides, gapmer designs) will further bolster the stability, specificity, and intracellular penetration of ASO and RNAi molecules. Such modifications can also help reduce off-target effects and immunogenicity.
- Structural Characterization of lncRNAs:
High-resolution technologies such as cryo-electron microscopy, advanced NMR techniques, and enhanced computational modeling will enable better elucidation of lncRNA structures. Such knowledge will facilitate the design of small molecule inhibitors that target specific structural motifs within lncRNAs.
- Combination Therapeutic Strategies:
Future therapies may involve combination treatments where lncRNA-targeting drugs are used alongside conventional chemotherapies or immunotherapies. For example, reversing drug resistance by knocking down an oncogenic lncRNA can improve the efficacy of an existing therapeutic regimen.
- CRISPR-Based Approaches:
Genome editing tools such as CRISPR/Cas9 and CRISPR interference (CRISPRi) are emerging as promising technologies to modulate lncRNA expression at the transcriptional level. Although there are ethical and technical challenges that must be overcome, these methods offer durable gene knockdown effects and may provide alternatives to transient RNA-based therapies.
- Expansion of Clinical Trials:
As more preclinical data emerge, early-phase clinical trials will increasingly test the safety and efficacy of lncRNA-targeting drugs in humans. Future clinical studies will be critical for establishing dosing regimens, identifying biomarkers of response, and determining long-term outcomes.
- Multidisciplinary Collaboration:
The complexity of lncRNA biology requires collaboration among molecular biologists, chemists, bioinformaticians, and clinicians. Leveraging such expertise will facilitate holistic approaches to drug design, clinical translation, and personalized medicine strategies.

Researchers are also beginning to explore RNA-targeted drug modalities beyond the traditional ASO, RNAi, and small molecules, including aptamers and ribozymes. The convergence of nanotechnology, synthetic biology, and high-throughput screening promises a new era of RNA-based therapies that are more precise, efficient, and adaptable to individual patient needs.

Conclusion

In summary, several drug modalities are available for targeting lncRNAs, each with unique mechanisms and challenges. Small molecule inhibitors work by binding directly to structured motifs within lncRNAs to interfere with their interactions or to change their conformation, providing the possibility of high tissue penetration and chemical optimization. Antisense oligonucleotides represent a versatile and clinically advanced approach, particularly effective in modulating nuclear lncRNAs by recruiting nucleases for transcript degradation or blocking functional sites, as demonstrated by recent clinical candidates like Andes-1537. RNA interference therapies utilize siRNAs and shRNAs to leverage the endogenous RISC pathway for targeted degradation of lncRNAs, though their efficiency depends on the subcellular localization of the target transcript and optimized delivery systems.

The drug discovery and development processes for lncRNA-targeting drugs have evolved considerably over recent years. Researchers integrate high-throughput sequencing, bioinformatics, chemical modifications, and innovative delivery platforms into a robust paradigm that spans from in vitro validation to in vivo preclinical studies and early-phase clinical trials. Clinical applications are emerging primarily in oncology and cardiology, where cell- and tissue-specific lncRNAs play pivotal roles in disease progression and therapeutic resistance.

However, challenges remain. Delivery, off-target effects, stability, and understanding complex lncRNA structures are key hurdles to overcome in order to translate these novel therapeutics to the clinic. Future research directions include the development of improved nanoparticle carriers, enhanced chemical modification techniques, in-depth structural studies, and the integration of CRISPR-based genome editing approaches. Collaborative, multidisciplinary approaches will be crucial in advancing the field and ensuring the safe, effective, and personalized application of lncRNA-targeting drugs.

In conclusion, as our understanding of lncRNA biology deepens and technological advances continue, the range of therapeutic options targeting lncRNAs is expected to expand dramatically. This progress holds great promise for treating a variety of diseases that currently have limited therapeutic options, ultimately paving the way toward more precise and effective treatments with fewer side effects. The future of lncRNA-targeting therapeutics is bright, contingent upon overcoming current challenges through innovation and interdisciplinary collaboration, thereby redefining the landscape of precision medicine in the years to come.

Discover Eureka LS: AI Agents Built for Biopharma Efficiency

Stop wasting time on biopharma busywork. Meet Eureka LS - your AI agent squad for drug discovery.

▶ See how 50+ research teams saved 300+ hours/month

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.