Global Journal of Pharmaceutical and Scientific Research (GJPSR)
Volume- 1, Issue-3, October- 2025
Article Received on 30/09/2025 Article Revised on 09/10/2025 Article Accepted on 24/10/2025
RNA-TARGETED SMALL MOLECULE DRUG DESIGN: CHALLENGES, OPPORTUNITIES, AND FUTURE DIRECTIONS
Prof. (Dr)Mohd. Wasiullah1*,Prof. (Dr) Piyush Yadav2*, Anshuman Yadav3
1. Principal , Department of Pharmacy, Prasad Institute of Technology, Jaunpur, Uttar Pradesh, India.
2. Head: Department of Pharma: Chemistry, Prasad Institute of Technology, Jaunpur, Uttar Pradesh, India.
3. Scholar - Department of Pharmacy, Prasad Institute of Technology, Jaunpur, Uttar Pradesh, India.
Corresponding Author: Anshuman Yadav, Research Scholar, Department of Pharmacy, Prasad institute of technology
Abstract
RNA, which includes coding mRNAs, non-coding RNAs, and structural viral elements that control gene expression, translation, and cellular homeostasis, has become a key and adaptable therapeutic target. Ribosome-directed antibiotics were once the prime example of RNA-targeting small molecules, but developments in structural biology, chemical biology, and computational modeling have broadened the RNA drug gable space to include viral RNA motifs, oncogenic non-coding RNAs, and epitranscriptomic modifications. To enable rational ligand design, contemporary approaches make use of RNA chemical probing, high-throughput and fragment-based screening, NMR/cryo-EM structural insights, and AI-driven predictive modeling. New approaches to long-standing problems with selectivity, conformational heterogeneity, and cellular transport include RIBOTAC-mediated RNA degradation, covalent or reversible covalent RNA binders, and combination therapy with RNA biologics. In order to provide a foundation for creating next-generation RNA-targeted treatments, this study discusses RNA structures, small-molecule design principles, discovery methodologies, case studies, obstacles, and upcoming approaches.
Keywords: RNA–ligand interactions, structural biology, computational drug discovery, machine learning, bioactive small molecule.
1. Introduction
In addition to acting as a messenger between DNA and protein, ribonucleic acid (RNA) also regulates gene expression, splicing, translation, and epigenetic processes, making it an essential and adaptable component of cellular life. The identification of non-coding RNAs (ncRNAs), such as microRNAs (miRNAs), long non-coding RNAs (lncRNAs), circular RNAs (circRNAs), and small nucleolar RNAs (snoRNAs), has expanded the functional repertoire of RNA in health and disease beyond the traditional mRNA and tRNA (Esteller, 2011; Slack & Chinnaiyan, 2019). RNA is a promising but underutilized therapeutic target since dysregulation of RNA molecules has been linked to cancer, viral infections, neurological illnesses, and metabolic problems (Garcia-Moreno & Chen, 2021).
Historically, RNA-targeted drug discovery has been limited to ribosome-targeting antibiotics, which exploit the structural differences between bacterial and eukaryotic ribosomes to hinder translation. Highly organized RNA motifs can produce druggable pockets, as demonstrated by aminoglycosides, macrolides, tetracyclines, and oxazolidinones, offering proof-of-concept for small-molecule regulation of RNA (Wilson, 2014; Dunkle & Cate, 2010). But as small-molecule targets, the great majority of RNA molecules—especially dynamic non-coding RNAs—remain relatively unexplored.
The field of RNA-targeted therapy has undergone a revolution thanks to recent technological developments. The identification of ligands that bind particular RNA patterns is made possible by high-throughput screening, fragment-based strategies, and chemical probing techniques (Warner et al., 2018). In the meanwhile, structural biology techniques like cryo-electron microscopy, NMR spectroscopy, and X-ray crystallography offer atomic-level understanding of RNA folds and RNA–ligand interactions (Ganser et al., 2019). By predicting RNA structures, ligand binding sites, and dynamic conformations, computational modeling and artificial intelligence (AI) have further improved RNA drug development by facilitating the logical design and optimization of small molecules (Townshend et al., 2021; Vasudevan & Disney, 2022).
The RNA druggable space is being expanded by new approaches that go beyond traditional binding. Recruiting nucleases for targeted RNA degradation (RIBOTACs), covalently or reversibly engaging RNA, or selectively modulating epitranscriptomic marks like pseudouridine and N6-methyladenosine (m–A) are all now possible with small molecules (Roundtree et al., 2017; Costales et al., 2019; Morgan et al., 2017). These developments promise improved functional control, potency, and selectivity, especially for dynamic or disease-specific RNAs.
Despite these developments, structural heterogeneity, conformational dynamics, off-target binding, and intracellular delivery hurdles provide particular difficulties for RNA-targeted small-molecule therapies (Al-Hashimi & Walter, 2008; Mayer & Janin, 2019). In order to overcome these obstacles, experimental, computational, and AI-driven methods must be integrated with knowledge from chemical and structural biology.
The structural and functional landscape of RNA, the justification for using small molecules to target RNA, design principles, experimental and computational approaches, case studies, difficulties, and new prospects are all covered in this review, which offers a thorough overview of RNA-targeted small-molecule drug design. This review attempts to direct the development of next-generation RNA therapies with potential for precision and personalized medical applications by summarizing recent developments.
2. RNA as a Drug Target: Structural and Functional Landscape
2.1 Classes of Therapeutically Relevant RNAs
Messenger RNA (mRNA).
Through organized components including splicing junctions, internal ribosome entry sites (IRES), and untranslated regions (UTRs), mRNAs control the expression of proteins. These areas create distinct secondary structures that can be specifically targeted by small compounds to alter splicing, translation, or stability, providing therapeutic options for viral infections, cancer, and genetic abnormalities (Disney et al., 2022).
Ribosomal RNA (rRNA).
The most therapeutically proven RNA medication target is rRNA, which also serves as the catalytic and structural core of the ribosome. Many antibiotics interfere with protein production by binding conserved rRNA motifs like the peptidyl transferase center and decoding site. Strong proof-of-concept for RNA-directed small-molecule therapies is provided by the effectiveness of rRNA-targeting antibiotics (Wilson, 2014).
Transfer RNA (tRNA).
Translation requires tRNAs, which have conserved secondary and tertiary structures. In addition to their normal function, tRNA-derived fragments and altered tRNA expression have been connected to neurological diseases and cancer. Disease-associated tRNA processing pathways offer new treatment prospects despite the difficulty of direct targeting (Kirchner & Ignatova, 2015).
Non-Coding RNAs (ncRNAs).
Non-coding RNAs play critical regulatory roles and are increasingly implicated in disease.
Viral and Pathogenic RNAs.
Stem loops and frameshifting elements are examples of conserved structural motifs found in viral RNAs that are necessary for translation and replication. Compared to antivirals that target proteins, small compounds that target these RNA structures can stop viral reproduction and possibly lower resistance (Hermann, 2016).
Table 1: Classes of Therapeutically Relevant RNAs and Their Potential Small-Molecule Targets
| RNA Class | Key Features | Disease/Relevance | Examples of Small-Molecule Strategies |
| mRNA | Linear, coding, modifiable 5′/3′ ends | Cancer, viral infections | Translation inhibitors, mRNA stabilizers |
| rRNA | Highly structured, essential for ribosome | Bacterial infections | Ribosome-targeting antibiotics (aminoglycosides, macrolides) |
| tRNA | Cloverleaf structure, modified nucleotides | Cancer, stress response | tRNA interaction modulators |
| miRNA | ~22 nt, regulates gene expression | Cancer, cardiovascular disease | Pre-miRNA binding small molecules, RIBOTACs |
| lncRNA | >200 nt, secondary & tertiary structures | Cancer, neurodegeneration | Structure-specific binders |
| circRNA | Covalently closed loop, stable | Cancer, viral infections | Small molecules disrupting RNA-protein interactions |
| Viral RNAs | Structured motifs (IRES, hairpins) | Viral infections | Viral RNA binders, translation inhibitors |
2.2 RNA Structural Hierarchy
The biological activity and druggability of RNA molecules are determined by their hierarchical structural arrangement. Unlike proteins, RNA folding is very dynamic, enabling access to temporary yet targetable conformations.
Primary, Secondary, and Tertiary Structure.
The nucleotide sequence of RNA determines its fundamental structure, which in turn determines base-pairing interactions that result in secondary structures like stems, loops, and junctions. These secondary components then come together to form intricate tertiary structures that are supported by base stacking, metal ions, and long-range interactions. Despite the polyanionic character of RNA, tertiary folding produces distinct three-dimensional pockets that can recognize specific small molecules (Doudna & Cech, 2002; Ganser et al., 2019).
Key RNA Structural Motifs.
RNA tertiary structures are built from recurrent motifs that serve as privileged binding sites for ligands:
These motifs are found in a variety of RNA classes and offer structural specificity that can be used to target certain small molecules (Hermann & Patel, 2000; Disney et al., 2022).

Figure 1: Structural Hierarchy of RNA and Common Druggable Motifs
2.3 RNA dynamics and conformational plasticity
Rather of occupying a single static shape, RNA molecules are intrinsically dynamic and inhabit ensembles of interconverting conformations. Weak base-pairing contacts, flexible sugar-phosphate backbones, and sensitivity to biological elements like ions, proteins, and metabolites are the causes of this conformational plasticity. Function, ligand recognition, and druggability are all heavily influenced by RNA dynamics (Ganser et al., 2019).
Base flipping, helix bending, loop restructuring, and tertiary contact creation are examples of the local and global structural changes that RNAs go through. Although these motions make it possible for RNAs to adjust to changing environments and binding partners, they also make structure-based drug design more difficult. Crucially, cryptic binding pockets that are not present in ground-state structures but are selectively targetable by small molecules can be revealed by temporary or low-population conformational states (Al-Hashimi & Walter, 2008).
Through induced fit, in which ligand binding stabilizes a novel RNA structure, or conformational selection, in which ligands bind pre-existing RNA conformers, small molecules can take advantage of RNA dynamics. Particularly in riboswitches, viral RNAs, and regulatory ncRNAs, both processes have been seen in RNA–ligand interactions. The therapeutic significance of RNA plasticity is highlighted by the fact that stabilization of particular RNA conformations can influence biological outcomes like translation, splicing, or RNA processing (Hermann, 2016; Disney et al., 2022).
3. Rationale for Small-Molecule Targeting of RNA
3.1 Comparison with Antisense and RNA-Based Biologics
Antisense oligonucleotides (ASOs), siRNAs, and RNA aptamers are examples of RNA-based biologics that have received regulatory approval for a number of genetic and metabolic illnesses by modulating gene expression through sequence-specific Watson–Crick base pairing. Despite this achievement, fundamental limitations include large molecular size, limited membrane permeability, endosomal entrapment, quick renal clearance, and reliance on chemical modifications or delivery vehicles (such lipid nanoparticles or conjugates) restrict their therapeutic applicability. These difficulties frequently limit tissue dispersion and make long-term treatment more difficult (Crooke et al., 2017).
On the other hand, tiny molecules use structure-based recognition instead of sequence complementarity to target RNA. This makes it possible for oligonucleotides to bind selectively to folded RNA motifs like hairpins, bulges, and junctions. Small compounds can access transitory or partially folded RNA states, act intracellularly without the need for delivery vehicles, and alter RNA function by stabilizing or destabilizing particular conformations. Crucially, RNAs implicated in translation, splicing, and RNA–protein interactions can be targeted by small compounds, extending the therapeutic spectrum beyond RNA degradation or knockdown (Warner et al., 2018; Disney et al., 2022).
3.2 Advantages of Small Molecules
Small compounds are appealing for RNA targeting because they have demonstrated pharmacological advantages. Oral bioavailability, cellular absorption, and wide tissue penetration—including the central nervous system—are made possible by their low molecular weight and adjustable lipophilicity. Small molecules have better chemical and metabolic stability than RNA biologics, allowing for a longer shelf life and easier formulation. Global accessibility and long-term treatment plans are supported by manufacturing, which is typically scalable and economical (Hermann, 2016).
Medicinal chemistry optimization enables the methodical enhancement of potency, selectivity, and pharmacokinetic characteristics from the standpoint of drug development. Additionally, small compounds can be designed to partially or context-dependently regulate RNA function without totally suppressing gene expression. When total RNA inhibition is undesired or harmful, this fine control is very helpful (Warner et al., 2018; Disney et al., 2022).
3.3 Therapeutic Contexts Favoring RNA-Targeted Small Molecules
RNA-targeted small compounds are particularly useful in disorders where pathogenic function depends on RNA structure. As shown by rRNA-targeting antibiotics and newly developed antiviral RNA ligands, conserved RNA elements in bacterial ribosomes and viral genomes provide verified and resistance-resilient targets in infectious illnesses. Tumor growth in oncology is driven by dysregulated non-coding RNAs and abnormal splicing processes; small drugs can specifically modify these RNA structures without changing genomic DNA (Ganser et al., 2019).
Additionally, repeat expansion diseases and other neurological and genetic disorders involving hazardous RNA gain-of-function pathways are excellent candidates for small molecules because they can restore cellular homeostasis by stabilizing or disrupting pathogenic RNA conformations. Additionally, the oral availability, reduced cost, and enhanced patient compliance of small molecules offer distinct advantages over injectable RNA biologics for chronic diseases requiring long-term therapy (Disney et al., 2022).
4. Design Principles for RNA-Targeted Small Molecules
4.1 Physicochemical Features of RNA-Binding Ligands
A strongly negatively charged phosphate backbone, shallow grooves, and few hydrophobic surfaces define the difficult chemical landscape of RNA. As a result, RNA-binding ligands frequently depart from the traditional "Rule-of-Five" guidelines developed for protein targets. Higher polar surface area, moderate molecular weight (300–600 Da), and richness in heteroaromatic scaffolds are characteristics of known RNA-binding small molecules that facilitate interactions with exposed bases and backbone phosphates (Warner et al., 2018).
Protonatable amines included in many RNA ligands increase affinity by electrostatic attraction; yet, an excessive positive charge encourages nonspecific binding to a large number of biological RNAs and nucleic acids. Therefore, charge is balanced with form complementarity and directional hydrogen bonding in effective RNA-targeting molecules. While limited structural flexibility lowers entropic penalties upon binding, planarity and semi-rigidity improve π-stacking with nucleobases. Crucially, in order to guarantee translational viability, medicinal chemistry efforts are increasingly focusing on drug-like characteristics (metabolic stability, permeability) (Disney et al., 2022).
4.2 Modes of RNA–Small Molecule Recognition
RNA–ligand recognition is multimodal and exploits structural features unique to folded RNAs.
Shape Complementarity
Different three-dimensional pockets made up of bulges, internal loops, junctions, and pseudoknots are produced by RNA secondary and tertiary folding. Higher specificity is attained by small molecules that geometrically complement these pockets as opposed to those that only use charge. It has been shown in riboswitches, viral RNA elements, and miRNA precursors that shape recognition is especially important for differentiating target RNAs from structurally similar but non-functional RNAs (Hermann & Patel, 2000).
Electrostatic Interactions
Initial complex formation is driven and residence time is increased by the electrostatic interaction between cationic ligands and the RNA phosphate backbone. This idea is demonstrated by aminoglycosides, which bind rRNA through a variety of ionic interactions. Modern RNA-targeted drug design, however, decreases reliance on nonspecific charge by integrating electrostatics with structural and hydrogen-bonding interactions, as electrostatics alone seldom confers selectivity (Guan & Disney, 2012).
Hydrogen Bonding and π–π Stacking
By interacting with base edges that are exposed in non-canonical RNA motifs, hydrogen bonds offer directionality and specificity. RNA-ligand complexes are stabilized by π–π stacking interactions between aromatic ligands and nucleobases, which are especially crucial in hairpins and bulges where bases are exposed to solvents. High-affinity binding depends on these interactions, which together make up for the restricted hydrophobicity of RNA surfaces (Warner et al., 2018).
4.3 Selectivity Determinants in RNA Recognition
Rather than primary sequence, three-dimensional fold recognition, non-canonical base pairing, and conformational dynamics are the main factors that determine selectivity in RNA targeting. Selective ligand engagement is made possible by the distinctive folds that RNAs participating in regulatory processes frequently assume. These folds can be temporary or condition-dependent. Selectivity is further improved by focusing on disease-specific RNA conformations or RNA processing intermediates (such as pre-miRNAs).
RNA dynamics are important because ligands can bind low-population conformers preferentially, stabilizing them and changing the functional equilibrium. This tactic lessens off-target effects and makes it possible to distinguish between closely related RNAs. By necessitating spatially precise binding geometries, multivalent ligands that interact with many motifs at once have also demonstrated improved selectivity (Ganser et al., 2019; Disney et al., 2022).
4.4 Structure–Activity Relationships (SAR) in RNA Targeting
Compared to protein-focused SAR, SAR investigations in RNA-targeted drug discovery reveal different concepts. Because of RNA flexibility and induced-fit effects, little changes like heteroatom substitution, ring expansion, or linker length variation can significantly change binding affinity and functional results. While substituent placement controls hydrogen-bonding networks and electrostatic complementarity, aromatic ring systems regulate the strength of π-stacking.
In compounds intended to stabilize tertiary folds or bridge nearby RNA motifs, linker stiffness and spatial orientation are particularly important. SAR optimization frequently necessitates repeated cycles integrating biophysical assays, chemical probing, and functional readouts rather than relying solely on static structural data because RNA adopts conformational ensembles rather than static structures (Guan & Disney, 2012; Warner et al., 2018).
5. Experimental and Computational Strategies in RNA-Targeted Drug Discovery
5.1 Experimental Approaches
High-Throughput and Fragment-Based Screening.
To find RNA-binding small compounds, high-throughput screening (HTS) platforms have been modified to use functional reporter assays, FRET, and fluorescence polarization. These methods enable the quick assessment of thousands of chemicals against structured RNA targets, such as viral RNA elements and riboswitches. Fragment-based screening (FBS) complements HTS by applying low-molecular-weight chemicals that efficiently survey RNA chemical space and bind shallow or temporary pockets. Following fragment hits, structure-guided tuning is used to enhance affinity and specificity while preserving advantageous drug-like characteristics (Patel et al., 2017; Thomas & Hergenrother, 2008).
Chemical Probing and Footprinting Methods.
Nucleotide-level examination of RNA structure and ligand-induced modifications is made possible by chemical probing methods. RNA flexibility, base pairing, and solvent accessibility are revealed by techniques including SHAPE, dimethyl sulfate (DMS) modification, and hydroxyl radical footprinting. By tracking conformational changes brought on by small compounds, these methods, when used in vitro or in cells, confirm target engagement and clarify the mechanism of action (Spitale et al., 2013).
Biophysical Techniques.
RNA–ligand interactions can be understood quantitatively and structurally using biophysical techniques. For the characterization of RNA dynamics, binding interfaces, and conformational exchange, NMR spectroscopy is especially useful. Large RNA assemblies and ribonucleoprotein complexes can be seen using cryo-electron microscopy (cryo-EM), while X-ray crystallography has produced atomic-resolution structures of RNA–small molecule complexes. Lead optimization is guided by the quantification of binding kinetics and thermodynamics using surface plasmon resonance (SPR) and isothermal titration calorimetry (ITC) (Butcher & Pyle, 2011).
5.2 Computational and AI-Driven Methods
RNA Structure Prediction and Modeling.
By offering structural hypotheses for ligand binding, computational modeling is essential to the development of RNA-targeted drugs. The accuracy of RNA models has increased because to improvements in secondary and tertiary structure prediction, which are backed by experimental limitations. For RNAs without high-resolution experimental structures, these techniques help identify druggable features and direct rational ligand design (Laing & Schlick, 2011).
Virtual Screening and Molecular Docking.
By mimicking RNA–ligand interactions, virtual screening makes it possible to quickly rank compounds for experimental testing. RNA flexibility, solvation effects, and non-canonical base interactions like stacking and backbone contacts are all taken into consideration by RNA-specific docking techniques. By combining docking with molecular dynamics simulations, hit discovery success can be increased by exploring conformational ensembles and identifying transient binding pockets (Erlacher & Micura, 2018).
Machine Learning and Generative AI in RNA Ligand Discovery.
Machine learning (ML) techniques are being used more and more to optimize lead compounds, classify RNA-binding chemical space, and forecast RNA–ligand interaction. Features controlling affinity and selectivity can be found using data-driven models trained on datasets of RNA-ligand interactions. Generative AI frameworks further enable de novo generation of RNA-focused chemical libraries, expediting discovery and lowering experimental attrition (Chen et al., 2020).
Table 2: Experimental and Computational Strategies for RNA-Targeted Small Molecules
| Strategy Type | Method/Tool | Key Advantages | Limitations/Challenges |
| Experimental | High-throughput screening (HTS) | Rapid identification of ligands | Limited to accessible RNA motifs |
| Experimental | Fragment-based screening | Efficient for weak binders | Requires structural knowledge |
| Experimental | Chemical probing/footprinting | Maps ligand binding & RNA structure | Labor-intensive, low throughput |
| Experimental | Biophysical techniques (NMR, X-ray, cryo-EM, SPR) | Atomic-level resolution | Requires purified RNA, cost-intensive |
| Computational | RNA structure prediction & modeling | Guides ligand design | Accuracy depends on input & algorithms |
| Computational | Virtual screening & molecular docking | Prioritizes candidates | May miss dynamic RNA conformations |
| Computational | Machine learning & AI | Explores large chemical space | Needs high-quality datasets |

Figure 2: Workflow of RNA-Targeted Small Molecule Drug Discovery
6. Case Studies of RNA-Targeted Small-Molecule Therapeutics
6.1 Clinically Approved RNA-Targeting Antibiotics
The most clinically proven RNA target for small compounds is still ribosomal RNA (rRNA). Aminoglycosides, macrolides, tetracyclines, lincosamides, and oxazolidinones are among the types of antibiotics that bind to particular rRNA regions in the bacterial ribosome to prevent translation. These medications achieve species selectivity by taking advantage of minute structural variations between bacterial and eukaryotic rRNAs.
For instance, macrolides engage with the 23S rRNA exit tunnel to prevent peptide elongation, whereas aminoglycosides bind the 16S rRNA decoding site to cause misreading of mRNA. These achievements verify RNA as a credible therapeutic target and show that organized RNA motifs can generate high-affinity, druggable regions. However, the necessity for next-generation RNA-targeted methods is highlighted by resistance resulting from rRNA mutations and chemical changes (Wilson, 2014; Dunkle & Cate, 2010).
6.2 Small Molecules Targeting Viral RNAs
Viral RNAs are appealing therapeutic targets because they have highly conserved structural components necessary for translation, replication, and immune evasion. RNA motifs including internal ribosome entry sites (IRES), frameshifting elements, and stem-loop regulatory sections of viruses like HIV, HCV, influenza, and SARS-CoV-2 have been targeted by small compounds.
For example, ligands binding the SARS-CoV-2 frameshift element decrease viral protein synthesis, whereas small compounds targeting the HIV-1 TAR RNA suppress Tat-mediated transcription. Since RNA structural components are frequently highly conserved for function, targeting viral RNA structures has the advantage of lower resistance when compared to protein targets (Hermann, 2016; Disney & Childs-Disney, 2021).
6.3 Targeting Oncogenic and Disease-Associated Non-Coding RNAs
A new area of RNA-targeted small-molecule therapies is represented by non-coding RNAs (ncRNAs). Tumor suppressor pathways are derepressed when small compounds specifically target structured precursors of oncogenic microRNAs (miRNAs), such as miR-21 and miR-155.
Similar to this, disease-associated long non-coding RNAs (lncRNAs) that are involved in transcriptional regulation and chromatin remodeling have modular structural domains that can be bound by ligands. It has been demonstrated that small compounds can alter downstream gene expression by interfering with lncRNA–protein interactions. These investigations show that functional selectivity can be attained by RNA-targeted small molecules without total RNA degradation, providing an alternative to antisense methods (Velagapudi et al., 2016; Matsui & Corey, 2017).
6.4 Lessons Learned from Successes and Failures
When taken as a whole, these case studies highlight important guidelines for effective RNA-targeted drug development. First, the efficacy of a treatment depends on the RNA target's structural definition and functional indispensability. Second, an over-reliance on electrostatic interactions results in toxicity and nonspecific binding, highlighting the significance of shape and hydrogen-bond recognition. Third, while RNA dynamics and conformational heterogeneity make structure-based design more difficult, they also present chances to take advantage of temporary binding pockets.
Poor selectivity, low cellular engagement, or a lack of knowledge about RNA structural biology in physiological situations are frequently the causes of RNA-targeted drug development failures. To close the gap between in vitro binding and in vivo efficacy, future success will depend on combining dynamic RNA models, high-resolution structural data, and iterative experimental validation (Thomas & Hergenrother, 2008).
7. Key Challenges in RNA-Targeted Small-Molecule Drug Design
7.1 Achieving High Selectivity over Cellular RNAs
Achieving selectivity in the densely packed cellular RNA environment is a major difficulty in the design of RNA-targeted drugs. RNAs have recurring structural motifs in unrelated transcripts because, in contrast to proteins, they are made up of only four chemically identical nucleotides. Promiscuous binding, which leads to off-target engagement with abundant RNAs like rRNA and tRNA, is frequently seen in small molecules that primarily rely on electrostatic interactions with the phosphate backbone. Therefore, identifying distinct three-dimensional folds, sequence-context-dependent motifs, and dynamic conformational states—rather than general RNA characteristics—is necessary to achieve selectivity (Ragno et al., 2020).
7.2 RNA Structural Heterogeneity and Dynamics
Rather than being static structures, RNA molecules are collections of interconverting conformations. Structure-based ligand design is complicated by this inherent flexibility since binding pockets may be shallow, ephemeral, or only filled under particular biological conditions. Conformational flexibility offers potential to selectively stabilize disease-relevant RNA states, but it also provides a challenge to rational targeting. However, it is still technically difficult to capture these dynamic ensembles experimentally at biologically relevant timescales, which limits the prediction accuracy of ligand design (Al-Hashimi & Walter, 2008).
7.3 Off-Target Effects and Toxicity Concerns
A significant barrier to the clinical translation of RNA-binding small compounds is still off-target binding. Protein synthesis can be hampered by non-specific interactions with mitochondrial or ribosomal RNA, which can result in cytotoxicity. Furthermore, phospholipidosis, cardiotoxicity, and CNS liabilities are linked to cationic and planar molecules, which are typical characteristics of RNA-binding ligands. Due of these safety issues, thorough toxicity profiling and early counter-screening against critical RNAs are required during lead optimization (Mayer & Janin, 2019).
7.4 Drug-Like Property Optimization and Delivery Barriers
Because RNA-binding ligands require polar surface area and hydrogen-bonding capacity, they frequently fail to meet traditional drug-likeness standards. It is therefore difficult to balance RNA affinity with oral bioavailability, metabolic stability, and membrane permeability. For many RNA targets, effective intracellular delivery—especially nuclear or organelle-specific localization—remains a challenge. To get beyond these restrictions, novel medicinal chemistry techniques as prodrugs, conformational constraint, and transporter exploitation are being investigated (Costales et al., 2020).
7.5 Translational and Regulatory Challenges
It is currently unclear how RNA-targeted small molecules are translated. Standardized biomarkers for RNA target engagement and pharmacodynamic response are scarce, in contrast to protein inhibitors. Evaluating mechanism-of-action, off-target RNA interactions, and long-term safety presents additional difficulties for regulatory bodies, especially when it comes to first-in-class RNA-binding drugs. For wider clinical application, it will be crucial to establish reliable translational frameworks, validated assays, and regulatory precedents (Bermann et al., 2021).
8. Emerging Opportunities and Innovative Solutions
8.1 Targeting RNA Epitranscriptomic Modifications
The RNA epitranscriptome includes more than 170 different chemical modifications, such as 2′-O-methylation, pseudouridine (Ψ), 5-methylcytidine (m³C), and N6-methyladenosine (m≥A). Beyond the basic sequence, these alterations produce structural and functional variety by controlling RNA stability, splicing, translation efficiency, and subcellular localization. Neurological conditions, viral infections, and cancer are all associated with dysregulation of epitranscriptomic markers. Disease-relevant RNA conformations can be stabilized or destabilized by small compounds that specifically identify or modify these changes.
For instance, METTL3 inhibitors, a mBy lowering the methylation of carcinogenic transcripts, a "writer" has shown anti-leukemic action. On the other hand, ligands that are made to bind m—A-modified RNA hairpins preferentially can alter RNA–protein interactions without changing unmodified transcripts, demonstrating the possibility of great selectivity (Roundtree et al., 2017; Barbieri & Kouzarides, 2020). Because epitranscriptome changes are frequently dynamically regulated in disease states but not in normal tissues, targeting the epitranscriptome also makes context-specific therapies possible.
8.2 RNA Degraders and Proximity-Induced Mechanisms
Endogenous ribonucleases are used by RNA-targeted degraders, including RIBOTACs, to catalytically break down particular RNAs. This method offers multiple benefits by converting occupancy-driven binding into catalytic RNA clearance.
RIBOTACs have been effectively used to achieve functional inhibition at sub-micromolar concentrations for viral RNAs (such SARS-CoV-2 frameshift elements) and carcinogenic miRNAs (like miR-21). This approach expands the therapeutic landscape for RNA targets by demonstrating the synergy between cellular machinery exploitation and structure-based recognition (Costales et al., 2019; Velagapudi & Disney, 2020).
8.3 Covalent and Reversible Covalent RNA Binders
For RNA treatments, covalent targeting techniques are becoming more popular. Covalent binders increase residence duration and potency by forming irreversible connections with ribose hydroxyls or nucleobase functional groups. By creating temporary connections in distinct RNA microenvironments, reversible covalent ligands provide improved specificity.
Recent examples include tiny compounds containing acrylamide that stabilize conformations that interfere with RNA–protein interactions by covalently engaging bulged uridine residues in disease-associated RNAs. When targeting dynamic or shallow RNA pockets, where traditional non-covalent ligands show insufficient affinity, this strategy is very beneficial (Morgan et al., 2017; Haniff et al., 2020).
Additionally, covalent techniques offer chances for allosteric modulation, which permits selective control over RNA function without total knockdown or degradation.
8.4 Combination Strategies with RNA Therapeutics and Biologics
Combining RNA-targeted small compounds with additional RNA therapies can improve their effectiveness, selectivity, and functional results. Examples include:
These combined approaches expand the range of druggable RNA, lower dosage requirements, and slow the emergence of resistance. For instance, in preclinical cancer models, small compounds that target oncogenic miRNA precursors have been demonstrated to enhance ASO-mediated miRNA silencing (Roberts et al., 2020; Disney & Childs-Disney, 2021).
8.5 Future Directions and Translational Potential
Emerging solutions highlight several translational opportunities:
When taken as a whole, these tactics hold the potential to overcome past constraints in cellular delivery, potency, and selectivity, opening the door for next-generation RNA-targeted treatments that are accurate, adaptable, and therapeutically translable.
9. Future Directions and Perspectives
9.1 Integration of AI, Multi-Omics, and Structural Biology
RNA-targeted drug development is about to undergo a revolution thanks to the convergence of artificial intelligence (AI), multi-omics data, and high-resolution structural biology. RNA folding, dynamic ensembles, and ligand-binding pockets can be predicted using AI and machine learning models, and disease-specific RNA targets can be found by multi-omics investigations (transcriptomics, epitranscriptomics, and interactomics). These methods are enhanced by structural biology techniques that offer atomic-level validation, such as cryo-EM, NMR, and X-ray crystallography.
For instance, structure-based discovery of SARS-CoV-2 RNA binders has been made possible by combining AI-guided modeling of viral RNA elements with SHAPE chemical probing, demonstrating the effectiveness of integrated techniques (Vasudevan & Disney, 2022; Townshend et al., 2021). By combining these fields, targets and ligands can be rationally prioritized, which lowers attrition in early discovery.
9.2 Personalized and Precision RNA-Targeted Therapies
By taking use of patient-specific RNA expression patterns, splice variations, or epitranscriptomic changes, RNA-targeted small molecules present prospects for precision medicine. For example, oncogenic miRNAs that are overexpressed in some malignancies could be specifically modulated by small compounds without affecting healthy cells. RNA-targeting techniques can be used with genomic profiling, single-cell RNA-seq, and RNA modification mapping to create customized treatments that reduce off-target toxicity and maximize efficacy (Garcia-Moreno & Chen, 2021).
In oncology, antiviral therapy, and neurological illnesses, where RNA expression and structural variability are context-dependent, this strategy is especially promising.
9.3 Expanding the RNA Druggable Space
The traditional RNA druggable space has been largely limited to rRNA, viral RNAs, and select non-coding RNAs. Emerging strategies are broadening this landscape to include:
Previously "undruggable" RNAs can now be targeted with highly selective small compounds by taking advantage of dynamic RNA motifs, transient conformations, and epitranscriptomic markers. This might possibly address unmet therapeutic needs (Morgan et al., 2017; Chen et al., 2020).
9.4 Outlook for Next-Generation RNA-Targeted Small Molecules
The future of RNA-targeted therapeutics will likely involve multi-modal approaches that combine:
Clinical applications will be made possible by next-generation compounds that balance drug-like characteristics, cell permeability, and specific RNA engagement. In the end, RNA-targeted small molecules may enhance or even outperform protein-targeted treatments in specific disease domains, especially when pathology is caused by RNA dysregulation (Disney & Childs-Disney, 2021; Haniff et al., 2020).
10. Conclusion
A quickly developing area of drug discovery is RNA-targeted small-molecule therapies, which provide chances to modify disease-relevant RNAs that were previously thought to be "undruggable." Rational design tactics that utilize form complementarity, hydrogen bonding, π-stacking, and electrostatic interactions to produce selectivity have been made possible by advances in our understanding of RNA structural hierarchy, dynamics, and epitranscriptomic changes. Potent RNA binders can be found more quickly by combining computational methods like molecular docking, AI-assisted modeling, and generative ligand design with experimental methods like high-throughput and fragment-based screening, chemical probing, and biophysical characterisation.
The potential and difficulties of RNA-targeted small molecules are demonstrated by clinically proven rRNA-targeting antibiotics and new approaches against viral RNAs and carcinogenic non-coding RNAs. The RNA druggable area is being expanded by novel strategies including RIBOTACs, covalent/reversible covalent binders, combination therapies, and epitranscriptomic targeting, but important obstacles like RNA structural heterogeneity, off-target toxicity, and drug-like property optimization still exist.
In the future, the rational design of next-generation RNA-targeted small molecules with enhanced potency, selectivity, and translational potential will be guided by the combination of AI, multi-omics, structural biology, and personalized medicine. A new age of medicines capable of treating a wide range of illnesses, from cancer and viral infections to genetic and neurological disorders, is heralded by these findings, which collectively position RNA as a flexible and clinically actionable target.
11. Acknowledgement
The authors would like to acknowledge the support and guidance of Institution for providing the necessary resources and research environment for the preparation of this review.
12. Conflict of Interest
The authors declare no conflicts of interest related to this review. This research was conducted independently, and no financial or personal relationships influenced the content of this manuscript.
13. References
| Article Type | Research Article |
|---|---|
| Journal Name | Global Journal of Pharmaceutical and Scientific Research |
| ISSN | 3108-0103 |
| Volume | Volume-1 |
| Issue | Issue 3, October-2025 |
| Corresponding Author | Prof. (Dr)Mohd. Wasiullah1*,Prof. (Dr) Piyush Yadav2*, Anshuman Yadav3 |
| Address | Principal , Department of Pharmacy, Prasad Institute of Technology, Jaunpur, Uttar Pradesh, India. 2. Head: Department of Pharma: Chemistry, Prasad Institute of Technology, Jaunpur, Uttar Pradesh, India. 3. Scholar - Department of Pharmacy, Prasad Institute of Technology, Jaunpur, Uttar Pradesh, India. |
| Received | ------ |
| Revised | ------ |
| Accepted | ------ |
| Published | ------ |
| Pages | ----- |