What are the therapeutic candidates targeting RdRp?

11 March 2025
Introduction to RdRp
RNA-dependent RNA polymerase (RdRp) is the central enzyme that drives the replication of RNA viral genomes. In virtually all RNA viruses—ranging from coronaviruses (such as SARS-CoV-2), flaviviruses (like hepatitis C virus and dengue virus), to picornaviruses (such as poliovirus)—RdRp catalyzes the copying of RNA from an RNA template. This replication activity makes RdRp indispensable for viral propagation because without continuous genome synthesis, viruses cannot produce the progeny needed to sustain an infection. The enzyme’s conserved catalytic motifs, present in the “palm,” “fingers,” and “thumb” subdomains, facilitate the binding of nucleotide substrates and metal ions that affect catalysis. Recent advances in structural biology—especially through cryo-electron microscopy and X-ray crystallography—have enabled researchers to resolve detailed structures of RdRp in complex with substrates, inhibitors, and cofactors, thus affirming its critical role in the viral life cycle.

Role of RdRp in Viral Replication
RdRp is responsible for the de novo synthesis of RNA strands by incorporating nucleoside triphosphates (NTPs) into a growing RNA chain, following the sequence of the original genomic RNA template. This enzyme not only drives the replication process but also plays a role in the transcription of viral mRNAs in some virus families, contributing to the expression of viral proteins required for assembly and immune evasion. Because RNA viruses generally lack proofreading activity, RdRp is associated with a relatively high error rate, which contributes both to viral evolution and sometimes to the rapid emergence of drug resistance. Nevertheless, this error-prone nature can be therapeutically exploited: certain antiviral drugs function as mutagenic nucleoside analogues that, when incorporated into the RNA chain, push the virus beyond the threshold of viability. The essential, centrally conserved role of RdRp in viral replication across diverse families thus makes it an exceptionally attractive target for the design of antiviral agents.

Importance of Targeting RdRp in Antiviral Therapy
Inhibiting RdRp can result in “replication catastrophe,” essentially blocking RNA synthesis so that the virus is rendered incapable of reproducing within the host cell. One of the principal advantages of targeting RdRp is its high degree of conservation among different RNA viruses—this structural conservation means that drugs directed against RdRp may have broad-spectrum antiviral activity. Additionally, because human cells do not possess a functional RNA-dependent RNA polymerase, inhibitors designed to target the viral RdRp can be highly selective, reducing the likelihood of deleterious off-target effects on host cellular replication machinery. This favorable therapeutic index, combined with the urgent public health needs seen in recent viral outbreaks such as COVID-19, underscores the critical importance of developing effective RdRp inhibitors in modern antiviral therapy.

Current Therapeutic Candidates
The landscape of antiviral drug development targeting RdRp consists of both approved agents and numerous compounds currently under experimental investigation. Through several rounds of high-throughput screening, structure-based molecular design, and computational docking guided by detailed RdRp structural models, researchers have identified multiple therapeutic candidates with differing mechanisms and pharmacokinetic profiles.

Approved Drugs Targeting RdRp
One of the most widely recognized and rigorously studied RdRp inhibitors is remdesivir. Initially developed for use in Ebola virus infection, remdesivir has gained regulatory approval for the treatment of COVID-19 in several jurisdictions. Remdesivir is a nucleotide analogue prodrug that, upon intracellular metabolism, is converted into its active triphosphate form (RTP). RTP is subsequently incorporated into the growing RNA strand by viral RdRp, causing delayed chain termination by stalling RNA synthesis at a few nucleotides downstream of incorporation. Molecular dynamic simulations have provided insight into remdesivir’s mechanism: the effective steric clash between the incorporated remdesivir moiety and key residues such as S861 prevents efficient translocation of the RNA duplex, culminating in inhibition of viral replication. In clinical settings, remdesivir has shown efficacy in reducing the duration of hospital stays and has been a frontline agent in combating SARS-CoV-2 infection.

Another approved therapeutic candidate that targets RdRp is favipiravir. Unlike remdesivir, favipiravir was originally developed as a treatment for influenza. Favipiravir functions as a ribonucleoside analogue; after being metabolized into its ribofuranosyl 5'-triphosphate form, it not only competes with natural nucleotides for incorporation into the viral RNA but also exerts mutagenic effects leading to error catastrophe during viral replication. Favipiravir’s broad-spectrum activity against RNA viruses, including influenza and potentially SARS-CoV-2, has spurred extensive clinical trials and investigations into its utility as an antiviral agent.

Sofosbuvir, a well-established antiviral approved for hepatitis C virus (HCV) infection, also acts as an RdRp inhibitor. Sofosbuvir is a nucleoside analogue that, once activated in hepatocytes, is incorporated into the viral RNA by HCV RdRp, leading to chain termination. Recent in vitro studies indicate that sofosbuvir may also have activity against other RNA viruses, including SARS-CoV-2, due to the conserved structural and mechanistic features of RdRp across RNA viral families. In clinical practice, sofosbuvir’s tolerability and effectiveness in HCV have prompted investigations into its potential repurposing for emerging RNA virus infections.

Other approved antivirals such as ribavirin have been traditionally used against a variety of RNA viruses. Ribavirin is recognized for its broad-spectrum antiviral activity, although its exact mechanism may involve both direct inhibition of RdRp activity and modulation of immune responses. Ribavirin can, to some degree, impair viral RNA synthesis though it is generally associated with higher toxicity and is often used in combination therapies to mitigate adverse effects.

Additionally, ledipasvir—a drug approved for the treatment of HCV when combined with sofosbuvir—has been explored as a complementary RdRp-targeting candidate. Clinical trial data and in vitro studies have examined its potential synergy with remdesivir, emphasizing that the combination may offer improved antiviral efficacy by simultaneously targeting distinct stages of the viral life cycle, including RNA replication and viral assembly.

In summary, the approved candidate drugs targeting RdRp include:
• Remdesivir – a nucleotide analogue causing delayed chain termination.
• Favipiravir – a ribonucleoside analogue inducing error catastrophe.
• Sofosbuvir – a nucleoside analogue effective against HCV with potential cross-activity.
• Ribavirin – a broad-spectrum antiviral with RdRp inhibitory effects.
• Ledipasvir – used in combination with sofosbuvir and explored for repurposing against other RNA viruses.

Experimental Compounds in Development
Beyond the approved drugs, a wide spectrum of experimental compounds are under evaluation, with many investigations focusing on refining the efficacy, specificity, and resistance profiles of RdRp inhibitors. These include experimental nucleoside and non-nucleoside analogues, small molecules discovered through high-throughput screening, as well as compounds designed using computational docking and molecular dynamics simulations.

One area of significant research has been the development of novel nucleoside analogues that either function as immediate chain terminators or induce error-prone replication upon incorporation into viral RNA. Several studies have utilized all-atom molecular dynamics simulations to compare the binding efficiencies and incorporation probabilities of candidate compounds such as RTP analogues versus natural ATP. For example, simulation studies have shown that remdesivir’s active triphosphate analogue is bound more strongly to the RdRp active site than ATP itself, an observation that has helped guide the design of next-generation nucleoside analogues with improved inhibitory potency.

Furthermore, experimental compounds such as molnupiravir (EIDD-2801) have garnered much interest. Molnupiravir is an orally available prodrug that converts to EIDD-1931, a ribonucleoside analogue incorporated by viral RdRp. Its mechanism differs from remdesivir; instead of causing immediate termination of RNA synthesis, it induces mutagenesis, eventually leading to non-viable virus progeny. Clinical trials on molnupiravir have progressively shown promising results, and it is one of the candidates that represent a new generation of broad-spectrum RdRp inhibitors targeting SARS-CoV-2.

Another promising direction is the identification of allosteric inhibitors and non-nucleoside compounds that bind to sites on RdRp distinct from the NTP entry channel. These compounds can induce conformational changes that indirectly hamper the enzyme’s catalytic activity. Computational docking studies have revealed a number of small molecules that stably interact with key residues in the active site and sometimes with accessory sites, inhibiting the replication process. Such candidates are still in the pre-clinical or early clinical phases, and many rely on structure-based design approaches using high-resolution structures of the RdRp holoenzyme.

Beyond nucleoside analogues, researchers are also exploring peptide-based inhibitors that mimic critical sites of interaction on the RdRp or interfere with the formation of the replication complex. A targeted approach involves designing peptides that compete for binding domains that are essential for the association of RdRp with viral cofactors such as nsp7 and nsp8 in coronaviruses. Although this strategy presents challenges in terms of peptide stability and delivery, advances in formulation chemistry and drug delivery systems continue to enhance their therapeutic viability.

An emerging trend is the repurposing of drugs with known cross-target activity. Through extensive in silico screening workflows, including reverse docking and deep learning–assisted binding affinity predictions, compounds originally developed for other indications have been identified as potential RdRp inhibitors. Examples include certain kinase inhibitors and allosteric modulators that, surprisingly, display high binding affinities for the RdRp active site in computational models. While these findings are promising, they require thorough experimental validation in biochemical and cell-based assays before clinical development can proceed.

Thus, the experimental therapeutic candidates targeting RdRp can be classified as:
• Next-generation nucleoside analogues—including refined RTP analogues designed via structure-based methods.
• Orally bioavailable prodrugs like molnupiravir (EIDD-2801) that induce viral mutagenesis.
• Non-nucleoside, allosteric inhibitors identified through computational docking and molecular dynamics simulations.
• Peptide inhibitors that disrupt RdRp complex formation.
• Repurposed small molecules and kinase inhibitors discovered via high-throughput in silico screening.

Mechanisms of Action
Understanding the inhibition mechanisms of RdRp-targeted therapeutics is essential for both refining current drug candidates and for designing new agents with improved efficacy and reduced adverse effects.

Inhibition Mechanisms of RdRp
RdRp inhibitors generally exert their antiviral effects through one or several convergent mechanisms. The primary mechanism seen with nucleoside analogues such as remdesivir is chain termination. When remdesivir is metabolized intracellularly into its active RTP form, it is incorporated into the nascent RNA chain. Biophysical and molecular dynamics studies have shown that once RTP is incorporated, its modified chemical structure—in particular, a 1′-cyano group—interferes sterically with adjacent residues in the active site (e.g., S861). This interference halts further nucleotide addition in a delayed manner (often 3 nucleotides post-incorporation), ultimately leading to premature termination of RNA synthesis.

Another well-established mechanism is the induction of lethal mutagenesis, as seen with favipiravir and molnupiravir. These drugs are incorporated into viral RNA and cause base mispairing during subsequent replication cycles. Favipiravir, through its ribonucleoside triphosphate derivative, leads to a higher error rate during RNA synthesis, and over successive replication rounds, the accumulation of errors renders the viral genome non-functional. Molnupiravir follows a similar mechanistic pathway by inducing mutagenesis, thus resulting in the collapse of the viral replication process.

In addition, non-nucleoside inhibitors function by binding allosterically to regions of the RdRp that are distant from the catalytic active site. This binding can result in conformational changes that misalign the catalytic residues or disrupt the formation of the functional replication complex. The advantage of allosteric inhibitors lies in their potential to retain activity even in the face of mutations near the active site, which are common due to the high replication error rate of RNA viruses. These inhibitors work either by reducing the binding affinity of natural nucleotides or by altering the overall structural dynamics of RdRp, thereby attenuating its enzymatic activity without directly competing with nucleotides.

Moreover, some experimental approaches seek to inhibit RdRp by targeting its interaction with essential viral cofactors. In coronaviruses, for instance, the RdRp functions as part of a multi-subunit complex that includes nsp7 and nsp8. By preventing or weakening the association between these proteins, peptide-based inhibitors or small molecules may destabilize the active replication complex. This approach, while still experimental, may offer a means to complement the action of nucleoside analogues, especially in viral infections where complex assembly is an additional layer of regulation.

Case Studies of Specific Inhibitors
Remdesivir serves as a paradigm for RdRp-targeting therapeutics. Detailed molecular studies have demonstrated how remdesivir’s incorporation into the RNA chain leads to stalling at specific nucleotide positions, a phenomenon frequently described as delayed chain termination. The crystal structures of the RdRp complex bound with remdesivir metabolites provide compelling evidence of the conformational changes induced by its incorporation, with key interactions being mapped to residues like K545, R553, and S861. These structural insights have been used to rationally optimize remdesivir analogues and guide the design of next-generation inhibitors.

Favipiravir also provides an illustrative case where an antiviral agent induces error catastrophe. Clinical and laboratory data demonstrate that favipiravir’s ribonucleoside analogue is preferentially incorporated by RdRp, causing mismatches during RNA synthesis. Over time, these mismatches accumulate to a level that is incompatible with viral survival. The broader spectrum of activity observed with favipiravir against other RNA viruses emphasizes the potential utility of modulating RdRp fidelity as a therapeutic strategy.

Molnupiravir is another notable case study. Unlike classic chain terminators, molnupiravir works by engaging RdRp and being misincorporated into viral RNA, thus inducing high mutation frequencies. This novel mechanism differentiates molnupiravir from drugs like remdesivir, while still targeting the same central enzyme. Clinical trials of molnupiravir have demonstrated reduced viral loads in infected individuals, and structural studies corroborate its binding within the RdRp active site, reinforcing its mechanism of action.

Recent in silico and in vitro studies have also highlighted several novel compounds that bind to RdRp’s NTP entry site and allosteric pockets. For instance, docking studies indicate that certain experimental inhibitors exhibit high binding affinity toward not only the catalytic site but also putative regulatory sites that modulate the enzyme’s activity. These insights, derived from extended molecular dynamics simulations and free energy perturbation analyses, support the development of dual-action inhibitors that combine chain termination with structural destabilization of the replication complex.

Another area of active investigation involves repurposed drugs discovered via computational analysis. Several compounds originally developed for non-antiviral indications have been predicted to bind RdRp with high specificity. These include agents that, while initially targeting cellular kinases or other enzymes, show unexpected affinity for conserved RdRp structural motifs. Although these agents are still in early evaluation phases, they represent an exciting new frontier in broad-spectrum antiviral development.

Challenges and Future Directions
While significant progress has been made in identifying and developing therapeutic candidates that target RdRp, numerous challenges remain. These challenges span the domains of drug design, clinical efficacy, resistance monitoring, and drug delivery, and they underscore the dynamic nature of antiviral drug development.

Current Challenges in Targeting RdRp
One major challenge in targeting RdRp is the rapid emergence of resistance. RNA viruses inherently exhibit high mutation rates due to the lack of proofreading functions in their RdRp. As a result, the viral genome can quickly accumulate mutations that reduce drug binding affinity or alter the conformation of the catalytic site. For instance, resistance mutations near residues critical for remdesivir binding have been documented in some in vitro studies, necessitating the design of drugs with high genetic barriers to resistance and the potential need for combination therapies to offset resistance development.

Another significant challenge pertains to the pharmacokinetic and pharmacodynamic limitations of RdRp inhibitors. Drugs such as remdesivir must undergo complex intracellular activation steps to be converted into their active triphosphate forms, and differences in metabolic activation across patients can lead to variable therapeutic outcomes. Similarly, the oral bioavailability of certain RdRp-targeting agents may be limited, which can hinder their widespread use, particularly in resource-limited settings where intravenous administration is less feasible.

Furthermore, achieving a balance between potency and toxicity remains a delicate issue. Although RdRp is absent in human cells and thus presents an attractive target, off-target effects, particularly in cellular metabolism or mitochondrial function, can occur with high doses of nucleoside analogues. Detailed studies of drug–target interactions using high-resolution structural methods and in-depth clinical pharmacology are essential to fine-tune the therapeutic window of these compounds.

Another challenge is the heterogeneity of RdRp across different virus families. While the enzyme is highly conserved, subtle sequence and structural differences can affect drug binding. This variability may limit the spectrum of activity for a given inhibitor, or on the contrary, may require the development of multiple compounds to address different viral RdRp effectively. Such complexities necessitate tailored approaches, including broad-spectrum agents that target invariant residues or structural regions that are conserved across multiple viruses.

The robustness of biochemical assays used to evaluate RdRp inhibition is also a notable hurdle. In vitro replication assays, often relying on purified enzymes and synthetic RNA templates, may not fully recapitulate the complexities of the intracellular environment, including the presence of viral cofactors or host proteins. Thus, ensuring that laboratory findings translate into clinical efficacy is a persistent challenge in drug development.

Future Research and Development Directions
Looking forward, several research directions hold promise for overcoming the current limitations in RdRp-targeted therapy. One key strategy is the continued integration of high-throughput screening with advanced computational techniques. Structure-based drug design, incorporating machine learning and deep neural network predictions, is increasingly being used to identify novel chemical scaffolds that bind RdRp with high affinity and specificity. Such approaches not only refine our understanding of binding interactions at an atomic level but also accelerate the identification of lead compounds from vast chemical libraries.

Another promising avenue is the development of combination therapies. Given the propensity of RNA viruses to develop resistance, using multiple drugs that target different steps in the viral replication cycle may provide synergistic effects and minimize resistance development. For example, combining nucleoside analogues (such as remdesivir or molnupiravir) with inhibitors that disrupt RdRp–cofactor interactions or with drugs targeting viral entry and release could form a multi-pronged strategy that suppresses viral replication more effectively than monotherapy.

Advancements in the field of drug repurposing also promise to expedite the pipeline of RdRp inhibitors. By mining large drug-relevant databases and employing signature reversal principles, researchers are identifying compounds with previously unrecognized activity against RdRp. Drugs that have already been approved for other indications—owing to their well-established safety profiles—can potentially be rapidly transitioned into clinical trials for emerging viral infections.

Additionally, evaluation of drug candidates in more physiologically relevant systems, such as three-dimensional organoids and animal models that recapitulate human infection, is crucial. These models can provide more accurate predictions of a compound’s efficacy and toxicity in vivo and can reveal pharmacodynamic interactions that might not be observable in conventional cell-based assays. The use of such advanced screening methods, combined with real-time monitoring of RdRp activity via innovative biosensors, will greatly enhance our ability to track the dynamics of viral replication and intervention.

Another area requiring focused attention is the optimization of drug delivery. Improving the bioavailability of RdRp inhibitors, especially those intended for oral administration, is paramount. Research into nanoparticle-based formulations, prodrug strategies, and targeted delivery systems may help ensure that active drug concentrations reach infected tissues with minimal systemic exposure. This is particularly important for drugs like remdesivir, which currently necessitate intravenous administration, and for future candidates that aim to expand treatment options beyond hospital settings.

On the regulatory and clinical fronts, the ongoing collaboration between industry, academia, and regulatory agencies is expected to streamline the development, evaluation, and deployment of RdRp inhibitors. Adaptive clinical trial designs that allow for modifications based on emerging efficacy and safety data, along with enhanced genomic surveillance to detect resistance mutations, can support more rapid and safe clinical translation of promising compounds.

Finally, understanding the interplay between viral replication mechanisms and host immune responses represents an exciting frontier. Combining RdRp inhibition with immune modulatory therapies might offer additive or synergistic effects, potentially leading to improved patient outcomes. As our understanding of the innate and adaptive immune responses to viral infections deepens, personalized therapeutic regimens that integrate RdRp inhibitors with immune-based interventions may become a reality, paving the way for next-generation antiviral strategies.

Conclusion
In conclusion, therapeutic candidates targeting RdRp constitute a diverse and evolving class of antiviral agents. On a broad-based level, the enzyme’s central role in the replication of RNA viruses and its absence from human biochemical pathways make it an ideal target for therapeutic intervention. Approved drugs such as remdesivir, favipiravir, sofosbuvir, ribavirin, and ledipasvir have already demonstrated clinical value by interfering with viral RNA synthesis through mechanisms including chain termination and the induction of error catastrophe. These successful agents have been characterized by detailed molecular studies that have elucidated critical binding interactions within the RdRp active site, leveraging high-resolution structural data and molecular dynamics simulations.

Simultaneously, experimental compounds continue to emerge from state-of-the-art screening techniques and computational models. Next-generation nucleoside analogues, non-nucleoside allosteric inhibitors, peptide-based disruptors of RdRp complex formation, and repurposed small molecules are being pursued to widen the spectrum of antiviral interventions and to overcome challenges such as resistance and suboptimal pharmacokinetic profiles. Inhibition strategies range from direct incorporation of nucleotide analogues that cause chain termination, to allosteric modulation that disturbs the enzyme’s conformational dynamics, demonstrating the multifaceted approaches researchers have employed to disable viral replication.

Nevertheless, targeting RdRp is not without its challenges. The high mutation rates of RNA viruses drive the rapid emergence of resistance, and achieving optimal drug activation and delivery remains a critical barrier. Bridging the translational gap between in vitro efficacy and clinical performance requires robust models and innovative methods to ensure that RdRp inhibitors display consistent activity in vivo. Future research directions that integrate advanced drug discovery methodologies, combination therapies, and improved delivery technologies offer a promising pathway to surmount these limitations.

Overall, the current state of therapeutic candidates targeting RdRp represents a convergence of detailed molecular insights, innovative drug design, and clinical urgency, reflecting both the progress achieved and the challenges that remain. Continued interdisciplinary research, leveraging the power of computational methods, high-throughput screening, and advanced clinical trial designs, is expected to further expand the arsenal of RdRp inhibitors and ultimately improve patient care in the face of viral epidemics. The progress in this field promises not only more effective treatments for current viral infections such as COVID-19 but also a broader range of antivirals capable of responding to emerging pathogens in future outbreaks.

In summary, the therapeutic landscape for targeting RdRp is characterized by a well-established group of approved drugs and an expanding pipeline of experimental candidates. These agents harness a variety of mechanisms to disrupt the essential process of viral RNA replication, ranging from chain termination to allosteric antagonism, and they hold promise for broad-spectrum antiviral therapy. With advances in structural biology, computational drug design, and personalized medicine approaches, future research is poised to overcome existing challenges and usher in a new era of antiviral therapeutics.

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.