How many FDA approved saRNAs are there?

17 March 2025

Introduction to Small Activating RNAs (saRNAs)

Small activating RNAs (saRNAs) are a class of small double‐stranded RNAs, typically around 21 nucleotides in length, that work via a mechanism known as RNA activation (RNAa). In contrast to small interfering RNAs (siRNAs) that mediate gene silencing by degrading messenger RNA (mRNA), saRNAs are designed to bind to promoter regions of target genes and enhance their transcription. This upregulation of gene expression is mediated by the recruitment of transcriptional activators and chromatin modifying complexes, which eventually leads to increased production of the encoded protein. The intrinsic nature of saRNAs offers a unique mode of action wherein a relatively small dose might yield a sustained increase in the levels of certain proteins, thereby representing a potential strategy for therapeutic applications in diseases characterized by the under-expression of beneficial genes.

Historical Development and Research Milestones
The concept of RNA activation emerged over a decade ago when researchers observed that a subset of small duplex RNAs, contrary to expectations, could upregulate target gene expression rather than silence it. Early investigations laid the foundation by demonstrating that these small RNAs could target genes like p21, VEGFA, and E-cadherin, thereby influencing cellular pathways that had previously been considered beyond pharmacological reach. With advances in understanding the molecular details of RNA-triggered transcriptional activation, research in this field gained momentum. In more recent years, the focus has shifted toward harnessing this mechanism to activate tumor suppressor genes and restore their function in disease conditions. Notably, preclinical studies have investigated saRNAs designed to activate the transcription factor CCAAT/enhancer-binding protein alpha (CEBPα), a critical regulator in liver function and a recognized tumor suppressor. These developments have pieced together a narrative of incremental advancements—from initial observations to targeted design—underscoring the therapeutic potential of saRNAs while also highlighting the challenges that must be addressed prior to clinical translation.

FDA Approval Process for saRNAs

Overview of FDA Approval Stages
The U.S. Food and Drug Administration (FDA) approval process for any therapeutic, including RNA-based agents, follows a rigorous multistage evaluation to ensure safety, efficacy, and quality. This process typically involves:

1. Preclinical Studies: Extensive laboratory and animal studies designed to investigate the pharmacodynamics, toxicity profiles, and potential off-target effects of the candidate molecule. Given the novelty of saRNAs, these studies emphasize the stability of the RNA, its biodistribution, and the mechanisms by which it increases gene expression. 
2. Investigational New Drug (IND) Application: Once promising preclinical data have been gathered, the developer needs to submit an IND application to the FDA. This ensures that the candidate saRNA can be safely administered to humans in early phase clinical trials. 
3. Clinical Trials (Phase I–III): 
- Phase I focuses on safety, dosing, and initial pharmacokinetic and pharmacodynamic parameters in a small group of healthy volunteers or patients. 
- Phase II expands the study to a larger patient population to assess the drug’s efficacy and further evaluate its safety profile. 
- Phase III involves large-scale testing to confirm efficacy, monitor adverse reactions, and compare the saRNA with standard or placebo treatments under controlled conditions. 
4. New Drug Application (NDA): After successful clinical evaluations, a comprehensive NDA is submitted, which includes all data from preclinical and clinical studies, manufacturing details, and proposed labeling. 
5. FDA Review and Approval: Further analysis by the FDA reviewers results in either approval, a request for additional data, or rejection of the therapeutic.

Each of these steps is tailored to the specific challenges posed by RNA-based therapies such as stability, delivery, immunogenicity, and manufacturing consistency—all issues that are particularly pronounced in novel modalities like saRNAs.

Specific Requirements for RNA-based Therapies
The FDA has established several specific requirements for RNA-based therapies given their unique characteristics. Among these are:

- Chemical Modification and Stability: RNA molecules, by nature, are susceptible to degradation by nucleases. Therefore, RNA therapeutics—saRNAs included—must be chemically modified to enhance their stability while preserving their functional integrity. 
- Delivery Systems: The delivery of RNA molecules into target cells remains one of the most significant hurdles. For an saRNA to be effective, it must be delivered intact into the cytoplasm, where it can reach the nucleus to exert its transcriptional activation effect. This often involves specialized carriers such as lipid nanoparticles (LNPs) that have demonstrated success in other RNA therapies (e.g., mRNA vaccines and siRNAs). 
- Off-target Effects & Specificity: RNA-based drugs need to be meticulously designed to minimize off-target effects that could lead to unintended gene activation or interference with other cellular processes. 
- Manufacturing and Scalability: Owing to their complex structures and modification requirements, the reproducibility and scalability of manufacturing processes are key considerations.

For saRNAs, these requirements are still under intensive investigation, and while there is notable optimism about their future applications, the data supporting their safety and efficacy in large human populations are still evolving.

Current FDA-Approved saRNAs

List of Approved saRNA Therapies
At the present time, there are no FDA-approved saRNAs. Despite significant preclinical research and early-phase clinical trials, no saRNA therapeutic has yet met the stringent requirements for FDA approval. For instance, MTL-CEBPA—a saRNA designed to upregulate the tumor suppressor gene CEBPα for hepatocellular carcinoma—is currently in early clinical development and undergoing evaluation in Phase I clinical trials. The absence of any FDA-approved saRNA can largely be attributed to the fact that the saRNA modality is still emerging as a therapeutic strategy compared to the better-established RNAi mechanisms underlying siRNA and antisense oligonucleotide therapies.

Therapeutic Areas and Indications
While saRNAs are being actively explored for various therapeutic applications—including cancer therapeutics aimed at reactivating tumor suppressor genes and potentially for genetic diseases associated with haploinsufficiency—their current status remains investigational. The therapeutic areas envisioned for saRNA applications include:

- Oncology: Activation of tumor suppressor genes such as CEBPα for liver and other cancers has been a primary focus, as seen in preclinical studies and early clinical trials. 
- Inflammatory Diseases: Some patent applications describe small RNA medicaments (which may sometimes include saRNA-based strategies) for the prevention and treatment of inflammatory diseases by modulating cytokine expression (e.g., IL-1β, IL-6, and TNF-α). Although these patents reflect a broad interest in using small RNAs for therapeutic activation, they do not translate into an FDA-approved saRNA product yet. 
- Potential Protein Replacement Therapies: There is ongoing research into using saRNAs to restore the expression of genes that are underexpressed in various disease conditions. However, these approaches remain at a very early stage of development.

Thus, while the landscape for RNA-based therapeutics is rich—with several siRNA, mRNA, and antisense products securing FDA approval—the specific category of saRNAs remains in the developmental pipeline. No saRNA product has yet advanced through all clinical trial phases to secure formal FDA approval.

Future Directions and Challenges

Ongoing Clinical Trials and Research
The future prospects for saRNA therapeutics are promising, with numerous ongoing clinical trials seeking to demonstrate both the safety and efficacy of these molecules. For example, the MTL-CEBPA saRNA is currently being evaluated in a Phase I clinical trial for hepatocellular carcinoma. This trial represents a significant milestone as it is the first clinical investigation of its kind, aiming to prove the concept that saRNAs can be effective in upregulating therapeutic genes in vivo. 

Alongside single-agent clinical trials, there is growing interest in combination therapies, where saRNAs may be used in conjunction with other treatment modalities to enhance therapeutic outcomes—for example, pairing them with immunotherapies or standard chemotherapeutic agents. Preclinical studies have also explored diverse delivery systems, including advanced LNP formulations and other nanocarriers, to overcome delivery challenges and improve the bioavailability of saRNAs. Moreover, advanced computational methods and high-throughput screening approaches are being developed to optimize saRNA design, thereby minimizing off-target effects and improving specificity.

Potential Challenges in saRNA Development
Despite the enthusiasm for saRNA-based therapies, several challenges still need to be addressed for successful translation from bench to bedside. These include: 

- Delivery and Cellular Uptake: Efficient intracellular delivery remains a critical challenge. Because saRNAs must reach the nucleus to activate transcription, the design of delivery systems that can navigate both cytoplasmic and nuclear membranes without triggering significant immune responses is paramount. 
- Stability and Pharmacokinetics: RNA molecules are inherently unstable due to RNase-mediated degradation. While chemical modification can enhance stability, ensuring that these modifications do not interfere with the RNA’s activity is a delicate balance that continues to require optimization. 
- Off-target Effects and Safety: Given that saRNAs work by activating gene transcription, there is a risk that they could inadvertently upregulate non-target genes, leading to unpredictable biological effects and potential toxicity. Enhanced sequence designs and comprehensive preclinical assessments are vital to mitigate such risks. 
- Regulatory and Manufacturing Considerations: With no precedent of an FDA-approved saRNA therapy, the regulatory environment for these molecules is still evolving. Developers must demonstrate not only robust clinical efficacy and safety but also maintain a high standard of manufacturing reproducibility and scalability. 
- Therapeutic Window and Dosing Challenges: Identifying the optimal dosing regimen is particularly critical in ensuring that the gene activation observed translates into meaningful therapeutic benefits without causing overactivation, which could lead to other pathogenic effects. Early-phase trials like that of MTL-CEBPA are expected to provide insights into these dynamics.

The ongoing research efforts to overcome these challenges are encouraging. Continued investment in delivery technologies and deeper mechanistic studies using advanced sequencing and proteomic approaches are likely to pave the way for more refined saRNA candidates in the future.

Conclusion
In summary, while small activating RNAs (saRNAs) represent an innovative and potentially transformative approach within the broader landscape of RNA therapeutics, they have not yet reached the stage of FDA approval. Despite their promising mechanism—whereby they specifically upregulate gene expression through RNA activation—and positive indications from preclinical studies and early-phase clinical trials (such as the ongoing evaluation of MTL-CEBPA in hepatocellular carcinoma), there remain significant challenges in terms of delivery, stability, specificity, and regulatory requirements. 

From a general perspective, the development of RNA therapeutics has seen significant achievements with approved siRNA, antisense oligonucleotides, and even mRNA-based vaccines; however, the subclass of saRNAs is still in its nascent stages relative to these more established technologies. In a more specific perspective, while numerous patent disclosures and early clinical investigations attest to the therapeutic potential of saRNAs in various indications—including cancer and inflammatory diseases—none of these candidates have yet traversed the full spectrum of clinical trials to achieve FDA approval, meaning the total number of FDA-approved saRNAs is currently zero. Finally, on a general note, the future of saRNA development hinges on overcoming the outlined challenges through advanced delivery mechanisms, improved chemical modifications, and refined clinical evaluation protocols, which may eventually lead to FDA-approved products that can address currently “undruggable” therapeutic targets. 

In conclusion, based on the available data from the synapse sources and the current state of clinical research, there are 0 FDA-approved saRNAs to date. This outcome highlights the emerging nature of the saRNA class and underscores the significant potential yet to be unlocked should the ongoing challenges be successfully addressed.

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.