What saRNAs are being developed?

17 March 2025
Introduction to Small Activating RNAs (saRNAs)
Small activating RNAs (saRNAs) are a relatively novel class of double-stranded RNA molecules that, unlike the classical small interfering RNAs (siRNAs) which function in RNA interference by silencing gene expression, instead upregulate the expression of target genes through a process known as RNA activation (RNAa). Their mechanism involves the binding of these short RNAs to complementary sequences in the promoter regions of genes, thereby influencing transcriptional machinery and chromatin structure to elevate gene expression. Because of their small size and ability to leverage the cellular transcription machinery without introducing exogenous genetic material, saRNAs have gained attention as a tool with potential for therapeutic applications in diseases where reactivation of suppressed genes is beneficial.

Definition and Mechanism of Action
saRNAs are typically around 21 nucleotides in length and are structurally similar to siRNAs; however, their mode of action is fundamentally different. Instead of guiding the RNA-induced silencing complex (RISC) to degrade messenger RNA, saRNAs target gene promoters and stimulate the recruitment of transcriptional co-factors and RNA polymerase II, which leads to increased transcription of the endogenous gene. This mechanism, known as RNA activation (RNAa), can result in sustained elevation of target gene expression without permanently altering the genome. The selectivity of saRNAs is achieved through base pairing between the antisense strand of the saRNA and sequences within the promoter region, and even minor alterations in the seed region of the saRNA can affect specificity and efficiency.

Historical Development and Significance
The concept of using RNA molecules to activate gene transcription was first described in the mid-2000s. Landmark studies demonstrated that short double-stranded RNAs targeting promoters of genes such as p21, VEGF, and E-cadherin could actually enhance their expression rather than silence them. This unexpected phenomenon flipped the conventional view of RNA therapeutics on its head. Over time, research groups around the world have validated that RNA activation is an evolutionarily conserved mechanism observed in various eukaryotic systems, thus establishing saRNAs not only as a promising investigative tool for gene regulation studies but also as potential therapeutic agents. The significance of saRNAs lies in their ability to “rescue” the downregulated expression of critical genes in conditions such as cancer and monogenic disorders, thereby “expanding the druggable genome” to targets that were previously considered undruggable.

Current saRNA Development Projects
The development of saRNAs has progressed from basic research into translational projects aimed at clinical utility. In several disease contexts—most notably in oncology—saRNAs are being harnessed to restore appropriate gene expression profiles and to modulate pathways that have gone awry.

Leading Research Institutions and Companies
A number of research institutions and biotechnology companies have emerged at the forefront of saRNA development. For example, MiNA Therapeutics, based in London, is a key player in the saRNA landscape. Their research focuses on developing saRNA-based therapeutics for liver disorders and cancer. One of their lead candidates is MTL-CEBPA, a saRNA designed to upregulate the transcription factor CCAAT/enhancer-binding protein alpha (CEBPα), a critical tumor suppressor in hepatocellular carcinoma (HCC). This candidate is being evaluated in clinical trials and represents a promising advancement in the use of saRNAs for protein replacement therapy.

Eli Lilly has also recognized the transformative potential of saRNA technology. Recently, Eli Lilly entered into a global research agreement with MiNA Therapeutics to explore saRNA-based drug candidates across key therapeutic areas, including cancer, immunology, and neurodegenerative diseases. This collaboration highlights industry interest in expanding the applications of saRNAs as a new modality that works at the gene level. Additionally, academic research groups have contributed significantly, helping to refine design criteria for saRNAs and elucidate their intracellular mechanisms of action.

Moreover, patent activity in this space further underscores active development. For instance, patents such as those for saRNA compositions and methods of use, and specifically for SIRT1-saRNA compositions designed to upregulate the SIRT1 gene, demonstrate that both small molecules acting on gene promoters and more complex constructs are under intellectual property protection. These filings are indicative of the growing confidence and investment in the technology among biotech companies and research institutions.

saRNAs in Clinical Trials
The translation of saRNA candidates from bench to bedside is underway, with several candidates advancing into clinical trials. MTL-CEBPA is one of the most notable examples. This saRNA is designed to reverse the downregulation of CEBPα—a transcription factor implicated in hepatocyte function and tumor suppression—in patients with advanced liver cancer. Clinical trial data to date have shown encouraging signals that support the potential for saRNA-mediated gene activation to overcome immunosuppressive effects in the tumor microenvironment.

Other clinical investigations have focused on different therapeutic areas. In colorectal cancer, for example, preclinical models have shown that saRNAs can induce the upregulation of tumor suppressor genes that are downregulated in cancer, thereby inhibiting cell proliferation and metastasis. Although most clinical trials have initially focused on cancer indications, the scope of saRNA development appears to be broadening to include chronic and monogenic disorders, where restoring the expression of a deficient protein can have profound therapeutic benefits.

Additionally, collaborations between pharmaceutical companies and biotech firms are accelerating clinical translation. The partnership between Eli Lilly and MiNA Therapeutics is anticipated to yield several drug candidates that will eventually be tested in human subjects. The active exploration in targeted delivery methods, such as using lipid nanoparticles and conjugate-based systems, is also central to these clinical trials. Ensuring efficient, safe, and cell-specific delivery remains a crucial aspect of current saRNA clinical development.

Applications of saRNAs
saRNAs are not merely research tools; their potential for broad therapeutic applications has been a major driver behind their development. Their ability to specifically upregulate gene expression opens novel avenues for treating a variety of diseases, especially where gene downregulation or haploinsufficiency is a central problem.

Therapeutic Applications
Therapeutically, saRNAs are being developed to treat diseases where restoration of endogenous gene expression could reverse pathological phenotypes. In oncology, several cancer-associated genes—including tumor suppressors—are frequently suppressed in malignant cells. By using saRNAs to upregulate these genes, researchers can potentially reinstate normal cell regulatory pathways. MTL-CEBPA, for example, represents a therapeutic approach where reactivating CEBPα expression in hepatocellular carcinoma has been shown to reverse the immunosuppressive state of myeloid cells and enhance the efficacy of concomitant anticancer drugs.

Beyond cancer, there are emerging applications in the treatment of monogenic disorders. Certain genetic diseases result from haploinsufficiency, where a mutant allele causes insufficient expression of a key protein. saRNAs offer a strategy to boost the expression of the remaining functional allele, thus ameliorating disease symptoms without the risks associated with viral gene therapy vectors. Similarly, saRNA applications are being explored in the context of metabolic disorders, immunodeficiencies, and even neurodegenerative diseases, where the restoration of a protein’s normal function can lead to marked clinical improvements.

The overall therapeutic advantage of saRNAs rests in their reversibility and precision. Unlike permanent gene editing techniques, saRNA-induced gene activation is transient and adjustable, allowing clinicians to titrate the therapeutic effect more safely. This facet makes them especially attractive for conditions where sustained overexpression might lead to deleterious off-target effects.

Potential in Gene Regulation
At the molecular level, saRNAs provide researchers with a powerful tool to dissect gene regulatory networks. By intuitively activating specific genes, scientists can probe the function of individual genes within larger signaling pathways and determine how their increased expression affects cellular phenotypes. This capability has already led to major insights in cancer biology, where understanding the interplay between oncogenes and tumor suppressor genes is crucial.

Furthermore, saRNAs have the potential to serve as tools for epigenetic reprogramming. Their interaction with promoter regions and subsequent recruitment of transcriptional co-factors may modify the local chromatin environment, thereby inducing epigenetic changes that persist beyond the immediate period of saRNA activity. This opens the door to using saRNAs not only as short-term therapeutics but also as modulators of long-term cellular differentiation and function.

The degree of specificity achieved by saRNAs is critical in these contexts. With optimized design rules—such as targeting promoter regions within –100 to –1000 base pairs of the transcription start site and calibrating GC content and thermodynamic stability at the ends—the efficiency of saRNA-mediated gene activation has been enhanced remarkably in preclinical models. Consequently, this refined design strategy is expected to improve both the potency and safety profile of saRNA-based therapeutics in clinical settings.

Challenges and Future Directions
In spite of the excitement surrounding saRNAs, several challenges must be overcome before these molecules can be widely adopted as standard therapeutic agents. Researchers are actively addressing issues related to delivery, specificity, immunogenicity, and regulatory approval.

Technical and Regulatory Challenges
One of the predominant technical challenges in saRNA development is ensuring efficient cellular delivery. Like many RNA-based therapeutics, saRNAs are inherently susceptible to degradation by nucleases, and they are prone to rapid clearance from the bloodstream. To circumvent these problems, various delivery systems such as lipid nanoparticles (LNPs), polymeric carriers, and ligand-conjugated approaches are being optimized for saRNA delivery. These advanced formulations not only protect the RNA molecules but also facilitate targeted uptake by the intended cell types.

Another technical aspect is the inherent variability in saRNA activity due to sequence-dependent effects. Because transcriptional activation is exquisitely sensitive to the precise location and sequence of the target site within a promoter region, only a fraction of designed saRNAs typically exhibit the desired upregulation. This “hit-or-miss” process necessitates the rigorous application of design rules developed by pioneering groups in the field. Improving in silico prediction models and high-throughput screening methods is critical to streamline the discovery and optimization of potent saRNAs.

Immunogenicity is additionally a concern. As with other RNA molecules, exogenous saRNAs can provoke immune responses if not properly modified. Chemical modifications are therefore employed to reduce recognition by pattern recognition receptors and mitigate unwanted inflammatory reactions. However, the balance between achieving sufficient stability/delivery and avoiding adverse immune activation remains delicate and is an active area of research.

Regulatory hurdles also represent a significant challenge. Because saRNAs constitute a novel therapeutic class with mechanisms distinct from traditional drugs, regulatory agencies require robust demonstration of both efficacy and safety. This challenge is compounded by the fact that saRNAs often work indirectly—by modulating transcription rather than directly targeting proteins—so correlating molecular endpoints with clinical outcomes requires comprehensive studies. In addition, standardized manufacturing protocols and quality controls for RNA-based products are still evolving, further complicating the approval process.

Future Prospects and Research Directions
Looking ahead, the prospects for saRNA therapeutics are very promising. Continued refinements in RNA chemistry and delivery technologies are likely to overcome many of the current obstacles. On the research front, scientists are exploring a number of avenues to improve the clinical potential of saRNAs:

1. Improved Design Algorithms: Enhanced bioinformatic tools that predict effective saRNA sequences based on promoter architecture and epigenetic context will increase the success rate of candidate molecules. These tools are expected to further narrow the “hit-or-miss” nature of current approaches, accelerating the translation from research to clinical application.

2. Novel Delivery Platforms: The rapid development of nanotechnology-based carriers—such as LNPs optimized for RNA delivery—will likely be repurposed for saRNAs. Innovations in conjugation techniques that combine targeting ligands with RNA-binding entities can ensure that saRNAs are delivered in a cell-type-specific manner, increasing therapeutic safety and efficacy.

3. Expanding Clinical Indications: While initial clinical trials have largely focused on oncology—particularly liver cancer and colorectal carcinoma—the potential applications of saRNAs extend to a wide range of conditions, including metabolic disorders, neurodegenerative diseases, and monogenic disorders. This broad potential is encouraging both for researchers and for investors aiming to fill previously unmet medical needs.

4. Combination Therapies: saRNAs are increasingly being seen not only as stand-alone agents but also as part of combination therapies. For instance, restoring the expression of a tumor suppressor gene in conjunction with immune checkpoint inhibitors may offer synergistic benefits in cancer treatment. Similar strategies may be applicable in other complex diseases where multiple pathways are dysregulated.

5. Long-lasting and Adaptive Therapies: By harnessing the potential for epigenetic reprogramming, saRNAs might not only provide transient boosts in gene expression but also induce more sustained changes in cell phenotype. This could be particularly useful in conditions where long-term augmentation of gene expression is required without the risks of permanent genomic alterations.

6. Integration with Personalized Medicine: With the advent of high-throughput sequencing and personalized genomic analysis, it becomes possible to design patient-specific saRNA molecules tailored to individual transcriptional deficiencies. This integration of saRNA technology with precision medicine approaches holds promise for customized therapies that maximize patient benefit while minimizing side effects.

7. Patent and IP Landscape Expansion: Ongoing patent filings regarding saRNA compositions and methods—such as those for SIRT1-saRNA—show that the intellectual property landscape is actively evolving. This not only validates the technological potential of saRNAs but also encourages further investment and innovation in this field.

8. Cross-disciplinary Collaborations: The strategic partnerships between major pharmaceutical companies like Eli Lilly and biotech innovators such as MiNA Therapeutics are expected to drive further advancements. These collaborations bring together expertise in drug development, delivery technologies, regulatory science, and basic RNA biology, creating a fertile environment for breakthrough therapies.

Conclusion
In summary, saRNAs represent a groundbreaking therapeutic modality that defies the traditional paradigm of RNA interference by actively stimulating gene expression. Defined as small double-stranded RNAs that operate via the RNA activation (RNAa) mechanism, saRNAs have emerged as potent tools for reactivating suppressed genes in a variety of disease contexts, particularly in oncology and monogenic disorders. The historical development of saRNAs—from initial observations in the mid-2000s to today’s advanced clinical trials—demonstrates a significant evolution driven by rigorous basic research and technological innovation.

Currently, leading players such as MiNA Therapeutics and Eli Lilly are pioneering saRNA-based therapies, with candidates like MTL-CEBPA making their way into clinical trials for hepatocellular carcinoma. Patent filings for saRNA compositions, including SIRT1-saRNA, indicate a robust commitment to expanding the intellectual property landscape in this domain. Moreover, the versatility of saRNAs makes them attractive for a wide range of applications: therapeutically, by reactivating tumor suppressor genes or compensating for haploinsufficiencies, and as investigative tools for dissecting gene regulation and epigenetic modifications.

Nonetheless, challenges remain. The technical hurdles include ensuring efficient and safe cellular delivery, optimizing sequence specificity, mitigating immunogenic responses, and navigating complex regulatory environments. Future research is expected to overcome these obstacles through improved design algorithms, advanced nanotechnology-based delivery systems, combination therapy strategies, and the integration of personalized medicine approaches. The expansion of clinical indications alongside the development of adaptive, long-lasting therapies is likely to further cement the role of saRNAs as a key player in next-generation therapeutics.

In conclusion, the ongoing development of saRNAs is multifaceted and dynamic, driven by a convergence of scientific discovery, technological innovation, and strategic industry collaborations. The field holds immense promise for transforming our approach to disease treatment by enabling precise, controllable, and safe upregulation of endogenous gene expression. As research efforts intensify and clinical trials expand, saRNAs are poised to contribute significantly to the armamentarium of RNA-based therapeutics, offering hope for patients with diseases that were once considered untreatable.

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