How many FDA approved miRNA are there?

17 March 2025
Introduction to miRNA

Definition and Biological Role
MicroRNAs (miRNAs) are a class of small, non-coding RNA molecules generally 18–25 nucleotides long that play a critical role in the post‐transcriptional regulation of gene expression. They modulate protein expression through mechanisms such as translational repression and mRNA degradation, thereby influencing a wide array of biological processes including differentiation, proliferation, apoptosis, and metabolism. Due to their capacity to target hundreds of messenger RNAs (mRNAs) by binding predominantly to the 3′ untranslated regions (UTRs), miRNAs are considered master regulators of cellular pathways. The intricate role of miRNAs in maintaining cellular homeostasis means that any dysregulation can contribute to disease pathogenesis, including cancer, cardiovascular disorders, and neurodegenerative diseases. From the early discovery in organisms such as Caenorhabditis elegans to extensive investigations in human tissues, the evolutionary conservation of miRNAs underscores their biological importance.

miRNA in Therapeutics
In recent years, the discovery of miRNAs’ regulatory abilities has sparked significant interest in their potential therapeutic application. Researchers have explored the use of miRNA mimics to restore the function of downregulated tumor-suppressive miRNAs and anti-miRNA oligonucleotides (antagomirs) to inhibit oncogenic miRNAs, thereby opening new avenues for the treatment of complex diseases. The ability of miRNAs to simultaneously regulate multiple target genes and influence entire signaling networks has made them attractive candidates for therapeutic intervention, especially in diseases with multifactorial etiologies. Despite intensive preclinical research and the initiation of several early-phase clinical trials, the translation of miRNA-based therapeutics from bench to bedside has encountered several challenges, including issues of delivery, stability, specificity, and toxicity. Moreover, the broad regulatory impact of miRNAs necessitates rigorous safety and off-target effect profiling before they can be considered for clinical use.

FDA Approval Process for miRNA

Regulatory Pathways
The U.S. Food and Drug Administration (FDA) follows stringent processes and guidelines for the approval of therapeutics, including RNA-based drugs. Regulatory pathways for nucleic acid-based therapies are complex due to the unique nature of these molecules. RNA-based therapies that have successfully navigated the FDA approval process include small interfering RNAs (siRNAs), antisense oligonucleotides (ASOs), and more recently, mRNA-based therapeutics such as the COVID-19 vaccines. The regulatory framework entails a thorough review of manufacturing processes, pharmacokinetics, pharmacodynamics, preclinical safety, and clinical efficacy.
For miRNA therapeutics, the pathway involves a similar rigor, but the FDA’s historical experience with miRNA molecules in a clinical setting is minimal. The growing body of RNA-based therapies has provided valuable lessons on the required standards; however, the inherent complexity of miRNA functions—namely their ability to regulate multiple genes simultaneously—adds an extra layer of challenge to the regulatory approval process. This complexity can influence target specificity and contributes to off-target effects, a primary safety concern from the FDA’s perspective.

Criteria for Approval
FDA approval of any novel therapeutic agent, including those based on RNA, requires demonstration of significant safety and efficacy profiles through multi-phase clinical trials. Key criteria include:

Preclinical Safety and Toxicology:
Detailed assessments, often in both in vitro and in vivo models, must confirm that the molecule does not induce adverse immunological or toxic effects. The preclinical evaluation must address issues such as degradation by serum nucleases, off-target binding, biodistribution, and the stability of the therapeutic candidate in biological systems.

Pharmacokinetics and Pharmacodynamics (PK/PD):
Characterization of the absorption, distribution, metabolism, and excretion (ADME) of the molecule is critical. For miRNA candidates, it is essential to establish that the molecule reaches the target tissue in an active form, maintains sufficient bioavailability, and achieves a therapeutic concentration without accumulating to toxic levels.

Clinical Efficacy and Safety:
Rigorous evaluation in clinical phases, particularly in phase III trials, is mandatory to confirm both the therapeutic benefit and the acceptable safety margin. The challenge for miRNA therapeutics lies in the need to understand the multifaceted mechanism of action and the potential for widespread gene regulation, which necessitates exceptionally detailed clinical endpoints and safety monitoring.

Manufacturing and Consistency:
The process of synthesizing miRNA mimics or inhibitors must be reproducible, scalable, and compliant with current Good Manufacturing Practices (cGMP). The complexity in producing these molecules often requires innovative chemical modifications to enhance stability, reduce immunogenicity, and optimize cell-specific delivery.

While these criteria are standard for all advanced therapeutic modalities, miRNA-based therapeutics have additional challenges stemming from their intrinsic biological properties, making the approval process even more scrutinized by regulatory authorities like the FDA.

Current FDA Approved miRNA Therapies

List and Description of Approved miRNA
Despite the extraordinary research efforts and promising preclinical data surrounding miRNA-based therapeutics, there remains a significant caveat when it comes to FDA approval. According to the accumulated evidence and specific commentary in the literature, there are currently no miRNA-based therapeutics that have been approved by the FDA.
For instance, a frequently cited observation in the field is that despite the enthusiasm around miRNAs as targets or drugs for human diseases, the US Food and Drug Administration has never approved nor intended to approve any miRNA-based therapeutic for clinical intervention. Unlike siRNA-based drugs, which have successfully transitioned into the clinic with approvals such as patisiran for polyneuropathy due to hereditary transthyretin-mediated amyloidosis, miRNA mimics or inhibitors remain in early-stage clinical evaluations or preclinical development.
This fundamental point about the regulatory landscape highlights that although many miRNA candidates are in various stages of clinical trials, the number of miRNA therapies that have reached the market is zero.

Clinical Applications and Indications
While miRNA therapeutics hold enormous potential for a broad range of clinical applications—from cancer treatment and cardiovascular diseases to neurodegenerative conditions—the lack of any FDA-approved miRNAs implies that there are no current clinical applications using these molecules as standalone approved drugs.
For example, clinical trials have explored miRNA candidates such as miravirsen (an inhibitor of miR-122) for hepatitis C virus infection and others in the realm of oncology; however, these remain investigational. Even though several RNA-based therapeutics are in the pipeline, the success of siRNA and mRNA therapies has overshadowed miRNA-based approaches in clinical approval. The gap between promising preclinical data and successful translation into approved therapies underscores the complexity of achieving a balance between efficacy and the safety profile required by regulatory bodies.

Challenges and Future Prospects

Challenges in miRNA Therapeutic Development
The journey from discovery to clinical translation of miRNA therapeutics is fraught with several challenges that have so far prevented any FDA approval. These challenges can be categorized into multiple domains:

Target Specificity and Off-Target Effects:
miRNAs typically bind to their target mRNAs via partially complementary seed regions, which inherently allows them to modulate multiple genes simultaneously. This promiscuity can lead to unintended downregulation of non-target genes, causing off-target effects that may manifest as toxicity or other adverse events. The potential for “too many targets” is one of the fundamental hurdles that has necessitated cautious regulatory evaluation.

Delivery Challenges:
Efficiently delivering miRNA therapeutics to the desired tissue or organ, while avoiding degradation and non-specific uptake, remains a significant technological and biological challenge. miRNAs need to navigate barriers such as the blood-brain barrier (BBB) when targeting neurodegenerative diseases, as well as peripheral distribution issues for other systemic diseases. Innovative delivery systems like lipid nanoparticles, conjugation with targeting ligands, and vector-based systems are under investigation; however, none have yet achieved the necessary precision and consistency for FDA approval.

Stability and Pharmacokinetics:
The intrinsic instability of RNA molecules in the presence of nucleases in the bloodstream requires chemical modifications to improve half-life and bioavailability. Modifications that enhance stability while preserving biological activity are still being optimized, and any alteration must be thoroughly vetted for potential immunogenicity and adverse pharmacodynamic alterations.

Immunogenicity and Toxicity:
The administration of RNA-based therapeutics has the potential to trigger innate immune responses, which can limit their therapeutic window. While some chemical modifications are designed to reduce immunogenicity, ensuring that these modifications do not interfere with the therapeutic function is an ongoing challenge. Moreover, because miRNAs regulate a broad spectrum of targets, the risk of unwanted toxicities due to cumulative off-target effects is non-negligible.

Manufacturing and Scalability:
The production of high-purity, chemically modified miRNA molecules in a scalable and cost-effective manner remains a technical hurdle. The reproducibility and quality control required under cGMP conditions impose additional layers of complexity that have yet to be fully overcome for miRNA therapeutics.

Future Directions in miRNA Research
Looking to the future, several strategies are under exploration to address the challenges in miRNA therapeutic development, which may eventually lead to FDA-approved products:

Enhanced Specificity Through Advanced Molecular Design:
Advances in computational biology and the development of more sophisticated target prediction algorithms promise to refine the understanding of miRNA-target interactions. This, coupled with the design of modified miRNA mimics and inhibitors that minimize off-target effects, could significantly improve the specificity of these agents. Furthermore, emerging strategies such as the use of “cluster analysis” of miRNA families might provide more targeted and context-specific therapeutic effects by harnessing the cumulative, yet controlled, regulation of gene networks.

Innovative Delivery Systems:
Research is increasingly focused on developing novel delivery platforms that can overcome the biological barriers faced by miRNA therapeutics. Nanoparticle-based delivery, extracellular vesicle encapsulation, and conjugation with targeting moieties (such as antibodies or peptides) represent promising avenues to improve tissue-specific delivery, reduce systemic exposure, and mitigate associated side effects. These delivery systems are essential not only for enhancing the clinical efficacy of miRNA drugs but also for ensuring that they meet regulatory safety benchmarks.

Rigorous Preclinical Models and Biomarker Development:
The development of robust preclinical models that better mimic human physiology is essential to validate the safety and efficacy of miRNA therapeutics. Improved biomarker panels for monitoring miRNA activity and off-target effects in both preclinical and clinical settings could facilitate a more accurate assessment of therapeutic impact and risk minimization, addressing one of the key concerns raised by nonclinical studies.

Collaborative and Multidisciplinary Approaches:
The challenges inherent in miRNA therapeutic development require interdisciplinary collaboration that spans molecular biology, clinical pharmacology, material science, and regulatory science. Collaborative efforts between academic groups, biopharmaceutical companies, and regulatory agencies can help accelerate the translation of promising preclinical findings into clinical therapies. Such collaborations are expected to pave the way for the next generation of RNA therapeutics, including those based on miRNAs.

Exploratory Clinical Trials and Adaptive Regulatory Pathways:
It is imperative to continue early-phase clinical trials with adaptive designs that allow for rapid modifications based on emerging data. These trial designs, potentially aided by breakthrough therapy designations and fast-track programs, could help overcome some of the current hurdles by providing a dynamic and responsive regulatory framework that adapts to the complexities of miRNA-based therapy.

Detailed Conclusion
In summary, extensive research on miRNA biology and their role as regulators of gene expression has illuminated their incredible potential as therapeutic agents. However, despite their promise, the clinical translation of miRNA-based therapeutics has been hindered by formidable challenges such as target specificity, delivery obstacles, stability, immunogenicity, and manufacturing hurdles. As detailed in the regulatory pathways and criteria established by the FDA, each aspect—from preclinical safety and PK/PD characterization to robust phase III clinical trials—must be rigorously addressed before a therapeutic can be approved.

A thorough evaluation of the literature, including detailed analyses of the challenges inherent in miRNA functional complexity and delivery systems, reveals a clear and consistent conclusion: there are currently no FDA-approved miRNA therapies. While other RNA-based therapies like siRNAs and mRNA vaccines have successfully navigated the approval process and achieved market authorization, miRNA-based therapeutics remain in the early phases of clinical development or even preclinical evaluation. This stark reality is rooted in the inherent biological properties of miRNAs—their ability to target multiple genes and influence entire pathways—which, while offering a promising avenue for treating multifactorial diseases, also poses significant hurdles in achieving the stringent safety and efficacy standards required by regulatory agencies.

Nonetheless, the future for miRNA-based therapeutics is not without hope. Advances in molecular design, innovative delivery systems, rigorous validation in preclinical models, and adaptive regulatory pathways hold the promise of eventually bridging the current gap. If these challenges can be systematically overcome, the potential for miRNAs to transform the therapeutic landscape is tremendous. Researchers and biopharmaceutical companies continue to invest and innovate in this space, fostering a multidisciplinary approach that may ultimately lead to the first FDA-approved miRNA drug.

In conclusion, based on the current state of research and the regulatory history documented in multiple peer-reviewed sources from synapse, the answer to the question "How many FDA approved miRNA are there?" is unequivocally zero. While the promise of miRNA-based therapeutics is compelling, and significant strides are being made in overcoming the developmental challenges, the clinical application of miRNA drugs has not yet reached the pivotal milestone of FDA approval. This conclusion emphasizes the need for continued research, innovation, and regulatory dialogue to fully harness the therapeutic potential of miRNAs in the future.

For an experience with the large-scale biopharmaceutical model Hiro-LS, please click here for a quick and free trial of its features

图形用户界面, 图示

描述已自动生成