A new era in molecular medicine
For decades, the central dogma of biology—DNA makes RNA makes protein—served as both a principle and a limitation. While DNA therapies aimed to rewrite the genetic code and protein biologics targeted cellular pathways, RNA remained an elusive middle ground: unstable, transient, and difficult to control.
That perception changed almost overnight. The global rollout of mRNA vaccines during the COVID-19 pandemic did more than stop a virus—it demonstrated, at scale, that RNA could be a reliable and rapidly adaptable therapeutic platform. Since then, RNA has moved from laboratory curiosity to clinical mainstay.
Today, RNA therapeutics encompass a spectrum of molecules—mRNA, siRNA, and antisense oligonucleotides (ASOs)—each leveraging the language of biology to modulate gene expression directly. For molecular biologists and drug developers, this is not just a new class of drugs; it’s a new way of thinking about disease intervention.
What are RNA therapeutics?

From antisense pioneers to mRNA innovators, RNA therapeutics have turned an informational molecule into a therapeutic engine.
ImageFX (2025)
RNA therapeutics are medicines that use ribonucleic acid molecules to treat or prevent disease by influencing the flow of genetic information. Unlike traditional drugs that target proteins downstream, RNA therapies work upstream—at the level of genetic messaging.
By designing RNA molecules that correspond to specific genetic sequences, scientists can either:
- Deliver instructions to make a protein (mRNA therapies).
- Silence a problematic gene (siRNA).
- Correct or modify a transcript (ASOs).
In other words, RNA drugs don’t just affect biology—they speak it fluently.
mRNA therapeutics: Turning cells into temporary factories
When messenger RNA (mRNA) entered the global vocabulary in 2020, it wasn’t through a scientific journal but through the public health playbook. The success of the Moderna and Pfizer-BioNTech vaccines validated decades of research in synthetic mRNA design, lipid nanoparticle (LNP) delivery, and immunology (Nat Nanotechnol, 2021; 16(9):1024–1038).
But mRNA’s potential reaches far beyond vaccines. These molecules can be designed to encode any therapeutic protein, effectively transforming the body’s own cells into miniature, temporary factories.
How mRNA works
Synthetic mRNA mimics the structure of naturally occurring transcripts—complete with a cap, untranslated regions, and a poly(A) tail. Once encapsulated in LNPs and delivered into the cytoplasm, the mRNA is read by ribosomes to produce the encoded protein. Because the RNA never enters the nucleus, there’s no risk of altering the genome.
Why it matters
The flexibility of mRNA is unmatched. A new protein drug can, in principle, be designed on a laptop, synthesized chemically, and tested within weeks. Beyond infectious disease, mRNA therapeutics are being explored for:
- Protein replacement in enzyme deficiencies or metabolic disorders.
- Cancer immunotherapy, encoding tumor antigens to stimulate immune recognition.
- Regenerative medicine, prompting cells to produce growth or repair factors.
The key challenge is delivery. While LNPs have revolutionized intramuscular and hepatic delivery, reaching other tissues—such as the heart, lung, or brain—remains difficult. Scientists are now experimenting with ionizable lipids, polymeric nanoparticles, and self-amplifying RNA to expand reach and reduce dosage.
As many scientists now emphasize, if mRNA was the breakthrough of 2020, delivery is the obsession of 2025—a sentiment echoed in recent reviews highlighting the central role of delivery systems in next-generation RNA therapeutics (Nat Rev Drug Discov, 2024; 23:671–673).
siRNA: The quiet power of gene silencing
If mRNA is the messenger, small interfering RNA (siRNA) is the censor. Rather than producing proteins, siRNA therapeutics prevent their production altogether by exploiting the cell’s natural RNA interference (RNAi) machinery.
Discovered in 1998 by Andrew Fire and Craig Mello, RNAi introduced a radical concept: short double-stranded RNA molecules could silence specific genes by destroying their messenger RNAs before translation (Nature, 1998; 391(6669):806–811).
How siRNA works
When siRNA enters the cytoplasm, one strand—the guide—is incorporated into the RNA-induced silencing complex (RISC). RISC then uses the siRNA sequence to recognize complementary mRNA molecules. Once bound, the Argonaute enzyme within RISC cleaves the target, effectively erasing that message from the cell’s playbook.
From discovery to therapy
It took nearly 20 years to turn this elegant biology into medicine. Patisiran (Onpattro®), approved in 2018, became the first siRNA drug on the market, treating hereditary transthyretin amyloidosis (hATTR) by knocking down mutant TTR transcripts in the liver (N Engl J Med, 2018; 379(1):11–21).
Since then, companies like Alnylam have brought multiple RNAi drugs to market, including Givosiran (Givlaari®) for acute hepatic porphyria and Inclisiran (Leqvio®) for LDL cholesterol reduction. All use sophisticated GalNAc conjugates that guide siRNA specifically to hepatocytes via the asialoglycoprotein receptor.
Strengths and limits
- High precision: siRNA recognizes a single target sequence, minimizing unintended effects.
- Long duration: Silencing can last for weeks or months after a single dose.
- Proven platform: RNAi has validated manufacturing and delivery technologies.
Still, most siRNA therapies remain liver-centric. Delivering RNAi drugs to other tissues—such as neurons or muscle—will require new carrier chemistries and targeting ligands.
Antisense oligonucleotides: Rewriting RNA messages
Long before mRNA vaccines or siRNA silencers, there were antisense oligonucleotides (ASOs)—short synthetic strands designed to bind RNA and alter its function. First conceptualized in the late 1970s, ASOs were the earliest attempt to use nucleic acids as drugs.
Unlike siRNA, which destroys mRNA via RISC, ASOs can block, degrade, or repair RNA through several mechanisms.
Mechanisms of action
Once bound to their target, ASOs can:
- Recruit RNase H1 to degrade the mRNA strand.
- Block translation by steric hindrance.
- Modulate splicing to skip or include specific exons—essentially editing RNA in situ.
Clinical breakthroughs
The ASO field came of age with Nusinersen (Spinraza®), the first FDA-approved therapy for spinal muscular atrophy (SMA). By altering splicing of the SMN2 gene, it restores production of functional SMN protein (Nat Neurosci, 2017; 20(4):497–499).
Other successes followed:
- Eteplirsen (Exondys 51®) for Duchenne muscular dystrophy, promoting exon skipping in the dystrophin gene.
- Inotersen (Tegsedi®), targeting TTR mRNA for hATTR amyloidosis.
Each of these breakthroughs reinforced the flexibility of ASOs to target previously untreatable genetic diseases.
The chemistry behind success
Modern ASOs are highly engineered molecules. Backbone modifications (such as phosphorothioate linkages) increase nuclease resistance, while sugar modifications (like 2′-O-methyl or locked nucleic acids) enhance binding affinity and stability.
Despite these advances, delivery and durability remain central challenges. Intrathecal administration, for instance, allows ASOs to reach the central nervous system but requires invasive dosing. Researchers are now testing systemic formulations and receptor-mediated uptake systems to broaden accessibility.
The promise and problems of RNA drugs
Taken together, RNA-based therapeutics have reshaped the drug discovery landscape. Their promise lies in precision and programmability: once a target sequence is known, a complementary RNA drug can be designed within weeks.
But biology remains a formidable adversary. RNA is inherently fragile, easily degraded, and sometimes immunostimulatory. Moreover, translating success in the liver to other organs—especially the brain, lung, and heart—requires new lipid chemistries and molecular carriers.
For now, the “delivery bottleneck” is what keeps many RNA therapeutics grounded in preclinical or early clinical stages. Yet, optimism runs high. As many experts have observed, translating RNA medicines into different tissues remains one of the greatest technical hurdles. As Science Translational Medicine summarized, the next wave of innovation will rely on teaching cells to “read” RNA safely and efficiently across organ systems (Sci Transl Med, 2024; 16:eadh3275).
What comes next
The next generation of RNA drugs is already taking shape. Self-amplifying RNA (saRNA) platforms promise lower doses and longer expression, while circular RNA (circRNA) molecules show increased stability and translation efficiency. Meanwhile, AI-guided design and high-throughput screening are helping predict secondary structures, off-target interactions, and delivery kinetics.
In the not-so-distant future, we may no longer distinguish between small molecules, biologics, and RNA drugs—they’ll coexist as tools within a unified molecular medicine toolkit.
Conclusion: Rewriting the rules of therapy
From antisense pioneers to mRNA innovators, RNA therapeutics have turned an informational molecule into a therapeutic engine. What began as a “missing middle” between genes and proteins has evolved into a versatile drug platform capable of silencing, correcting, or expressing virtually any gene.
For molecular biologists, this represents more than a new class of medicine—it’s a rewriting of biology itself, where the transcript becomes the treatment and the code becomes the cure.
References
This article was created with the assistance of Generative AI and has undergone editorial review before publishing.















