We've updated our Privacy Policy to make it clearer how we use your personal data. We use cookies to provide you with a better experience. You can read our Cookie Policy here.
Dr. Katie Minns received her PhD in Biomedical Sciences in 2016. She developed her passion for molecular biology as a microbiologist in a contract research organization and as a healthcare scientist team leader at Public Health England. Katie moved into science communication in 2021.
RNA therapeutics are redefining how diseases can be treated, targeting the root cause or pathway of a disease and advancing treatments for previously “undruggable” conditions. However, despite their potential, challenges with delivery, stability and immune responses remain key hurdles. Overcoming these obstacles is critical to unlocking the full power of RNA-based medicines.
This listicle explores four major RNA therapeutic modalities, highlighting their breakthroughs, challenges and future potential.
Download this listicle to discover:
Four key RNA-based treatment strategies and their mechanisms
Milestones in RNA therapeutic development and success stories
Current challenges and how researchers are overcoming them
1
Listicle
RNA therapeutics are a versatile drug class made from ribonucleic acid that have rapidly gained momentum
over the last decade, redefining how diseases can be treated. Conventional drug modalities, such as
small molecules, typically target disease symptoms. However, RNA therapeutics go to the root cause or
pathway of the disease. This means they can be designed for targets previously considered “undruggable”.1
These therapeutics were slow to take off until 2016, when Spinraza was approved to treat spinal muscular
atrophy. Fast forward to the start of 2025, and 34 RNA therapies have been approved globally for
clinical use, with many more in the development pipeline.2
RNA therapeutics have an advantage over small molecules and antibodies, in that they can be developed
quickly by using existing design and delivery technologies.3 They are chemically synthesized, which
allows for rapid production4 and they have the benefits of a vast selection of disease targets, robust and
long-lasting efficacy and a high development success rate.5
This modality has now been used to treat conditions such as cancer, cardiometabolic and CNS disorders,
rare genetic diseases and used as vaccines.2 It has also been used for personalized medicine.6
There are challenges for this drug type, largely with delivery and uptake into target cells. In addition,
these therapeutics are unstable and can be degraded by nucleases.7
In this listicle, we explore the key types of RNA therapeutics, milestones in their development history,
success stories and challenges.
mRNA vaccines
mRNA vaccines are well known for their successful application against COVID-19 in 2020. However, they
first appeared in 1995 for a cancer treatment in mice. By 2013, the first clinical trial in humans began
using an mRNA vaccine for infectious disease.8,9 Then in 2020, the mRNA-based COVID-19 vaccines were
rapidly developed, by inserting new mRNA sequences into existing mRNA design and delivery methods
for fast clinical testing. Emergency use approvals came in 2020, with full approvals in 2021.8
After a flurry of activity with a series of vaccines all targeting COVID-19, 2024 saw the approval of mRESVIA
for respiratory syncytial virus (RSV).10 This is the first commercially available mRNA vaccine to target
a different infectious disease, highlighting the potential for more mRNA-based preventive treatments. In
fact, for any pathogen with a known protein target, it should be possible to produce an mRNA vaccine.4
The mechanism for these vaccines involves encoding antigens in mRNA and delivering them into the
host cell cytoplasm via a non-viral delivery route, where they are then expressed and stimulate a specific
immune response.4
RNA Therapeutics: Four Key
Modalities Reshaping Medicine
Katie Minns, PhD
RNA THERAPEUTICS: FOUR KEY MODALITIES RESHAPING MEDICINE 2
Listicle
Despite the rapid success of this vaccine platform, there are challenges. mRNA is unstable and susceptible
to degradation, so the vaccines must be enclosed in a delivery system. Lipid nanoparticles (LNPs) are
used to facilitate uptake into cells through endocytosis, while also allowing escape once inside the cell for
translation in the cytoplasm. 7
Cold storage is another challenge for these vaccines, with some needing to be kept at temperatures
below -60 °C.3 This proved to be a challenge for vaccinating care home residents during the COVID-19
pandemic, forcing early batches of available vaccine to be diverted to alternative high-priority groups.11
While the vaccine must be able to generate a strong immune response, excessive immunogenicity is a
risk for adverse reactions. Modifications to the mRNA structural elements, such as chemically altering
nucleotides and adding poly(A) tails, can minimize these effects and improve stability.12
Antisense oligonucleotides
Antisense oligonucleotides (ASOs) are short, single-stranded oligos, approximately 12–24 bases long.13
They target endogenous RNA, including noncoding RNA and mRNA3 and affect the expression of specific
target RNAs by two mechanisms. Either triggering RNase H1 or ribozymes to cleave target mRNA, or by
regulating gene expression. This can occur in a number of ways, including altering splice patterns to skip
or include particular exons, inhibiting or activating translation, via mRNA decay, or by blocking microRNAs
from binding to target mRNA.13
Fomivirsen was the first RNA therapeutic approved for use in 1998 for the treatment of retinitis caused
by cytomegalovirus.14 Since then, 13 ASOs have been approved for use, mostly targeting genetic diseases,
with 4 approved for treating Duchenne muscular dystrophy (DMD) — an X-linked genetic disorder mostly
affecting young boys.3
Other ASO success stories include the 2024 approval of imetelstat for myelodysplastic syndrome,15
representing a novel approach to treating this type of cancer.16 This class of RNA therapeutic also
represents the first example of a drug being made for a single patient, showing the true potential of
personalized medicine.6
One of the main challenges of ASO therapeutics is enabling them to reach their target sites. Modifications
of the phosphate backbone, ribose ring and bases are often used to overcome this, improving nuclease
resistance and delivery.3 Conjugating the therapeutic oligo to a ligand or using nanoparticles can also
aid delivery.13
Small interfering RNA
Small interfering RNAs (siRNAs) are short double-stranded RNAs, around 21–25 nucleotides long, that
are used in a process called RNA interference (RNAi).17
After uptake into the cell, siRNAs are cut by an endoribonuclease called Dicer, then loaded onto a protein
complex including Argonaute. The strands of the RNA duplex are separated and the “passenger” strand is
ejected. The “guide” strand, that is complementary to the mRNA sequence of interest, remains bound to
Argonaute and together become the RNA-induced silencing complex (RISC). This directs silencing by cleaving
the target sequence or repressing translation, resulting in decreased production of specific proteins.17
The first siRNA therapeutic approved for clinical use was patisiran in 2018 for the treatment of hereditary
transthyretin-mediated amyloidosis – a rare, progressive and often fatal disease. As with other types of
RNA THERAPEUTICS: FOUR KEY MODALITIES RESHAPING MEDICINE 3
Listicle
RNA therapeutic, delivery is a major challenge for siRNA. Patisiran mitigates this by utilizing a LNP delivery
mechanism.18 However, LNPs have their own challenges, including that they trigger an innate immune
response which can cause adverse effects.19 The five siRNAs approved since patisiran all use conjugation
with N-acetylgalactsamine (GalNAc). GalNAc binds the asialoglycoprotein receptor, which is highly expressed
on the surface of hepatocytes, facilitating efficient uptake of siRNAs bound to it.20,21
Other challenges of siRNAs include stability and protection from nucleases. Modifications, such as phosphorothioate
and methylene insertion, can be used to protect the backbone of the oligonucleotide.22
One benefit of the RNAi technique is the abundance of targets, offering flexibility to drug developers and
clinicians. For example, two siRNA drugs, lumasiran and nedosiran, are both designed to treat primary
hyperoxaluria – characterized by the excessive synthesis of oxalate in the liver, leading to kidney disease
– but they differ in their specific targets and mechanisms of action.21,23
RNA aptamers
RNA aptamers are single-stranded oligonucleotides that fold into well-defined structures that specifically
bind to proteins and other targets.1,13 They can be used as antagonists to block interactions between proteins,
or receptors and ligands. Or they can also function as delivery agents when conjugated to drugs.13
Aptamers function in a similar manner to antibodies but have several advantages when compared to
them. Their flexible nature means they can fold into or around a wide range of targets, they have high
thermal stability, and because they are chemically synthesized, they can have rapid, scalable production.24
The first aptamer drug to be approved was Macugen, in 2004, for treating wet age-related macular degeneration
(wet AMD) – a cause of blindness. This was discontinued in 2020,3 although in 2023 a new RNA
aptamer drug, Izervay, was approved to treat geographic atrophy secondary to AMD.25 This remains the
only RNA aptamer commercially available, although there are others in development.26
Unlike the other RNA therapeutic types, delivery is not such a challenge due to aptamer targets typically
being extracellular. However, they are unstable and at risk of clearance. Base modifications and conjugation
with biocompatible polymers, such as polyethylene glycol (PEG) can help to mitigate these problems.3
Future outlook
As the field advances, RNA therapeutics continue to expand into new applications and disease areas,
offering hope to those with rare diseases or previously untreatable conditions. As new solutions are
found to address the challenges with delivery and stability, the potential of these platforms will be unleashed
further.
Related technologies, including CRISPR gene editing and RNA editing, are also expanding the possibilities
for precise genetic interventions. These approaches complement RNA therapeutics, potentially offering even
more targeted and long-lasting treatments. However, the permanent nature of gene editing carries additional
risk, meaning that RNA therapeutics are a safer option while offering many of the same benefits. 3
Sponsored by
RNA THERAPEUTICS: FOUR KEY MODALITIES RESHAPING MEDICINE 4
Listicle
References
1. Damase TR, Sukhovershin R, Boada C, et al. The limitless future of RNA therapeutics. Front Bioeng Biotechnol. 2021;9. doi:
10.3389/fbioe.2021.628137
2. American Society of Gene and Cell Therapy. Gene, cell & RNA therapy landscape report. Q3 2024 quarterly data report.
https://www.asgct.org/global/documents/asgct-citeline-q3-2024-report.aspx. Published 2024. Accessed January 28,
2025.
3. Curreri A, Sankholkar D, Mitragotri S, Zhao Z. RNA therapeutics in the clinic. Bioengineering & Transla Med.
2023;8(1);e10374. doi: 10.1002/btm2.10374
4. Jackson NAC, Kester KE, Casimiro D, et al. The promise of mRNA vaccines: a biotech and industrial perspective. npj Vaccines.
2020;5. doi: 10.1038/s41541-020-0159-8
5. Guo S, Zhang M, Huang Y. Three ‘E’ challenges for siRNA drug development. Trends Mol Med. 2024;30(1);13-24. doi:
10.1016/j.molmed.2023.10.005
6. Kim J, Hu C, Moufawad El Achkar C, et al. Patient-customized oligonucleotide therapy for a rare genetic disease. N Engl J
Med. 2019;381(17):1644-1652. doi: 10.1056/NEJMoa1813279
7. Kutikuppala LVS, Kourampi I, Kanagala RSD, et al. Prospects and challenges in developing mRNA vaccines for infectious
diseases and oncogenic viruses. Med Sci. 2024;22;12(2):28. doi: 10.3390/medsci12020028
8. Kim YK. RNA therapy: rich history, various applications and unlimited future prospects. Exp Mol Med. 2022; 54,455–465.
doi: 10.1038/s12276-022-00757-5
9. Alberer M, Gnad-Vogt U, Hong HS, et al. Safety and immunogenicity of a mRNA rabies vaccine in healthy adults: an
open-label, non-randomised, prospective, first-in-human phase 1 clinical trial. The Lancet. 2017;390(10101),1511-1520.
doi: 10.1016/S0140-6736(17)31665-3
10. MRESVIA. FDA. https://www.fda.gov/vaccines-blood-biologics/vaccines/mresvia. Published December 26, 2024. Accessed
January 01, 2025.
11. BMJ. Vaccinating the UK: how the covid vaccine was approved, and other questions answered. 2020;371. doi: 10.1136/bmj.
m4759
12. Kim S, Sekhon SS, Shin WR, et al. Modifications of mRNA vaccine structural elements for improving mRNA stability and
translation efficiency. Mol Cell Toxicol. 2022; 18,1–8. doi: 10.1007/s13273-021-00171-4
13. Zhu Y, Zhu L, Wang X, et al. RNA-based therapeutics: an overview and prospectus. Cell Death Dis. 2022;13,644. doi:
10.1038/s41419-022-05075-2
14. Fomivirsen approved for CMV retinitis: first antisense drug. AIDS Treat News. 1998; 4;(302):7. PMID: 11365764.
15. FDA approves imetelstat for low- to intermediate-1 risk myelodysplastic syndromes with transfusion-dependent anemia.
FDA. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-imetelstat-low-intermediate-
1-risk-myelodysplastic-syndromes-transfusion-dependent. Published June 06, 2024. Accessed January 28, 2025.
16. Lennox AL, Huang F, Behrs MK, et al. Imetelstat, a novel, first-in-class telomerase inhibitor: Mechanism of action, clinical,
and translational science. Clin Transl Sci. 2024;18;17(11):e70076. doi: 10.1111/cts.70076
17. Wilson RC, Doudna JA. Molecular mechanisms of RNA interference. Annual Reviews. 2013;42:217-239. doi: 10.1146/annurev-
biophys-083012-130404
18. Zhang X, Goel V, Robbie GJ. Pharmacokinetics of patisiran, the first approved RNA interference therapy in patients
with hereditary transthyretin-mediated amyloidosis. Journal of Clinical Pharmacology. 2019;60(5);573-585. doi: 10.1002/
jcph.1553
19. Lee Y, Jeong M, Park J, et al. Immunogenicity of lipid nanoparticles and its impact on the efficacy of mRNA vaccines and
therapeutics. Exp Mol Med 2023;55,2085–2096. doi: 10.1038/s12276-023-01086-x
20. An G. Pharmacokinetics and pharmacodynamics of GalNAc-conjugated siRNAs. J Clin Pharma. 2023; 64(1),45-47. doi:
10.1002/jcph.2337
21. Syed YY. Nedosiran: First approval. Drugs. 2023; 83,1729-1733. doi: 10.1007/s40265-023-01976-4
22. Yamada K, Hariharan VN, Caiazzi J, et al. Enhancing siRNA efficacy in vivo with extended nucleic acid backbones. Nat
Biotechnol. 2024. doi: 10.1038/s41587-024-02336-7
23. Garrelfs SF, Frishberg Y, Hulton SA, et al. Lumasiran, an RNAi Therapeutic for Primary Hyperoxaluria Type 1. N Engl J
Med. 2021;384,1216-1226. doi: 10.1056/NEJMoa2021712
24. Byun J. Recent progress and opportunities for nucleic acid aptamers. Life. 2021;11(3),193. doi: 10.3390/life11030193
25. Drug trials snapshots: IZERVAY. FDA. https://www.fda.gov/drugs/drug-approvals-and-databases/drug-trials-snapshots-
izervay. Published October 11, 2024. Accessed January 01, 2025.
26. Santarpia G, Carnes E. Therapeutic applications of aptamers. Int J Mol Sci. 2024; 25, 6742. doi: 10.3390/ijms25126742
Sponsored by
Download the List for FREE Now!
Information you provide will be shared with the sponsors for this content. Technology Networks or its sponsors may contact you to offer you content or products based on your interest in this topic. You may opt-out at any time.