White Paper on RNA Therapeutics
This is the first article of a series on the state of emerging therapeutic modalities and related
technologies. In this installment, we review the RNA therapeutics scientific and clinical development
landscape with emphasis on oncology, immunology, neurology, & diabetes applications. I will start each
of these installments with operational definitions & essential background of the therapeutic modality
used to frame the overview. Then I will provide a brief history on the field from its first appearance
through the present day. We will then explore the present state of the field including current
opportunities & applications in the disease areas of interest. Finally, each installment will end with a
look at future prospects & potential developments in the field as they relate to the clinical development
landscape.
Background & Definitions:
Ribonucleic acid (RNA) is an integral part of the central dogma of biology where genetic information is
encoded in the deoxyribonucleic acid (DNA) sequence of a cell which is then transcribed into RNA, some
of which are then translated into proteins which are the functional (or not) molecules for cellular
processes. Traditional medicine has focused on targeting proteins which are uniquely associated with a
diseased cell or which may be malfunctioning to cause a disease state. RNA therapeutics broadly can
refer to the treatment or prevention of disease utilizing RNA-based agents or by targeting RNAs whose
expression is related to the disease state. For this article, we will use this definition when referring to
RNA therapeutics as a field.
History:
Nucleic acids (the base units of DNA & RNA) were discovered by Friedrich Miescher in the last half of the
19th century. It wasn’t until the 1930s that biologists began to recognize a relationship of nucleic acids to
cellular growth and development. In 1947, Andre Boivin and Roger Vendrely published the hypothesis
that DNA governed the assembly of RNA which in turn controlled the production of cytoplasmic
enzymes. In the 1950’s, work by Rosalind Franklin, Francis Crick, and James Watson revealed the double
helical structure of DNA followed by the postulation of Crick for the existence of messenger RNA (mRNA)
that serves as a template for protein production. In 1961, Watson and colleagues published in Nature
that they had isolated these mRNA intermediates. Over the next three decades, scientists continued to
discover new forms and functions of RNAs until a breakthrough experiment by Jon Wolff demonstrated
that in vivo transcribed mRNA or plasmid DNA directly injected into the muscle tissue of mice were able
to enter the cells and were translated into proteins in the cytoplasm (1). This experiment and the
subsequent discovery of the RNA interference (RNAi) process 8 years later by Andre Fire & Craig Mello
sparked a surge in RNA research giving rise to the field of RNA therapeutics.
Mechanisms of RNA Therapeutics:
The direct introduction of synthesized mRNAs can be used for protein or peptide replacement or
introduction. Once mRNAs reach the cytoplasm of target cells, they take advantage of the cell’s
translational machinery (ribosomes & tRNA) to produce a peptide or protein of interest with therapeutic
benefit. These mRNAs can be chemically modified are designed to increase their stability in the cell, but
ultimately, liner mRNA transcripts are degraded fairly rapidly in most cells by nucleases making this type
of therapy more transient than other methods. One potential solution for improved stability is being
leveraged by Orna Therapeutics in the form of engineered circular RNAs (oRNA) that are inherently less
suitable to nuclease degradation than linear RNAs. Merck (MSD) recently announced a collaboration
with Orna based on the promise of the improved in vivo stability of the oRNA platform verses linear
mRNA.
RNAi refers to the delivery of RNAs (or RNA/DNA hybrids) such as antisense oligonucleotides (ASOs or
AONs) or short interfering RNAs (siRNAs) designed to bind nucleic acid targets to degrade, sequester, or
block active domains of the target. ASOs can also be chemically modified with added labels or tags to
identify subcellular localizations of targets or to isolate targeted transcripts. This has applications in drug
design, basic research, diagnostics, and for measuring a patient’s response to treatment or disease
progression when paired with medical imaging modalities such as positron emission tomography (PET or
PET-CT) (2). An additional application involves chemically modified ASO-mediated control of RNA
splicing which can be directed to block the incorporation of mutated exons (exon skipping) to rescue
expression of near-full length, functional proteins such as in Duchenne Muscular Dystrophy (DMD) (3).
ASO-mediated steric hindrance can also affect gene regulation by interfering with epigenetic
modification & chromatin state mediated by long non-coding RNAs (lncRNAs) (4). In March of this year,
NextRNA Therapeutics launched with $9.3M from seed financing and $46.8M from Series A on a
platform aimed to disrupt the binding activity of non-coding RNAs, particularly lncRNAs, with small
molecules.
Development of RNA vaccines are also increasing for applications beyond infectious disease including
cancer vaccines. The premise of mRNA vaccines is that delivery of a synthetic mRNA encoding cancerspecific epitopes found on the surface of malignant cells are expressed in antigen-presenting cells (APCs)
which facilitate activation of the innate & adaptive immune system. In this way, the patients own
immune system is trained to recognize cancer cells and eliminate them. In contrast to the immune
stimulatory effect for most vaccines, a recent publication in Science describes the development of a 1
methylpseudouridine-modified mRNA vaccine that may reduce the damaging autoimmune response for
disorders such as multiple sclerosis (MS) (5). They find that maintenance of this therapeutic effect is
dependent on expression of immune checkpoint signaling including PD-1 & CTLA-4 signaling. Treatment
with this modified mRNA vaccine reduced proinflammatory cytokine release in animal models of MS
suggesting a possible application for CRS management experienced with certain cell therapies.
Gene editing traditionally is mediated by DNA plasmids encoding the gene-editing enzyme machinery
and guide RNA (gRNA) sequences to accomplish targeted edits within cells. We will discuss gene editing
in greater detail as the subject for the next installment. These plasmids are often delivered using
transfection reagents or viral vectors which have variable efficacy based on cell type & significant safety
risks. A drawback of DNA plasmid transfection for in vivo editing applications is that the persistent
expression of gene editing components can increase the risk of off-target effects and of altering cellular
phenotypes in unpredictable ways. Researchers at Tufts University recently described lipid nanoparticle
(LNP) delivery of CRISPR-Cas9 ribonucleoprotein (RNP) co-packaged with a guide RNA (gRNA) or LNP
delivery of CRISPR-Cas9 mRNA resulting in efficient, transient gene editing with non-integrating Cas9
expression (6). This represents a significant opportunity for the future development of in vivo gene
editing therapeutic approaches leveraging the systems and lessons from RNA therapeutics to combine
these therapeutic modalities.
RNA aptamers are sometimes referred to as chemical antibodies. These molecules are short nucleic acid
sequences (DNA or RNA) that bind to specific targets (proteins, DNA, or RNA) similar to traditional
protein antibodies, but they are much more versatile than traditional antibodies and have distinct
properties that differentiate them from other RNA-based therapies. Aptamers, like ASOs, can be
chemically modified to reduce immunogenicity, resist degradation, enhance potency, or attach labels.
There are also peptide-based aptamers with broad therapeutic applications. I will discuss aptamers as a
therapeutic modality in the 3rd installment of this series.
Challenges – Delivery/stability, structure/chemical synthesis, & off-target effects/immunogenicity:
As mentioned previously, cells in the body including malignant cells have a host of mechanisms for
degrading and recycling RNAs in the cytoplasm and nucleus. Additionally, RNA is inherently unstable,
especially in linear form. Your skin and the surfaces around you have nuclease enzymes that rapidly
degrade RNA meaning the study, production, storage, and administration of RNA-based therapeutics
require special processes & conditions to protect the integrity of these molecules. Many ASOs employ a
“gapmer” design strategy with 3 or 5 modified RNA bases on each end of the oligo separated by 8 to 12
oligodeoxynucleotide gap. This design along with the base modifications enhance the binding of the ASO
to the target. Adding phosphorothioate (PS) modified linkages to the backbone of ASOs further
improves their stability and activity by protecting the oligo from nuclease degradation while promoting
RNase H1-mediated degradation of the ASO target. PS modifications also introduce chiral centers into
the oligonucleotide, which can be controlled using stereoselective synthesis methods which can further
control the binding affinity, stability, and other properties of ASOs (7). Furthermore, PS gapmer ASOs
acting through RNase H1 degradation are not cleaved in the process meaning the activity of these ASOs
can persist for long periods of time and high levels of activity can be achieved with fewer molecules in
the cell. Still other modifications can produce an ASO which binds but does not degrade its target
serving to disrupt activity by steric hindrance or even can be linked to a tag or marker to label the target
molecule.
RNA therapies can sometimes be delivered “naked” in suspension by direct injection or administration
to the targeted site. These RNAs can then enter the cell via a process called gymnotic uptake (gymnosis)
by which transmembrane transporters bring the extracellular RNAs into the cell (8). RNAs can be
chemically modified to increase the efficiency of uptake and can be designed to enter the cell nucleus or
to remain in the cytoplasm depending on the subcellular location of the intended target. RNA therapies
have also commonly utilized lipid nanoparticle (LNP) packaging and delivery for systemic administration.
There are advantages and disadvantages to this approach, the largest drawback being accumulation in
the liver and spleen (unless those are the target organs). Kumthekar et.al. have described the use of
spherical nucleic acids (SNAs) that consist of a gold nanoparticle core with covalently linked
oligonucleotides for RNAi applications in the CNS (9). There are numerous alternative nanoparticle
delivery systems in development including self-assembled RNA nanoparticles like the phi29 pRNA
hexameric ring formation which can be modified for in vivo delivery applications (10,11). Delivery using
nanoparticles is a complex bioengineering problem. Patisiran (Alnylam Pharmaceuticals siRNA for
polyneuropathy FDA approved in 2018), Tozinameran (Pfizer-BioNTech COVID-19 vaccine approved in
2021), and Elasomeran (Moderna’s conditionally approved COVID-19 vaccine) all deliver RNA payloads
via LNPs. Two large differentiation points across these products demonstrate the complexity of LNP-RNA
design. While the two COVID vaccines can be administered intramuscularly in seconds, Patisiran is
administered by IV with over 70min infusion time (12). Patisiran must be stored between 2 & 8 degrees
C and will lose efficacy if frozen. The Pfizer/BioNTech vaccine must be stored between -60 & -90 degrees
C while the Moderna vaccine can be stored from -15 to -50 degrees C. These differences are in part
because of the stability of siRNA vs mRNA, but also due to the composition & design of the LNP (12).
Off-target and undesired host immune activation are additional challenges for RNA therapy design. Base
constrained ASO modifications such as locked nucleic acids (LNA), 2’-O-methoxyethyl (2’-MOE)
oligonucleotides, and 2’,4’-constrained ethyl (cEt) modifications can further enhance binding affinity,
target specificity, and resistance to nucleases. However, LNA ASOs have been found to have neurotoxic
and hepatotoxic effects. Base modifications within CpG motifs of oligonucleotides can reduce potentially
toxic immunogenicity. AUM Biotech’s proprietary FANA technology has the advantages of self-delivery,
reduced off-target effects, and improved stability over other RNA modalities. AUM Biosciences is the
clinical stage development arm leveraging modified ASOs for oncology applications.
Applications of RNA Therapeutics:
RNA therapies in the U.S. marketplace are relatively young. The first RNA-based FDA approval was the
ASO Fomivirsen for cytomegalovirus retinitis (CMV) delivered via intraocular injection in 1998. It was
developed by Ionis Pharmaceuticals (formerly Isis Pharmaceuticals) and licensed to Novartis to address
the high incidence of CMV in HIV/AIDs patients. Ionis also secured FDA approval for the first ASO,
Eteplirsen, to affect RNA-splicing by steric binding to promote exon skipping and restore mostly
functioning dystrophin protein to DMD patients. Alnylam Pharmaceuticals secured the first FDA
approval for Patisiran, the first siRNA used to treat polyneuropathy caused by a form of hereditary
amyloidosis (13). BNT162b2, now Comirnaty, was the first mRNA vaccine granted Emergency Use
Authorization (EUA) and then the first FDA-approved mRNA vaccine having been developed by BioNTech
and Pfizer. There are several disease-specific applications in development described below:
NeurologyThere are 10 FDA-approved RNA-based therapeutics for the treatment of neurological diseases including
8 ASOs from 1998 (fomivirsen) to 2021(Casimersen), 1 siRNA (patisiran 2018), and 1 RNA aptamer
(pegaptanib 2004) (14). Four of the approved ASOs are for DMD. These are examples of splice-switching
ASOs (ssASOs). A host of neurological and neurodegenerative diseases are related to alternative splicing
or mutations in exon-encoding regions that produce malignant or non-functional proteins. Preclinical
studies suggest ssASOs may have therapeutic applications in dementia-causing diseases and
“tauopathies” by reducing or preventing plaque formations (15,16). A challenge for all drugs is delivery
to the brain and crossing the blood-brain barrier (BBB). Intrathecal (IT) administration is a common CNS
delivery route introducing drugs into the subarachnoid space and cerebrospinal fluid (CSF) for efficient
distribution in the CNS compartment. Intraventricular injections are another route entering CSF
circulation and intravitreal or intraocular injections are also common routes of administration. A class of
arginine-rich cell-penetrating peptides conjugated to ASOs have shone to cross the BBB in animal
models (17). Exosomes and nanoparticles have also been decorated with brain-targeting peptides for
enhanced delivery to the brain.
OncologyImmunotherapeutic approaches in oncology are a rapidly growing market segment across the globe.
RNA therapies are crossing into this space through RNA cancer vaccines which are targeted to APCs to
train the host immune cells to recognize and eliminate cancer cells (18). A PD-L1 targeted siRNA
conjugated to a magnetic nanocarrier has demonstrated robust pancreatic cancer tumor reduction
preclinically that thanks to the magnetic conjugate, can also be used to monitor therapy response by
MRI (19). Some RNA therapies are delivered with engineered viral vectors because of their payload
capacities and long history of use as vectors in biotechnology. Inorganic gold or iron-oxide nanoparticle
delivery system have theragnostic potential both in targeted delivery of the drug, but also enabling noninvasive visualization to identify distal metastases or gauge tumor mass and invasion into normal
parenchyma (20). Photothermal and photodynamic therapies are being combined with RNA therapies
for additive or synergistic anti-tumor effects including highly-specific RNAi targeting where focused
electromagnetic or x-ray wavelengths trigger release of the RNAi payload only in the specific targeted
site (21). RNA therapies could represent a revolution in the treatment of cancer and other diseases by
opening new therapeutic targets to pathways or malignant programs once thought “un-druggable.” It is
possible the species of RNAs such as lncRNAs or circRNAs can serve as master regulators of upstream
disease-programs by affecting cellular programs at the epigenetic level (22). Enfuego Therapeutics
founded by Chad Pecot at UNC-CH received STTR Phase 1 funding in 2020 to design ASOs to elusive
oncology targets previously thought “undruggable.”
ImmunologyClearly, mRNA vaccines against COVID-19 have left their mark on the RNA and immunology landscapes
for protection against infectious diseases. As mentioned in the previous section, RNA therapies including
RNA cancer vaccines are bridging the two modalities and demonstrating the versatility of RNAimmunotherapy combinations. Traditional vaccines are based on live attenuated infectious peptides or
whole killed pathogens to stimulate immune response and memory. RNA-based vaccines can be used to
express one or more specific epitopes of an infectious organism on APCs producing safe, long-lasting
immune responses and have the potential for rapid development and production (23). Another
potential advantage for mRNA vaccines is described in a 2017 manuscript by Stitz et.al. where they
developed a thermostable mRNA vaccine against rabies (24). Most vaccines require special storage
conditions including cryopreservation from production until administration to the patient. The
thermostable mRNA vaccine described here represents opportunities for greater access to life-saving
prophylactic vaccines even in remote areas that lack modern refrigeration.
DiabetesType 1 diabetes is a lifelong disease that could potentially be cured with xenotransplantation or
allotransplantation of pancreatic tissue, but these procedures are highly invasive with significant
mortality and morbidity (25). Recent efforts to engineer pancreatic islet cells for transplantation are now
leveraging gene editing and RNAi methods to potentiate immune responses, reduce risk of acute or
chronic rejection, and improve outcomes for transplant patients (26). In 2006, researchers at the
University of Bracelona, demonstrated that transient RNAi against Phosphoenolpyruvate carboxykinase
(PEPCK) lowered blood glucose and improved glucose tolerance in diabetic animal models suggesting an
alternative treatment mechanism for diabetes (27). Diabetic patients often suffer from chronic wounds
related to the disease that can be life-threatening. Two recent articles have described improved diabetic
wound healing using engineered miR-31 exosomes or engineered hydrogels containing siRNAs which
reduce inflammatory signaling and promote wound healing programs (28,29). This again demonstrates
the breath of therapeutic applications and opportunities for RNA-based therapies.
Conclusions & Future Directions:
The RNA world is vast, diverse, and our understanding of RNA biology is constantly evolving. A class of
circular RNAs (circRNAs) which were discovered more than 4 decades ago have become attractive
therapeutic targets and engineered synthetic circRNAs are gaining commercial interest as an RNA-based
therapeutic platform. Similar to NextRNA Therapeutics previously mentioned, some companies like
Ribometrix are developing small molecules targeting malignant RNAs based on their unique 3D
structure. The global pandemic has attracted more interest than ever before in the field. Scientific
knowledge and commercial interest in RNA-based therapeutics are on the rise. As of August 18, 2022,
1,376 Scientific publications of RNA therapies appear in NCBI PubMed records since the early 1990’s
(PubMed). Nearly 81% of these were published in the past decade and more than 40% since 2020. This
represents a 22.56% CAGR of publications from 2015 to 2021. There are 407 U.S. patent filings for “RNA
vaccines” with nearly 72% filed since 2015 and over 31% of all patents filed since 2020 (USPTO).
However, there are still opportunities for advancement and discovery in RNA therapy field. Less than 5%
of oncology/cancer-related U.S. patents include RNAi applications and only around 0.1% of oncology
patents mention lncRNAs. Targeted in vivo delivery remains challenging for many disease applications.
Methods for discovering disease-related targets and subsequent design of RNA therapies with high
efficacy, specificity, and favorable safety profiles are rapidly evolving. High-throughput screening
approaches for candidate selection and testing can be costly. Numerous in silico-based approaches for
rational design of RNA drugs are under development including a commercial digital platform (Benchling)
that has garnered attention from numerous pharmaceutical & biotechnology companies.
Staying on top of the RNA scientific and clinical development landscape can be challenging, especially
given the rapid growth of new companies and the pace of new discoveries in this space. There are a
multitude of promising preclinical and clinical stage RNA-based or RNA targeting products and
platforms. For pharmaceutical, CRO, and biotechnology companies in or planning to enter this space,
opportunities abound, but having the right partners with the knowledge and resources to navigate the
RNA therapeutic/biotechnology landscape can help jumpstart your program(s) while avoiding pitfalls or
setbacks.
Want to learn more?
Abbreviations:
RNA – ribonucleic acid
DNA – deoxyribonucleic acid
mRNA – messenger RNA
ncRNA – non-coding RNA
lncRNA – long non-coding RNA
miRNA – microRNA
rRNA – ribosomal RNA
RNAi – RNA interference
siRNA – silencing or short/small interfering RNA
LNP – lipid nanoparticles
circRNA – circular RNA
cET - 2’,4’-constrained ethyl base modification
ASO/ANO – antisense oligonucleotides
About the author:
Christian T. Stackhouse, Ph.D.
Christian has more than a decade of primary scientific research experience in the Life Sciences with
more than 8years of experience in translational oncology research. He received his doctorate in
Neuroscience from the University of Alabama at Birmingham working principally in the areas of neurooncology and RNA biology. Before joining Sedulo as a Client Engagement Manager, he completed a
postdoctoral fellowship at Duke University in the hematology-oncology division of the Department of
Pediatrics. Christian has numerous peer-reviewed scientific publications, abstracts, and has presented at
national & international conferences for basic science, epidemiology, oncology, and computer science.
His most recent article: “An in vivo model of glioblastoma radiation resistance identifies long non-coding
RNAs and targetable kinases” was published in JCI Insight in August 2022.
References:
1) Wolff, J. A., Malone, R. W., Williams, P., Chong, W., Acsadi, G., Jani, A., & Felgner, P. L. (1990).
Direct gene transfer into mouse muscle in vivo. Science (New York, N.Y.),- Pt 1),-. https://doi.org/10.1126/science-) Saleem A, Matthews JC, Ranson M, Callies S, André V, Lahn M, Dickinson C, Prenant C, Brown G,
McMahon A, Talbot DC, Jones T, Price PM. Molecular Imaging and Pharmacokinetic Analysis of
Carbon-11 Labeled Antisense Oligonucleotide LY- in Cancer Patients. Theranostics.
2011;1:290-301. doi: 10.7150/thno/v01p0290. Epub 2011 Jun 1. PMID:-; PMCID:
PMC-) Matsuo M. Antisense Oligonucleotide-Mediated Exon-skipping Therapies: Precision Medicine
Spreading from Duchenne Muscular Dystrophy. JMA J. 2021 Jul 15;4(3):232-240. doi:-/jmaj-. Epub 2021 Jul 9. PMID:-; PMCID: PMC-) Stackhouse, C. T., Gillespie, G. Y., & Willey, C. D. (2020). Exploring the Roles of lncRNAs in GBM
Pathophysiology and Their Therapeutic Potential. Cells, 9(11), 2369.
https://doi.org/10.3390/cells-Miao, L., Zhang, Y., & Huang, L. (2021). mRNA vaccine for
cancer immunotherapy. Molecular cancer, 20(1), 41. https://doi.org/10.1186/s-) Krienke, C., Kolb, L., Diken, E., Streuber, M., Kirchhoff, S., Bukur, T., Akilli-Öztürk, Ö., Kranz, L. M.,
Berger, H., Petschenka, J., Diken, M., Kreiter, S., Yogev, N., Waisman, A., Karikó, K., Türeci, Ö., &
Sahin, U. (2021). A noninflammatory mRNA vaccine for treatment of experimental autoimmune
encephalomyelitis. Science (New York, N.Y.),-), 145–153.
https://doi.org/10.1126/science.aay3638
6) Qiu, M., Glass, Z., Chen, J., Haas, M., Jin, X., Zhao, X., Rui, X., Ye, Z., Li, Y., Zhang, F., & Xu, Q.
(2021). Lipid nanoparticle-mediated codelivery of Cas9 mRNA and single-guide RNA achieves
liver-specific in vivo genome editing of Angptl3. Proceedings of the National Academy of
Sciences of the United States of America, 118(10), e-.
https://doi.org/10.1073/pnas-) Iwamoto, N., Butler, D., Svrzikapa, N., Mohapatra, S., Zlatev, I., Sah, D., Meena, Standley, S. M.,
Lu, G., Apponi, L. H., Frank-Kamenetsky, M., Zhang, J. J., Vargeese, C., & Verdine, G. L. (2017).
Control of phosphorothioate stereochemistry substantially increases the efficacy of antisense
oligonucleotides. Nature biotechnology, 35(9), 845–851. https://doi.org/10.1038/nbt.3948
8) Stein CA, Hansen JB, Lai J, Wu S, Voskresenskiy A, Høg A, Worm J, Hedtjärn M, Souleimanian N,
Miller P, Soifer HS, Castanotto D, Benimetskaya L, Ørum H, Koch T. Efficient gene silencing by
delivery of locked nucleic acid antisense oligonucleotides, unassisted by transfection reagents.
Nucleic Acids Res. 2010 Jan;38(1):e3. doi: 10.1093/nar/gkp841. Epub 2009 Oct 23. PMID:-; PMCID: PMC-) Kumthekar P, Ko CH, Paunesku T, et al. A first-in-human phase 0 clinical study of RNA
interference-based spherical nucleic acids in patients with recurrent glioblastoma. Sci Transl
Med. 2021;13(584):eabb3945. doi:10.1126/scitranslmed.abb3945
10) Germer K, Leonard M, Zhang X. RNA aptamers and their therapeutic and diagnostic applications.
Int J Biochem Mol Biol. 2013 Mar 31;4(1):27-40. PMID:-; PMCID: PMC-) Guo P. The emerging field of RNA nanotechnology. Nat Nanotechnol. 2010 Dec;5(12):833-42.
doi: 10.1038/nnano-. Epub 2010 Nov 21. PMID:-; PMCID: PMC-) Suzuki Y, Ishihara H. Difference in the lipid nanoparticle technology employed in three approved
siRNA (Patisiran) and mRNA (COVID-19 vaccine) drugs. Drug Metab Pharmacokinet. 2021
Dec;41:100424. doi: 10.1016/j.dmpk-. Epub 2021 Oct 10. PMID:-; PMCID:
PMC-) Adams, D., Gonzalez-Duarte, A., O'Riordan, W. D., Yang, C. C., Ueda, M., Kristen, A. V., Tournev,
I., Schmidt, H. H., Coelho, T., Berk, J. L., Lin, K. P., Vita, G., Attarian, S., Planté-Bordeneuve, V.,
Mezei, M. M., Campistol, J. M., Buades, J., Brannagan, T. H., 3rd, Kim, B. J., Oh, J., … Suhr, O. B.
(2018). Patisiran, an RNAi Therapeutic, for Hereditary Transthyretin Amyloidosis. The New
England journal of medicine, 379(1), 11–21. https://doi.org/10.1056/NEJMoa-) Anthony K. RNA-based therapeutics for neurological diseases. RNA Biol. 2022;19(1):176-190.
doi: 10.1080/-. Epub 2021 Dec 31. PMID:-; PMCID:
PMC-) Chang JL, Hinrich AJ, Roman B, et al. Targeting Amyloid-β precursor protein, APP, splicing with
antisense oligonucleotides reduces toxic amyloid-β production. Mol Ther. 2018;26:-) Kalbfuss B, Mabon SA, Misteli T. Correction of alternative splicing of tau in frontotemporal
dementia and parkinsonism linked to chromosome 17. J Biol Chem. 2001;276:-) Du L, Kayali R, Bertoni C, Fike F, Hu H, Iversen PL, Gatti RA. Arginine-rich cell-penetrating peptide
dramatically enhances AMO-mediated ATM aberrant splicing correction and enables delivery to
brain and cerebellum. Hum Mol Genet. 2011 Aug 15;20(16):3151-60. doi: 10.1093/hmg/ddr217.
Epub 2011 May 16. PMID:-; PMCID: PMC-) Shae D., Baljon J.J., Wehbe M., Becker K.W., Sheehy T.L., Wilson J.T. At the bench: Engineering
the next generation of cancer vaccines. J. Leukoc. Biol. 2019 doi: 10.1002/JLB.5BT-R.
19) Yoo B, Jordan VC, Sheedy P, Billig AM, Ross A, Pantazopoulos P, Medarova Z. RNAi-Mediated PDL1 Inhibition for Pancreatic Cancer Immunotherapy. Sci Rep. 2019 Mar 18;9(1):4712. doi:
10.1038/s-. PMID:-; PMCID: PMC-) Revia RA, Stephen ZR, Zhang M. Theranostic Nanoparticles for RNA-Based Cancer Treatment.
Acc Chem Res. 2019 Jun 18;52(6):-. doi: 10.1021/acs.accounts.9b00101. Epub 2019
May 28. PMID:-; PMCID: PMC-) Li, K., Zhang, Y., Hussain, A., Weng, Y., & Huang, Y. (2021). Progress of Photodynamic and RNAi
Combination Therapy in Cancer Treatment. ACS biomaterials science & engineering, 7(9),-. https://doi.org/10.1021/acsbiomaterials.1c-) Stackhouse, C. T., Anderson, J. C., Yue, Z., Nguyen, T., Eustace, N. J., Langford, C. P., Wang, J.,
Rowland Iv, J. R., Xing, C., Mikhail, F. M., Cui, X., Alrefai, H., Bash, R. E., Lee, K. J., Yang, E. S.,
Hjelmeland, A. B., Miller, C. R., Chen, J. Y., Gillespie, G. Y., & Willey, C. D. (2022). An in vivo
model of glioblastoma radiation resistance identifies long non-coding RNAs and targetable
kinases. JCI insight, e148717. Advance online publication.
https://doi.org/10.1172/jci.insight-) Zhang C, Maruggi G, Shan H, Li J. Advances in mRNA Vaccines for Infectious Diseases. Front
Immunol. 2019 Mar 27;10:594. doi: 10.3389/fimmu-. PMID:-; PMCID:
PMC-) Stitz L, Vogel A, Schnee M, Voss D, Rauch S, Mutzke T, Ketterer T, Kramps T, Petsch B. A
thermostable messenger RNA based vaccine against rabies. PLoS Negl Trop Dis. 2017 Dec
7;11(12):e-. doi: 10.1371/journal.pntd-. PMID:-; PMCID: PMC-) Troppmann C. (2010). Complications after pancreas transplantation. Current opinion in organ
transplantation, 15(1), 112–118. https://doi.org/10.1097/MOT.0b013e-) Li F, Mahato RI. RNA interference for improving the outcome of islet transplantation. Adv Drug
Deliv Rev. 2011 Jan-Feb;63(1-2):47-68. doi: 10.1016/j.addr-. Epub 2010 Dec 13.
PMID:-; PMCID: PMC-) Gómez-Valadés, A. G., Vidal-Alabró, A., Molas, M., Boada, J., Bermúdez, J., Bartrons, R., &
Perales, J. C. (2006). Overcoming diabetes-induced hyperglycemia through inhibition of hepatic
28)
29)
30)
31)
phosphoenolpyruvate carboxykinase (GTP) with RNAi. Molecular therapy : the journal of the
American Society of Gene Therapy, 13(2), 401–410.
https://doi.org/10.1016/j.ymthe-
Huang J, Yu M, Yin W, Liang B, Li A, Li J, Li X, Zhao S, Liu F. Development of a novel RNAi therapy:
Engineered miR-31 exosomes promoted the healing of diabetic wounds. Bioact Mater. 2021 Feb
20;6(9):-. doi: 10.1016/j.bioactmat-. PMID:-; PMCID:
PMC-.
Yang, L., Zhang, L., Hu, J., Wang, W., & Liu, X. (2021). Promote anti-inflammatory and
angiogenesis using a hyaluronic acid-based hydrogel with miRNA-laden nanoparticles for chronic
diabetic wound treatment. International journal of biological macromolecules, 166, 166–178.
https://doi.org/10.1016/j.ijbiomac-
https://patft.uspto.gov/netahtml/PTO/search-bool.html
https://pubmed.ncbi.nlm.nih.gov/