The era of RNA interference medicines: the clinical landscape of synthetic gene silencing drugs

Authors

  • Bruno M. D. C. Godinho RNA Therapeutics Institute, University of Massachusetts Medical School. Worcester, USA.
  • Anastasia Khvorova Lisbon School of Health Technology, Polytechnic Institute of Lisbon. Lisbon, Portugal.

DOI:

https://doi.org/10.25758/set.2227

Keywords:

RNA interference, siRNA, miRNA, Therapeutic oligonucleotides, Gene knockdown, Drug development

Abstract

Therapeutic oligonucleotides, such as small interfering RNAs (siRNAs), provide a simple and effective tool to modulate the expression of any gene. siRNAs harness the RNA interference (RNAi) pathway to degrade disease-associated messenger RNAs (mRNAs). The inherent sequence specificity and potency of siRNAs make them ideal drug candidates that are expected to transform drug development and our approach to human health. However, the first wave of clinical trials was not immediately successful and temporarily dampened the excitement over this newly discovered technology. Most studies did not meet desired efficacy and failed to achieve clinically-relevant endpoints. Poor chemical design, lack of enzymatic stability, and inadequate delivery strategies were found to be the main issues stifling success. Recent advancements in RNA chemistry, biology, and mechanistic understanding of factors that define oligonucleotide pharmacokinetic/pharmacodynamic behavior have resulted in a fundamental shift in the clinical landscape of this novel class of therapeutic modalities. As a result, there has been a dramatic increase in both the number of clinical trials and, more importantly, the level of observed clinical efficacy. In 2018, we witnessed a major landmark for the field with the first formulation-based RNAi therapeutic, Patisiran (OnpattroTM), being approved by the Food and Drug Administration and the European Medicines Agency. Approximately a year later, another breakthrough was achieved with the approval of Givosiran (GIVLAARITM), the first siRNA using the conjugate-mediated approach for targeted delivery to hepatocytes. Both these approvals brought revitalized hope and enthusiasm to the field, and have restored the interest in RNAi, as a powerful disease-modifying therapeutic strategy for a variety of genetically-defined disorders. This review gives an overview of the clinical landscape of synthetic RNAi drugs, contextualizing how advances in RNAi chemistry and formulation strategies have helped define the clinical utility of this promising class of drugs.

Downloads

Download data is not yet available.

References

Napoli C, Lemieux C, Jorgensen R. Introduction of a chimeric chalcone synthase gene into petunia results in reversible co-suppression of homologous genes in trans. Plant Cell. 1990;2(4):279-89.

Fire A, Xu S, Montgomery MK, Kostas SA, Driver SE, Mello CC. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature. 1998;391(6669):806-11.

Elbashir SM, Martinez J, Patkaniowska A, Lendeckel W, Tuschl T. Functional anatomy of siRNAs for mediating efficient RNAi in Drosophila melanogaster embryo lysate. EMBO J. 2001;20(23):6877-88.

Wood H. FDA approves patisiran to treat hereditary transthyretin amyloidosis. Nat Rev Neurol. 2018;14(10):570.

Corey DR. RNA learns from antisense. Nat Chem Biol. 2007;3:8.

Haussecker D. The business of RNAi therapeutics in 2012. Mol Ther Nucleic Acids. 2012;1(2):e8.

Khvorova A, Watts JK. The chemical evolution of oligonucleotide therapies of clinical utility. Nat Biotechnol. 2017;35(3):238-48.

Plasterk RH. RNA silencing: the genome's immune system. Science. 2002;296(5571):1263-5.

Meister G, Tuschl T. Mechanisms of gene silencing by double-stranded RNA. Nature. 2004;431(7006):343-9.

Huang W. MicroRNAs: biomarkers, diagnostics, and therapeutics. In: Huang J, Borchert GM, Dou D, Huan J, Lan W, Tan M, et al., editors. Bioinformatics in MicroRNA research. New York: Springer; 2017. p. 57-67. ISBN 9781493970469

Thomas CE, Ehrhardt A, Kay MA. Progress and problems with the use of viral vectors for gene therapy. Nat Rev Genet. 2003;4(5):346-58.

Couto LB, High KA. Viral vector-mediated RNA interference. Curr Opin Pharmacol. 2010;10(5):534-42.

Marshall E. Gene therapy death prompts review of adenovirus vector. Science. 1999;286(5448):2244-5.

Ginn SL, Amaya AK, Alexander IE, Edelstein M, Abedi MR. Gene therapy clinical trials worldwide to 2017: an update. J Gene Med. 2018;20(5):e3015.

Borel F, Kay MA, Mueller C. Recombinant AAV as a platform for translating the therapeutic potential of RNA interference. Mol Ther. 2014;22(4):692-701.

Boudreau RL, Spengler RM, Hylock RH, Kusenda BJ, Davis HA, Eichmann DA, et al. siSPOTR: a tool for designing highly specific and potent siRNAs for human and mouse. Nucleic Acids Res. 2012;41(1):e9.

Naito Y, Ui-Tei K. siRNA design software for a target gene-specific RNA interference. Front Genet. 2012;3(102).

Reynolds A, Leake D, Boese Q, Scaringe S, Marshall WS, Khvorova A. Rational siRNA design for RNA interference. Nat Biotechnol. 2004;22(3):326-30.

Birmingham A, Anderson E, Sullivan K, Reynolds A, Boese Q, Leake D, et al. A protocol for designing siRNAs with high functionality and specificity. Nat Protoc. 2007;2(9):2068-78.

Godinho BM, Coles AH, Khvorova A. Conjugate-mediated delivery of RNAi-based therapeutics: enhancing pharmacokinetics-pharmacodynamics relationships of medicinal oligonucleotides. In: Agrawal S, Gait MJ, editors. Advances in nucleic acid therapeutics. Royal Society of Chemistry; 2019. p. 206-32. ISBN 9781788015714

Kleinman ME, Yamada K, Takeda A, Chandrasekaran V, Nozaki M, Baffi JZ, et al. Sequence- and target-independent angiogenesis suppression by siRNA via TLR3. Nature. 2008;452(7187):591-7.

Robbins M, Judge A, Liang L, McClintock K, Yaworski E, MacLachlan I. 2′-O-methyl-modified RNAs Act as TLR7 Antagonists. Mol Ther. 2007;15(9):1663-9.

Sledz CA, Holko M, de Veer MJ, Silverman RH, Williams BRG. Activation of the interferon system by short-interfering RNAs. Nat Cell Biol. 2003;5(9):834-9.

Schlee M, Hornung V, Hartmann G. siRNA and isRNA: two edges of one sword. Mol Ther. 2006;14(4):463-70.

Watts JK, Deleavey GF, Damha MJ. Chemically modified siRNA: tools and applications. Drug Discov Today. 2008;13(19-20):842-55.

Haraszti RA, Roux L, Coles AH, Turanov AA, Alterman JF, Echeverria D, et al. 5΄-Vinylphosphonate improves tissue accumulation and efficacy of conjugated siRNAs in vivo. Nucleic Acids Res. 2017;45(13):7581-92.

Yin H, Kanasty RL, Eltoukhy AA, Vegas AJ, Dorkin JR, Anderson DG. Non-viral vectors for gene-based therapy. Nat Rev Genet. 2014;15(8):541-55.

Guzman-Villanueva D, El-Sherbiny IM, Herrera-Ruiz D, Vlassov AV, Smyth HD. Formulation approaches to short interfering RNA and MicroRNA: challenges and implications. J Pharm Sci. 2012;101(11):4046-66.

Osborn MF, Coles AH, Biscans A, Haraszti RA, Roux L, Davis S, et al. Hydrophobicity drives the systemic distribution of lipid-conjugated siRNAs via lipid transport pathways. Nucleic Acids Res. 2018;47(3):1070-81.

Zimmermann TS, Karsten V, Chan A, Chiesa J, Boyce M, Bettencourt BR, et al. Clinical proof of concept for a novel hepatocyte-targeting GalNAc-siRNA conjugate. Mol Ther. 2017;25(1):71-8.

Springer AD, Dowdy SF. GalNAc-siRNA conjugates: leading the way for delivery of RNAi therapeutics. Nucleic Acid Ther. 2018;28(3):109-18.

Garba AO, Mousa SA. Bevasiranib for the treatment of wet, age-related macular degeneration. Ophthalmol Eye Dis. 2010;2:75-83.

Kaiser PK, Symons RA, Shah SM, Quinlan EJ, Tabandeh H, Do DV, et al. RNAi-based treatment for neovascular age-related macular degeneration by Sirna-027. Am J Ophthalmol. 2010;150(1):33-9. e2.

Peddi V, Ratner L, Cooper M, Gaber O, Feng S, Tso P, et al. Treatment with QPI-1002, a short interfering (SI) RNA for the prophylaxis of delayed graft function: abstract# 2967. Transplantation. 2014;98:153.

Koldehoff M, Steckel NK, Beelen DW, Elmaagacli AH. Therapeutic application of small interfering RNA directed against bcr-abl transcripts to a patient with imatinib-resistant chronic myeloid leukaemia. Clin Exp Med. 2007;7(2):47-55.

Davis ME, Zuckerman JE, Choi CHJ, Seligson D, Tolcher A, Alabi CA, et al. Evidence of RNAi in humans from systemically administered siRNA via targeted nanoparticles. Nature. 2010;464(7291):1067-70.

Rozema DB, Lewis DL, Wakefield DH, Wong SC, Klein JJ, Roesch PL, et al. Dynamic polyConjugates for targeted in vivo delivery of siRNA to hepatocytes. Proc Nat Acad Sci. 2007;104(32):12982-7.

Rupaimoole R, Slack FJ. MicroRNA therapeutics: towards a new era for the management of cancer and other diseases. Nat Rev Drug Discov. 2017;16(3):203-22.

Schluep T, Lickliter J, Hamilton J, Lewis DL, Lai CL, Lau JY, et al. Safety, tolerability, and pharmacokinetics of ARC‐520 injection, an RNA interference‐based therapeutic for the treatment of chronic hepatitis B virus infection, in healthy volunteers. Clin Pharmacol Drug Dev. 2017;6(4):350-62.

Turner AM, Stolk J, Bals R, Lickliter JD, Hamilton J, Christianson DR, et al. Hepatic-targeted RNA interference provides robust and persistent knockdown of alpha-1 antitrypsin levels in ZZ patients. J Hepatol. 2018;69(2):378-84.

Adams D, Gonzalez-Duarte A, O’Riordan WD, Yang C-C, Ueda M, Kristen AV, et al. Patisiran, an RNAi therapeutic, for hereditary transthyretin amyloidosis. N Engl J Med. 2018;379(1):11-21.

Pavco P, Libertine L, Young V, Woolery-Lloyd H, Cauwenbergh G. RXI-109 treatment to reduce the formation of keloids following keloidectomy. J Am Acad Dermatol. 2015;72(5 Suppl 1):AB273.

Mullard A. RNAi hits another rut. Nat Rev Drug Discov. 2016;15(11):738.

Ray KK, Landmesser U, Leiter LA, Kallend D, Dufour R, Karakas M, et al. Inclisiran in patients at high cardiovascular risk with elevated LDL cholesterol. N Engl J Med. 2017;376(15):1430-40.

Huang Y. Preclinical and clinical advances of GalNAc-decorated nucleic acid therapeutics. Mol Ther Nucleic Acids. 2017;6:116-32.

Ando Y, Coelho T, Berk JL, Cruz MW, Ericzon B-G, Ikeda S-i, et al. Guideline of transthyretin-related hereditary amyloidosis for clinicians. Orphanet J Rare Dis. 2013;8:31.

Suhr OB, Coelho T, Buades J, Pouget J, Conceicao I, Berk J, et al. Efficacy and safety of patisiran for familial amyloidotic polyneuropathy: a phase II multi-dose study. Orphanet J Rare Dis. 2015;10:109.

Benson MD, Waddington-Cruz M, Berk JL, Polydefkis M, Dyck PJ, Wang AK, et al. Inotersen treatment for patients with hereditary transthyretin amyloidosis. N Engl J Med. 2018;379(1):22-31.

Sardh E, Harper P, Balwani M, Stein P, Rees D, Bissell DM, et al. Phase 1 Trial of an RNA interference therapy for acute intermittent porphyria. N Engl J Med. 2019;380(6):549-58.

Brandão PR, Titze-de-Almeida SS, Titze-de-Almeida R. Leading RNA interference therapeutics part 2: silencing delta-aminolevulinic acid synthase 1, with a focus on Givosiran. Mol Diagn Ther. 2019 December 2. [Online ahead of print].

Published

2022-07-29

Issue

Section

Artigos de Revisão

How to Cite

The era of RNA interference medicines: the clinical landscape of synthetic gene silencing drugs. (2022). Saúde & Tecnologia, 21, 05-17. https://doi.org/10.25758/set.2227