Chemical engineering of therapeutic siRNAs for allele-specific gene silencing in vivo in CNS

Small interfering RNAs (siRNAs) are a new class of drugs, exhibiting sequence-driven, potent, and sustained silencing of gene expression in vivo. We recently demonstrated that siRNA chemical architectures can be optimized to provide efficient delivery to the CNS. Many genetically-defined neurodegenerative disorders are autosomal dominant favoring selective silencing of the mutant allele. In some cases, successful targeting of the mutant allele requires targeting of a single nucleotide polymorphism (SNP) heterozygosity. Using Huntington’s disease as a model, we demonstrate allele-specific RNAi-based silencing of gene expression in vivo and in neurons differentiated from HD patient-derived iPSCs. A series of in vitro screens, with chemical and thermodynamic optimization, identified compounds with >50-fold selectivity for the mutant HD-causing allele, based on a single nucleotide difference. The optimized compound exhibits selective silencing of mutant huntingtin (HTT) protein in patient derived cells and throughout the HD mouse brain, providing a demonstration of SNP-based allele-specific RNAi silencing of gene expression in vivo in the CNS. The ability to target a disease-causing allele using RNAi-based therapies could be applied to a wide range of dominant CNS disorders, where maintenance of wild-type expression is essential.

[1]  A. Southwell,et al.  Antisense Oligonucleotide Therapy: From Design to the Huntington Disease Clinic , 2022, BioDrugs.

[2]  M. DiFiglia,et al.  Structurally constrained phosphonate internucleotide linkage impacts oligonucleotide-enzyme interaction, and modulates siRNA activity and allele specificity , 2021, Nucleic acids research.

[3]  Katie Kingwell Double setback for ASO trials in Huntington disease , 2021, Nature Reviews Drug Discovery.

[4]  E. Nordh,et al.  Long-term safety and efficacy of patisiran for hereditary transthyretin-mediated amyloidosis with polyneuropathy: 12-month results of an open-label extension study , 2020, Lancet Neurology.

[5]  N. Svrzikapa,et al.  Investigational Assay for Haplotype Phasing of the Huntingtin Gene , 2020, Molecular therapy. Methods & clinical development.

[6]  M. Manoharan,et al.  Investigating the pharmacodynamic durability of GalNAc–siRNA conjugates , 2020, Nucleic acids research.

[7]  Lawrence A Leiter,et al.  Two Phase 3 Trials of Inclisiran in Patients with Elevated LDL Cholesterol. , 2020, The New England journal of medicine.

[8]  H. Yin,et al.  Striatal Projection Neurons Require Huntingtin for Synaptic Connectivity and Survival. , 2020, Cell reports.

[9]  M. Hayden,et al.  Potent and sustained huntingtin lowering via AAV5 encoding miRNA preserves striatal volume and cognitive function in a humanized mouse model of Huntington disease , 2019, Nucleic acids research.

[10]  M. Hayden,et al.  A Comprehensive Haplotype Targeting Strategy for Allele-Specific HTT Suppression in Huntington Disease. , 2019, American journal of human genetics.

[11]  P. Zamore,et al.  High-Throughput Analysis Reveals Rules for Target RNA Binding and Cleavage by AGO2. , 2019, Molecular cell.

[12]  M. DiFiglia,et al.  A divalent siRNA chemical scaffold for potent and sustained modulation of gene expression throughout the central nervous system , 2019, Nature Biotechnology.

[13]  Lisa M Anderson,et al.  Huntingtin suppression restores cognitive function in a mouse model of Huntington’s disease , 2018, Science Translational Medicine.

[14]  V. Mattis,et al.  Human Huntington's Disease iPSC-Derived Cortical Neurons Display Altered Transcriptomics, Morphology, and Maturation. , 2018, Cell reports.

[15]  A. Khvorova,et al.  Functional features defining the efficacy of cholesterol-conjugated, self-deliverable, chemically modified siRNAs , 2018, Nucleic acids research.

[16]  K. G. Rajeev,et al.  RNA octamer containing 2'-OMe, 4'- Cbeta-OMe U. , 2018 .

[17]  S. Solomon,et al.  Patisiran, an RNAi Therapeutic, for Hereditary Transthyretin Amyloidosis , 2018, The New England journal of medicine.

[18]  M. Moore,et al.  Comparison of partially and fully chemically-modified siRNA in conjugate-mediated delivery in vivo , 2018, Nucleic acids research.

[19]  S. Milstein,et al.  Advanced siRNA Designs Further Improve In Vivo Performance of GalNAc-siRNA Conjugates , 2018, Molecular therapy : the journal of the American Society of Gene Therapy.

[20]  Zicai Liang,et al.  Site-Specific Modification Using the 2′-Methoxyethyl Group Improves the Specificity and Activity of siRNAs , 2017, Molecular therapy. Nucleic acids.

[21]  P. Dietrich,et al.  Elimination of huntingtin in the adult mouse leads to progressive behavioral deficits, bilateral thalamic calcification, and altered brain iron homeostasis , 2017, PLoS genetics.

[22]  Shihua Li,et al.  CRISPR/Cas9-mediated gene editing ameliorates neurotoxicity in mouse model of Huntington’s disease , 2017, The Journal of clinical investigation.

[23]  Daniel G. Anderson,et al.  Advances in the delivery of RNA therapeutics: from concept to clinical reality , 2017, Genome Medicine.

[24]  S. Hersch,et al.  KEAP1-modifying small molecule reveals muted NRF2 signaling responses in neural stem cells from Huntington's disease patients , 2017, Proceedings of the National Academy of Sciences.

[25]  M. Hayden,et al.  A novel humanized mouse model of Huntington disease for preclinical development of therapeutics targeting mutant huntingtin alleles , 2017, Human molecular genetics.

[26]  anastasia. khvorova,et al.  The chemical evolution of oligonucleotide therapies of clinical utility , 2017, Nature Biotechnology.

[27]  Lawrence M. Lifshitz,et al.  Visualization of self-delivering hydrophobically modified siRNA cellular internalization , 2016, Nucleic acids research.

[28]  M. Hayden,et al.  Design, Characterization, and Lead Selection of Therapeutic miRNAs Targeting Huntingtin for Development of Gene Therapy for Huntington's Disease , 2016, Molecular therapy. Nucleic acids.

[29]  A. Khvorova,et al.  Hydrophobically Modified siRNAs Silence Huntingtin mRNA in Primary Neurons and Mouse Brain , 2015, Molecular therapy. Nucleic acids.

[30]  M. Hayden,et al.  Huntingtin Haplotypes Provide Prioritized Target Panels for Allele-specific Silencing in Huntington Disease Patients of European Ancestry. , 2015, Molecular therapy : the journal of the American Society of Gene Therapy.

[31]  L. Hood,et al.  Sequence-Level Analysis of the Major European Huntington Disease Haplotype. , 2015, American journal of human genetics.

[32]  M. Moore,et al.  Single-Molecule Imaging Reveals that Argonaute Reshapes the Binding Properties of Its Nucleic Acid Guides , 2015, Cell.

[33]  M. Hayden,et al.  HD iPSC-derived neural progenitors accumulate in culture and are susceptible to BDNF withdrawal due to glutamate toxicity. , 2015, Human molecular genetics.

[34]  Fritz Eckstein,et al.  Phosphorothioates, essential components of therapeutic oligonucleotides. , 2014, Nucleic acid therapeutics.

[35]  N. Déglon,et al.  Allele-Specific Silencing of Mutant Huntingtin in Rodent Brain and Human Stem Cells , 2014, PloS one.

[36]  M. Wood,et al.  Allele-specific silencing of mutant Ataxin-7 in SCA7 patient-derived fibroblasts , 2014, European Journal of Human Genetics.

[37]  T. Taksir,et al.  Silencing mutant huntingtin by adeno-associated virus-mediated RNA interference ameliorates disease manifestations in the YAC128 mouse model of Huntington's disease. , 2014, Human gene therapy.

[38]  K. Ye,et al.  Single modification at position 14 of siRNA strand abolishes its gene‐silencing activity by decreasing both RISC loading and target degradation , 2013, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[39]  C. Fabián Flores-Jasso,et al.  Argonaute Divides Its RNA Guide into Domains with Distinct Functions and RNA-Binding Properties , 2012, Cell.

[40]  G. Deleavey,et al.  Designing chemically modified oligonucleotides for targeted gene silencing. , 2012, Chemistry & biology.

[41]  L. Shihabuddin,et al.  Sustained Therapeutic Reversal of Huntington's Disease by Transient Repression of Huntingtin Synthesis , 2012, Neuron.

[42]  D. Corey,et al.  Silencing disease genes in the laboratory and the clinic , 2012, The Journal of pathology.

[43]  S. Freier,et al.  Potent and selective antisense oligonucleotides targeting single-nucleotide polymorphisms in the Huntington disease gene / allele-specific silencing of mutant huntingtin. , 2011, Molecular therapy : the journal of the American Society of Gene Therapy.

[44]  M. Manoharan,et al.  Unexpected origins of the enhanced pairing affinity of 2′-fluoro-modified RNA , 2010, Nucleic acids research.

[45]  M. Mogi,et al.  Listening to the sound of silence between men and women , 2010, Hypertension Research.

[46]  S. Shukla,et al.  Exploring Chemical Modifications for siRNA Therapeutics: A Structural and Functional Outlook , 2010, ChemMedChem.

[47]  C. Gellera,et al.  A majority of Huntington's disease patients may be treatable by individualized allele-specific RNA interference , 2009, Experimental Neurology.

[48]  B. Landwehrmeyer,et al.  Five siRNAs Targeting Three SNPs May Provide Therapy for Three-Quarters of Huntington's Disease Patients , 2009, Current Biology.

[49]  Thomas J Hudson,et al.  CAG expansion in the Huntington disease gene is associated with a specific and targetable predisposing haplogroup. , 2009, American journal of human genetics.

[50]  N. Minakawa,et al.  Synthesis and characterization of 2′-modified-4′-thioRNA: a comprehensive comparison of nuclease stability , 2009, Nucleic acids research.

[51]  R. Friedlander,et al.  Allele‐specific silencing of mutant Huntington’s disease gene , 2009, Journal of neurochemistry.

[52]  Carlos Cepeda,et al.  Full-Length Human Mutant Huntingtin with a Stable Polyglutamine Repeat Can Elicit Progressive and Selective Neuropathogenesis in BACHD Mice , 2008, The Journal of Neuroscience.

[53]  Stefan L Ameres,et al.  The impact of target site accessibility on the design of effective siRNAs , 2008, Nature Biotechnology.

[54]  K. G. Rajeev,et al.  Therapeutic silencing of mutant huntingtin with siRNA attenuates striatal and cortical neuropathology and behavioral deficits , 2007, Proceedings of the National Academy of Sciences.

[55]  J. Schelter,et al.  Designing siRNA That Distinguish between Genes That Differ by a Single Nucleotide , 2006, PLoS genetics.

[56]  J. Lieberman,et al.  Determinants of specific RNA interference-mediated silencing of human beta-globin alleles differing by a single nucleotide polymorphism. , 2006, Proceedings of the National Academy of Sciences of the United States of America.

[57]  M. Hayden,et al.  Wild‐type huntingtin protects neurons from excitotoxicity , 2006, Journal of neurochemistry.

[58]  Elena Cattaneo,et al.  Normal huntingtin function: an alternative approach to Huntington's disease , 2005, Nature Reviews Neuroscience.

[59]  P. Zamore,et al.  Kinetic analysis of the RNAi enzyme complex , 2004, Nature Structural &Molecular Biology.

[60]  Phillip D Zamore,et al.  Sequence-Specific Inhibition of Small RNA Function , 2004, PLoS biology.

[61]  T. Tuschl,et al.  Functional anatomy of siRNAs for mediating efficient RNAi in Drosophila melanogaster embryo lysate , 2001, The EMBO journal.

[62]  Michael S. Levine,et al.  Inactivation of Hdh in the brain and testis results in progressive neurodegeneration and sterility in mice , 2000, Nature Genetics.

[63]  G. Varani,et al.  The G x U wobble base pair. A fundamental building block of RNA structure crucial to RNA function in diverse biological systems. , 2000, EMBO reports.

[64]  A. Fire,et al.  Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans , 1998, Nature.

[65]  R. Carraway,et al.  Huntingtin is a cytoplasmic protein associated with vesicles in human and rat brain neurons , 1995, Neuron.

[66]  P. D. Cook,et al.  Uniformly modified 2'-deoxy-2'-fluoro phosphorothioate oligonucleotides as nuclease-resistant antisense compounds with high affinity and specificity for RNA targets. , 1993, Journal of medicinal chemistry.

[67]  Ryan M Spengler,et al.  Artificial miRNAs Targeting Mutant Huntingtin Show Preferential Silencing In Vitro and In Vivo. , 2015, Molecular therapy. Nucleic acids.

[68]  M. Manto,et al.  [Autosomal dominant spinocerebellar ataxia]. , 1999, Revue medicale de Bruxelles.