WO2024196804A1 - Factor viii splice-modulating antisense oligonucleotides and methods of use - Google Patents
Factor viii splice-modulating antisense oligonucleotides and methods of use Download PDFInfo
- Publication number
- WO2024196804A1 WO2024196804A1 PCT/US2024/020275 US2024020275W WO2024196804A1 WO 2024196804 A1 WO2024196804 A1 WO 2024196804A1 US 2024020275 W US2024020275 W US 2024020275W WO 2024196804 A1 WO2024196804 A1 WO 2024196804A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- aso
- exon
- splicing
- interest
- splice
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P7/00—Drugs for disorders of the blood or the extracellular fluid
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/7088—Compounds having three or more nucleosides or nucleotides
- A61K31/7105—Natural ribonucleic acids, i.e. containing only riboses attached to adenine, guanine, cytosine or uracil and having 3'-5' phosphodiester links
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K9/00—Medicinal preparations characterised by special physical form
- A61K9/0012—Galenical forms characterised by the site of application
- A61K9/0019—Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/745—Blood coagulation or fibrinolysis factors
- C07K14/755—Factors VIII, e.g. factor VIII C (AHF), factor VIII Ag (VWF)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/11—DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
- C12N15/113—Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K9/00—Medicinal preparations characterised by special physical form
- A61K9/08—Solutions
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2310/00—Structure or type of the nucleic acid
- C12N2310/10—Type of nucleic acid
- C12N2310/11—Antisense
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2310/00—Structure or type of the nucleic acid
- C12N2310/30—Chemical structure
- C12N2310/31—Chemical structure of the backbone
- C12N2310/315—Phosphorothioates
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2310/00—Structure or type of the nucleic acid
- C12N2310/30—Chemical structure
- C12N2310/32—Chemical structure of the sugar
- C12N2310/321—2'-O-R Modification
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2320/00—Applications; Uses
- C12N2320/30—Special therapeutic applications
- C12N2320/33—Alteration of splicing
Definitions
- Noncoding sequences interrupt protein coding information (exons) in most human genes.
- conserved sequences known as splice sites (ss) demarcate exon-intron boundaries (1 ).
- Messenger RNA (mRNA) biogenesis requires intron removal from precursor transcripts and exon ligation (2, 3).
- the spliceosome assembles on each intron to catalyze the splicing reaction. This process involves the ordered assembly of five uracil-rich small nuclear ribonucleoprotein particles (U snRNPs) and hundreds of protein factors (4)(5).
- Exon definition is an initial spliceosome assembly step where splice site recognition occurs (6).
- U1 snRNP recognizes the 5'ss while the 3'ss and polypyrimidine tract bind U2 snRNP auxiliary factor (U2AF) (7-10).
- U2AF U2 snRNP auxiliary factor
- ASOs Antisense oligonucleotides
- Their facile design exploits the chemical language of nucleic acid base pairing interactions.
- problems including toxicity, delivery and stability hindered clinical translation (21 ).
- Notable examples include the splice-modulating drugs Nusinersen and Milasen. 15 years in development, Nusinersin was the first FDA approved cure for spinal muscular atrophy (23-25). By contrast, Milasen is a patientspecific ASO for treatment of Batten's disease, developed in only 16 months (26).
- the F8 gene encodes a protease required for activation of the coagulation cascade.
- F8 deficiency causes Hemophilia A (HA), a potentially lethal inherited bleeding disorder.
- HA Hemophilia A
- aberrant splicing of the F8 pre-m RNA contributes to HA etiology (27-30).
- compositions comprising the splice-modulating ASOs.
- the compositions are formulated for administration to a subject.
- Methods of treating Hemophilia A (HA) in a subject in need thereof are also provided.
- the subject exhibits aberrant splicing of Factor VIII (F8) exon 16, and the methods comprise administering to the subject a therapeutically effective amount of a composition of the present disclosure.
- FIG. 1 In vivo splicing assays reveal a highly fragile exon susceptible to mutation- induced aberrant splicing.
- A HA-causing mutations were tested for their impact on splicing across the F8 locus. Their positions are denoted by a black bar and their identifying number in the Human Gene Mutation Database (HGMD).
- B Each test exon was cloned into the HBB minigene to create a heterologous splicing reporter.
- C A representative agarose gel showing the various HA-causing mutations that can induce aberrant splicing of F8 exon 16, one of the most fragile exons discovered from the present splicing assays.
- FIG. 2 ASO interference mapping (AIM) reveals splice-modulating ASOs for F8 exon 16 and a highly splicing-sensitive mutation in exon 16.
- A A mock schematic of AIM. Each ASO used in the present AIM experiments is 18 nucleotides in length, and are designed using ribose sugars that are heavily modified. The 2’-OH is modified to contain a methoxyethyl group (2’-MOE, highlighted), and the phosphate backbone is modified to a phosphorothioate backbone (highlighted). Each 18-mer ASO is contiguous by design, tiling across exon 16 and its flanking introns with no overlaps between each ASO.
- FIG. 3 SHAPE probing identifies a native RNA structure (TSL-3-15) that is uniquely positioned at the 3’ss of F8 exon 16.
- TTL-3-15 native RNA structure
- FIG. 3 A normalized SHAPE reactivity vs. arc diagram plot comparing WT exon 16 and the A333G mutant. The plot uses SHAPE data that is plotted on the top portion of the plot, where each bar indicates the normalized SHAPE reactivity for each nucleotide position. The bottom portion of the plot depicts RNA structure predictions using the normalized SHAPE reactivity as a folding constraint. Each arc represents a base pairing interaction between the respective nucleotides involved within the sequence to form a given RNA structure.
- RNA structures unique to the WT or A333G mutant are depicted by their respective annotations. RNA structures that are shared and found in both sequence contexts are annotated black. A schematic model of exon 16 and its flanking introns are shown at the bottom of the plot to illustrate relative positions of SHAPE data. TSL-3-15 is specifically annotated to illustrate its position. (B) SHAPE-driven secondary structure prediction of TSL-3-15 depicted in its two dimensional structure. Cis- regulatory elements, core splicing signals, and ASOs are also annotated within the structure. Black represents core splicing signals such as the branchpoint motif, the poly-Y tract, and the consensus 3’ and 5’ss dinucleotides (these are explicitly indicated by arrows).
- Red-orange represents the bioinformatically predicted hnRNPAI binding sites.
- Each specific ASO and their target sequence within TSL-3-15 is annotated by a distinct color and sequence complementarity, respectively.
- All SHAPE probing data generated were done in vitro using the SHAPE reagent 2A3, and all subsequent data analysis was performed in RNA Framework.
- FIG. 4 A combination of ASOs targeting TSL-3-15 can additively enhance splicing of a highly splicing-sensitive mutation by increasing 3’ss accessibility.
- a representative agarose gel depicting the results from the in vivo splicing assays testing duo and trio ASO combinations’ ability to modulate splicing. Each splicing assay condition is annotated as shown in the matrix above the gel. Expected mRNA isoforms including or excluding the test exon are also annotated to the left of the agarose gel.
- B A plot quantifying the results from (A) using the PSI ratio.
- Each ASOs ability to significantly modulate splicing is annotated by color and corresponding effect (e.g., enhance or suppress splicing).
- C An overlay plot comparing normalized 2A3 reactivities between two distinct SHAPE probing conditions used to probe the A333G mutant.
- One SHAPE condition probes A333G with ASOs present (annotated light blue), and the other condition probes A333G without ASOs present (annotated light red).
- Admixing of colors where this is indistinguishable overlap represents similar SHAPE reactivity values between the two probing conditions at that nucleotide position.
- the nucleotide positions where the ASOs bind, in addition to important splicing signals, are annotated in the plot.
- FIG. 5 hnRNPAI cooperates with TSL-3-15 to amplify inhibitory effects at the 3’ss of F8 exon 16.
- A Representative Western blot and agarose gel depicting results from the hnRNPAI - ASO competition assay. Each condition tested in the assay is annotated as shown in the matrix above the gel. Epitopes targeted by specific antibodies in the Western blots are indicated to the left of their respective blots. Expected mRNA isoforms including or excluding the test exon are also annotated to the left of the agarose gel.
- B A plot quantifying the results from (A) using the PSI ratio.
- FIG. 6 A combination of ASOs targeting TSL-3-15 can reverse aberrant splicing for a broad array of Hemophilia A associated variants of exon 16 by increasing 3’ss accessibility and blocking hnRNPAI binding.
- A A UCSC Genome Browser screenshot depicting the F8 exon 16 locus and the positions of HA-causing mutations tested in this study. The 3’ and 5’ splice sites are annotated in addition to TSL-3-15.
- Successful ASOs targeting TSL-3-15 are depicted using the same color scheme as previously shown in Fig. 3B.
- B A normalized SHAPE reactivity vs.
- arc diagram plot comparing WT exon 16 to multiple HA-causing mutations that induce aberrant splicing of exon 16.
- the plot uses SHAPE data for each sequence context (i.e., WT or MT) that is plotted on the top portion of the plot, where each bar indicates the normalized SHAPE reactivity at a nucleotide position for a given sequence context.
- the bottom portion of the plot depicts RNA structure predictions for each sequence context using their respective normalized SHAPE reactivity as a folding constraint.
- Each arc represents a base pairing interaction between the respective nucleotides involved within the sequence to form a given RNA structure.
- RNA structures unique to the WT or a specific MT are depicted by their respective color annotations.
- RNA structures that are shared and found in both WT or MT sequence contexts are annotated black.
- a schematic model of exon 16 and its flanking introns are shown at the bottom of the plot to illustrate relative positions of SHAPE data.
- TSL-3-15 is specifically annotated in yellow to illustrate its position.
- C A representative agarose gel depicting the results from the in vivo splicing assays testing the trio ASO combinations’ ability to reverse aberrant splicing of exon 16 induced by other HA-causing mutations.
- Each splicing assay condition included in this specific assay is annotated as shown in the matrix above the gel.
- Expected mRNA isoforms including or excluding the test exon are also annotated to the left of the agarose gel.
- FIG. 7 The loss of a critical ESE in F8 exon 16 presumably amplifies the inhibitory nature of TSL-3-15 to alter exon definition and splicing fidelity.
- a functional ESE recruits a positive splicing factor that controls the structure-function mechanism comprising TSL-3-15 and hnRNPAI at the 3’ss of exon 16. Doing so appears regulates the inhibitory effects of TSL-3-15 and hnRNPAI cooperation, increasing the accessibility of the 3’ss to the splicing machinery.
- (B) A schematic depicting the loss of the ESE in exon 16 due to the A333G mutation. Losing the ESE diminishes the ability to regulate TSL-3-15 and hnRNPAI cooperation at the 3’ss exon 16, leading to decreased accessibility of the 3’ss.
- (C) A schematic depicting the trio ASO combinations’ ability to reverse A333G-induced aberrant splicing of exon 16 by destabilizing TSL-3-15, and preventing the recruitment of hnRNPAI to the 3’ss. Collectively, the data-supported model indicates that the trio ASOs block the recruitment of a negative splicing factor and to increase the accessibility of the 3’ss to the splicing machinery.
- TSL-3-15 is annotated by a simplified depiction of an RNA secondary structure at the 3’ss of exon 16.
- RBPs binding to TSL-3-15 and this region such as hnRNPAI and U2AF are respectively annotated.
- the predicted ESE is annotated within exon 16, and its binding partner, presumably an RBP like SR proteins that are known to enhance splicing, is depicted as well.
- the loss of the ESE by the A333G mutation is annotated within exon 16.
- FIG. 8 mRNA isoform levels quantified by a two-step end-labeled RT-PCR assay and capillary electrophoresis indicating that the majority of HA-causing mutations failed to induce exon skipping in a heterologous reporter context.
- FIG. 9 mRNA isoform levels quantified by a two-step end-labeled RT-PCR assay and capillary electrophoresis indicating the effects of mutations in exon 7, 11 , 16 and 18 on exon inclusion.
- FIG. 10 SHAPE probing data of HA mutants of exon 16.
- FIG. 11 In vivo splicing assay data for a duo combination of ASOs.
- FIG. 12 Data demonstrating that a single ASO such as 91 -108 is capable of increasing the SHAPE activities for nucleotides comprising the poly-Y tract.
- aspects of the present disclosure include splice-modulating antisense oligonucleotides (ASOs).
- the ASOs target a terminal stem loop structure at the 3’ end of intron 15 (TSL-3-15) of a Factor VIII (F8) pre-mRNA.
- TSL-3-15 intron 15
- F8 Factor VIII
- Such ASOs are based on the inventors’ surprising discovery of the TSL-3-15 via RNA chemical probing, and of ASOs that target the TSL- 3-15 and rescue aberrant splicing of Factor VIII exon 16 resulting from point mutations in the gene encoding Factor VIII. Details regarding the ASOs of the present disclosure will now be provided.
- a splice-modulating ASO of the present disclosure hybridizes to the F8 pre-mRNA at all or a portion of positions 1 -18, 37-54, 55-72, 73-90, 91 -108, or 469-486.
- such an ASO may hybridize to 10 or more, 1 1 or more, 12 or more, 13 or more, 14 or more, 15 or more, 16 or more, 17, or each of the contiguous nucleotides at positions 1 -18, 37- 54, 55-72, 73-90, 91 -108, or 469-486.
- a splice-modulating ASO of the present disclosure hybridizes to the F8 pre-mRNA at all or a portion of positions 55-72, 73-90, or 91-108.
- such an ASO hybridizes to 10 or more, 1 1 or more, 12 or more, 13 or more, 14 or more, 15 or more, 16 or more, 17, or each of the contiguous nucleotides at positions 55-72, 73- 90, or 91 -108.
- ASOs are typically small (-15-30 nucleotides), synthetic, single-stranded nucleic acid polymers.
- the ASOs comprise nucleotide modifications. Such modifications may impart useful properties, e.g. increase the biological stability of the ASOs (e.g. nuclease resistance), enhance target binding, increase tissue uptake and/or increase the physical stability of the duplex formed between the ASOs and target nucleic acids, etc..
- the ASO induces steric block of a target sequence, and in such a way that it does not induce target cleavage via RNase H recruitment.
- an ASO may comprise a chemistry which does not support RNase H cleavage (e.g., does not generate consecutive runs of DNA or DNA-like bases).
- an ASO may comprise a “mixmer” pattern in which the ASO may comprise two or more different nucleic acid chemistries, but runs of more than 2 or 3 DNA or DNA-like bases (which would support RNase H-mediated cleavage) are avoided.
- the ASO of the present disclosure may comprise DNA, RNA, and/or nucleotide analogues.
- the nucleotide analogues may be peptide nucleic acid (PNA), FANA, DANA, LNA, and other branched nucleic acids (ENA, cEt), phosphorodiamidate morpholino oligomer (PMO), and/or tricyclo DNA.
- the ASO comprises an abasic site, i.e., the absence of a purine (adenine and guanine) or a pyrimidine (thymine, uracil and cytosine) nucleobase.
- the ASO comprises a 3' to 5' phosphodiester (PO) linkage as naturally found in DNA or RNA.
- the ASO may comprise a modified internucleoside linkage, e.g. a phosphotriester linkage, a phosphorothioate (PS) linkage, a boranophosphate linkage, a phosphorodiamidate linkage, a phosphoamidate linkage, and/or a thiopho sphoramidate linkage.
- the modified internucleoside linkage may be other modifications known in the art.
- the ASO comprises one or more asymmetric centers and thus give rise to enantiomers, diasteromers, and other stereoisomeric configurations, e.g. R, S.
- stereochemistry may be constrained at one or more modified internucleoside linkages.
- the oligonucleotide may comprise repeated left-left-right (or SSR) chiral PS centers.
- the ASO comprises a sugar moiety as found in naturally occurring RNA (e.g., a ribofuranosyl) or a sugar moiety as found in naturally occurring DNA (e.g., a deoxyribofuranosyl).
- the ASO may comprise a modified sugar moiety, i.e. a substituted sugar moiety or a sugar surrogate.
- Substituted sugar moieties include furanosyls comprising substituents at the 2'-position , the 3'-position, the 5 '-position and/or the 4'-position.
- a substituted sugar moiety may be a bicyclic sugar moiety (BNA).
- Sugar surrogates include morpholino, cyclohexeynl and cyclohexitol.
- the modified sugar moiety may comprise a 2'-O-methyl, 2'-O-methoxyethyl (2'-O-MOE), 2'-O-aminopropyl, 2'-deoxy, 2'-O- propyl (2'-O-AP), 2'-O-dimethylaminoethyl (2'O-DMAOE), 2'-O- dimethylaminopropyl (2'-O-DMAP), 2'-O-dimethylaminoethyloxyethyl (2'O-DMAEOE), or 2'0-N- methylacetoamido (2'0-NMA) modification or a locked or bridged ribose conformation (e.g., LNA, cEt or ENA).
- the modified sugar moiety may comprise other modifications known in the art.
- the ASO comprises a terminal modification at its 5' and/or 3' end, such as a vinyl phosphonate, and/or inverted terminal bases.
- the ASO comprises a nucleobase as found in naturally occurring RNA and DNA (i.e. adenine (A), thymine (T), uracil (U), guanine (G), cytosine (C), inosine (I), and 5-methyl C).
- the oligonucleotide may comprise a modified nucleobase, e.g. 5- hyrdoxymethylcytosine, 5-formylcytosine, and 5-carboxycytosine. The inclusion of 5'methylcytosine may enhance base pairing by modifying the hydrophobic nature of the oligonucleotide.
- each of the sugar moieties in the ASO may comprise a 2'-O-methoxyethyl (2'MOE) modification and each of the internucleoside linkages may be a phosphorothioate (i.e. a fully PS-MOE oligonucleotide).
- PS modifications are known to result in resistance to a broad spectrum of nucleases and increase protein binding, which also improves tissue uptake.
- 2'MOE modifications are known to enable enhanced binding affinity to the target mRNA with minimal toxicity and reduce plasma protein binding.
- the ASO is produced using chemical synthesis and/or enzymatic ligation reactions using procedures known in the art.
- An ASO may be produced biologically using an expression vector into which the oligonucleotide is sub-cloned in an antisense orientation, e.g., RNA transcribed from the inserted oligonucleotide will be of an antisense orientation to the target nucleic acid of interest.
- compositions comprising any of the ASOs of the present disclosure.
- a composition of the present disclosure includes the ASO present in a liquid medium.
- the liquid medium may be an aqueous liquid medium, such as water, a buffered solution, or the like.
- One or more additives such as a salt (e.g., NaCI, MgCI 2 , KOI, MgSO 4 ), a buffering agent (a Tris buffer, N-(2-Hydroxyethyl)piperazine-N'-(2-ethanesulfonic acid) (HEPES), 2-(N-Morpholino)ethanesulfonic acid (MES), 2-(N-Morpholino)ethanesulfonic acid sodium salt (MES), 3-(N-Morpholino)propanesulfonic acid (MOPS), N-tris[Hydroxymethyl]methyl- 3-aminopropanesulfonic acid (TAPS), etc.), a solubilizing agent, a detergent (e.g., a non-
- a pharmaceutical composition of the present disclosure includes an ASO of the present disclosure, and a pharmaceutically acceptable carrier.
- the ASO can be incorporated into a variety of formulations for therapeutic administration. More particularly, the ASO can be formulated into pharmaceutical compositions by combination with appropriate, pharmaceutically acceptable excipients or diluents, and may be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, injections, inhalants and aerosols.
- Formulations of the ASOs for administration to an individual are generally sterile and may further be free of detectable pyrogens or other contaminants contraindicated for administration to a patient according to a selected route of administration.
- the ASOs can be administered in the form of their pharmaceutically acceptable salts, or they may also be used alone or in appropriate association, as well as in combination, with other pharmaceutically active compounds.
- the following methods and carriers/excipients are merely examples and are in no way limiting.
- the ASOs can be used alone or in combination with appropriate additives to make tablets, powders, granules or capsules, for example, with conventional additives, such as lactose, mannitol, corn starch or potato starch; with binders, such as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins; with disintegrators, such as corn starch, potato starch or sodium carboxymethylcellulose; with lubricants, such as talc or magnesium stearate; and if desired, with diluents, buffering agents, moistening agents, preservatives and flavoring agents.
- conventional additives such as lactose, mannitol, corn starch or potato starch
- binders such as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins
- disintegrators such as corn starch, potato starch or sodium carboxymethylcellulose
- lubricants such as talc or magnesium stearate
- the ASOs can be formulated for parenteral (e.g., intravenous, subcutaneous, intraarterial, intraosseous, intramuscular, intracerebral, intracerebroventricular, intrathecal, etc.) administration.
- parenteral e.g., intravenous, subcutaneous, intraarterial, intraosseous, intramuscular, intracerebral, intracerebroventricular, intrathecal, etc.
- the ASOs are formulated for injection by dissolving, suspending or emulsifying the ASOs in an aqueous or non-aqueous solvent, such as vegetable or other similar oils, synthetic aliphatic acid glycerides, esters of higher aliphatic acids or propylene glycol; and if desired, with conventional additives such as solubilizers, isotonic agents, suspending agents, emulsifying agents, stabilizers and preservatives.
- an aqueous or non-aqueous solvent such as vegetable or other similar oils, synthetic ali
- compositions that include the ASOs may be prepared by mixing the ASOs having the desired degree of purity with optional physiologically acceptable carriers, excipients, stabilizers, surfactants, buffers and/or tonicity agents.
- Acceptable carriers, excipients and/or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid, glutathione, cysteine, methionine and citric acid; preservatives (such as ethanol, benzyl alcohol, phenol, m-cresol, p-chlor-m-cresol, methyl or propyl parabens, benzalkonium chloride, or combinations thereof); amino acids such as arginine, glycine, ornithine, lysine, histidine, glutamic acid, aspartic acid, isoleucine, leucine, alanine, phenylalanine, tyrosine, try
- the pharmaceutical composition may be in a liquid form, a lyophilized form or a liquid form reconstituted from a lyophilized form, wherein the lyophilized preparation is to be reconstituted with a sterile solution prior to administration.
- the standard procedure for reconstituting a lyophilized composition is to add back a volume of pure water (typically equivalent to the volume removed during lyophilization); however solutions comprising antibacterial agents may be used for the production of pharmaceutical compositions for parenteral administration.
- An aqueous formulation of the ASOs may be prepared in a pH-buffered solution, e.g., at pH ranging from about 4.0 to about 7.0, or from about 5.0 to about 6.0, or alternatively about 5.5.
- buffers that are suitable for a pH within this range include phosphate-, histidine-, citrate-, succinate-, acetate-buffers and other organic acid buffers.
- the buffer concentration can be from about 1 mM to about 100 mM, or from about 5 mM to about 50 mM, depending, e.g., on the buffer and the desired tonicity of the formulation.
- a tonicity agent may be included to modulate the tonicity of the formulation.
- Example tonicity agents include sodium chloride, potassium chloride, glycerin and any component from the group of amino acids, sugars as well as combinations thereof.
- the aqueous formulation is isotonic, although hypertonic or hypotonic solutions may be suitable.
- the term "isotonic" denotes a solution having the same tonicity as some other solution with which it is compared, such as physiological salt solution or serum.
- Tonicity agents may be used in an amount of about 5 mM to about 350 mM, e.g., in an amount of 100 mM to 350 mM.
- a surfactant may also be added to the formulation to reduce aggregation and/or minimize the formation of particulates in the formulation and/or reduce adsorption.
- Example surfactants include polyoxyethylensorbitan fatty acid esters (Tween), polyoxyethylene alkyl ethers (Brij), alkylphenylpolyoxyethylene ethers (Triton-X), polyoxyethylene-polyoxypropylene copolymer (Poloxamer, Pluronic), and sodium dodecyl sulfate (SDS).
- suitable polyoxyethylenesorbitan-fatty acid esters are polysorbate 20, (sold under the trademark Tween 20TM) and polysorbate 80 (sold under the trademark Tween 80TM).
- Suitable polyethylene-polypropylene copolymers are those sold under the names Pluronic® F68 or Poloxamer 188TM.
- suitable Polyoxyethylene alkyl ethers are those sold under the trademark BrijTM.
- Example concentrations of surfactant may range from about 0.001% to about 1% w/v.
- a lyoprotectant may also be added in order to protect the ASO against destabilizing conditions during a lyophilization process.
- known lyoprotectants include sugars (including glucose and sucrose); polyols (including mannitol, sorbitol and glycerol); and amino acids (including alanine, glycine and glutamic acid). Lyoprotectants can be included, e.g., in an amount of about 10 mM to 500 nM.
- the pharmaceutical composition includes the ASO, and one or more of the above-identified components (e.g. , a surfactant, a buffer, a stabilizer, a tonicity agent) and is essentially free of one or more preservatives, such as ethanol, benzyl alcohol, phenol, m- cresol, p-chlor-m-cresol, methyl or propyl parabens, benzalkonium chloride, and combinations thereof.
- a preservative is included in the formulation, e.g., at concentrations ranging from about 0.001 to about 2% (w/v).
- aspects of the present disclosure further include methods of treating Hemophilia A (HA) in a subject in need thereof, e.g., a subject who exhibits aberrant splicing of Factor VIII (F8) exon 16.
- the methods comprise administering to the subject a therapeutically effective amount of a composition of the present disclosure, e.g., a composition formulated for administration to a subject comprising any of the ASOs or desired combinations thereof of the present disclosure.
- the ASOs of the present disclosure may be administered via any suitable route of administration.
- an ASO or combination thereof of the present disclosure is administered to the subject via parenteral administration.
- parenteral routes of administration that find use in practicing the methods of the present disclosure include intravenous (IV) infusion and subcutaneous (SC) injection.
- the ASOs of the present disclosure may be administered in a composition in a therapeutically effective amount.
- therapeutically effective amount is meant a dosage sufficient to produce a desired result, e.g., an amount sufficient to effect beneficial or desired therapeutic (including preventative) results, such as a reduction in a symptom of HA, as compared to a control.
- the therapeutically effective amount is sufficient to reduce the clotting time of the subject, which in some instances may be assessed via a Prothrombin time (PT) test.
- PT Prothrombin time
- An effective amount can be administered in one or more administrations.
- aspects of the present disclosure include methods for treating HA in the subject.
- treatment is meant at least an amelioration of one or more symptoms associated with the HA of the subject, where amelioration is used in a broad sense to refer to at least a reduction in the magnitude of a parameter, e.g. symptom, associated with the HA being treated.
- amelioration also includes situations where the HA, or at least one or more symptoms associated therewith, are completely inhibited, e.g., prevented from happening, or stopped, e.g., terminated, such that the individual no longer suffers from the HA, or at least the symptoms that characterize the HA.
- kits find use in practicing the methods of the present disclosure, e.g., methods of treating Hemophilia A (HA) in a subject in need thereof.
- HA Hemophilia A
- kits of the present disclosure comprises any of the ASOs or desirable combinations thereof of the present disclosure (e.g., present in a composition formulated for administration to a subject, such as any of the compositions of the present disclosure), and instructions for administering the ASO or combination thereof to the subject.
- the kits of the present disclosure may include any of the ASOs having any of the features described above in the section relating to the Splice-Modulating Antisense Oligonucleotides of the present disclosure, which are not reiterated herein for purposes of brevity.
- kits of the present disclosure may include a quantity of the ASO or combination of ASOs, present in unit dosages, e.g., ampoules, or a multi-dosage format.
- the kits may include one or more (e.g., two or more) unit dosages (e.g., ampoules) of an ASO or combination of ASOs of the present disclosure.
- unit dosage refers to physically discrete units suitable as unitary dosages for human and animal subjects, each unit containing a predetermined quantity of the ASO or combination of ASOs calculated in an amount sufficient to produce the desired effect.
- kits may include a single multi dosage amount of the ASO or combination of ASOs.
- the instructions (e.g., instructions for use (I FU)) included in the kits may be recorded on a suitable recording medium.
- the instructions may be printed on a substrate, such as paper or plastic, etc.
- the instructions may be present in the kits as a package insert, in the labeling of the container of the kit or components thereof (i.e., associated with the packaging or sub-packaging) etc.
- the instructions are present as an electronic storage data file present on a suitable computer readable storage medium, e.g., portable flash drive, etc.
- the actual instructions are not present in the kit, but means for obtaining the instructions from a remote source (e.g., via the internet) are provided.
- An example of this embodiment is a kit that includes a web address where the instructions can be viewed and/or from which the instructions can be downloaded.
- the means for obtaining the instructions is recorded on a suitable substrate.
- the present disclosure also provides ASO interference mapping (AIM) methods of identifying a cis region that affects splicing of an exon of interest.
- the methods comprise testing tiled or non-overlapping ASOs of known sequence that span the exon of interest and its flanking introns or portions thereof in a splicing reporter assay.
- the splicing reporter assay reports splicing of the exon of interest, and an ASO that affects splicing of the exon of interest identifies the target sequence of the ASO as a sequence comprising a cis region that affects splicing of the exon of interest.
- the methods comprise testing tiled or non-overlapping ASOs of known sequence that span the exon of interest and its flanking introns or portions thereof in a splicing reporter assay.
- the splicing reporter assay reports splicing of the exon of interest, and wherein an ASO that affects splicing of the exon of interest identifies the ASO as an ASO that affects splicing of an exon of interest.
- the splicing reporter assay comprises co-transfecting an individual ASO and a splicing reporter comprising the exon of interest and its flanking introns or portions thereof into cells present in a confinement, such that each confinement corresponds to an ASO that targets a specific position of the exon of interest or its flanking introns or portions thereof; and qualitatively or quantitively assaying for splicing of the exon of interest.
- the splicing reporter assay comprises co-transfecting an individual ASO and a splicing reporter comprising the exon of interest and its flanking introns or portions thereof into cells present in a confinement (e.g., a tube, well, or the like), such that each confinement corresponds to an ASO that targets a specific position of the exon of interest or its flanking introns or portions thereof; and qualitatively or quantitively assaying for splicing of the exon of interest.
- a confinement e.g., a tube, well, or the like
- the exon of interest and its flanking introns or portions thereof are the wild-type exon of interest and its flanking introns or portions thereof. In other embodiments, the exon of interest and its flanking introns or portions thereof comprise a mutation. The mutation
- compositions comprising such ASOs, including compositions formulated for administration to a subject in need thereof or also provided, as are methods comprising administering such compositions to an individual in need thereof, e.g., where the ASO rescues a defect in splicing of the exon of interest in the subject.
- a splice-modulating antisense oligonucleotide that targets a terminal stem loop structure at the 3’ end of intron 15 (TSL-3-15) of a Factor VIII (F8) pre-mRNA.
- composition comprising the splice-modulating ASO of any one of clauses 1 to 9.
- composition of clause 10 comprising one, two, or each of: a splice-modulating ASO that hybridizes to the F8 pre-mRNA at all or a portion of positions 55-72; a splice-modulating ASO that hybridizes to the F8 pre-mRNA at all or a portion of positions 73-90; and a splice-modulating ASO that hybridizes to the F8 pre-mRNA at all or a portion of positions 91 -108.
- composition of clause 10 or clause 1 1 formulated for administration to a subject.
- composition of clause 13 formulated for intravenous administration to the subject.
- a method of treating Hemophilia A (HA) in a subject in need thereof, wherein the subject exhibits aberrant splicing of Factor VIII (F8) exon 16 the method comprising administering to the subject a therapeutically effective amount of the composition of any one of clauses 12 to 14.
- An ASO interference mapping (AIM) method of identifying a cis region that affects splicing of an exon of interest comprising: testing tiled or non-overlapping ASOs of known sequence that span the exon of interest and its flanking introns or portions thereof in a splicing reporter assay, wherein the splicing reporter assay reports splicing of the exon of interest, and wherein an ASO that affects splicing of the exon of interest identifies the target sequence of the ASO as a sequence comprising a cis region that affects splicing of the exon of interest.
- AIM ASO interference mapping
- the splicing reporter assay comprises: co-transfecting an individual ASO and a splicing reporter comprising the exon of interest and its flanking introns or portions thereof into cells present in a confinement, such that each confinement corresponds to an ASO that targets a specific position of the exon of interest or its flanking introns or portions thereof; and qualitatively or quantitively assaying for splicing of the exon of interest.
- a method of identifying an ASO that affects splicing of an exon of interest comprising: testing tiled or non-overlapping ASOs of known sequence that span the exon of interest and its flanking introns or portions thereof in a splicing reporter assay, wherein the splicing reporter assay reports splicing of the exon of interest, and wherein an ASO that affects splicing of the exon of interest identifies the ASO as an ASO that affects splicing of an exon of interest.
- the splicing reporter assay comprises: co-transfecting an individual ASO and a splicing reporter comprising the exon of interest and its flanking introns or portions thereof into cells present in a confinement, such that each confinement corresponds to an ASO that targets a specific position of the exon of interest or its flanking introns or portions thereof; and qualitatively or quantitively assaying for splicing of the exon of interest.
- composition comprising the splice-modulating ASO of clause 29 or clause 30.
- composition of clause 31 formulated for administration to a subject.
- composition of clause 32 formulated for parenteral administration to the subject.
- composition of clause 33 formulated for intravenous administration to the subject.
- mRNA isoform levels are quantified by a two-step end-labeled RT-PCR assay and capillary electrophoresis.
- Fig. 8 mutations in exon 7, 11 , 16, and 18 had striking effects on exon inclusion, indicating that these exons may be susceptible to aberrant splicing.
- Fig. 1 C shows splicing assays for 16 HA-causing variants of exon 16.
- Exon 16 encodes a large portion of the A3 domain which is required for efficient blood clotting (32).
- Clinical data repositories indicate that the coding sequence for the A3 domain is frequently disturbed by missense mutations that are linked to HA (33, 34).
- Our exon 16 splicing assays support the notion that aberrant splicing is a contributing etiology of HA as a result of skipping important coding sequences from F8 mRNA.
- there are multiple HA-causing point mutations that can readily impact inclusion of exon 16, indicating that exon 16 is fragile and susceptible to aberrant splicing.
- we identify that the A333G mutation induces the highest degree of exon 16 skipping, seeing a significant 5.23-fold decrease in its splicing relative to the WT.
- ASO interference mapping ASO interference mapping
- each individual ASO is co-transfected with the WT exon 16 splicing reporter into each well of a 96-well tissue culture plate culturing HEK293T cells. Effectively, each well corresponds to an ASO that targets a specific position of exon 16 or its flanking introns.
- AIM analysis on WT exon 16 indicates that all ASOs targeting the exon, except for a few, strongly inhibited its splicing (Fig. 2C). By contrast, individual ASOs targeting the flanking introns had little impact on exon 16 splicing relative to our controls.
- a couple of ASOs that target the intronic region downstream of the 5’ss, 469-486 and 505-522, are indicated to be statistically significant in enhancing splicing relative to the WT control with no ASO.
- ASOs that target the flanking intronic sequences upstream of the 3’ss appear to modulate splicing positively or negatively in a subtle, non-statistically significant manner.
- ASOs 1 -18, 37-54, 55-72, and 91 -108 target regions upstream and adjacent to the 3'ss, with 37-54 exhibiting the largest rescue effect by increasing splicing by 2.015-folds (P-value ⁇ 0.0001 ).
- our AIM data indicates that targeting regions upstream or downstream of F8 exon 16 with ASOs may rescue splicing of splicing-sensitive mutants by perturbing the influence of inhibitory elements found in the flanking introns.
- RNA structures in F8 exon 16 we performed selective 2’-hydroxyl acylation analyzed by primer extension and mutational profiling coupled to high-throughput sequencing (SHAPE-MaP-seq) on in vitro transcribed RNA. All in vitro RNA corresponding to WT exon 16 or its HA mutants contain the same sequence context as we tested in our splicing reporter assays.
- SHAPE-MaP-seq high-throughput sequencing
- RNA structure profiles for several splicing-sensitive variants of F8 exon 16 that we discovered, comparing each of their 2A3 reactivities and SHAPE-driven folding predictions to WT exon 16.
- SHAPE probing of this mutant shows that it primarily induces local rearrangements to RNA structures nearby, as well as in creating some long-range base-pairing interactions that involve the flanking introns (Fig. 3A).
- Fig. 3A Surprisingly, when comparing the entire structure profile to the WT, most of the RNA structures detected in the A333G mutant appear unchanged.
- TSL-3-15 may contain any functional binding sites for RBPs
- RBPmap to identify RBP consensus motifs within the structure (36).
- RBPmap recognized two hnRNPAI binding motifs within TSL-3-15 (Fig. 3B, binding sites are highlighted in dark red bubbles with white text).
- the first predicted motif is found at nucleotide positions 84-90 (UUAGGGA), and the second predicted motif is found at nucleotide positions 99-105 (CUAAGGA).
- these predicted hnRNPAI binding sites are ISS-15-1 and ISS-15-2, respectively. Based on published research, these predicted binding sites contain a motif that contains or highly resembles the hnRNPAI consensus motif, UAGG (37, 38). These predicted hnRNPAI binding sites are positioned within the three-way junction of TSL-3-15. Intriguingly, ASOs 73-90 and 91 -108 directly binds ISS-15-1 and ISS-15-2, respectively. These ASOs, when used individually, improved splicing of the A333G mutant.
- Example 4 Combinations of ASOs additively enhance exon 16 splicing in the A333G Mutant By Destabilizing TSL-3-15
- the data supports our structure-function hypothesis and illuminates a mechanism of action for our ASOs where the reversal of aberrant splicing is achieved by destabilizing TSL-3-15 to increase the accessibility of the poly-Y tract to U2AF.
- ASOs and a splicing reporter were co-transfected with either an empty expression vector, or an hnRNPAI expression vector.
- Conditions with the hnRNPAI expression vector should lead to the overexpression of hnRNPAI , which hypothetically will inhibit splicing of exon 16. If splicing inhibition directed by hnRNPAI is attenuated with ASOs that target the predicted silencers ISS-15-1 and ISS-15-2, this would therefore indicate that hnRNPAI indeed interacts with TSL-3-15.
- WT wild-type F8 exons
- PCR products were ligated into pACT7_SC14 (HBB minigene reporter from Lynch Lab) using homology-based cloning technology (In-Fusion HD Cloning kit from Takara Bio).
- MT Mutant splicing reporter constructs were then sequence-validated using Sanger sequencing to confirm successful cloning and identity of splicing reporters.
- each F8 point mutation and therefore each MT F8 exon presented in this study, is based on the nucleotide being mutated (e g., A>C), and its position within the sequence context tested (i.e., length of flanking introns included and size of exon tested).
- HEK293T cells (ATCC) were cultured in 6-well tissue culture plates (CytoOne, USA Scientific) using Dulbecco’s Modified Eagle Medium (Gibco, supplemented with 10% FBS) at 37°C, 5% CO2. The cells were transiently transfected at -60-80% confluency with 2.5ug of each F8 splicing reporter using Lipofectamine 2000 (Invitrogen). Total RNA was harvested from cells 24-hours post-transfection using the Direct-zol RNA Miniprep kits (Zymo Research). Each in vivo splicing assay was performed a minimum of three times.
- ASOs complementary to F8 exon 16 and flanking introns were designed from the reverse complement of the F8 sense sequence, creating non-overlapping 18-mers.
- F8 exon 16 ASOs were designed to contiguously tile across the exon and its flanking introns.
- ASOs were synthesized by Integrated DNA Technologies (IDT). Each ASO is designated by their complementary positions in the F8 exon 16 reporter.
- HEK293T cells ATCC were cultured in 96-well tissue culture plates (Perkin Elmer) as described above.
- HEK293T cells (ATCC) were cultured in 6-well tissue culture plates as described above. Cells were co-transfected with 1250ng of the WT splicing reporter, 1250ng of either an empty expression vector or a T7-tagged hnRNPAI expression vector, and 50pmol of ASO(s) as described above. Total RNA and protein were isolated 24-hours post transfection using a RSB lysis buffer (10mM Tris pH 7.0, 100mM NaCI, 5mM MgCI2, 0.5% NP40, 0.5% Triton X-100, and EDTA-free Protease Inhibitor Cocktail (Roche)).
- RSB lysis buffer 10mM Tris pH 7.0, 100mM NaCI, 5mM MgCI2, 0.5% NP40, 0.5% Triton X-100, and EDTA-free Protease Inhibitor Cocktail (Roche)).
- the cell lysate was then centrifuged at 10,000 x g for 10 minutes at 4°C. The supernatant was then collected and aliquoted for two separate applications. The first aliquot, comprising -90% of the cell lysate, was prepared for total RNA purification using the Direct-zol RNA Miniprep kits from Zymo Research. The remaining -10% of the cell lysate was then homogenized into a denaturing buffer solution containing 4X NuPAGETM LDS Sample Buffer in preparation for polyacrylamide gel electrophoresis (InvitrogenTM NuPAGETM, 4 to 12%, Bis-Tris, 1.0-1.5 mm, Mini Protein Gels), and subsequent Western blots.
- 4X NuPAGETM LDS Sample Buffer in preparation for polyacrylamide gel electrophoresis (InvitrogenTM NuPAGETM, 4 to 12%, Bis-Tris, 1.0-1.5 mm, Mini Protein Gels), and subsequent Western blots.
- RNA was then used as input for all first-strand cDNA synthesis using Multiscribe Reverse Transcriptase (Applied Biosystems).
- the resulting cDNA was then used as a template for endpoint PCR amplification using specific primers that detect our mRNA splicing reporter isoforms.
- the forward primer of the pair contains a 5’FAM modification.
- the resulting amplicons were then analyzed using agarose gel electrophoresis to empirically evaluate mRNA isoforms detected. The abundance of each 5’FAM labeled mRNA isoform is quantified using capillary electrophoresis and fragment analysis (UC Berkeley, DNA Sequencing Center).
- each sample is suspended in a formamide solution that contains a proper size standard for sizing detected fragments (GeneScan 1200 Liz, Applied Biosystems). Analysis was performed in PeakScanner (Thermofisher). Calculating Splicing Efficiency using Percent-Spliced-ln (PSI) Index Formula
- the mean PSI for a given reporter context is then calculated using all its respective replicates for a corresponding experiment.
- Statistical significance in the differences between the mean PSI of the control group(s) vs the experimental group(s) is determined using analysis of variance (ANOVA), and Dunett’s post-hoc test. All statistical tests for PSI analysis were done in GraphPad Prism 9. Values are determined to be statistically significant if calculated the P-value is below an alpha value of ⁇ 0.05.
- RNA was purified by denaturing PAGE and eluted from gel slices overnight in 10 mM Tris pH 7.5, 480 mM sodium acetate, 1 mM EDTA, 0.1 % SDS. Following ethanol precipitation transcripts were resuspended in ddH2O and quantified by spectrophotometry.
- F8 exon 16 in vitro transcribed pre-mRNA sequences were first denatured by incubating at 95°C for 3 minutes in 65mM Na-HEPES (pH 8.0). The denatured RNA was then allowed to slowly cool to room temperature (RT) for 15 minutes, after which MgCI2 was supplemented to 1 mM for a total volume of 15pL and incubated at RT for an additional 5 minutes.
- RT room temperature
- MgCI2 was supplemented to 1 mM for a total volume of 15pL and incubated at RT for an additional 5 minutes.
- 2-aminopyridine-3-carboxylic acid imidazolide (2A3) was added to a final concentration of 100mM and incubated for 2 minutes at 37°C.
- RNA Clean & Concentrator- 5 from Zymo Research.
- RNAs were fragmented to a median size of 200nt by incubation at 94°C for 1 minute using NEBNext® Magnesium RNA Fragmentation Kit and then purified using NEB’s recommended ethanol precipitation protocol. Purified RNA was then prepared for reverse transcription, incubating the RNA with 1 pL of 10mM dNTPs and 2pL of 20pM random hexamers at 70°C for 5 minutes, followed by immediate transfer to ice.
- Reverse transcription reactions were then supplemented with 4 L of 5X RT buffer (250mM Tris-HCI pH 8.3, 375mM KCI), 2pL of 0.1 M DTT, 1 L of 120 mM MnCI2, 10 U of SUPERase RNase Inhibitor, and 200 U of Superscript II Reverse Transcriptase (SSII) [ThermoFisher Scientific, cat. 18064014] to a final volume of 20 L. These reactions were then incubated at 25°C for 10 minutes to allow for partial primer extension, followed by incubation at 42°C for 3 hours to enable efficient extension. SSII was then heat- inactivated by incubation at 75°C for 20 minutes.
- 5X RT buffer 250mM Tris-HCI pH 8.3, 375mM KCI
- 2pL of 0.1 M DTT 1 L of 120 mM MnCI2
- 10 U of SUPERase RNase Inhibitor 10 U
- SSII Superscript II Reverse Transcripta
- Reverse transcription reactions were then supplemented with EDTA to a final concentration of 6mM to chelate Mn2+ ions and incubated at RT for 5 minutes. MgCI 2 was then added to a final concentration of 6mM for each reaction. Reverse transcription reactions were then used as input for NEBNext® UltraTM II DNA library Prep Kit for Illumina® (New England Biolabs, cat. E7645L), using NEBNext Multiplex Oligos for Illumina® (Unique Dual index UMI Adaptors DNA Set 1 , cat. E7395). Subsequent reactions were performed following manufacturer instructions.
- Reads produced from Illumina libraries were pre-processed and mapped using the rf-map module (parameters: -b2 -mp “-no-mixed -no-discordant” -bs) ensuring only paired-end mates with expected mate orientation were considered with Bowtie2.
- the mutational signal was obtained using the rf-count module (parameters: -m -pp -nd -ni) enabling mutation counts of reads produced from properly paired mates.
- Mutational signal was normalized relative to an unmodified control using parameters (-sm 3 -nm 1 -mu 0.05), based on a scoring method from Siegfried et al., 2014 and further normalized using the 2-8% normalization approach provided by RNA Framework. Normalized reactivities were then supplied to RNAstructure to generate data-driven predicted structure models.
- Ovalbumin gene evidence for a leader sequence in mRNA and DNA sequences at the exon-intron boundaries. Proc. Natl. Acad. Sci. U. S. A., 75, 4853-4857.
- RBPmap a web server for mapping binding sites of RNA-binding proteins. Nucleic Acids Res., 42, W361-7.
- HNRNPA1 promotes recognition of splice site decoys by U2AF2 in vivo. Genome Res., 28, 689-698.
- Hotspot exons are common targets of splicing perturbations. Nat. Commun., 12, 2756.
- RNA folding affects the recruitment of SR proteins by mouse and human polypurinic enhancer elements in the fibronectin EDA exon. Mol. Cell. Biol., 24, 1387-1400.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Engineering & Computer Science (AREA)
- Genetics & Genomics (AREA)
- Organic Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Molecular Biology (AREA)
- Medicinal Chemistry (AREA)
- Public Health (AREA)
- Pharmacology & Pharmacy (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Veterinary Medicine (AREA)
- Biomedical Technology (AREA)
- Zoology (AREA)
- Animal Behavior & Ethology (AREA)
- Biochemistry (AREA)
- Epidemiology (AREA)
- General Engineering & Computer Science (AREA)
- Hematology (AREA)
- Wood Science & Technology (AREA)
- Biotechnology (AREA)
- Biophysics (AREA)
- Plant Pathology (AREA)
- Physics & Mathematics (AREA)
- Microbiology (AREA)
- Dermatology (AREA)
- Toxicology (AREA)
- Gastroenterology & Hepatology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Diabetes (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
Description
Claims
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US202363453036P | 2023-03-17 | 2023-03-17 | |
| US63/453,036 | 2023-03-17 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2024196804A1 true WO2024196804A1 (en) | 2024-09-26 |
Family
ID=92842362
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/US2024/020275 Pending WO2024196804A1 (en) | 2023-03-17 | 2024-03-15 | Factor viii splice-modulating antisense oligonucleotides and methods of use |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2024196804A1 (en) |
Citations (4)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US20190390197A1 (en) * | 2015-10-09 | 2019-12-26 | Wave Life Sciences Ltd. | Oligonucleotide compositions and methods thereof |
| US20210254163A1 (en) * | 2011-07-28 | 2021-08-19 | The Regents Of The University Of California | Exonic splicing enhancers and exonic splicing silencers |
| US20230045557A1 (en) * | 2019-12-19 | 2023-02-09 | Nippon Shinyaku Co., Ltd. | Antisense nucleic acid enabling exon skipping |
| WO2023012481A2 (en) * | 2021-08-04 | 2023-02-09 | Oxford University Innovation Limited | Method |
-
2024
- 2024-03-15 WO PCT/US2024/020275 patent/WO2024196804A1/en active Pending
Patent Citations (4)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US20210254163A1 (en) * | 2011-07-28 | 2021-08-19 | The Regents Of The University Of California | Exonic splicing enhancers and exonic splicing silencers |
| US20190390197A1 (en) * | 2015-10-09 | 2019-12-26 | Wave Life Sciences Ltd. | Oligonucleotide compositions and methods thereof |
| US20230045557A1 (en) * | 2019-12-19 | 2023-02-09 | Nippon Shinyaku Co., Ltd. | Antisense nucleic acid enabling exon skipping |
| WO2023012481A2 (en) * | 2021-08-04 | 2023-02-09 | Oxford University Innovation Limited | Method |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| US20230250439A1 (en) | Polynucleotide secondary structure | |
| US20250270563A1 (en) | Targeted augmentation of nuclear gene output | |
| CN106661580B (en) | Antisense oligonucleotides for treating pompe disease | |
| Qin et al. | Hsa_circ_0001649: a circular RNA and potential novel biomarker for hepatocellular carcinoma | |
| JP2022078069A (en) | Methods and Compositions for Specific Inhibition of α-1 Antitrypsin by Double-stranded RNA | |
| Ibáñez-Cabellos et al. | Acute depletion of telomerase components DKC1 and NOP10 induces oxidative stress and disrupts ribosomal biogenesis via NPM1 and activation of the P53 pathway | |
| EP3083960B1 (en) | Ras exon 2 skipping for cancer treatment | |
| Jalan et al. | Decoding the ‘fifth’nucleotide: impact of RNA pseudouridylation on gene expression and human disease | |
| US20200157546A1 (en) | Antisense oligonucleotides for modulating htra1 expression | |
| WO2024196804A1 (en) | Factor viii splice-modulating antisense oligonucleotides and methods of use | |
| Posey et al. | Target DNA structure plays a critical role in RAG transposition | |
| US20220372475A1 (en) | Inhibitors Of RNA Editing And Uses Thereof | |
| JP2024522148A (en) | Treatment of psoriasis with interferon-induced helicase C domain 1 (IFIH1) inhibitors | |
| CA3219247A1 (en) | Inhibitory nucleic acids for factor h family proteins | |
| CA3207341A1 (en) | Compositions for treatment of conditions and diseases associated with polycystin expression | |
| US20230203493A1 (en) | Biomarkers Related to Oral Squamous Cell Carcinoma and Methods of Diagnosis and Treatment Thereof | |
| EP3256601B1 (en) | Methods for detecting therapeutic nucleic acids | |
| US20180010188A1 (en) | Method for detecting nonsense-mediated rna decay | |
| Tse | RNA Splicing Mechanisms in Rare Disease | |
| WO2025166258A1 (en) | Rna-guided control of protein secretion and membrane protein expression | |
| Tennakoon | Transfer RNA Missense Suppressors in Nature and Disease | |
| WO2025038613A1 (en) | Therapeutic modulation of snora13 for treatment of ribosomopathies | |
| CA3210480A1 (en) | Treatment of inflammation with glucocorticoids and angiopoietin-like 7 (angptl7) inhibitors | |
| WO2024076934A2 (en) | Compositions and methods for modulation of cftr | |
| KR20240099232A (en) | Glaucoma treatment using Rho Guanine Nucleotide Exchange Factor 12 (ARHGEF12) inhibitors |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 24775476 Country of ref document: EP Kind code of ref document: A1 |
|
| WWE | Wipo information: entry into national phase |
Ref document number: 2024775476 Country of ref document: EP |
|
| NENP | Non-entry into the national phase |
Ref country code: DE |
|
| ENP | Entry into the national phase |
Ref document number: 2024775476 Country of ref document: EP Effective date: 20251017 |
|
| ENP | Entry into the national phase |
Ref document number: 2024775476 Country of ref document: EP Effective date: 20251017 |
|
| ENP | Entry into the national phase |
Ref document number: 2024775476 Country of ref document: EP Effective date: 20251017 |
|
| ENP | Entry into the national phase |
Ref document number: 2024775476 Country of ref document: EP Effective date: 20251017 |
|
| ENP | Entry into the national phase |
Ref document number: 2024775476 Country of ref document: EP Effective date: 20251017 |