WO2017205384A1 - Compositions and methods for inhibiting target rna expression - Google Patents

Compositions and methods for inhibiting target rna expression Download PDF

Info

Publication number
WO2017205384A1
WO2017205384A1 PCT/US2017/034019 US2017034019W WO2017205384A1 WO 2017205384 A1 WO2017205384 A1 WO 2017205384A1 US 2017034019 W US2017034019 W US 2017034019W WO 2017205384 A1 WO2017205384 A1 WO 2017205384A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
target rna
target
antisense oligonucleotides
enzyme
Prior art date
Application number
PCT/US2017/034019
Other languages
French (fr)
Inventor
Sudhir Agrawal
Reina IMPROGO
Jiang WAYNE
Original Assignee
Idera Pharmaceuticals, Inc.
Priority date (The priority date 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 date listed.)
Filing date
Publication date
Application filed by Idera Pharmaceuticals, Inc. filed Critical Idera Pharmaceuticals, Inc.
Publication of WO2017205384A1 publication Critical patent/WO2017205384A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-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
    • C12N15/1135Non-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 against oncogenes or tumor suppressor genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-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
    • C12N15/1137Non-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 against enzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • C12N2320/34Allele or polymorphism specific uses

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Biomedical Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • Molecular Biology (AREA)
  • Organic Chemistry (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Microbiology (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Virology (AREA)
  • Oncology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates to third generation antisense (3GA) compounds and the therapeutic and prophylactic use of 3GA compounds to selectively down-regulate the expression of a first allele (e.g., a mutant), even when the first allelic mRNA differs from a second allele (e.g., wild-type) by only a single nucleotide, as is the case with certain mutations, for example, point mutations.

Description

COMPOSITIONS AND METHODS FOR INHIBITING TARGET RNA EXPRESSION
PRIORITY
This application claims priority to, and the benefit of, U.S. Provisional Application No. 62/340,226 filed May 23, 2016, which is hereby incorporated by reference in its entirety.
BACKGROUND OF THE INVENTION
Functional nucleic acids controlling the expression of target genes in vivo are an innovative class of pharmaceuticals that include antisense technology and RNA inhibition (RNAi). Both antisense gene silencing and RNAi make use of sequence-specific binding of DNA and/or RNA based oligonucleotides to cellular RNA targets, and the functional inhibition that results therefrom. This oligonucleotide-based inhibition is the result of one or more cellular mechanisms, which may include direct (steric) blockage of translation and/or triggering degradation of the RNA target. These gene-silencing approaches have the disadvantage of resulting in off-target silencing, which includes impacts on the expression of non-target RNA having similar sequences to the target. These off-target effects can complicate the use of oligonucleotide- based gene silencing where the RNA target is similar in sequence to non-target RNA. For example, dominant, gain-of-function gene mutations in heterozygotes bearing a mutant copy and a wild type copy of the gene are not typically considered ideal targets for oligonucleotide-based gene silencing.
Gene silencing approaches that can discriminate target and non-target RNA potentially differing by only a single nucleotide are needed, including the ability to suppress only the expression of a mutant allele causing a disease or condition without influencing the expression of the corresponding wild-type allele. BRIEF SUMMARY OF THE INVENTION
The present invention relates to "third generation antisense" (3GA) compounds and the therapeutic and prophylactic use of 3GA compounds to selectively down-regulate the expression of a target RNA. Gene silencing activity of 3GA compounds is sensitive to mis- matches at positions 9, 10, and 11 (from the oligonucleotide 5' end), making 3GA compounds particularly useful for discriminating target and non-target RNAs that differ by only one or a few nucleotides. In accordance with embodiments of the invention, the 3GA compounds distinguish target and non-target sequences that differ by a single nucleotide. For example, the 3GA compounds can distinguish an RNA sequence of a mutant allele (e.g., a deleterious mutant allele), from a second (e.g., wild-type) allele. In some embodiments, the deleterious mutation is defined by a single nucleotide substitution, for example, a point mutation. The compounds, compositions and methods described herein provide for single nucleotide discrimination and selective down-regulation of expression of target RNAs relative to non-target RNAs. The invention provides synthetic 3GA compounds and pharmaceutical compositions comprising two or more oligonucleotides that are complementary to one or more target RNAs, wherein the oligonucleotides are linked through their 5' ends. The target RNA has 1, 2, 3 or more mutations relative to a related, non-target RNA. The 3GA compound is specific for a region of the target RNA comprising a mutation (e.g., a deleterious mutation), and wherein the 9th, 10th, and/or 11th nucleotides from the 5' end of the component antisense oligonucleotides of the 3GA compound align with and are complementary to at least one mutation in the target RNA, preferably at least one deleterious mutation. The invention provides 3GA compounds which are sensitive to a single mismatch at nucleotides 9, 10, and 11 from the 5' end and hence able to selectively differentiate between target and non-target RNAs at the level of one nucleotide.
In other aspects, the invention provides 3GA compounds and methods that target an RNA (e.g., mRNA or miRNA) that differs from a non-target RNA (mRNA or miRNA) by no more than 1, 2, or 3 nucleotides in the target region. Thus, the 3GA compounds can distinguish between highly similar sequences. In various embodiments, the target RNA encodes an oncogene, such as BRAF, or a Ras protein such as H-Ras, K-Ras, or N-Ras. These oncogenes contain point mutations responsible for their tumorigenic activity in cells. In some embodiments, the target RNA and non-target RNA encode an enzyme, where a point mutation results in stronger activity (or abnormal activity) of the encoded enzyme, as compared to the enzyme encoded by the non-target RNA. For example, the target and non- target RNA may encode a kinase, e.g., that may harbor a mutation associated with development or progression of cancer. In still other embodiments, the target RNA and non- target RNA encode a transcriptional activator, such as MYD88. In some embodiments, the target and non-target RNA encode a G-protein coupled receptor. In some embodiments, the invention provides methods for treating a patient having a disease or disorder correlated with the presence of a target RNA, such as a dominant gain-of-function mutation allele. In various embodiments, the methods comprise administering to the patient a 3GA compound according to the invention in a therapeutically effective amount. Other aspects and embodiments of the invention will be apparent from the following detailed description of the invention.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 is a conceptual representation of exemplary 3GA compounds according to the invention. Figure 1 shows a conceptual wild-type allele (SEQ ID NO: l) and a corresponding mutant allele (SEQ ID NO:2) having a single point mutation (G > T). Figure 1 further illustrates 3GA compounds designed to target the mutant allele specifically, by aligning the oligonucleotide such that the point mutations is complementary to position 9 (SEQ ID NO:3), 10 (SEQ ID NO:4), or 11 (SEQ ID NO:5) of the oligonucleotide. Figure 2 demonstrates that mismatches at the 9th, 10th, and/or 11th nucleotide from the 5' end of the component antisense oligonucleotides of the 3GA compounds reduces the efficiency of target cleavage (SEQ ID NO: l). 3GA compounds with or without introduced mismatches are SEQ ID NOS: 6 to 10. Figure 3 demonstrates that mutations (mismatches) at the 9 , 10 , and/or 11 nucleotides from the 5' end of the component antisense oligonucleotides of the 3GA compounds also reduced gene silencing in cell culture (shown for PCSK9 target mRNA).
Figure 4 depicts the BRAF sequence (SEQ ID NO: 11) and the T to A nucleotide change in the BRAF V600E mutation sequence (SEQ ID NO: 12).
Figure 5 depicts the section of the wild-type BRAF (SEQ ID NO: 13) and BRAF V600E sequence (SEQ ID NO: 14) and a 3GA compound comprising two oligonucleotides each complementary to the wild-type sequence but for a mismatch at position 11, and which is fully complementary to the V600E sequence (SEQ ID NO: 15). Figure 6 demonstrates that a 3GA compound targeting the BRAF V600E mutation selectively inhibits BRAF V600E expression and not wild-type expression in cell culture.
Figure 7 depicts the MyD88 wild-type sequence (SEQ ID NO: 16) and the T to C nucleotide change in the MyD88 L265P mutation sequence (SEQ ID NO: 17).
Figure 8 depicts the section of the wild-type MyD88 (SEQ ID NO: 18) and MyD88 L265P sequence (SEQ ID NO: 19) and a 3GA compound targeting the L265P sequence (SEQ ID NO: 20), but having a mismatch at position 11 upon hybridization with the wild-type sequence.
Figure 9 demonstrates that a 3GA compound targeting the MyD88 L265P mutation selectively inhibits MyD88 L265P expression and not expression of the wild-type RNA. DETAILED DESCRIPTION OF THE INVENTION
The present invention relates to third generation antisense (3GA) compounds and the therapeutic and prophylactic use of 3GA compounds to selectively down-regulate the expression of an RNA comprising a target sequence, over non-target RNA comprising a nucleotide sequence similar to the target (e.g., differing by at least one nucleotide from the target). For example, the RNA target can be expressed from a first allele (e.g., a mutant allele), even when the first allelic mRNA differs from a second allele (e.g., wild-type allele) by only a single nucleotide, as is the case with certain mutations, for example, point mutations. The 3GA compounds of the present invention are capable of single nucleotide discrimination and selective down-regulation of expression of their target alleles. The methods of the present invention are applicable to the treatment of diseases caused by dominant, gain-of-function type of gene mutations, such as gain-of-function point mutations.
As used herein, the term "3GA compound" refers to a compound comprising at least two antisense oligonucleotides linked, directly or indirectly, through their 5' ends, such that the compound comprises at least two 3' ends. 3GA compounds provide for efficient gene silencing. See U.S. Patent 8,431,544, which is hereby incorporated by reference. Also see Bhagat et al., Novel oligonucleotides containing two 3'-ends complementary to target mRNA show optimal gene-silencing activity, J Med Chem. 2011 Apr 28;54(8):3027-36.
The term "3' ", when used directionally, generally refers to a region or position in a polynucleotide or oligonucleotide 3' (toward the 3 'end of the nucleotide) from another region or position in the same polynucleotide or oligonucleotide. The term "3' end" generally refers to the 3' terminal nucleotide of the component oligonucleotide. The term "5' ", when used directionally, generally refers to a region or position in a polynucleotide or oligonucleotide 5' (toward the 5 'end of the nucleotide) from another region or position in the same polynucleotide or oligonucleotide. The term "5' end" generally refers to the 5' terminal nucleotide of the component oligonucleotide. For example, where two or more single- stranded antisense oligonucleotides are linked at their 5' ends, there is a linkage between the 5' terminal nucleotides of the oligonucleotides which may be directly via 5', 3' or 2' hydroxyl groups, or indirectly, via a non-nucleotide linker. Such linkages may also be via a nucleoside, utilizing both 2' and 3' hydroxyl positions of the nucleoside. Such linkages may also employ a functionalized sugar or nucleobase of a 5' terminal nucleotide.
The 3GA compounds in accordance with embodiments of the invention are able to discriminate between RNAs expressed from two alleles in a cell or organism. The term "allele" refers to one of two alternate forms of a gene that can have the same locus on homologous chromosomes. Two different alleles may be responsible for alternative traits, e.g., one allele can be dominant over the other. The term "dominant allele" refers to an allele from which a trait is preferentially manifested as a phenotype.
The alleles in some embodiments encode proteins having biologically different activities. For example, one of the alleles may encode an abnormally functioning protein while the other allele may encode a normally functioning protein. In this case, the allele encoding the abnormally functioning protein may act as a dominant allele, resulting in the abnormal function of the protein exhibited as a phenotype in the individual (e.g., a disease phenotype). Specific examples thereof include alleles responsible for autosomal dominantly inherited diseases.
In various embodiments, the 3GA compounds comprise oligonucleotides that are completely complementary to a region of the target RNA. In this context, "fully complementary" or "completely complementary" means each nucleobase of an oligonucleotide has a complementary nucleobase in the target sequence or region. The term "oligonucleotide" refers to a polynucleoside formed from a plurality of linked nucleoside units, which may include, for example, deoxyribonucleotides or ribonucleotides, natural or modified nucleotides, phosphodi ester or modified linkages, natural bases or modified bases natural sugars or modified sugars, or combinations of these components. The nucleoside residues can be coupled to each other by any of the numerous known internucleoside linkages. Such internucleoside linkages include, without limitation, phosphodiester, phosphorothioate, phosphorodithioate, methylphosphonate, alkylphosphonate, alkylphosphonothioate, phosphotriester, phosphoramidate, siloxane, carbonate, carboalkoxy, acetamidate, carbamate, morpholino, borano, thioether, bridged phosphoramidate, bridged methylene phosphonate, bridged phosphorothioate, and sulfone internucleoside linkages. The term "oligonucleotide" also encompasses polynucleosides having one or more stereospecific internucleoside linkage (e.g., (Rp)- or (Sp)- phosphorothioate, alkylphosphonate, or phosphotriester linkages). In certain exemplary embodiments, these internucleoside linkages may be phosphodiester, phosphorothioate or phosphorodithioate linkages, or combinations thereof. In exemplary embodiments, the nucleotides of the synthetic oligonucleotides are linked by at least one phosphorothioate internucleotide linkage. The phosphorothioate linkages may be mixed Rp and Sp enantiomers, or they may be stereoregular or substantially stereoregular in either Rp or Sp form (see Iyer et al. (1995) Tetrahedron Asymmetry 6: 1051-1054). In certain embodiments, one or more of the oligonucleotides within the antisense compositions of the invention contain one or more locked nucleotides (e.g., 2'-0,4'-C-methylene-b-D- ribofuranosyl nucleic acids), wherein the ribose is modified with a bond between the 2' and 4' carbons, which fixes the ribose in the 3'-endo structural conformation.
The term "biologic instability" generally refers to a molecule's ability to be degraded and subsequently inactivated in vivo. For oligonucleotides, such degradation results from exonuclease activity and/or endonuclease activity, wherein exonuclease activity refers to cleaving nucleotides from the 3' or 5' end of an oligonucleotide, and endonuclease activity refers to cleaving phosphodiester bonds at positions other than at the ends of the oligonucleotide. Modifications to the oligonucleotide can be selected to render the 3GA compound more stable in vivo.
The oligonucleotides may include a modified heterocyclic base, a modified sugar moiety, or any combination thereof. In some embodiments, the modified nucleoside or nucleotide derivative is a non-natural pyrimidine or purine nucleoside. For purposes of the invention, a modified nucleoside or nucleotide derivative, a pyrimidine or purine analog or non-naturally occurring pyrimidine or purine can be used interchangeably and refers to a nucleoside that includes a non-naturally occurring base and/or non-naturally occurring sugar moiety. For purposes of the invention, a base is considered to be non-natural if it is not guanine, cytosine, adenine, thymine or uracil and a sugar is considered to be non- natural if it is not β-ribo-furanoside or 2'-deoxyribo-furanoside. In some embodiments, the 3GA compounds comprise oligonucleotides having one or more 2'-0-substituted nucleotides. The term "2'-0-substituted" means substitution of the 2' position of the pentose moiety. Exemplary substitutions include -O-lower alkyl group containing 1-6 carbon atoms (for example, but not limited to, 2'-0-methyl or 2'-0- ethyl), or with an -O-aryl or allyl group having 2-6 carbon atoms, wherein such alkyl, aryl or allyl group may be unsubstituted or may be substituted. Exemplary 2'-0-substituted nucleotides include 2'-0-methoxyethyl. In some embodiments the 3GA compound comprises one or more oligonucleotides that have from one to ten, such as two, three, four, five, six, seven, or eight 2'-0-substituted nucleotides (e.g., 2'-0-methyl or 2'-0- methoxy ethyl).
The term "point mutation" refers to a single-base substitution observed in a target nucleotide sequence compared with the corresponding nucleotide sequence of a non-target sequence (e.g., a wild-type or normal allele). In this context, the "wild-type allele" refers to common naturally occurring alleles in the allele population of the same type of gene, wherein a protein encoded by this allele has normal function and/or activity. The point mutation may be any of congenitally occurring mutations and postnatally acquired mutations. Further point mutations include missense mutations that bring about amino acid substitution, silent mutations that do not result in amino acid substitution but causes change to a degenerate codon, a nonsense mutation that leads to the appearance of a stop codon, and a mutation at a splicing site. In certain embodiments, the point mutation is a dominant point mutation. A "dominant point mutation" refers to a point mutation that confers a dominant trait on the allele, or a dominant mutation-associated (or -linked) point mutation in one transcript.
"Targeting" or "targeted" means the process of design and selection of a gene silencing compound that will specifically hybridize to a target nucleic acid and induces a desired effect. "Target gene", "target allele", "target nucleic acid," "target RNA," "target mRNA," and "target RNA transcript" all refer to a nucleic acid whose expression is to be selectively inhibited or silenced. A "target allele" is an allele whose expression is to be selectively inhibited or silenced. "Target segment", "target region", and "target site" all refer to the sequence of nucleotides of a target nucleic acid to which a 3GA compound is targeted.
The invention provides 3GA compounds and methods for the selective silencing of a mutant/target mRNA allele, while allowing another non-target mRNA or allele to remain relatively or substantially unaffected (e.g., allele specific inhibition of expression). Without wishing to be bound by any particular theory, it is believed that binding interactions between the target RNA and the 3GA compound cause preferential cleavage of the target RNA (e.g., expressed from a mutant allele) over the non-target RNA (e.g., wild-type mRNA), leaving the non-target RNA substantially less affected. In some embodiments, the non-target RNA remains expressed at a level that provides for wild-type or near wild-type protein or gene functionality.
"Allele specific inhibition of expression" refers to the ability to significantly inhibit expression of one allele of a gene over another, e.g., when both alleles are present in the same cell. For example, the alleles can differ by one, two, or three or more nucleotides in the target region. In some embodiments, one allele is associated with disease causation, e.g., a disease correlated to a dominant gain-of-function mutation.
The target allele may specify the amino acid sequence of a mutant protein associated with a pathological condition. For example, the protein may be a gain-of- function (e.g., a dominant gain-of-function) mutant protein. In a preferred aspect, the mutant protein is associated with a disease or disorder which is correlated with expression of a particular allele of a gene, e.g., a dominant gain-of-function mutation. The term "gain- of-function mutation" as used herein, refers to any mutation in a gene in which the protein encoded by said gene (i.e., the mutant protein) acquires a function not normally associated with the protein (i.e., the wild type protein) causes or contributes to a disease or disorder. The gain-of-function mutation can be a deletion, addition, or substitution of a nucleotide or nucleotides in the gene which gives rise to the change in the function of the encoded protein. In one embodiment, the gain-of-function mutation is a point mutation. In one embodiment, the gain-of-function mutation is a translocation.
In one embodiment, the gain-of-function mutation changes the function of the mutant protein or causes interactions with other proteins. In another embodiment, the gain- of-function mutation causes a decrease in or removal of normal wild-type protein, for example, by interaction of the altered, mutant protein with said normal, wild-type protein. The trait in which the dominant point mutation is involved is not particularly limited and is preferably a trait to be suppressed. Examples thereof include a mutation involved in the onset of a disease and a mutation involved in abnormal morphology. Gain- of-function disorders are a class of disease or disorders characterized by a gain-of-function mutation. For example, such disorders may include amyotrophic lateral sclerosis, Huntington's disease, Alzheimer's disease, Parkinson's disease, as well as cancer.
Other examples of gain-of-function mutations include the KIT receptor, which has been linked to a number of gastrointestinal stromal tumors. Naturally occurring mutations in G protein alpha subunits and in G protein-coupled receptors have been linked to a number of human diseases, including endocrine disorders. Germline loss of function mutations in the ubiquitously expressed Gs-alpha gene have been identified as the cause of generalized hormone resistance and dysmorphic features in the inherited disorder pseudohypoparathyroidism type la. Somatic gain-of-function mutations in Gs-alpha have been identified as the cause of the McCune-Albright syndrome, a sporadic disorder in which affected individuals have varying combinations of endocrine hyperfunction, cafe-au- lait skin pigmentation, and polyostotic fibrous dysplasia. These mutant genes and conditions may be targeted with 3GA compounds in accordance with the invention.
Further, gain-of-function mutations in the thyrotropin receptor (TSHR, a G-protein coupled receptor) are correlated with toxic follicular thyroid adenoma, a condition caused by excessive quantities of thyroid hormones. Gain-of-function mutations in TSH receptor genes have also been linked to hereditary toxic thyroid hyperplasia, another condition caused by excessive quantities of thyroid hormones. Mutations of the superoxide dismutase (SOD) gene have been linked to certain familial forms of ALS. Mutations in protein-tyrosine phosphatase, nonreceptor-type 11 (PTPN11) have been correlated with Noonan syndrome, an autosomal dominant disorder characterized by dysmorphic facial features, proportionate short stature and heart disease. Hereditary pancreatitis is associated with mutations in human cationic trypsinogen. Brachydactyly type B (BDB), an autosomal dominant disorder characterized by terminal deficiency of the fingers and toes, is believed to be associated with dominant gain-of-function mutation in ROR2, which encodes an orphan receptor tyrosine kinase, von Willebrand disease, particularly Type 2A and 2B, is another disease which may be associated with a dominant gain-of-function mutation. A dominant gain-of-function mutation has been described in p53 that results in oncogenic activation of that gene. In addition, Creutzf el dt- Jakob disease has been associated with a dominant gain-of-function mutation in the prion protein gene, the PRNP E200K mutation. Testotoxicosis is an autosomal dominant condition caused by a gain-of-function mutation in the LH receptor.
In some embodiments, the target RNA encodes an oncogene, such as BRAF, or a Ras protein such as H-Ras, K-Ras, or N-Ras. These oncogenes contain point mutations responsible for their tumorigenic activity in cells. For example, the 3GA compound may comprise oligonucleotides that are complementary to a T > A mutation at position 9, 10, 11 of the oligonucleotides (numbered from the 5' end). In some embodiments, the 3GA compound may comprise oligonucleotides that are complementary to a T > A mutation at position 11 of the oligonucleotides (numbered from the 5' end). An exemplary 3GA compound targeting the BRAF oncogene is provided as SEQ ID NO: 15. The compound shown as SEQ ID NO: 15 may contain oligonucleotide modifications as disclosed herein, including one or more 2'-0-substituted nucleotides and one or more phosphorothioate linkages.
In some embodiments, the target RNA and non-target encode an enzyme, where a point mutation results in stronger activity (or abnormal activity) of the encoded enzyme, as compared to the enzyme encoded by the non-target RNA. For example, the target and non- target RNA may encode a kinase. In various embodiments, the enzyme is an oxidoreductase, transferase, hydrolase, lyase, isomerase, or ligase. In some embodiments, the enzyme is a superoxide dismutase or a triglyceride hydrolase. In still other embodiments, the target RNA and non-target RNA encode a transcriptional activator, such as MYD88. The MYD88 L265P variant is the most prevalent mutation in patients with Waldenstrom's macroglobulinemia (WM), a type of non-Hodgkin's lymphoma. MYD88 L265P often results from a T→C transversion. Signaling studies showed that the mutant protein that is encoded by MYD88 L265P triggers tumor growth through the activation of nuclear factor kappa light-chain enhancer of activated B cells (NF-κΒ) by Bruton's tyrosine kinase. (Treon et al., MYD88 Mutations and Response to Ibrutinib in Waldenstrom's Macroglobulinemia, N Engl J Med 2015; 373 :584-586 (2015)). SEQ ID NO: 20 is a 3GA compound targeting MYD88 L265P encoding mRNA. The compound shown as SEQ ID NO: 20 may contain oligonucleotide modifications as disclosed herein, including one or more 2'-0-substituted nucleotides and one or more phosphorothioate linkages.
By inhibiting the expression of such proteins, e.g., allele-specific dominant gain-of- function mutant proteins, valuable information regarding the function of said proteins as well as therapeutic benefits may be obtained.
The 3GA compound are capable of single nucleotide discrimination between target RNA (e.g., expressed from a "mutant allele" or "target allele") and a non-target RNA (e.g., expressed from a wild-type allele), the mutant or target RNA having 1, 2, 3 or more mutations relative to the non-target RNA. In various embodiments, the 3GA compound comprises a sequence complementary to a region of the target RNA (e.g., from 15 to 25 nucleotide) encoding a gain-of-function point mutation, said region comprising 1, 2, or 3 point mutations. The 3GA compound comprises a sequence comprising 1, 2, or 3 nucleotides aligned opposite of and complementary to the 1, 2, 3 or more point mutations, wherein at least one nucleotide complementary to a point mutation is located at the 9th, 10th, and/or 11th nucleotide positions from the 5' end of a component antisense oligonucleotide. The 3GA compound directs target-specific cleavage of an RNA containing the point mutations, whereas a non-target RNA (which results in a mismatch at positions 9, 10, and/or 11), is degraded at a substantially lower level/rate. In certain embodiments the mutant allele comprises 1 point mutation. In some embodiments the mutant allele comprises 2 point mutations. In other embodiments the mutant allele comprises 3 point mutations. In some embodiment the mutant allele comprises more than 3 point mutations. In various embodiments, a deleterious point mutation is complementary to position 11 of the component oligonucleotides. The 3GA compounds comprise two or more single-stranded antisense oligonucleotides linked at their 5' ends, wherein the antisense oligonucleotides are complementary to a nucleic acid sequence of a target RNA, wherein the target RNA differs by at least one base pair from a non-target or wild-type RNA, and wherein the 9th, 10th, and/or 11th nucleotides from the 5' end of the component antisense oligonucleotides is aligned opposite of and complementary to at least one different nucleotide within the target RNA. As such, the 3GA compounds when hybridized to a non-target, wild-type allele, will produce at least one mismatch at the 9th, 10th, and/or 11th nucleotides of the antisense oligonucleotide (as determined from the 5' end). In some embodiments, the target RNA is expressed from a gain-of-function allele. In some embodiments, the difference between the target allele and the wild type allele is a single nucleotide, e.g., a point mutation. In some embodiments, the target RNA and the non-target RNA are messenger-RNA. In other embodiments, the target RNA and non-target RNA are miRNAs, such that the 3GA compound differentiates related miRNAs or iso-miRs, based on designing complementarity to the target miRNA at positions 9, 10, 11 (e.g., position 11) of the oligonucleotides, with a mismatch to the non-target RNAs predicted at these/this position.
The 3GA compound should be specific for a target region that differs by at least one base pair between the non-target (e.g., wild-type) and target (e.g., mutant) RNA, e.g., a target region comprising the gain-of-function mutation. In some embodiments the difference between the target RNA and the non-target RNA is a single nucleotide, e.g., a point mutation. In some embodiments, where the gain-of-function mutation is associated with one or more other mutations in the same gene, the 3GA compound can be targeted to any of the mutations. In some embodiments, the 3GA is targeted to a target region that does not comprise a known mutation but does comprise an allelic variation of the wild- type (reference) sequence.
The component antisense oligonucleotides of the 3GA compounds are typically at least 14 nucleotides in length. In some embodiments the component antisense oligonucleotides of the 3GA compounds are, independently, 15 to 40 nucleotides long. In some embodiments the component antisense oligonucleotides of the 3GA compounds are, independently, 17 to 30 nucleotides long. In some embodiments the component antisense oligonucleotides of the 3GA compounds are, independently, 18 to 23 nucleotides long. In some embodiments the component antisense oligonucleotides of the 3GA compounds are each 19 nucleotides long. Thus, the component oligonucleotides of the oligonucleotide- based compounds of the invention are preferably 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39 or 40 nucleotides in length.
In some embodiments, the general structure of the 3GA compounds may be described by the following formula:
NiN2N3N4N5N6N7N8N9NioNiiNi2Ni3Ni4Ni5Ni6Ni7Ni8Ni9...N„-3' (Formula I)
wherein L is a nucleotide linker or non-nucleotide linker; wherein N^N19 and N1-N19, at each occurrence, is independently a nucleotide or nucleotide derivative; wherein Nm and Nn, at each occurrence, are independently a nucleotide or nucleotide derivative; wherein m and n are independently numbers from 0 to 10; wherein the sequences of N^N19 and Ni- Nig are the same or different; and wherein N9, N10, and/or N11 and N9, N10, and/or Nn are aligned opposite and complementary to one or more mutations in the mutant/target RNA. In some embodiments, the sequences of N^N19 and N1-N19 are the same. In some embodiments, m and n are from 0 to 2. In some embodiments, m is 0 or 1. In some embodiments, n is 0 or 1. In some embodiments, m and n are 0. In some embodiments, L is a non-nucleotide linker.
Figure 1 shows a conceptual structural representation of a 3GA compound. As shown in Figure 1, an exemplary wild-type allele and a mutant allele having a single G to T point mutation are provided. The 3GA compounds are complementary to the mutant allele wherein the 9th, 10th or 11th nucleotide position from the 5' end of the component antisense oligonucleotides is aligned opposite and complementary to the point mutation ("T") within the mutant allele (e.g., 3GA 9th, 3GA 10th and 3GA 11th, respectively). In some embodiments, the 3GA compound comprises oligonucleotides that are complementary to the point mutation at position 11 of the oligonucleotide. In some embodiments, where the mutant allele comprises 2 consecutive mutations, the 3GA compounds are complementary to the corresponding RNA wherein one of the 9th, 10th or 11th nucleotide positions from the 5' end of the component antisense oligonucleotides is aligned opposite with and complementary to one of the point mutations, or wherein the 9th and 10th or 10th and 11th nucleotide positions from the 5' end of the component antisense oligonucleotides are aligned opposite with and complementary to the point mutations. Where the mutant allele comprises 2 mutations with an intervening wild- type nucleotide, the 3GA compounds are complementary to the mutant allele wherein one of the 9th, 10th or 11th nucleotide positions from the 5' end of the component antisense oligonucleotides is aligned opposite with and complementary to one of the point mutations, or wherein the 9th and 11th nucleotide positions from the 5' end of the component antisense oligonucleotides are aligned opposite with and complementary to the point mutation. Where the mutant allele comprises 3 consecutive mutations, the 3GA compounds are complementary to the mutant allele wherein at least one of the 9th, 10th or 11th nucleotide positions from the 5' end of the component antisense oligonucleotides is aligned opposite with and complementary to one of the point mutations, or wherein each of the 9th 10th and 11th nucleotide positions from the 5' end of the component antisense oligonucleotides are aligned opposite with and complementary to the point mutations.
As a result of the linking of two or more antisense oligonucleotides at their 5' ends, the 3GA compounds have two or more accessible 3' ends. The linkage at the 5' ends of the component oligonucleotides is independent of the other oligonucleotide linkages and may be directly via 5', 3' or 2' hydroxyl groups, or indirectly, via a non-nucleotide linker or a nucleoside, utilizing either the 2' or 3' hydroxyl positions of the nucleoside. Linkages may also utilize a functionalized sugar or nucleobase of a 5' terminal nucleotide. In some embodiments, the linkage at the 5'-ends is via a non-nucleotidic linker.
The term "linker" generally refers to any moiety that can be attached to an oligonucleotide by way of covalent bonding through a sugar, a base, or the backbone. Such linker can be either a non-nucleotide linker or a nucleoside linker.
Representative non-nucleotide linkers are set forth in Table 1. Table 1: Representative Non-Nucleotide Linkers
Figure imgf000018_0001
thane
Glycerol (1,2,3-Propanetriol)
Figure imgf000018_0002
l)propane
1,2,4-Butanetriol
2-
2
Figure imgf000018_0003
2-Amino-2-(hy droxymethy 1)- 1 ,3-propanediol
1,1,1 -Tris(hy droxymethy l)ethane
Figure imgf000018_0004
N-[Tris(hy droxymethy l)methyl]acrylamide Table 1: Continued
Figure imgf000019_0001
1 ,3 -Di(hydroxyethoxy)-2-hy droxyl-propane cis- 1 , 3 , 5 -Cy clone xanetriol
Figure imgf000019_0002
cis- 1 ,3 ,5 -Tri(hydroxy methyl)cyclohexane
Figure imgf000019_0003
3,5, -Di(hy droxy methy l)phenol
Figure imgf000019_0004
l
1 , 3 , 5 , -Tri(hy droxy methy l)benzene Table 1: Continued
Figure imgf000020_0001
1 ,6-anhydro-P-D-Glucose 4.6-Nitropyrogallol
Figure imgf000020_0002
l,3,5-Tris(2-hydroxyethyl)-Cyanuric
Figure imgf000020_0003
Figure imgf000020_0004
3,5,7-Trihydroxyflavone Table 1: Continued
HO' "' v "OH
,OH 1,5-Pentanediol
HO'
Ethylene glycol
Figure imgf000021_0001
1,3 -Propanediol 2,4-Pentanediol
Figure imgf000021_0002
1 ,2-Propanediol
1,6-Hexanediol
Figure imgf000021_0003
2,5-Hexanediol
Figure imgf000021_0004
Table 1: Continued
Figure imgf000022_0001
2-( 1 -Aminopropyl)- 1 ,3 -propanediol
1,8-Octanediol ose
Figure imgf000022_0002
1,12-Dodecanediol
Figure imgf000022_0003
Triethylene glycol
Figure imgf000022_0004
Tetraethylene glycol
Figure imgf000022_0005
Hexaethylene glycol
In some embodiments, the small molecule linker is glycerol or a glycerol homolog of the formula HO-(CH2)0-CH(OH)-(CH2)/ OH, wherein o and p independently are integers from 1 to about 6, from 1 to about 4 or from 1 to about 3. In some other embodiments, the small molecule linker is a derivative of l,3-diamino-2-hydroxypropane. Some such derivatives have the formula
HO-(CH2)m-C(0) H-CH2-CH(OH)-CH2- HC(0)-(CH2)m-OH, wherein m is an integer from 0 to about 10, from 0 to about 6, from 2 to about 6 or from 2 to about 4.
Some non-nucleotide linkers permit attachment of more than two oligonucleotides. For example, the small molecule linker glycerol has three hydroxyl groups to which such oligonucleotides may be covalently attached. Some 3GA compounds comprise two or more oligonucleotides linked to a nucleotide or a non-nucleotide linker. Such oligonucleotides according to the invention are referred to as being "branched".
In some embodiments, the 3GA compounds comprise two or more antisense oligonucleotides having identical sequences. For example, the 3GA compound may comprise two or more antisense oligonucleotides having different sequences. In certain embodiments, the 3GA compound comprises two or more antisense oligonucleotides substantially complementary to a target sequence. In certain embodiments, the 3GA compound comprises two antisense oligonucleotides having identical sequences, and optionally structures. In certain embodiments, the 3GA compound comprises two antisense oligonucleotides having different sequences. The 3GA compounds comprise two antisense oligonucleotides substantially complementary to a target sequence. 3GA compounds that comprise antisense oligonucleotides having identical sequences are able to bind to a specific RNA via Watson-Crick hydrogen bonding interactions and silence expression. 3GA compounds that comprise different sequences are able to bind to two or more different regions of the same RNA target or bind to two or more different RNA target(s) and silence expression. Such compounds are comprised of heteronucleotide sequences complementary to target RNA and form stable duplex structures through Watson-Crick hydrogen bonding.
Other modifications of 3GA compounds include those that are internal or at the end(s) of the oligonucleotide molecule and include additions to the molecule, such as cholesterol, cholesteryl, or diamine compounds with varying numbers of carbon residues between the amino groups and terminal ribose, deoxyribose and phosphate modifications which cleave, or crosslink to the opposite chains or to associated enzymes or other proteins which bind to the genome.
3GA compounds may be synthesized as described in US Patent 8,431,544, which is hereby incorporated by reference in its entirety.
The 3GA compounds can be incorporated into pharmaceutical compositions. Such compositions typically include the 3GA compound and a pharmaceutically acceptable carrier. In some embodiments, the pharmaceutically acceptable carrier comprises saline, solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration. Supplementary active compounds can also be incorporated into the compositions.
The term "pharmaceutically acceptable" means a non-toxic material that does not interfere with the effectiveness of a compound according to the invention or the biological activity of a compound according to the invention. The term "physiologically acceptable" refers to a non-toxic material that is compatible with a biological system such as a cell, cell culture, tissue, or organism. Preferably, the biological system is a living organism, such as a mammal, particularly a human.
The term "carrier" generally encompasses any excipient, diluent, filler, salt, buffer, stabilizer, solubilizer, oil, lipid, lipid containing vesicle, microspheres, liposomal encapsulation, or other material for use in pharmaceutical formulations. It will be understood that the characteristics of the carrier, excipient or diluent will depend on the route of administration for a particular application. The preparation of pharmaceutically acceptable formulations containing these materials is described in, for example, Remington's Pharmaceutical Sciences, 18th Edition, ed. A. Gennaro, Mack Publishing Co., Easton, PA, 1990.
A pharmaceutical composition is formulated to be compatible with its intended route of administration. Examples of routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, oral, intramuscular, transdermal (topical), transmucosal, and rectal administration. Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide. The parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic. In some aspects, the invention provides a method for inhibiting target RNA expression, the method comprising contacting a cell with a 3GA compound, or a composition comprising a 3GA compound. For example, the cell may express both target and non-target RNA as described above. In some embodiments, a component oligonucleotide of the 3GA compound is complementary to an RNA expressed from a first allele in the cell (the target RNA), but is not fully complementary to an RNA expressed from a second allele (e.g., non-target RNA) in the cell. Thus, when the component oligonucleotide hybridizes to the target RNA, it is fully complementary at positions 9, 10, 11 of the oligonucleotide (as determined from its 5' end), thereby triggering destruction of the target RNA. On the other hand, when the component oligonucleotide hybridizes to the second, non-target RNA, there will be at least one mismatch at positions 9, 10, and 11, such that the non-target allele will not be degraded at the rate/level of the target RNA. In some embodiments, the oligonucleotides are completely complementary to the target RNA, but have a mismatch at position 11 of the oligonucleotide when hybridized to the non-target RNA. The invention also provides a method for inhibiting allele specific gene expression in a mammal, the method comprising administering to the mammal a 3GA compound, or a composition comprising a 3GA compound. Exemplary mammals include, without limitation, humans, non-human primates, rats, mice, cats, dogs, horses, cattle, cows, pigs, sheep and rabbits. In some embodiments, the mammal is a human. The mammalian subject (e.g., human) is heterozygous for the gene of interest, and thus has an allele that expresses the target RNA, and a corresponding allele that expresses non-target RNA. The target and non-target RNAs (which can be expressed from a mutant and corresponding wild-type allele, respectively) may differ by only one nucleotide in the target region. In some embodiments, a component oligonucleotide of the 3GA compound is complementary to a portion of the target RNA, but is not fully complementary to a portion of the non-target RNA (e.g., produced by the wild-type allele). Thus, when the component oligonucleotide hybridizes to the target RNA, it is fully complementary at positions 9, 10, 11 of the oligonucleotide (as determined from its 5' end), thereby triggering destruction of the target RNA. On the other hand, when the component oligonucleotide hybridizes to the second, non-target RNA, there will be at least one mismatch at positions 9, 10, and 11, such that the non-target RNA will not be degraded at the rate or level of the target RNA.
The present invention provides for both prophylactic and therapeutic methods of treating a subject at risk of (or susceptible to) a disorder, or having a disorder, associated with expression of a particular allele of a gene, e.g., a dominant gain-of-function mutation. The term "treatment" generally refers to an approach intended to obtain a beneficial or desired result, which may include alleviation of symptoms, or delaying or ameliorating a disease progression.
The presence or predisposition to the disease can be confirmed by determining all or part of the genotype of the patient using routine methods, generally including that portion of the genotype of the patient that is known to be associated with a disease. The treatment can include administering 3GA compounds, or compositions comprising 3GA compounds, complementary to one or more target sites on one or more target alleles (e.g., RNAs). The treatment can include administering 3GA compounds, or compositions comprising 3GA compounds, complementary to a target site on a target allele. A mixture of different 3GA compounds, or compositions comprising 3GA compounds, may be administered together or sequentially, and the mixture may be varied over occasion.
In some embodiments, the patient has a disease or disorder correlated with the presence of a dominant, gain-of-function mutation allele. In some embodiments, the patient is a cancer patient testing positive for a BRAF mutation, or a mutation in a Ras protein such as H-Ras, K-Ras, or N-Ras. These oncogenes contain point mutations responsible for their tumorigenic activity in cells. For example, the 3GA compound may comprise oligonucleotides that are complementary to a T > A mutation at position 9, 10, 11 of the oligonucleotides (numbered from the 5' end). In some embodiments, the 3GA compound may comprise oligonucleotides that are complementary to a T > A mutation at position 11 of the oligonucleotides (numbered from the 5' end). An exemplary 3GA compound targeting the BRAF oncogene is provided as SEQ ID NO: 15. The compound shown as SEQ ID NO: 15 may contain oligonucleotide modifications as disclosed herein, including one or more 2'-0-substituted nucleotides and one or more phosphorothioate linkages.
In still other embodiments, the patient may have Waldenstrom's Macroglobulinemia, and the target RNA and non-target RNA may encode MYD88. The MYD88 L265P variant is the most prevalent mutation in patients with Waldenstrom's macroglobulinemia (WM), a type of non-Hodgkin's lymphoma. MYD88 L265P often results from a T→C transversion. Signaling studies showed that the mutant protein that is encoded by MYD88 L265P triggers tumor growth through the activation of nuclear factor kappa light-chain enhancer of activated B cells (NF-κΒ) by Bruton's tyrosine kinase. SEQ ID NO: 20 is a 3GA compound targeting MYD88 L265P encoding mRNA. The compound shown as SEQ ID NO: 20 may be contain oligonucleotide modifications as disclosed herein, including one or more 2'-0-substituted nucleotides and one or more phosphorothioate linkages.
In some embodiments, the target RNA and non-target encode an enzyme, where a point mutation results in stronger activity (or abnormal activity) of the encoded enzyme, as compared to the enzyme encoded by the non-target RNA. For example, the patient may have cancer associated with a heightened kinase activity, and the target and non-target RNA may encode the kinase, which is targeted by the 3GA compound. In various embodiments, the target RNA encodes an enzyme that is an oxidoreductase, transferase, hydrolase, lyase, isomerase, or ligase, and which harbors a mutation associated with disease. In some embodiments, the enzyme is a superoxide dismutase or a triglyceride hydrolase.
In various other embodiments, the disease or disorder comprises one or more of amyotrophic lateral sclerosis (ALS), Huntington's disease, Alzheimer's disease, Parkinson's disease, gastrointestinal stromal tumors, endocrine disorders, pseudo hypo-parathyroidism type IA, the McCune- Albright syndrome, toxic follicular thyroid adenoma, hereditary toxic thyroid hyperplasia, familial forms of ALS, Noonan syndrome, Hereditary pancreatitis, brachydactyly type B (BDB), Waldenstrom's macroglobulinemia (WM), non-Hodgkin's lymphoma, von Willebrand disease, e.g. Type 2A and 2B, diseases result from mutations in the BRAF proto-oncogene, oncogenic diseases results from gain-of-function mutation in p53, Creutzfeldt-Jakob disease, and Testotoxicosis.
The examples below are intended to further illustrate certain preferred embodiments of the invention, and are not intended to limit the scope of the invention.
EXAMPLES Example 1: Altering Excision of Targeted RNA by Positional Modification of 3GA Compounds
Mouse PCSK9 Cell Culture and Knockdown Experiments
Hepal-6 cells (ATCC) were seeded in 12-well cluster plates in DMEM + 10% FBS, 24 hours prior to transfection. Cells were then transiently transfected with 50 nM oligonucleotides of interest using Lipofectamine 2000. After 16 hours, total RNA isolation was performed using Qiagen's RNeasy Kit. cDNA synthesis was then performed using Applied Biosystem's High-Capacity cDNA Reverse Transcription Kit. This was followed by quantitative real-time PCR using a PCSK9 TaqMan gene expression assay (Applied Biosystems). Gene expression was calculated using the 2"DDCT method, with PPIB as the endogenous control.
RNA-Ligase Mediated Rapid Amplification of cDNA Ends (RLM-RACE^ RLM-RACE was performed using Ambion's FirstChoice RLM-RACE kit. Briefly, total RNA was ligated to a 5' adapter using T4 RNA Ligase. First strand cDNA synthesis was then performed using a PCSK9-specific primer. This was followed by two rounds of nested PCR using PCSK9-specific primers. PCR products were then run in an agarose gel and bands of interest were excised and purified using Qiagen's QIAquick Gel Extraction Kit. Samples were then cloned into the pCR™4-TOPO sequencing vector using Invitrogen's TOPO-TA cloning kit and submitted for Sanger sequencing.
As shown in Figure 1, a 3GA compound complementary to a target sequence was modified by introducing a mismatched nucleotide at the 9th, 10th, and/or 11th nucleotide from the 5' end of the component antisense oligonucleotides.
Figure 2 demonstrates that modifying a 3GA compound to introduce a mismatched nucleotide at the 9th, 10th, and/or 11th nucleotide from the 5' end of the component antisense oligonucleotides reduces the efficiency of cleavage of the target sequence. Additionally, Figure 3 demonstrates that these modifications of a 3GA compound also resulted in the loss of gene silencing.
Example 2: Targeting Genes with a Gain-Of-Function Point Mutation
BRAF Cell Culture and Knockdown Experiments
The BRAF proto-oncogene encodes a serine/threonine kinase that is mutated in 15% of all human cancers. In melanomas, the incidence of BRAF mutations can reach as high as 60%. The most common BRAF mutation is V600E (nucleotide change: 1799T>A), which comprises 80% of all BRAF mutations (Figure 4). Thus, the difference between the wild-type BRAF and BRAF V600E is a single point mutation. This mutation occurs in the kinase domain of the BRAF protein and leads to constitutive kinase activity. 3GA compound described according to the invention were employed against the BRAF V600E mutation (Figure 5).
A MCF10A cells expressing either BRAF wild-type (ATCC and Sigma) or BRAF V600E (Sigma) were seeded in 12-well cluster plates in MEGM media (Lonza), 24 hours prior to transfection. Cells were then transiently transfected with 50 nM of the 3GA compound using Lipofectamine 2000. After 16 hours, total RNA isolation was performed using Qiagen's RNeasy Kit. cDNA synthesis was then performed using Applied Biosystem's High-Capacity cDNA Reverse Transcription Kit. This was followed by quantitative real-time PCR using a BRAF TaqMan gene expression assay (Applied Biosystems) or custom castPCR assays (Applied Biosystems) specific for either wild-type or V600E BRAF transcripts. Gene expression was calculated using the 2-DDCT method, with PPIB as the endogenous control.
As shown in Figure 6, the BRAF V600E specific 3GA compounds selectively inhibited BRAF V600E expression. BRAF wild-type expression was unaffected.
MyD88 Cell Culture and Knockdown Experiments
Whole-genome sequencing identified the A4YD88 L265P variant as the most prevalent mutation in patients with Waldenstrom's macroglobulinemia (WM), a type of non- odgSdirs lymphoma. In 93 to 97% of patients with this disorder, aSlele-specific polymerase-chain-reaction (AS-PCR) assays identified MYD88 L265P, which results from a T→C transversion (Figure 7). Signaling studies showed that the mutant protein that is encoded by A4YD88 L265P triggers tumor growth through the activation of nuclear factor kappa light-chain enhancer of activated B cells (NF-κΒ) by Bruton's tyrosine kinase, (Treon et al., MYD88 Mutations and Response to Ibrutinib in Waldenstrom's Macroglobulinemia, N Engl J Med 2015; 373 :584-586 (2015)). 3GA compound described according to the invention are employed against the MYD88 L265P mutation (Figure 8).
Cells expressing either MyD88 wild-type or MyD88 L265P mutation are seeded in 12-well cluster plates in MEGM media (Lonza), 24 hours prior to transfection. Cells are then transiently transfected with 50 nM of the 3GA compound using Lipofectamine 2000. After 16 hours, total RNA isolation is performed using Qiagen's RNeasy Kit. cDNA synthesis is then performed using Applied Biosystem's High-Capacity cDNA Reverse Transcription Kit. This is followed by quantitative real-time PCR using an assay specific for either wild-type or MyD88 L265P transcripts. Gene expression is calculated using the 2-DDCT method, against an endogenous control. As shown in Figure 9, 3GA oligonucleotides specific for MyD88 L265P can selectively inhibit MyD88 L265P expression while MyD88 wild-type expression was unaffected.
EQUIVALENTS
Those skilled in the art will recognize, or be able to ascertain, using no more than routine experimentation, numerous equivalents to the specific substances and procedures described herein. For example, antisense oligonucleotides that overlap with the oligonucleotides may be used. Such equivalents are considered to be within the scope of this invention, and are covered by the following claims.
While this invention has been particularly shown and described with references to preferred embodiments thereof, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the scope of the invention encompassed by the appended claims.

Claims

1. A compound comprising two or more single- stranded antisense oligonucleotides linked at their 5' ends, wherein the antisense oligonucleotides are complementary to one or more nucleic acid sequences of a target RNA having a point mutation relative to a non- target RNA, wherein the 9th, 10th, and/or 11th nucleotides from the 5' end of an antisense oligonucleotide is aligned opposite of and complementary to at least one point mutation.
2. The compound of claim 1, comprising two antisense oligonucleotides linked at their 5' ends.
3. The compound of claim 1, wherein the sequences of the antisense oligonucleotides are different.
4. The compound of claim 3, wherein the antisense oligonucleotides are complementary to different regions of the same target RNA.
5. The compound of claim 1, wherein the sequences of the antisense oligonucleotides are the same.
6. The compound of claim 1, wherein the antisense oligonucleotides are independently 15 to 40 nucleotides in length.
7. The compound of claim 1, wherein the antisense oligonucleotides are independently 18 to 23 nucleotides in length.
8. The compound of claim 1, wherein the antisense oligonucleotides are 19 nucleotides in length.
9. The compound of any one of claims 1 to 8, wherein the target RNA is expressed from a mutant allele, and the non-target RNA is expressed from a wild-type allele.
10. The compound of claim 9, wherein the point mutation is a gain-of-function mutation.
11. The compound of claim 10, wherein the difference between the target RNA and the wild-type RNA is a single nucleotide.
12. The compound of claim 1, wherein the target RNA is messenger RNA (mRNA) or miRNA.
13. The compound of any one of claims 1 to 12, wherein the antisense oligonucleotides are linked to each other through a nucleotide linkage.
14. The compound of any one of claims 1 to 12, wherein the antisense oligonucleotides are linked to each other through a non-nucleotide linker.
15. The compound of any one of claims 1 to 14, wherein the antisense oligonucleotides comprise one or more modified nucleotides.
16. The compound of claim 15, wherein the modified oligonucleotide has at least one internucleotide linkage selected from the group consisting of alkylphosphonate, phosphorothioate, phosphorodithioate, methylphosphonate, and non-nucleotide linker.
17. The compound according to claim 16, wherein the modified oligonucleotide comprises at least one 2 -O-substituted nucleotide.
18. The compound of claim 17, wherein the 2'-0-substitution is selected from 2'-0- methyl, 2'-0-methoxy, 2'-0-ethoxy, 2'-0-methoxyethyl, 2'-0-alkyl, 2'-0-aryl, and 2'-0- allyl.
19. The compound of any one of claims 1 to 18, wherein the target RNA encodes an oncogene, which is optionally BRAF, or a Ras protein selected from H-Ras, K-Ras, or N- Ras.
20. The compound of any one of claims 1 to 18, wherein the target RNA and non- target encode an enzyme.
21. The compound of claim 20, wherein the target RNA has a point mutation resulting in stronger activity of the encoded enzyme, as compared to the enzyme encoded by the non-target RNA.
22. The compound of claim 21, wherein the enzyme is a kinase.
23. The compound of claim 21, wherein the enzyme is an oxidoreductase, transferase, hydrolase, lyase, isomerase, or Ligase.
24. The compound of claim 23, wherein the enzyme is a hydrolase, which is optionally a triglyceride hydrolase.
25. The compound of any one of claims 1 to 18, wherein the target RNA and non- target RNA encode a transcriptional activator, which is optionally MYD88.
26. A composition comprising a compound of any one of claims 1 to 25 and a physiologically acceptable carrier.
27. A method for inhibiting expression of a target RNA is a cell, the method comprising contacting a cell with a compound according to any one of claims 1 to 25 or a composition according to claim 26.
28. The method of claim 27, wherein the cell expresses the target RNA and the non- target RNA.
29. A method for inhibiting the expression of a target RNA in an organism, the method comprising administering to a mammal the compound according to any one of claims 1 to
25 or a composition according to claim 26.
30. The method of claim 29, wherein the mammal expresses the target RNA and the non-target RNA.
31. The method of claim 30, wherein the mammal is a human.
32. The method according to claim 31, wherein the compound or composition is delivered parenterally.
33. The method of any one of claims 30 to 32, wherein the target RNA encodes an oncogene, which is optionally BRAF, or a Ras protein selected from H-Ras, K-Ras, or N- Ras.
34. The method of any one of claims 30 to 33, wherein the target RNA and non-target encode an enzyme.
35. The method of claim 34, wherein the target RNA has a point mutation resulting in stronger activity of the encoded enzyme, as compared to the enzyme encoded by the non- target RNA.
36. The method of claim 35, wherein the enzyme is a kinase.
37. The method of claim 35, wherein the enzyme is an oxidoreductase, transferase, hydrolase, lyase, isomerase, or ligase.
38. The method of claim 37, wherein the enzyme is a hydrolase, which is optionally a triglyceride hydrolase.
39. The method of any one of claims 30 to 32, wherein the target RNA and non-target RNA encode a transcriptional activator, which is optionally MYD88.
PCT/US2017/034019 2016-05-23 2017-05-23 Compositions and methods for inhibiting target rna expression WO2017205384A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201662340226P 2016-05-23 2016-05-23
US62/340,226 2016-05-23

Publications (1)

Publication Number Publication Date
WO2017205384A1 true WO2017205384A1 (en) 2017-11-30

Family

ID=60412558

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2017/034019 WO2017205384A1 (en) 2016-05-23 2017-05-23 Compositions and methods for inhibiting target rna expression

Country Status (1)

Country Link
WO (1) WO2017205384A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020191177A1 (en) * 2019-03-21 2020-09-24 Sudhir Agrawal Antisense oligonucleotides for allele specificity

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030099959A1 (en) * 1995-04-12 2003-05-29 Kandimalla Ekambar R. Cooperative oligonucleotides
US20060270624A1 (en) * 1991-12-24 2006-11-30 Isis Pharmaceuticals, Inc. Gapped 2' modified oligonucleotides
US20070259827A1 (en) * 2006-01-25 2007-11-08 University Of Massachusetts Compositions and methods for enhancing discriminatory RNA interference
US20110082186A1 (en) * 2009-08-27 2011-04-07 Idera Pharmaceuticals, Inc. Compositions for inhibiting gene expression and uses thereof
US8501703B2 (en) * 2005-08-30 2013-08-06 Isis Pharmaceuticals, Inc. Chimeric oligomeric compounds for modulation of splicing

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060270624A1 (en) * 1991-12-24 2006-11-30 Isis Pharmaceuticals, Inc. Gapped 2' modified oligonucleotides
US20030099959A1 (en) * 1995-04-12 2003-05-29 Kandimalla Ekambar R. Cooperative oligonucleotides
US8501703B2 (en) * 2005-08-30 2013-08-06 Isis Pharmaceuticals, Inc. Chimeric oligomeric compounds for modulation of splicing
US20070259827A1 (en) * 2006-01-25 2007-11-08 University Of Massachusetts Compositions and methods for enhancing discriminatory RNA interference
US20110082186A1 (en) * 2009-08-27 2011-04-07 Idera Pharmaceuticals, Inc. Compositions for inhibiting gene expression and uses thereof

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020191177A1 (en) * 2019-03-21 2020-09-24 Sudhir Agrawal Antisense oligonucleotides for allele specificity

Similar Documents

Publication Publication Date Title
TWI660968B (en) Antagonat compositions and methods of use
JP6280045B2 (en) Method for regulating expression of metastasis associated with metastasis in lung adenocarcinoma 1 (METASTASIS-ASSOCIATED-IN-LUN-ADENOCARCINOMA-TRANSCRIPT-1: MALAT-1)
CA2872304C (en) Organic compositions to treat kras-related diseases
EP3390666A1 (en) Compositions and methods for treatment of kidney diseases
EA035433B1 (en) Modulators of complement factor b
EP3390635A1 (en) Antisense oligomers for treatment of tuberous sclerosis complex
WO2016115490A1 (en) Compounds and methods for modulation of dux4
CA3005254A1 (en) Compositions and methods for treatment of retinitis pigmentosa 18 and retinitis pigmentosa 13
EP3810776A1 (en) Oligonucleotides for modulating scn9a expression
WO2016138278A2 (en) Compositions for inhibiting checkpoint gene expression and uses thereof
US20090136957A1 (en) Methods and compositions for regulating cell cycle progression via the miR-106B family
KR20200128446A (en) Diagnostic markers for treating cell proliferative disorders with telomerase inhibitors
WO2021097437A1 (en) Oligonucleotide interference treatments of prostate cancer
WO2016149618A1 (en) Method for regulation of telomere-length
WO2017205384A1 (en) Compositions and methods for inhibiting target rna expression
TW201119681A (en) Compositions and methods for inhibiting expression of KIF10 genes
US9932585B2 (en) Manipulating microRNA for the management of neurological diseases or conditions and compositions related thereto
US20220372475A1 (en) Inhibitors Of RNA Editing And Uses Thereof
US11149274B2 (en) Methods and compositions for managing vascular conditions
WO2022086935A1 (en) Targeting xist and rna methylation for x reactivation therapy
CN115916978A (en) Oligonucleotides
US20220073932A1 (en) Antisense oligonucleotides for allele specificity
JPWO2007126150A1 (en) Novel therapeutic composition for cancer
EP2689013A1 (en) Compositions for inhibiting gene expression and uses thereof
WO2023196944A2 (en) Oligonucleotide interference treatments of prostate cancer

Legal Events

Date Code Title Description
NENP Non-entry into the national phase

Ref country code: DE

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 17803429

Country of ref document: EP

Kind code of ref document: A1

122 Ep: pct application non-entry in european phase

Ref document number: 17803429

Country of ref document: EP

Kind code of ref document: A1