NZ623476B2 - Modified nucleosides, nucleotides, and nucleic acids, and uses thereof - Google Patents

Modified nucleosides, nucleotides, and nucleic acids, and uses thereof Download PDF

Info

Publication number
NZ623476B2
NZ623476B2 NZ623476A NZ62347612A NZ623476B2 NZ 623476 B2 NZ623476 B2 NZ 623476B2 NZ 623476 A NZ623476 A NZ 623476A NZ 62347612 A NZ62347612 A NZ 62347612A NZ 623476 B2 NZ623476 B2 NZ 623476B2
Authority
NZ
New Zealand
Prior art keywords
optionally substituted
jessb
annotation
independently
alkyl
Prior art date
Application number
NZ623476A
Other versions
NZ623476A (en
Inventor
Stephane Bancel
Fougerolles Antonin De
Paul Hatala
Atanu Roy
Jason P Schrum
Suhaib Siddiqi
Original Assignee
Moderna Therapeutics 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 Moderna Therapeutics Inc filed Critical Moderna Therapeutics Inc
Priority claimed from PCT/US2012/058519 external-priority patent/WO2013052523A1/en
Publication of NZ623476A publication Critical patent/NZ623476A/en
Publication of NZ623476B2 publication Critical patent/NZ623476B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/193Colony stimulating factors [CSF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/0008Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition
    • A61K48/0025Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition wherein the non-active part clearly interacts with the delivered nucleic acid
    • A61K48/0033Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition wherein the non-active part clearly interacts with the delivered nucleic acid the non-active part being non-polymeric
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • A61K48/0066Manipulation of the nucleic acid to modify its expression pattern, e.g. enhance its duration of expression, achieved by the presence of particular introns in the delivered nucleic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/0075Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the delivery route, e.g. oral, subcutaneous
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • A61K9/0021Intradermal administration, e.g. through microneedle arrays, needleless injectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/5123Organic compounds, e.g. fats, sugars
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • C07H21/02Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids with ribosyl as saccharide radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/53Colony-stimulating factor [CSF]
    • C07K14/535Granulocyte CSF; Granulocyte-macrophage CSF
    • 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
    • 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
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/67General methods for enhancing the expression

Abstract

Discloses an isolated mRNA encoding a polypeptide of interest, said isolated mRNA comprising: (a) a sequence of n number of linked nucleosides, (b) a 5’ UTR, (c) a 3’ UTR, and (d) at least one 5? cap structure, wherein said isolated mRNA is fully modified with 1-methylpseudouridine, wherein said isolated mRNA, when administered to peripheral blood mononuclear cells (PBMCs), provides Protein:Cytokine (P:C) ratios of greater than 100 for TNF alpha and greater than 100 for IFNalpha after about eighteen or more hours, and wherein said P:C ratios are higher than those of a corresponding mRNA comprising pseudouridine (?) in place of 1-methylpseudouridine. id isolated mRNA, when administered to peripheral blood mononuclear cells (PBMCs), provides Protein:Cytokine (P:C) ratios of greater than 100 for TNF alpha and greater than 100 for IFNalpha after about eighteen or more hours, and wherein said P:C ratios are higher than those of a corresponding mRNA comprising pseudouridine (?) in place of 1-methylpseudouridine.

Description

MODIFIED NUCLEOSIDES, NUCLEOTIDES, AND NUCLEIC ACIDS, AND USES THEREOF REFERENCE TO THE SEQUENCE LISTING The present ation is being filed along with a Sequence g in electronic format.
The Sequence Listing file, entitled M009SQLST.txt, was created on October 3, 2012 and is 9,859 bytes in size. The information in electronic format of the Sequence Listing is incorporated herein by reference in its ty.
CROSS REFERENCE TO RELATED APPLICATIONS This application claims priority to US. Provisional Patent Application No. 61/542,533, filed October 3, 201 l, entitled Modified Nucleosides, Nucleotides, and Nucleic Acids, and Uses Thereof, the contents of which are incorporated by reference in its entirety.
TECHNICAL FIELD The present disclosure provides compositions and methods using modified nucleic acids to modulate cellular function. The modified nucleic acids of the invention may encode peptides, polypeptides or multiple proteins. The encoded molecules may be used as therapeutics and/or diagnostics.
OUND OF THE INVENTION lly occurring RNAs are synthesized from four basic ribonucleotides: ATP, CTP, UTP and GTP, but may contain post-transcriptionally modified tides. r, approximately one hundred different nucleoside modifications have been identified in RNA (Rozenski, J, Crain, P, and key, J. (1999). The RNA Modification Database: 1999 update. Nucl Acids Res 27: 196- 197). The role of nucleoside modifications on the immune-stimulatory potential and on the translation efficiency ofRNA, however, is r.
There are multiple problems with prior methodologies of effecting protein sion. For example, heterologous DNA introduced into a cell can be inherited by daughter cells (whether or not the heterologous DNA has integrated into the chromosome) or by offspring. Introduced DNA can integrate into host cell genomic DNA at some frequency, resulting in alterations and/or damage to the host cell c DNA. In addition, multiple steps must occur before a protein is made. Once inside the cell, DNA must be transported into the s where it is transcribed into RNA. The RNA ribed from DNA must then enter the cytoplasm where it is translated into protein.
This need for le processing steps creates lag times before the generation of a protein of interest. Further, it is difficult to obtain DNA expression in cells; frequently DNA enters cells but is not expressed or not expressed at reasonable rates or concentrations. This can be a particular problem when DNA is introduced into cells such as primary cells or modified cell lines.
There is a need in the art for biological modalities to address the modulation of intracellular translation of nucleic acids.
SUMMARY OF THE INVENTION [0006a] The invention as claimed herein is described in the following items 1 to 71: 1. An isolated mRNA ng a polypeptide of interest, said isolated mRNA comprising: (a) a sequence of n number of linked sides, (b) a 5’ UTR, (c) a 3’ UTR, and (d) at least one 5′ cap structure, wherein said isolated mRNA is fully modified with 1-methylpseudouridine, n said isolated mRNA, when administered to peripheral blood mononuclear cells ), provides n:Cytokine (P:C) ratios of greater than 100 for TNF-alpha and greater than 100 for pha after about eighteen or more hours, and wherein said P:C ratios are higher than those of a corresponding mRNA comprising pseudouridine (ψ) in place of 1-methylpseudouridine. 2. The isolated mRNA of item 1, further comprising a poly-A tail. 3. The isolated mRNA of item 1 or 2 which is purified. 4. The isolated mRNA of any one of items 1-3, wherein the at least one 5′ cap structure is selected from the group consisting of Cap0, Cap1, ARCA, inosine, N1-methyl-guanosine, 2′fluoroguanosine , 7-deaza-guanosine, 8-oxo-guanosine, 2-amino-guanosine, LNA-guanosine, and 2- azido-guanosine.
. The isolated mRNA of any one of items 1-4, wherein the sequence of n number of linked nucleosides comprises at least one chemical cation of a nucleoside is located in the nucleoside base and/or sugar portion of the side. 8090949_1 (GHMatters) P96706.NZ ation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb 6. The isolated mRNA of item 5, wherein the at least one chemical modification located in a nucleoside base and the side base has the formula: XII-a, XII-b, or XII-c: wherein: denotes a single or double bond; X is O or S; U and W are each independently C or N; V is O, S, C or N; wherein when V is C then R1 is H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, halo, or –ORc, wherein C1-20 alkyl, C2-20 alkenyl, C2-20 alkynyl are each optionally substituted with –OH, , - SH, -C(O)Rc, -C(O)ORc, -NHC(O)Rc, or -NHC(O)ORc; and wherein when V is O, S, or N then R1 is absent; R2 is H, -ORc, -SRc, , or halo; or when V is C then R1 and R2 together with the carbon atoms to which they are attached can form a 5- or 6-membered ring optionally substituted with 1-4 substituents selected from halo, -OH, -SH, -NRaRb, C1-20 alkyl, C2-20 alkenyl, C2-20 alkynyl, C1-20 alkoxy, or C1-20 thioalkyl; R3 is H or C1-20 alkyl; R4 is H or C1-20 alkyl; wherein when denotes a double bond then R4 is absent, or NR4 , taken together, forms a positively charged N substituted with C1-20 alkyl; Ra and Rb are each independently H, C1-20 alkyl, C2-20 alkenyl, C2-20 l, or C6-20 aryl; Rc is H, C1-20 alkyl, C2-20 alkenyl, phenyl, benzyl, a polyethylene glycol group, or an polyethylene glycol group. 7. The isolated mRNA of item 6, n the nucleoside base has the formula: XII-b wherein: [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb ation] jessb ed set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb R3 is C1-20 alkyl. 8. The isolated mRNA of item 7, wherein R3 is C1-4 alkyl. 9. The isolated mRNA of item 7, wherein R3 is CH3.
. The isolated mRNA of any one of items 1-6, wherein the sequence of n number of linked nucleosides does not include pseudouridine (ψ) or 5-methyl-cytidine (m5C). 11. A pharmaceutical composition comprising the isolated mRNA of any one of items 1-10 and a ceutically acceptable excipient. 12. The pharmaceutical composition of item 11, wherein the excipient is selected from a solvent, s t, non-aqueous solvent, dispersion media, diluent, dispersion, sion aid, surface active agent, isotonic agent, ning or emulsifying agent, preservative, lipid, lipidoids liposome, lipid nanoparticle, core-shell nanoparticles, polymer, lipoplex, e, protein, cell, hyaluronidase, and mixtures thereof. 13. Use of the isolated mRNA of any one of items 1-10 in the preparation of a medicament for expressing a polypeptide of interest in a mammalian subject. 14. The use of item 13, wherein the mRNA is formulated.
. The use of item 13, wherein the medicament is ed to be administered at a total daily dose of between 1 ug and 150 ug. 16. The use of item 15, wherein the medicament is designed to be administered by injection. 17. The use of item 15, wherein the medicament is designed to be administered intradermally, subcutaneously, or intramuscularly. 18. The use of item 13, wherein the medicament is designed to produce levels of the polypeptide of interest in the serum of the mammal of at least 50 pg/mL at least two hours after administration. 19. The use of item 18, wherein the medicament is designed to produce levels of the polypeptide of interest in the serum of the mammal of at least 50 pg/mL for at least 72 hours after administration.
[Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb ation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb . The use of item 19, wherein the medicament is designed to produce levels of the polypeptide of interest in the serum of the mammal of at least 60 pg/mL for at least 72 hours after administration. 21. The use of item 13, wherein the medicament is ed to be administered in two or more equal or unequal split doses. 22. The use of item 21 wherein the medicament is designed to produce higher levels of the polypeptide in the mammalian subject by administering split doses than by administering the same total daily dose as a single administration. 23. The use of item 13, wherein the mammalian subject is a human patient in need of an increased level of the polypeptide of interest. 24. The use of item 23, wherein the increased level of the polypeptide of interest is detectable in a bodily fluid of said t.
. The use of item 24, n the bodily fluid is selected from the group ting of peripheral blood, serum, plasma, ascites, urine, cerebrospinal fluid (CSF), sputum, saliva, bone marrow, synovial fluid, aqueous humor, amniotic fluid, cerumen, breast milk, broncheoalveolar lavage fluid, semen, prostatic fluid, cowper's fluid or pre-ejaculatory fluid, sweat, fecal matter, hair, tears, cyst fluid, pleural and peritoneal fluid, rdial fluid, lymph, chyme, chyle, bile, titial fluid, menses, pus, sebum, vomit, vaginal secretions, mucosal secretion, stool water, pancreatic juice, lavage fluids from sinus cavities, bronchopulmonary aspirates, blastocyl cavity fluid, and umbilical cord blood. 26. The use of item 23, n the medicament is designed to be administered according to a dosing regimen which occurs over the course of hours, days, weeks, months, or years. 27. The use of item 16, wherein ion is achieved by using one or more devices selected from multi-needle injection s, catheter or lumen systems, and ultrasound, electrical or radiation based systems. 28. The use of item 21, wherein the amount of mRNA administered in any dose is substantially equal. 29. The use of item 21, wherein the medicament is designed for administration of a first dose, a second dose or any of a plurality of doses at substantially the same time.
[Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb ionNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb . The use of item 13, n the medicament is designed to be administered as a single unit dose between about 10 mg/kg and about 500 mg/kg. 31. The use of item 13, wherein the medicament is designed to be administered as a single unit dose between about 1.0 mg/kg and about 10 mg/kg. 32. The use of item 13, wherein the medicament is designed to be administered as a single unit dose between about 0.001 mg/kg and about 1.0 mg/kg. 33. The isolated mRNA of any one of items 1-10, n the sequence of n number of linked nucleosides comprises at least one chemical modification, and the chemical modification includes ing or substituting an atom of a pyrimidine nucleobase with an amine, an SH, a methyl or ethyl, or a chloro or fluoro. 34 The isolated mRNA of any one of items 1-10, wherein the sequence of n number of linked nucleosides comprise the a (Ia): Y1 Y5 B R3 R4 R5 R1' R1" R2" m " Y2 R2'm' Y3 P (Ia), or a pharmaceutically acceptable salt or isomer thereof, U is O, S, N(RU)nu, or C(RU)nu, wherein nu is an integer from 0 to 2 and each RU is, independently, H, halo, or optionally substituted alkyl; is a single or double bond; is a single bond or absent; each of R1’, R2’, R1”, R2”, R1, R2, R3, R4, and R5, if present, is, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally tuted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted lkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, ally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, or absent; wherein the combination of R3 with one or more of R1’, R1”, R2’, R2”, or R5 can join together to form optionally substituted alkylene or optionally substituted heteroalkylene and, taken together with the carbons to which they are attached, provide an ally substituted heterocyclyl; wherein the combination of R5 with one [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb ation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb or more of R1’, R1”, R2’, or R2” can join together to form optionally substituted alkylene or optionally substituted heteroalkylene and, taken together with the carbons to which they are ed, provide an optionally substituted heterocyclyl; and n the combination of R4 and one or more of R1’, R1”, R2’, R2”, R3, or R5 can join together to form optionally substituted alkylene or optionally substituted alkylene and, taken together with the carbons to which they are attached, provide an optionally substituted heterocyclyl; each of m’ and m” is, independently, an integer from 0 to 3; each of Y1, Y2, and Y3, is, independently, O, S, Se, -NRN1-, optionally substituted alkylene, or optionally tuted heteroalkylene, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, or absent; each Y4 is, independently, H, hydroxy, thiol, boranyl, optionally substituted alkyl, optionally substituted l, optionally substituted alkynyl, optionally substituted alkoxy, ally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted koxy, optionally substituted alkoxyalkoxy, or ally substituted amino; each Y5 is, ndently, O, S, Se, optionally substituted alkylene, or optionally substituted heteroalkylene; n is an integer from 1 to 0; and B is a nucleobase.
. The isolated mRNA of item 34, wherein B is not pseudouridine (ψ) or 5-methyl-cytidine (m5C). 36. The ed mRNA of item 34 or 35, wherein: U is O or C(RU)nu, wherein nu is an integer from 1 to 2 and each RU is, independently, H, halo, or optionally substituted alkyl; each of R1, R1’, R1”, R2, R2′, and R2”, if present, is, independently, H, halo, hydroxy, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl; each of R3 and R4 is, independently, H, halo, hydroxy, optionally substituted alkyl, or optionally substituted alkoxyalkoxy; each of Y1, Y2, and Y3, is, independently, O, S, Se, -NRN1-, optionally substituted ne, or optionally substituted heteroalkylene, wherein RN1 is H, optionally substituted alkyl, optionally substituted l, or optionally substituted alkynyl; each Y4 is, independently, H, hydroxy, thiol, boranyl, optionally tuted alkyl, optionally tuted alkenyl, optionally substituted alkynyl, optionally tuted alkoxy, [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb ation] jessb MigrationNone set by jessb ation] jessb ed set by jessb [Annotation] jessb None set by jessb [Annotation] jessb ionNone set by jessb [Annotation] jessb Unmarked set by jessb optionally substituted alkenyloxy, optionally tuted alkynyloxy, optionally substituted thioalkoxy, or optionally substituted amino; each Y5 is, independently, O or optionally substituted alkylene; and n is an integer from 10 to 10,000. 37. The isolated mRNA of item 36, wherein each of R1, R1’, and R1”, if present, is H. 38. The isolated mRNA of item 37, wherein each of R 2, R2′, and R2”, if present, is, independently, H, halo, hydroxy, optionally substituted , or optionally substituted alkoxyalkoxy. 39. The isolated mRNA of item 38, wherein each of R2, R2′, and R2”, if present, is H. 40. The isolated mRNA of item 39, wherein each of R 1, R1’, and R1”, if present, is, independently, H, halo, hydroxy, optionally substituted alkoxy, or optionally substituted alkoxyalkoxy. 41. The isolated mRNA of item 34, wherein the sequence of n number of linked nucleotides comprise the Formula (IIa): Y1 Y5 B R5 R1 R3 R4 3 PY (IIa), or a pharmaceutically acceptable salt or stereoisomer thereof. 42. The isolated mRNA of item 41, wherein the sequence of n number of linked sides comprise the Formula (IIb) or (IIc): Y1 Y5 B B U Y1 Y5 U R5 R1 R5 R1 R4 R3 R4 R2 R2 Y2 Y2 3 PY 3 PY Y4 Y4 (IIb), or (IIc), or a pharmaceutically acceptable salt thereof. ation] jessb None set by jessb [Annotation] jessb ionNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb 43. The isolated mRNA of item 42, wherein the sequence of n number of linked nucleosides comprise the Formula (IIb-1), (IIb-2), or (IIc-1)-(IIc-4): Y 1 Y5 B U Y1 Y5 B R2' R2 Y2 Y2 3 PY 3 PY Y4 Y4 (IIb-1), (IIb-2), Y1 Y5 B U Y1 Y5 B R1 R1 R2 R2 Y2 Y2 3 PY 3 PY Y4 Y4 (IIc-1), ), Y1 Y5 B U Y1 Y5 B R3 R3 R2 R2 Y2 Y2 3 PY 3 PY Y4 Y4 (IIc-3), or (IIc-4), or a pharmaceutically acceptable salt thereof. 44. The isolated mRNA of item 34, wherein the sequence of n number of linked nucleosides se the Formula (IId): Y1 Y5 B R5 R1 R3 R4 3 PY (IId), or a pharmaceutically acceptable salt or stereoisomer thereof.
[Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb 45. The isolated mRNA of item 43, wherein the sequence of n number of linked sides comprise the Formula (IIe) or (IIf): Y1 Y5 B Y1 Y5 B U U R5 R1 R5 R1 R3 R4 R3 R4 R2 R2 Y2 Y2 3 PY 3 PY Y4 Y4 (IIe) or (IIf), or a ceutically acceptable salt thereof. 46. The isolated mRNA of item 34, wherein each of said linked nucleotides independently have one of Formulas (IIg)-(IIj): Y1 Y5 B Y1 Y5 B U U R3 R4 R3 R4 R5 R1' R1" R5 R1' R1" R2" R2" Y2 R2' Y2 R2' Y3 P Y3 P Y4 Y4 (IIg), (IIh), Y1 Y5 B3 Y1 Y5 B3 U U R3 Rb3 R3 Rb3 R5 B1 B2 R5 B1 B2 Rb2 Rb2 Y2 Rb1 Y2 Rb1 Y3 P Y3 P Y4 Y4 (IIi), or (IIj),or a pharmaceutically acceptable salt or isomer thereof.
[Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb ionNone set by jessb [Annotation] jessb Unmarked set by jessb 47. The isolated mRNA of item 34, wherein the sequence of n number of linked nucleosides comprise the Formula (IIk): Y1 Y5 B R5 R1' R3 R4 Y2 m 3 PY (IIk) , or a pharmaceutically acceptable salt or stereoisomer thereof. 48. The isolated mRNA of item 47, wherein the sequence of n number of linked nucleosides se the Formula (IIl): Y1 Y5 R3 R4 Y3 P (IIl), or a pharmaceutically acceptable salt or stereoisomer f. 49. The isolated mRNA of item 47, wherein the sequence of n number of linked nucleosides comprise the Formula (IIm): Y1 Y5 B R3 R4 R5 R1' R1" Y2 R2' Y3 P (IIm), or a pharmaceutically acceptable salt or isomer thereof, wherein each of R1’, R1”, R2′, and R2” is, independently, H, halo, hydroxy, optionally tuted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally tuted alkynyloxy, optionally substituted aminoalkoxy, ally substituted alkoxyalkoxy, or absent; and wherein the combination of R2′ and R3 or the combination of R2” and R3 can be taken together to form optionally substituted alkylene or optionally substituted heteroalkylene.
[Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb ation] jessb Unmarked set by jessb 50. The isolated mRNA of any one of items 37-49, wherein U is O or C(RU)nu, wherein nu is an integer from 1 to 2 and each RU is, independently, H, halo, or optionally substituted alkyl; each of R1 and R2 is, independently, H, halo, hydroxy, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxy, optionally substituted amino, azido, ally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl; each of R3 and R4 is, independently, H or optionally substituted alkyl; each of Y1, Y2, and Y3, is, independently, O, S, Se, -NRN1-, ally substituted ne, or ally substituted heteroalkylene, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, or optionally substituted alkynyl; each Y4 is, independently, H, hydroxy, thiol, boranyl, optionally substituted alkyl, optionally substituted alkenyl, optionally tuted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted thioalkoxy, or optionally substituted amino; each Y5 is, independently, O or optionally tuted alkylene; and n is an r from 10 to 10,000. 51. The isolated mRNA of item 34, n the sequence of n number of linked sides comprise the a (IIn): Y1 Y5 B R3 R4 3 PY (IIn), or a ceutically acceptable salt or stereoisomer thereof, wherein U is O or C(RU)nu, wherein nu is an integer from 1 to 2 and each RU is, independently, H, halo, or optionally substituted alkyl; each of R1 and R4 is, independently, H, halo, hydroxy, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl; [Annotation] jessb None set by jessb ation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb R3′ is O, S, or , n RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl; R3” is optionally substituted alkylene or optionally substituted heteroalkylene; each of Y1, Y2, and Y3, is, independently, O, S, Se, -NRN1-, optionally substituted alkylene, or optionally substituted heteroalkylene, wherein RN1 is H, optionally substituted alkyl, optionally substituted l, or optionally substituted alkynyl; each Y4 is, independently, H, hydroxy, thiol, boranyl, optionally substituted alkyl, optionally tuted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted koxy, or optionally substituted amino; each Y5 is, independently, O, S, optionally substituted alkylene (e.g., methylene), or optionally substituted heteroalkylene; and n is an integer from 10 to 10,000. 52. The isolated mRNA of item 34, wherein the sequence of n number of linked sides comprise Formula (IIn-1) or ): Y1 Y5 B U Y1 Y5 B R3" R3" R3' O Y2 Y2 3 PY 3 PY Y4 Y4 (IIn-1) or (IIn-2), or a pharmaceutically acceptable salt or stereoisomer thereof. 53. The isolated mRNA of any one of items 34-52, wherein each B independently has a formula selected from a (b1)-(b5): T1' T1" R12c R12c R12c R12a R10 R10 V1 N R12a N N N N V2 T2" T2" N N R11 R11 N N O T2' O (b1), (b2), (b3), (b4), or R10 R12c N O (b5), or a ceutically able salt or stereoisomer thereof, [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb ation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb wherein is a single or double bond; each of T1’, T1”, T2′, and T2” is, independently, H, optionally substituted alkyl, optionally substituted alkoxy, or optionally substituted thioalkoxy, or the combination of T1’ and T1” or the combination of T2′ and T2” join together (e.g., as in T2) to form O (oxo), S (thio), or Se o); each of V1 and V2 is, independently, O, S, N(RVb)nv, or C(RVb)nv, wherein nv is an integer from 0 to 2 and each RVb is, independently, H, halo, optionally substituted amino acid, optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted alkenyl, optionally substituted l, optionally substituted , optionally substituted alkenyloxy, optionally substituted loxy, optionally substituted yalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted acylaminoalkyl, optionally substituted alkoxycarbonylalkyl, ally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, or ally substituted carbonylalkoxy; R10 is H, halo, optionally substituted amino acid, hydroxy, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aminoalkyl, optionally substituted hydroxyalkyl, optionally tuted hydroxyalkenyl, optionally substituted hydroxyalkynyl, ally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted alkoxy, optionally substituted carbonylalkyl, ally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally tuted alkoxycarbonylalkoxy, optionally substituted carboxyalkoxy, ally substituted carboxyalkyl, or optionally substituted carbamoylalkyl; R11 is H or optionally substituted alkyl; R12a is H, optionally substituted alkyl, ally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl, optionally substituted carboxyalkyl, optionally substituted carboxyalkoxy, optionally substituted carboxyaminoalkyl, or optionally substituted carbamoylalkyl; and R12c is H, halo, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted thioalkoxy, optionally substituted amino, optionally substituted yalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, ally substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl. 54. The isolated mRNA of any one of items 34-53, wherein B ses Formula (b6)-(b9): [Annotation] jessb None set by jessb [Annotation] jessb ionNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb ation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb R12c R12c T1' T1" R12c R12a R12a R12b R12a V3 N V3 N R12b N N N N W1 T2'' W1 W2 W2 T2 T2" T2" T2' T2' T2' (b6), (b7), (b8), or (b9), or a pharmaceutically acceptable salt or isomer thereof, wherein is a single or double bond; each of T1’, T1”, T2′, and T2” is, independently, H, optionally substituted alkyl, optionally substituted alkoxy, or ally substituted thioalkoxy, or the combination of T1’ and T1” join together or the combination of T2′ and T2” join together to form O (oxo), S (thio), or Se (seleno), or each T1 and T2 is, independently, O (oxo), S (thio), or Se (seleno); each of W1 and W2 is, independently, N(RWa)nw or C(RWa)nw, n nw is an integer from 0 to 2 and each RWa is, independently, H, optionally substituted alkyl, or optionally substituted alkoxy; each V3 is, independently, O, S, N(RVa)nv, or C(RVa)nv, wherein nv is an integer from 0 to 2 and each RVa is, independently, H, halo, optionally substituted amino acid, optionally substituted alkyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted alkenyl, ally substituted alkynyl, optionally substituted heterocyclyl, optionally substituted alkheterocyclyl, optionally substituted alkoxy, optionally substituted loxy, or optionally substituted alkynyloxy, optionally tuted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted acylaminoalkyl, optionally tuted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylacyl, ally substituted alkoxycarbonylalkoxy, optionally substituted carboxyalkyl, optionally tuted carboxyalkoxy, optionally substituted carboxyaminoalkyl, or optionally substituted carbamoylalkyl, and wherein RVa and R12c taken er with the carbon atoms to which they are attached can form optionally substituted cycloalkyl, optionally substituted aryl, or optionally substituted heterocyclyl; R12a is H, ally substituted alkyl, optionally tuted hydroxyalkyl, ally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, ally substituted carboxyalkyl, optionally substituted carboxyalkoxy, optionally substituted carboxyaminoalkyl, optionally substituted carbamoylalkyl, or absent; R12b is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally tuted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, [Annotation] jessb None set by jessb [Annotation] jessb ionNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb ionNone set by jessb [Annotation] jessb ed set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb optionally substituted aminoalkynyl, ally tuted alkaryl, optionally substituted heterocyclyl, optionally tuted alkheterocyclyl, optionally substituted amino acid, optionally substituted alkoxycarbonylacyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted alkoxycarbonylalkyl, ally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted carboxyalkyl, optionally substituted carboxyalkoxy, optionally substituted carboxyaminoalkyl, or optionally substituted carbamoylalkyl, wherein the combination of R12b and T1’ or the combination of R12b and R12c can join together to form optionally substituted heterocyclyl; and R12c is H, halo, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted thioalkoxy, optionally substituted amino, optionally substituted lkyl, optionally substituted lkenyl, or optionally substituted aminoalkynyl. 55. The isolated mRNA of item 54, wherein R12a, R12b, R12c, or RVa is substituted with - (CH2)s2(OCH2CH2)s1(CH2)s3OR’, wherein s1 is an integer from 1 to 10, each of s2 and s3, independently, is an r from 0 to 10, and R’ is H or C1-20 alkyl); or - NRN1(CH2)s2(CH2CH2O)s1(CH2)s3NRN1, wherein s1 is an integer from 1 to 10, each of s2 and s3, independently, is an integer from 0 to 10, and each RN1 is, independently, hydrogen or optionally substituted C1-6 alkyl. 56. The isolated mRNA of claim 54, wherein B comprises Formula (b28)-(b31): T1 T1 T1 RVb' R12a RVb' R12a R12b R12a N N N N RVb" N T2 N T2 T2 (b28), (b29), (b30), or RVb' R12a N T2 (b31), or a pharmaceutically acceptable salt or stereoisomer thereof. 57. The isolated mRNA of any one of items 34-56, wherein B comprises Formula (b10)-(b14): [Annotation] jessb None set by jessb ation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb ation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb R13a R13b R13b R13a R13b R13a R13b N N N N R14 R14 R16 V5 N N N V4 N R15 N T3" R15 N T3" R15 N T3" R15 T3" T3' T3' T3' T3' (b10), (b11), (b12), V4 N R15 N T3" (b13), or (b14), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each of T3′ and T3” is, independently, H, optionally substituted alkyl, optionally substituted alkoxy, or optionally substituted thioalkoxy, or the combination of T3′ and T3” join together to form O (oxo), S (thio), or Se (seleno); each V4 is, independently, O, S, N(RVc)nv, or C(RVc)nv, wherein nv is an r from 0 to 2 and each RVc is, independently, H, halo, optionally substituted amino acid, optionally substituted alkyl, ally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, ally tuted alkenyloxy, optionally substituted heterocyclyl, optionally substituted alkheterocyclyl, or optionally substituted alkynyloxy, wherein the combination of R13b and RVc can be taken together to form optionally substituted heterocyclyl; each V5 is, independently, N(RVd)nv, or C(RVd)nv, wherein nv is an integer from 0 to 2 and each RVd is, independently, H, halo, optionally substituted amino acid, ally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted , optionally substituted alkenyloxy, optionally substituted heterocyclyl, optionally substituted alkheterocyclyl, or optionally substituted alkynyloxy; each of R13a and R13b is, independently, H, optionally substituted acyl, optionally substituted acyloxyalkyl, optionally substituted alkyl, or optionally substituted alkoxy, wherein the combination of R13b and R14 can be taken together to form optionally substituted heterocyclyl; each R14 is, independently, H, halo, hydroxy, thiol, optionally substituted acyl, optionally substituted amino acid, optionally tuted alkyl, ally substituted kyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl, ally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted alkoxy, optionally tuted alkenyloxy, ally substituted loxy, optionally substituted aminoalkoxy, ally substituted alkoxyalkoxy, optionally substituted acyloxyalkyl, optionally substituted amino, azido, optionally substituted aryl, optionally substituted heterocyclyl, optionally tuted alkheterocyclyl, optionally tuted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl; and [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb ionNone set by jessb [Annotation] jessb Unmarked set by jessb each of R15 and R16 is, independently, H, optionally substituted alkyl, optionally substituted alkenyl, or optionally substituted alkynyl. 58. The isolated mRNA of item 57, wherein B comprises a (b36): R13a R13b R13b R13a R13b N N T1 N R14 R14 R16 R14 R14 N N N N R13a R15 N T3 R15 N T3 N R15 N (b32), (b33), R13b (b34), R13b R14a R15 N R14b (b35), or (b36) or a pharmaceutically acceptable salt or stereoisomer thereof. 59. The isolated mRNA of any one of items 34-58, wherein B comprises Formula (b15)-(b17): T4' T4" T5' T5" R23 V5 R18 N N N V6 R21 R24 N R19a N N N N N R19b (b15), R22 (b16), or T5' T5" N R18 N N T6' R22 (b17) or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each of T4’, T4”, T5′, T5”, T6’, and T6” is, independently, H, optionally tuted alkyl, or optionally substituted alkoxy, and wherein the combination of T4’ and T4” or the combination of T5′ and T5” or the combination of T6’ and T6” join together form O (oxo), S , or Se (seleno); each of V5 and V6 is, ndently, O, S, N(RVd)nv, or nv, wherein nv is an integer from 0 to 2 and each RVd is, independently, H, halo, thiol, optionally substituted amino acid, cyano, amidine, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, ally substituted aminoalkynyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted loxy, optionally substituted thioalkoxy, or optionally substituted amino; and [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb ed set by jessb [Annotation] jessb None set by jessb [Annotation] jessb ionNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb ionNone set by jessb [Annotation] jessb Unmarked set by jessb each of R17, R18, R19a, R19b, R21, R22, R23, and R24 is, independently, H, halo, thiol, ally substituted alkyl, ally substituted alkenyl, optionally substituted alkynyl, optionally substituted thioalkoxy, optionally substituted amino, or optionally substituted amino acid. 60. The isolated mRNA of item 59, wherein B comprises Formula (b37)-(b40): T4 T4' T4 N R18 N R18 N N N N R19a R19a R19a N N N N N N N N R19b (b37), R19b (b38), R19b (b39), or N R18 N R19a N N R19b (b40), ), or a pharmaceutically acceptable salt or stereoisomer thereof. 61. The isolated mRNA of any one of items 34-58, wherein B comprises Formula (b18)-(b20): R26a R26b R26b N N V7 V7 R28 N N R25 R25 N N R27 N N R27 (b18), (b19), or N N R27 (b20), or a pharmaceutically acceptable salt or stereoisomer thereof wherein each V7 is, independently, O, S, N(RVe)nv, or C(RVe)nv, wherein nv is an integer from 0 to 2 and each RVe is, independently, H, halo, optionally substituted amino acid, optionally substituted alkyl, ally substituted alkenyl, optionally substituted alkynyl, ally substituted alkoxy, optionally substituted alkenyloxy, or ally substituted alkynyloxy; each R25 is, independently, H, halo, thiol, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted thioalkoxy, or optionally substituted amino; [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb each of R26a and R26b is, independently, H, optionally substituted acyl, optionally substituted amino acid, optionally tuted oylalkyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted alkoxy, or polyethylene glycol group, or an amino-polyethylene glycol group; each R27 is, independently, H, ally substituted alkyl, optionally substituted alkenyl, optionally substituted l, optionally substituted alkoxy, optionally substituted thioalkoxy, or optionally substituted amino; each R28 is, independently, H, optionally tuted alkyl, optionally substituted alkenyl, or optionally substituted alkynyl; and each R29 is, independently, H, optionally substituted acyl, optionally substituted amino acid, optionally substituted carbamoylalkyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl, ally substituted hydroxyalkenyl, optionally substituted alkoxy, or ally substituted amino. 62. The isolated mRNA of item 61, wherein B comprises Formula (b41)-(b43): R26a R26b R26a R26b R26a R26b N N N N N N N N N N N R27 N N N N (b41), (b42), or (b43), or a pharmaceutically acceptable salt or stereoisomer thereof. 63. The ed mRNA of item 61, wherein R26a, R26b, or R29 is substituted with -(CH2)s2(OCH2CH2)s1(CH2)s3OR’, wherein s1 is an integer from 1 to 10, each of s2 and s3, independently, is an integer from 0 to 10, and R’ is H or C1-20 alkyl); or CH2)s2(CH2CH2O)s1(CH2)s3NRN1, wherein s1 is an integer from 1 to 10, each of s2 and s3, independently, is an integer from 0 to 10, and each RN1 is, ndently, hydrogen or optionally tuted C1-6 alkyl. 64. The isolated mRNA of any one of items 34-63, n B comprises Formula (b21): xaX12 R12a N N (b21), or a pharmaceutically acceptable salt or stereoisomer thereof, [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb ation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb wherein X12 is, independently, O, S, optionally substituted alkylene, or optionally substituted alkylene; xa is an integer from 0 to 3; R12a is H, optionally substituted alkyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, or absent; and T2 is O, S, or Se. 65. The isolated mRNA of any one of items 34-64, wherein B comprises Formula (b22): O T1 R10' R12a N N R11 N T2 (b22), or a ceutically acceptable salt or stereoisomer thereof, wherein R10’ is, independently, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted l, optionally substituted aryl, optionally substituted heterocyclyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, ally substituted aminoalkynyl, optionally substituted alkoxy, optionally substituted carbonylalkyl, optionally tuted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted carboxyalkoxy, optionally substituted carboxyalkyl, or optionally tuted carbamoylalkyl; R11 is H or optionally substituted alkyl; R12a is H, optionally substituted alkyl, optionally substituted aminoalkyl, optionally tuted aminoalkenyl, optionally substituted lkynyl, or absent; and each of T1 and T2 is, independently, O, S, or Se. 66. The isolated mRNA of any one of items 34-65, n B comprises Formula (b23): R10 R12a R11 N T2 (b23), wherein R10 is optionally substituted heterocyclyl or optionally substituted aryl; R11 is H or optionally substituted alkyl; R12a is H, optionally substituted alkyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, or absent; and each of T1 and T2 is, independently, O, S, or Se. 67. The isolated mRNA of any one of items 34-66, n B comprises Formula (b24) or (b25): [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb ionNone set by jessb [Annotation] jessb ed set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb R13a R13b R13a R13b O N O N R14' N N R14' N N H H R15 N T3 R15 N T3 (b24) or (b25), wherein T3 is O, S, or Se; each of R13a and R13b is, independently, H, optionally substituted acyl, optionally substituted alkyl, or optionally substituted alkoxy, n the combination of R13b and R14 can be taken together to form optionally substituted heterocyclyl; R14’ is, independently, optionally substituted alkyl, optionally substituted alkenyl, ally substituted alkynyl, optionally substituted aryl, optionally substituted heterocyclyl, optionally substituted alkheterocyclyl, optionally substituted alkaryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynynyl, optionally substituted alkoxy, optionally substituted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally tuted carbonylalkynyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted carboxyalkoxy, optionally substituted carboxyalkyl, or optionally substituted oylalkyl; and each R15 is, ndently, H, optionally substituted alkyl, optionally substituted alkenyl, or optionally substituted alkynyl. 68. The isolated mRNA of any one of items 34-67, wherein B comprises Formula (b26) or (b27): (b26) or (b27). 69. The ed mRNA of any one of items 34-68, wherein B ses Formula (b43). 70. The isolated mRNA of claim 34, wherein said isolated mRNA is prepared from one or more building blocks selected from BB-1 to BB-274, or a pharmaceutically acceptable salts or stereoisomers thereof. 71. The isolated mRNA of item 34, wherein said isolated mRNA is ed from one or more building blocks selected from compounds 1-50, or pharmaceutically acceptable salts or stereoisomers thereof.
The present disclosure es, inter alia, modified nucleosides, modified nucleotides, and modified c acids which can exhibit a reduced innate immune response when introduced into a population of cells, both in vivo and ex vivo.
The present invention provides polynucleotides which may be isolated or purified.
These polynucleotides may encode one or more polypeptides of interest and comprise a sequence of n number of linked sides or nucleotides comprising at least one modified nucleoside or nucleotide as compared to the chemical structure of an A, G, U or C nucleoside or nucleotide.
The polynucleotides may also contain a 5’ UTR comprising at least one Kozak sequence, a 3’ UTR, and at least one 5′ cap structure. The isolated polynucleotides may further contain a poly-A tail and may be purified.
The isolated polynucleotides of the invention also comprise at least one 5′ cap structure selected from the group consisting of Cap0, Cap1, ARCA, inosine, N1-methyl- guanosine, 2′fluoro-guanosine, 7-deaza-guanosine, 8-oxo-guanosine, 2-amino-guanosine, LNA- guanosine, and 2-azido-guanosine.
Modifications of the polynucleotides of the invention may be on the nucleoside base and/or sugar portion of the nucleosides which comprise the polynucleotide.
In some ments, the modification is on the base and is ed from the group consisting of pseudouridine or N1-methylpseudouridine.
In some embodiments, the modified side is not pseudouridine (ψ) or 5-methylcytidine (m5C).
In some embodiments, multiple modifications are included in the modified nucleic acid or in one or more individual nucleoside or nucleotide. For e, modifications to a nucleoside may include one or more modifications to the nucleobase and the sugar.
In some embodiments are provided novel building blocks, e.g., nucleosides and nucleotides for the preparation of modified cleotides and their method of synthesis and manufacture.
The present invention also provides for pharmaceutical compositions comprising the modified polynucleotides described herein. These may also further include one or more pharmaceutically acceptable excipients selected from a solvent, aqueous solvent, non-aqueous t, dispersion media, diluent, dispersion, sion aid, surface active agent, isotonic agent, thickening or emulsifying agent, preservative, lipid, lipidoids liposome, lipid nanoparticle, core-shell nanoparticles, polymer, lipoplexe peptide, protein, cell, hyaluronidase, and mixtures f.
Methods of using the polynucleotides and modified nucleic acids of the invention are also provided. In this instance, the poynucleotides may be formulated by any means known in the art or administered Via any of several routes including injection by intradermal, subcutaneous or intramuscular means.
Administration of the modified nucleic acids of the inventin may be Via two or more equal or unequal split doses. In some embodiments, the level of the polypeptide produced by the subject by administering split doses of the polynucleotide is r than the levels ed by administering the same total daily dose of polynucleotide as a single administration.
Detection of the d nucleic acids or the encoded polypeptides may be performed in the bodily fluid of the subject or patient where the bodily fluid is selected from the group consisting ofperipheral blood, serum, plasma, ascites, urine, cerebrospinal fluid (CSF), sputum, saliva, bone marrow, al fluid, aqueous humor, ic fluid, cerumen, breast milk, broncheoalveolar lavage fluid, semen, prostatic fluid, 's fluid or pre—ejaculatory fluid, sweat, fecal matter, hair, tears, cyst fluid, pleural and peritoneal fluid, pericardial fluid, lymph, chyme, chyle, bile, interstitial fluid, menses, pus, sebum, vomit, vaginal secretions, mucosal secretion, stool water, pancreatic juice, lavage fluids from sinus cavities, opulmonary aspirates, blastocyl cavity fluid, and umbilical cord blood.
In some embodiments, administration is according to a dosing n which occurs over the course of hours, days, weeks, months, or years and may be achieved by using one or more devices selected from multi-needle injection systems, catheter or lumen systems, and ound, electrical or radiation based systems.
Unless otherwise defined, all cal and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs.
Methods and materials are described herein for use in the present disclosure; other, le methods and materials known in the art can also be used. The materials, s, and examples are illustrative only and not intended to be limiting. All publications, patent applications, patents, sequences, database entries, and other references mentioned herein are incorporated by reference in their ty. In case of conflict, the present specification, including definitions, will control.
Other features and advantages of the present disclosure will be apparent from the following detailed description and figures, and from the claims.
BRIEF DESCRIPTION OF THE DRAWINGS The foregoing and other s, features and advantages will be apparent from the following description of particular ments of the invention, as illustrated in the accompanying drawings in which like reference characters refer to the same parts throughout the ent Views.
The drawings are not necessarily to scale, emphasis instead being placed upon illustrating the principles of various embodiments of the invention. provides the spectrum and graphs of the analytical results for N4-Me-CTP (NTP of compound I). Figure 1A es the nuclear magnetic resonance (NMR) spectrum in DMSO and Figure 1B provides the NMR spectrum in D20, Figure 1C provides the mass spectrometry (MS) results, and Figure 1D is the high mance liquid chromatography (HPLC) results for N4— methylcytidine (N4—Me-cytidine, compound 1). shows the HPLC s for N4—Me-CTP (NTP of compound 1). provides the analytical results for 2’-OMe-N, N—di—Me-CTP (NTP of compound 2).
Figure 3A provides the NMR spectrum. Figure 3B provides the MS results. Figure 3C provides HPLC s for 2’-O-methyl—N4, N4-dimethylcytidine (2’-OMe—N,N—di—Me-cytidjne, compound 2). shows the HPLC results for 2’-OMe—N, N—di-Mc—CTP (NTP of compound 2). provides the HPLC results for 5-methoxycarbonylmethoxy-UTP (NTP of compound 3). provides the analytical results of 3-methyl pseudouridine und 4). Figure 6A provides the NIVIR um of 3-methyl pseudouridine (compound 4) and Figure 6B provides the HPLC results for yl pseudouridine (compound 4). provides the analytical s of S-TBDMS-OCHz-cytidine (compound 6). Figure 7A provide the NIVIR spectrum, Figure 7B provides the MS results, and Figure 7C provides the HPLC results for S-TBDMS-OCHz-cytidine (compound 6). es the analytical results of uoromethyl uridine (compound 8). Figure 8A provides the NMR spectrum, Figure 8B provides MS results, and Figure 8C provides HPLC results for 5-trifluoromethyl uridine (compound 8). provides the NMR spectrum results for of 5-(methoxycarbonyl) methyl uridine (compound 9). provides a graph g the variability of protein (GCSF; line B) and cytokjne (interferon-alpha (IFNa); line A and tumor necrosis factor-alpha (TNFa); line C) expression as function of percent modification.
DETAILED DESCRIPTION The present disclosure es, inter alia, d nucleosides, modified nucleotides, and modified nucleic acids that exhibit improved therapeutic properties including, but not limited to, a reduced innate immune response when introduced into a population of cells.
As there remains a need in the art for therapeutic modalities to address the myriad of barriers nding the efficacious tion of intracellular translation and processing ofnucleic acids encoding ptides or fragments thereof, the inventors have shown that certain modified mRNA sequences have the potential as therapeutics with benefits beyond just evading, avoiding or diminishing the immune response.
The present invention addresses this need by providing nucleic acid based compounds or polynucleotides which encode a polypeptide of interest (e. g., modified mRNA) and which have structural and/or chemical features that avoid one or more of the problems in the art, for example, features which are useful for optimizing nucleic acid-based therapeutics while retaining structural and fimctional integrity, overcoming the threshold of expression, improving expression rates, half life and/or protein concentrations, optimizing protein localization, and avoiding deleterious bio- ses such as the immune response and/or ation pathways.
Provided herein, in part, are cleotides encoding polypeptides of interest which have been chemically modified to improve one or more of the ity and/or clearance in tissues, or uptake and/or kinetics, cellular access by the compositions, engagement with translational machinery, mRNA half-life, translation efficiency, immune evasion, protein tion capacity, secretion efficiency (when applicable), accessibility to circulation, protein ife andfor tion of a cell’s status, function and/or activity.
The modified nucleosides, nucleotides and c acids of the invention, including the combination of modifications taught herein have superior properties making them more suitable as therapeutic modalities.
It has been determined that the “all or none” model in the art is sorely insufficient to describe the ical phenomena associated with the therapeutic utility of modified mRNA. The present inventors have determined that to improve protein tion, one may consider the nature of the modification, or combination of modifications, the percent ation and survey more than one cytokine or metric to determine the efficacy and risk profile of a ular modified mRNA.
In one aspect of the invention, methods of determining the effectiveness of a modified mRNA as compared to unmodified involves the measure and analysis of one or more cytokines whose expression is triggered by the administration of the exogenous c acid of the invention.
These values are compared to administration of an umodified nucleic acid or to a standard metric such as cytokine se, PolyIC, R-848 or other standard known in the art.
One example of a standard metric developed herein is the e of the ratio of the level or amount of encoded polypeptide (protein) produced in the cell, tissue or organism to the level or amount of one or more (or a panel) of nes whose expression is red in the cell, tissue or organism as a result of administration or contact with the modified nucleic acid. Such ratios are referred to herein as the nsztokine Ratio or “PC” Ratio. The higher the PC ratio, the more efficacioius the modified nucleic acid ucleotide encoding the protein measured). Preferred PC Ratios, by cytokine, of the present invention may be greater than 1, greater than 10, greater than 100, greater than 1000, greater than 10,000 or more. Modified nucleic acids having higher PC Ratios than a modified nucleic acid of a different or unmodified construct are preferred.
The PC ratio may be further qualified by the percent modification present in the polynucleotide. For example, normalized to a 100% d nucleic acid, the protein production as a function of cytokine (or risk) or cytokine profile can be determined.
In one embodiment, the present invention provides a method for determining, across chemistries, cytokines or percent modification, the relative efficacy of any particular modified polynucleotide by comparing the PC Ratio of the modified nucleic acid (polynucleotide).
In another ment, the chemically modified mRNA are substantially non toxic and non mutagenic.
In one embodiment, the modified sides, modified nucleotides, and modified nucleic acids can be chemically modified on the major groove face, thereby disrupting major groove binding partner interactions, which may cause innate immune responses. Further, these modified nucleosides, modified nucleotides, and modified nucleic acids can be used to deliver a payload, e.g, detectable or therapeutic agent, to a biological target. For example, the nucleic acids can be covalently linked to a payload, e.g. a detectable or therapeutic agent, through a linker attached to the nucleobase or the sugar moiety. The compositions and methods described herein can be used, in viva and in vitro, both extracellarly or intracellularly, as well as in assays such as cell free assays.
In some ments, the present disclosure provides compounds comprising a nucleotide that disrupts binding of a major groove interacting, e.g. binding, r with a nucleic acid, wherein the nucleotide has decreased binding affinity to major groove interacting panners.
In r aspect, the present disclosure provides nucleotides that contain chemical modifications, wherein the nucleotide has altered binding to major groove interacting partners.
In some embodiments, the chemical modifications are d on the major groove face of the nucleobase, and wherein the al modifications can include replacing or substituting an atom of a pyrimidine nucleobase with an amine, an SH, an alkyl (e.g., methyl or ethyl), or a halo (e.g., chloro or fluoro).
In another aspect, the present sure provides al modifications located on the sugar moiety of the tide.
In r aspect, the t disclosure provides chemical modifications located on the phosphate backbone of the nucleic acid.
In some embodiments, the chemical modifications alter the electrochemistry on the major groove face of the nucleic acid.
] In another , the present disclosure provides nucleotides that contain chemical modifications, n the nucleotide reduces the cellular innate immune response, as compared to the ar innate immune induced by a corresponding unmodified nucleic acid.
In another aspect, the present disclosure provides nucleic acid sequences comprising at least two nucleotides, the nucleic acid sequence comprising a nucleotide that disrupts binding of a major groove interacting partner with the nucleic acid ce, wherein the nucleotide has sed binding ty to the major groove binding partner.
In another aspect, the present disclosure provides compositions comprising a compound as described herein. In some embodiments, the composition is a reaction mixture. In some embodiments, the composition is a pharmaceutical composition. In some embodiments, the composition is a cell culture. In some embodiments, the composition further comprises an RNA polymerase and a cDNA template. In some embodiments, the ition further comprises a nucleotide ed from the group consisting of adenosine, cytosine, ine, and uracil.
In a further aspect, the present disclosure provides methods of making a pharmaceutical formulation comprising a physiologically active secreted n, comprising transfecting a first population ofhuman cells with the pharmaceutical nucleic acid made by the methods described herein, wherein the secreted protein is active upon a second tion of human cells.
In some embodiments, the secreted protein is capable of interacting with a receptor on the surface of at least one cell present in the second population.
In some embodiments, the secreted n is ocyte—Colony ating Factor (G— CSF).
In some embodiments, the second population contains myeloblast cells that express the GCSF receptor.
In certain embodiments, provided herein are combination therapeutics containing one or more modified nucleic acids ning translatable regions that encode for a protein or proteins that boost a mammalian subject’s immunity along with a protein that induces antibody-dependent cellular toxitity. For example, provided are eutics containing one or more c acids that encode trastuzumab and granulocyte-colony stimulating factor (G—CSF). In particular, such combination therapeutics are useful in Her21L breast cancer patients who develop induced resistance to trastuzumab. (See, e.g., Albrecht, Irnmunotherapy. 2(6):795-8 (2010)).
In one embodiment, it is intended that the compounds of the present sure are stable.
It is further appreciated that certain features of the present disclosure, which are, for clarity, described in the context of te embodiments, can also be provided in combination in a single embodiment. Conversely, various features of the present disclosure which are, for brevity, described in the context of a single embodiment, can also be provided separately or in any suitable subcombination.
Modified Nucleotidesa Nucleosides and cleotides of the invention Herein, in a nucleotide, side or polynucleotide (such as the nucleic acids of the invention, e.g., mRNA molecule), the terms “modification” or, as appropriate, ed” refer to ation with respect to A, G, U or C ribonucleotides. lly, herein, these terms are not intended to refer to the ribonucleotide modifications in naturally occurring 5'—terminal mRNA cap moieties. In a polypeptide, the term “modification” refers to a modification as compared to the cal set of 20 amino acids, moiety) The modifications may be various distinct modifications. In some embodiments, where the c acid is an mRNA, the coding region, the flanking regions and/or the terminal regions may n one, two, or more (optionally different) nucleoside or nucleotide modifications. In some embodiments, a modified polynucleotide introduced to a cell may exhibit reduced degradation in the cell, as compared to an unmodified polynucleotide.
The polynucleotides can include any useful modification, such as to the sugar, the nucleobase, or the internucleoside linkage (e.g. to a linking phosphate / to a phosphodiester linkage/ to the phosphodiester backbone). For example, the major groove of a cleotide, or the major groove face of a nucleobase may comprise one or more modifications. One or more atoms of a pyrimidine nucleobase (e. g. on the major groove face) may be replaced or substituted with optionally substituted amino, optionally substituted thiol, optionally substituted alkyl (e. g., methyl or ethyl), or halo (e.g., chloro or fluoro). In certain embodiments, modifications (e.g., one or more modifications) are present in each of the sugar and the internucleoside linkage. Modifications according to the present invention may be modifications of cleic acids (RNAs) to deoxyribonucleic acids (DNAs), e.g., the substitution of the 2’OH of the ribofuranysyl ring to 2’H, threose c acids (TNAs), glycol nucleic acids (GNAs), peptide nucleic acids (PNAs), locked nucleic acids (LNAs) or hybrids thereof). Additional modifications are described herein As described herein, the polynucleotides of the invention do not substantially induce an innate immune response of a cell into which the cleotide (e.g., mRNA) is introduced. es of an induced innate immune response e 1) increased expression of pro-inflammatory cytokines, 2) activation of intracellular PRRs (RIG-I, MDAS, etc, and/or 3) termination or reduction in protein translation.
In n embodiments, it may desirable for a modified nucleic acid molecule introduced into the cell to be degraded intracellulary. For example, degradation of a modified nucleic acid molecule may be preferable if precise timing of protein production is desired. Thus, in some embodiments, the invention provides a modified nucleic acid molecule containing a degradation , which is capable of being acted on in a directed manner within a cell. In another aspect, the present disclosure provides polynucleotides comprising a nucleoside or nucleotide that can disrupt the binding of a major groove interacting, e.g. binding, r with the polynucleotide (e.g., where the modified nucleotide has decreased binding affinity to major groove interacting partner, as compared to an unmodified nucleotide).
The polynucleotides can ally include other agents (e.g., RNAi-inducing agents, RNAi agents, , shRNAs, miRNAs, antisense RNAs, ribozymes, tic DNA, tRNA, RNAs that induce triple helix formation, aptamers, vectors, etc.). In some embodiments, the polynucleotides may include one or more messenger RNAs (mRNAs) having one or more modified nucleoside or nucleotides (i.e., modified mRNA molecules). Details for these polynucleotides follow.
Palynucleotides The polynucleotides of the invention es a first region of linked nucleosides encoding a polypeptide of interest, a first flanking region located at the 5’ us of the first region, and a second flanking region located at the 3’ terminus of the first region.
In some embodiments, the polynucleotide (e. g., the first region, first g region, or second flanking region) includes 11 number of linked nucleosides having a (Ia) or a (Ia-1) or a pharmaceutically acceptable salt or stereoisomer thereof, wherein U is O, S, N(RU)nu, or C(RU)nu, wherein nu is an integer from 0 to 2 and each RU is, independently, H, halo, or ally substituted alkyl; - - - is a single bond or absent; each of R1,, R2), R1", R2”, R1, R2, R3, R4, and R5, if present, is, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally tuted loxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted arninoalkynyl, or absent; wherein the combination of R3 with one or more of R1), R1", R2,, R2", or R5 (e.g., the combination of R1: and R3, the combination of R1" and R3, the ation ofRT and R3, the combination of RT and R3, or the combination of R5 and R3) can join together to form optionally substituted alkylene or ally substituted heteroalkylene and, taken together with the carbons to which they are attached, provide an optionally substituted heterocyclyl (e.g., a bicyclic, tricyclic, or tetracyclic cyclyl); wherein the combination of R5 with one or more of RF, R1", RT, or RT (e.g., the combination of R1) and R5, the combination of R1" and R5, the combination of R2, and R5, or the combination of R2” and R5) can join together to form ally substituted alkylene or ally substituted heteroalkylene and, taken er with the carbons to which they are ed, provide an optionally substituted heterocyclyl (e. g., a bicyclic, tricyclic, or tetracyclic heterocyclyl); and wherein the combination of R4 and one or more of RF, R1", R2), R2”, R3, or R5 can join together to form optionally substituted alkylene or optionally substituted heteroalkylene and, taken together with the carbons to which they are attached, provide an optionally substituted heterocyclyl (e.g., a bicyclic, tricyclic, or yclic heterocyclyl); each of m’ and m” is, independently, an integer from 0 to 3 (e. g., from 0 to 2, from 0 to 1, from 1 to 3, or from 1 to 2); each of Y1, Y2, and Y3, is, independently, O, S, Se, -NRN1-, optionally substituted alkylene, or optionally substituted heteroalkylene, wherein RN1 is H, optionally substituted alkyl, ally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, or absent; each Y4 is, independently, H, hydroxy, thiol, boranyl, ally substituted alkyl, optionally substituted alkenyl, optionally tuted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted loxy, optionally substituted thioalkoxy, optionally substituted alkoxyalkoxy, or optionally substituted amino; each Y5 is, independently, O, S, Se, optionally substituted ne (eigi, methylene), or optionally substituted heteroalkylene; n is an integer from 1 to 100,000; and B is a nucleobase (e.g., a purine, a pyrimidine, or derivatives thereof), wherein the combination ofB and R1», the combination ofB and R2,, the combination ofB and R1“, or the combination ofB and R2» can, taken together with the carbons to which they are ed, optionally form a bicyclic group (e.g., a bicyclic cyclyl) or wherein the combination of B, RI“, and R3 or the combination of B, R2”, and R3 can optionally form a tricyclic or tetracyclic group (e.g., a lic or tetracyclic heterocyclyl, such as in Formula (IIo)—(IIp) herein).
In some embodiments, the polynucleotide es a modified ribose. In some embodiments, the polynucleotide (e. g., the first , the first flanking region, or the second g region) includes 11 number of linked nucleosides having Formula (Ia-2)-(Ia—5) or a pharmaceutically acceptable salt or stereoisomer thereof.
(Ia-3), (Ia-4), (Ia-5).
In some embodiments, the polynucleotide (e. g., the first region, the first flanking region, or the second flanking region) includes 11 number of linked nucleosides having Formula (lb) or Formula (lb-I): [<37]l\1_ R4R13' -- R3_//1L>[R1R4 R R5 YIZ R5 Y3=F:’4 Y3=F|'4 (Ib- 1) or a pharmaceutically acceptable salt or stereoisomer thereof, n U is O, S, N(RU)m,, or C(RU)nu, wherein nu is an integer from 0 to 2 and each RU is, independently, H, halo, or optionally substituted alkyl; - — — is a single bond or ; each of R1, Ry, R3", and R4 is, independently, H, halo, hydroxy, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally tuted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted lkynyl, or ; and wherein the ation of R1 and RT or the combination of R1 and R3" can be taken together to form optionally substituted alkylene or optionally substituted heteroalkylene (e. g., to produce a locked nucleic acid); each R5 is, independently, H, halo, hydroxy, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, or ab sent; each of Y1, Y2, and Y3 is, independently, O, S, Se, NRN1-, optionally substituted alkylene, or optionally substituted heteroalkylene, wherein RNl is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl; each Y4 is, independently, H, hydroxy, thiol, boranyl, optionally tuted alkyl, ally substituted alkenyl, optionally tuted alkynyl, optionally substituted , optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted alkoxyalkoxy, or ally substituted amino; n is an integer from 1 to 100,000; and B is a nucleobase.
In some embodiments, the polynucleotide (e. g., the first region, first flanking region, or second flanking region) es n number of linked nucleosides having Formula (Ic): (Ic), or a phannaceutically acceptable salt or stereoisomer thereof, wherein U is O, S, N(RU)nu, or C(RU)m,, wherein nu is an r from 0 to 2 and each RU is, independently, H, halo, or optionally substituted alkyl; — — — is a single bond or ; each of B1, B2, and B3 is, independently, a nucleobase (e. g., a purine, a pyrimidine, or derivatives thereof, as described herein), H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally tuted alkynyloxy, optionally substituted aminoalkoxy, optionally tuted alkoxyalkoxy, ally substituted yalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl, wherein one and only one of B1, B2, and B3 is a nucleobase; each of RM, RM, Rm, R3, and R5 is, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted loxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, ally substituted yalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl; each of Y1, Y2, and Y3, is, independently, O, S, Se, -NRN1-, optionally substituted alkylene, or optionally substituted heteroalkylene, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl; each Y4 is, independently, H, hydroxy, thiol, boranyl, optionally substituted alkyl, optionally substituted alkenyl, ally substituted l, optionally tuted alkoxy, ally substituted alkenyloxy, optionally substituted alkynyloxy, optionally tuted thioalkoxy, optionally substituted alkoxyalkoxy, or optionally substituted amino; each Y5 is, independently, O, S, Se, optionally substituted ne (e. g., methylene), or optionally substituted heteroalkylene; n is an integer from 1 to 100,000; and wherein the ring including U can include one or more double bonds.
In ular embodiments, the ring including U does not have a double bond between U- CB3R"3 or between CB3Rb3-CBZRb2.
In some embodiments, the polynucleotide (e. g., the first region, first flanking region, or second flanking region) includes 11 number of linked nucleosides having Formula (Id): iiy1iy5 IB Y3:P—* (Id), or a pharmaceutically acceptable salt or stere01somer thereof, wherein U [S. . . . 7 7 O, S, N(RU nu, or C(RU)nu, wherein nu is an integer from 0 to 2 and each RU is, independently, H, halo, or optionally substituted alkyl; each R3 is, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally tuted , optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally tuted amino, azido, ally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl; each of Y1, Y2, and Y3, is, independently, O, S, Se, -NRN1-, optionally substituted alkylene, or optionally substituted heteroalkylene, wherein RN1 is H, optionally substituted alkyl, optionally tuted alkenyl, optionally substituted alkynyl, or optionally substituted aryl; ] each Y4 is, independently, H, y, thiol, boranyl, optionally substituted alkyl, ally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted thioalkoxy, optionally substituted alkoxyalkoxy, or ally substituted amino; each Y5 is, independently, O, S, optionally tuted ne (eg, methylene), or optionally substituted heteroalkylene; n is an integer from 1 to 100,000; and B is a nucleobase (e.g., a purine, a dine, or derivatives thereof).
In some embodiments, the polynucleotide (e. g., the first region, first flanking region, or second flanking region) includes n number of linked nucleosides having Formula (Ie): (Ie), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each of U’ and U” is, ndently, O, S, N(RU)nu, or C(RU)nu, wherein nu is an integer from 0 to 2 and each RU is, ndently, H, halo, or optionally substituted alkyl; each R6 is, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl; ] each Y5, is, independently, O, S, optionally substituted alkylene (e. g., methylene or ethylene), or optionally substituted heteroalkylene; n is an integer from 1 to 100,000; and B is a base (e.g., a purine, a pyrimidine, or derivatives thereof).
] In some ments, the polynucleotide (e. g., the first , first g region, or second flanking region) includes 11 number of linked nucleosides having Formula (If) or (If—l): (If), (If-l), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each of U’ and U” is, ndently, O, S, N, N(RU)uu, or C(RU)uu, wherein nu is an integer from 0 to 2 and each RU is, independently, H, halo, or optionally substituted alkyl (e.g., U’ is O and U” is N); - - - is a single bond or absent; each of R1,, R2,, R1”, R2”, R3, and R4 is, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted , optionally substituted alkenyloxy, optionally tuted loxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, ally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted lkynyl, or absent; and wherein the combination ofRF and R3, the combination ofR1“ and R3, the combination of R2, and R3, or the combination ofRT and R3 can be taken together to form optionally substituted alkylene or optionally substituted heteroalkylene (e. g., to produce a locked nucleic acid);each of m’ and m” is, ndently, an integer from 0 to 3 (e. g., from 0 to 2, from 0 to 1, from 1 to 3, or from 1 to 2); each of Y1, Y2, and Y3, is, independently, O, S, Se, -NRN1-, optionally substituted alkylene, or optionally substituted heteroalkylene, wherein RN1 is H, optionally substituted alkyl, optionally substituted l, optionally substituted alkynyl, optionally substituted aryl, or absent; each Y4 is, independently, H, hydroxy, thiol, boranyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted , optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted koxy, optionally substituted alkoxyalkoxy, or ally substituted amino; ] each Y5 is, ndently, O, S, Se, optionally substituted alkylene (e.g., methylene), or optionally substituted heteroalkylene; n is an integer from 1 to 100,000; and B is a nucleobase (e.g., a purine, a dine, or derivatives thereof).
In some embodiments of the polynucleotides (e.g., Formulas (Ia)-(Ia-5), (Ib)-(If-l), (Ila)- (IIp), (IIb-l), (IIb-2), (IIc-l)-(IIc-2), (IIn-l), (IIn—2), (IV1), and (IXa)-(IXr)), the ring including U has one or two double bonds.
In some embodiments of the polynucleotides (e.g., Formulas a-5), (Ib)-(If-l), (Ila)- (IIp), (IIb-l), (IIb-2), (IIc-1)-(IIc-2), (IIn-l), (IIn-2), (IVa)-(IV1), and (IXa)-(IXr)), each of R', R1, and RI”, if present, is H. In further embodiments, each of R2, R2,, and R2”, if present, is, independently, H, halo (e. g., fluoro), hydroxy, optionally substituted alkoxy (e,g., methoxy or ethoxy), or ally substituted alkoxyalkoxy. In particular embodiments, alkoxyalkoxy is - (CH2)52(OCH2CH2)51(CH2)530R’, wherein s1 is an integer from 1 to 10 (eg., from 1 to 6 or from 1 to 4), each of s2 and 53, independently, is an integer from 0 to 10 (e.g., from O to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R’ is H or Cmo alkyl). In some embodiments, s2 is 0, S] is l or 2, s3 is 0 or 1, and R’ is C1_6 alkyl.
In some ments of the polynucleotides (e.g., as (Ia)-(Ia—5), (Ib)-(If), (Ila)- (Ilp), (IIb-l), (IIb-2), (IIc—1)-(IIc-2), (IIn—l), (IIn-2), (IVa)-(IV1), and (IXa)-(IXr)), each of R2, RT, and RT, if present, is H. In further ments, each of R1, R1,, and R1”, if t, is, independently, H, halo (e. g., fluoro), hydroxy, optionally substituted alkoxy (e. g., y or ethoxy), or optionally substituted alkoxyalkoxy. In particular ments, alkoxyalkoxy is - (CH2)52(OCH2CH2)51(CH2)S3OR’, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e. g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R’ is H or C140 . In some embodiments, s2 is 0, s1 is l or 2, s3 is 0 or 1, and R’ is C1_6 alkyl.
In some embodiments of the polynucleotides (e.g., Formulas (Ia)-(Ia-5), (Ib)-(If-l), (Ila)— (IIp), (IIb- l), (IIb-2), (IIc—1)-(IIc-2), (IIn—l), (IIn-2), (IVa)-(IV1), and (IXa)-(IXr)), each ofR3, R4, and R5 is, independently, H, halo (e.g., fluoro), hydroxy, optionally substituted alkyl, optionally substituted alkoxy (e.g., methoxy or ethoxy), or optionally substituted alkoxyalkoxy. In ular embodiments, R3 is H, R4 is H, R5 is H, or R3, R4, and R5 are all H. In ular embodiments, R3 is CH alkyl, R4 is C1_6 alkyl, R5 is C1_6 alkyl, or R3, R4, and R5 are all CH; alkyl. In particular embodiments, R3 and R4 are both H, and R5 is C1_6 alkyl.
In some embodiments of the polynucleotides (e.g., Formulas (Ia)-(Ia-5), (Ib)—(If—l), (Ila)- (IIp), (IIb- l), (IIb-2), (IIc— l )-(IIc-2), (IIn—l), (IIn-2), (IVa)-(IV1), and (IXa)-(IXr)), R3 and R5 join together to form optionally substituted alkylene or optionally substituted heteroalkylene and, taken together with the carbons to which they are attached, provide an optionally substituted heterocyclyl (e.g., a bicyclic, lic, or tetracyclic heterocyclyl, such as trans-3’,4’ analogs, wherein R3 and R5 join together to form heteroalkylene (e.g., b1O(CH2)b20(CH2)b3-, wherein each of bl , b2, and b3 are, independently, an integer from 0 to 3).
In some embodiments of the polynucleotides (e.g., Formulas (Ia)-(Ia-5), (Ib)-(If-l), (Ila)- (IIp), (IIb-l), ), (IIc—l)—(IIc—2), (IIn-l), (IIn-2), (IVa)-(IVl), and (IXa)-(I_Xr)), R3 and one or more of R1, R1”, R2,, R2”, or R5join together to form optionally tuted ne or optionally substituted heteroalkylene and, taken together with the carbons to which they are attached, provide an optionally substituted heterocyclyl (e.g., a bicyclic, lic, or tetracyclic heterocyclyl, R3 and one or more of R1», R1", R2,, R2”, or R5 join together to form heteroalkylene (e.g., - (CH2)bIO(CH2)bZO(CHZ)b3-, wherein each of b1, b2, and b3 are, independently, an integer from 0 to In some embodiments of the polynucleotides (e.g., Formulas (Ia)-(Ia—5), (Ib)-(If-l), (Ila)- (IIp), (IIb-l), (IIb-2), (IIc—l)—(IIc—2), (IIn—l), (IIn-2), (IVa)-(IV1), and (IXa)-(IXr)), R5 and one or more of Rli, R1", R2,, or R2” join together to form optionally substituted alkylene or optionally substituted heteroalkylene and, taken together with the carbons to which they are attached, provide an optionally substituted heterocyclyl (e.g., a ic, tricyclic, or tetracyclic heterocyclyl, R5 and one or more of R1), R1“, R2,, or R2” join together to form heteroalkylene (e. g., - (CH2),10(CH2)b20(CH2)b3-, wherein each of bl, b2, and b3 are, independently, an integer from 0 to In some embodiments of the polynucleotides (e.g., Formulas (Ia)-(Ia-5), (Ib)-(If-l), (Ila)— (IIp), (IIb— I), (IIb-2), (IIc—1)-(IIc-2), (IIn— 1), (1111-2), (IVa)-(IV1), and (IXr)), each Y2 is, independently, O, S, or -NRN1-, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl. In ular embodiments, Y2 is NRNL, wherein RN1 is H or optionally substituted alkyl (e. g., C1_6 alkyl, such as methyl, ethyl, isopropyl, or n—propyl).
In some embodiments of the polynucleotides (e.g., Formulas (Ia)-(Ia-5), (Ib)-(If—l), (Ila)- (IIp), (IIb- l), (IIb-2), (IIc—1)-(IIc-2), (IIn— 1), (1111-2), (IVa)-(IV1), and (IXa)—(IXr)), each Y3 is, independently, O or S.
] In some ments of the polynucleotides (e.g., Formulas (Ia)-(Ia-5), (Ib)—(If—l), (Ila)- (IIp), (IIb- l), ), (IIc— l )-(IIc-2), (IIn- 1), (1111-2), (IVa)-(IV1), and (IXa)-(IXr)), R1 is H; each R2 is, independently, H, halo (e.g., fluoro), y, optionally substituted alkoxy (e.g., methoxy or ethoxy), or optionally substituted alkoxyalkoxy (e. g., -(CH2)52(OCH2CH2)S1(CH2)S3OR’, n s1 is an integer from I to 10 (e.g., from I to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R’ is H or C140 alkyl, such as n 52 is 0, 51 is 1 or 2, s3 is 0 or 1, and R’ is CH; alkyl); each Y2 is, independently, O or , wherein RNl is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally tuted aryl (e,g,, wherein RNI is H or optionally substituted alkyl (e.g., C14; alkyl, such as , ethyl, isopropyl, or n-propyl)); and each Y3 is, independently, O or S (e.g., S). In fiuther embodiments, R3 is H, halo (e.g., fluoro), hydroxy, optionally tuted alkyl, optionally substituted alkoxy (e. g., methoxy or ethoxy), or optionally tuted alkoxy. In yet further embodiments, each Y1 is O or -N'RNl-, , independently, wherein RN] is H, optionally substituted alkyl, ally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl (e. g., wherein RNl is H or optionally substituted alkyl (e.g., CL 6 alkyl, such as methyl, ethyl, isopropyl, or n-propyl)); and each Y4 is, independently, H, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, ally substituted thioalkoxy, optionally substituted alkoxyalkoxy, or optionally substituted amino.
In some embodiments of the polynucleotides (e.g., Formulas (Ia)-(Ia-5), (Ib)-(If-l), (Ila)- (Ilp), (IIb— l), (IIb-2), (IIc— 1 )—(IIc-2), (IIn— 1), (1111-2), (IVa)-(IV1), and (IXr)), each R1 is, independently, H, halo (e. g., fluoro), hydroxy, optionally tuted alkoxy (e. g., methoxy or ethoxy), or optionally substituted alkoxyalkoxy (e. g., -(CH2)52(OCH2CH2)S1(CH2)530R’, wherein S] is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R’ is H or Cmo alkyl, such as wherein s2 is 0, s1 is 1 or 2, s3 is 0 or 1, and R’ is C1_6 alkyl); R2 is H; each Y2 is, independently, O or -NRN1-, wherein RNl is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl (e.g., wherein RN1 is H or optionally substituted alkyl (e. g., C1_6 alkyl, such as methyl, ethyl, isopropyl, or n-propyl)); and each Y3 is, independently, O or S (e.g., S). In r embodiments, R3 is H, halo (e.g., fluoro), hydroxy, optionally substituted alkyl, optionally substituted alkoxy (e.g., methoxy or ethoxy), or optionally substituted alkoxyalkoxy. In yet further ments, each Y1 is O or - , independently, NRN'-, wherein RNl is H, optionally substituted alkyl, optionally substituted alkenyl, ally substituted alkynyl, or ally substituted aryl (e.g., wherein RNl is H or optionally substituted alkyl (e.g., CH; alkyl, such as methyl, ethyl, isopropyl, or n-propyl)); and each Y4 is, independently, H, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted thioalkoxy, optionally tuted alkoxy, or optionally substituted amino, In some embodiments of the polynucleotides (e.g., Formulas (Ia)-(Ia-5), (Ib)-(If-l), (Ila)- (IIp), ), (Ilb-2), (IIc-1)-(IIc-2), (IIn-l), (IIn-2), (IVa)—(IV1), and (IXI)), the ring including U is in the [3-D (e.g., B-D-ribo) configuration.
In some embodiments of the polynucleotides (e.g., Formulas (Ia)-(Ia-5), (Ib)-(If-l), (IIa)- (IIp), (IIb-l), ), (IIc-1)-(IIc-2), ), (IIn-2), (IVa)—(IV1), and (IXa)-(IXr)), the ring including U is in the a—L (e. g., a-L-ribo) configuration.
In some embodiments of the polynucleotides (e.g., Formulas (Ia)-(Ia-5), (Ib)-(If-l), (IIa)- (IIp), (IIb-l), (IIb-2), (IIc-1)-(IIc-2), (IIn-l), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr)), one or more B is not pseudouridine (w) or yl-cytidine (mSC).
] In some embodiments, about 10% to about 100% of 11 number of B nucleobases is not 1|; or mSC (e.g., from 10% to 20%, from 10% to 35%, from 10% to 50%, from 10% to 60%, from 10% to 75%, from 10% to 90%, from 10% to 95%, from 10% to 98%, from 10% to 99%, from 20% to 35%, from 20% to 50%, from 20% to 60%, from 20% to 75%, from 20% to 90%, from 20% to 95%, from % to 98%, from 20% to 99%, from 20% to 100%, from 50% to 60%, from 50% to 75%, from 50% to 90%, from 50% to 95%, from 50% to 98%, from 50% to 99%, from 50% to 100%, from 75% to 90%, from 75% to 95%, from 75% to 98%, from 75% to 99%, and from 75% to 100% ofn number of B is not w or mSC). In some embodiments, B is not w or msC.
In some embodiments of the polynucleotides (e.g., Formulas (Ia)-(Ia-5), (Ib)-(If-l), (IIa)— (IIp), (IIb— l), (IIb-2), (IIc-1)-(IIc-2), (IIn-l), ), (IVa)—(IV1), and (IXa)-(IXr)), when B is an unmodified nucleobase ed from cytosine, guanine, uracil and adenine, then at least one of Y1, Y2, or Y3 is not 0.
In some embodiments, the polynucleotide includes a modified ribose. In some embodiments, the polynucleotide (e. g., the first region, the first flanking region, or the second flanking region) es 11 number of linked nucleosides having Formula (IIa)-(IIc): . (Ila), (IIb), or (IIc), or a pharmaceutically acceptable salt or stereoisomer thereof. In particular embodiments, U is O or C(RU)nu, wherein nu is an integer from 0 to 2 and each RU is, independently, H, halo, or ally substituted alkyl (e.g., U is iCHr or iCHi). In other embodiments, each of R1, R2, R3, R4, and R5 is, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally tuted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted yalkoxy, optionally substituted amino, azido, ally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, or absent (e. g., each R1 and R2 is, independently H, halo, hydroxy, optionally substituted alkyl, or optionally substituted alkoxy; each R3 and R4 is, independently, H or optionally substituted alkyl; and R5 is H or hydroxy), and; is a single bond or double bond.
] In particular embodiments, the polynucleotide (e. g., the first region, the first flanking , or the second flanking region) includes 11 number of linked nucleosides having Formula (IIbl )—(IIb-2): _ 1_Y5 Y1_Y5 B Y3: Y3=||3 (IIb- l) or (IIb-2) or a pharmaceutically able salt or stereoisomer thereof. In some embodiments, U is O or C(RU)nu, wherein nu is an integer from 0 to 2 and each RU is, independently, H, halo, or optionally substituted alkyl (etgt, U is —CH2— or — CH—). In other ments, each of R1 and R2 is, independently, H, halo, y, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally tuted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally tuted amino, azido, optionally tuted aryl, optionally substituted lkyl, ally substituted aminoalkenyl, optionally substituted aminoalkynyl, or absent (e.g., each R1 and R2 is, independently, H, halo, hydroxy, optionally tuted alkyl, or optionally substituted alkoxy, e. g., H, halo, hydroxy, alkyl, or alkoxy). In particular embodiments, R2 is hydroxy or optionally substituted alkoxy (e.g., methoxy, ethoxy, or any described herein).
In particular embodiments, the polynucleotide (e.g., the first region, the first flanking region, or the second flanking region) includes 11 number of linked nucleosides having Formula (IIc- l)-(IIc—4): Y1—Y5 ——Y1—Y5 B WoR2 Y3=P Y3=P — (IIc—l), — — ), 1 5 Y —Y B 1 5 Y —Y B U O R3 R3 R2 WR2 Y2 Y2 Yazlla Y3:||3 l4 l4 — (IIc-3), or (IIc-4), or a pharmaceutically acceptable salt or stereoisomer thereof.
In some embodiments, U is O or C(RU)nu, wherein nu is an r from 0 to 2 and each RU is, independently, H, halo, or optionally substituted alkyl (e.g., U is fiHr or 43H7). In some embodiments, each of R1, R2, and R3 is, ndently, H, halo, hydroxy, thiol, c,ptionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, ally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, or absent (e.g., each R1 and R2 is, independently, H, halo, hydroxy, optionally substituted alkyl, or optionally tuted alkoxy, e.g., H, halo, hydroxy, alkyl, or alkoxy; and each R3 is, independently, H or optionally substituted alkyl)). In particular embodiments, R2 is optionally substituted alkoxy (e.g., methoxy or ethoxy, or any described herein). In particular embodiments, R1 is optionally substituted alkyl, and R2 is hydroxy. In other embodiments, R1 is hydroxy, and R2 is optionally tuted alkyl. In r embodiments, R3 is optionally substituted alkyl.
In some embodiments, the polynucleotide es an acyclic d ribose. In some embodiments, the polynucleotide (e. g., the first region, the first flanking region, or the second flanking region) includes n number of linked nucleosides having Formula (IId)-(IIt): __Y1_Y5 B {5 R1 R3 R4 Y3=I|= — (IId), — — (He), or Y‘—Y5 B R R1 R3 7'14 Y3=|l3 — — (III), or a pharrnaceutically acceptable salt or stereoisomer thereof.
In some embodiments, the polynucleotide includes an acyclic modified hexitol. In some embodiments, the polynucleotide (e. g., the first region, the first flanking region, or the second flanking region) includes 11 number of linked nucleosides having a (IIg)—(IIj): Y1_Y5 (11g), (1111), (IIi), or (IIj), or a pharmaceutically acceptable salt or stereoisomer thereof.
In some embodiments, the polynucleotide es a sugar moiety having a contracted or an expanded ribose ring. In some embodiments, the polynucleotide (e. g., the first region, the first flanking region, or the second flanking region) includes 11 number of linked nucleosides having Formula (IIk)-(IIm): Y1_Y5 UB WR51 4 2 (11k), (III), or (Hm), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each of R1,, R1”, R23, and RT is, independently, H, halo, hydroxy, optionally substituted alkyl, optionally substituted , optionally substituted alkenyloxy, optionally substituted loxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, or absent; and wherein the combination ofRT and R3 or the combination of R2" and R3 can be taken er to form optionally substituted alkylene or ally substituted heteroalkylene, In some embodiments, the cleotide es a locked modified ribose. In some embodiments, the polynucleotide (e. g., the first region, the first flanking region, or the second flanking region) includes n number of linked nucleosides having Formula (IIn): (IIn), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein R37 is O, S, or -NRN1-, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl and R3" is optionally substituted alkylene (e.g., -CH2-, 2-, or -CH2CH2CH2-) or optionally substituted alkylene (e.g., — CHZNH-, -CH2CH2NH-, -CH20CH2-, or -CH2CH20CH2-) (e.g., R3’ is o and R3" is optionally substituted alkylene (e. g., -CH2-, -CH2CH2-, or -CH2CH2CH2-)).
In some embodiments, the polynucleotide (e. g., the first region, the first g region, or the second flanking region) includes 11 number of linked sides having Formula (IIn-l )-(II— n2): (IIn—l) or — — (IIn—2), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein R3, is O, S, or -NRN1-, wherein RNl is H, optionally substituted alkyl, optionally tuted alkenyl, optionally substituted alkynyl, or optionally substituted aryl and Ry is optionally substituted alkylene (e. g., -CH2-, -CH2CH2-, or -CH2CH2CH2-) or optionally substituted heteroalkylene (e. g., -, -CH2CH2NH-, -CH20CH2-, or - 0CH2-) (e.g., R3) is O and R3" is optionally substituted alkylene (e.g, -CH2-, -CH2CH2-, or - CH2CH2CH2-)).
In some embodiments, the polynucleotide includes a locked modified ribose that forms a tetracyclic heterocyclyl. In some embodiments, the polynucleotide (e,g, the first region, the first flanking region, or the second flanking region) includes 11 number of linked nucleosides having a (110): /U\T4T2' ——Y1—Y5 R12a U\IR4T2' ,Rlza /lzTN/T1I .. All-TN 12c R3: M V R37\ 3-K" 5H Y?R3A:v‘lT1.. Y2 R V Y3=i=— ”I": — l4 l4 7 (110) or 7 7 (Hp), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein R12“, R12“, T1,, ,, T2", V1, and V3 are as described herein.
Any of the formulas for the polynucleotides can include one or more nucleobases described herein (e.g., Formulas b43)).
In one embodiment, the present invention provides methods of preparing a polynucleotide comprising at least one nucleotide that disrupts binding of a major groove interacting partner with the nucleic acid, wherein the cleotide comprises 11 number of nucleosides having Formula (Ia), as defined herein: (la), the method comprising reacting a compound of Formula (IIIa), as defined herein: Y6 P—Y‘ Y5 (IIIa), with an RNA polymerase, and a cDNA template.
In a further embodiment, the present invention provides methods of amplifying a polynucleotide comprising at least one nucleotide that disrupts binding of a major groove binding partner with the polynucleotide sequence, the method comprising: reacting a compound of Formula (Illa), as defined herein, with a primer, a cDNA te, and an RNA polymerase, In one embodiment, the present invention provides methods of preparing a polynucleotide comprising at least one nucleotide that disrupts binding of a major groove interacting partner with the nucleic acid, wherein the polynucleotide ses 11 number of nucleosides having Formula (Ia- l), as defined herein: (Ia-l), the method comprising reacting a compound ofFormula (IIIa—l), as defined herein: Y6 I'D—Y1 Y5 (IIIa—l), with an RNA rase, and a cDNA te, In a r embodiment, the present invention provides methods of amplifying a polynucleotide comprising at least one nucleotide (e. g., d mRNA molecule) that disrupts binding of a major groove binding partner with the polynucleotide sequence, the method comprising: reacting a compound of Formula (IIIa— 1 ), as defined herein, with a , a cDNA te, and an RNA polymerase.
In one embodiment, the present ion provides methods of preparing a polynucleotide comprising at least one nucleotide that disrupts binding of a major groove interacting partner with the nucleic acid ce, wherein the cleotide comprises n number of nucleosides having Formula (Ia-2), as defined herein: (Ia-2), the method comprising reacting a compound of Formula (IIIa— 2), as defined herein: Y6 I'D—Y1 Y5 q (IIIa—2), with an RNA polymerase, and a cDNA template.
In a further embodiment, the present invention provides methods of amplifying a polynucleotide comprising at least one nucleotide (e. g., modified mRNA molecule) that dismpts binding of a major groove binding partner with the polynucleotide, the method comprising reacting a compound ofFormula (IIIa—2), as defined herein, with a primer, a cDNA template, and an RNA polymerase.
In some embodiments, the reaction may be ed from 1 to about 7,000 times. In any of the embodiments , B may be a nucleobase of a (b1)—(b43).
The cleotides can optionally include 5’ and/or 3’ flanking regions, which are described herein.
Modified Nucleotides and Nucleosides The present ion also includes the building blocks, e. g., modified ribonucleosides, modified ribonucleotides, of the polynucleotides, e.g., modified RNA ( or mRNA) molecules. For example, these ng blocks can be useful for preparing the polynucleotides of the invention.
In some embodiments, the building block molecule has Formula (IIIa) or (IIIa-l): (IIIa), (IIIa— 1) or a ceutically acceptable salt or stereoisomer thereof, wherein the substituents are as described herein (e.g., for Formula (Ia) and (Ia-1)), and wherein when B is an unmodified nucleobase selected from ne, guanine, uracil and adenine, then at least one of Y1, Y2, or Y3 is not 0.
In some embodiments, the building block molecule, which may be incorporated into a polynucleotide, has Formula (IVa)-(IVb): YfE‘Ylbf $4130, (5W< (IVa) or HO OH (IVb), or a pharmaceutically acceptable salt or isomer thereof, wherein B is as bed herein (e.g., any one of (bl)- (1343))- In particular embodiments, Formula (IVa) or (IVb) is combined with a modified uracil (erg, any one of formulas (b1)-(b9), (b23), and (b28)—(b31), such as formula (b1), (b8), (b28), (b29), or (b30)). In particular embodiments, Formula (IVa) or (IVb) is combined with a d cytosine (e.g., any one of formulas (b10)—(b14), (b24), (b25), and (b36), such as formula (MO) or (b32)). In particular embodiments, Formula (IVa) or (IVb) is combined with a modified gianine (e.g., any one of formulas (b15)-(bl7) and (b37)-(b40)). In particular embodiments, Formula (IVa) or (IVb) is combined with a modified adenine (e.g., any one of formulas (b18)-(b20) and (b4l)- (M3))- In some embodiments, the building block molecule, which may be incorporated into a polynucleotide, has Formula (IVc)—(IVk): Y6 1 P—Y \6 $44 cf“ HO R2 (IVc) H6R2,(1Vd) Y6 Y6‘< R2(IVe), Y6 Y6 >m(IVg), Y6 Y6 H5 i= (1V1) Y6 F|>—Y1 5 Y6 Y4 r /U R3‘ R1 HO CI (IVk), or HO 1 (W1), or a pharmaceutically acceptable salt or stereoisomer f, wherein B is as described herein (e.g., any one of(bl)-(b43)).
In particular embodiments, one of Formulas (IVc)-(IVk) is combined with a modified uracil (e.g., any one of formulas (bl)-(b9), (b21)-(b23), and (b28)-(b31), such as formula (bl), (b8), (b28), (b29), or .
In particular embodiments, one of Formulas (IVc)-(IVk) is combined with a modified cytosine (e.g., any one of formulas (b10)—(b14), (b24), (b25), and (b32)-(b36), such as formula (blO) or (b32)).
In particular embodiments, one of Formulas (IVc)-(IVk) is combined with a modified guanine (e.g., any one of formulas (b15)-(b17) and (b37)—(b40)).
In ular embodiments, one of Formulas (IVc)-(IVk) is combined with a modified e (e.g., any one of formulas (b18)-(b20) and (b41)—(b43)).
] In other embodiments, the building block molecule, which may be incorporated into a cleotide has Formula (Va) or (Vb): Y6 "_ I'34 1 5 (U Y6 Y I Z rR3\\ (Vb), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein B is as described herein (e.g., any one of (bl)-(b43)).
In other embodiments, the building block molecule, which may be orated into a polynucleotide has Formula (IXa)-(IXd): Y3 Y3 Y6 I'D—Y1 5 Y6 5 0 I'l3—Y1 H6 [3 (IXa), H6 I3r(IXb), HO CI (IXc), or On. (IXd), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein B is as described herein (e. g., any one of (b1)-(b43)).
In particular embodiments, one of as (IXa)—(IXd) is ed with a modified uracil (e.g., any one of as (b1)-(b9), (b21)-(b23), and (b28)-(b31), such as formula (b1), (b8), (b28), (b29), or (b30)). In particular embodiments, one of Formulas (IXa)—(IXd) is combined with a modified cytosine (e.g., any one of formulas (b10)—(b14), (b24), (b25), and (b32)-(b36), such as formula (blO) or (b32)).
In particular embodiments, one of Formulas (IXa)-(IXd) is combined with a modified guanine (e.g., any one of formulas (b15)—(b17) and (b37)-(b40)).
In particular embodiments, one of as (IXd) is combined with a modified adenine (e.g., any one of formulas (b18)—(b20) and (b41)-(b43)).
In other embodiments, the building block le, which may be incorporated into a polynucleotide has Formula (IXe)-(IXg): HO R2 (IXg), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein B is as bed herein (e. g., any one of (b1)—(b43)).
In particular embodiments, one of Formulas (IXe)-(IXg) is combined with a modified uracil (e.g., any one of formulas (b1)-(b9), (b21)-(b23), and (b28)-(b31), such as formula (bl), (b8), (b28), (b29), or (1330)).
In ular embodiments, one of Formulas (IXe)-(IXg) is combined with a modified ne (e.g., any one of formulas (b10)—(b14), (b24), (b25), and (b32)-(b36), such as formula (blO) or .
In particular embodiments, one of Formulas (IXe)-(IXg) is combined with a modified guanine (e.g., any one of formulas (b15)-(b17) arid (b37)—(b40)).
In particular embodiments, one of as (IXe)-(IXg) is combined with a modified adenine (e.g., any one of formulas (b18)-(b20) and (b41)—(b43)).
In other embodiments, the building block molecule, which may be incorporated into a polynucleotide has Formula (IXh)-(IXk): r3 % (Y 1 Y6 P—Yl 5 Y P—Y 5 r w r w , . H35 - 1 HO OH (IXj), or HO OH(IXk), or a pharmaceutically able salt or stereoisomer thereof, wherein B is as described herein (e.g., any one of (bl)— (b43)). In ular embodiments, one of Formulas (IXh)-(IXk) is combined with a modified uracil (erg, any one of formulas b9), (b21)—(b23), and (b28)—(b31), such as formula (b1), (b8), (b28), (b29), or (b30)). In particular embodiments, one of Formulas (IXh)-(IXk) is combined with a modified cytosine (e.g., any one of formulas (b10)-(b14), (b24), (b25), and (b32)-(b36), such as formula (b 10) or (b32)).
In particular embodiments, one of Formulas (IXh)-(IXk) is combined with a modified guanine (e.g., any one of formulas (b15)-(bl7) and (b37)—(b40)). In ular embodiments, one of Formulas (IXh)-(IX'.k) is combined with a modified adenine (e. g., any one of formulas (bl 0) and (b4])-(b43)).
In other embodiments, the building block molecule, which may be incorporated into a polynucleotide has a (IXl)-(IXr): o o (I? (3 HO I'il-O fill—o B Ho I'D-O I'D-O B OH BH2 0 OH CH3 0 r2 1 r2 r1 0 Se 0 II II II HO I'D—o I'D—o B HO I'D—o B OH OH 0 OH 0 r2 r1 HO: :OH(IXn), Ho Rm», 9 9 H0 ”LO B HO F|’—O OH O OH O HO bCH3(IXr) or a pharmaceutically acceptable salt or isomer thereof, wherein each rl and r2 is, independently, an integer from 0 to 5 (e.g., from 0 to 3, from 1 to 3, or from 1 to 5) and B is as described herein (e.g., any one of(bl)-(b43)).
In ular embodiments, one of Formulas (IXl)-(IXI) is combined with a modified uracil (e.g., any one of formulas (bl)—(b9), (b23), and (b28)-(b3l), such as formula (bl), (b8), (b28), (b29), or (b30)).
In particular embodiments, one of Formulas (IXl)-(IXI) is combined with a modified cytosine (e.g., any one of as (b10)-(bl4), (b24), (b25), and (b32)-(b36), such as formula (MO) or (b32)).
In particular embodiments, one of Formulas (IXl)-(IXI) is combined with a modified guanine (e.g., any one of formulas (b15)-(b17) and (b37)-(b40)). In particular embodiments, one of Formulas (IXl)-(I.XI) is combined with a modified adenine (e.g., any one of formulas (bl 8)-(b20) and (b41)-(b43)).
In some embodiments, the building block le, which may be incorporated into a polynucleotide can be selected from the group consisting of: N \N N / </ I A o 3 l J 9 N N/ N N HO I'D—O NH2 Ho 5—0 | o r r H6 oH (BB- 1), HO OH (BB- 2), \NH Cl N / o <’ | J o < | / 9 II N ” N N HO I'D-O N HO O I'D-O O OH OH r r HO OH (BB- 3), HO OH (BB- 4), NH 0 N /CH3 N N </ NH O l J o </ l II N J N n N HO I'D—o HO N 0 I'D-O o 0H OH r r HO OH (BB- 5), HO OH (BB- 6), N \N / N | </ I 9 % Ax N N NH2 HO I'D—o N HO 5-0 o I o OH OH I' r H6 :OH (BB- 7), H6 bH (BB- 8), o 0 ,CH2 H <,” N N | NH (I? A O:< I A N (u) HO I'D—o N NH2 N N o Ho F|’—O NH2 OH OH r r HO OH (BB- 9), H6 bH (BB- 10), CI 0 N \ N / NH <3 flufi 9 (Cf: HO I'D—o N NH2 o HO 'ID'0 N NH OH OH r r H6 6H (BB- 11), and HO: :OH (BB— 12), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each r is, ndently, an integer from 0 to 5 (e.g., from 0 to 3, from 1 to 3, or from 1 to 5).
In some embodiments, the building block molecule, which may be incorporated into a polynucleotide can be selected from the group consisting of: HO #0 N Ho o F|’-O 0 OH H r H6 6H (BB- 13), H6 OH (BB- 14), 0e 9 \N® \ea \N N \N 9 </N | g o </ | g HO "_ II: o_ N N 0 HO II: o OH OH r r H6 6H (BB-15), H6 6H (BB-16), NH2 9 /N \N’0 o < | II N /)6 Ho I'D—o N H6 6H (BB-17), Hoe—o </ | HO J I'D—o o N N OH ‘ _ r 6 2') H6 :OH (BB- 18), x (BB— 19), and 0 flNH HO fi'uo N/go HO OH (BB- 20), or a pharmaceutically able salt or stereoisomer thereof, wherein each r is, independently, an r from 0 to 5 (eg, from O to 3, from 1 to 3, or from 1 to 5) and s] is as described herein.
In some embodiments, the building block molecule, which may be incorporated into a nucleic acid (e.g., RNA, mRNA, polynucleotide), is a modified uridine (e,g, selected from the group consisting of: (BB- 22), (BB- 23), (BB- 26), (BB- 27), Y6 —Y1 N\N/§O Y4 0 ), H6 :OH (BB- 29), JOL ,CH3 HN N (BB- 33), OCH3 (BB- 34), (BB- 35), (BB- 38), HO OH (BB- 39), /CH3 NW W< CF3 OACFso (BB- 40), (BB- 41), / OH l HW (BB- 42), H6 6H (BB- 43), (BB- 44), (BB- 45), / moc (BB-46), H6 OCHa (BB- 47), H6 6H (BB- 48), H6 6H (BB- 49), O O HN 0Fmoc (BB- 50), (BB- 53), (BB- 54), (BB- 55), (BB-56), (BB-5?), (BB- 59), N’CH3 (BB- 60), (BB- 6]), HO OH (BB- 62), HO OCH3 (BB_ 63), J\N NHFmoc Y6 FI’—Y1 O Y4 0 HO: :OH (BB-64), j]: c02H H3c\ NvNHZ Y6 F|’—Y1 O Y4 0 H6 EDH (BB-65), OH (BB- 67), (BB- 70), H3C\NJLN/CH3 HNANN K3 / \ X3 o Y6 I'D—Y1 O Y1 Y4 0 Y4 0 HO: 2OH (BB-71), HO OH (BB-72), HN NW I3 / Y —F|’—Y1 Y4 0 HO EDH (BB- 74), HO OH (BB- 75), HN/lkN/v I3 / Y I'D—Y Y4 0 HO 6H (BB-76), A /OH HN NW HN Y3 / Y3 / ll 0 Y —F|’-Y1 )(6—'F|'>—Y1 0 Y4 0 Y4 O r r H6 :OH (BB- 77), H6 6H (BB- 78), O O HO OH (BB-81), HO OH (BB-82), OH (BB- 83), (BB- 84), (BB- 85), Y6 Y6 HO CH3 (BB- 86), (BB- 87), X3 (LNH Y6 1 N 0 Y6 Y4 0 i: —.
Hd\b (BB- 88), (BB- 89), Y6 Yeé H6 El (BB- 90), (BB- 91), (BB- 97), (BB- 98), (BB- 99), o s LNH H3C\NJJ\NH Y3 Y3 ll \ H \ Y6 F|’—Y1 S Y6 I'D-Y1 O Y4 0 Y4 0 I' l" H6 6H (BB- 100), HO: 6H (BB- 101), OH (BB- 102), HNAN/\N/VSOJmoc Y3 / Y —I?—Y1 Y4 0 (BB- 103), (BB- 104), (BB- 105), (BB- 106), (BB- 107), (BB- 108), OCH3 (BB-109), H6 6H 0), H6 6H (BB-111), (BB- 112), HO OH (BB- 113), HO bCHg HO , E)CH3(BB-115), HNJLNH ii HN NH Y3 Y3 I%—Y1 \ || \ Y6 1 O . O Y6 F|’—Y1 Y4 0 Y4 O r r HO i= (BB- 116), HO CI (313- 117), 1 (BB-119), i i HN NH HN NH ‘63 \ I3 \ Y6 F|’—Y1 O Y6 FI’—Y1 O Y4 0 Y4 0 r r H6 CH3 HO (BB- 120), OCH3 (BB-121) HNJLNH ii HN NH I3 Y3 \ II \ Y6 P—Y1 1 O Y6 Ff—Y 0 $4 0 Y4 0 r CH3 r HO OH HO (BB- 122), bCH3 (BB-123), HO OH (BB- 124), and (BB- 125), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein Y1, Y3, Y4, Y6, and r are as bed herein (e.g., each I is, independently, an integer from 0 to 5, such as from O to 3, from 1 to 3, or from 1 to 5)).
In some embodiments, the building block molecule, which may be incorporated into a polynucleotide is a modified cytidine (e. g., ed from the group consisting of: (BB- 127), (BB- 129), (BB- 130), HO OH (BB- 131), ,CH3 (BB- 132), H5 6H (BB- 133), (BB- 135), (BB- 137), NHAc NH2 TBDMS\ o |\N Y3 N/gO Y6 F|’—Y1 Y4 0 EDH (BB- 138), H6 6H (BB- 139), (BB- 140), CH3 (1313- 141), (BB- 143), (BB- 145), H6 :Br (BB- 146), (BB- 147), Ho E3H3 (BB- 148), (BB- 149), (3 6H (BB- 151), (BB- 153), (BB- 155), H6 6H (BB— 156), H6 6H (BB- 157), COszoc /\/\/I\NHFm°c (BB- 158), and (BB- 159) or a pharmaceutically acceptable salt or stereoisomer thereof, wherein Y1, Y3, Y4, Y6, and r are as described herein (e. g., each r is, independently, an integer from 0 to 5, such as from 0 to 3, from 1 to 3, or from 1 to 5)). For example, the building block molecule, which may be incorporated into a polynucleotide can be: o o o “301 o | 1H HO o_ N/go HO I'|='—o N 0 OH 0 OH H6 6H (BB- 160) or H6 6H (BB-161),ora pharmaceutically acceptable salt or stereoisomer thereof, wherein each r is, independently, an integer from 0 to 5 (e.g., from 0 to 3, from 1 to 3, or from 1 to 5).
In some embodiments, the building block molecule, which may be orated into a polynucleotide is a modified adenosine (e.g., ed from the group consisting of: N \ 3 N 1 < f) y6 Fl)_1 N N Y4 0 r CH3 H6 :OH (BB- 163), Yeé .
H6 6H (BB- 165), N \ 3 N x </ 113 Y6 I'D—Y1 N N Y4 0 H6 i= N \ 3 / N Y6 <15 F|>—Y1 N N Y4 0 H6 :Br (BB- 168), (BB- 169), Y3 / \N n I Y6 g I'D—Y1 N N Y4 0 HO E3'43 (BB- 170), (BB- 171), N \ N Y3 / | Y6 _ 1 N N/J\/ Y4 0 H6 6H (BB- 172), Y3 / |\N Y6 1 N NM Y4 0 HO: :OH (BB-173), N \ N Y3 <’ | v6 aw N NM Y4 0 H6 6H (BB- 174), II < f1 Y6 lf—1 N N/ OCH3 Y4 0 H6 6H (BB- 175), H6 OH (BB- 177), (BB- 178), H6 6H (BB- 179), NH2 NHZ N N Y3 \N H c-</ | Y3 \N n /J " \—</ | Y6 1'3— 1 N N/ Y F|’—Y1 N /J Y4 0 Y4 0 r r HO OH (BB- 180), HO OH (BB- 181), NH2 NH2 < N N y3 / \N Y3 / n I || I Y6 A A FI)_Y1 N N Y6 F|)_Y1 N N Y4 0 Y4 0 r r HO OH (BB- 182), HO CH (33- 183), \“(3 Br—</ Y6 F|’-1 N Y4 0 H6 :OH (BB- 185), || |—</ 1"“ N Y4 0 (BB- 186), H6 6H (BB- 187), || \s—</N Y6 Fl)_ 1 N Y4 0 (BB- 188), H6 6H (BB- 189), 3 N || s—</ Y6 Fl)_ 1 N Y4 0 (BB- 190), H6 6H (BB- 191), I3 S—<’ FI’_Y1 N Y4 0 H6 10H Y3 / Y4 0 (BB- 194), H6 6H (BB- 195), Fl)— 1 N Y4 0 (BB- 196), H6 6H (BB— 197), (BB- 198), Y3 < \N u | 6 1 N A Y F|’— N Y4 0 HO :OH (BB- 199), and HNAVONNHZ N 5 Y3 </ \N " I 6 F|’—Y1 N A Y N Y4 0 H6 6H (BB- 200) or a pharmaceutically acceptable salt or stereoisomer thereof, n Y1, Y3, Y4, Y6, and r are as described herein (e.g., each r is, independently, an integer from 0 to 5, such as from 0 to 3, from 1 to 3, or from 1 to 5)).
In some embodiments, the building block molecule, which may be orated into a polynucleotide, is a modified guanosine (e. g., selected from the group consisting of: 3 </ NH K ' A Y6 If-Y1 N N NH2 Y4 0 r CH3 HO OH (BB-201), I NH2 Y4 0 H6 63"13 (BB- 202), Y6 1” N/ NH2 H6 6H (BB- 203), HO 0 (BB- 204), H6 F (BB- 205), I N NH2 Y4 0 HO: El (BB- 206), I3 < If:/ NH Y6 'IL 1 N N NH2 Y4 0 H6 Er (BB-207), H6 EDI (BB- 208), N NH2 HO CH3 (BB-209), I N NH2 Y4 0 HO bCH3 (BB- 210), 3 NH Y6 FI)_Y1 N N N/\ Y4 O H H5 6H 1), 3 / NH K ' A Y6 If— 1 N N N Y4 O H H6 6H (BB- 212), -7]- I N Y4 O H HO: :OH 3), OCH3 N \ II < I 1 Y6 53-1 N / N NH2 Y4 0 H6 6H (BB-214), N \ ll < I 1 Y6 If—1 / N N NH2 Y4 0 H6 EDI (BB-215), O/\/ N \ ll < . 1 Y6 If-1 / N N NH2 Y4 0 H6 6H (BB- 216), " < P“ 6 1 N 7k Y If- N NH2 Y4 0 H6 :OH (BB-217), 3 NH 6 > Y E— N N NH2 Y4 0 H6 6H (BB-218), \_</N Y3 NH " I A Y6 If- 1 N N NH2 Y4 0 H6 6H (BB-219), H6 6H (BB- 220), 3 / NH I l 6 1 N 6i\ Y I?— N NH2 Y4 0 HO: 2OH (BB-221), 3 NH I H” ' A Y6 I'D— 1 N - N NH2 Y4 0 H6 EDH 2), 3 NH I Mf: Y6 If— 1 N N NH2 Y4 0 H6 6H (BB-223), N/i”NH2 H6 6H (BB-224), N NH2 Y4 0 HO: :OH (BB-225), 3 NH x «fl Y6 I'D- N N NH2 Y4 0 H6 6H 6), \ N v3 / ll I Y6 I'D—1 N N/1HNH2 Y4 0 H6 6H (BB-227), ( N I3 H Y6 If— 1 . N N NH2 Y4 0 H6 6H (BB- 228), I N NH2 Y4 0 H6 5H 9), ? o 3 NH I S—</ f: Y6 I'D-Y1 N N NH2 Y4 0 H6 6H (BB-230), 3 S—</ If:NH I Y6 If—Y1 N N NH2 Y4 0 H6 6H (BB-231), x3 <’ fl“ Y6 FI)_Y1 N N/ N/ Y4 O H H6 6H (BB-232), I N N Y4 0 | HO: 6H 3), v3 xfiNH I A Y6 I'3'_1 <N '4 N NH2 Y o H6 6H (BB- 234), v3 / II I Y6 N/1H "34—1 ON NH2 H6 6H (BB-235), +4 N NH2 HO OH (BB- 236), and Y F." N N NH2 Y4 o HO OH (BB- 237), or a pharmaceutically acceptable salt or stereoisomer f, wherein Y1, Y3, Y4, Y6, and r are as described herein (e. g., each r is, independently, an integer from 0 to 5, such as from 0 to 3, from 1 to 3, or from 1 to 5)).
In some embodiments, the major groove chemical modification can include replacement ofC group at C-5 of the ring (e.g., for a pyrimidine nucleoside, such as cytosine or uracil) with N (e.g., replacement of the >CH group at C-5 with >NRNl group, wherein RNl is H or optionally substituted alkyl). For example, the building block molecule, which may be incorporated into a polynucleotide can be: HN NH \N NH ll \ || \ HO P—O O HO P—O O | | OH 0 OH 0 r r HO 6H (BB- 238) or HO 6H (BB- 239) or AOL JOL HN N,CH3 H30\N N,CH3 9 \ 9 \ HO F|’—O O O . HO Ff-O OH 0 OH O r r HO 6H (BB- 240) or HO 6H (BB- 241), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each r is, independently, an integer from 0 to 5 (e.g., from 0 to 3, from 1 to 3, or from 1 to 5).
In another embodiment, the major groove chemical ation can include ement ofthe hydrogen at C-5 of cytosine with halo (e.g., Br, Cl, F, or I) or optionally substituted alkyl (e.g., methyl). For example, the building block molecule, which may be incorporated into a polynucleotide can be: HO 6H (BB- 243) or NHAc AcO |\N o N’go F|’—O OH 0 H6 6H (BB- 244) or H5 6H (BB- 245), ora pharmaceutically acceptable salt or stereoisomer thereof, wherein each r is, independently, an r from 0 to 5 (e.g., from 0 to 3, from 1 to 3, or from 1 to 5).
In yet a further embodiment, the major groove chemical modification can include a fused ring that is formed by the NH2 at the C-4 on and the carbon atom at the C-5 position. For example, the building block molecule, which may be incorporated into a polynucleotide can be: HO OH (BB- 246), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each 1‘ is, independently, an integer from 0 to 5 (e. g., from O to 3, from 1 to 3, or from 1 to 5).
Modifications on the Sugar The modified nucleosides and nucleotides (e.g., building block molecules), which may be incorporated into a cleotide (e.g., RNA or mRNA, as described herein), can be modified on the sugar of the ribonucleic acid. For example, the 2’ hydroxyl group (OH) can be modified or replaced with a number of different substituents, Exemplary substitutions at the ition e, but are not limited to, H, halo, optionally substituted CM alkyl; optionally substituted CH; alkoxy; optionally substituted 06.10 aryloxy; optionally substituted C3_s cycloalkyl; optionally substituted C34; cycloalkoxy; optionally substituted C6_10 aryloxy; optionally substituted C640 aryl-C1_6 alkoxy, ally tuted C142 (heterocyclyl)oxy; a sugar (e.g., ribose, pentose, or any described herein); a polyethyleneglycol (PEG), -O(CH2CHZO)nCH2CHZOR, where R is H or optionally substituted alkyl, and n is an integer from 0 to 20 (e. g., from 0 to 4, from 0 to 8, from 0 to 10, from 0 to 16, from 1 to 4, from 1 to 8, from 1 to 10, from 1 to 16, from 1 to 20, from 2 to 4, from 2 to 8, from 2 to 10, from 2 to 16, from 2 to 20, from 4 to 8, from 4 to 10, from 4 to 16, and from 4 to 20); “locked” c acids (LNA) in which the 2’-hydroxyl is ted by a C1_6 alkylene or CH; heteroalkylene bridge to the bon of the same ribose sugar, where exemplary bridges included methylene, propylene, ether, or amino bridges; aminoalkyl, as defined herein; aminoalkoxy, as defined herein; amino as defined herein; and amino acid, as defined herein Generally, RNA includes the sugar group ribose, which is a 5-membered ring having an oxygen. Exemplary, non-limiting d nucleotides include replacement of the oxygen in ribose (e.g., with S, Se, or alkylene, such as methylene or ethylene); addition of a double bond (e.g., to replace ribose with entenyl or cyclohexenyl); ring ction of ribose (e.g., to form a 4- membered ring of cyclobutane or oxetane); ring expansion of ribose (e.g., to form a 6- or 7— membered ring having an additional carbon or heteroatom, such as for anhydrohexitol, altritol, mannitol, cyclohexanyl, exenyl, and morpholino that also has a phosphoramidate backbone); multicyclic forms (e. g., tricyclo; and “unlocked” forms, such as glycol nucleic acid (GNA) (e.g., R- GNA or S-GNA, where ribose is replaced by glycol units attached to phosphodjester bonds), threose c acid (TNA, where ribose is replace with (x-L-threofuranosyl-(3’—>2’)) and peptide nucleic acid (PNA, where 2-amino-ethyl-glycine linkages replace the ribose and phosphodjester backbone).
The sugar group can also contain one or more carbons that s the opposite stereochemical configuration than that of the corresponding carbon in ribose. Thus, a polynucleotide molecule can include nucleotides containing, e.g., arabinose, as the sugar.
Modifications on the Nucleobase The present disclosure provides for modified nucleosides and nucleotides. As described herein “nucleoside” is defined as a compound ning a sugar molecule (e. g., a pentose or ribose) or derivative f in combination with an organic base (e. g., a purine or pyrimidine) or a derivative f (also referred to herein as “nucleobase”). As described herein, “nucleotide” is defined as a nucleoside including a phosphate group. In some embodiments, the nucleosides and nucleotides described herein are generally chemically modified on the major groove face.
Exemplary non-limiting modifications include an amino group, a thiol group, an alkyl group, a halo group, or any described herein. The modified nucleotides may by synthesized by any useful method, as described herein (e.g., chemically, enzymatically, or recombinantly to include one or more modified or tural nucleosides).
The modified nucleotide base pairing encompasses not only the standard adenosine— thymine, adenosine—uracil, or guanosine-cytosine base pairs, but also base pairs formed between tides and/or modified tides sing non-standard or modified bases, wherein the arrangement of hydrogen bond donors and hydrogen bond acceptors permits en bonding between a andard base and a standard base or between two complementary non-standard base structures. One example of such andard base pairing is the base pairing between the modified nucleotide inosine and adenine, cytosine or .
] The modified nucleosides and nucleotides can include a modified nucleobase. Examples ofnucleobases found in RNA include, but are not limited to, adenine, guanine, cytosine, and uracil.
Examples of nucleobase found in DNA include, but are not limited to, adenine, guanine, cytosine, and thymine. These nucleobases can be modified or wholly replaced to provide polynucleotide molecules having enhanced properties, e. g., resistance to ses, stability, and these properties may manifest through disruption of the g of a major groove binding r. For example, the nucleosides and nucleotides described can be chemically modified on the major groove face. In some embodiments, the major groove chemical modifications can include an amino group, a thiol group, an alkyl group, or a halo group.
Table 1 below identifies the chemical faces of each canonical nucleotide. s identify the atoms comprising the respective chemical regions.
Table 1 Watson-Crld< MEJ'or Groove Minor Groove Base-pairing Face Face Face 1 NH, -9 i —P. |‘ 3 l Cvtidine: W320 0 0 0-330 0 3? . . . OH Pyrlmldlnes 0" anon cm, _0 V H ..°. | —9.
Urldlne: o-Eja o o 0—3: a (FEED—k3? cum anon anon N N 0 \ ”A 0 I o Adenosine: 0-1.5: 0:5. fig 03:0 (ful 0‘13? fig? 6, Pun‘nes H STE”o m 1‘? dl'". 8 (5' ”H ,9 (3‘.
Guanoslne: 047:0 m, o—P-_c| ° 33 6 ‘23; who-go In some embodiments, B is a modified uracil. Exemplary modified uracils include those having Formula (b 1 )-(b5): T1. T1" R120 R12c 1X R120 /R12a A 12a R10 R10 VI N N N/R \N \N I n ‘ V2 % | N n \N T2. R11 0 R11 N -'|['2' \N O ' (b1), W'W (b2), W'W (133), M?" (134), 0r R10 12c N[no W-w (b5), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein ] \\ is a single or double bond; each of T1,, T1”, T2,, and T2” is, independently, H, ally substituted alkyl, optionally tuted alkoxy, or optionally substituted thioalkoxy, or the combination of TI‘ and T1“ or the combination of T2, and T2» join together (e.g., as in T2) to form 0 (0X0), S (thio), or Se (seleno); each of V1 and V2 is, independently, O, S, N(RVb)nv, or C(RW)“, wherein nv is an integer from 0 to 2 and each RVb is, independently, H, halo, optionally substituted amino acid, optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted aminoalkyl (e. g., substituted with an N- protecting group, such as any described herein, e.g., trifluoroacetyl), optionally substituted lkenyl, optionally substituted aminoalkynyl, optionally substituted acylaminoalkyl (e.g., substituted with an N-protecting group, such as any described herein, e. g., trifluoroacetyl), optionally substituted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, or optionally substituted alkoxycarbonylalkoxy (e.g., optionally substituted with any substituent described herein, such as those selected from (l)-(21) for alkyl); R10 is H, halo, optionally tuted amino acid, hydroxy, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aminoalkyl, optionally substituted hydroxyalkyl, ally substituted hydroxyalkenyl, ally substituted yalkynyl, ally substituted aminoalkenyl, optionally substituted lkynyl, optionally substituted alkoxy, optionally substituted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted carboxyalkoxy, optionally substituted carboxyalkyl, or optionally substituted carbamoylalkyl; R11 is H or optionally substituted alkyl; R128 is H, optionally substituted alkyl, ally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted yalkynyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted lkynyl, optionally substituted carboxyalkyl (e. g., ally substituted with y), optionally substituted yalkoxy, optionally substituted carboxyaminoalkyl, or optionally substituted carbamoylalkyl; ] R120 is H, halo, optionally tuted alkyl, optionally tuted alkoxy, optionally substituted thioalkoxy, optionally substituted amino, optionally substituted yalkyl, optionally tuted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted aminoalkyl, ally substituted aminoalkenyl, or optionally tuted aminoalkynyl.
Other exemplary modified uracils include those having Formula (b6)-(b9): R12c R12c 1 . 1 .. 3* 12C /R12a 3k /R12a R12:3 \|/: ‘ N R12b\T><T R12” j: Y: ‘ N N N/ \N \ N W JVTZ" W‘ K 2" " \thl2 \ 2 2 2 vy T V?T2, RXT1",. ' (b6), ‘ (b7), WIW‘ (b8), or W (b9), or a pharmaceutically acceptable salt or stereoisomer f, n \ is a single or double bond; ] each of T1,, T1”, T2,, and T2" is, independently, H, optionally substituted alkyl, optionally substituted alkoxy, or optionally substituted thioalkoxy, or the combination of T1, and T1” join er (e.g,. as in T1) or the combination of T2, and TT join together (e. g., as in T2) to form 0 (0x0), S (thio), or Se (seleno), or each T1 and T2 is, independently, 0 (0X0), S (thio), or Se (seleno); each ofW1 and W2 is, independently, N(Rwa)DW or C(Rwa)“, wherein nw is an integer from 0 to 2 and each RW8 is, independently, H, optionally substituted alkyl, or optionally substituted each V3 is, independently, O, S, nv, or C(Rva)nv, wherein nV is an integer from 0 to 2 and each RV?1 is, independently, H, halo, optionally substituted amino acid, optionally substituted alkyl, optionally substituted hydroxyalkyl, optionally tuted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted heterocyclyl, optionally substituted erocyclyl, optionally substituted alkoxy, optionally substituted alkenyloxy, or optionally substituted alkynyloxy, optionally substituted lkyl (e. g., substituted with an N—protecting group, such as any described herein, e.g., trifluoroacetyl, or lkyl), optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted inoalkyl (e. g., substituted with an ecting group, such as any described herein, e.g., trifluoroacetyl), optionally tuted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally substituted carbonylacyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted carboxyalkyl (e. g., optionally substituted with y and/or an O-protecting group), optionally substituted carboxyalkoxy, optionally substituted carboxyaminoalkyl, or optionally substituted carbamoylalkyl (e.g., optionally substituted with any substituent described herein, such as those selected from (l)-(21) for alkyl), and wherein RV3 and R120 taken together with the carbon atoms to which they are attached can form optionally substituted cycloalkyl, optionally substituted aryl, or optionally substituted heterocyclyl (e. g., a 5- or 6-membered ring); R128 is H, ally substituted alkyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted carboxyalkyl (e. g., ally tuted with hydroxy and/or an 0-protecting group), optionally substituted carboxyalkoxy, ally tuted carboxyaminoalkyl, optionally tuted carbamoylalkyl, or absent; R12b is H, optionally substituted alkyl, optionally substituted alkenyl, optionally tuted alkynyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted lkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted alkaryl, optionally substituted heterocyclyl, optionally substituted alkheterocyclyl, optionally substituted amino acid, optionally substituted alkoxycarbonylacyl, optionally substituted alkoxycarbonylalkoxy, optionally tuted alkoxycarbonylalkyl, optionally substituted carbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylalkoxy, optionally tuted carboxyalkyl (e.g., optionally substituted with hydroxy and/or an 0-protecting group), optionally substituted carboxyalkoxy, optionally substituted carboxyaminoalkyl, or optionally substituted carbamoylalkyl, wherein the combination of R12b and T1, or the combination of R12b and Rm can join together to form optionally substituted heterocyclyl; and Rm is H, halo, optionally tuted alkyl, optionally substituted alkoxy, optionally substituted koxy, optionally substituted amino, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl.
Further exemplary modified uracils include those having Formula (b28)-(b31): T1 T1 T1 RVb' R12a / RVb' / R123 R12b N \ A R122: I l I N N RVb" N T2 NkTZ KKK-r2 ' (1328), (b29), “t” (b30), or RVb' R1 2a Nk 2 T . (b3l), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein ] each of T1 and T2 is, independently, O (oxo), S (thio), or Se o); each RVb’ and RV” is, independently, H, halo, optionally substituted amino acid, optionally tuted alkyl, optionally substituted haloalkyl, ally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted alkenyl, ally tuted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted loxy, optionally substituted aminoalkyl (e. g., tuted with an N—protecting group, such as any described herein, e.g., trifluoroacetyl, or sulfoalkyl), optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally tuted acylaminoalkyl (e.g., substituted with an ecting group, such as any described , e.g., trifluoroacetyl), optionally substituted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylacyl, ally substituted alkoxycarbonylalkoxy, optionally substituted carboxyalkyl (e.g., optionally substituted with hydroxy and/or an O-protecting group), optionally substituted carboxyalkoxy, optionally substituted carboxyaminoalkyl, or optionally substituted carbamoylalkyl (e. g., optionally substituted with any substituent described herein, such as those selected from (l)-(21) for alkyl) (e. g., RVb’ is optionally substituted alkyl, optionally substituted alkenyl, or optionally substituted aminoalkyl, e. g., substituted with an N—protecting group, such as any bed herein, e. g., trifluoroacetyl, or sulfoalkyl); R123 is H, optionally substituted alkyl, optionally substituted carboxyaminoalkyl, optionally substituted aminoalkyl (e. g., e.g., tuted with an N—protecting group, such as any described herein, e.g., trifluoroacetyl, or sulfoalkyl), optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl; and R121) is H, optionally substituted alkyl, optionally tuted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl, optionally tuted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted aminoalkyl, ally substituted aminoalkenyl, optionally substituted aminoalkynyl (e.g., e.g., substituted with an N-protecting group, such as any described herein, e. g., trifluoroacetyl, or sulfoalkyl), optionally substituted alkoxycarbonylacyl, ally substituted alkoxycarbonylalkoxy, optionally substituted alkoxycarbonylalkyl, ally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally tuted alkoxycarbonylalkoxy, optionally substituted carboxyalkoxy, optionally substituted carboxyalkyl, or optionally substituted carbamoylalkyl.
In particular embodiments, T1 is 0 (0x0), and T2 is S (thio) or Se (seleno). In other embodiments, T1 is S (thio), and T2 is 0 (0x0) or Se o). In some embodiments, RV”, is H, optionally substituted alkyl, or ally substituted alkoxy.
In other embodiments, each R123 and R12b is, ndently, H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted hydroxyalkyl. In particular embodiments, R1221 is H. In other embodiments, both R128 and R12.) are In some embodiments, each RWY of R12b is, independently, optionally substituted aminoalkyl (e. g., substituted with an N—protecting group, such as any described herein, e.g., roacetyl, or lkyl), optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, or optionally substituted acylaminoalkyl (e. g., tuted with an N—protecting group, such as any described herein, e. g., trifluoroacetyl). In some embodiments, the amino and/or alkyl of the optionally substituted aminoalkyl is substituted with one or more of optionally substituted alkyl, optionally substituted alkenyl, optionally substituted sulfoalkyl, optionally substituted carboxy (e.g., tuted with an O-protecting , optionally substituted hydroxy (e. g., substituted with an 0- protecting group), optionally substituted carboxyalkyl (e.g., substituted with an O-protecting group), optionally substituted alkoxycarbonylalkyl (e. g., substituted with an O-protecting group), or N- protecting group. In some embodiments, optionally tuted aminoalkyl is substituted with an optionally substituted sulfoalkyl or optionally substituted alkenyl. In particular embodiments, R123 and RW’ are both H. In particular embodiments, T1 is 0 (0X0), and T2 is S (thio) or Se (seleno).
In some embodiments, RV”, is optionally substituted alkoxycarbonylalkyl or optionally substituted carbamoylalkyl.
In particular embodiments, the optional substituent for R123, Rm, Rm, or RVa is a polyethylene glycol group (e.g., -(CH2)52(OCH2CH2)51(CH2)53OR’, wherein s1 is an r from 1 to (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an r from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R’ is H or C140 alkyl); or an amino-polyethylene glycol group (e.g., CH2)s2(CH2CH20)51(CH2)53NRN1, wherein s] is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and 53, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each RNl is, independently, hydrogen or optionally substituted C1_6 alkyl).
In some embodiments, B is a modified cytosine. Exemplary modified cytosines include compounds ofFormula (b 10)—(b14): 13a 13b 13b 13a 13b 13a 13b R R \N NR R \N/R R R \vs NkN V4J§N R15 NJV I ilrli/JYI—T: R15)\N/J:J3l REM??? “1“" (1)10),R “T” (b11),“"7w (b12), “’“x‘” (bl3) or v4/§N R15)\NI T3..
I T3 vav- I (bl4), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each of T3, and T3” is, independently, H, optionally substituted alkyl, optionally substituted alkoxy, or optionally substituted thioalkoxy, or the combination of T3, and T3” join together (e.g., as in T3) to form 0 (oxo), S (thio), or Se (seleno); each V4 is, ndently, O, S, N(va)nv, 0r C(va)nv, wherein nV is an integer from 0 to 2 and each RVC is, independently, H, halo, optionally substituted amino acid, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted l, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted heterocyclyl, optionally tuted alkheterocyclyl, or optionally substituted alkynyloxy (e.g., optionally substituted with any substituent described herein, such as those selected from (l)—(2l) for , n the combination of R13b and RVC can be taken together to form optionally substituted heterocyclyl; each V5 is, ndently, N(RVd)nv, or nv, wherein nv is an integer from 0 to 2 and each RVd is, independently, H, halo, optionally substituted amino acid, optionally tuted alkyl, optionally substituted alkenyl, ally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted heterocyclyl, optionally substituted alkheterocyclyl, or ally substituted alkynyloxy (e.g., optionally substituted with any substituent described herein, such as those selected from (l)-(21) for alkyl) (e.g., V5 is —CH or N); each of R13a and R131) is, independently, H, optionally substituted acyl, optionally substituted acyloxyalkyl, optionally substituted alkyl, or optionally substituted alkoxy, wherein the combination of R131) and R14 can be taken together to form optionally substituted heterocyclyl; each R14 is, independently, H, halo, hydroxy, thiol, optionally substituted acyl, optionally substituted amino acid, optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted l, ally substituted alkynyl, ally substituted hydroxyalkyl (e.g., substituted with an 0-protecting group), optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, ally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted acyloxyalkyl, optionally substituted amino (e.g., -NHR, wherein R is H, alkyl, aryl, or oryl), azido, optionally tuted aryl, optionally substituted heterocyclyl, optionally substituted alkheterocyclyl, optionally substituted aminoalkyl, optionally substituted lkenyl, or optionally substituted aminoalkynyl; and each of R15 and R16 is, ndently, H, optionally substituted alkyl, optionally substituted alkenyl, or optionally substituted alkynyl.
Further exemplary modified cytosines include those having Formula (b32)-(b35): R13a\N/R13b N’R13b T1 R13a\N/R13b R14 R14 16 R14 R14 \ N N’R \N \ I ‘ IN (R13a R15 N/kTS R15 N/KTa VII“ R15 N M)“, MAM WW 13b w'w . (b32), i (b33), . R (b34), or . (b35), or a pharmaceutically acceptable salt or isomer thereof, wherein each of T1 and T3 is, independently, O (oxo), S (thio), or Se (seleno); each of R1321 and R131) is, ndently, H, optionally tuted acyl, optionally tuted yalkyl, optionally substituted alkyl, or optionally substituted alkoxy, n the combination of R131) and R14 can be taken together to form optionally substituted heterocyclyl; each R14 is, independently, H, halo, hydroxy, thiol, optionally tuted acyl, optionally substituted amino acid, optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl (e.g., substituted with an 0-protecting group), optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted alkoxy, optionally substituted loxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally tuted alkoxy, optionally substituted acyloxyalkyl, optionally substituted amino (e.g., -NHR, wherein R is H, alkyl, aryl, or phosphoryl), azido, optionally substituted aryl, optionally substituted heterocyclyl, optionally substituted alkheterocyclyl, optionally substituted aminoalkyl (e.g., hydroxyalkyl, alkyl, alkenyl, or alkynyl), ally substituted aminoalkenyl, or ally substituted aminoalkynyl; and ] each of R15 and R16 is, independently, H, optionally substituted alkyl, optionally substituted alkenyl, or optionally substituted alkynyl (e. g., R15 is H, and R16 is H or ally substituted alkyl).
In some embodiments, R15 is H, and R16 is H or optionally substituted alkyl. In particular embodiments, R14 is H, acyl, or hydroxyalkyl. In some ments, R14 is halo. In some embodiments, both R14 and R15 are H. In some embodiments, both R15 and R16 are H. In some ments, each of R14 and R15 and R16 is H. In further embodiments, each of R133 and R13]) is independently, H or optionally substituted alkyl. r non-limiting examples of modified cytosines include compounds of Formula (b36): R143 | IN R15 N/J\R14b . (b36) or a ceutically acceptable salt or stereoisomer thereof, wherein each R13b is, independently, H, optionally substituted acyl, optionally substituted acyloxyalkyl, optionally substituted alkyl, or optionally substituted alkoxy, wherein the combination of R13') and R141) can be taken together to form optionally substituted heterocyclyl; each R1421 and R141) is, ndently, H, halo, hydroxy, thiol, optionally substituted acyl, optionally substituted amino acid, optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally tuted hydroxyalkyl (e.g., substituted with an 0-protecting group), ally substituted hydroxyalkenyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally tuted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted yalkyl, optionally substituted amino (e.g., -NHR, wherein R is H, alkyl, aryl, phosphoryl, optionally substituted aminoalkyl, or optionally substituted carboxyaminoalkyl), azido, optionally substituted aryl, optionally substituted heterocyclyl, optionally substituted alkheterocyclyl, optionally tuted aminoalkyl, ally substituted aminoalkenyl, or optionally tuted aminoalkynyl; each of R15 is, independently, H, optionally tuted alkyl, optionally substituted alkenyl, or optionally substituted alkynyl.
In particular embodiments, R14b is an optionally substituted amino acid (e. g., optionally substituted lysine). In some embodiments, R145 is H.
In some embodiments, B is a modified guanine. Exemplary modified guanines include compounds ula (b15)—(b17): fr 5' MfrR23 T5' T5" N (R18 21 / /&R24 N 17 / R fig 1'2”" (b15) (b16), or W‘” l?” (bl7), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein Each of T4), Tl", T5,, T5”, Téy, and T6” is, independently, H, optionally substituted alkyl, or optionally substituted alkoxy, and wherein the combination of T4) and T4” (e. g., as in T) or the combination of T5, and T5“ (e. g., as in T5) or the combination of T6, and 16" join together (e.g., as in T6) form 0 (oxo), S (thio), or Se (seleno); each of V5 and V6 is, ndently, O, S, N(RVd)uv, 0r C(RVd)nv, wherein nV is an integer from 0 to 2 and each RVd is, independently, H, halo, thiol, optionally substituted amino acid, cyano, amidine, optionally substituted aminoalkyl, optionally substituted lkenyl, optionally substituted aminoalkynyl, ally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally tuted alkenyloxy, optionally substituted alkynyloxy (e. g., optionally substituted with any substituent described herein, such as those selected from (l)-(21) for alkyl), optionally tuted thioalkoxy, or optionally substituted amino; and each of R17, R18, R193, Rm, R21, R22, R23, and R24 is, independently, H, halo, thiol, optionally substituted alkyl, optionally tuted alkenyl, optionally substituted alkynyl, optionally substituted thioalkoxy, optionally substituted amino, or optionally substituted amino acid.
Exemplary modified guanosines include nds of Formula (b37)-(b40): 1.4 T4' 1“ miN R13 N \ </ N r / N 193 A 19a / / 193 h" N’R h“ N’R n“ ’ R N N N N I | W | l R19b (1337), . R19b (b38), I R19b (b39), or ’ R13 ’ R19a N N W | ' Rm, (b40), or a ceutically acceptable salt or stereoisomer thereof, wherein each of T4, is, independently, H, optionally substituted alkyl, or ally substituted alkoxy, and each T4 is, independently, O (oxo), S (thio), or Se (seleno); each of R18, R193, ngb, and R21 is, independently, H, halo, thiol, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted thioalkoxy, optionally substituted amino, or optionally substituted amino acid.
In some embodiments, R18 is H or optionally substituted alkyl. In further embodiments, T4 is oxo. In some embodiments, each ofRM1 and R19b is, independently, H or optionally tuted alkyl.
In some embodiments, B is a modified adenine. Exemplary modified adenines include compounds ofFormula (b l 8)—(b20): 26a 26b 26b R R R \ / / N N‘ R29 V7 \ V7 (R28 V7 N N \ N R </ l A 25 </ 25 R i A R </ i A “5:“ N R27 WNW N R27 W]: N R27 . (bl 8), ‘ (b19), or ‘ (b20), or a pharmaceutically able salt or isomer f, wherein each V7 is, independently, O, S, N(Rve)nv, 0r C(Rve)nv, wherein nV is an integer from 0 to 2 and each RVe is, independently, H, halo, optionally substituted amino acid, ally substituted alkyl, optionally substituted alkenyl, optionally substituted l, optionally substituted alkoxy, optionally substituted alkenyloxy, or optionally substituted alkynyloxy (etgt, optionally substituted with any substituent described herein, such as those selected from (1)-(21) for alkyl); each R25 is, independently, H, halo, thiol, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, ally substituted thioalkoxy, or optionally substituted amino; each of R263 and R261) is, independently, H, optionally substituted acyl, optionally substituted amino acid, ally substituted carbamoylalkyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, ally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally tuted alkoxy, or polyethylene glycol group (e. g., 52(OCH2CH2)51(CH2)530R’, wherein S] is an r from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of $2 and 53, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R’ is H or C140 ; or an amino-polyethylene glycol group (e.g., -NRN1(CH2)52(CH2CHZO)51(CH2)53NRN', wherein S] is an r from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of 52 and 53, independently, is an integer from 0 to 10 (e. g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each RNl is, independently, hydrogen or optionally substituted CH; alkyl); each R27 is, independently, H, optionally substituted alkyl, optionally substituted l, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted thioalkoxy, or ally substituted amino; each R28 is, independently, H, optionally substituted alkyl, optionally substituted alkenyl, or optionally substituted alkynyl; and each R29 is, independently, H, optionally substituted acyl, optionally substituted amino acid, optionally substituted carbamoylalkyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted alkoxy, or optionally substituted amino.
Exemplary modified adenines include compounds of Formula (b41)-(b43): R263\ N/R26b R26a\ N/R26b R26a\N/R26b N \ N N \ N \ </ I R254</ ‘ ‘ / /)N </ /)N N N R27 N N N N w-‘M (b41), W‘W‘ (b42), or "‘1” (b43), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each R25 is, independently, H, halo, thiol, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted l, optionally substituted thioalkoxy, or optionally substituted amino; each of R263 and R261) is, independently, H, optionally substituted acyl, optionally substituted amino acid, optionally substituted carbamoylalkyl, optionally tuted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl, ally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted alkoxy, or polyethylene glycol group (e. g., -(CH2)52(OCH2CH2)51(CH2)530R’, wherein S] is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of $2 and 53, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R’ is H or C140 alkyl); or an amino-polyethylene glycol group (e.g., CH2)52(CH2CHZO)51(CH2)53NRN', wherein S] is an r from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of 52 and 53, independently, is an integer from 0 to 10 (e. g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each RNl is, independently, hydrogen or optionally tuted CH; alkyl); and each R27 is, independently, H, optionally substituted alkyl, optionally substituted alkenyl, ally substituted alkynyl, optionally substituted alkoxy, optionally substituted thioalkoxy, or optionally substituted amino.
In some embodiments, R2661 is H, and R26b is optionally substituted alkyl. In some embodiments, each of R263 and R26b is, independently, optionally tuted alkyl. In particular embodiments, R27 is ally substituted alkyl, optionally substituted alkoxy, or optionally substituted koxy. In other embodiments, R25 is ally substituted alkyl, optionally substituted alkoxy, or optionally tuted thioalkoxy.
In particular embodiments, the optional sub stituent for R263, R2“, or R29 is a polyethylene glycol group (e. g., -(CH2)52(OCH2CH2)51(CH2)530R’, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of 52 and s3, independently, is an integer from 0 to 10 (eg, from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R’ is H or C1_20 alkyl); or an amino- polyethylene glycol group (e.g., -NRN1(CH2)52(CH2CH20)31(CH2)53NRN1, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of $2 and s3, independently, is an r from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each RN1 is, independently, hydrogen or optionally substituted C1.6 alkyl).
In some embodiments, B may have Formula (b21): ‘ (b21), wherein X12 is, independently, O, S, optionally substituted alkylene (e. g., methylene), or optionally substituted heteroalkylene, xa is an integer from 0 to 3, and R123 and T2 are as described herein.
In some ments, B may have Formula (b22): 0 T1 R10I\N N/R12a H | 11 k2 R N T I (b22), wherein R10) is, independently, optionally tuted alkyl, optionally substituted l, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heterocyclyl, optionally substituted aminoalkyl, ally substituted aminoalkenyl, optionally tuted aminoalkynyl, optionally substituted alkoxy, optionally substituted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylalkoxy, optionally tuted carboxyalkoxy, optionally substituted carboxyalkyl, or ally substituted carbamoylalkyl, and R11, R12“, T1, and T2 are as described herein.
In some embodiments, B may have Formula (b23): R10 N’R12a R11 N/KTz I (b23), wherein R10 is optionally substituted heterocyclyl (e. g., ally substituted furyl, optionally substituted l, or optionally substituted pyrrolyl), optionally substituted aryl (e.g., optionally tuted phenyl or optionally substituted naphthyl), or any substituent described herein (e. g., for R10) ;and wherein R11 (e. g., H or any substituent described herein), R12a (e.g., H or any substituent described herein), T1 (e. g., oxo or any substituent described herein), and T2 (e. g., oxo or any substituent described herein) are as described herein.
In some embodiments, B may have Formula (b24): o N R14\N \ N H l R15 N/k-I-S I (b24), wherein R14, is, independently, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted l, optionally substituted aIyl, optionally tuted heterocyclyl, optionally substituted alkaryl, optionally substituted alkheterocyclyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted alkoxy, optionally substituted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted carboxyalkoxy, optionally substituted carboxyalkyl, or optionally tuted carbamoylalkyl, and R133, Rm, R15, and T3 are as described herein.
In some embodiments, B may have Formula (b25): R13a\ ’ R13b o N R14'/\N \ N HR15 I NAT?’ Mi" (b25), wherein R14, is optionally substituted heterocyclyl (e.g., optionally substituted furyl, optionally substituted thienyl, or optionally substituted pyrrolyl), optionally substituted aryl (e.g., optionally substituted phenyl or optionally substituted yl), or any substituent described herein (e. g., for R14 or R”); and n R133 (e. g., H or any substituent described herein), R13b (e. g., H or any substituent bed herein), R15 (e.g., H or any substituent described herein), and T3 (e.g., oxo or any substituent described herein) are as bed herein.
In some embodiments, B is a nucleobase selected from the group consisting of cytosine, guanine, adenine, and uracil. In some embodiments, B may be: YN‘N/ \r/Nw/ NH2 (urn, <>11qu 9 </N \fi/O N N/J N N/) N N/J . ~.w (b26) or “-W (1327)- In some embodiments, the modified nucleobase is a modified , Exemplary nucleobases and nucleosides having a modified uracil include pseudouridine (w), none ribonucleoside, S-aza-uridine, uridine, 2-thioaza-uridine, 2-thio-uridine (szU), 4-thio- undine (s4U), 4-thio-pseudouridine, 2-thio-pseudouridine, 5-hydroxy-u1idine (hosU), S-aminoallyl- undme, 5-halo-uridine (e. g., -uxidineor 5-bromo-u1idine), 3-methyl-u1idine (m3U), 5- methoxy-uridine (mosU), uridine 5-oxyacetic acid (cmosU), undine 5-oxyacetic acid methyl ester (mcmoSU), 5-carboxymethyl-uridine (cmsU), 1-carboxymethyl-pseudouridine, 5- caIboxyhydxoxymethyl-uridine (chmsU), 5-carboxyhydroxymethyl—uridine methyl ester (mchmsU), oxycarbonylmethyl-undme (mcmSU), 5-methoxycarbonylmethyl—2-thio-uridine (mcm5s2U), -aminomethyl-Z-thio-uridine (nmsszU), 5-methylaminomethyl-u1idine (nmmsU), 5- methylaminomethylthio-u1idine zU), 5-methylaminomethylseleno-uiidine (mnmsser), -carbamoylmethyl-u1idine (ncmSU), 5-caIboxymethylaminomethyl-uridine (cmnmSU), 5- carboxymethylaminomethylthio-uridine (cmnmsszU), 5-propynyl-uridine, 1-propynyl- pseudouridine, inomethyl-u1idine (rmSU), 1-taurinomethyl—pseudouridine, 5-taurinomethyl thio-uridine(rm5s2U), inomethylthio-pseudouridine, 5-methyl-uridine (mSU, i.e., having the nucleobase deoxythymine), yl-pseudouridine (mlw), 5-methylthio-u1idine (msszU), l- melhyl—4—thio-pseudouridine (m1s4w), 4-thiomethyl-pseudouridine, 3-methyl-pseudouridine (m3\|/), 2—thio- 1-methyl—pseudouridine, 1-methyl— 1-deaza-pseudouridine, 2-thio-1 -methyl-l -deaza— pseudouridine, ouridine (D), dihydropseudouridine, 5,6-dihydrouridine, 5-methyldihydrouridine (mSD), 2-thio-dihydrouridine, 2-thio-dihydropseudouridine, 2-meth0xy-uridine, 2- methoxythio-uridine, 4-methoxy-pseudouridine, 4-methoxythio-pseudouridine, N1 -methyl— pseudouridine, 3-(3-aminocarboxypropyl)uridine (acp3U), 1-methyl(3-amino ypropyl)pseudouridine (acp3 w), 5-(isopentenylaminomethyl)uridine (inmSU), 5- (isopentenylaminomethyl)thio-uridine (inmsszU), (x-thio-uridine, 2’-O-methyl-uridine (Um), 5,2’- O—dimethyl-uridine (mSUm), ethyl—pseudouridine (wm), 2-thio-2’-O-methyl—uridine (s2Um), -methoxycarbonylmethyl-2’-O-methyl-uridine (mcmSUm), 5-carbamoylmethyl-2’-O-methyl-uridine (ncm5Um), 5-carboxymethylaminomethyl—Z’-O-methyl-uridine (cmnmSUm), 3,2’-O-dimethyl- uridine (m3Um), and 5-(isopentenylaminomethyl)—2’-O-methyl-uridine m), l-thio-uridine, deoxythymidine, 2'-F-ara-uridine, 2'-F-uridine, 2'-OH-ara-uridine, 5-(2-carbomethoxyvinyl) uridine, and 5-[3-(1-E-propenylamino)uridine.
] In some ments, the modified nucleobase is a modified cytosine Exemplary nucleobases and nucleosides having a modified ne include 5-aza—cytidine, 6-aza-cytidine, pseudoisocytidine, 3-methyl-cytidine (m3C), N4-acetyl-cytidine (ac4C), 5-formyl-cytidine (fSC), N4— methyl-cytidine (m4C), 5-methyl-cytidine (mSC), 5-halo-cytidine (eg‘, 5-iodo-cytidine), 5- hydroxymethyl-cytidine (thC), l-methyl-pseudoisocytidine, pyrrolo-cytidine, pyrrolo- pseudoisocytidine, 2-thio-cytidine (52C), methyl-cytidine, 4-thio-pseudoisocytidine, 4-thiol-methyl-pseudoisocytidine , 4-thiomethyl-l-deaza-pseudoisocytidine, l-methyl- l-deaza- pseudoisocytidine, zebularine, S-aza-zebularine, yl-zebularine, 5-azathio-zebularine, 2- thio-zebularine, 2-methoxy—cy‘tidine, 2-methoxymethyl-cytidine, oxy-pseudoisocytidine, 4— methoxy-l-methyl-pseudoisocytidine, lysidine (kgC), a—thio-cytidine, 2’-O-methyl—cytidine (Cm), ,2’-O-dimethyl-cytidine (mSCm), N4-acetyl-2’-O-methyl-cytidine (ac4Cm), N4,2’-O-dimethylcytidine (m4Cm), 5-formyl—2’-O-methyl-cytidine (fSCm), N4,N4,2’-O-t1imethyl-cytidine (m42Cm), l- thio-cytidine, 2'-F-ara-cytidine, 2'-F-cytidine, and 2'-OH-ara-cytidine.
In some embodiments, the modified nucleobase is a d adenine. Exemplary nucleobases and nucleosides having a d adenine include 2-amino-pu1ine, 2, 6-diaminopurine, 2-amino—6—halo-pu1ine (e.g., 2-aminochloro-purine), 6-halo-purine (e. g., 6-chloro-purine), 2- amino-6—methyl-purine, 8-azido-adenosine, 7-deaza—adenine, 7-deaza—8-aza—adenine, a—2- amino—purine, 7-deazaaza—2-amino-purine, 7-deaza—2,6-diaminopurine, a—8-aza—2,6- diaminopurine, l-methyl—adenosine (mlA), 2-methyl—adenine (mZA), hyl—adenosine (m6A), 2-methylthio-N6-methyl-adenosine (mszmfiA), N6-isopentenyl—adenosine (iéA), 2-methylthio-N6— isopentenyl-adenosine (mszifiA), N6-(cis-hydroxyisopentenyl)adenosine (ioéA), 2-methylthio—N6- (cis-hydroxyisopentenyl)adenosine (msziosA), N6-glycinylcarbamoyl-adenosine (géA), N6— threonylcarbamoyl-adenosine (téA), hyl-N6-threonylcarbamoyl-adenosine (métGA), 2- methylthio-N6-threonylcarbamoyl—adenosine (mszgéA), N6,N6-dimethyl-adenosine (m62A), N6- hydroxynorvalylcarbamoyl—adenosine (hnGA), 2-methylthio-N6-hydroxynorvalylcarbamoyl— adenosine (mszhnéA), N6-acetyl—adenosine (acsA), 7-methyl-adenine, 2-methylthio-adenine, 2- methoxy-adenine, (x-thio-adenosine, 2’-O-methyl—adenosine (Am), N6,2’-O-dimethyl-adenosine (mfiAm), N6,N6,2’-O-trimethyl-adenosine (mézAm), l,2’-O-dimethyl-adenosine (mlAm), 2’-O- ribosyladenosine (phosphate) (Ar(p)), 2-amino-N6-methyl—purine, -adenosine, 8—azido- adenosine, ra-adenosine, 2'-F-adenosine, 2'-OH-ara-adenosine, and N6-( l 9-amino-pentaoxanonadecyl)-adenosine.
In some embodiments, the modified base is a modified guanine, Exemplary nucleobases and nucleosides having a modified guanine e inosine (I), l-methyl-inosine (mil), wyosine (imG), methylwyosine (mimG), 4-demethyl-wyosine (imG—14), isowyosine (imG2), sine (yW), peroxywybutosine (OzyW), hydroxywybutosine (OHyW), undermodifled hydroxywybutosine (OHyW*), 7-deaza-guanosine, ine (Q), epoxyqueuosine (0Q), galactosyl- queuosine (galQ), mannosyl—queuosine (manQ), 7-cyanodeaza—guanosine (prer), ?- aminomethyldeaza-guanosine (prte), archaeosine (G+), 7-deazaaza-guanosine, 6-thio- guanosine, 6-thiodeaza-guanosine, 6-thiodeazaaza-guanosine, 7-methyl-guanosine (m7G), methyl-guanosine, 7-methyl-inosine, 6-methoxy-guanosine, 1-methyl-guanosine (mlG), N2-methyl-guanosine (mZG), N2,N2-dimethyl-guanosine (mzzG), N2,7-dimethyl-guanosine (m2‘7G), N2, N2,7-dimethyl-guanosine (m2’2'7G), 8-oxo-guanosine, 7-methyloxo-guanosine, l-methyl thio-guanosine, N2-methylthio-guanosine, N2,N2-dimethyl—6-thio-guanosine, o-guanosine, 2’-O-methyl—guanosine (Gm), N2-methyl-2’-O-methyl-guanosine (szm), N2,N2-dimethyl-2’-O- -guanosine (mzsz), l-methyl-2’-O-methyl-guanosine (mle), N2,7-dimethyl-2’-O-methyl- ine (m2‘7Gm), 2’-O-methyl-inosine (Im), l,2’-O-dimethyl—inosine (mllm), 2’-O- ribosylguanosine (phosphate) (Gr(p)) , l-thio-guanosine, O6-methyl-guanosine, 2'-F-ara-guanosine, and 2'-F-guanosine.
] In some embodiments, the nucleotide can be modified on the major groove face. For example, such modifications include replacing hydrogen on C-5 of uracil or cytosine with alkyl (e.g., methyl) or halo.
The nucleobase of the nucleotide can be independently selected from a purine, a pyrimidine, a purine or pyrimidine analog. For example, the nucleobase can each be independently selected from adenine, cytosine, guanine, uracil, or nthine. In r embodiment, the nucleobase can also e, for example, lly-occurring and synthetic derivatives of a base, including lo [3 ,4—d]pyrimidines, 5-methylcytosine (5-me—C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2-thiocytosine, 5-propynyl uracil and cytosine, 6-azo uracil, cytosine and thymine, il (pseudouracil), 4-thiouracil, 8-halo (e.g., 8-bromo), 8-amino, l, 8-thioalkyl, 8-hydroxyl and other 8-substituted adenines and guanines, 5-halo particularly S-bromo, 5-trifluoromethyl and other -substituted uracils and cytosines, 7-methylguanine and 7-methyladenine, 8-azaguanine and 8- azaadenine,deazaguanine,7-deazaguannie,3-deazaguanine,deazaadenine,7-deazaadenine,3- deazaadenine, lo [3 ,4-d]pyrimidine, imidazo [ l ,5-a] 1 ,3 ,5 triazinones, 9-deazapurines, imidazo[4,5-d]pyrazines, thiazolo[4,5-d]pyrimidines, pyrazin-Z-ones, 1,2,4-t1iazine, pyridazine; and 1,3,5 triazine. When the nucleotides are depicted using the shorthand A, G, C, T or U, each letter refers to the representative base and/or tives thereof, e.g., A includes adenine or adenine analogs, e.g., 7-deaza adenine).
In some embodiments, the modified nucleotide is a compound of Formula XI: wherein: \\ denotes a single or a double bond; — - -denotes an optional single bond; U is O, S, -NRa-, or -CRaRb- when \ s a single bond, or U is -CRa- when \‘\~ denotes a double bond; Z is H, C142 alkyl, or C640 aryl, or Z is absent when \ denotes a double bond; and Z can be -CRaRb- and form a bond with A; A is H, OH, NHR wherein R: alkyl or aryl or phosphoryl, sulfate, -NH2, N3, azido, -SH, N an amino acid, or a peptide comprising 1 to 12 amino acids; D is H, OH, NHR n R: alkyl or aryl or oryl, -NH2, -SH, an amino acid, a peptide comprising 1 to 12 amino acids, or a group of Formula XII: or A and D together with the carbon atoms to which they are attached form a 5-membered ring; X is O or S; each of Y1 is independently selected from —ORa1, Rb1, and —SR“1; each of Y2 and Y3 are independently selected from O, -CRaRb-, NRc, S or a linker comprising one or more atoms selected from the group consisting of C, O, N, and S; n is 0, 1,2, or 3; In is 0, 1,2 or 3; B is nucleobase; ] R3 and Rb are each independently H, C142 alkyl, C242 alkenyl, C242 alkynyl, or C620 aryl; Rc is H, C142 alkyl, C242 alkenyl, phenyl, benzyl, a polyethylene glycol group, or an amino-polyethylene glycol group; ] R31 and R1,1 are each independently H or a rion; and —ORCl is OH at a pH of about 1 or —ORCl is O- at physiological pH; ] provided that the ring encompassing the variables A, B, D, U, Z, Y2 and Y3 cannot be ribose.
In some embodiments, B is a nucleobase selected from the group consisting of cytosine, guanine, adenine, and uracil.
In some ments, the nucleobase is a pyrimidine or derivative thereof.
In some embodiments, the modified nucleotides are a compound of Formula XI-a: XI-a.
In some embodiments, the modified nucleotides are a compound of Formula XI—b: Y1 ||=I’— 3 . Y B ORc1 HO OH XI-b.
In some embodiments, the modified nucleotides are a compound of Formula XI-cl , XI-c2, or XI-c3: 2.‘ >..‘ >..‘ Y1 I'f—Y Y3 B Y1 FI’— Y3 B Y1 E—Y Y3 B ORCI ORc1 ORG1 n Wx 2 n Q n HO A HO OH Ho A, XI-cl XI-c2 XI-c3 ] In some embodiments, the modified nucleotides are a nd of Formula XI: Y1‘<B—Y%Y3’..‘ B WU n Z _ D A wherein: \\ denotes a single or a double bond; , , —denotes an optional single bond; U is O, S, -NRa-, or -CRaRb- when ‘\~ denotes a single bond, or U is -CRa- when \ s a double bond; Z is H, C142 alkyl, or C640 aryl, or Z is absent when \\ denotes a double bond; and Z can be - and form a bond with A; A is H, OH, sulfate, -NH2, -SH, an amino acid, or a peptide comprising 1 to 12 amino acids; D is H, OH, -NH2, -SH, an amino acid, a peptide comprising 1 to 12 amino acids, or a group of Formula XII: Y1 ||3=X or A and D together with the carbon atoms to which they are attached form a 5—membered ring; X is O or S; each of Y1 is independently selected from 70R“, -NRa1Rb1, and 7SRa1; each of Y2 and Y3 are independently selected from O, -CRaRb-, NRc, S or a linker comprising one or more atoms selected from the group consisting of C, O, N, and S; n is 0, 1,2, or 3; In is 0, 1,2 or 3; B is a nucleobase of Formula XIII: ”It N R5 XIII V is N or positively charged NRC; R3 is NRCRd, -OR", or -SR“; R4 is H or can ally form a bond with Y3; R5 is H, -NRCRd, or -ORa; Ra and Rb are each independently H, C142 alkyl, C242 alkenyl, C242 alkynyl, or C640 aryl; Rc is H, C1.” alkyl, C242 alkenyl, , benzyl, a polyethylene glycol group, or an amino—polyethylene glycol group; R31 and Rb1 are each independently H or a counterion; and 70Rc1 is OH at a pH of about 1 or 70R“ is O’ at physiological pH.
In some embodiments, B is: <fiN \ N wherein R3 is 70H, -SH, or ”“2 In some embodiments, B is: N‘N/ In some embodiments, B is: /N \‘3’0 <N IN/) In some embodiments, the modified nucleotides are a compound of Formula I-d: In some embodiments, the d nucleotides are a compound selected from the group consisting of: N \ N o o o II <f ‘9 o—B—o—Ig—o—P—o N NANHZ be ('39 69 HO OH (BB- 247), / NH 0 o o I A eo—I'l3'—o—1'I>'—o—fi"—o N N ““2 06 0e 06 HO OH (BB- 248), H3C\O N \N 9 9 9 film eO—P—O—IID—O—IID—O N NH2 (36 0e 06 HO OH (BB-249), / ‘N o o o | J 6 o P o P o_ll_ _ll_ _|I_ N P o N Ce 0e 06 HO OH (BB- 250), 9 9 9 9 o—P—o—P—o—P—o ” HO OH (BB-251), N \ N o o o <’ | J @o—P—o—P—o—P—o N N 09 0e 06 HO OH (BB- 252), N \ N o o o (N | g eo—P—o—P—o—P—o N 06 09 o6w HO OH (BB- 253) N N’CH3 o o o </ l II II II g 6 N 0—P—o—P—o—P—o N 06 0e 06>w HO OH (BB- 254), 9 9 9 <”NNfLN/CHS' A eo-fi-o-fi-o-Ff-o N NH2 09 09 09V HO OH (BB-255), 0 o o 0=< | A 6 ll 11 II N O-IlD-O-l'f-O-Ffo N NH2 06 06 Dev HO OH (BB- 256), 0 o o </ | g 6 _II_ _II_ _||_ N o Ff 0 Fl’ 0 Fl’ 0 N 06 06 09V HO OH (BB- 257), and o o <’ | II II 9 6 NANHZ O—llD—O—IID-O-IID—O ON 0e 0e 06 HO OH (BB- 258), or a pharmaceutically acceptable salt thereof.
] In some embodiments, the modified nucleotides are a compound selected from the group consisting of: NW NW 9 (N | / 9 9 9 | N <N/ eO-Ff-O N o eO-Ff-O-Ff-O-Ff-O o HO OH (BB— 259), HO OH (BB- 260), 0 Mg»I N o H2N dLofi e P—o—P—O‘Q e 06) 09 06 HO OH (BB-261), H0 0H (BB- 262), e e \N® \N® \N \N o <’ l ,J o o o </ | II N II II II N A o—P—o N e N o o—P—o—P—o—P—o o 09w 06 06 06w HO OH (BB- 263), HO OH (BB- 264), 89 Se \e \e N N \N o o o o </ l E) ll <’N N) g 9 N 0—P—o N o o—F}—o—F}—o—F}—o o 09 06 Ge 09w HO OH (BB- 265), HO OH (BB-266), NH2 6 NH2 6 N \N/0 N \N’0 o </ | ® e / 0 o o </ I E) / n N N E) o—F}—o—F}—o—F}—o N N o—F}—o o o 06w 06 09 06w HO OH (BB- 267), HO OH (BB- 268), HO OH (BB- 269), H‘N/\/\O/\/O\/\O 9 9 9 <1, ' a OWO 9 O-lf-O-I‘D—O—IID-O N 09 0e 0e HO OH (BB- 270), 0 o 99 (if? 000 (If? (Hi—0 0 o—fil—o—fiL—o N o6w 09 09 09w o o o 0 \fi (BB- 271), \l/\ (BB- 272), o o o flNH NKO o o o flNH eO-IIIDI—O 9 o—fil—o—fiL—o—fil—o N’£o 0e 06 09 09 HO OH (BB- 273), and HO OH (BB- 274), or a pharmaceutically acceptable salt thereof.
Modifications on the Internucleoside Linkage The modified tides, which may be incorporated into a polynucleotide molecule, can be modified on the internucleoside linkage (e. g., phosphate backbone). Herein, in the context of the polynucleotide backbone, the phrases “phosphate” and “phosphodiester” are used interchangeably.
Backbone phosphate groups can be modified by replacing one or more of the oxygen atoms with a different substituent. Further, the modified nucleosides and nucleotides can e the wholesale ement of an unmodified phosphate moiety with another internucleoside linkage as described herein. Examples of modified phosphate groups include, but are not limited to, phosphorothioate, phosphoroselenates, boranophosphates, boranophosphate esters, hydrogen phosphonates, phosphoramidates, orodiamidates, alkyl or aryl phosphonates, and phosphotriesters.
Phosphorodithioates have both non-linking oxygens replaced by sulfur. The phosphate linker can also be modified by the replacement of a linking oxygen with en (bridged phosphoramidates), sulfur ed phosphorothioates), and carbon ed methylene-phosphonates).
The (x-thio substituted phosphate moiety is provided to confer stability to RNA and DNA polymers h the unnatural orothioate backbone linkages. Phosphorothioate DNA and RNA have increased nuclease resistance and subsequently a longer half-life in a cellular nment. While not wishing to be bound by theory, phosphorothioate linked polynucleotide molecules are ed to also reduce the innate immune response through weaker binding/activation of cellular innate immune molecules.
In specific embodiments, a modified nucleoside includes an alpha-thio-nucleoside (e.g., l—thiophosphate)-adenosine, l-thiophosphate)-cytidine (a—thio-cytidine), 5’-O-(l- thiophosphate)—guanosine, 5’-O-(l-thiophosphate)-uridine, or 5’-O-(l-thiophosphate)- pseudouridine).
Other internucleoside linkages that may be employed according to the present invention, including internucleoside linkages which do not n a phosphorous atom, are described herein below.
Combinations of Modified Sugarsa basesa and Internucleoside Linkages The polynucleotides of the invention can include a combination of ations to the sugar, the nucleobase, and/or the internucleoside linkage. These combinations can include any one or more modifications described herein. For examples, any of the nucleotides described herein in Formulas (Ia), (Ia— l )-(Ia—3), (Ib)-(If), (IIa)-(IIp), (IIb- 1 ), (IIb-2), (IIc— 1)-(IIc—2), (IIn- 1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr) can be combined with any of the nucleobases described herein (e.g., in Formulas (bl)—(b43) or any other described herein).
Synthesis of Polynucleotide Molecules ] The polynucleotide molecules for use in accordance with the invention may be prepared according to any useful technique, as described herein. The modified nucleosides and nucleotides used in the synthesis of polynucleotide molecules disclosed herein can be prepared from readily available starting materials using the following general methods and procedures. Where typical or preferred s conditions (e.g., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are provided, a skilled artisan would be able to optimize and develop additional process conditions. Optimum reaction conditions may vary with the particular reactants or solvent used, but such conditions can be determined by one skilled in the art by routine optimization procedures.
The processes described herein can be monitored according to any suitable method known in the art. For example, t formation can be monitored by spectroscopic means, such as nuclear magnetic resonance spectroscopy (e.g., 1H or 13C) infrared spectroscopy, spectrophotometry (e.g., UV-visible), or mass spectrometry, or by chromatography such as high performance liquid tography (HPLC) or thin layer chromatography.
Preparation of polynucleotide molecules of the present invention can involve the protection and deprotection of various chemical groups. The need for protection and deprotection, and the selection of riate ting groups can be readily ined by one skilled in the art.
The chemistry of protecting groups can be found, for example, in Greene, et al., Protective Groups in c Synthesis, 2d. Ed., Wiley & Sons, 1991, which is incorporated herein by nce in its entirety.
] The reactions of the processes described herein can be carried out in suitable solvents, which can be readily selected by one of skill in the art of organic synthesis. Suitable solvents can be substantially nonreactive with the starting materials (reactants), the intermediates, or products at the temperatures at which the ons are carried out, i.e., atures which can range from the solvent’s freezing ature to the solvent’s boiling temperature. A given on can be d out in one solvent or a mixture of more than one solvent. Depending on the particular reaction step, suitable solvents for a particular reaction step can be selected.
Resolution of racemic mixtures of modified polynucleotides or nucleic acids (e.g., polynucleotides or d mRNA molecules) can be carried out by any of numerous methods known in the art. An example method includes fractional recrystallization using a “chiral resolving acid” which is an optically active, salt-forming organic acid. Suitable resolving agents for fractional tallization methods are, for example, optically active acids, such as the D and L forms of tartaric acid, diacetyltartaric acid, dibenzoyltartaric acid, mandelic acid, malic acid, lactic acid or the various optically active camphorsulfonic acids. Resolution of racemic mixtures can also be carried out by elution on a column packed with an lly active resolving agent (e.g., dinitrobenzoylphenylglycine). Suitable elution solvent composition can be determined by one skilled in the art. d nucleosides and nucleotides (e.g., building block molecules) can be prepared according to the synthetic methods described in Ogata et al., J. Org. Chem 74:2585-2588 (2009); Purmal et al., Nucl. Acids Res. 22(1): 72-78, (1994); Fukuhara et al., Biochemistry, 1(4): 563-568 ; and Xu et al., Tetrahedron, 48(9): 1729-1740 , each of which are incorporated by reference in their ty.
The polynucleotides of the invention may or may not be uniformly modified along the entire length of the le. For example, one or more or all types of nucleotide (e.g., purine or pyrimidine, or any one or more or all of A, G, U, C) may or may not be uniformly modified in a polynucleotide of the invention, or in a given predetermined sequence region thereof. In some embodiments, all nucleotides X in a polynucleotide of the invention (or in a given sequence region thereof) are modified, wherein X may any one of nucleotides A, G, U, C, or any one ofthe cmnbmmkmsA+G,A+U,A+C,G+U,G+C,U+C,A+G+U,A+G+C,G+U+CorA+G+C.
Different sugar modifications, nucleotide modifications, and/or ucleoside linkages (e.g., backbone structures) may exist at various positions in the polynucleotide. One of ordinary skill in the art will appreciate that the nucleotide analogs or other ation(s) may be located at any position(s) of a polynucleotide such that the function of the polynucleotide is not substantially decreased. A modification may also be a 5’ or 3’ al modification. The polynucleotide may contain from about 1% to about 100% modified nucleotides (either in on to overall nucleotide content, or in relation to one or more types of nucleotide, i.e. any one or more of A, G, U or C) or any intervening percentage (e. g., from 1% to 20%, from 1% to 25%, from 1% to 50%, from 1% to 60%, from 1% to 70%, from 1% to 80%, from 1% to 90%, from 1% to 95%, from 10% to 20%, from % to 25%, from 10% to 50%, from 10% to 60%, from 10% to 70%, from 10% to 80%, from 10% to 90%, from 10% to 95%, from 10% to 100%, from 20% to 25%, from 20% to 50%, from 20% to 60%, from 20% to 70%, from 20% to 80%, from 20% to 90%, from 20% to 95%, from 20% to 100%, from 50% to 60%, from 50% to 70%, from 50% to 80%, from 50% to 90%, from 50% to 95%, from 50% to 100%, from 70% to 80%, from 70% to 90%, from 70% to 95%, from 70% to 100%, from 80% to 90%, from 80% to 95%, from 80% to 100%, from 90% to 95%, from 90% to 100%, and from 95% to 100%).
In some embodiments, the cleotide includes a modified pyrimidine (e.g., a modified uracil/uridinex’U or modified cytosine/cytidine/C). In some embodiments, the uracil or uridine ally: U) in the polynucleotide molecule may be replaced with from about 1% to about 100% of a modified uracil or modified uridine (e.g., from 1% to 20%, from 1% to 25%, from 1% to 50%, from 1% to 60%, from 1% to 70%, from 1% to 80%, from 1% to 90%, from 1% to 95%, from 10% to 20%, from 10% to 25%, from 10% to 50%, from 10% to 60%, from 10% to 70%, from 10% to 80%, from 10% to 90%, from 10% to 95%, from 10% to 100%, from 20% to 25%, from 20% to 50%, from 20% to 60%, from 20% to 70%, from 20% to 80%, from 20% to 90%, from 20% to 95%, from 20% to 100%, from 50% to 60%, from 50% to 70%, from 50% to 80%, from 50% to 90%, from 50% to 95%, from 50% to 100%, from 70% to 80%, from 70% to 90%, from 70% to 95%, from 70% to 100%, from 80% to 90%, from 80% to 95%, from 80% to 100%, from 90% to 95%, from 90% to 100%, and from 95% to 100% of a modified uracil or modified uridine). The modified uracil or uridine can be replaced by a compound having a single unique structure or by a plurality of compounds having different structures (e.g., 2, 3, 4 or more unique structures, as described herein).
In some embodiments, the cytosine or cytidine (generally: C) in the polynucleotide molecule may be replaced with from about 1% to about 100% of a modified cytosine or modified cytidine (e.g., from 1% to 20%, from 1% to 25%, from 1% to 50%, from 1% to 60%, from 1% to 70%, from 1% to 80%, from 1% to 90%, from 1% to 95%, from 10% to 20%, from 10% to 25%, from 10% to 50%, from % to 60%, from 10% to 70%, from 10% to 80%, from 10% to 90%, from 10% to 95%, from 10% to 100%, from 20% to 25%, from 20% to 50%, from 20% to 60%, from 20% to 70%, from 20% to 80%, from 20% to 90%, from 20% to 95%, from 20% to 100%, from 50% to 60%, from 50% to 70%, from 50% to 80%, from 50% to 90%, from 50% to 95%, from 50% to 100%, from 70% to 80%, from 70% to 90%, from 70% to 95%, from 70% to 100%, from 80% to 90%, from 80% to 95%, from 80% to 100%, from 90% to 95%, from 90% to 100%, and from 95% to 100% ofa modified cytosine or d cytidine). The d cytosine or ne can be replaced by a compound having a single unique structure or by a plurality of compounds having different structures (e.g., 2, 3, 4 or more unique structures, as described herein).
In some embodiments, the t disclosure provides methods of synthesizing a polynucleotide (e. g., the first region, first flanking region, or second flanking region) ing n number of linked nucleosides having Formula (Ia-1): (Ia-1), comprising: a) reacting a nucleotide of Formula (IV-1): Y17Y5 B RN-”R5 4 d m' (IV-1), with a phosphoramidite nd of Formula (V-1): P1—Y1—Y5 B Y YN (v-1), wherein Y9 is H, hydroxy, phosphoryl, pyrophosphate, sulfate, amino, thiol, optionally substituted amino acid, or a peptide (e.g., including from 2 to 12 amino ; and each P1, P2, and P3 is, independently, a suitable protecting group; and 0 denotes a solid t; to provide a polynucleotide of Formula (VI-1): P1—Y1—Y5 B 0 P2), m (VI-1), and b) oxidizing or sulfurizing the polynucleotide of Formula (V) to yield a polynucleotide of Formula (VII-l): 0 m' (VII-1), and ] c) removing the protecting groups to yield the polynucleotide of Formula (Ia).
In some embodiments, steps a) and b) are repeated from 1 to about 10,000 times. In some embodiments, the methods further comprise a nucleotide selected from the group consisting of A, C, G and U adenosine, cytosine, guanosine, and uracil. In some embodiments, the nucleobase may be a pyrimidine or derivative thereof. In some embodiments, the polynucleotide is translatable.
Other components of polynucleotides are optional, and are beneficial in some embodiments. For example, a 5’ untranslated region (UTR) and/or a 3’UTR are provided, wherein either or both may independently contain one or more different nucleotide modifications. In such ments, nucleotide cations may also be present in the atable region. Also provided are polynucleotides containing a Kozak sequence.
Combinations of Nucleotides ] Further examples of modified nucleotides and modified nucleotide combinations are provided below in Table 2. These combinations of modified nucleotides can be used to form the polynucleotides of the invention. Unless otherwise noted, the modified nucleotides may be completely tuted for the natural nucleotides of the polynucleotides of the invention. As a nonlimiting e, the natural nucleotide uridine may be tuted with a modified side described herein. In another non-limiting e, the natural nucleotide uridine may be partially substituted (e.g., about 0.1%, 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 99.9%) with at least one of the modified nucleoside disclosed herein.
Table 2 Modified Nucleotide Modified Nucleotide Combination -cytidine a—thio-cytidine/S-iodo-uridine a—thio-cytidine/N 1-methy1-pseudo-uridine a—thio-cytidine/a-thio-uridine a—thio-cytidine/S-methyl-uridine -cytidine/pseudo-uridine about 50% of the cytosines are a—thio-cytidine pseudoisooytidine pseudoisocytidine/5 -iodo-uridine pseudoisocytidine/N1-methyl-pseudouridine pseudoisocytidine/a—thio-uridine pseudoisocytidine/S -methy1-uridine pseudoisocytidine/pseudoutidine about 25% of cytosines are pseudoisocytidine pseudoisocytidine/about 50% of uridines are N1 -methy1-pseudouridine and about 50% ofuridines are pseudouridine pseudoisocytidine/about 25% of uridines are N1 -methy1-pseudouridine and about 25% ofuridines are pseudouridine (e.g., 25% N1-methy1-pseudoun'dine/75% pseudouridine) pyIrolo-cytidine lyrrolo-cytidine/S-iodo-uridine o-cytidine/N 1 -methy1-pseudouridine pyrrolo-cytidine/(x-thio-un'dine pyrrolo-cytidine/S-methy1-uridine pyrrolo-cytidine/pseudouridine about 50% of the cytosines are pyrrolo-cytidine -methyl—cytidine 5-methyl-cytidine/5-iodo-uridine -methyl-cytidine/N 1 -methy1-pseudoun'dine -methyl-cytidine/0L-thio-un'dine -methy1-cytidine/5-methy1-uridine -methyl-cytidine/pseudoun'dine about 25% of cytosines are 5-methy1-cytidine about 50% of cytosines are 5-methy1-cytidine -methyl-cytidine/5-methoxy-uridine -methy1-cytidine/5-bromo-uridine -methyl-cytidine/2-thio-uridine S-methyl-cytidine/about 50% of es are 2-tl1io-midine about 50% of uridines are yl-cytidine/ about 50% of un'dines are 2- thio-uridine N4-acetyl-cytidine N4-acety1—cytidine /5-iodo-uridine ty1-cytidine thy1-pseudouridine N4-acety1-cytidine /0L-thio-un'dine N4-acety1-cytidine /5-methy1-uridine N4-acety1-cytidine /pseudouridine about 50% of cytosines are ty1-cytidine about 25% of cytosines are N4-acety1-cytidine N4-acety1-cytidine /5-methoxy-uridine N4-acetyl-cytidine /5-bromo-uridine N4-acetyl-cytidine /2-thio-uridine about 50% of cytosines are N4-acetyl-cytidine/ about 50% ines are 2-thio-uridine Certain modified nucleotides and nucleotide combinations have been ed by the current inventors. These findings are described in US. Provisional Application No 61/404,413, filed on October 1, 2010, entitled Engineered Nucleic Acids and Methods of Use Thereof, US. Patent Application No 13/251,840, filed on October 3, 2011, entitled Modified Nucleotides, and Nucleic Acids, and Uses Thereof, now abandoned, US. Patent Application No 13/481,127, filed on May 25, 2012, entitled Modified tides, and Nucleic Acids, and Uses Thereof, International Patent Publication No W02012045075, filed on October 3, 201 1, entitled Modified Nucleosides, Nucleotides, And c Acids, and Uses Thereof, US. Patent Publication No US20120237975 filed on October 3, 201 1, entitled Engineered Nucleic Acids and Method of Use Thereof, and International Patent Publication No W02012045082, which are incorporated by reference in their entireties.
Further examples of modified nucleotide combinations are provided below in Table 3.
These combinations of modified nucleotides can be used to form the polynucleotides ofthe invention.
Table 3 Modified Nucleotide Modified Nucleotide Combination d cytidine having one or more modified cytidine with (b10)/pseudouridine nucleobases 0f Formula (1310) modified cytidine with (b10)/N1-methy1-pseudouridine modified cytidine with (b10)/5-methoxy-uridine modified cytidine with (b10)/5-methy1-utidine modified cytidine with (b10)/5-bromo-uridine modified cytidine with (b10)/2-thio-uridine about 50% of cytidine substituted with d ne (b10)/ about 50% of uridines are 2-thio-uridine modified cytidine having one or more modified cytidine with pseudouridine PaSES 0f Formula 0332) modified cytidine with (b32)/N1-methyl-pseudouridine modified cytidine with 5-methoxy-uridine d cytidine with (b32)/5-methyl-uridine modified cytidine with (b32)/5-bromo-uridine d cytidine with (b32)/2-thio-uridine about 50% of cytidine substituted with modified cytidine (b32)/ about 50% of uridines are 2-thio-uridine modified e having one or more modified uridine with (b1)/ N4-acetyl-cytidine bases of Formula (b1) modified uridine with (b1)/ y1-cytidine modified uridine having one or more modified uridine with (b8)/ N4-acety1-cytidine nucleobases of a 038) modified uridine with (b8)/ 5-methy1-cytidine modified uridine having one or more modified e with (b28)/ N4-acety1—cytidine nuoleobases of Formula (b28) modified uridine with (b28)/ 5-methy1—cytidine d uridine having one or more modified uridine with (b29)/ N4-acety1—cytidine nucleobases of Formula 0’29) modified e with (b29)/ 5-methy1—cytidine modified uridine having one or more modified uridine with (b3 0)/ N4-acety1—cytidine nucleobases of Formula (b3 0) modified uridine with (b3 0)/ 5-methy1—cytidine In some embodiments, at least 25% of the cytosines are replaced by a compound of Formula (b10)-(b14), (b24), (b25), or (b32)-(b35) (e. g., at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or about 100% of, e. g., a compound of Formula (MO) or (b32))- In some ments, at least 25% of the uracils are replaced by a compound ofFormula (bl)-(b9), (b21)—(b23), or (b28)-(b31) (e. g., at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or about 100% of, e. g., a compound of Formula (b1), (b8), (b28), (b29), or (b30))- In some embodiments, at least 25% of the cytosines are replaced by a compound of Formula (b10)-(bl4), (b24), (b25), or (b32)-(b35) (e.g. Formula (b10) or (b32)), and at least 25% of the uracils are replaced by a compound of a (b1)-(b9), (b21)—(b23), or (b28)-(b3l) (e.g.
Formula (bl), (b8), (b28), (b29), or (b30)) (e.g., at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or about 100%).
Modifications including Linker and a Payload The nucleobase of the nucleotide can be covalently linked at any chemically appropriate position to a payload, e,g,, detectable agent or therapeutic agent. For example, the nucleobase can be deaza-adenosine or deaza-guanosine and the linker can be attached at the C-7 or C-8 positions of the deaza-adenosine or deaza-guanosine. In other embodiments, the base can be cytosine or uracil and the linker can be attached to the N-3 or C-5 positions of cytosine or uracil. Scheme 1 below depicts an exemplary modified nucleotide wherein the nucleobase, adenine, is attached to a linker at the C-7 carbon of 7-deaza adenine. In addition, Scheme 1 s the ed nucleotide with the linker and payload, e.g., a detectable agent, incorporated onto the 3’ end of the mRNA.
Disulfide cleavage and 1,2-addition of the thiol group onto the propargyl ester releases the able agent. The remaining structure (depicted, for e, as pApCSParg in Scheme 1) is the inhibitor.
The ale for the structure of the modified nucleotides is that the tethered inhibitor sterically interferes with the ability of the polymerase to incorporate a second base. Thus, it is al that the tether be long enough to affect this function and that the inhibiter be in a stereochemical orientation that inhibits or prohibits second and follow on nucleotides into the growing polynucleotide strand.
Scheme 1 HN o ““2 / oW5,SWNW N, o | \ 0 NH; \N N ACapless pCpCS Farg \N O O o o,?,o,§,o,§,o N o 0' 0' 0' rod, 0 OH OH .dp‘o 703F1430‘ oration NH2 HN w O l 2 OH OH N/KO Cleavage of 8-8 bond '0\ P00 O NHZ m .01 \0 N/ \ : 0W SH -o’\\F’\’o RNA~w§ l\\ I WI 0- N N o OH OH : OH RNA”Ewl KNN/l 0 04% S + 7 OH OH ] The term “linker” as used herein refers to a group of atoms, e.g., lO-l,000 atoms, and can be comprised of the atoms or groups such as, but not limited to, carbon, amino, alkylamino, oxygen, sulfur, sulfoxide, sulfonyl, carbonyl, and imine. The linker can be attached to a modified nucleoside or nucleotide on the nucleobase or sugar moiety at a first end, and to a payload, e.g., able or therapeutic agent, at a second end. The linker is of sufficient length as to not interfere with incorporation into a nucleic acid sequence.
Examples of chemical groups that can be incorporated into the linker include, but are not limited to, an alkyl, alkene, an alkyne, an amido, an ether, a thioether, an or an ester group. The linker chain can also se part of a saturated, unsaturated or aromatic ring, including polycyclic and heteroaromatic rings wherein the aromatic ring is an aryl group containing from one to four heteroatoms, N, O or S. Specific examples of linkers include, but are not limited to, unsaturated alkanes, polyethylene glycols, and dextran polymers.
For example, the linker can include ethylene or propylene glycol monomeric units, e.g., diethylene glycol, dipropylene glycol, triethylene glycol, tripropylene , tetraethylene glycol, or tetraethylene glycol. In some embodiments, the linker can include a divalent alkyl, l, and/or alkynyl moiety. The linker can include an ester, amide, or ether .
Other es include cleavable moieties within the linker, such as, for example, a disulfide bond (-S-S-) or an azo bond (-N:N-), which can be cleaved using a reducing agent or photolysis. A cleavable bond incorporated into the linker and attached to a d nucleotide, when cleaved, results in, for example, a short “scar” or chemical modification on the nucleotide. For example, after cleaving, the resulting scar on a nucleotide base, which formed part of the modified nucleotide, and is incorporated into a cleotide strand, is unreactive and does not need to be chemically lized. This increases the ease with which a uent nucleotide can be incorporated during cing of a nucleic acid polymer template. For example, conditions include the use of tris(2-carboxyethyl)phosphine (TCEP), dithiothreitol (DTT) and/or other ng agents for cleavage of a disulfide bond. A selectively severable bond that includes an amido bond can be cleaved for example by the use of TCEP or other reducing agents, and/or ysis. A selectively severable bond that includes an ester bond can be cleaved for example by acidic or basic hydrolysis.
Payload The methods and compositions described herein are useful for delivering a payload to a biological target. The payload can be used, e.g., for labeling (e.g., a detectable agent such as a fluorophore), or for therapeutic purposes (e.g., a cytotoxin or other therapeutic agent).
Payload: Therapeutic Agents In some embodiments the payload is a therapeutic agent such as a cytotoxin, radioactive ion, chemotherapeutic, or other therapeutic agent. A cytotoxin or cytotoxic agent includes any agent that is detrimental to cells. Examples include taxol, cytochalasin B, gramicidin D, um bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, bicin, daunorubicin, dihydroxy cin dione, ntrone, mithramycin, actinomycin D, I- dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, puromycin, maytansinoids, e.g., maytansinol (see US. Pat. No. 020), CC-1065 (see US. Pat. Nos. ,475,092, 5,585,499, 5,846,545) and analogs or homologs thereof. Radioactive ions include, but are not limited to iodine (e.g., iodine 125 or iodine 131), strontium 89, phosphorous, palladium, cesium, iridium, phosphate, , yttrium 90, Samarium 153 and praseodymium. Other therapeutic agents include, but are not limited to, antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6- thioguanine, cytarabine, 5-fluorouracil azine), alkylating agents (e.g., mechlorethamine, thioepa chlorambucil, CC-1065, melphalan, stine (BSNU) and lomustine (CCNU), cyclothosphamide, busulfan, dibromomannitol, streptozotocin, cin C, and cis- dichlorodiamine platinum (II) (DDP) cisplatin), cyclines (e.g., daunorubicin (formerly daunomycin) and doxorubicin), antibiotics (e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, and anthramycin (AMC)), and itotic agents (e.g., vincristine, vinblastine, taxol and maytansinoids).
Payload'Detectable Agents Examples of detectable substances include various organic small molecules, inorganic compounds, nanoparticles, enzymes or enzyme substrates, cent als, luminescent materials, bioluminescent materials, chemiluminescent materials, radioactive materials, and contrast agents. Such optically-detectable labels include for example, without limitation, 4-acetamido-4'- isothiocyanatostilbene—2,2'disulfonic acid; acridine and derivatives: acridine, ne isothiocyanate; 5-(2’-aminoethyl)aminonaphthalene-1 -sulfonic acid (EDANS); o-N-[3- ulfonyl)phenyl]naphthalimide-3,5 disulfonate; N-(4-anilino-l-naphthyl)maleimide; anthranilamide; BODIPY; Brilliant Yellow; coumarin and derivatives; coumarin, 7-amino-4— methylcoumarin (AMC, Coumarin 120), 7-aminotrifluoromethylcouluarin (Coumaran 15]); cyanine dyes; cyanosine; 4’,6-diaminidino-Z-phenylindole (DAPI); 5’ 5"-dibromopyrogallol- sulfonaphthalein (Bromopyrogallol Red); 7-diethylamino(4’-isothiocyanatophenyl)—4- methylcoumarin; diethylenetriamine pentaacetate; 4,4 ’-diisothiocyanatodihydro-stilbene-2,2 '- disulfonic acid; 4,4'-diisothiocyanatostilbene-2,2’-disulfonic acid; 5-[dimethylamino]-naphthalene-l- sulfonyl chloride (DNS, dansylchloride); 4-dimethylaminophenylazophenyl—4’-isothiocyanate (DABITC); eosin and derivatives; eosin, eosin isothiocyanate, erythrosin and derivatives; erythrosin B, erythrosin, isothiocyanate; ethidium; fluorescein and derivatives; 5-carb0xyflu0rescein (FAM), -(4,6-dichlorotriazinyl)amin0fluorescein (DTAF), 2 ',7 '-dimeth0xy-4'5'-dichlor0 carboxyfluorescein, fluorescein, fluorescein isothiocyanate, QFITC, (XRITC); camine; IRl44; ; Malachite Green isothiocyanate; 4-methylumbelliferone0rth0 cresolphthalein; yrosine; pararosaniline; Phenol Red; B-phycoerythrin; o-phthaldialdehyde; pyrene and derivatives: pyrene, pyrene butyrate, succinimidyl 1-pyrene; te quantum dots; Reactive Red 4 ronTM Brilliant Red 3B-A) rhodamine and derivatives: 6-carboxy-X-rh0damine (ROX), 6— carboxyrhodamine (R6G), lissamine rhodamine B sulfonyl chloride rhodamine (Rhod), rhodamine B, rhodamine 123, rhodamine X isothiocyanate, sulforhodamine B, sulforhodamine 101, sulfonyl chloride derivative of sulforhodamine 101 (Texas Red); N,N,N',N'tetramethyl-6— yrhodamine (TAMRA); tetramethyl rhodamine; tetramethyl rhodamine ocyanate (TRITC); riboflavin; rosolic acid; terbium chelate derivatives; Cyanine-3 (Cy3); Cyanine—S (CyS); Cyanine-5.5 (Cy5.5), Cyanine-7 (Cy7); IRD 700; IRD 800; Alexa 647; La Jolta Blue; phthalo cyanine; and naphthalo cyanine. In some embodiments, the able label is a fluorescent dye, such as Cy5 and Cy3.
Examples luminescent material includes luminol; examples of inescent materials include luciferase, luciferin, and aequorin.
] Examples of suitable radioactive material e 18F, 67Ga, 81mKr, 82Rb, 111k], 123I, mXe, 201TI, ”51, 35S, 14C, or 3H, 99'“Tc (e. g., as pertechnetate etate(VII), TcO4') either directly or indirectly, or other radioisotope able by direct counting of radioemission or by scintillation counting.
In addition, contrast agents, e.g., contrast agents for MRI or NMR, for X-ray CT, Raman imaging, optical nce tomography, absorption imaging, ultrasound imaging, or thermal imaging can be used. ary contrast agents include gold (e.g., gold nanoparticles), gadolinium (e.g., chelated Gd), iron oxides (e,g,, superparamagnetic iron oxide (SPIO), monocrystalline iron oxide nanoparticles ), and ultrasmall superparamagnetic iron oxide (USPIO)), manganese chelates (e.g., Mn-DPDP), barium sulfate, iodirrated contrast media (iohexol), microbubbles, or perfluorocarbons can also be used.
In some embodiments, the detectable agent is a non-detectable pre—cursor that becomes detectable upon activation. Examples include fluorogenic tetrazine—fluorophore constructs (e.g., tetrazine-BODIPY FL, tetrazine-Oregon Green 488, or tetrazine—BODIPY TMR-X) or enzyme activatable fluorogenic agents (e.g., SE (VisEn Medical».
When the compounds are enzymatically d with, for example, horseradish peroxidase, ne phosphatase, or luciferase, the enzymatic label is detected by ination of conversion of an riate substrate to product.
] In vitro assays in which these compositions can be used include enzyme linked irnrnunosorbent assays (ELISAs), irnmunoprecipitations, immunofluorescence, enzyme assay (EIA), radioirnmunoassay (RIA), and Western blot analysis.
Labels other than those described herein are contemplated by the present disclosure, including other optically-detectable labels. Labels can be attached to the modified nucleotide of the t disclosure at any position using standard tries such that the label can be removed from the incorporated base upon cleavage of the cleavable linker.
Payload: Cell Penetrating ds In some embodiments, the modified nucleotides and modified nucleic acids can also include a payload that can be a cell penetrating moiety or agent that enhances intracellular delivery ofthe compositions. For example, the compositions can include a cell-penetrating peptide sequence that facilitates delivery to the ellular space, e.g., HIV-derived TAT peptide, penetratins, transportans, or hCT derived cell-penetrating peptides, see, e.g., Caron et al., (2001) Mol Ther. 3(3):310-8; Langel, Cell-Penetrating Peptides: Processes and Applications (CRC Press, Boca Raton FL 2002); El-Andaloussi et al., (2005) Curr Pharrn Des. l l(28):3597-6l l; and Deshayes et al., (2005) Cell Mol Life Sci. 62(16):]839-49. The compositions can also be formulated to include a cell penetrating agent, e.g., liposomes, which e ry of the compositions to the intracellular space.
Payloadfliological Targets The modified nucleotides and modified nucleic acids described herein can be used to deliver a payload to any biological target for which a specific ligand exists or can be generated. The ligand can bind to the biological target either covalently or non-covalently.
Exemplary biological targets include biopolymers, e.g., antibodies, nucleic acids such as RNA and DNA, proteins, enzymes; exemplary proteins include enzymes, receptors, and ion channels. In some embodiments the target is a tissue— or cell-type specific marker, e.g., a protein that is expressed specifically on a ed tissue or cell type. In some embodiments, the target is a receptor, such as, but not d to, plasma membrane receptors and nuclear ors; more specific es include G—protein-coupled receptors, cell pore proteins, orter proteins, surface-expressed antibodies, HLA proteins, MHC proteins and growth factor receptors.
Synthesis of Modified Nucleotides The modified nucleosides and nucleotides disclosed herein can be prepared from readily available starting materials using the following l methods and procedures. It is tood that where typical or red process conditions (i.e., reaction atures, times, mole ratios of reactants, solvents, pressures, etc.) are given; other process conditions can also be used unless otherwise stated. Optimum reaction conditions may vary with the particular reactants or solvent used, but such ions can be determined by one skilled in the art by e optimization procedures.
The processes described herein can be monitored according to any suitable method known in the art. For example, product formation can be monitored by spectroscopic means, such as nuclear magnetic resonance spectroscopy (e.g., 1H or 13C) infrared spectroscopy, spectrophotometry (e.g., UV-visible), or mass spectrometry, or by chromatography such as high mance liquid chromatography (HPLC) or thin layer chromatography.
Preparation of modified nucleosides and nucleotides can involve the protection and deprotection of various chemical groups. The need for protection and deprotection, and the selection of appropriate protecting groups can be readily determined by one skilled in the art. The chemistry of protecting groups can be found, for e, in Greene, et a1., Protective Groups in Organic Synthesis, 2d. Ed, Wiley & Sons, 1991, which is orated herein by reference in its ty.
The ons of the ses described herein can be carried out in suitable solvents, which can be readily selected by one of skill in the art of organic synthesis. Suitable solvents can be substantially nonreactive with the starting materials (reactants), the intermediates, or products at the temperatures at which the reactions are carried out, i.e., atures which can range from the solvent’s freezing temperature to the solvent’s boiling temperature. A given reaction can be carried out in one solvent or a mixture of more than one t. Depending on the particular reaction step, le solvents for a particular reaction step can be selected.
Resolution of racemic es of modified nucleosides and nucleotides can be carried out by any of numerous methods known in the art. An example method includes fractional recrystallization using a l resolving acid” which is an optically active, salt-forming organic acid. Suitable resolving agents for fractional tallization methods are, for example, optically active acids, such as the D and L forms of tartaric acid, diacetyltartaric acid, dibenzoyltartaric acid, mandelic acid, malic acid, lactic acid or the various optically active camphorsulfonic acids.
Resolution of racemic mixtures can also be carried out by elution on a column packed with an optically active resolving agent (e.g., dinitrobenzoylphenylglycine). Suitable elution solvent composition can be determined by one skilled in the art.
Exemplary syntheses of modified tides, which are incorporated into a polynucleotides, e. g., RNA or mRNA, are provided below in Scheme 2 through Scheme 12. Scheme 2 provides a general method for phosphorylation of nucleo sides, including modified nucleosides.
Scheme 2 /N‘N/ N‘N me \~ 4 \ N) 4 \ N) N 1)POC13 fi fi g N H0 —> 0 90—7—0—7—0—7—0 2) Pymphosphate O 0 O 0 e 06) OH OH OH OH Various protecting groups may be used to l the reaction. For example, Scheme 3 provides the use of multiple protecting and deprotecting steps to promote phosphorylation at the 5’ on of the sugar, rather than the 2’ and 3’ yl groups.
Scheme 3 / \ 2 Acetone/H+ H0 H0 N OH OH 0T0 Argo / \ /) DowexH N OH CH "Wu,” Ph3CCl \N 1) OH- 39003 \N) %\\A0Pflo 0 3) Pyrophosphate 4) H+ 8/\ 88O/P\O/\\Pg\ OH OH Pth/O RCPh3 d nucleotides can be synthesized in any useful manner. Schemes 4, 5, and 8 e exemplary methods for synthesizing modified nucleotides having a modified purine nucleobase; and Schemes 6 and 7 provide exemplary methods for synthesizing modified nucleotides having a modified pseudouridine or pseudoisocytidine, respectively.
Scheme 4 N CH N N/ 3 </ NH I </ ‘ / )\ N NAM N N NH2 2 Ho HO CHgl/heat , O OH OH OH OH 1)POC13 2) Pymphosphate o o o </ A II II II N N NH2 0 o—IT—o—T—o—Fl’—0 Ce 06 06 0 OH OH Schemes </” \N | A 1? a? 1? N e o T o N NH2 o o 1)P0C]3 T T —> 09 09 Ce 0 2)Pymphosphate 0H OH OH OH Scheme 6 RBr/Heat R = alkyl, l, HO H0 ally], and benzyl OH OH OH OH 1)P0C13 2) Pyrophosphafe Scheme 7 RBr/Heat R = alkyl, alkenyl, HO H0 ally], and benzyl OH OH OH 2) Pyrophosphate Scheme 8 NHCH3 N \ </ N I N \N N A N l NH2 </ / H0 N CH3NH2/Heat N NH2 OH OH OH OH 1) Pocl3 2) Pymphosphate NHCH3 N \ </ N o o o N A || || II N NHz 6 O—P—O—P—O—PJO | | | o o o e e 09 OH OH Schemes 9 and 10 provide exemplary ses of modified nucleotides. Scheme 11 provides a non-limiting biocatalytic method for producing nucleotides.
Scheme 9 AcOOH 3Pd(0) AcO OAc A020 Enzymatic Hydrolysis Hogm‘ HO OAc 0 . (1)0504 (2) Acetone, TsOH 0 O 0 NH ’o,, 0w NH ‘6 (1)(Et0)2POCI-120Ts OvNfi . 00 )r (2)TMSI| )r (1) DCC, Morpholine (2) Pyrophosphate Scheme 10 0 H Y ””2 Pth(Pd) 0 N / / _,Hoeg_~( \ I \ CH2COCH3 HO OH HO OH COCHg 1)H' 2) 'OH, heat 0 H H N \ 0 U) 1) POC|3 j: HO j <— 0 HO OH N 2) Pyrophosphate 0 OH O:R'_OH H07)” 0 ‘OH Scheme 11 HO\ B O —>‘_O\:/OWOenzyme ATP yeast enzymes 2:0 (IF; OH OH OH OH '32074 OH OH Scheme 12 es an exemplary synthesis of a modified uracil, where the N] position on the major groove face is d with Rm, as provided elsewhere, and the 5’-position of ribose is phosphorylated. T1, T2, R123, Rm, and r are as provided herein. This synthesis, as well as optimized versions thereof, can be used to modify the major groove face of other pyrimidine nucleobases and purine nucleobases (see e. g., Formulas (b1)-(b43)) and/or to install one or more phosphate groups (e. g., at the 5’ position of the . This alkylating reaction can also be used to include one or more optionally substituted alkyl group at any reactive group (e. g., amino group) in any nucleobase described herein (e.g., the amino groups in the Watson-Crick base-pairing face for cytosine, uracil, e, and guanine).
Scheme 12 T1 T1 T1 12a 12b\ R12a 12b 12a R R HNAN/R NJKN/ \NJKN/R \ T2 \ 2 ea T POC131) 9 HO 0‘15“” HO 2)Pyrophospate HO P—O 0 o OH OH OH OH OH OH Modified nucleosides and nucleotides can also be prepared according to the synthetic methods described in Ogata et al. Journal nic Chemistry 74:2585-2588, 2009; Purmal et al.
Nucleic Acids Research 22(1): 72-78, 1994; ra et a1. mistry 1(4): 563-568, 1962; and Xu et al. Tetrahedron 48(9): 1729-1740, 1992, each of which are incorporated by reference in their entirety.
Modified Nucleic Acids The present disclosure es nucleic acids (or polynucleotides), including RNAs such as mRNAs that contain one or more modified nucleosides (termed “modified nucleic acids”) or tides as described herein, which have useful properties including the lack of a substantial induction of the innate immune response of a cell into which the mRNA is introduced. Because these modified c acids enhance the efficiency of protein production, intracellular retention of nucleic acids, and ity of contacted cells, as well as possess reduced immunogenicity, these nucleic acids having these properties are also termed “enhanced nucleic acids” .
In addition, the present disclosure provides nucleic acids, which have decreased binding affinity to a major groove interacting, e.g. binding, partner. For example, the c acids are comprised of at least one nucleotide that has been chemically modified on the major groove face as described herein.
The term “nucleic acid,” in its broadest sense, includes any compound and/or substance that is or can be incorporated into an oligonucleotide chain. In this context, the term nucleic acid is used mously with polynucleotide. Exemplary nucleic acids for use in accordance with the present disclosure include, but are not limited to, one or more ofDNA, RNA ing messenger mRNA (mRNA), hybrids thereof, RNAi-inducing agents, RNAi agents, siRNAs, shRNAs, miRNAs, antisense RNAs, ribozymes, catalytic DNA, RNAs that induce triple helix formation, aptamers, s, eta, described in detail herein.
Provided are modified nucleic acids containing a translatable region and one, two, or more than two different nucleoside modifications. In some embodiments, the modified nucleic acid exhibits d degradation in a cell into which the nucleic acid is introduced, relative to a corresponding unmodified nucleic acid. Exemplary nucleic acids include ribonucleic acids , deoxyribonucleic acids (DNAs), threose nucleic acids (TNAs), glycol nucleic acids (GNAs), or a hybrid thereof. In preferred embodiments, the modified nucleic acid includes messenger RNAs (mRNAs). As described herein, the c acids of the present disclosure do not substantially induce an innate immune se of a cell into which the mRNA is introduced.
In certain embodiments, it is desirable to intracellularly degrade a modified nucleic acid introduced into the cell, for example if precise timing of n production is desired. Thus, the present disclosure provides a modified nucleic acid containing a degradation domain, which is capable of being acted on in a directed manner within a cell.
Other components of nucleic acid are optional, and are beneficial in some embodiments.
For example, a 5’ slated region (UTR) and/or a 3’UTR are provided, n either or both may independently contain one or more different side modifications. In such ments, nucleoside modifications may also be present in the translatable region. Also provided are nucleic acids containing a Kozak sequence.
Additionally, provided are nucleic acids containing one or more ic nucleotide sequences capable of being excised from the nucleic acid.
Further, provided are nucleic acids containing an internal ribosome entry site (IRES). An IRES may act as the sole ribosome binding site, or may serve as one of multiple ribosome g sites of an mRNA. An mRNA ning more than one functional ribosome binding site may encode several peptides or polypeptides that are translated independently by the ribosomes ("multicistronic mRN "). When nucleic acids are provided with an IRES, further optionally provided is a second translatable region. es of IRES sequences that can be used according to the present disclosure include without limitation, those from picornaviruses (e.g. FMDV), pest viruses (CFFV), polio viruses (PV), encephalomyocarditis viruses , nd—mouth disease viruses (FMDV), hepatitis C viruses (HCV), classical swine fever viruses (CSFV), murine leukemia virus (MlV), simian immune deficiency viruses (SIV) or cricket paralysis viruses (CrPV).
In another aspect, the present sure provides for nucleic acid sequences comprising at least two tides, the nucleic acid sequence comprising a nucleotide that disrupts binding of a major groove binding partner with the nucleic acid sequence, n the nucleotide has decreased binding affinity to the major groove binding partner.
In some embodiments, the nucleic acid is a compound of Formula XI-a: _§_Y1_F|,:X 0Rc1 XI—a wherein: \‘\\ denotes an optional double bond; — — —denotes an optional single bond; U is O, S, -NRa-, or -CRaRb- when ‘\~ denotes a single bond, or U is -CRa- when ‘\ denotes a double bond; ] A is H, OH, phosphoryl, pyrophosphate, sulfate, -NH2, -SH, an amino acid, a peptide comprising 2 to 12 amino acids; X is O or S; each of Y1 is independently selected from 70R“, -NRa1Rb1, and iSRal; each of Y2 and Y3 are independently selected from O, -CRaRb-, NRC, S or a linker comprising one or more atoms selected from the group consisting of C, O, N, and S; R8 and Rb are each independently H, C142 alkyl, C242 alkenyl, C242 alkynyl, or C640 aryl; Rc is H, C142 alkyl, C242 alkenyl, phenyl, benzyl, a polyethylene glycol group, or an amino-polyethylene glycol group; R81 and Rb1 are each independently H or a counterion; ] —ORCl is OH at a pH of about 1 or —ORCl is O' at physiological pH; and B is base; provided that the ring encompassing the les A, B, D, U, Z, Y2 and Y3 cannot be ribose.
In some embodiments, B is a nucleobase of Formula XII-a, XII-b, or XII-c: R2 o RLYJ§N31 4 R\N R2 U‘W/kx RAD Rage XII-a XII-b XII-c wherein: \ denotes a single or double bond; ] X is O or S; U and W are each independently C or N; V is O, S, C or N; wherein when V is C then R1 is H, C1_6 alkyl, C1_6 alkenyl, C1_6 alkynyl, halo, or 70R: wherein C1_20 alkyl, C240 alkenyl, C240 alkynyl are each ally tuted with QH, -NRaRb, — SH, -C(O)R°, -C(O)ORC, -NHC(O)RC, or )OR°; and wherein when V is O, S, or N then R1 is absent; R2 is H, -OR°, -SRC, -NRaRb, or halo; or when V is C then R1 and R2 together with the carbon atoms to which they are attached can form a 5- or 6-membered ring optionally substituted with 1-4 substituents selected from halo, - OH, -SH, - aRb, C1_20 alkyl, C240 alkenyl, C240 alkynyl, C1_20 alkoxy, or C140 kyl; R3 is H or 01.20 alkyl; R4 is H or C140 alkyl; wherein when ‘\~ denotes a double bond then R4 is absent, or N-R“, taken together, forms a positively charged N substituted with C1_20 alkyl; R8 and Rb are each independently H, C1_20 alkyl, C240 alkenyl, C240 alkynyl, or C640 aryl; Rc is H, C140 alkyl, C240 alkenyl, phenyl, benzyl, a polyethylene glycol group, or an amino-polyethylene glycol group.
In some embodiments, B is a base of Formula XII-a1, XII-a2, XII-a3, XII-a4, or XII-a5: IL: ofR Rn; RELRfiHNA 0 XII-a1 XII-a2 XII-a3 XII-a4 XII-a5.
In some embodiments, the nucleobase is a pyrimidine or derivative thereof.
In some embodiments, the nucleic acid ns a plurality of structurally unique nds ofFormula XI-a.
In some embodiments, at least 25% of the cytosines are replaced by a compound of Formula XI-a (e.g., at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or about 100%).
In some embodiments, at least 25% of the uracils are replaced by a compound of Formula XI-a (e.g., at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or about 100%).
In some embodiments, at least 25% of the cytosines and 25% of the uracils are replaced by a compound of Formula XI-a (e. g., at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or about 100%).
In some embodiments, the nucleic acid is atable.
In some embodiments, when the nucleic acid includes a tide modified with a linker and payload, for example, as described herein, the nucleotide modified with a linker and d is on the 3’ end of the nucleic acid.
Maior Groove Interacting Partners As described herein, the phrase “major groove interacting r” refers RNA recognition receptors that detect and respond to RNA s through interactions, 6.g. binding, with the major groove face of a nucleotide or nucleic acid. As such, RNA ligands comprising modified nucleotides or nucleic acids as described herein decrease interactions with major groove binding partners, and therefore se an innate immune response, or expression and secretion of pro- inflammatory cytokines, or both.
Example major groove interacting, ag, binding, partners include, but are not limited to the ing nucleases and helicases. Within membranes, TLRs (loll-like Receptors) 3, Z and 8 can respond to single- and double-stranded RNAs. Within the cytoplasm, members of the superfamily 2 class of DEX(D/H) helicases and ATPases can sense RNAs to initiate antiviral ses. These helicases include the RIG-I (retinoic acid-inducible gene 1) and MDA5 (melanoma differentiation- associated gene 5). Other examples include laboratory ofgenetics and logy 2 (LGP2), HIN- 200 domain containing proteins, or Helicase-domain ning proteins.
Prevention or reduction of innate cellular immune response The term "innate immune response" includes a cellular response to exogenous single stranded nucleic acids, generally of viral or bacterial origin, which es the ion of cytokine expression and release, particularly the interferons, and cell death. Protein synthesis is also reduced during the innate cellular immune response. While it is advantageous to eliminate the innate immune response in a cell which is red by introduction of exogenous nucleic acids, the present disclosure es modified nucleic acids such as mRNAs that substantially reduce the immune response, including interferon signaling, without entirely eliminating such a response. In some embodiments, the immune response is reduced by 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 99.9%, or greater than 99.9% as compared to the immune response induced by a corresponding unmodified nucleic acid. Such a reduction can be measured by expression or activity level of Type 1 interferons or the sion of interferon-regulated genes such as the toll—like receptors (e.g., TLR7 and TLR8). Reduction or lack of induction of innate immune response can also be measured by decreased cell death following one or more administrations of modified RNAs to a cell population; e.g., cell death is 10%, 25%, 50%, 75%, 85%, 90%, 95%, or over 95% less than the cell death frequency observed with a ponding unmodified nucleic acid. Moreover, cell death may affect fewer than 50%, 40%, 30%, 20%, 10%, 5%, 1%, 0.1%, 0.01% or fewer than 0.01% of cells ted with the modified c acids.
In some embodiments, the modified nucleic acids, including polynucleotides and/or mRNA molecules are d in such a way as to not , or induce only minimally, an immune response by the recipient cell or organism. Such evasion or avoidance of an immune response trigger or activation is a novel feature of the modified polynucleotides of the t invention.
The present disclosure provides for the repeated introduction (e.g., transfection) of modified c acids into a target cell population, e.g., in vitro, ex vivo, or in vivo. The step of contacting the cell population may be repeated one or more times (such as two, three, four, five or more than five times). In some embodiments, the step of contacting the cell population with the modified nucleic acids is repeated a number of times ent such that a predetermined efficiency of protein translation in the cell population is achieved. Given the reduced cytotoxicity of the target cell population provided by the c acid modifications, such repeated transfections are achievable in a diverse array of cell types in vitro and/or in viva.
Polypeptide variants Provided are nucleic acids that encode variant ptides, which have a certain identity with a reference polypeptide sequence. The term "identity" as known in the art, refers to a relationship between the sequences of two or more peptides, as determined by comparing the sequences. In the art, "identity" also means the degree of sequence dness between peptides, as determined by the number of matches between s of two or more amino acid es. "Identity" measures the t of identical matches n the smaller of two or more sequences with gap alignments (if any) addressed by a particular mathematical model or computer program (i.e., "algorithms"). Identity of related es can be readily calculated by known s. Such methods include, but are not limited to, those described in Computational Molecular Biology, Lesk, A. M., ed., Oxford sity Press, New York, 1988; Biocomputing: atics and Genome Projects, Smith, D. W., ed., Academic Press, New York, 1993; Computer Analysis of Sequence Data, Part 1, Griffin, A. M., and Griffin, H. G., eds., Humana Press, New Jersey, 1994; Sequence Analysis in Molecular Biology, von Heinje, G., Academic Press, 1987; Sequence Analysis Primer, Gribskov, M. and Devereux, J., eds., M. Stockton Press, New York, 1991; and Carillo et al., SIAM J.
Applied Math. 48, 1073 (1988).
In some embodiments, the polypeptide t has the same or a similar activity as the reference polypeptide. Alternatively, the variant has an d activity (e.g., increased or decreased) relative to a reference polypeptide. Generally, variants of a particular polynucleotide or polypeptide of the present disclosure will have at least about 40%, 45%, 50%, 55%, 60%, 65%, 70%, ?5%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to that particular nce polynucleotide or polypeptide as determined by sequence alignment programs and parameters described herein and known to those skilled in the art.
As recognized by those skilled in the art, protein fragments, onal protein domains, and homologous proteins are also considered to be within the scope of this present disclosure. For example, provided herein is any protein fragment of a reference n (meaning a polypeptide sequence at least one amino acid residue shorter than a reference polypeptide sequence but otherwise identical) 10, 20, 30, 40, 50, 60, 70, 80, 90, 100 or greater than 100 amino acids in length In another example, any n that includes a stretch of about 20, about 30, about 40, about 50, or about 100 amino acids which are about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, or about 100% identical to any of the sequences described herein can be utilized in accordance with the t disclosure. In certain embodiments, a protein sequence to be utilized in accordance with the present disclosure includes 2, 3, 4, 5, 6, 7, 8, 9, 10, or more mutations as shown in any of the sequences provided or referenced herein.
Polypeptide libraries Also provided are polynucleotide libraries containing nucleoside modifications, n the polynucleotides individually contain a first nucleic acid sequence encoding a ptide, such as an antibody, protein binding partner, ld n, and other polypeptides known in the art.
Preferably, the polynucleotides are mRNA in a form suitable for direct introduction into a target cell host, which in turn synthesizes the encoded polypeptide.
In certain embodiments, multiple variants of a protein, each with different amino acid modification(s), are produced and tested to determine the best t in terms of pharmacokinetics, stability, biocompatibility, and/or biological activity, or a biophysical property such as expression level. Such a library may contain 10, 102, 103, 104, 105, 106, 107, 108, 109, or over 109 possible variants (including substitutions, deletions of one or more es, and insertion of one or more residues).
Polypeptide-nucleic acid complexes Proper protein translation involves the physical aggregation of a number of polypeptides and nucleic acids associated with the mRNA. Provided by the present disclosure are protein-nucleic acid complexes, containing a translatable mRNA having one or more nucleoside modifications (e.g., at least two ent nucleoside modifications) and one or more polypeptides bound to the mRNA.
Generally, the proteins are provided in an amount effective to t or reduce an innate immune response of a cell into which the complex is uced.
Untranslatable d nucleic acids As described herein, provided are mRNAs having sequences that are substantially not translatable. Such mRNA is effective as a vaccine when administered to a mammalian subject.
Also provided are modified nucleic acids that contain one or more noncoding regions.
Such modified nucleic acids are generally not translated, but are capable of binding to and tering one or more translational machinery component such as a ribosomal protein or a transfer RNA (tRNA), thereby effectively reducing protein expression in the cell. The modified nucleic acid may contain a small nucleolar RNA (sno-RNA), micro RNA ), small interfering RNA (siRNA) or Piwi-interacting RNA (piRNA).
Synthesis of Modified Nucleic Acids Nucleic acids for use in accordance with the present disclosure may be prepared according to any available technique including, but not limited to chemical synthesis, enzymatic synthesis, which is generally termed in vitro transcription, tic or chemical cleavage of a longer precursor, etc. Methods of synthesizing RNAs are known in the art (see, e.g., Gait, MJ. (ed.) Oligonucleotide synthesis: a practical approach, Oxford [Oxfordshire], Washington, DC: IRL Press, 1984; and Herdewijn, P. (ed.) Oligonucleotide synthesis: methods and applications, Methods in Molecular Biology, v. 288 (Clifton, NJ.) Totowa, N.J.: Humana Press, 2005; both of which are incorporated herein by nce).
Modified nucleic acids need not be uniformly modified along the entire length ofthe molecule. Different nucleotide modifications and/or backbone structures may exist at various positions in the nucleic acid. One of ry skill in the art will appreciate that the nucleotide analogs or other modification(s) may be located at any position(s) of a nucleic acid such that the on of the nucleic acid is not substantially decreased. A modification may also be a 5’ or 3’ al modification. The nucleic acids may contain at a minimum one and at maximum 100% modified nucleotides, or any intervening percentage, such as at least 5% modified nucleotides, at least 10% modified nucleotides, at least 25% modified nucleotides, at least 50% modified nucleotides, at least 80% modified tides, or at least 90% modified nucleotides. For example, the nucleic acids may n a modified pyrimidine such as uracil or cytosine. In some embodiments, at least 5%, at least 10%, at least 25%, at least 50%, at least 80%, at least 90% or 100% of the uracil in the nucleic acid is ed with a modified uracil. The d uracil can be replaced by a compound having a single unique structure, or can be replaced by a plurality of compounds having different structures (e.g., 2, 3, 4 or more unique structures). In some embodiments, at least 5%, at least 10%, at least 25%, at least 50%, at least 80%, at least 90% or 100% of the cytosine in the c acid is ed with a modified cytosine. The modified cytosine can be replaced by a compound having a single unique structure, or can be replaced by a plurality of compounds having different structures (e.g., 2, 3, 4 or more unique structures).
] Generally, the shortest length of a modified mRNA of the present disclosure can be the length of an mRNA sequence that is sufficient to encode for a dipeptide. In another embodiment, the length of the mRNA sequence is ent to encode for a tripeptide. In another embodiment, the length of an mRNA sequence is sufficient to encode for a tetrapeptide. In another embodiment, the length of an mRNA sequence is sufficient to encode for a pentapeptide. In r embodiment, the length of an mRNA sequence is sufficient to encode for a hexapeptide. In another embodiment, the length of an mRNA sequence is sufficient to encode for a heptapeptide. In another embodiment, the length of an mRNA sequence is sufficient to encode for an octapeptide. In another embodiment, the length of an mRNA sequence is sufficient to encode for a nonapeptide. In another embodiment, the length of an mRNA ce is sufficient to encode for a decapeptide.
Examples of dipeptides that the modified nucleic acid sequences can encode for include, but are not limited to, carnosine and anserine.
In a further embodiment, the mRNA is greater than 30 nucleotides in length. In another embodiment, the RNA le is greater than 35 tides in length. In another ment, the length is at least 40 nucleotides. In r ment, the length is at least 45 nucleotides. In another ment, the length is at least 55 nucleotides. In another ment, the length is at least 60 nucleotides. In another embodiment, the length is at least 60 nucleotides. In another embodiment, the length is at least 80 nucleotides. In another embodiment, the length is at least 90 nucleotides. In another embodiment, the length is at least 100 nucleotides. In another embodiment, the length is at least 120 nucleotides. In another embodiment, the length is at least 140 nucleotides.
In another embodiment, the length is at least 160 nucleotides. In another ment, the length is at least 180 nucleotides. In another embodiment, the length is at least 200 nucleotides. In another embodiment, the length is at least 250 nucleotides. In another embodiment, the length is at least 300 nucleotides. In another embodiment, the length is at least 350 nucleotides. In r embodiment, the length is at least 400 nucleotides. In another embodiment, the length is at least 450 nucleotides.
In another embodiment, the length is at least 500 nucleotides. In another embodiment, the length is at least 600 nucleotides. In another embodiment, the length is at least 700 nucleotides. In another embodiment, the length is at least 800 nucleotides. In another embodiment, the length is at least 900 nucleotides. In another embodiment, the length is at least 1000 nucleotides. In r embodiment, the length is at least 1100 nucleotides. In another embodiment, the length is at least 1200 nucleotides. In another embodiment, the length is at least 1300 tides. In another embodiment, the length is at least 1400 tides. In another ment, the length is at least 1500 nucleotides. In another ment, the length is at least 1600 nucleotides. In another embodiment, the length is at least 1800 nucleotides. In another embodiment, the length is at least 2000 nucleotides. In r embodiment, the length is at least 2500 nucleotides. In another embodiment, the length is at least 3000 nucleotides. In r embodiment, the length is at least 4000 nucleotides. In another embodiment, the length is at least 5000 nucleotides, or r than 5000 nucleotides.
For example, the modified nucleic acids described herein can be prepared using methods that are known to those skilled in the art of c acid synthesis.
In some embodiments, the present disclosure provides methods, e.g., enzymatic, of preparing a nucleic acid sequence comprising a nucleotide that disrupts binding of a major groove binding partner with the nucleic acid sequence, wherein the nucleic acid sequence comprises a compound ofFormula XI-a: P—‘F\ 3 B (BRc1 ,9? 2 A —§—Y‘—;=x ORc1 XI-a wherein: the nucleotide has decreased binding affinity to the major groove binding partner; \ denotes an optional double bond; - - -denotes an optional single bond; U is O, S, -NRa-, or -CRaRb- when \ denotes a single bond, or U is -CR3- when \‘\~ denotes a double bond; A is H, OH, phosphoryl, pyrophosphate, sulfate, -NH2, -SH, an amino acid, a peptide comprising 2 to 12 amino acids; X is O or S; each of Y1 is independently selected from —OR“, -N'Ra1Rb1, and —SRa1; each of Y2 and Y3 are independently selected from O, -CRaRb-, NR°, S or a linker comprising one or more atoms selected from the group consisting of C, O, N, and S; R3 and Rb are each independently H, €1.12 alkyl, C2.12 alkenyl, C2.12 alkynyl, or C620 aryl; Rc is H, CH2 alkyl, C242 alkenyl, phenyl, benzyl, a polyethylene glycol group, or an amino-polyethylene glycol group; R31 and RM are each independently H or a counterion; 70R“ is OH at a pH of about 1 or 70R“ is O’ at physiological pH; and B is nucleobase; provided that the ring encompassing the variables A, B, D, U, Z, Y2 and Y3 cannot be ribose the method comprising reacting a compound of Formula XIII: Y‘ P—Y2 3 B 6R“ ’,U n 2 j Y1 A XIII with an RNA polymerase, and a cDNA template.
In some ments, the reaction is repeated from 1 to about 7,000 times.
In some embodiments, B is a nucleobase of a XII-a, XII-b, or XII-c: R‘Y 1 f “N”R4 R‘N3 i R3\ f NH N \N U~ka v0 RAD XII-a XII-b XII-c ] wherein: \ denotes a single or double bond; X is O or S; U and W are each ndently C or N; V is O, S, C or N; n when V is C then R1 is H, C1_6 alkyl, CM alkenyl, C1_6 alkynyl, halo, or —OR°, wherein C120 alkyl, C220 alkenyl, C2.2o alkynyl are each ally substituted with —OH, -NRaRb, - SH, -C(O)R°, -C(O)OR°, -NHC(O)R°, or -NHC(O)OR°; and wherein when V is O, S, or N then R1 is absent; R2 is H, -OR°, -SR°, -NRaRb, or halo; or when V is C then R1 and R2 together with the carbon atoms to which they are attached can form a 5- or ered ring optionally substituted with 1-4 substituents selected from halo, - OH, -SH, -NRaRb, C140 alkyl, C240 alkenyl, C240 alkynyl, C1_20 alkoxy, or Cmo thioalkyl; ] R3 is H or C140 alkyl; R4 is H or C140 alkyl; wherein when \ s a double bond then R4 is absent, or N-R", taken er, forms a positively charged N substituted with C1_20 alkyl; R3 and Rb are each ndently H, C1_20 alkyl, C240 alkenyl, C240 alkynyl, or C640 aryl; Rc is H, C140 alkyl, C240 alkenyl, phenyl, benzyl, a polyethylene glycol group, or an amino-polyethylene glycol group.
In some embodiments, B is a base of Formula XII-a1 or , XII-a2, XII-a3, XII-a4, XII-a5: it: PR4 b: fiefix”NA XII-a XII-a2 XII-a3 XII-a4 XII-a5.
In some embodiments, the methods further comprise a nucleotide ed from the group consisting of ine, cytosine, guanosine, and uracil.
In some embodiments, the nucleobase is a pyrimidine or derivative thereof.
In another aspect, the present disclosure provides for methods of amplifying a nucleic acid sequence sing a nucleotide that disrupts binding of a major groove binding partner with the nucleic acid sequence, the method comprising: X :eacting a compound of Formula XI-d: Y'—P——Y2-P—Y2—P——Y2 ORC1 ORC1 ORC1Z%_zA/B XI-d wherein: the nucleotide has decreased binding affinity to the major groove binding partner; ‘\ s a single or a double bond; - - -denotes an optional single bond; U is O, S, -NRa-, or -CRaRb- when \ denotes a single bond, or U is -CRa- when ‘\~ denotes a double bond; ] Z is H, 01.12 alkyl, or C640 aryl, or Z is absent when \ denotes a double bond; and Z can be -CRaRb- and form a bond with A; A is H, OH, phosphoryl, pyrophosphate, sulfate, -NH2, -SH, an amino acid, or a peptide comprising 1 to 12 amino acids; ] X is O or S; each of Y1 is independently selected from 70R“, -NRa1Rb1, and 7SRal; ] each of Y2 and Y3 are ndently selected from O, -CRaRb-, NRC, S or a linker comprising one or more atoms selected from the group consisting of C, O, N, and S; n is 0, 1, 2, or 3; In is 0, 1, 2 or 3; B is nucleobase; R8 and Rb are each ndently H, C142 alkyl, C242 alkenyl, C242 alkynyl, or C640 aryl; R“ is H, C142 alkyl, C242 alkenyl, phenyl, benzyl, a polyethylene glycol group, or an amino-polyethylene glycol group; R81 and Rbl are each independently H or a counterion; and 70Rc1 is OH at a pH of about 1 or 70R“ is O’ at physiological pH; provided that the ring encompassing the variables A, B, D, U, Z, Y2 and Y3 cannot be ribose with a primer, a cDNA template, and an RNA polymerase.
In some embodiments, B is a nucleobase of Formula XII-a, XII-b, or XII-c: R2 o R‘VJ‘\‘N’R1 4 R‘NJLNH3 R‘N \N3 f a Js \ ‘vy x o o XII-a XII-b XII-c ‘\ denotes a single or double bond; X is O or S; U and W are each independently C or N; ] V is O, S, C or N; wherein when V is C then R1 is H, CH; alkyl, C14; alkenyl, C1.6 alkynyl, halo, or —OR°, wherein C140 alkyl, Cmo alkenyl, C240 alkynyl are each ally substituted with —OH, -NRaRb, - SH, -C(O)R°, -C(O)OR°, -NHC(O)RC, or )OR°; and wherein when V is O, S, or N then R1 is absent; R2 is H, -ORC, -SRC, -NRaRb, or halo; or when V is C then R1 and R2 together with the carbon atoms to which they are attached can form a 5— or 6-membered ring optionally substituted with 1-4 substituents selected from halo, - OH, -SH, —NRaRb, C1_20 alkyl, C240 alkenyl, C240 alkynyl, C1_20 alkoxy, or C140 thioalkyl; R3 is H or C140 alkyl; R4 is H or C140 alkyl; wherein when ‘\ denotes a double bond then R4 is absent, or N—R4, taken together, forms a vely charged N substituted with C1_20 alkyl; ] R3 and Rb are each independently H, C1_20 alkyl, C240 alkenyl, C240 alkynyl, or C640 aryl; Rc is H, C140 alkyl, C240 alkenyl, phenyl, benzyl, a polyethylene glycol group, or an amino-polyethylene glycol group.
] In some embodiments, B is a nucleobase of Formula XII-a1, XII-a2, XII-a3, XII—a4, or XII—a5: N’K NN 0 XII-a1 XII-a2 XII-a3 XII-a4 XII-a5.
In some embodiments, the methods r comprise a nucleotide selected from the group consisting of adenosine, cytosine, guanosine, and uracil.
In some embodiments, the nucleobase is a pyrimidine or derivative f, In some embodiments, the present disclosure provides for methods of synthesizing a ceutical nucleic acid, comprising the steps of: a) providing a complementary deoxyribonucleic acid (cDNA) that encodes a pharmaceutical protein of interest; b) selecting a nucleotide that is known to disrupt a binding of a major groove binding partner with a nucleic acid, n the nucleotide has decreased binding affinity to the major groove binding partner; and c) contacting the provided cDNA and the selected nucleotide with an RNA polymerase, under conditions such that the pharmaceutical nucleic acid is synthesized.
In further embodiments, the ceutical nucleic acid is a ribonucleic acid (RNA).
] In still a further aspect of the present disclosure, the modified nucleic acids can be prepared using solid phase synthesis methods.
] In some embodiments, the present disclosure provides methods of synthesizing a nucleic acid comprising a compound of Formula XI—a: -§—Y'—P—W\ C'JRc1 ,IUZ/B 2 A ,___1 0R61 XI-a wherein: \‘\~ denotes an optional double bond; ] - - —denotes an optional single bond; U is O, S, -NRa-, or -CRaRb- when ‘\~ s a single bond, or U is -CRa- when ‘\ denotes a double bond; A is H, OH, phosphoryl, pyrophosphate, sulfate, -NH2, -SH, an amino acid, a peptide comprising 2 to 12 amino acids; ] X is O or S; each of Y1 is independently selected from 70R“, -NRa1Rb1, and 7SRa1; each of Y2 and Y3 are independently selected from O, -CRaRb-, NRC, S or a linker comprising one or more atoms selected from the group consisting of C, O, N, and S; R3 and Rb are each ndently H, C142 alkyl, C242 alkenyl, C242 alkynyl, or CMO aryl; ] Rc is H, C142 alkyl, C242 alkenyl, phenyl, benzyl, a hylene glycol group, or an amino-polyethylene glycol group; R31 and Rbl are each independently H or a counterion; —OR°1 is OH at a pH of about 1 or —ORCl is O— at physiological pH; and B is nucleobase; provided that the ring encompassing the les A, B, U, Z, Y2 and Y3 cannot be ribose; comprising: a) reacting a nucleotide of Formula XIII-a: 3513 B Gt “’2 XIII-a with a phosphoramidite compound of Formula XIII-b: P1_Y\Y32 B 2);?’U 2 A—P2 XXIII-b wherein:0 denotes a solid support; and Pl, P2 and P3 are each independently le ting groups; to provide a nucleic acid of Formula XIV-a: Pl—Y2\Y3 B XIV-a and b) oxidizing or sulfurizing the nucleic acid of Formula XIV-a to yield a nucleic acid of Formula XIVb: PLYzy3 B 2);?,u 1 A—P2 I:\ T, O—Pr XIV-b and c) removing the protecting groups to yield the c acid of Formula XI-a In some embodiments, the methods further comprise a nucleotide selected from the group consisting of adenosine, cytosine, guanosine, and uracil.
In some embodiments, B is a nucleobase of Formula XIII: Mfii «yw N R5 XIII V is N or positively charged NRC; R3 is NRCRd, -OR“, or -SR“; R4 is H or can optionally form a bond with Y3; R5 is H, -NRCRd, or -ORa; R8 and Rb are each independently H, C142 alkyl, C242 l, C242 alkynyl, or C620 aryl; Rc is H, C142 alkyl, C242 alkenyl, phenyl, benzyl, a polyethylene glycol group, or an polyethylene glycol group.
In some embodiments, steps a) and b) are repeated from 1 to about 10,000 times.
Uses of d Nucleic Acids Therapeutic Agents The modified nucleic acids described herein can be used as therapeutic agents. For example, a modified nucleic acid bed herein can be administered to an animal or subject, wherein the modified nucleic acid is translated in vivo to e a eutic e in the animal or t. Accordingly, provided herein are compositions, methods, kits, and reagents for treatment or prevention of disease or conditions in humans and other s. The active therapeutic agents of the present disclosure include modified nucleic acids, cells containing modified nucleic acids or polypeptides translated from the modified nucleic acids, ptides translated from modified nucleic acids, cells contacted with cells containing modified nucleic acids or polypeptides translated from the modified nucleic acids, tissues containing cells containing modified nucleic acids and organs containing tissues containing cells containing modified nucleic acids.
Provided are methods of inducing translation of a synthetic or inant polynucleotide to e a polypeptide in a cell tion using the modified nucleic acids described herein.
Such translation can be in viva, ex viva, in culture, or in vitra. The cell population is contacted with an effective amount of a composition containing a nucleic acid that has at least one nucleoside ation, and a translatable region encoding the polypeptide. The tion is contacted under conditions such that the nucleic acid is localized into one or more cells of the cell population and the recombinant polypeptide is translated in the cell from the nucleic acid.
An effective amount of the ition is provided based, at least in part, on the target tissue, target cell type, means of administration, physical characteristics of the nucleic acid (e.g., size, and extent of modified nucleosides), and other determinants. In general, an effective amount of the composition es efficient protein production in the cell, preferably more efficient than a composition containing a corresponding unmodified nucleic acid. Increased efficiency may be demonstrated by increased cell transfection (i.e., the percentage of cells transfected with the nucleic acid), increased protein ation from the nucleic acid, decreased nucleic acid degradation (as demonstrated, e.g., by increased duration of protein translation from a modified nucleic acid), or reduced innate immune response of the ho st cell or improve therapeutic y.
Aspects of the present disclosure are directed to methods of inducing in viva translation of a recombinant polypeptide in a ian subject in need thereof. Therein, an effective amount of a composition containing a nucleic acid that has at least one nucleoside modification and a translatable region encoding the polypeptide is administered to the subject using the delivery methods described herein. The nucleic acid is provided in an amount and under other conditions such that the nucleic acid is localized into a cell or cells of the subject and the recombinant polypeptide is translated in the cell from the c acid. The cell in which the nucleic acid is zed, or the tissue in which the cell is present, may be targeted with one or more than one rounds of nucleic acid administration.
Other aspects of the present disclosure relate to transplantation of cells containing modified nucleic acids to a mammalian subject. stration of cells to mammalian subjects is known to those of ordinary skill in the art, such as local implantation (e.g., topical or aneous administration), organ delivery or systemic injection (e.g., intravenous ion or inhalation), as is the formulation of cells in pharrnaceutically acceptable carrier. Compositions containing modified nucleic acids are formulated for administration intramuscularly, transarterially, eritoneally, intravenously, intranasally, aneously, endoscopically, transdermally, or intrathecally. In some embodiments, the composition is formulated for extended release.
The t to whom the therapeutic agent is administered suffers from or is at risk of developing a disease, disorder, or deleterious condition. Provided are methods of identifying, diagnosing, and classifying subjects on these bases, which may include clinical diagnosis, biomarker levels, genome-wide association studies (GWAS), and other methods known in the art.
In certain embodiments, the administered modified nucleic acid directs production of one or more recombinant polypeptides that provide a functional activity which is substantially absent in the cell in which the recombinant polypeptide is translated. For example, the missing onal activity may be enzymatic, structural, or gene regulatory in nature.
In other embodiments, the administered modified c acid s production of one or more recombinant polypeptides that replace a polypeptide (or multiple polypeptides) that is substantially absent in the cell in which the inant ptide is translated. Such absence may be due to c mutation of the encoding gene or regulatory pathway thereof. In other embodiments, the administered modified nucleic acid directs production of one or more recombinant polypeptides to supplement the amount of polypeptide (or multiple polypeptides) that is present in the cell in which the recombinant polypeptide is translated. atively, the recombinant polypeptide functions to antagonize the activity of an endogenous protein present in, on the surface of, or secreted from the cell. Usually, the ty of the endogenous protein is deleterious to the subject, for example, due to mutation of the endogenous protein resulting in altered activity or localization. Additionally, the recombinant polypeptide antagonizes, directly or indirectly, the activity of a biological moiety present in, on the surface of, or secreted from the cell. Examples of antagonized biological moieties include lipids (e.g., cholesterol), a otein (e.g., low density lipoprotein), a nucleic acid, a carbohydrate, or a small molecule toxin.
] The recombinant proteins described herein are engineered for localization within the cell, potentially within a specific compartment such as the s, or are engineered for secretion from the cell or translocation to the plasma membrane of the cell.
As described herein, a useful feature of the modified nucleic acids of the present disclosure is the capacity to reduce, evade, avoid or ate the innate immune response of a cell to an exogenous c acid. Provided are methods for performing the titration, reduction or elimination of the immune response in a cell or a population of cells. In some embodiments, the cell is contacted with a first composition that contains a first dose of a first exogenous nucleic acid including a translatable region and at least one nucleoside modification, and the level of the innate immune response of the cell to the first exogenous nucleic acid is determined. Subsequently, the cell is contacted with a second composition, which includes a second dose of the first exogenous nucleic acid, the second dose containing a lesser amount of the first ous nucleic acid as compared to the first dose. atively, the cell is ted with a first dose of a second exogenous nucleic acid. The second exogenous nucleic acid may contain one or more modified nucleosides, which may be the same or different from the first exogenous nucleic acid or, alternatively, the second exogenous nucleic acid may not n modified nucleosides. The steps of contacting the cell with the first composition and!or the second composition may be repeated one or more times. onally, efficiency of protein production (e.g., protein translation) in the cell is optionally determined, and the cell may be re—transfected with the first and/or second composition repeatedly until a target protein production efficiency is achieved.
Merapeuticsfor diseases and conditions ] Provided are methods for treating or preventing a symptom of diseases characterized by missing or aberrant protein activity, by replacing the g protein activity or overcoming the aberrant protein activity. Because of the rapid initiation of protein production following introduction ofmodified mRNAs, as compared to viral DNA vectors, the compounds of the t sure are particularly advantageous in ng acute diseases such as sepsis, stroke, and myocardial infarction.
Moreover, the lack of transcriptional regulation of the modified mRNAs of the present disclosure is advantageous in that te titration of protein production is able. Multiple diseases are characterized by missing (or substantially diminished such that proper protein function does not occur) protein activity. Such proteins may not be present, are present in very low quantities or are essentially non-functional. The present disclosure provides a method for treating such conditions or diseases in a subject by introducing nucleic acid or cell-based eutics containing the modified nucleic acids provided herein, wherein the modified nucleic acids encode for a protein that replaces the n activity g from the target cells of the subject.
Diseases characterized by dysfunctional or aberrant protein activity include, but not limited to, cancer and proliferative diseases, genetic diseases (e.g., cystic fibrosis), autoimmune diseases, diabetes, neurodegenerative es, cardiovascular diseases, and metabolic diseases. The present disclosure es a method for treating such conditions or diseases in a subject by introducing nucleic acid or ased therapeutics containing the d nucleic acids ed herein, wherein the modified nucleic acids encode for a protein that nizes or otherwise overcomes the aberrant protein activity present in the cell of the subject.
Specific examples of a ctional protein are the missense or nonsense mutation variants of the cystic fibrosis transmembrane tance regulator (CFTR) gene, which produce a dysfimctional or nonfunctional, respectively, n variant of CFTR protein, which causes cystic Thus, provided are methods of treating cystic fibrosis in a ian subject by contacting a cell of the subject with a modified nucleic acid having a translatable region that encodes a functional CFTR polypeptide, under conditions such that an effective amount of the CTFR polypeptide is present in the cell. Preferred target cells are epithelial cells, such as the lung, and methods of administration are ined in View of the target tissue; i.e., for lung delivery, the RNA molecules are formulated for administration by inhalation.
In another embodiment, the present disclosure provides a method for treating hyperlipidemia in a subject, by introducing into a cell population of the subject with a modified mRNA molecule encoding Sortilin, a protein recently characterized by genomic studies, thereby ameliorating the ipidemia in a subject. The SORTI gene encodes a Golgi network (TGN) transmembrane protein called Sortilin. Genetic studies have shown that one of five individuals has a single nucleotide polymorphism, rs12740374, in the 1p13 locus of the SORT] gene that predisposes them to having low levels of low-density lipoprotein (LDL) and very-low-density lipoprotein (VLDL). Each copy of the minor allele, present in about 30% of , alters LDL cholesterol by 8 mg/dL, while two copies of the minor allele, present in about 5% of the population, lowers LDL cholesterol 16 mg/dL. Carriers of the minor allele have also been shown to have a 40% sed risk of dial infarction. Functional in viva studies in mice describes that overexpression of SORTI in mouse liver tissue led to significantly lower LDL-cholesterol levels, as much as 80% lower, and that silencing SORT] increased LDL cholesterol approximately 200% (Musunuru K et al.
From noncoding variant to phenotype via SORT1 at the 1p13 cholesterol locus. Nature 2010; 466: 714-721).
Methods ofcellular nucleic acid del'wery Methods of the present disclosure enhance nucleic acid delivery into a cell population, in viva, ex viva, or in culture. For example, a cell culture containing a plurality of host cells (e.g., eukaryotic cells such as yeast or ian cells) is contacted with a composition that contains an enhanced nucleic acid having at least one side modification and, optionally, a translatable region. The composition also generally contains a transfection reagent or other compound that increases the efficiency of enhanced c acid uptake into the host cells. The enhanced nucleic acid exhibits enhanced retention in the cell population, relative to a corresponding unmodified nucleic acid. The ion of the enhanced nucleic acid is greater than the retention ofthe unmodified nucleic acid. In some embodiments, it is at least about 50%, 75%, 90%, 95%, 100%, 150%, 200% or more than 200% greater than the ion of the unmodified nucleic acid. Such retention advantage may be achieved by one round of transfection with the enhanced nucleic acid, or may be obtained following repeated rounds of transfection.
In some ments, the enhanced nucleic acid is delivered to a target cell population with one or more additional nucleic acids. Such ry may be at the same time, or the enhanced c acid is delivered prior to delivery of the one or more additional nucleic acids. The additional one or more nucleic acids may be modified nucleic acids or unmodified nucleic acids. It is tood that the initial presence of the enhanced nucleic acids does not substantially induce an innate immune response of the cell population and, moreover, that the innate immune response will not be activated by the later presence of the unmodified nucleic acids. In this regard, the enhanced c acid may not itself contain a translatable region, if the protein desired to be t in the target cell population is translated from the unmodified nucleic acids. ing Maieties In ments of the present disclosure, modified nucleic acids are provided to express a protein-binding partner or a receptor on the surface of the cell, which functions to target the cell to a specific tissue space or to interact With a specific moiety, either in vivo or in vitro. Suitable protein- binding partners include antibodies and functional fragments thereof, scaffold proteins, or peptides.
Additionally, modified c acids can be employed to direct the synthesis and extracellular localization of lipids, carbohydrates, or other biological moieties.
Permanent Gene Expression ing A method for epigenetically silencing gene expression in a mammalian subject, comprising a nucleic acid where the translatable region encodes a polypeptide or polypeptides capable of directing sequence—specific histone H3 methylation to initiate chromatin formation and reduce gene transcription around specific genes for the purpose of silencing the gene. For example, a f—function mutation in the Janus Kinase 2 gene is sible for the family of Myeloproliferative Diseases.
Delivery ofa Detectable or eutic Agent to a Biological Target The modified nucleosides, modified nucleotides, and d nucleic acids described herein can be used in a number of different scenarios in which delivery of a substance (the “payload”) to a biological target is desired, for example delivery of detectable substances for detection of the target, or delivery of a therapeutic agent. Detection methods can e both imaging in vitro and in vivo imaging methods, e.g., immunohistochemistry, bioluminescence g (BLI), Magnetic Resonance Imaging (MRI), positron emission aphy (PET), electron microscopy, X-ray computed tomography, Raman imaging, l coherence tomography, tion imaging, thermal imaging, fluorescence reflectance imaging, fluorescence microscopy, fluorescence molecular tomographic imaging, nuclear magnetic resonance g, X-ray imaging, ultrasound imaging, coustic imaging, lab assays, or in any situation where tagging/staining/imaging is required.
For example, the modified sides, modified nucleotides, and modified nucleic acids described herein can be used in reprogramming induced pluripotent stem cells (iPS cells), which can then be used to directly track cells that are transfected compared to total cells in the cluster. In another example, a drug that is attached to the modified nucleic acid via a linker and is cently labeled can be used to track the drug in viva, eg, ellularly. Other examples include the use of a modified nucleic acid in reversible drug delivery into cells.
The modified nucleosides, modified nucleotides, and modified nucleic acids described herein can be used in intracellular ing of a payload, e.g., detectable or therapeutic agent, to specific organelle. Exemplary intracellular targets can include the nuclear localization for advanced mRNA processing, or a nuclear localization sequence (NLS) linked to the mRNA containing an inhibitor.
In addition, the modified sides, modified nucleotides, and modified nucleic acids described herein can be used to r therapeutic agents to cells or tissues, e.g., in living animals.
For example, the modified nucleosides, d nucleotides, and modified nucleic acids described herein can be used to deliver highly polar chemotherapeutics agents to kill cancer cells. The d nucleic acids attached to the therapeutic agent through a linker can facilitate member permeation allowing the therapeutic agent to travel into a cell to reach an ellular target.
In another e, the d nucleosides, modified nucleotides, and modified nucleic acids can be attached to a Viral inhibitory peptide (VIP) through a cleavable linker. The cleavable linker will release the VIP and dye into the cell. In another example, the modified nucleosides, modified nucleotides, and modified c acids can be attached through the linker to a ADP- ribosylate, which is responsible for the s of some bacterial toxins, such as cholera toxin, diphtheria toxin, and pertussis toxin. These toxin proteins are bosyltransferases that modify target proteins in human cells. For example, cholera toxin ADP-ribosylates G proteins, g massive fluid secretion from the lining of the small intestine, resulting in life-threatening diarrhea.
Pharmaceutical Compositions The present disclosure provides proteins generated from modified mRNAs. ceutical compositions may optionally comprise one or more additional therapeutically active substances. In accordance with some embodiments, a method of administering pharmaceutical compositions comprising a modified nucleic acide encoding one or more proteins to be delivered to a subject in need thereof is provided. In some embodiments, compositions are administered to humans. For the purposes of the present disclosure, the phrase “active ient” generally refers to a protein, protein encoding or protein-containing complex as described herein.
] Although the descriptions of pharmaceutical compositions provided herein are principally directed to pharmaceutical itions which are suitable for administration to humans, it will be understood by the skilled artisan that such compositions are generally suitable for administration to animals of all sorts. Modification of pharmaceutical compositions suitable for administration to humans in order to render the compositions suitable for administration to various animals is well understood, and the ordinarily skilled veterinary pharrnacologist can design and/or perform such modification with merely ry, if any, experimentation. Subjects to which administration of the pharmaceutical itions is contemplated include, but are not limited to, humans and/or other es; s, including commercially relevant mammals such as cattle, pigs, horses, sheep, cats, dogs, mice, and/or rats; and/or birds, including commercially relevant birds such as ns, ducks, geese, and/or turkeys. ations of the pharmaceutical compositions described herein may be prepared by any method known or hereafter developed in the art of pharmacology. In general, such preparatory methods include the step of bringing the active ingredient into association with an excipient and/or one or more other accessory ingredients, and then, if necessary and/or desirable, g and/or packaging the product into a desired single- or multi-dose unit.
A pharmaceutical composition in accordance with the t disclosure may be prepared, packaged, and/or sold in bulk, as a single unit dose, and/or as a plurality of single unit doses. As used herein, a “unit dose” is discrete amount of the pharmaceutical composition comprising a predetermined amount of the active ingredient. The amount of the active ingredient is generally equal to the dosage of the active ingredient which would be administered to a subject and/or a convenient fraction of such a dosage such as, for e, one-half or one-third of such a dosage.
Relative s of the active ingredient, the pharrnaceutically acceptable excipient, and/or any additional ingredients in a pharmaceutical composition in accordance with the present disclosure will vary, depending upon the identity, size, and/or condition of the subject treated and further depending upon the route by which the composition is to be administered. By way of example, the ition may comprise between 0.1% and 100% (w/w) active ingredient.
Pharmaceutical formulations may additionally comprise a pharmaceutically able excipient, which, as used herein, includes any and all solvents, dispersion media, diluents, or other liquid vehicles, dispersion or suspension aids, e active , isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired. Remington’s The Science and ce ofPharmacy, 215' Edition, A. R.
Gennaro ncott, Williams & Wilkins, Baltimore, MD, 2006; incorporated herein by reference) discloses various excipients used in formulating pharmaceutical compositions and known techniques for the preparation thereof. Except insofar as any conventional excipient medium is incompatible with a substance or its derivatives, such as by producing any rable biological effect or otherwise interacting in a deleterious manner with any other component(s) of the ceutical composition, its use is contemplated to be within the scope of this present disclosure.
In some embodiments, a pharrnaceutically acceptable excipient is at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% pure. In some embodiments, an excipient is approved for use in humans and for veterinary use. In some embodiments, an excipient is ed by United States Food and Drug Administration. In some embodiments, an excipient is pharmaceutical grade. In some embodiments, an excipient meets the standards of the United States Pharmacopoeia (USP), the an Pharmacopoeia (EP), the h Pharmacopoeia, and/or the International Pharmacopoeia.
Pharmaceutically acceptable excipients used in the manufacture of ceutical compositions include, but are not limited to, inert diluents, dispersing and/or granulating , surface active agents and/or emulsifiers, disintegrating agents, binding agents, preservatives, buffering agents, ating agents, and/or oils. Such excipients may optionally be included in pharmaceutical ations. Excipients such as cocoa butter and suppository waxes, coloring agents, coating agents, ning, ng, and/or perfuming agents can be present in the composition, according to the judgment of the ator.
Exemplary diluents include, but are not limited to, calcium carbonate, sodium carbonate, calcium phosphate, dicalcium phosphate, calcium sulfate, calcium hydrogen phosphate, sodium phosphate lactose, sucrose, cellulose, microcrystalline cellulose, kaolin, mannitol, sorbitol, inositol, sodium chloride, dry starch, cornstarch, powdered sugar, eta, and/or ations thereof.
Exemplary granulating and/or dispersing agents include, but are not limited to, potato starch, corn , tapioca starch, sodium starch glycolate, clays, alginic acid, guar gum, citrus pulp, agar, bentonite, cellulose and wood products, natural sponge, cation-exchange resins, calcium carbonate, silicates, sodium carbonate, cross-linked poly(vinyl-pyrrolidone) (crospovidone), sodium carboxymethyl starch (sodium starch glycolate), carboxymethyl ose, cross-linked sodium carboxymethyl ose (croscarmellose), methylcellulose, pregelatinized starch (starch 1500), microcrystalline starch, water insoluble starch, calcium carboxymethyl cellulose, magnesium aluminum silicate (Veegum), sodium lauryl sulfate, nary ammonium compounds, eta, and/or combinations thereof.
Exemplary surface active agents and/or emulsifiers include, but are not limited to, natural emulsifiers (e.g. acacia, agar, alginic acid, sodium alginate, tragacanth, chondrux, cholesterol, xanthan, pectin, gelatin, egg yolk, casein, wool fat, cholesterol, wax, and lecithin), colloidal clays (e.g. bentonite num silicate] and Veegum® [magnesium aluminum silicate]), long chain amino acid derivatives, high molecular weight alcohols (e.g. stearyl alcohol, cetyl l, oleyl alcohol, triacetin earate, ethylene glycol distearate, glyceryl monostearate, and propylene glycol monostearate, polyvinyl alcohol), carbomers (e.g. carboxy polymethylene, polyacrylic acid, acrylic acid polymer, and carboxyvinyl polymer), carrageenan, cellulosic derivatives (6.g. carboxymethylcellulose sodium, powdered cellulose, hydroxymethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, methylcellulose), sorbitan fatty acid esters (ag. polyoxyethylene sorbitan monolaurate ®20], polyoxyethylene sorbitan ®60], yethylene sorbitan monooleate [Tween®80], sorbitan monopalmitate [Span®40], sorbitan monostearate [Span®60], sorbitan tristearate [Span®65], glyceryl monooleate, sorbitan monooleate [Span®80]), yethylene esters (e.g. polyoxyethylene monostearate [My1j®45], polyoxyethylene hydrogenated castor oil, hoxylated castor oil, polyoxymethylene stearate, and Solutol®), sucrose fatty acid esters, polyethylene glycol fatty acid esters (e.g. Cremophor®), polyoxyethylene ethers, (e.g. polyoxyethylene lauryl ether [Brij®30]), poly(vinyl-pyrrolidone), diethylene glycol monolaurate, triethanolamine oleate, sodium oleate, potassium oleate, ethyl oleate, oleic acid, ethyl laurate, sodium lauryl sulfate, Pluronic®F 68, Poloxamer®l 88, onium bromide, cetylpyridinium de, benzalkonium chloride, docusate , etc. and/or combinations thereof. ary binding agents include, but are not d to, starch (e.g. cornstarch and starch paste); gelatin; sugars (e.g. sucrose, glucose, dextrose, dextrin, molasses, e, lactitol, mannitol,); natural and synthetic gums (e.g. acacia, sodium alginate, extract of Irish moss, panwar gum, ghatti gum, mucilage of isapol husks, carboxymethylcellulose, methylcellulose, ethylcellulose, hydroxyethylcellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, microcrystalline cellulose, cellulose acetate, poly(vinyl-pyrrolidone), magnesium aluminum silicate m®), and larch arabogalactan); alginates; polyethylene oxide; polyethylene glycol; nic m salts; silicic acid; polymethacrylates; waxes; water; alcohol; etc.; and combinations thereof. ary preservatives may include, but are not limited to, antioxidants, chelating agents, antimicrobial vatives, antifungal preservatives, alcohol preservatives, acidic preservatives, and/or other preservatives. ary antioxidants include, but are not limited to, alpha tocopherol, ascorbic acid, l palmitate, butylated hydroxyanisole, butylated hydroxytoluene, monothioglycerol, potassium sulfite, propionic acid, propyl gallate, sodium ate, sodium bisulfite, sodium metabisulflte, and/or sodium sulfite. Exemplary chelating agents include ethylenediaminetetraacetic acid , citric acid monohydrate, disodium edetate, ssium edetate, edetic acid, c acid, malic acid, oric acid, sodium edetate, tartaric acid, and/or trisodium edetate. Exemplary crobial preservatives include, but are not limited to, benzalkonium chloride, benzethonium chloride, benzyl alcohol, bronopol, cetrimide, cetylpyridinium chloride, chlorhexidine, chlorobutanol, chlorocresol, chloroxylenol, cresol, ethyl alcohol, glycerin, hexetidine, imidurea, , phenoxyethanol, phenylethyl alcohol, phenylmercuric nitrate, ene glycol, and/or thimerosal. Exemplary antifungal preservatives include, but are not limited to, butyl paraben, methyl paraben, ethyl n, propyl paraben, benzoic acid, hydroxybenzoic acid, potassium benzoate, potassium sorbate, sodium benzoate, sodium nate, and/or sorbic acid. Exemplary l vatives include, but are not limited to, ethanol, polyethylene glycol, phenol, phenolic compounds, bisphenol, chlorobutanol, hydroxybenzoate, and/or phenylethyl alcohol. Exemplary acidic preservatives include, but are not limited to, Vitamin A, Vitamin C, Vitamin E, beta-carotene, citric acid, acetic acid, dehydroacetic acid, ascorbic acid, sorbic acid, and/or phytic acid. Other preservatives include, but are not limited to, tocopherol, tocopherol acetate, deteroxime mesylate, cetrimide, butylated hydroxyanisol (BHA), butylated ytoluened (BHT), ethylenediamine, sodium lauryl sulfate (SLS), sodium lauryl ether sulfate (SLES), sodium bisulfite, sodium metabisulfite, potassium sulfite, potassium sulfite, Glydant Plus®, Phenonip®, methylparaben, Germall®115, Germaben®II, Neolonem, Kathonm, and/or Euxyl®.
Exemplary buffering agents include, but are not limited to, citrate buffer solutions, acetate buffer solutions, phosphate buffer solutions, ammonium chloride, calcium carbonate, calcium chloride, calcium citrate, calcium glubionate, calcium gluceptate, calcium ate, d—gluconic acid, calcium glycerophosphate, calcium lactate, propanoic acid, calcium levulinate, pentanoic acid, dibasic calcium phosphate, oric acid, tribasic calcium phosphate, calcium hydroxide phosphate, potassium acetate, potassium chloride, potassium gluconate, potassium es, dibasic potassium phosphate, monobasic potassium phosphate, potassium phosphate mixtures, sodium acetate, sodium bicarbonate, sodium chloride, sodium citrate, sodium lactate, dibasic sodium ate, monobasic sodium phosphate, sodium phosphate mixtures, tromethamine, magnesium hydroxide, aluminum hydroxide, c acid, pyrogen-free water, isotonic saline, Ringer’s solution, ethyl alcohol, etc, and/or combinations thereof.
Exemplary lubricating agents include, but are not limited to, magnesium stearate, calcium stearate, stearic acid, silica, talc, malt, glyceryl behanate, hydrogenated vegetable oils, polyethylene glycol, sodium benzoate, sodium acetate, sodium chloride, leucine, magnesium lauryl sulfate, sodium lauryl e, etc, and combinations thereof Exemplary oils include, but are not limited to, almond, apricot kernel, avocado, babassu, bergamot, black current seed, borage, cade, camomile, canola, caraway, camauba, castor, cinnamon, cocoa butter, coconut, cod liver, coffee, corn, cotton seed, emu, eucalyptus, evening primrose, fish, flaxseed, geraniol, gourd, grape seed, hazel nut, hyssop, pyl myristate, jojoba, kukui nut, lavandin, lavender, lemon, litsea cubeba, macademia nut, mallow, mango seed, meadowfoam seed, mink, nutmeg, olive, orange, orange roughy, palm, palm , peach , peanut, poppy seed, pumpkin seed, rapeseed, rice bran, ry, safflower, sandalwood, sasquana, savoury, sea buckthom, sesame, shea butter, silicone, soybean, sunflower, tea tree, thistle, tsubaki, vetiver, walnut, and wheat germ oils. Exemplary oils e, but are not limited to, butyl stearate, caprylic triglyceride, capric triglyceride, cyclomethicone, diethyl te, icone 360, isopropyl ate, mineral oil, odecanol, oleyl alcohol, silicone oil, and/or combinations thereof.
] Liquid dosage forms for oral and parenteral administration include, but are not limited to, pharrnaceutically able emulsions, mulsions, solutions, suspensions, syrups, and/or elixirs. In addition to active ingredients, liquid dosage forms may comprise inert ts commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene , 1,3-butylene glycol, dirnethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, oral compositions can include adjuvants such as g agents, emulsifying and suspending agents, sweetening, flavoring, and/or perfuming agents. In certain ments for parenteral stration, compositions are mixed with solubilizing agents such as Cremophor®, alcohols, oils, modified oils, glycols, polysorbates, cyclodextrins, polymers, and/or combinations thereof.
Injectable ations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing agents, wetting agents, and/or suspending agents. Sterile injectable preparations may be sterile injectable solutions, suspensions, and/or emulsions in nontoxic parenterally acceptable diluents and/or solvents, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P., and isotonic sodium chloride solution. Sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed including synthetic mono- or diglycerides. Fatty acids such as oleic acid can be used in the preparation of injectables.
Injectable formulations can be sterilized, for example, by filtration through a bacterialretaining filter, and/or by incorporating sterilizing agents in the form of sterile solid compositions which can be ved or dispersed in sterile water or other sterile injectable medium prior to use.
In order to prolong the effect of an active ient, it is often ble to slow the absorption of the active ient from subcutaneous or intramuscular inj ection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water lity. The rate of absorption of the drug then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle. Injectable depot forms are made by forming microencapsule matrices of the drug in radable polymers such as polylactide—polyglycolide. Depending upon the ratio of drug to polymer and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable ations are prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissues.
Compositions for rectal or l stration are lly suppositories which can be prepared by mixing compositions with suitable non-irritating excipients such as cocoa , polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body ature and therefore melt in the rectum or vaginal cavity and release the active ingredient.
Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, an active ingredient is mixed with at least one inert, ceutically acceptable excipient such as sodium citrate or dicalcium phosphate and/or fillers or extenders (e.g. starches, lactose, e, glucose, mannitol, and silicic acid), s (e.g. carboxymethylcellulose, tes, gelatin, polyvinylpyrrolidinone, sucrose, and acacia), humectants (e.g. glycerol), disintegrating agents (e.g. agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate), solution retarding agents (e.g. paraffin), absorption rators (e. g. quaternary ammonium compounds), wetting agents (6.g. cetyl alcohol and glycerol monostearate), absorbents (e.g, kaolin and ite clay), and ants (e.g. talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate), and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may se buffering agents.
Solid compositions of a similar type may be ed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like. Solid dosage forms of tablets, s, capsules, pills, and granules can be prepared with gs and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally comprise opacifying agents and can be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include ric substances and waxes. Solid compositions of a similar type may be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene s and the like.
Dosage forms for topical and/or transdermal stration of a composition may include ents, , creams, lotions, gels, powders, solutions, , inhalants and/or patches.
Generally, an active ingredient is admixed under sterile conditions with a pharrnaceutically acceptable excipient and/or any needed preservatives and/or buffers as may be required.
Additionally, the present disclosure contemplates the use of transdermal patches, which ofien have the added advantage of providing controlled delivery of a compound to the body. Such dosage forms may be prepared, for example, by dissolving and/or dispensing the compound in the proper medium. Alternatively or additionally, rate may be controlled by either ing a rate controlling membrane and/or by dispersing the nd in a polymer matrix and/or gel. le devices for use in delivering intraderrnal pharmaceutical compositions described herein include short needle devices such as those described in US. Patents 4,886,499; 5,190,521; ,328,483; 288; 4,270,537; 235; 5,141,496; and 5,417,662. Intradermal compositions may be administered by devices which limit the effective penetration length of a needle into the skin, such as those described in PCT publication WO 99/34850 and functional equivalents thereof. Jet injection devices which deliver liquid compositions to the dermis Via a liquid jet injector and/or via a needle which pierces the stratum corneum and produces a jet which reaches the dermis are suitable.
Jet injection devices are described, for example, in US. s 381; 302; 5,334,144; ,993,412; 5,649,912; 5,569,189; 5,704,911; 5,383,851; 5,893,397; 5,466,220; 5,339,163; ,312,335; 5,503,627; 5,064,413; 5,520,639; 4,596,556; 4,790,824; 4,941,880; 4,940,460; and PCT publications WO 97/37705 and W0 97/13537. Ballistic powder/particle delivery s which use compressed gas to accelerate vaccine in powder form through the outer layers of the skin to the dermis are suitable. Alternatively or additionally, tional syringes may be used in the classical mantoux method of intradermal administration.
Formulations suitable for topical administration e, but are not limited to, liquid and/or semi liquid preparations such as liniments, lotions, oil in water and/or water in oil emulsions such as creams, ointments and/or pastes, and/or solutions and/or suspensions. Topically- administrable formulations may, for example, comprise from about 1% to about 10% (w/w) active ingredient, although the concentration of active ingredient may be as high as the solubility limit of the active ingredient in the solvent. Formulations for topical administration may further comprise one or more of the additional ingredients described herein.
] A pharmaceutical composition may be prepared, packaged, and/or sold in a formulation suitable for pulmonary administration Via the buccal cavity. Such a formulation may comprise dry les which comprise the active ingredient and which have a diameter in the range from about 0.5 nm to about 7 nm or from about 1 nm to about 6 nm. Such compositions are iently in the form of dry powders for administration using a device comprising a dry powder oir to which a stream of propellant may be directed to disperse the powder and/or using a self propelling solvent/powder dispensing container such as a device comprising the active ingredient dissolved and/or suspended in a low-boiling propellant in a sealed container. Such powders comprise particles wherein at least 98% of the particles by weight have a diameter greater than 0.5 nm and at least 95% of the particles by number have a er less than 7 nm. Alternatively, at least 95% ofthe particles by weight have a diameter greater than 1 nm and at least 90% of the particles by number have a diameter less than 6 nm. Dry powder compositions may include a solid fine powder diluent such as sugar and are conveniently provided in a unit dose form.
Low boiling propellants generally include liquid propellants having a boiling point of below 65 °F at atmospheric re. lly the lant may constitute 50% to 99.9% (w/w) of the composition, and active ingredient may constitute 0.1% to 20% (w/w) of the composition. A propellant may further comprise additional ingredients such as a liquid non-ionic and/or solid anionic surfactant and/or a solid diluent (which may have a particle size of the same order as particles comprising the active ingredient). ceutical itions formulated for pulmonary delivery may provide an active ingredient in the form of droplets of a solution and/or suspension. Such formulations may be prepared, packaged, and/or sold as aqueous and/or dilute alcoholic solutions arid/or suspensions, optionally sterile, comprising active ingredient, and may conveniently be stered using any nebulization and/or atomization device. Such formulations may further comprise one or more additional ingredients including, but not limited to, a flavoring agent such as rin sodium, a volatile oil, a buffering agent, a surface active agent, and/or a preservative such as methylhydroxybenzoate. Droplets ed by this route of administration may have an average diameter in the range from about 0.1 nm to about 200 nm.
Formulations described herein as being useful for ary delivery are useful for intranasal delivery of a pharmaceutical composition. Another formulation suitable for intranasal administration is a coarse powder comprising the active ingredient and having an average particle from about 0.2 urn to 500 um. Such a ation is administered in the manner in which snuff is taken, i.e. by rapid tion through the nasal passage from a container of the powder held close to the nose.
Formulations suitable for nasal administration may, for example, comprise from about as little as 0.1% (w/w) and as much as 100% (w/w) of active ient, and may comprise one or more ofthe additional ingredients described herein. A pharmaceutical composition may be prepared, packaged, and/or sold in a formulation suitable for buccal stration. Such formulations may, for example, be in the form of tablets and/or es made using conventional methods, and may, for example, 0.1% to 20% (w/w) active ingredient, the balance comprising an orally dissolvable and/or degradable composition and, optionally, one or more of the additional ingredients described herein. Alternately, formulations suitable for buccal administration may comprise a powder and/or an aerosolized and/or ed solution and/or suspension comprising active ingredient. Such powdered, aerosolized, and/or lized formulations, when dispersed, may have an average particle and/or droplet size in the range from about 0.1 nm to about 200 nm, and may further comprise one or more of any onal ingredients described herein.
A pharmaceutical composition may be prepared, packaged, and/or sold in a formulation suitable for ophthalmic administration. Such formulations may, for example, be in the form of eye drops including, for example, a 0. 1/1.0% (w/w) solution and/or suspension of the active ingredient in an aqueous or oily liquid excipient. Such drops may further comprise buffering agents, salts, and/or one or more other of any additional ingredients described herein. Other opthalmically-administrable formulations which are useful include those which comprise the active ingredient in microcrystalline form and/or in a mal preparation. Ear drops and/or eye drops are contemplated as being within the scope of this present disclosure.
General considerations in the formulation and/or manufacture of pharmaceutical agents may be found, for example, in Remington: The Science and Practice ofPharmacy 21St ed., Lippincott Williams & s, 2005 (incorporated herein by reference).
Administration The present disclosure provides methods comprising administering proteins or complexes in accordance with the t disclosure to a subject in need thereof. Proteins or complexes, or pharmaceutical, imaging, diagnostic, or prophylactic itions thereof, may be administered to a t using any amount and any route of administration effective for preventing, treating, diagnosing, or imaging a disease, er, and/or condition (e.g., a e, disorder, and/or condition relating to working memory deficits). The exact amount required will vary from t to t, depending on the species, age, and general ion of the t, the severity of the disease, the particular composition, its mode of administration, its mode of activity, and the like.
Compositions in accordance with the present disclosure are typically formulated in dosage unit form for ease of administration and uniformity of dosage. It will be understood, however, that the total daily usage of the itions of the present disclosure will be decided by the attending physician within the scope of sound medical judgment. The specific therapeutically effective, prophylactically effective, or riate imaging dose level for any ular patient will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of stration, route of stration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed; and like factors well known in the medical arts.
Proteins to be delivered and/or pharmaceutical, prophylactic, stic, or imaging compositions thereofmay be administered to animals, such as mammals (e.g., humans, domesticated s, cats, dogs, mice, rats, etc.). In some ments, pharmaceutical, prophylactic, diagnostic, or imaging compositions thereof are administered to humans.
Proteins to be red and/or pharmaceutical, prophylactic, diagnostic, or g compositions thereof in accordance with the present disclosure may be administered by any route.
In some embodiments, proteins and/or pharmaceutical, prophylactic, diagnostic, or g compositions thereof, are administered by one or more of a variety of routes, including oral, intravenous, intramuscular, intra—arterial, intramedullary, intrathecal, subcutaneous, intraventricular, transdermal, interderrnal, rectal, intravaginal, intraperitoneal, topical (e.g. by powders, nts, creams, gels, lotions, and/or drops), mucosal, nasal, buccal, enteral, vitreal, intratumoral, sublingual; by intratracheal instillation, bronchial instillation, and/or inhalation; as an oral spray, nasal spray, and/or aerosol, and/or through a portal vein catheter. In some ments, proteins or complexes, and/or pharmaceutical, prophylactic, diagnostic, or imaging compositions thereof, are administered by systemic intravenous ion. In ic embodiments, proteins or complexes and/or pharmaceutical, prophylactic, diagnostic, or g compositions thereofmay be administered intravenously and/or orally. In specific embodiments, ns or complexes, and/or pharmaceutical, prophylactic, diagnostic, or imaging compositions f, may be administered in a way which allows the protein or x to cross the blood-brain barrier, vascular barrier, or other epithelial barrier.
However, the present disclosure encompasses the delivery of proteins or complexes, and/or pharmaceutical, prophylactic, diagnostic, or imaging compositions thereof, by any appropriate route taking into consideration likely advances in the sciences of drug delivery.
In general the most appropriate route of administration will depend upon a variety of factors including the nature of the protein or x comprising proteins ated with at least one agent to be delivered (e.g., its stability in the environment of the gastrointestinal tract, tream, etc), the condition of the patient (e.g., whether the patient is able to tolerate particular routes of administration), etc. The present disclosure encompasses the delivery of the pharmaceutical, prophylactic, stic, or imaging compositions by any appropriate route taking into consideration likely advances in the sciences of drug delivery.
] In certain embodiments, compositions in accordance with the present disclosure may be administered at dosage levels sufficient to deliver from about 0.0001 mg/kg to about 100 mg/kg, from about 0.01 mg/kg to about 50 mg/kg, from about 0.1 mg/kg to about 40 mg/kg, from about 0.5 mg/kg to about 30 mg/kg, fiom about 0.01 mg/kg to about 10 mg/kg, from about 0.1 mg/kg to about mg/kg, or from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired eutic, diagnostic, prophylactic, or imaging effect. The desired dosage may be delivered three times a day, two times a day, once a day, every other day, every third day, every week, every two weeks, every three weeks, or every four weeks. In certain embodiments, the desired dosage may be delivered using multiple administrations (e.g., two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, en, or more administrations).
Proteins or xes may be used in combination with one or more other therapeutic, lactic, diagnostic, or imaging agents. By “in combination with,” it is not intended to imply that the agents must be administered at the same time and/or ated for delivery together, although these methods of delivery are within the scope of the present disclosure. Compositions can be administered concurrently with, prior to, or subsequent to, one or more other desired therapeutics or medical procedures. In general, each agent will be administered at a dose and/or on a time schedule determined for that agent. In some embodiments, the present disclosure encompasses the delivery of pharmaceutical, prophylactic, diagnostic, or imaging compositions in combination with agents that improve their bioavailability, reduce and/or modify their metabolism, inhibit their excretion, and/or modify their distribution within the body.
It will further be appreciated that therapeutically, prophylactically, diagnostically, or imaging active agents utilized in combination may be administered together in a single composition or administered separately in different compositions. In general, it is expected that agents utilized in combination with be utilized at levels that do not exceed the levels at which they are utilized individually. In some embodiments, the levels utilized in combination will be lower than those utilized individually.
The particular combination of therapies (therapeutics or procedures) to employ in a ation regimen will take into t ibility of the desired therapeutics and/or procedures and the desired therapeutic effect to be achieved. It will also be appreciated that the therapies employed may achieve a desired effect for the same er (for example, a composition useful for treating cancer in accordance with the t sure may be administered concurrently with a chemotherapeutic agent), or they may achieve different effects (e.g., control of any adverse effects).
The present sure provides a variety of kits for conveniently and/or effectively carrying out methods of the present disclosure. Typically kits will se sufficient amounts and/or numbers of components to allow a user to perform multiple treatments of a subject(s) and/or to perform multiple experiments.
In one aspect, the sure provides kits for protein production, comprising a first isolated nucleic acid sing a translatable region and a nucleic acid modification, wherein the nucleic acid is capable of evading or avoiding ion of an innate immune response of a cell into which the first isolated nucleic acid is introduced, and packaging and instructions.
In one , the disclosure es kits for protein production, comprising: a first isolated modified nucleic acid comprising a translatable region, provided in an amount effective to produce a desired amount of a protein encoded by the translatable region when introduced into a target cell; a second nucleic acid comprising an tory nucleic acid, provided in an amount effective to substantially inhibit the innate immune response of the cell; and packaging and instructions.
In one aspect, the sure provides kits for protein production, comprising a first isolated nucleic acid comprising a translatable region and a nucleoside modification, wherein the nucleic acid exhibits reduced degradation by a cellular nuclease, and packaging and instmctions.
In one aspect, the sure provides kits for protein production, comprising a first isolated nucleic acid comprising a translatable region and at least two different nucleoside modifications, n the nucleic acid ts reduced degradation by a cellular nuclease, and packaging and instructions.
In one aspect, the disclosure provides kits for n production, comprising a first isolated nucleic acid comprising a translatable region and at least one nucleoside modification, wherein the c acid ts reduced degradation by a cellular nuclease; a second c acid comprising an tory nucleic acid; and packaging and instructions.
In some embodiments, the first isolated nucleic acid comprises messenger RNA (mRNA).
In some embodiments the mRNA comprises at least one nucleoside selected from the group consisting of pyridinone ribonucleoside, S-aza-uridine, 2-thioaza-uridine, 2-thiouridine, 4-thio- pseudouridine, 2-thio-pseudouridine, 5-hydroxyuridine, 3-methyluridine, 5-carboxymethyl-uridine, l-carboxymethyl-pseudouridine, 5-propynyl-uridine, ynyl-pseudou1idine, 5- taurinomethyluridine, l-taurinomethyl-pseudouridine, 5-tau1inomethylthio-uridine, l- taurinomethylthio-uridine, 5-methyl-uridine, 1-methyl-pseudou1idine, 4-thiomethylpseudouridine , 2-thiomethyl-pseudouridine, 1-methyldeaza—pseudouridine, 2-thio- 1 l deaza-pseudouridine, dihydrouridine, dihydropseudouridine, 2-thio-dihydrouridine, 2-thio- dihydropseudouridine, 2-methoxyuridine, 2-methoxy-4—thio-uridine, 4-methoxy—pseudou1idine, 4— methoxythio-pseudouridine or any sed herein.
In some embodiments, the mRNA comprises at least one nucleoside selected from the group consisting of 5-aza—cytidine, pseudoisocytidine, 3-methyl-cytidine, N4-acetylcytidine, 5- formylcytidine, N4-methylcytidine, 5-hydroxymethylcytidine, 1-methyl-pseudoisocytidine, pyrrolo— cytidine, o-pseudoisocytidine, 2-thio-cytidine, 2-thiomethyl-cytidine, 4-thiopseudoisocytidine , 4-thiomethyl-pseudoisocytidine, 4-thiomethyldeaza-pseudoisocytidine, l-methyl—l—deaza—pseudoisocytidine, zebularine, 5-aza-Zebularine, 5-methyl-Zebularine, 5-aza-2— thio-zebularine, 2-thio-zebularine, 2-methoxy-cytidine, 2-methoxymethyl—cytidine, 4-methoxy— pseudoisocytidine, 4-methoxymethyl-pseudoisocytidine or any disclosed .
In some embodiments, the mRNA comprises at least one nucleoside selected from the group consisting of 2-aminopurine, 2, 6-diaminopurine, 7-deaza—adenine, 7-deaza—8-aza—adenine, 7- deaza—2-aminopurine, 7-deazaazaaminopurine, 7-deaza-2,6-diaminopurine, 7-deaza—8—aza—2,6- diaminopurine, l-methyladenosine, N6-methyladenosine, N6-isopentenyladenosine, s— hydroxyisopentenyl)adenosine, 2-methylthio-N6-(cis-hydroxyisopentenyl) ine, N6— glycinylcarbamoyladenosine, N6-threonylcarbamoyladenosine, 2-methylthio-N6-threonyl carbamoyladenosine, N6,N6-dimethyladenosine, 7-methyladenine, 2-methylthio-adenine, 2- methoxy-adenine or any disclosed herein.
] In some embodiments, the mRNA comprises at least one nucleoside ed from the group consisting of inosine, l-methyl-inosine, wyosine, wybutosine, 7-deaza—guanosine, ?-deaza—8- aza-guanosine, 6-thio-guanosine, deaza—guanosine, 6-thiodeaza—8-aza—guanosine, 7- methyl-guanosine, 6—thiomethyl—guanosine, ylinosine, 6-methoxy—guanosine, l- methylguanosine, N2-methylguanosine, N2,N2-dimethylguanosine, 8-oxo-guanosine, 7-methyl oxo-guanosine, l-methylthio-guanosine, N2-methylthio-guanosine, N2,N2-dimethylthio- guanosine or any sed herein.
In another aspect, the sure provides compositions for protein production, comprising a first isolated nucleic acid comprising a translatable region and a nucleoside modification, wherein the nucleic acid exhibits reduced degradation by a ar nuclease, and a mammalian cell suitable for ation of the translatable region of the first nucleic acid.
Definitions ] At various places in the present specification, substituents of compounds of the present disclosure are disclosed in groups or in . It is specifically ed that the present disclosure include each and every dual subcombination of the members of such groups and ranges. For example, the term “CM alkyl” is specifically intended to dually disclose methyl, ethyl, C3 alkyl, C4 alkyl, C5 alkyl, and C5 alkyl.
About: As used herein, the term “about” means +/- 10% of the recited value.
Administered in combination: As used herein, the term “administered in combination” or “combined administration” means that two or more agents are administered to a subject at the same time or within an interval such that there may be an overlap of an effect of each agent on the patient.
In some ments, they are administered within about 60, 30, 15, 10, 5, or 1 minute of one another. In some embodiments, the administrations of the agents are spaced ently closely together such that a combinatorial (e.g., a synergistic) effect is achieved.
Animal: As used herein, the term “animal” refers to any member of the animal kingdom.
In some embodiments, “animal” refers to humans at any stage of development. In some embodiments, “animal” refers to non-human animals at any stage of development. In certain embodiments, the non-human animal is a mammal (e.g., a rodent, a mouse, a rat, a rabbit, a monkey, a dog, a cat, a sheep, cattle, a primate, or a pig). In some embodiments, animals include, but are not limited to, mammals, birds, es, ians, fish, and worms. In some embodiments, the animal is a transgenic animal, genetically-engineered animal, or a clone.
Antigens ofinterest or desired antigens: As used herein, the terms “antigens of interest” or “desired antigens” e those proteins and other ecules provided herein that are immunospecifically bound by the antibodies and fragments, mutants, variants, and alterations thereof described herein. Examples of antigens of interest include, but are not d to, insulin, insulin- like growth factor, hGH, tPA, cytokines, such as interleukins (IL), e. g., IL-1, IL-2, IL-3, 11-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-1 1, IL-12, IL-13, IL-l4, IL-15, IL-16, [L-l7, IL-18, interferon (IFN) alpha, IFN beta, IFN gamma, IFN omega or IFN tau, tumor necrosis factor (TNF), such as TNF alpha and TNF beta, TNF gamma, TRAIL; G—CSF, GM-CSF, M-CSF, MCP-l and VEGF.
Approximately: As used herein, the term “approximately” or “about,” as d to one or more values of interest, refers to a value that is similar to a stated reference value. In certain embodiments, the term “approximately” or “about” refers to a range of values that fall within 25%, %, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, or less in either ion (greater than or less than) of the stated reference value unless otherwise stated or otherwise evident from the t (except where such number would exceed 100% of a possible value).
Associated with: As used herein, the terms “associated wit ,” “conjugated,” “linked,” “attached,” and “tethered,” when used with respect to two or more moieties, means that the moieties are physically associated or connected with one another, either directly or Via one or more additional moieties that serves as a linking agent, to form a structure that is sufficiently stable so that the moieties remain physically associated under the conditions in which the structure is used, e.g. physiological conditions. An “association” need not be strictly h direct covalent chemical bonding. It may also suggest ionic or hydrogen bonding or a hybridization based connectivity ently stable such that the “associated” entities remain physically associated.
Biocompatible: As used herein, the term “biocompatible” means ible will] living cells, tissues, organs or s posing little to no risk of injury, toxicity or rejection by the immune system. radable: As used herein, the term “biodegradable” means capable ofbeing broken down into innocuous products by the action of living things.
Biologically active: As used herein, the phrase gically active” refers to a characteristic of any substance that has activity in a biological system and/or organism. For instance, a substance that, when administered to an organism, has a biological effect on that organism, is considered to be ically active. In particular embodiments, a polynucleotide of the present invention may be considered biologically active if even a portion of the polynucleotide is ically active or mimics an activity considered biologically relevant. al terms: The following provides the definition of various chemical terms from “acyl” to “thiol.” The term “acyl,” as used herein, represents a hydrogen or an alkyl group (e.g., a kyl group), as defined herein, that is attached to the parent molecular group through a carbonyl group, as defined herein, and is exemplified by formyl (i.e., a carboxyaldehyde , acetyl, trifluoroacetyl, nyl, butanoyl and the like. Exemplary unsubstituted acyl groups include from 1 to 7, from 1 to 11, or from 1 to 21 carbons. In some embodiments, the alkyl group is further substituted with l, 2, 3, or 4 substituents as described herein.
The term “acylamino,” as used herein, represents an acyl group, as defined herein, attached to the parent molecular group though an amino group, as defined herein (i.e., 7N(RN1)- C(O)-R, where R is H or an optionally tuted CM, C140, or C1_20 alkyl group (e. g., haloalkyl) and RNl is as defined herein). Exemplary tituted acylamino groups e from 1 to 41 carbons (e.g., from 1 to 7, from 1 to 13, from 1 to 21, from 2 to 7, from 2 to 13, from 2 to 21, or from 2 to 41 carbons). In some embodiments, the alkyl group is further substituted with 1, 2, 3, or 4 substituents as described herein, and/or the amino group is iNHz or , wherein RNl is, independently, OH, N02, NHz, NRNZZ, SOZORNZ, SOZRNZ, SORNZ, alkyl, aryl, acyl (e. g., acetyl, trifluoroacetyl, or others described herein), or alkoxycarbonylalkyl, and each RNZ can be H, alkyl, or aryl.
] The term “acylaminoalkyl,” as used herein, represents an acyl group, as defined herein, attached to an amino group that is in turn attached to the parent molecular group though an alkyl group, as defined herein (i.e., ialkyl-N(RN1)-C(O)-R, where R is H or an optionally substituted C14,, C140, or C140 alkyl group (e. g., haloalkyl) and RNl is as defined herein). Exemplary unsubstituted acylamino groups include from 1 to 41 carbons (e.g., from 1 to 7, from 1 to 13, from 1 to 21, from 2 to 7, from 2 to 13, from 2 to 21, or from 2 to 41 carbons). In some embodiments, the alkyl group is further substituted with l, 2, 3, or 4 tuents as described , and/or the amino group is iNHz or iNl—IRNI, wherein RNl is, independently, OH, N02, NHz, NRNZZ, SOZORNZ, SOZRNZ, SORNz, alkyl, aryl, acyl (e.g., acetyl, trifluoroacetyl, or others described herein), or alkoxycarbonylalkyl, and each RN2 can be H, alkyl, or aryl.
The term “acyloxy,” as used herein, represents an acyl group, as defined herein, attached to the parent molecular group though an oxygen atom (i.e., )—R, where R is H or an optionally substituted C143, €1.10, or C140 alkyl group). Exemplary unsubstituted acyloxy groups include from 1 to 21 carbons (e.g., from 1 to 7 or from 1 to 11 carbons). In some embodiments, the alkyl group is further substituted with 1, 2, 3, or 4 substituents as described herein.
The term “acyloxyalkyl,” as used herein, represents an acyl group, as defined herein, attached to an oxygen atom that in turn is attached to the parent lar group though an alkyl group (i.e., —alkyl-O-C(O)-R, Where R is H or an optionally substituted C1.6, C140, or C140 alkyl group). ary unsubstituted acyloxyalkyl groups e from 1 to 21 carbons (e.g., from 1 to 7 or from 1 to 11 carbons). In some embodiments, the alkyl group is, independently, further substituted with 1, 2, 3, or 4 substituents as described herein.
] The term yl,” as used herein, represents an aryl group, as defined herein, attached to the parent molecular group through an ne group, as defined herein. Exemplary unsubstituted alkaryl groups are from 7 to 30 carbons (e. g., from 7 to 16 or from 7 to 20 carbons, such as CH; alk- C640 aryl, CHO alk-C6_10 aryl, or C140 alk-C6_10 aryl). In some embodiments, the alkylene and the aryl each can be further substituted with 1, 2, 3, or 4 substituent groups as defined herein for the respective groups. Other groups preceded by the prefix “alk-” are defined in the same manner, where “alk” refers to a C1_6 alkylene, unless otherwise noted, and the attached chemical structure is as defined herein.
] The term “alkcycloalkyl” represents a cycloalkyl group, as defined , attached to the parent molecular group through an alkylene group, as defined herein (e. g., an alkylene group of from 1 to 4, from 1 to 6, from 1 to 10, or form 1 to 20 carbons). In some ments, the alkylene and the cycloalkyl each can be further substituted with 1, 2, 3, or 4 tuent groups as defined herein for the respective group.
The term “alkenyl,” as used herein, represents monovalent straight or branched chain groups of, unless otherwise specified, from 2 to 20 carbons (e. g., from 2 to 6 or from 2 to 10 carbons) containing one or more carbon-carbon double bonds and is exemplified by ethenyl, l- propenyl, 2-propenyl, 2-methylpropenyl, 1-butenyl, 2-butenyl, and the like. Alkenyls include both cis and trans isomers. Alkenyl groups may be optionally tuted with 1, 2, 3, or 4 substituent groups that are selected, independently, from amino, aryl, lkyl, or heterocyclyl (e.g., aryl), as defined herein, or any of the exemplary alkyl substituent groups described herein.
The term “alkenyloxy” represents a chemical substituent of formula —OR, where R is a C2. alkenyl group (e.g., C24; or C2.1o alkenyl), unless otherwise specified. Exemplary alkenyloxy groups include ethenyloxy, propenyloxy, and the like. In some embodiments, the alkenyl group can be further substituted with 1, 2, 3, or 4 substituent groups as defined herein (e. g., a hydroxy group).
The term “alkheteroary ” refers to a heteroaryl group, as defined herein, attached to the parent molecular group through an alkylene group, as defined herein. Exemplary unsubstituted alkheteroaryl groups are from 2 to 32 carbons (e.g., from 2 to 22, from 2 to 18, from 2 to 1?, from 2 to 16, from 3 to 15, from 2 to 14, from 2 to 13, or from 2 to 12 carbons, such as C1.6 alk-CHZ heteroaryl, C140 alk-Cm heteroaryl, or Cmo alk-CHZ heteroaryl). In some embodiments, the alkylene and the heteroaryl each can be further substituted with 1, 2, 3, or 4 substituent groups as defined herein for the respective group. Alkheteroaryl groups are a subset of alkheterocyclyl groups.
The term terocyclyl” represents a heterocyclyl group, as defined herein, attached to the parent molecular group through an alkylene group, as defined herein. Exemplary unsubstituted alkheterocyclyl groups are from 2 to 32 carbons (e. g., from 2 to 22, from 2 to 18, from 2 to 17, from 2 to 16, from 3 to 15, from 2 to 14, from 2 to 13, or from 2 to 12 carbons, such as C1_6 alk-CHZ heterocyclyl, C140 alk-CHZ heterocyclyl, or C140 alk-CHZ heterocyclyl). In some embodiments, the alkylene and the heterocyclyl each can be further substituted with 1, 2, 3, or 4 substituent groups as defined herein for the respective group.
The term “alkoxy” represents a chemical substituent of formula 70R, where R is a C140 alkyl group (e.g., CM or C140 alkyl), unless otherwise specified. ary alkoxy groups include y, ethoxy, propoxy (e. g., n-propoxy and isopropoxy), t-butoxy, and the like. In some embodiments, the alkyl group can be further substituted with 1, 2, 3, or 4 substituent groups as defined herein (e.g., hydroxy or alkoxy).
The term “alkoxyalkoxy” represents an alkoxy group that is substituted with an alkoxy group. Exemplary unsubstituted alkoxy groups include between 2 to 40 carbons (e.g., from 2 to 12 or from 2 to 20 s, such as C1_6 alkoxy-CM alkoxy, C140 alkoxy-C140 , or C140 alkoxy-C140 alkoxy). In some embodiments, the each alkoxy group can be further substituted with l, 2, 3, or 4 tuent groups as defined herein.
] The term “alkoxyalkyl” represents an alkyl group that is substituted with an alkoxy group.
Exemplary unsubstituted alkoxyalkyl groups include between 2 to 40 carbons (e.g., from 2 to 12 or from 2 to 20 carbons, such as CM alkoxy-CH; alkyl, C140 alkoxy-C140 alkyl, or C140 alkoxy-C140 alkyl). In some embodiments, the alkyl and the alkoxy each can be r substituted with l, 2, 3, or 4 substituent groups as defined herein for the tive group.
] The term ycarbonyl,” as used herein, represents an alkoxy, as defined herein, attached to the parent lar group through a carbonyl atom (e. g., -C(O)-OR, where R is H or an optionally substituted C143, C140, or C140 alkyl group). Exemplary unsubstituted alkoxycarbonyl include from 1 to 21 carbons (e.g., from 1 to 11 or from 1 to 7 carbons). In some embodiments, the alkoxy group is further substituted with 1, 2, 3, or 4 substituents as described herein.
The term “alkoxycarbonylacyl,” as used herein, represents an acyl group, as defined herein, that is substituted with an alkoxycarbonyl group, as defined herein (e.g., -C(O) -alkyl-C(O)- OR, where R is an ally substituted C1_6, CHO, or C140 alkyl group). Exemplary unsubstituted alkoxycarbonylacyl include from 3 to 41 carbons (e.g., from 3 to 10, from 3 to 13, from 3 to 17, from 3 to 21, or from 3 to 31 carbons, such as C1_6 alkoxycarbonyl-CH, acyl, C140 alkoxycarbonyl- C140 acyl, or Cmo carbonyl-Cmo acyl). In some embodiments, each alkoxy and alkyl group is further independently substituted with 1, 2, 3, or 4 substituents, as described herein (e.g., a hydroxy group) for each group.
The term “alkoxycarbonylalkoxy,” as used herein, represents an alkoxy group, as defined herein, that is substituted with an alkoxycarbonyl group, as defined herein (e.g., -O-alkyl-C(O)-OR, whwRfimmfimemwammdQfiCHmmCkaflgwmdkmmMymwmmmw alkoxycarbonylalkoxy include from 3 to 41 carbons (e.g., from 3 to 10, from 3 to 13, from 3 to 17, from 3 to 21, or from 3 to 31 carbons, such as C1_6 alkoxycarbonyl-C1_6 , C140 alkoxycarbonyl-CHO alkoxy, or C140 alkoxycarbonyl-Cmo alkoxy). In some embodiments, each alkoxy group is r independently substituted with 1, 2, 3, or 4 substituents, as described herein (e.g., a hydroxy group).
] The term “alkoxycarbonylalkyl,” as used herein, represents an alkyl group, as defined herein, that is substituted with an alkoxycarbonyl group, as defined herein (e.g., -C(O)—OR, whmRqummmwwmmemflmmmQfififlgWMJkWmemwmmw alkoxycarbonylalkyl include from 3 to 41 carbons (e.g., from 3 to 10, from 3 to 13, from 3 to 17, from 3 to 21, or from 3 to 31 carbons, such as C1_6 carbonyl-C1_6 alkyl, C140 alkoxycarbonyl- C140 alkyl, or C140 alkoxycarbonyl—C1_20 alkyl). In some embodiments, each alkyl and alkoxy group is further independently substituted with 1, 2, 3, or 4 substituents as described herein (eg, a hydroxy goup) The term “alkoxycarbonylakenyl,” as used herein, represents an alkenyl group, as defined herein, that is substituted with an alkoxycarbonyl group, as defined herein (e.g., -alkenyl-C(O)-OR, where R is an ally substituted C140, C140, or C1.6 alkyl group). Exemplary unsubstituted alkoxycarbonylalkenyl include from 4 to 41 carbons (e.g., from 4 to 10, from 4 to 13, from 4 to 17, from 4 to 21, or from 4 to 31 carbons, such as CH; alkoxycarbonyl-C2_6 alkenyl, C140 alkoxycarbonyl-C2.10 alkenyl, or C140 alkoxycarbonyl-CMO alkenyl). In some embodiments, each alkyl, alkenyl, and alkoxy group is r independently substituted with l, 2, 3, or 4 substituents as described herein (e.g., a hydroxy group).
The term “alkoxycarbonylalkynyl,” as used herein, represents an alkynyl group, as defined herein, that is substituted with an alkoxycarbonyl group, as defined herein (e.g., -alkynyl-C(O)-OR, whwRSmmmMMWmmmmemmQmmflhwkflgwmfifimmMymwmmmw alkoxycarbonylalkynyl include from 4 to 41 carbons (e. g., from 4 to 10, from 4 to 13, from 4 to 17, from 4 to 21, or from 4 to 31 carbons, such as C1_6 alkoxycarbonyl-CM l, C140 alkoxycarbonyl-C2_10 alkynyl, or C1_20 alkoxycarbonyl—CMO alkynyl). In some ments, each alkyl, alkynyl, and alkoxy group is further independently substituted with 1, 2, 3, or 4 substituents as described herein (e.g., a hydroxy group).
The term “alkyl,” as used herein, is ive of both straight chain and branched chain saturated groups from 1 to 20 carbons (e.g., from 1 to 10 or from 1 to 6), unless otherwise specified.
Alkyl groups are exemplified by , ethyl, n- and iso-propyl, n-, sec-, iso- and tert-butyl, neopentyl, and the like, and may be optionally substituted with one, two, three, or, in the case of alkyl groups of two carbons or more, four substituents ndently selected from the group consisting of: (l) C1_6 alkoxy; (2) C1_6 alkylsulfinyl; (3) amino, as defined herein (e. g., unsubstituted amino (i.e., -NH2) or a substituted amino (i.e., -N(RN1)2, where RNl is as defined for amino); (4) C640 H; alkoxy; (5) azido; (6) halo; (7) (C23 heterocyclyl)oxy; (8) hydroxy, optionally substituted with an ecting group; (9) nitro; (10) oxo (e.g., carboxyaldehyde or acyl); (l 1) C14 spirocyclyl; (l2) thioalkoxy; (l3) thiol; (l4) -C02RA’, optionally substituted with an ecting group and where RA, is selected from the group consisting of (a) C140 alkyl (e.g., C1_6 alkyl), (b) C240 alkenyl (erg, CM alkenyl), (c) C640 aryl, (d) hydrogen, (e) C1_6 alk-C6_1o aryl, (f) amino-C140 alkyl, (g) polyethylene glycol of -(CH2)52(OCH2CH2)51(CH2)530R’, wherein s1 is an r from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R’ is H or C1_20 alkyl, and (h) amino- polyethylene glycol of -NRN1(CH2)52(CH2CH20)51(CH2)S3NRN1, wherein S] is an integer from 1 to (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 4me0w6melwkfimnhoamfimnhomxmdwmeb, independently, hydrogen or optionally substituted CH; alkyl; (15) -C(O)N'RB’RC‘, where each of RB, and RC5 is, independently, selected from the group consisting of (a) hydrogen, (b) C1.6 alkyl, (0) C640 aryl, and (d) CM alk-C5_10 aryl; (16) -SOZRD', where RD: is selected from the group ting of (a) CM. alkyl, (b) C540 aryl, (c) CH; alk-C5_10 aryl, and (d) hydroxy; (17) -SO¢NRESRF‘, where each of RE’ and RF is, independently, selected from the group consisting of (a) hydrogen, (b) CM alkyl, (0) C640 aryl and (d) CH; _1o aryl; (18) G’, where RG’ is selected from the group consisting of (a) C140 alkyl (e.g., C1_5 alkyl), (b) C240 alkenyl (e.g, C24, alkenyl), (c) C640 aryl, ((1) hydrogen, (e) CM alk-C6_10 aryl, (f) amino-C1_zo alkyl, (g) polyethylene glycol of - (CHz)sz(OCH2CH2)51(CH2)530R’, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of $2 and 53, independently, is an integer from 0 to 10 (e. g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R’ is H or C140 alkyl, and (h) amino-polyethylene glycol of - H2)52(CH2CH20)S1(CH2)53NRN1, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and 53, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each RNl is, independently, en or optionally substituted C1_6 alkyl; (19) -NRH’C(O)RI’, wherein RH, is selected from the group consisting of (al) hydrogen and (b1) C1_6 alkyl, and R1, is selected from the group consisting of (a2) C1_20 alkyl (e.g., C1_6 alkyl), (b2) C240 l (e. g., C2_6 alkenyl), (c2) C640 aryl, (d2) hydrogen, (e2) C1_6 alk—C6_lo aryl, (f2) amino-C140 alkyl, (g2) hylene glycol of -(CH2),2(OCH2CH2),1(CH2),30R’, wherein s] is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R’ is H or C140 alkyl, and (h2) polyethylene glycol of -NRN1(CH2)52(CH2CH20)51(CH2)S3NRNI, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e. g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each RNl is, independently, hydrogen or optionally substituted CM alkyl; (20) - NRJ’C(O)ORK‘, wherein Ry is selected from the group consisting of (a1) hydrogen and (b1) CH; alkyl, and RK' is selected from the group consisting of (a2) C140 alkyl (e.g., C1_6 alkyl), (b2) C240 alkenyl (e.g., CM alkenyl), (c2) C640 aryl, (d2) en, (e2) C14, alk-C6_10 aryl, (f2) amino-C140 alkyl, (g2) polyethylene glycol of -(CH2)52(OCH2CH2)51(CH2)530R’, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and 33, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R’ is H or €1.20 alkyl, and (h2) amino-polyethylene glycol of -NRN1(CH2)52(CH2CHZO)51(CH2)53N'RN1, wherein s] is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of 52 and 53, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each RNl is, independently, hydrogen or ally substituted C1_6 alkyl; and (21) amidine. In some embodiments, each of these groups can be further substituted as described herein. For e, the alkylene group of a Cl-alkaryl can be r tuted with an oxo group to afford the respective aryloyl substituent.
The term “alkylene” and the prefix “alk-,” as used herein, represent a saturated divalent hydrocarbon group derived from a straight or branched chain saturated hydrocarbon by the removal of two hydrogen atoms, and is exemplified by ene, ethylene, isopropylene, and the like. The term “CK.y alkylene” and the prefix “CK.y alk-” represent alkylene groups having between x and y s. Exemplary values for x are 1, 2, 3, 4, 5, and 6, and exemplary values for y are 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, or 20 (e.g., C1_6, C140, C240, CM, C240, or C240 alkylene). In some embodiments, the alkylene can be further substituted with 1, 2, 3, or 4 substituent groups as defined herein for an alkyl group.
] The term “alkylsulfinyl,” as used herein, ents an alkyl group attached to the parent molecular group through an -S(O)- group. Exemplary unsubstituted alkylsulfinyl groups are from 1 to 6, from 1 to 10, or from 1 to 20 carbons. In some embodiments, the alkyl group can be further substituted with 1, 2, 3, or 4 tuent groups as defined herein.
The term “alkylsulfmylalkyl,” as used , represents an alkyl group, as defined herein, substituted by an alkylsulfinyl group. Exemplary unsubstituted alkylsulfinylalkyl groups are from 2 to 12, from 2 to 20, or from 2 to 40 carbons. In some embodiments, each alkyl group can be further tuted with 1, 2, 3, or 4 substituent groups as defined herein.
The term “alkynyl,” as used herein, represents monovalent straight or branched chain groups from 2 to 20 carbon atoms (e. g., from 2 to 4, from 2 to 6, or from 2 to 10 carbons) containing a carbon-carbon triple bond and is exemplified by ethynyl, l-propynyl, and the like. Alkynyl groups may be optionally substituted with 1, 2, 3, or 4 substituent groups that are selected, independently, from aryl, cycloalkyl, or heterocyclyl (e. g., heteroaryl), as defined herein, or any of the exemplary alkyl substituent groups described herein.
The term “alkynyloxy” represents a al substituent of formula —OR, where R is a C2. alkynyl group (e.g., C24; or C2.1o alkynyl), unless otherwise specified. Exemplary alkynyloxy groups include ethynyloxy, propynyloxy, and the like. In some embodiments, the alkynyl group can be further substituted with 1, 2, 3, or 4 substituent groups as defined herein (e. g., a hydroxy group).
The term “amidine,” as used herein, represents a —C(=NH)NH2 group, The term “amino,” as used , represents —N(RN1)2, wherein each RNl is, ndently, H, OH, N02, N(RN2)2, SOZORNZ, , SORNZ, an N-protecting group, alkyl, l, alkynyl, alkoxy, aryl, alkaryl, cycloalkyl, alkcycloalkyl, carboxyalkyl (e.g., optionally substituted with an 0-protecting group, such as optionally substituted koxycarbonyl groups or any described herein), sulfoalkyl, acyl (e.g., acetyl, roacetyl, or others described herein), alkoxycarbonylalkyl (e. g., optionally substituted with an 0-protecting group, such as optionally substituted arylalkoxycarbonyl groups or any described herein), heterocyclyl (e. g., heteroaryl), or alkheterocyclyl (e. g., eroaryl), wherein each of these recited RNl groups can be optionally substituted, as defined herein for each group; or two RN1 combine to form a heterocyclyl or an N- protecting group, and n each RNZ is, independently, H, alkyl, or aryl. The amino groups of the invention can be an unsubstituted amino (i.e., iNHz) or a substituted amino (i.e., 7N(RN1)2). In a preferred embodiment, amino is iNHz or iNHRNl, wherein RNl is, independently, OH, N02, NHZ, NRNZZ, SOZORNZ, SOZRNZ, SORNZ, alkyl, carboxyalkyl, sulfoalkyl, acyl (e. g., acetyl, trifluoroacetyl, or others described herein), alkoxycarbonylalkyl (e.g., t-butoxycarbonylalkyl) or aryl, and each RN2 can be H, Cmo alkyl (e.g., C1_6 alkyl), or C640 aryl.
The term “amino acid,” as described herein, refers to a molecule having a side chain, an amino group, and an acid group (e. g., a carboxy group of£02H or a sulfo group of 7S03H), wherein the amino acid is attached to the parent lar group by the side chain, amino group, or acid group (e.g., the side chain). In some embodiments, the amino acid is attached to the parent molecular group by a carbonyl group, where the side chain or amino group is attached to the carbonyl group. Exemplary side chains include an optionally substituted alkyl, aryl, heterocyclyl, alkaryl, alkheterocyclyl, aminoalkyl, oylalkyl, and carboxyalkyl. Exemplary amino acids include alanine, arginine, gine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, histidine, hydroxynorvaline, isoleucine, leucine, lysine, methionine, norvaline, ine, phenylalanine, proline, pyrrolysine, selenocysteine, serine, taurine, threonine, tryptophan, tyrosine, and . Amino acid groups may be optionally substituted with one, two, three, or, in the case of amino acid groups of two carbons or more, four substituents independently selected from the group consisting of: (1) CM ; (2) C1_6 alkylsulfinyl; (3) amino, as defined herein (e. g., unsubstituted amino (i.e., -NH2) or a substituted amino (i.e., -N(RN1)2, where RNI is as defined for amino); (4) C640 aryl-Cl.6 alkoxy; (5) azido; (6) halo; (7) (C23 heterocyclyl)oxy; (8) hydroxy; (9) nitro; (10) 0x0 (e.g., yaldehyde or acyl); (11) C1.7 spirocyclyl; (12) thioalkoxy; (l3) thiol; (14) -C02RA’, where RA, is selected from the group consisting of (a) C140 alkyl (e.g., C1_6 alkyl), (b) C240 l (e.g., CM alkenyl), (c) C640 aryl, ((1) hydrogen, (e) C1.6 alk-C5.1o aryl, (f) amino-C140 alkyl, (g) polyethylene glycol of -(CH2)52(OCH2CH2)51(CH2)53OR’, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an r from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R’ is H or C1_20 alkyl, and (h) amino- polyethylene glycol of -NRN1(CH2)52(CH2CH20)s1(CH2),3NRN1, n S] is an integer from 1 to (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each RNl is, independently, hydrogen or optionally substituted CM alkyl; (15) -C(O)NRB’RCY, where each of RBY and RC) is, independently, selected from the group consisting of (a) hydrogen, (b) CH; alkyl, (c) C640 aryl, and (d) CM alk-C5_10 aryl; (16) -SOZRD’, where RD, is selected from the group consisting of (a) CM alkyl, (b) C540 aryl, (c) C1_6 alk-C6_10 aryl, and (d) hydroxy; (17) -SOZNRE’RF’, where each of RE) and RF, is, independently, selected from the group consisting of (a) hydrogen, (b) C1_6 alkyl, (c) C640 aryl and (d) CM alk-C6_10 aryl; (18) -C(O)RG), where RGY is selected from the group consisting of (a) C140 alkyl (e.g., C1_6 alkyl), (b) C240 alkenyl (e. g., CM alkenyl), (c) C640 aryl, (d) hydrogen, (e) C1_6 alk-CHO aryl, (t) amino-C140 alkyl, (g) polyethylene glycol of - (CH2)52(OCH2CH2)51(CH2)530R’, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and 53, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R’ is H or C140 alkyl, and (h) amino-polyethylene glycol of - NRN1(CH2)52(CH2CH20)S1(CH2)53NRN1, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of 52 and 53, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each RNl is, ndently, hydrogen or optionally substituted C1_6 alkyl; (19) -NRH’C(O)RI’, wherein RH, is selected from the group ting of (a1) hydrogen and (bl) CH; alkyl, and R1, is selected from the group consisting of (a2) C140 alkyl (e.g., CH, alkyl), (b2) C240 l (e. g., CM alkenyl), (c2) C640 aryl, (d2) hydrogen, (e2) C1_6 alk—C6_lo aryl, (f2) amino-C140 alkyl, (g2) hylene glycol of -(CH2)52(OCH2CH2)51(CH2)530R’, wherein s] is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R’ is H or C140 alkyl, and (112) polyethylene glycol of -N'RN1(CH2)52(CH2CH20)81(CH2)53NRN', wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e. g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each RNl is, independently, hydrogen or optionally substituted C1_6 alkyl; (20) - NRJ’C(O)ORKi, wherein Ry is ed from the group consisting of (a1) hydrogen and (bl) CH. alkyl, and R“ is selected from the group consisting of (a2) C140 alkyl (e.g., C1.6 alkyl), (b2) C240 alkenyl (e.g., C245 alkenyl), (02) C640 aryl, (d2) hydrogen, (e2) C1_6 _10 aryl, (f2) amino-C140 alkyl, (g2) polyethylene glycol of -(CH2)sz(OCHzCH2)31(CH2)530R’, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R’ is H or C140 alkyl, and (h2) amino-polyethylene glycol of -NRN1(CH2)52(CH2CHZO)51(CH2)53NRN1, wherein s] is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each RNl is, independently, hydrogen or optionally substituted C1_6 alkyl; and (21) amidine. In some embodiments, each of these groups can be further substituted as described herein.
The term “aminoalkoxy,” as used herein, represents an alkoxy group, as defined herein, substituted by an amino group, as defined herein. The alkyl and amino each can be further substituted with 1, 2, 3, or 4 substituent groups as described herein for the respective group (e.g., COZRAY, where RA, is ed from the group consisting of (a) C1_6 alkyl, (b) C640 aryl, (c) hydrogen, and (d) C1_6 alk-C6_1o aryl, e.g., carboxy).
The term “aminoalkyl,” as used herein, represents an alkyl group, as defined herein, substituted by an amino group, as defined . The alkyl and amino each can be further substituted with l, 2, 3, or 4 tuent groups as described herein for the respective group (e.g., CO2RA’, where RA, is selected from the group ting of (a) C1_6 alkyl, (b) C640 aryl, (c) hydrogen, and (d) C1_6 _10 aryl, e.g., carboxy, and/or an N—protecting group).
The term “aminoalkenyl,” as used herein, represents an alkenyl group, as defined herein, substituted by an amino group, as defined herein. The l and amino each can be filrther tuted with l, 2, 3, or 4 substituent groups as described herein for the respective group (e.g., COzRA’, where RA, is selected from the group consisting of (a) C1_6 alkyl, (b) C640 aryl, (0) hydrogen, and (d) CH; alk-C5_10 aryl, e.g., y, and/or an N—protecting group).
The term “aminoalkynyl,” as used , represents an alkynyl group, as defined herein, substituted by an amino group, as defined herein. The alkynyl and amino each can be further substituted with l, 2, 3, or 4 substituent groups as described herein for the respective group (e.g., COZRA’, where RA, is selected from the group consisting of (a) CM alkyl, (b) C640 aryl, (c) hydrogen, and (d) CH; .1o aryl, e.g., carboxy, and/or an N-protecting group).
The term “aryl,” as used herein, represents a mono-, bicyclic, or multicyclic carbocyclic ring system having one or two aromatic rings and is exemplified by , naphthyl, 1,2- dihydronaphthyl, 1,2,3,4-tetrahydronaphthyl, anthracenyl, phenanthrenyl, fluorenyl, indanyl, indenyl, and the like, and may be optionally substituted with 1, 2, 3, 4, or 5 substituents independently ed from the group consisting of: (1) C1_7 acyl (e.g., carboxyaldehyde); (2) C140 alkyl (e.g., C1_6 alkyl, C14, alkoxy-C1_5 alkyl, C1_5 alkylsulfmyl—CH; alkyl, CM alkyl, azido-C1_ 6 alkyl, (carboxyaldehyde)—C1_5 alkyl, halo-CM alkyl (e. g., perfluoroalkyl), hydroxy-C1_6 alkyl, nitro- C1_6 alkyl, or C1_5thioalkoxy-C1_6 alkyl); (3) C140 alkoxy (e.g., C1_6 alkoxy, such as perfluoroalkoxy); (4) C14, alkylsulfinyl; (5) C640 aryl; (6) amino; (7) C1_6 alk-C6_1o aryl; (8) azido; (9) C34; lkyl; (10) C1_6 alk-C3_8 cycloalkyl; (11) halo; (12) C142 heterocyclyl (e.g., C142 heteroaryl); (13) (C142 heterocyclyl)oxy; (14) hydroxy; (15) nitro; (16) C140 thioalkoxy (e.g., CM thioalkoxy); (17) 7(CH2)qCOZRAy, where q is an integer from zero to four, and RA) is selected from the group ting of (a) C1_6 alkyl, (b) C640 aryl, (c) hydrogen, and (d) C1_6 alk-CHO aryl; (18) 7 CONRB)RC’, where q is an integer from zero to four and where RBY and RC, are independently selected from the group consisting of (a) hydrogen, (b) C1_6 alkyl, (c) C640 aryl, and (d) CM alk—CHO aryl; (l9) 7(CH2)qSOZRD), where q is an integer from zero to four and where RD) is selected from the group consisting of (a) alkyl, (b) C640 aryl, and (c) alk-C6_1o aryl; (20) 7(CH2)qSOZNRE)RF,, where q is an integer from zero to four and where each of RE, and RF, is, independently, selected from the group consisting of (a) hydrogen, (b) C1_6 alkyl, (c) C640 aryl, and (d) C1_6 alk-C6_10 aryl; (21) thiol; (22) C640 aryloxy; (23) C3_g cycloalkoxy; (24) C640 aryl-C1_6 alkoxy; (25) C1_6 alk-CHZ heterocyclyl (e.g., CM alk-CHZ heteroaryl); (26) C240 alkenyl; and (27) C240 l. In some embodiments, each of these groups can be further substituted as described herein. For example, the alkylene group of a Cl-alkaryl or a C1-alkheterocyclyl can be further substituted with an oxo group to afford the respective aryloyl and (heterocyclyl)oyl sub stituent group.
The term “arylalkoxy,” as used herein, represents an alkaryl group, as defined , attached to the parent lar group through an oxygen atom. Exemplary unsubstituted arylalkoxy groups include from 7 to 30 carbons (e.g., from 7 to 16 or from 7 to 20 s, such as C640 aryl-CH, alkoxy, C540 aryl-CHo alkoxy, or C540 aryl-Cmo alkoxy). In some embodiments, the arylalkoxy group can be substituted With 1, 2, 3, or 4 substituents as defined herein The term “arylalkoxycarbonyl,” as used herein, represents an arylalkoxy group, as defined herein, attached to the parent molecular group h a yl (e. g., -C(O)-O-alkyl-aryl).
Exemplary unsubstituted arylalkoxy groups include from 8 to 31 carbons (e. g., from 8 to l? or from 8 to 21 carbons, such as C540 aryl-CH; alkoxy-carbonyl, C640 aryl-CHO alkoxy-carbonyl, or C640 aryl-C140 -carbonyl). In some embodiments, the arylalkoxycarbonyl group can be substituted with l, 2, 3, or 4 substituents as defined herein.
] The term “aryloxy” represents a chemical substituent of formula —OR’, where R’ is an aryl group of 6 to 18 carbons, unless otherwise specified. In some embodiments, the aryl group can be substituted with 1, 2, 3, or 4 substituents as defined herein.
The term “aryloyl,” as used herein, represents an aryl group, as defined herein, that is attached to the parent molecular group through a carbonyl group. Exemplary unsubstituted aryloyl groups are of 7 to 11 carbons. In some embodiments, the aryl group can be substituted with l, 2, 3, or 4 substituents as defined herein.
The term “azido” represents an —N3 group, which can also be represented as —N:N:N.
The term “bicyclic,” as used herein, refer to a structure having two rings, which may be aromatic or non-aromatic. Bicyclic structures include spirocyclyl groups, as defined herein, and two rings that share one or more bridges, where such bridges can include one atom or a chain including two, three, or more atoms. ary bicyclic groups include a bicyclic yclyl group, where the first and second rings are carbocyclyl groups, as defined herein; a bicyclic aryl groups, where the first and second rings are aryl groups, as defined herein; bicyclic heterocyclyl groups, where the first ring is a heterocyclyl group and the second ring is a carbocyclyl (e.g., aryl) or heterocyclyl (e.g., heteroaryl) group; and bicyclic heteroaryl groups, where the first ring is a heteroaryl group and the second ring is a yclyl (e. g., aryl) or heterocyclyl (e. g., heteroaryl) group. In some embodiments, the bicyclic group can be substituted with l, 2, 3, or 4 tuents as defined herein for cycloalkyl, heterocyclyl, and aryl groups.
] The term “boranyl,” as used , represents 7B(RB1)3, where each RBl is, independently, selected from the group consisting of H and optionally substituted alkyl. In some ments, the boranyl group can be substituted with 1, 2, 3, or 4 substituents as defined herein for alkyl.
The terms “carbocyclic” and “carbocyclyl,” as used herein, refer to an optionally substituted C342 monocyclic, bicyclic, or tricyclic structure in which the rings, which may be aromatic or non-aromatic, are formed by carbon atoms. yclic structures include cycloalkyl, cycloalkenyl, and aryl groups.
The term “carbamoyl,” as used herein, represents —C(O)-N(RN1)2, where the meaning of each RNl is found in the definition of “amino” provided herein.
The term “carbamoylalkyl,” as used herein, represents an alkyl group, as defined herein, substituted by a carbamoyl group, as defined herein. The alkyl group can be further substituted with l, 2, 3, or 4 substituent groups as described herein.
The term “carbamyl,” as used herein, refers to a carbamate group having the ure (:O)OR or -OC(:O)N(RN1)2, where the meaning of each RNl is found in the definition of “amino” provided herein, and R is alkyl, cycloalkyl , alkcycloalkyl, aryl, alkaryl, heterocyclyl (e.g., heteroaryl), or alkheterocyclyl (e. g., alkheteroaryl), as defined herein.
] The term “carbonyl,” as used herein, represents a C(O) group, which can also be represented as C:O.
The term “carboxyaldehyde” represents an acyl group having the ure 7CHO.
The term “carboxy,” as used herein, means 7COZH.
The term “carboxyalkoxy,” as used herein, represents an alkoxy group, as defined herein, tuted by a carboxy group, as defined herein. The alkoxy group can be further substituted with l, 2, 3, or 4 substituent groups as described herein for the alkyl group, and the carboxy group can be optionally substituted with one or more O-protecting groups.
The term “carboxyalkyl,” as used herein, represents an alkyl group, as defined herein, substituted by a carboxy group, as defined . The alkyl group can be r substituted with l, 2, 3, or 4 substituent groups as described herein, and the carboxy group can be optionally substituted with one or more O-protecting groups.
The term “carboxyaminoalkyl,” as used herein, represents an lkyl group, as defined herein, substituted by a y, as defined herein. The carboxy, alkyl, and amino each can be further substituted with l, 2, 3, or 4 substituent groups as described herein for the respective group (e.g., COzRAi, where RAV is selected from the group consisting of (a) C1_6 alkyl, (b) C640 aryl, (0) hydrogen, and (d) CH; _10 aryl, e.g., carboxy, and/or an N—protecting group, and/or an 0- protecting group).
The term “cyano,” as used herein, represents an —CN group.
The term “cycloalkoxy” represents a chemical tuent of formula —OR, where R is a C34; lkyl group, as defined herein, unless otherwise specified. The cycloalkyl group can be further tuted with 1, 2, 3, or 4 substituent groups as described herein. ary unsubstituted cycloalkoxy groups are from 3 to 8 carbons. In some ment, the cycloalkyl group can be further substituted with 1, 2, 3, or 4 substituent groups as described herein.
The term “cycloalkyl,” as used herein represents a monovalent saturated or unsaturated non-aromatic cyclic hydrocarbon group from three to eight carbons, unless otherwise specified, and is exemplified by cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, bicycle heptyl, and the like. When the cycloalkyl group includes one carbon-carbon double bond, the cycloalkyl group can be referred to as a “cycloalkenyl” group. Exemplary cycloalkenyl groups include cyclopentenyl, cyclohexenyl, and the like. The cycloalkyl groups of this invention can be optionally substituted with: (1) CH acyl (e.g., carboxyaldehyde); (2) C140 alkyl (e.g., C1_6 alkyl, CH, alkoxy-CM alkyl, C1. 6 alkylsulfinyl-C1_5 alkyl, amino-CH; alkyl, azido-C1_6 alkyl, (carboxyaldehyde)-C1_6 alkyl, halo-CM alkyl (e.g., perfluoroalkyl), hydroxy-C1_6 alkyl, C1_6 alkyl, or C1_6 thioalkoxy-C1_(, alkyl); (3) C1. alkoxy (e.g., CM , such as roalkoxy); (4) C1_6 alkylsulfmyl; (5) C640 aryl; (6) amino; (7) C1_6 alk-C5_10 aryl; (8) azido; (9) C34; cycloalkyl; (10) C1_6 alk—C3_8 cycloalkyl; (11) halo; (12) C1. 12 heterocyclyl (e. g., C142 heteroaryl); (13) (C142 heterocyclyl)oxy; (14) hydroxy; (15) nitro; (16) C1. thioalkoxy (e. g., C1_6 thioalkoxy); (17) 7(CH2)qCOZRAY, where q is an integer from zero to four, and RA, is selected from the group consisting of (a) C1_6 alkyl, (b) C640 aryl, (c) hydrogen, and ((1) CM _1o aryl; (18) {CH2)qCONRBYRC,, where q is an integer from zero to four and where RB, and RC, are independently ed from the group consisting of (a) hydrogen, (b) C640 alkyl, (0) C6. aryl, and (d) C14, _1o aryl; (19) 7(CH2)qS02RDY, where q is an integer from zero to four and where RD, is selected from the group consisting of (a) C640 alkyl, (b) C640 aryl, and (c) CH; alk-C6_w aryl; (20) 7(CH2)qS02NREVRF), where q is an integer from zero to four and where each ofRE, and RF, is, independently, selected from the group consisting of (a) hydrogen, (b) C640 alkyl, (c) CH0 aryl, and (d) C145 alk-C6_10 aryl; (21) thiol; (22) C640 aryloxy; (23) C3_8 cycloalkoxy; (24) C640 aryI-Cl_6 alkoxy; (25) Cm alk-CHZ heterocyclyl (e.g., CM alk-CHZ heteroaryl); (26) oxo; (27) C240 alkenyl; and (28) C240 alkynyl. In some embodiments, each of these groups can be further substituted as described herein. For example, the ne group of a Cl-alkaryl or a Cl-alkheterocyclyl can be further substituted with an oxo group to afford the respective aryloyl and (heterocyclyl)oyl substituent group.
The term “diastereomer,” as used herein means stereoisomers that are not mirror images of one another and are non-superimposable on one another.
The term “effective amount” of an agent, as used herein, is that amount sufficient to effect beneficial or desired results, for example, clinical results, and, as such, an “effective amount” depends upon the context in which it is being applied. For example, in the context of administering an agent that treats cancer, an ive amount of an agent is, for example, an amount sufficient to achieve treatment, as defined herein, of cancer, as compared to the response obtained without administration of the agent.
The term “enantiomer,” as used herein, means each individual optically active form of a compound of the ion, having an optical purity or omeric excess (as determined by methods standard in the art) of at least 80% (i.e., at least 90% of one enantiomer and at most 10% of the other enantiomer), preferably at least 90% and more preferably at least 98%.
The term “halo,” as used herein, represents a halogen selected from bromine, chlorine, , or fluorine.
The term “haloalkoxy,” as used herein, represents an alkoxy group, as defined herein, substituted by a halogen group (i.e., F, Cl, Br, or I). A haloalkoxy may be tuted with one, two, three, or, in the case of alkyl groups of two carbons or more, four halogens. Haloalkoxy groups include perfluoroalkoxys (e. g., -OCF3), -OCHF2, -OCH2F, -OCC13, -OCH2CH2Br, - OCHZCH(CH2CH2Br)CH3, and H3. In some embodiments, the haloalkoxy group can be further tuted with l, 2, 3, or 4 tuent groups as bed herein for alkyl .
The term “haloalkyl,” as used herein, represents an alkyl group, as defined herein, substituted by a halogen group (i.e., F, Cl, Br, or I). A haloalkyl may be substituted with one, two, three, or, in the case of alkyl groups of two carbons or more, four halogens. Haloalkyl groups include perfluoroalkyls (e.g., -CF3), -CHF2, -CH2F, -CC13, -CH2CH2Br, -CH2CH(CH2CH2Br)CH3, and -CHICH3. In some embodiments, the haloalkyl group can be further substituted with l, 2, 3, or 4 substituent groups as bed herein for alkyl groups.
The term “heteroalkylene,” as used herein, refers to an alkylene group, as d herein, in which one or two of the constituent carbon atoms have each been replaced by nitrogen, oxygen, or sulfur. In some embodiments, the heteroalkylene group can be further tuted with l, 2, 3, or 4 substituent groups as described herein for alkylene groups.
The term oaryl,” as used , represents that subset of heterocyclyls, as defined herein, which are aromatic: i.e., they contain 4n+2 pi electrons within the mono- or multicyclic ring system. ary unsubstituted heteroaryl groups are of 1 to 12 (e.g., 1 to 11, l to 10, l to 9, 2 to 2lem2w9MMMm.hmmemmwmmmflWMWmaflfiwmmmquhLL 3, or 4 substituents groups as defined for a heterocyclyl group.
The term “heterocyclyl,” as used herein represents a 5-, 6- or 7-membered ring, unless otherwise specified, ning one, two, three, or four heteroatoms independently selected from the group consisting of nitrogen, oxygen, and sulfur. The 5-membered ring has zero to two double bonds, and the 6- and 7-membered rings have zero to three double bonds. Exemplary tituted heterocyclyl groups are of] to 12 (e.g., 1 to 11, l to 10, l to 9, 2 to 12, 2 to 11, 2 to 10, or 2 to 9) carbons. The term “heterocyclyl” also represents a cyclic compound having a bridged multicyclic structure in which one or more carbons and/or heteroatoms bridges two non-adjacent members of a monocyclic ring, e.g., a quinuclidinyl group. The term “heterocyclyl” es bicyclic, tricyclic, and tetracyclic groups in which any of the above heterocyclic rings is fused to one, two, or three carbocyclic rings, e.g., an aryl ring, a cyclohexane ring, a cyclohexene ring, a cyclopentane ring, a cyclopentene ring, or another monocyclic heterocyclic ring, such as indolyl, quinolyl, isoquinolyl, tetrahydroquinolyl, benzofuryl, benzothienyl and the like. Examples of fused hdmnqubnwm&Hnmmwsmmlifififlflwmmdwmnmddfime Hdflmwdmsmdmk pyrrolyl, pyrrolinyl, idinyl, pyrazolyl, pyrazolinyl, pyrazolidinyl, imidazolyl, imidazolinyl, imidazolidinyl, pyridyl, piperidinyl, homopiperidinyl, pyrazinyl, piperazinyl, pyrimidjnyl, pyridazinyl, oxazolyl, oxazolidinyl, isoxazolyl, isoxazolidiniyl, morpholinyl, thiomorpholinyl, thiazolyl, thiazolidinyl, isothiazolyl, azolidinyl, indolyl, indazolyl, quinolyl, isoquinolyl, quinoxalinyl, oquinoxalinyl, quinazolinyl, cinnolinyl, phthalazinyl, benzimidazolyl, benzothiazolyl, benzoxazolyl, benzothiadiazolyl, furyl, thienyl, thiazolidinyl, isothiazolyl, triazolyl, tetrazolyl, oxadiazolyl (e. g., 1,2,3-oxadiazolyl), purinyl, thiadjazolyl (e.g., 1,2,3-thiadiazolyl), tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, dihydroindolyl, dihydroquinolyl, tetrahydroquinolyl, tetrahydroisoquinolyl, dihydroisoquinolyl, l, dihydropyranyl, dithiazolyl, benzofuranyl, isobenzofuranyl, benzothienyl, and the like, including dihydro and tetrahydro forms thereof, where one or more double bonds are reduced and ed with ens. Still other exemplary heterocyclyls include: 2,3 ,4,5-tetrahydrooxo-oxazolyl; 2,3-dihydrooxo-lH-imidazolyl; 2,3 ,4,5-tetrahydrooxo-lH—pyrazolyl (e.g., 2,3,4,5-tetrahydro phenyl-S-oxo-lH-pyrazolyl); 2,3,4,5-tetrahydro-2,4-dioxo-lH-imidazolyl (erg, 2,3,4,5-tetrahydro- 2,4-dioxomethylphenyl-lH-imidazolyl); 2,3-dihydrothioxo-1,3,4-oxadiazolyl (e.g., 2,3- othioxophenyl—1,3,4—oxadiazolyl); 4,5-dihydrooxo- azolyl (e. g., 4,5-dihydro-3— methylamino 5-oxo-1H-triazolyl); 1,2,3,4-tetrahydro-2,4-dioxopyridinyl (e‘g., l,2,3,4—tetrahydro- 2,4-dioxo-3,3-diethylpyridinyl); 2,6-dioxo-piperidinyl (e.g., 2,6-dioxoethylphenylpiperidinyl); l ,6-dihydrooxopyridiminyl; 1,6-dihydro-4—oxopyrimidinyl (e. g., 2-(methylthio)-1 ,6-dihydro-4— oxo-S-methylpyrimidinyl); 1,2,3,4—tetrahydro-2,4-dioxopy1imidinyl(e.g., 1,2,3,4—tetrahydro-2,4- dioxoethylpyrimidinyl); l,6-dihydrooxo-pyridazinyl (e. g., 1,6-dihydrooxo yridazinyl); 1,6-dihydrooxo-l ,2,4—triazinyl (e. g., l,6-dihydroisopropyloxo-l,2,4- triazinyl); 2,3-dihydrooxo- lH-indolyl (e.g., 3,3-dimethyl-2,3-dihydrooxo- lH-indolyl and 2,3- dihydro-Z—oxo-3,3’-spiropropane-lH—indol-l-yl); l,3-dihydro-l-oxo-2H—iso-indolyl; 1,3-dihydrol ,3-dioxo—2H—iso-indolyl; lH-benzopyrazolyl (e.g., l-(ethoxycarbonyl)- lH-benzopyrazolyl); 2,3— dihydro—2—oxo- lH-benzimidazolyl (e. g., 3-ethyl-2,3-dihydrooxo- lH-benzimidazolyl); 2,3- dihydro—2—oxo-benzoxazolyl (e.g., 5-chloro-2,3-dihydrooxo-benzoxazolyl); 2,3-dihydrooxo— benzoxazolyl; 2-oxo-2H-benzopyranyl; nzodioxanyl; 1,3-benzodioxanyl; 2,3-dihydro oxo,4H— nzothiazinyl; 3 ,4-dihydro-4—oxo-3H—quinazolinyl (e.g. , yl-3 ,4-dihydro—4—oxo— 3H—quinazolinyl); 1,2,3 ,4—tetrahydro-2,4-dioxo-3H—quinazolyl (e.g., l-ethyl-l ,2,3,4-tetrahydro—2,4— 3H-quinazolyl); l,2,3,6-tetrahydro-2,6-dioxo-7H—purinyl (e.g., l,2,3,6-tetrahydro—l,3- dimethyl-2,6-dioxo-7 H -purinyl); l,2,3,6-tetrahydro-2,6-dioxo-l H ipurinyl (e. g., 1,23,6— tetrahydro-3,7-dimethyl—2,6-dioxo-l H -purinyl); 2-oxobenz[c,d]indolyl; l,l-dioxo-2H-naphth[l,8- c,d]isothiazolyl; and l,8-naphthylenedicarboxamido. Additional cyclics include 3,3a,4,5,6,6a— dro-pyrrolo [3 ,4-b]pyrrol—(2H)—yl, and 2,5-diazabicyclo[2.2. l]heptanyl, homopiperazinyl (or diazepanyl), tetrahydropyranyl, dithiazolyl, benzofuranyl, benzothienyl, oxepanyl, nyl, azocanyl, oxecanyl, and thiocanyl. Heterocyclic groups also include groups of the a “’7\ \ / E' where E’ is selected from the group consisting of -N- and -CH-; F’ is selected from the group consisting of , -NH-CH2-, -NH-C(O)—, -NH-, -CH=N—, -CH2-NH-, -C(O)-NH—, -CH=CH—, - CH2-, -CH2CH2-, —CH20-, -OCH2-, -O-, and -S-; and G’ is selected from the group consisting of - CH- and -N-. Any of the heterocyclyl groups mentioned herein may be optionally substituted with one, two, three, four or five substituents independently selected from the group consisting of: (1) C1. 7 acyl (e.g., carboxyaldehyde ); (2) C140 alkyl (e. g., C“; alkyl, C1.6 alkoxy-C1.6 alkyl, C1.6 akylsulfmyl-C1_5 alkyl, amino-CM alkyl, azido-C1_6 alkyl, (carboxyaldehyde)-C1.6 alkyl, halo-CH, alkyl (e.g., perfluoroalkyl), hydroxy-C1_5 alkyl, nitro-CH; alkyl, or CH; thioalkoxy-CM alkyl); (3) C1. alkoxy (e.g., CM alkoxy, such as perfluoroalkoxy); (4) C1.6 alkylsulfinyl; (5) C640 aryl; (6) amino; (7) CM alk-C6_10 aryl; (8) azido; (9) C343 lkyl; (10) C14, alk-C3_8 cycloalkyl; (l 1) halo; (12) C1_ 12 heterocyclyl (e. g., C242 heteroaryl); (13) (C142 heterocyclyl)oxy; (14) hydroxy; (15) nitro; (16) C1_ koxy (e.g., C1_6 thioalkoxy); (17) -(CH2)qCOZRAY, where q is an integer from zero to four, and RAY is selected from the group consisting of (a) C1_6 alkyl, (b) C640 aryl, (0) hydrogen, and (d) CM alk—C6_1o aryl; (18) -(CH2)qCONRB)RCY, where q is an integer from zero to four and where RBI and RC) are independently selected from the group consisting of (a) en, (b) C14, alkyl, (c) C640 aryl, and (d) C1_5 alk-C5_1o aryl; (19) -(CH2)qS02RDy, where q is an integer from zero to four and where RD, is selected from the group consisting of (a) C1_6 alkyl, (b) C640 aryl, and (C) CH _10 aryl; (20) -(CH2)qS02NRE)RF’, where q is an integer from zero to four and where each of RE, and RF) is, independently, selected from the group consisting of (a) hydrogen, (b) C1_6 alkyl, (0) C640 aryl, and (d) C”, alk-C5_10 aryl; (21) thiol; (22) C640 aryloxy; (23) C3_8 cycloalkoxy; (24) arylalkoxy; (25) C1_6 alk—CHZ heterocyclyl (e.g., C1_6 alk-C1_12 heteroaryl); (26) 0x0; (27) (C142 heterocyclyl)imino; (28) C240 alkenyl; and (29) C240 alkynyl. In some embodiments, each of these groups can be further substituted as described herein. For example, the ne group of a Cl-alkaryl or a C1- alkheterocyclyl can be further substituted with an oxo group to afford the respective aryloyl and (heterocyclyl)oyl sub stituent group.
The term “(heterocyclyl) imino,” as used herein, represents a heterocyclyl group, as defined herein, attached to the parent lar group through an imino group. In some embodiments, the heterocyclyl group can be substituted with 1, 2, 3, or 4 tuent groups as defined herein.
The term “(heterocyclyl)oxy,” as used , represents a heterocyclyl group, as defined herein, attached to the parent molecular group through an oxygen atom. In some embodiments, the heterocyclyl group can be tuted with 1, 2, 3, or 4 substituent groups as defined .
The term “(heterocyclyl)oyl,” as used herein, represents a heterocyclyl group, as defined herein, attached to the parent molecular group through a carbonyl group. In some embodiments, the cyclyl group can be substituted With 1, 2, 3, or 4 substituent groups as defined herein.
The term “hydrocarbon,” as used herein, represents a group consisting only of carbon and hydrogen atoms.
The term “hydroxy,” as used herein, represents an —OH group. In some ments, the hydroxy group can be substituted with 1, 2, 3, or 4 substituent groups (e.g., O-protecting ) as defined herein for an alkyl.
] The term “hydroxyalkenyl,” as used herein, represents an alkenyl group, as defined , substituted by one to three hydroxy groups, with the proviso that no more than one hydroxy group may be attached to a single carbon atom of the alkyl group, and is exemplified by dihydroxypropenyl, hydroxyisopentenyl, and the like. In some embodiments, the hydroxyalkenyl group can be tuted with 1, 2, 3, or 4 substituent groups (e. g., O-protecting groups) as defined herein for an alkyl.
The term “hydroxyalkyl,” as used herein, represents an alkyl group, as defined herein, substituted by one to three hydroxy groups, with the proviso that no more than one y group may be attached to a single carbon atom of the alkyl group, and is exemplified by hydroxymethyl, dihydroxypropyl, and the like. In some embodiments, the hydroxyalkyl group can be substituted with 1, 2, 3, or 4 substituent groups (e. g., O-protecting groups) as defined herein for an alkyl.
The term “hydroxyalkynyl,” as used herein, represents an alkynyl group, as defined herein, substituted by one to three hydroxy , with the proviso that no more than one hydroxy group may be attached to a single carbon atom of the alkyl group. In some embodiments, the hydroxyalkynyl group can be substituted with 1, 2, 3, or 4 substituent groups (e.g., O-protecting groups) as defined herein for an alkyl.
The term “isomer,” as used herein, means any tautomer, isomer, enantiomer, or diastereomer of any compound of the invention. It is recognized that the compounds of the ion can have one or more chiral centers and/or double bonds and, therefore, exist as isomers, such as double-bond isomers (i.e., geometric E/Z isomers) or diastereomers (e.g., enantiomers (i.e., (+) or (-)) or cis/trans isomers). According to the invention, the chemical structures depicted herein, and therefore the compounds of the invention, encompass all of the corresponding stereoisomers, that is, both the stereomerically pure form (e. g., geometrically pure, enantiomerically pure, or diastereomerically pure) and enantiomeric and isomeric mixtures, e.g., racemates. Enantiomeric and isomeric mixtures of compounds of the invention can typically be ed into their component enantiomers or stereoisomers by well-known methods, such as chiral-phase gas chromatography, chiral-phase high performance liquid chromatography, crystallizing the nd as a chiral salt x, or crystallizing the nd in a chiral solvent. omers and stereoisomers can also be obtained from stereomerically or enantiomerically pure intermediates, reagents, and catalysts by well-known asymmetric synthetic The term “N—protected ” as used herein, refers to an amino group, as defined herein, to which is attached one or two N—protecting groups, as defined herein.
The term “N-protecting group,” as used herein, represents those groups intended to protect an amino group against undesirable reactions during synthetic procedures. Commonly used N- protecting groups are sed in Greene, “Protective Groups in Organic Synthesis,” 3rd Edition (John Wiley & Sons, New York, 1999), which is incorporated herein by reference. N—protecting groups include acyl, aryloyl, or carbamyl groups such as formyl, acetyl, propionyl, pivaloyl, t- butylacetyl, 2-chloroacetyl, 2-bromoacetyl, roacetyl, oroacetyl, phthalyl, o- nitrophenoxyacetyl, (x-chlorobutyryl, benzoyl, 4-chlorobenzoyl, 4-bromobenzoyl, 4-nitrobenzoyl, and chiral auxiliaries such as protected or unprotected D, L or D, o acids such as alanine, leucine, alanine, and the like; sulfonyl-containing groups such as benzenesulfonyl, p- toluenesulfonyl, and the like; carbamate forming groups such as oxycarbonyl, pchlorobenzyloxycarbonyl , p-methoxybenzyloxycarbonyl, obenzyloxycarbonyl, 2- nitrobenzyloxycarbonyl, p-bromobenzyloxycarbonyl, 3,4-dimethoxybenzyloxycarbonyl, 3,5—dimethoxybenzyloxycarbonyl, 2,4—dimethoxybenzyloxycarbonyl, 4-methoxybenzyloxycarbonyl, 2—nitro-4,5-dimethoxybenzyloxycarbonyl, 3 ,4,5-trimethoxybenzyloxycarbonyl, 1-(p-biphenylyl)- l - methylethoxycarbonyl, 0L,(x-dimethyl—3,5-dimethoxybenzyloxycarbonyl, benzhydryloxy carbonyl, t- butyloxycarbonyl, diisopropylmethoxycarbonyl, isopropyloxycarbonyl, ethoxycarbonyl, methoxycarbonyl, allyloxycarbonyl, 2,2,2,-trichloroethoxycarbonyl, phenoxycarbonyl, 4— nitrophenoxy carbonyl, fluorenylmethoxycarbonyl, cyclopentyloxycarbonyl, adamantyloxycarbonyl, cyclohexyloxycarbonyl, phenylthiocarbonyl, and the like, alkaryl groups such as benzyl, triphenylrnethyl, benzyloxymethyl, and the like and silyl groups, such as trimethylsilyl, and the like. Preferred ecting groups are formyl, acetyl, benzoyl, pivaloyl, t- butylacetyl, alanyl, phenylsulfonyl, benzyl, t—butyloxycarbonyl (Boc), and benzyloxycarbonyl (Cbz).
The term “nitro,” as used herein, represents an —N02 group.
The term “0-protecting group,” as used herein, represents those groups intended to t an oxygen containing (e. g., phenol, hydroxyl, or carbonyl) group against undesirable reactions during synthetic procedures. Commonly used 0-protecting groups are disclosed in Greene, ctive Groups in Organic Synthesis,” 3‘d Edition (John Wiley & Sons, New York, 1999), which is incorporated herein by reference. Exemplary 0-protectirig groups e acyl, aryloyl, or carbamyl groups, such as formyl, acetyl, propionyl, pivaloyl, t—butylacetyl, 2-chloroacetyl, 2- bromoacetyl, trifluoroacetyl, trichloroacetyl, phthalyl, o-nitrophenoxyacetyl, a—chlorobutyryl, benzoyl, 4-chlorobenzoyl, 4-bromobenzoyl, t—butyldimethylsilyl, tii-isa-propylsilyloxymethyl, 4,4'- dimethoxytrityl, isobutyryl, phenoxyacetyl, 4-isopropylpehenoxyacetyl, dimethylformamidino, and 4-nitrobenzoyl; arbonyl groups, such as acyl, acetyl, propionyl, pivaloyl, and the like; optionally substituted arylcarbonyl groups, such as benzoyl; silyl groups, such as trimethylsilyl (TMS), tert—butyldimethylsilyl (TBDMS), tri-iso-propylsilyloxymethyl (TOM), triisopropylsilyl (TIPS), and the like; ether-forming groups with the hydroxyl, such methyl, methoxymethyl, tetrahydropyranyl, benzyl, p-methoxybenzyl, trityl, and the like; alkoxycarbonyls, such as methoxycarbonyl, ethoxycarbonyl, isopropoxycarbonyl, ropoxycarbonyl, n-butyloxycarbonyl, isobutyloxycarbonyl, sec-butyloxycarbonyl, t-butyloxycarbonyl, 2-ethylhexyloxycarbonyl, cyclohexyloxycarbonyl, methyloxycarbonyl, and the like; alkoxyalkoxycarbonyl groups, such as methoxymethoxycarbonyl, ethoxymethoxycarbonyl, oxyethoxycarbonyl, 2- ethoxyethoxycarbonyl, 2-butoxyethoxycarbonyl, 2-methoxyethoxymethoxycarbonyl, allyloxycarbonyl, propargyloxycarbonyl, 2-butenoxycarbonyl, 3-methylbutenoxycarbonyl, and the like; haloalkoxycarbonyls, such as 2-chloroethoxycarbonyl, roethoxycarbonyl, 2,2,2— trichloroethoxycarbonyl, and the like; optionally substituted arylalkoxycarbonyl groups, such as benzyloxycarbonyl, ylbenzyloxycarbonyl, p-methoxybenzyloxycarbonyl, p- nitrobenzyloxycarbonyl, 2,4—dinitrobenzyloxycarbonyl, methylbenzyloxycarbonyl, p- chlorobenzyloxycarbonyl, p-bromobenzyloxy-carbonyl, ylmethyloxycarbonyl, and the like; and optionally substituted aryloxycarbonyl groups, such as phenoxycarbonyl, p- nitrophenoxycarbonyl, o-nitrophenoxycarbonyl, 2,4-dinitrophenoxycarbonyl, p-methyl- phenoxycarbonyl, m-methylphenoxycarbonyl, o-bromophenoxycarbonyl, 3,5- dimethylphenoxycarbonyl, rophenoxycarbonyl, 2-chloro-4—nitrophenoxy-carbonyl, and the like); substituted alkyl, aryl, and alkaryl ethers (e. g., trityl; methylthiomethyl; ymethyl; benzyloxymethyl; siloxymethyl; 2,2,2,-trichloroethoxymethyl; tetrahydropyranyl; tetrahydrofuranyl; ethoxyethyl; l-[2-(trimethylsilyl)ethoxy]ethyl; 2-trimethylsilylethyl; t-butyl ether; p-chlorophenyl, p-methoxyphenyl, p-nitrophenyl, benzyl, p-methoxybenzyl, and nitrobenzyl); silyl ethers (e.g., trimethylsilyl; triethylsilyl; triisopropylsilyl; dimethylisopropylsilyl; t-butyldimethylsilyl; t- butyldiphenylsilyl; tribenzylsilyl; triphenylsilyl; and diphenymethylsilyl); carbonates (e.g., methyl, methoxymethyl, 9-fluorenylmethyl; ethyl; 2,2,2-trichloroethyl; 2-(trimethylsilyl)ethyl; vinyl, allyl, nitrophenyl; benzyl; methoxybenzyl; methoxybenzyl; and nitrobenzyl); carbonyl-protecting groups (e.g., acetal and ketal groups, such as dimethyl acetal, 1,3-dioxolane, and the like; acylal ; and dithiane groups, such as 1,3-dithianes, 1,3-dithiolane, and the like); carboxylic acid- protecting groups (e.g., ester groups, such as methyl ester, benzyl ester, t—butyl ester, sters, and the like; and ine groups.
] The term “oxo” as used herein, represents :0.
The term “perfluoroalkyl,” as used herein, represents an alkyl group, as defined herein, where each en radical bound to the alkyl group has been replaced by a fluoride radical.
Perfluoroalkyl groups are exemplified by trifluoromethyl, pentafluoroethyl, and the like.
The term oroalkoxy,” as used herein, ents an alkoxy group, as defined herein, where each en radical bound to the alkoxy group has been replaced by a fluoride radical.
Perfluoroalkoxy groups are exemplified by romethoxy, pentafluoroethoxy, and the like.
The term “spirocyclyl,” as used herein, represents a C24 alkylene diradical, both ends of which are bonded to the same carbon atom of the parent group to form a spirocyclic group, and also a CM heteroalkylene diradical, both ends of which are bonded to the same atom. The heteroalkylene radical forming the spirocyclyl group can containing one, two, three, or four heteroatoms independently selected from the group consisting of nitrogen, oxygen, and sulfur. In some ments, the yclyl group includes one to seven carbons, excluding the carbon atom to which the diradical is attached. The spirocyclyl groups of the invention may be optionally substituted with l, 2, 3, or 4 substituents provided herein as optional substituents for cycloalkyl and/or heterocyclyl groups.
The term “stereoisomer,” as used , refers to all possible different isomeric as well as conformational forms which a compound may possess (e. g., a compound of any formula described herein), in particular all possible stereochemically and conformationally isomeric forms, all diastereomers, enantiomers and/or conformers of the basic molecular structure. Some compounds of the present invention may exist in different tautomeric forms, all of the latter being included within the scope of the present invention.
The term alkyl,” as used herein, represents an alkyl group, as defined herein, substituted by a sulfo group of —SO3H. In some ments, the alkyl group can be further substituted with l, 2, 3, or 4 tuent groups as bed herein, and the sulfo group can be further substituted with one or more 0-protecting groups (e.g., as described herein).
The term “sulfonyl,” as used herein, represents an -S(O)2- group.
The term “thioalkaryl,” as used , represents a chemical substituent of formula iSR, where R is an alkaryl group. In some ments, the alkaryl group can be further substituted with l, 2, 3, or 4 substituent groups as described herein.
The term “thioalkheterocyclyl,” as used herein, represents a chemical substituent of formula 7SR, where R is an alkheterocyclyl group. In some embodiments, the alkheterocyclyl group can be further substituted with l, 2, 3, or 4 substituent groups as described herein.
The term “thioalkoxy,” as used herein, represents a chemical substituent of formula 7SR, where R is an alkyl group, as defined herein. In some embodiments, the alkyl group can be further substituted with l, 2, 3, or 4 substituent groups as bed herein.
Compound: As used herein, the term “compound,” is meant to include all isomers, geometric isomers, tautomers, and isotopes of the structures depicted.
The compounds described herein can be asymmetric (e.g., having one or more stereocenters). All stereoisomers, such as enantiomers and diastereomers, are intended unless otherwise indicated. Compounds of the present disclosure that contain asymmetrically substituted carbon atoms can be isolated in optically active or racemic forms. Methods on how to prepare optically active forms from optically active starting materials are known in the art, such as by resolution of racemic mixtures or by stereoselective synthesis. Many geometric isomers of , C:N double bonds, and the like can also be present in the compounds described herein, and all such stable isomers are contemplated in the present disclosure. Cis and trans geometric s of the compounds of the present disclosure are described and may be isolated as a e of isomers or as separated isomeric forms.
Compounds of the present disclosure also include tautomeric forms. Tautomeric forms result from the swapping of a single bond with an adjacent double bond and the concomitant migration of a proton. Tautomeric forms include ropic tautomers which are isomeric protonation states having the same empirical a and total charge. Examples prototropic tautomers include ketone — enol pairs, amide — imidic acid pairs, lactam — lactim pairs, amide — imidic acid pairs, enamine — imine pairs, and annular forms where a proton can occupy two or more ons of a heterocyclic system, such as, 1H- and 3H-imidazole, 1H-, 2H— and 4H- 1,2,4-triazole, 1H- and 2H- isoindole, and 1H- and 2H-pyrazole. Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution.
] Compounds of the present disclosure also include all of the isotopes of the atoms occurring in the intermediate or final compounds. “Isotopes” refers to atoms having the same atomic number but different mass numbers resulting from a different number of neutrons in the nuclei. For example, isotopes of hydrogen include tritium and deuterium.
The compounds and salts of the present disclosure can be prepared in combination with solvent or water molecules to form solvates and hydrates by routine methods.
Conserved: As used herein, the term “conserved” refers to nucleotides or amino acid residues of a polynucleotide sequence or polypeptide sequence, tively, that are those that occur unaltered in the same on of two or more sequences being ed. Nucleotides or amino acids that are relatively conserved are those that are conserved amongst more related sequences than nucleotides or amino acids appearing elsewhere in the sequences.
In some embodiments, two or more sequences are said to be “completely conserved” if they are 100% identical to one another. In some embodiments, two or more sequences are said to be y conserved” if they are at least 70% identical, at least 80% identical, at least 90% cal, or at least 95% identical to one another. In some embodiments, two or more sequences are said to be “highly conserved” if they are about 70% identical, about 80% identical, about 90% identical, about 95%, about 98%, or about 99% identical to one another. In some embodiments, two or more sequences are said to be rved” if they are at least 30% identical, at least 40% identical, at least 50% cal, at least 60% identical, at least 70% identical, at least 80% identical, at least 90% identical, or at least 95% identical to one another. In some embodiments, two or more ces are said to be “conserved” if they are about 30% identical, about 40% cal, about 50% identical, about 60% identical, about 70% identical, about 80% identical, about 90% cal, about 95% identical, about 98% identical, or about 99% identical to one r. Conservation of sequence may apply to the entire length of an oligonucleotide or polypeptide or may apply to a portion, region or feature thereof.
Cyclic or Cyclized: As used herein, the term “cyclic” refers to the presence of a continuous loop. Cyclic molecules need not be circular, only joined to form an unbroken chain of subunits. Cyclic molecules such as the mRNA of the present invention may be single units or multimers or comprise one or more components of a complex or higher order structure.
Cytostaric: As used , tatic” refers to inhibiting, ng, suppressing the growth, division, or multiplication of a cell (e.g., a mammalian cell (e.g., a human cell)), bacterium, virus, fungus, oan, parasite, prion, or a combination thereof. ] xic: As used herein, “cytotoxic” refers to killing or causing injurious, toxic, or deadly effect on a cell (e.g., a mammalian cell (e.g., a human cell)), bacterium, virus, fungus, protozoan, parasite, prion, or a combination thereof.
Delivery: As used herein, “delivery” refers to the act or manner of delivering a nd, substance, entity, moiety, cargo or payload.
Delivery Agent: As used herein, “delivery agent” refers to any substance which facilitates, at least in part, the in vivo delivery of a polynucleotide to targeted cells.
Destabilized: As used , the term “destable,” “destabilize,” or “destabilizing region” means a region or molecule that is less stable than a starting, wild-type or native form ofthe same region or molecule.
Detectable label: As used herein, “detectable label” refers to one or more markers, signals, or moieties which are attached, incorporated or associated with another entity that is y detected by methods known in the art including raphy, fluorescence, chemiluminescence, enzymatic ty, ance and the like. Detectable labels e radioisotopes, fluorophores, chromophores, enzymes, dyes, metal ions, ligands such as biotin, avidin, streptavidin and haptens, quantum dots, and the like. Detectable labels may be d at any position in the peptides or proteins disclosed herein. They may be within the amino acids, the peptides, or proteins, or located at the N- or C- termini.
Digest: As used herein, the term “digest” means to break apart into smaller pieces or components. When referring to polypeptides or proteins, digestion results in the production of peptides.
Distal: As used herein, the term “distal” means situated away from the center or away from a point or region of interest.
Encodedprotein cleavage signal: As used herein, ed protein cleavage signal” refers to the nucleotide sequence which encodes a protein cleavage signal. ered." As used herein, embodiments of the invention are “engineered” when they are designed to have a feature or property, whether structural or chemical, that varies from a starting point, wild type or native molecule.
Expression: As used herein, “expression” of a nucleic acid sequence refers to one or more of the following events: (1) production of an RNA te from a DNA sequence (e.g., by ription); (2) processing of an RNA transcript (e.g., by splicing, editing, 5’ cap formation, and/or 3’ end processing); (3) ation of an RNA into a polypeptide or protein; and (4) post- translational modification of a polypeptide or protein.
Feature: As used herein, a “feature” refers to a characteristic, a property, or a distinctive element.
Formulation: As used herein, a “formulation” includes at least a polynucleotide and a delivery agent.
Fragment: A “fragment,” as used herein, refers to a portion. For example, fragments of proteins may se polypeptides obtained by digesting full-length protein isolated from cultured cells.
Functional: As used herein, a “functional” biological le is a biological molecule in a form in which it exhibits a property and/or activity by which it is terized.
Homology: As used herein, the term “homology” refers to the overall relatedness between polymeric molecules, e.g. between nucleic acid molecules (e.g. DNA molecules and/or RNA molecules) and/or n polypeptide les. In some embodiments, polymeric molecules are considered to be “homologous” to one r if their sequences are at least 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99% identical or similar. The term “homologous” necessarily refers to a comparison between at least two sequences ucleotide or polypeptide sequences). In accordance with the invention, two polynucleotide sequences are considered to be homologous if the ptides they encode are at least about 50%, 60%, 70%, 80%, 90%, 95%, or even 99% for at least one stretch of at least about 20 amino acids. In some embodiments, homologous cleotide sequences are characterized by the ability to encode a stretch of at least 4—5 uniquely specified amino acids. For polynucleotide sequences less than 60 nucleotides in length, homology is determined by the ability to encode a stretch of at least 4—5 uniquely specified amino acids. In accordance with the ion, two protein ces are considered to be homologous if the proteins are at least about 50%, 60%, 70%, 80%, or 90% identical for at least one stretch of at least about 20 amino acids.
Idemily: As used herein, the term “identity” refers to the overall relatedness between polymeric molecules, e,g,, between oligonucleotide molecules (6.g. DNA molecules and/or RNA molecules) and/or between polypeptide molecules. Calculation of the percent identity of two polynucleotide sequences, for example, can be performed by aligning the two sequences for optimal comparison purposes (e.g., gaps can be introduced in one or both of a first and a second nucleic acid sequences for optimal alignment and non-identical sequences can be disregarded for comparison purposes). In certain embodiments, the length of a sequence aligned for comparison purposes is at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or 100% of the length of the reference sequence. The nucleotides at corresponding nucleotide positions are then compared. When a position in the first sequence is occupied by the same nucleotide as the corresponding position in the second sequence, then the molecules are identical at that on. The percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which needs to be introduced for optimal alignment of the two sequences. The ison of sequences and ination of percent identity n two sequences can be lished using a mathematical algorithm. For example, the percent identity between two nucleotide sequences can be determined using s such as those described in Computational Molecular Biology, Lesk, A. M., ed., Oxford University Press, New York, 1988; Biocomputing: Informatics and Genome Projects, Smith, D. W., ed., Academic Press, New York, 1993; Sequence Analysis in Molecular Biology, von Heinje, G., ic Press, 1987; Computer is of Sequence Data, Part I, Griffin, A. M., and Griffin, H. G., eds., Humana Press, New Jersey, 1994; and Sequence Analysis Primer, Gribskov, M. and Devereux, J., eds., M Stockton Press, New York, 1991; each of which is orated herein by reference. For example, the t identity between two nucleotide sequences can be determined using the algorithm of Meyers and Miller (CABIOS, 1989, 4:1 l-l7), which has been orated into the ALIGN program (version 2.0) using a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4. The percent ty between two nucleotide sequences can, alternatively, be determined using the GAP m in the GCG software e using an NWSgapdna.CMP matrix. Methods commonly employed to determine percent identity between sequences include, but are not limited to those disclosed in Carillo, H., and Lipman, D., SIAM J Applied Math, 48: 1073 (1988); incorporated herein by reference. Techniques for determining identity are codified in publicly available computer ms. Exemplary er software to determine homology between two sequences include, but are not limited to, GCG program package, ux, 1., et al., Nucleic Acids ch, 12(1), 387 (1984)), BLASTP, BLASTN, and FASTA Altschul, S. F. et al., J. Molec. Biol, 215, 403 (1990)). ] t expression ofa gene: As used herein, the phrase “inhibit sion of a gene” means to cause a reduction in the amount of an expression product of the gene. The sion product can be an RNA transcribed from the gene (e.g., an mRNA) or a polypeptide translated from an mRNA transcribed from the gene. Typically a reduction in the level of an mRNA results in a reduction in the level of a polypeptide translated therefrom. The level of expression may be determined using standard techniques for measuring mRNA or protein.
In vitro: As used herein, the term “in vitro” refers to events that occur in an artificial environment, e.g., in a test tube or reaction vessel, in cell culture, in a Petri dish, etc, rather than within an sm (e.g., animal, plant, or microbe).
In vivo: As used herein, the term “in vivo” refers to events that occur within an organism (e.g., animal, plant, or microbe or cell or tissue thereof).
Isolated: As used herein, the term “isolated” refers to a substance or entity that has been separated from at least some of the components with which it was associated (whether in nature or in an experimental setting). Isolated substances may have varying levels of purity in reference to the substances from which they have been associated. Isolated substances and/or es may be separated from at least about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, or more of the other components with which they were initially associated. In some embodiments, ed agents are more than about 80%, about 85%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or more than about 99% pure. As used herein, a substance is “pure” if it is substantially free of other components. Substantially isolated: By “substantially isolated” is meant that the compound is substantially separated from the environment in which it was formed or detected.
Partial separation can include, for example, a composition enriched in the nd of the present disclosure. Substantial separation can e compositions containing at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 97%, or at least about 99% by weight of the compound of the t disclosure, or salt thereof. Methods for isolating compounds and their salts are routine in the art.
Linker." As used , a linker refers to a group of atoms, e.g, 10-1,000 atoms, and can be comprised of the atoms or groups such as, but not limited to, carbon, amino, mino, oxygen, sulfur, sulfoxide, sulfonyl, carbonyl, and imine. The linker can be attached to a modified nucleoside or nucleotide on the base or sugar moiety at a first end, and to a payload, e. g., a detectable or therapeutic agent, at a second end. The linker may be of sufficient length as to not interfere with incorporation into a nucleic acid sequence. The linker can be used for any useful purpose, such as to form multimers (e.g., through linkage of two or more polynucleotides) or ates, as well as to administer a payload, as described herein. Examples of chemical groups that can be incorporated into the linker e, but are not limited to, alkyl, alkenyl, alkynyl, amido, amino, ether, her, ester, alkylene, heteroalkylene, aryl, or heterocyclyl, each of which can be optionally substituted, as described herein. Examples of linkers include, but are not limited to, unsaturated alkanes, polyethylene glycols (e.g., ethylene or propylene glycol monomeric units, e.g., diethylene glycol, ylene glycol, triethylene glycol, tripropylene glycol, tetraethylene glycol, or thylene glycol), and n polymers, Other examples include, but are not limited to, cleavable moieties within the linker, such as, for example, a rde bond (-S-S-) or an azo bond (-N:N-), which can be cleaved using a reducing agent or photolysis. Non-limiting examples of a selectively cleavable bond include an amido bond can be cleaved for example by the use of -carboxyethyl)phosphine (TCEP), or other reducing agents, and/or photolysis, as well as an ester bond can be cleaved for example by acidic or basic hydrolysis.
Modified: As used herein “modified” refers to a changed state or ure of a molecule ofthe invention. Molecules may be modified in many ways including chemically, structurally, and functionally. In one embodiment, the mRNA molecules of the present ion are modified by the introduction of non-natural nucleosides and/or nucleotides, e. g., as it relates to the natural ribonucleotides A, U, G, and C. Noncanonical nucleotides such as the cap structures are not considered “modified” although they differ from the chemical structure of the A, C, G, U ribonucleotides.
] Naturally occurring: As used herein, “naturally occurring” means existing in nature without artificial aid.
Non-human vertebrate: As used herein, a “non human vertebrate” es all vertebrates except Homo sapiens, including wild and domesticated species. Examples of non-human rates include, but are not limited to, mammals, such as alpaca, banteng, bison, camel, cat, cattle, deer, dog, donkey, gayal, goat, guinea pig, horse, llama, mule, pig, rabbit, reindeer, sheep water buffalo, and yak. get: As used herein, “off target” refers to any unintended effect on any one or more target, gene, or cellular transcript.
Open readingflame: As used herein, “open reading frame” or “ORF” refers to a ce which does not contain a stop codon in a given reading frame.
Operably linked: As used herein, the phrase “operably linked” refers to a functional connection between two or more molecules, constructs, transcripts, entities, moieties or the like. pe: As used herein, a “paratope” refers to the antigen-binding site of an antibody. t: As used herein, “patient” refers to a subject who may seek or be in need of treatment, es treatment, is receiving ent, will receive treatment, or a subject who is under care by a trained professional for a ular disease or condition.
Optionally substituted: Herein a phrase of the form “optionally substituted X” (e.g., optionally substituted alkyl) is ed to be equivalent to “X, wherein X is optionally substituted” (e.g., “alkyl, wherein said alkyl is ally substituted”). It is not intended to mean that the feature “X” (e.g. alkyl) per se is optional.
Peptide: As used herein, “peptide” is less than or equal to 50 amino acids long, e.g., about ,10, 15, 20, 25, 30, 35, 40, 45, or 50 amino acids long.
Pharmaceutically acceptable: The phrase “pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
] Pharmaceutically acceptable excipients: The phrase “pharmaceutically acceptable excipient,” as used herein, refers any ingredient other than the compounds bed herein (for example, a vehicle capable of suspending or dissolving the active compound) and having the properties of being substantially nontoxic and non-inflammatory in a patient. Excipients may include, for example: herents, antioxidants, binders, coatings, compression aids, disintegrants, dyes (colors), emollients, emulsifiers, fillers (diluents), film formers or coatings, flavors, fragrances, glidants (flow enhancers), lubricants, preservatives, printing inks, sorbents, suspensing or dispersing agents, sweeteners, and waters of hydration. Exemplary excipients include, but are not limited to: butylated hydroxytoluene (BHT), calcium carbonate, calcium phosphate (dibasic), calcium stearate, croscarmellose, crosslinked polyvinyl pyrrolidone, citric acid, crospovidone, cysteine, ethylcellulose, gelatin, hydroxypropyl cellulose, hydroxypropyl methylcellulose, lactose, magnesium stearate, maltitol, mannitol, nine, methylcellulose, methyl paraben, microcrystalline cellulose, polyethylene glycol, polyvinyl idone, povidone, pregelatinized starch, propyl paraben, retinyl palmitate, shellac, silicon dioxide, sodium carboxymethyl ose, sodium citrate, sodium starch glycolate, sorbitol, starch (corn), stearic acid, sucrose, talc, titanium dioxide, vitamin A, vitamin E, vitamin C, and l.
Pharmaceutically acceptable salts: The present disclosure also includes pharmaceutically able salts of the compounds described herein. As used herein, “pharmaceutically acceptable salts” refers to derivatives of the disclosed compounds n the parent compound is modified by converting an existing acid or base moiety to its salt form (e.g., by reacting the free base group with a le c acid). Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. Representative acid addition salts include acetate, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, e, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptonate, ophosphate, lfate, heptonate, hexanoate, romide, hydrochloride, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl e, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, e, oleate, oxalate, ate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartratc, thiocyanate, toluenesulfonate, undecanoate, te salts, and the like. entative alkali or alkaline earth metal salts include , lithium, potassium, calcium, magnesium, and the like, as well as nontoxic ammonium, quaternary ammonium, and amine cations, including, but not limited to ammonium, ethylammonium, tetraethylamrnonium, amine, dimethylamine, trimethylamine, triethylamine, ethylamine, and the like. The pharmaceutically acceptable salts of the present disclosure include the conventional non-toxic salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. The pharmaceutically acceptable salts of the present disclosure can be sized from the parent compound which contains a basic or acidic moiety by conventional chemical methods. lly, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, eous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of le salts are found in Remington ’s ceutical es, 17th ed., Mack Publishing Company, Easton, Pa, 1985, p. 1418, Pharmaceutical Salts: Properties, Selection, and Use, P.H. Stahl and CG. Wermuth (eds), Wiley-VCH, 2008, and Berge et al., Journal ofPharmaceutical Science, 66, 1-19 (1977), each of which is incorporated herein by reference in its entirety. cokinetic: As used herein, “pharmacokinetic” refers to any one or more properties of a molecule or compound as it relates to the determination of the fate of substances administered to a living organism. Pharmacokjnetics is divided into several areas including the extent and rate of absorption, distribution, lism and excretion. This is ly ed to as ADME where: (A) Absorption is the process of a substance entering the blood circulation; (D) Distribution is the dispersion or dissemination of substances throughout the fluids and tissues of the body; (M) Metabolism (or Biotransformation) is the irreversible transformation of parent compounds into daughter metabolites; and (E) Excretion (or Elimination) refers to the elimination of the substances from the body. In rare cases, some drugs irreversibly accumulate in body tissue.
Pharmaceutically acceptable solvate: The term “pharmaceutically acceptable solvate,” as used herein, means a compound of the invention n molecules of a suitable solvent are incorporated in the l lattice. A suitable solvent is physiologically tolerable at the dosage administered. For example, solvates may be prepared by crystallization, recrystallization, or precipitation from a solution that includes organic solvents, water, or a mixture thereof. Examples of suitable ts are l, water (for example, mono-, di—, and tri-hydrates), N- methylpyrrolidinone (NMP), dirnethyl sulfoxide (DMSO), N,N’-dimethylformamide (DMF), N,N’- dimethylacetamide (DMAC), l,3-dimethylimidazolidinone (DMEU), 1,3-djmethyl-3,4,5,6- tetrahydro(lH)-pyrimidinone (DMPU), acetonitrile (ACN), propylene glycol, ethyl acetate, benzyl alcohol, 2-pyrrolidone, benzyl benzoate, and the like. When water is the solvent, the solvate is ed to as a “hydrate.” ] ochemical: As used herein, “physicochemical” means of or ng to a physical and/or chemical property.
Preventing: As used herein, the term “preventing” refers to partially or completely delaying onset of an infection, e, disorder and/or condition; partially or completely delaying onset of one or more symptoms, features, or clinical manifestations of a ular infection, disease, er, andfor condition; partially or completely delaying onset of one or more symptoms, features, or manifestations of a particular infection, disease, disorder, and/or condition; partially or completely delaying progression from an infection, a particular disease, disorder and/or condition; and/or decreasing the risk of developing pathology associated with the infection, the disease, disorder, and/or condition.
Prodrug: The present disclosure also includes prodrugs of the compounds described herein. As used , “prodrugs” refer to any substance, molecule or entity which is in a form predicate for that sub stance, molecule or entity to act as a therapeutic upon al or physical alteration. Prodrugs may by covalently bonded or tered in some way and which e or are converted into the active drug moiety prior to, upon or after administered to a mammalian subject.
Prodrugs can be prepared by modifying functional groups present in the compounds in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compounds. Prodrugs include compounds n hydroxyl, amino, sulfhydryl, or carboxyl groups are bonded to any group that, when administered to a mammalian subject, cleaves to form a free yl, amino, sulflrydryl, or carboxyl group respectively. Preparation and use of s is discussed in T. Higuchi and V. Stella, “Pro-drugs as Novel Delivery Systems,” Vol. 14 ofthe A.C.S.
Symposium Series, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, both of which are hereby incorporated by reference in their entirety.
Proliferate: As used herein, the term “proliferate” means to grow, expand or increase or cause to grow, expand or increase rapidly. “Proliferative” means having the ability to proliferate.
“Anti-proliferative” means having properties counter to or inapposite to proliferative properties. n cleavage site: As used herein, “protein cleavage site” refers to a site where controlled cleavage of the amino acid chain can be lished by chemical, tic or photochemical means.
Protein cleavage signal: As used herein “protein cleavage signal” refers to at least one amino acid that flags or marks a ptide for cleavage.
Protein ofinterest: As used herein, the terms “proteins of interest” or “desired proteins” include those ed herein and fragments, mutants, variants, and alterations thereof.
Proximal: As used herein, the term “proximal” means situated nearer to the center or to a point or region of interest.
Purified: As used herein, “purify,” “purified,” “purification” means to make substantially pure or clear from ed components, al defilement, admixture or imperfection.
Sample: As used herein, the term “sample” or “biological sample” refers to a subset of its tissues, cells or component parts (e. g. body fluids, including but not limited to blood, mucus, lymphatic fluid, synovial fluid, cerebrospinal fluid, saliva, amniotic fluid, amniotic cord blood, urine, vaginal fluid and semen). A sample further may include a nate, lysate or extract prepared from a whole organism or a subset of its tissues, cells or component parts, or a fraction or portion thereof, including but not limited to, for example, plasma, serum, spinal fluid, lymph fluid, the al sections of the skin, respiratory, intestinal, and genitourinary tracts, tears, saliva, milk, blood cells, , organs. A sample r refers to a medium, such as a nutrient broth or gel, which may contain cellular components, such as proteins or nucleic acid molecule.
Signal Sequences: As used , the phrase “signal sequences” refers to a sequence which can direct the transport or localization of a protein.
Significant or Significantly: As used herein, the terms “significant” or “significantly” are used synonymously with the term “substantially.” Single unit dose: As used herein, a “single unit dose” is a dose of any eutic administed in one t one time/single single point of contact, i.e., single administration event.
Similarity: As used herein, the term “similarity” refers to the overall relatedness between ric molecules, e.g. between polynucleotide molecules (e.g. DNA molecules andfor RNA molecules) and/0r between polypeptide molecules. Calculation of percent similarity of polymeric molecules to one another can be performed in the same manner as a calculation of percent identity, except that calculation of percent similarity takes into account conservative substitutions as is understood in the art.
] Split dose: As used herein, a “split dose” is the division of single unit dose or total daily dose into two or more doses.
." As used herein “stable” refers to a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction e, and preferably capable of formulation into an efficacious therapeutic agent. ized: As used herein, the term “stabilize”, lized,” “stabilized region” means to make or become stable.
Subject: As used herein, the term “subject” or “patient” refers to any organism to which a composition in accordance with the invention may be administered, e.g., for experimental, diagnostic, prophylactic, and/or therapeutic purposes. l subjects include animals (e.g, mammals such as mice, rats, rabbits, non-human primates, and humans) and/or plants.
Substantially: As used herein, the term “substantially” refers to the qualitative condition of exhibiting total or near-total extent or degree of a characteristic or ty of interest. One of ordinary skill in the biological arts will understand that biological and chemical phenomena rarely, if ever, go to completion and/or proceed to completeness or achieve or avoid an absolute result. The term “substantially” is ore used herein to capture the potential lack of completeness inherent in many ical and chemical phenomena.
Substantially equal: As used herein as it relates to time differences between doses, the term means plus/minus 2%.
Substantially simultaneously: As used herein and as it relates to plurality of doses, the term means within 2 seconds.
Sufleringfrom: An individual who is ring from” a disease, disorder, and/or condition has been diagnosed with or displays one or more symptoms of a disease, disorder, and/or condition.
Susceptible to: An individual who is “susceptible to” a disease, er, andfor condition has not been diagnosed with and/or may not exhibit ms of the disease, disorder, and/or condition but harbors a propensity to develop a disease or its symptoms. In some embodiments, an individual who is susceptible to a disease, disorder, and/or ion (for example, ) may be characterized by one or more of the following: (1) a genetic mutation associated with development of the disease, disorder, and/or condition; (2) a genetic polymorphism associated with development of the disease, disorder, and/or condition; (3) increased and/or decreased expression andfor activity of a protein and/or nucleic acid associated With the disease, disorder, and/or condition; (4) habits and/or lifestyles associated with development of the disease, disorder, and/or condition; (5) a family history of the disease, disorder, and/or condition; and (6) re to and/or infection with a e associated with development of the disease, disorder, and/or condition In some ments, an individual who is susceptible to a disease, disorder, and/or ion will develop the disease, disorder, andfor condition. In some embodiments, an individual who is susceptible to a disease, disorder, andfor condition will not develop the disease, disorder, and/or condition.
Synthetic: The term “synthetic” means ed, prepared, and/or manufactured by the hand of man. Synthesis of polynucleotides or polypeptides or other molecules of the present invention may be chemical or enzymatic.
Targeted Cells: As used herein, “targeted cells” refers to any one or more cells of interest.
The cells may be found in vitro, in vivo, in situ or in the tissue or organ of an organism. The organism may be an animal, preferably a mammal, more preferably a human and mo st preferably a patient.
Therapeutic Agent: The term “therapeutic agent” refers to any agent that, when administered to a subject, has a therapeutic, diagnostic, and/or prophylactic effect and/or elicits a desired biological and/or pharmacological effect. eutically eflective amount: As used herein, the term “therapeutically effective amount” means an amount of an agent to be delivered (e.g., nucleic acid, drug, therapeutic agent, diagnostic agent, lactic agent, etc.) that is sufficient, when administered to a subject suffering from or susceptible to an infection, disease, disorder, and/or condition, to treat, improve symptoms of, diagnose, prevent, and/or delay the onset of the infection, disease, er, and/or ion. [0081 l] Therapeutically eflective outcome: As used , the term “therapeutically ive outcome” means an outcome that is sufficient in a subject suffering from or susceptible to an infection, e, disorder, and/or condition, to treat, improve symptoms of, diagnose, prevent, and/or delay the onset of the infection, disease, disorder, and/or condition.
Total daily dose: As used herein, a “total daily dose” is an amount given or ibed in 24 hr period. It may be administered as a single unit dose.
Transcriptionfactor: As used herein, the term “transcription factor” refers to a DNA- binding protein that regulates transcription ofDNA into RNA, for example, by activation or sion of transcription. Some transcription s effect regulation of transcription alone, while others act in concert with other proteins. Some transcription factor can both activate and repress transcription under certain conditions. In general, transcription factors bind a specific target sequence or sequences highly similar to a c consensus sequence in a regulatory region of a target gene. Transcription s may regulate transcription of a target gene alone or in a complex with other molecules.
Treating: As used herein, the term ing” refers to partially or completely alleviating, ameliorating, improving, relieving, delaying onset of, inhibiting progression of, reducing severity of, and/or reducing nce of one or more symptoms or features of a particular infection, disease, disorder, andfor condition. For example, “treating” cancer may refer to inhibiting survival, growth, and/or spread of a tumor. Treatment may be administered to a t who does not exhibit signs of a disease, disorder, and/or ion and/or to a subject who exhibits only early signs of a disease, disorder, and/or condition for the purpose of decreasing the risk of developing pathology associated with the disease, disorder, and/or condition.
Unmodified: As used herein, “unmodified” refers to any nce, compound or molecule prior to being changed in any way. Unmodified may, but does not always, refer to the wild type or native form of a biomolecule. les may undergo a series of modifications whereby each modified molecule may serve as the ified” starting molecule for a subsequent modification.
Eguivalents and Scope Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments in accordance with the invention described herein. The scope of the present invention is not intended to be limited to the above Description, but rather is as set forth in the appended claims.
In the claims, articles such as “a,” “an,” and “the” may mean one or more than one unless indicated to the contrary or otherwise t from the t. Claims or descriptions that include “or” n one or more members of a group are considered ed if one, more than one, or all of the group members are present in, employed in, or otherwise relevant to a given product or process unless indicated to the contrary or otherwise evident from the t. The invention includes embodiments in which exactly one member of the group is present in, employed in, or otherwise relevant to a given product or process. The invention includes embodiments in which more than one, or all of the group members are t in, employed in, or otherwise relevant to a given product or process.
It is also noted that the term “comprising” is intended to be open and permits but does not require the inclusion of additional elements or steps. When the term “comprising” is used herein, the term “consisting of” is thus also encompassed and disclosed.
Where ranges are given, endpoints are included. Furthermore, it is to be tood that unless otherwise indicated or otherwise evident from the context and understanding of one of ordinary skill in the art, values that are expressed as ranges can assume any specific value or subrange within the stated ranges in different embodiments of the ion, to the tenth of the unit of the lower limit of the range, unless the t clearly es otherwise.
In addition, it is to be understood that any particular embodiment of the present invention that falls within the prior art may be explicitly excluded from any one or more of the claims. Since such embodiments are deemed to be known to one of ordinary skill in the art, they may be excluded even if the ion is not set forth explicitly . Any particular embodiment of the compositions of the invention (e.g., any nucleic acid or protein encoded thereby; any method of production; any method of use; etc.) can be excluded from any one or more , for any reason, whether or not related to the existence of prior art.
All cited sources, for example, references, publications, databases, database entries, and art cited herein, are incorporated into this ation by reference, even if not expressly stated in the citation. In case of conflicting statements of a cited source and the instant application, the statement in the instant application shall control.
EXAMPLES The present disclosure is further described in the following examples, which do not limit the scope of the disclosure described in the claims.
Example 1. d mRNA In Vitro Transcription A. Materials and s Modified mRNAs according to the invention are made using standard laboratory methods and materials for in vitro transcription with the exception that the nucleotide mix contains modified nucleotides. The open reading frame (ORF) of the gene of interest is flanked by a 5' untranslated region (UTR) ning a strong Kozak translational initiation signal and an alpha-globin 3' UTR terminating with an oligo(dT) sequence for templated addition of a polyA tail for mRNAs not incorporating adenosine analogs. Adenosine-containing mRNAs are synthesized without an oligo (dT) ce to allow for post-transcription poly (A) polymerase poly-(A) tailing.
The modified mRNAs may be modified to reduce the cellular innate immune response.
The modifications to reduce the ar response may include uridine (w) and S-methyl- cytidine (SmeC, Smc or msC). (See, Kariko K et al. Immunity 23:165-75 (2005), Kariko K et al. Mol Ther 16:1833-40 (2008), Anderson BR et al. NAR (2010); herein incorporated by reference).
The ORF may also include various am or downstream additions (such as, but not limited to, B-globin, tags, etc.) may be ordered from an optimization service such as, but limited to, DNA2.0 (Menlo Park, CA) and may contain multiple cloning sites which may have XbaI recognition. Upon receipt of the construct, it may be reconstituted and transformed into chemically competent E. coli.
For the t invention, NEB pha Competent E. coli are used. Transformations are performed according to NEB instructions using 100 ng of plasmid. The protocol is as follows: Thaw a tube ofNEB 5-alpha Competent E. coli cells on ice for 10 s.
Add 1-5 [11 containing 1 pg—100 ng of plasmid DNA to the cell mixture. Carefully flick the tube 4-5 times to mix cells and DNA. Do not vortex.
Place the mixture on ice for 30 minutes. Do not mix.
Heat shock at 42°C for exactly 30 seconds. Do not mix.
Place on ice for 5 s. Do not mix.
Pipette 950 pl of room temperature SOC into the mixture.
Place at 37°C for 60 minutes. Shake vigorously (250 rpm) or rotate.
Warm selection plates to 37°C.
Mix the cells thoroughly by flicking the tube and inverting.
Spread 50-100 ul of each on onto a ion plate and incubate overnight at 37°C.
Alternatively, incubate at 30°C for 24-36 hours or 25°C for 48 hours.
A single colony is then used to inoculate 5 ml ofLB growth media using the riate antibiotic and then allowed to grow (250 RPM, 37° C) for 5 hours. This is then used to inoculate a 200 ml culture medium and allowed to grow overnight under the same ions.
To isolate the plasmid (up to 850 pg), a maxi prep is performed using the Invitrogen PURELINKTM HiPure Maxiprep Kit (Carlsbad, CA), following the manufacturer’s instructions.
In order to generate cDNA for In Vitro ription (IVT), the plasmid (an Example of which is shown in Figure 3) is first linearized using a restriction enzyme such as XbaI. A typical ction digest with XbaI will comprise the following: Plasmid 1.0 pg; 10x Buffer 1.0 pl; XbaI 1.5 pl; dH20 up to 10 pl; incubated at 37° C for 1 hr; If performing at lab scale (< Spg), the reaction is cleaned up using Invitrogen’s PURELINKTM PCR Micro Kit bad, CA) per manufacturer’s instructions. Larger scale purifications may need to be done with a product that has a larger load capacity such as Invitrogen’s rd PURELINKTM PCR Kit (Carlsbad, CA). Following the cleanup, the linearized vector is quantified using the NanoDrop and analyzed to confirm linearization using agarose gel electrophoresis.
B. Agarose Gel Electrophoresis of modified mRNA Individual modified mRNAs (200-400 ng in a 20 pl ) are loaded into a well on a non-denaturing 1.2% Agarose E—Gel (Invitrogen, Carlsbad, CA) and run for 12-15 minutes according to the manufacturer protocol.
C. Agarose Gel Electrophoresis of RT-PCR products Individual reverse transcribed-PCR ts (200-400ng) are loaded into a well of a non— denaturing 1.2% e E-Gel (Invitrogen, Carlsbad, CA) and run for 12-15 s according to the manufacturer protocol.
D. Nanodrop modified mRNA fication and UV spectral data Modified mRNAs in TE buffer (1 pl) are used for Nanodrop UV absorbance readings to quantitate the yield of each modified mRNA from an in vitro transcription reaction (UV absorbance traces are not shown).
Example 2. Modified mRNA Transfection A. Reverse ection For experiments performed in a 24-well collagen-coated tissue culture plate, Keratinocytes are seeded at a cell density of l x 105. For experiments performed in a 96-well collagen-coated tissue culture plate, Keratinocytes are seeded at a cell density of 0.5 x 105. For each modified mRNA to be transfected, modified mRNA: RNAIMAXTM are prepared as described and mixed with the cells in the multi-well plate within 6 hours of cell seeding before cells had adhered to the tissue e plate.
B. Forward Transfection In a 24-well collagen-coated tissue culture plate, nocytes are seeded at a cell density of 0.7 x 105. For experiments performed in a 96-well collagen-coated tissue culture plate, nocytes are seeded at a cell density of 0.3 x 105. Keratinocytes are then grown to a confluency of>70% for over 24 hours. For each modified mRNA to be transfected, modified mRNA: RNAIMAXTM are prepared as described and ected onto the cells in the multi-well plate over 24 hours after cell seeding and adherence to the tissue culture plate.
C. Modified mRNA Translation Screen: G-CSF ELISA Keratinocytes are grown in EpiLife medium with Supplement S7 from Invitrogen at a confluence of>70%. Keratinocytes are e transfected with 300 11g of the indicated ally modified mRNA complexed with RNAIMAXTM from Invitrogen. atively, keratinocytes are forward transfected with 300 ng modified mRNA complexed with RNAIMAXTM from Invitrogen.
The RNA: RNAIMAXTM complex is formed by first incubating the RNA with Supplement-free EPILIFE® media in a 5X volumetric on for 10 minutes at room temperature.
] In a second vial, RNAIMAXTM t is ted with Supplement-free EPHJFE® Media in a 10X volumetric dilution for 10 minutes at room temperature. The RNA vial is then mixed with the RNAIMAXTM vial and incubated for 20-30 at room temperature before being added to the cells in a drop-wise fashion. Secreted huG—CSF tration in the culture medium is measured at 18 hours post-transfection for each of the chemically modified mRNAs in triplicate.
Secretion ofHuman Granulocyte-Colony Stimulating Factor (G-CSF) from transfected human keratinocytes is quantified using an ELISA kit from Invitrogen or R&D s (Minneapolis, MN) following the manufacturers recommended instructions.
D. Modified mRNA Dose and Duration: G—CSF ELISA Keratinocytes are grown in EPILIFE® medium with Supplement S7 from Invitrogen at a confluence of>70%. Keratinocytes are reverse transfected with Ong, 46.875ng, 93.75ng, 187.5ng, 375ng, 750ng, or 1500ng d mRNA complexed with RNAIMAXTM from Invitrogen. The modified mRNA: RNAIMAXTM complex is formed as described. Secreted F concentration in the culture medium is measured at 0, 6, 12, 24, and 48 hours post-transfection for each concentration of each modified mRNA in triplicate. Secretion of Human Granulocyte-Colony Stimulating Factor (G-CSF) from transfected human keratinocytes is quantified using an ELISA kit from Invitrogen or R&D Systems following the manufacturers recommended instructions. e 3. ar Innate Immune Response to Modified Nucleic Acids: ta ELISA and TNF—alpha ELISA An enzyme-linked immunosorbent assay (ELISA) for Human Tumor Necrosis Factor-a ("INF-(1), Human Interferon-l3 (IFN-B) and Human Granulocyte-Colony Stimulating Factor (G-CSF) secreted from in viva-transfected Human Keratinocyte cells is tested for the detection of a cellular innate immune se.
Keratinocy‘tes are grown in EPILIFE® medium with Human nocyte Growth Supplement in the absence of ortisone from Invitrogen at a confluence of >70%.
Keratinocytes are reverse transfected with Ong, 93.75ng, 187.5ng, 375ng, 750ng, 1500ng or 3000ng of the ted chemically modified mRNA complexed with RNAIMAXTM from Invitrogen as described in triplicate. Secreted TNF-(x in the culture medium is measured 24 hours post- transfection for each of the chemically modified mRNAs using an ELISA kit from Invitrogen ing to the manufacturer protocols.
Secreted IFN—[i is ed 24 hours post-transfection for each of the chemically modified mRNAs using an ELISA kit from Invitrogen according to the manufacturer protocols.
Secreted hu—G—CSF concentration is measured at 24 hours post-transfection for each ofthe ally modified mRNAs. Secretion of Human Granulocyte-Colony Stimulating Factor (G- CSF) from transfected human keratinocytes is quantified using an ELISA kit from ogen or R&D Systems (Minneapolis, MN) following the manufacturers recommended instructions. These data indicate which modified mRNA are capable eliciting a reduced cellular innate immune response in comparison to l and other chemically modified polynucleotides or reference compounds by measuring exemplary type 1 cytokines TNF-alpha and IFN—beta.
Example 4. Human Granulocfie—Colony ating Factor-modified mRNA-induced Cell Proliferation Assay ] Human keratinocytes are grown in E® medium with Supplement S7 from Invitrogen at a confluence of >70% in a 24-well collagen-coated TRANSWELL® (Corning, Lowell, MA) co-culture tissue culture plate. Keratinocytes are reverse transfected with 750mg of the indicated chemically modified mRNA complexed with RNAIMAXTM from Invitrogen as described in tnplicate. The modified mRNA: RNAIMAXTM complex is formed as described. Keratinocyte media is exchanged 6-8 hours post-transfection. 42-hours post-transfection, the 24-well TRANSWELL® plate insert with a 0.4um-pore semi-permeable polyester membrane is placed into the hu-G-CSF modified mRNA-transfected keratinocyte containing culture plate.
Human myeloblast cells, Kasumi-l cells or KG-l (0.2 X 105 cells), are seeded into the insert well and cell proliferation is quantified 42 hours post-co-culture tion using the CyQuant Direct Cell Proliferation Assay (Invitrogen) in a 100-120 ul volume in a 96-well plate. modified mRNA-encoding hu-G—CSF-induced myeloblast cell proliferation is expressed as a percent cell proliferation normalized to untransfected keratinocyte/myeloblast co-culture control wells. Secreted hu—G-CSF concentration in both the nocyte and last insert co-culture wells is ed at 42 hours o-culture initiation for each modified mRNA in duplicate. Secretion of Human Granulocyte—Colony Stimulating Factor (G-CSF) is quantified using an ELISA kit from Invitrogen following the manufacturers recommended ctions.
Transfected SF modified mRNA in human keratinocyte feeder cells and untransfected human myeloblast cells are detected by RT-PCR. Total RNA from sample cells is extracted and lysed using RNAEASY® kit (Qiagen, Valencia, CA) according to the manufacturer instructions. Extracted total RNA is ted to RT-PCR for specific amplification ofmodified mRNA-G—CSF using CRIPT® M-MuLV Taq RT-PCR kit (New d BioLabs, Ipswich, MA) according to the manufacturer instructions with SF-specific primers. RT—PCR products are Visualized by 1.2% agarose gel electrophoresis.
Example 5. Cytotoxicifl and Apoptosis This experiment trates cellular Viability, xity and apoptosis for distinct modified mRNA-in vitro transfected Human Keratinocyte cells. Keratinocytes are grown in EPILIFE® medium with Human nocyte Growth Supplement in the absence of hydrocortisone from Invitrogen at a confluence of >70%. nocytes are reverse transfected with Ong, 46.875ng, 93.75ng, 187.5ng, 375ng, 750ng, 1500ng, 3000ng, or 6000ng of modified mRNA complexed with RNAIMAXTM from Invitrogen. The modified mRNA: RNAIMAXTM complex is formed. Secreted huG-CSF concentration in the culture medium is measured at 0, 6, 12, 24, and 48 hours post- transfection for each concentration of each modified mRNA in triplicate. Secretion ofHuman Granulocyte—Colony Stimulating Factor (G-CSF) from transfected human nocytes is quantified using an ELISA kit from Invitrogen or R&D Systems following the manufacturers recommended instructions. Cellular Viability, cytotoxicity and apoptosis is measured at O, 12, 48, 96, and 192 hours post-transfection using the APOTOX-GLOTM kit from Promega (Madison, WI) according to manufacturer instructions.
Example 6. Co-Culture Environment The modified mRNA comprised of ally-distinct modified nucleotides encoding human Granulocyte-Colony Stimulating Factor (G—CSF) may stimulate the cellular proliferation of a transfection incompetent cell in co-culture environment. The co-culture includes a highly transfectable cell type such as a human keratinocyte and a transfection incompetent cell type such as a white blood cell (WBC). The d mRNA encoding G-CSF may be transfected into the highly transfectable cell allowing for the production and secretion of G-CSF protein into the extracellular environment where G—CSF acts in a paracrine—like manner to stimulate the white blood cell sing the G—CSF receptor to proliferate. The expanded WBC population may be used to treat immune-compromised patients or partially titute the WBC population of an irnrnunosuppressed patient and thus reduce the risk of unistic infections.
In r example, a highly transfectable cell such as a fibroblast are transfected with n growth factors to support and simulate the growth, maintenance, or differentiation ofpoorly transfectable nic stem cells or induced otent stem cells.
Exam le 7. 5’-Guanosine Ca in on Modified Nucleic Acids modified mRNAs A. Materials and Methods The cloning, gene synthesis and vector sequencing was performed by DNA2.0 Inc.
(Menlo Park, CA). The ORF was restriction digested using XbaI and used for cDNA synthesis using tailed-or tail-less—PCR. The tailed-PCR cDNA product was used as the template for the modified mRNA sis reaction using 25mM each modified nucleotide mix (all modified nucleotides were custom synthesized or purchased from TriLink h, San Diego, CA except pyrrolo—C triphosphate purchased from Glen Research, Sterling VA; unmodifed tides were purchased from Epicenter Biotechnologies, Madison, WI) and CellScript MEGASCRIPTTM (Epicenter Biotechnologies, Madison, WI) complete mRNA synthesis kit. The in vitro transcription reaction was run for 4 hours at 37°C. Modified mRNAs incorporating adenosine analogs were poly (A) tailed using yeast Poly (A) Polymerase (Affymetrix, Santa Clara, CA). PCR reaction used HiFi PCR 2X MASTER MIXTM (Kapa Biosystems, Woburn, MA). Modified mRNAs were post- transcriptionally capped using recombinant Vaccinia Virus Capping Enzyme (New England BioLabs, Ipswich, MA) and a recombinant ethyltransferase (Epicenter Biotechnologies, Madison, WI) to generate the 5’-guanosine Capl structure. Cap 2 structure and Cap 2 structures may be ted using additional 2’-o-methyltransferases. The In vitro transcribed mRNA product was run on an agarose gel and Visualized. Modified mRNA was purified with Ambioanpplied Biosystems (Austin, TX) MEGAClear RNATM purification kit. PCR used PURELIN'KTM PCR purification kit (Invitrogen, Carlsbad, CA). The t was fied on NANODROPTM UV Absorbance (ThermoFisher, Waltham, MA). Quality, UV absorbance quality and Visualization of the product was performed on an 1.2% agarose gel. The product was resuspended in TE .
B. 5’ Capping Modified Nucleic Acid (mRNA) Structure 5’-capping of modified mRNA may be completed concomitantly during the in vitratranscription reaction using the following chemical RNA cap analogs to generate the nosine cap structure according to manufacturer protocols: 3 'Me—m7G(5')ppp(5')G (the ARCA cap); G(5')ppp(5')A; G(5')ppp(5')G; m7G(5')ppp(5')A; m7G(5')ppp(5')G (New England BioLabs, Ipswich, MA). 5 ’—capping of modified mRNA may be completed ranscriptionally using a Vaccinia Virus Capping Enzyme to generate the “Cap 0” structure: m7G(5')ppp(5')G (New d BioLabs, Ipswich, MA). Cap 1 ure may be generated using both Vaccinia Virus Capping Enzyme and a 2’-O methyl-transferase to generate: m7G(5')ppp(5')G—2’-O-methyl. Cap 2 structure may be generated from the Cap 1 structure followed by the 2’-o-methylation of the 5’-antepenultimate nucleotide using a 2’-O methyl-transferase. Cap 3 structure may be generated from the Cap 2 structure followed by the 2’-o-methylation of the 5’-preantepenultimate nucleotide using a 2’-O methyl-transferase. Enzymes are preferably derived from a inant source.
When transfected into mammalian cells, the modified mRNAs have a stability of 12-18 hours or more than 18 hours, e.g., 24, 36, 48, 60, 72 or greater than 72 hours.
Example 8. Synthesis of N4-methyl cytidine und 1) and N4-methyl CTP g STP of said compound) -2l9- 0 o I I NH ‘ 1,2,4-triazole HO N O TBDMSC' TBDMSO N/go m. k o 9 azole $0; Eth, MeCN OH OH O OTBDMS \ CH N\ HN/ 3 | MeNH2 l N/go /& 40% aq. solution TBDMSO N O TBDMSO —> O :0: MeCN TBDMSO OTBDMS TBDMS'O OTBDMS HN/ 3 HN/CHa l 1) P(O)(0Me)3 \ N TBAFIHZO H0 & 2) POCI3 l N o ll 8 E —.-O*P*O*P*O*P—O N o THF 3) TBAPP l l l O 4) TEAB 0' 0' 0' 0H OH 4Et3NH+ OH OH compound1 hyl cytidine CTP C10H15N305 MOI. Wt: 257.24 Uridine was silylated to provide a trisilylated compound, which was purified by column, activated with re—distilled POClg/triazole under anhydrous condition, and then followed by nucleophilic substitution with 40% methylamine aqueous solution. N4—Methyl-2’,3’,5’-tri—O- TBDMS-cytidine was thus obtained after chromatographic purification. The resultant product was deprotected with TBAF and then purified with an l-ethyl acetate (3:1) solvent system to obtain compound 1. The final product was characterized by NMR (in DMSO); MS: 258 (M + H)+, 280 (M + Na)+, and 296 (M + K)+; and HPLC: purity, 99.35% (FIGS. lA-lD). HPLC, purity 98% (.
Exam le 9. S nthesis of 2’-0Me—N N-di—Me—c idine com ound 2 and 2’-0Me-N N-di—Me— CTP gNTP of said compound! //—N o o \N NH NH 1,2,4-triazole \ N | & LMSCL l POCIa,oc I A “O N 0 TBDMS‘O N o EtaN,MeCN TBDMS—O N o imidazole :0: o O 0H OCH: TBDMS’O °CH3 TBDMS’O 0C": H c3 ,CH \N 3 H30\ ,CH3 MezNH-HCI ‘ k \N E13N TBDMsfio TBAF/HZO l N o 0 N&O MeCN THF 0 ,0 OCH TBDMS 3 OH OCH: compoundz 2'-tri-O-methy|cy‘tidine C12H19N305 Mol. Wt.: 285.30 H3C\N/CH3 1) P(0)(0Me)3 \ N o o o 2)P°C's H H H l k ’ '0 F" 0 fi’ 0 F" 0 N o 3)TBAPP o 0- o- o. 4)TEAB 4Et3NH+ OH OCH: 2'-0Me-N,N-di-Me-CTP d by ion-exchange column, lyophilized purifed by e phase comlumn, lyophilized 2’-O-Methyluridine was silylated to give the di-silylated compound. Purified 2’—O— methyl-3 ’,5 ’-di—O-TBDMS uridine was activated with re-distilled POC13 and imidazole under anhydrous condition, followed by the nucleophilic substitution with dimethylamine hydrochloride under triethylamine environment to trap HCl. ediate compound N4,N4,2’-tri-O—methyl-3’,5’- bis-O-TBDMS uridine was purified by flash chromatography and obtained as a white foam. The resultant compound was de—protected with TBAF and then purified to provide ~400 mg final product compound 2 as white foam. ES MS: m/z 308 (NI + Na)+, 386 (M + H)+; HPLC: purity, 99.49% (FIGS. 3A-3C).
To synthesize the ponding NTP, 70 mg of side compound 2 provided 23 mg of2’-OMe-N,N-di-Me-CTP after purification Via ion-exchange and reverse phase s. HPLC: purity, 95% (.
Exam le 10. S nthesis of 5-methox carbon lmethox uridine com ound3 and 5- methox carbon lmethox -UTP NTP of said com ound o o NH I NH | N/go —',,”B'2, HO H0 ”0 N 0 —~ . H20 HO HO N/go H0 0 2)pyndlne O control ature HO OH HO OH HO OH 3'3 3-h 3-c 0 0 Ho HO I E m HO N O acetone HO N O INO\ NaOH or KOH “—0, o methoxy o m (control) '“.
Ho OH OX0 34‘] 3-9 Br \ O o O O HN O\)J\O/ HN 0%0/ 2x I O 3mm» 3 a R A ‘ N Ho —3, o N 0 Dillioiliofllio 3)TBAPP o 0' 0' 0- 4)TEAB H0 0H + HO OH compoundii 'M90COCH20'UTP ~methoxycarbony| methoxy uridine Uridine 3-a in water was treated with excess amount of bromine and then flushed with air to remove bromine. The reaction mixture was d with pyridine at a controlled speed and temperature. During the reaction, unstable bromo-intermediate 3-b gradually converted to di- hydroxyl intermediate 3-c, which ably dehydrated to the stable 5-hydroxyuridine 3—d. Then, the 5-hydroxyuridine was protected with a 2’,3’-isopropylidene group to provide compound 3-g.
Reaction with compound 3-f provided compound 3. 60-70 mg of the nucleoside provided >21 mg of the desired sphate after two HPLC colunm purification and two lyophilization steps. HPLC: purity, 98% (.
Exam lell. S nthesis of 3-meth l seudouridine com ound4 and 3-meth l seudo—UTP gNTP of said compound! \ HN NH N NH HO 0 0 \ \ A020. DMF MO o POMCI (9 equiv), AcO 0 —. 0 —. O Et N,3 ridine -30 C,DMAP FY HO OH pseudouridine AcO OAc AcO OAc 4—a 4.}, 4-c o 0 OANAN,Me O HNAN,Me \ \ DMF_DMA AcO o NH3-MeOH HO o o —. o AcO OAc HO OH compound 4 4-d 3—methyl pseudouridine 1) P<oy<0Me)3 HNAN e O O O 3 H H H pioipio o 3) TBAPP \ l l o 4) TEAB 0- O- O- 4 Et3NH+ HO OH 3-Me-pseudo-UTP ] Pseudouridine 4-a was reacted with AcZO to provide acetyl-protected pseudouridine 4—b.
Then, N] was selectively protected with POM to provide compound 4-c. Methylation ofN3, followed by deprotected, provided compound 4 (N400 mg). Molecular formula: C10H14N206, molecular : 258.23g/mol; appearance: white solid; storage ions: store at 25 OC; HPLC: purity, 98.51%; 1H NMR (DMSO'dg): 5 11.17 (d, 1H, J: 3.0 Hz), 7.56 (d, 1H, J: 3.6 Hz), 4.91 (d, 1H, J: 3.6 Hz), 4.79 (t, 1H, J: 4.2Hz), 4.70 (d, 1H, J: 4.2Hz), 4.49 (d, 1H, J: 3.0Hz), 3.82-3.88 (m, 2H), 3.66-3.67 (m, 1H), 3.57-3.61 (m, 1H), 3.40-3.47 (m, 1H), 3.09 (s, 3H); MS: 281 (M + Na)+) (FIGS. 6A and 6B).
] Alternative routes could be applied to obtain compound 4. For example, pseudouridine could be reacted with an O-protecting group (e. g., as described herein, such as TMS) and reacted with an N-protecting group (e.g., as described herein, such as acetyl at N1). Then, N3 of the nucleobase could be reacted with an alkylating agent (e. g., dimethylamine/dimethoxymethyl) to provide compound 4 having N- and O-protecting groups. Finally, the resultant compound would be deprotected (e.g., under basic ions, such as NHg/MCOH) to provide compound 4.
N/ 0H N/ OAC I HN OAc \ A A ] 1)POCI3 NH3 02x,“ O N fi. TrO TrCl 0 N TrO 0 W zole O H o o o o 0x0 X X 5-g 5-h 5-i NHAC NHAc N/ OAc l N/ OAc A020 0%N ‘ acid 4» TrO —» 0%N 7:0; :0: 0 0 X 5:J OH OH compound 5 Uridine 5-a was ted to obtain isopropylidene compound 5-b, which was reacted with (CHCO)... Acetic acid with catalyst amount of TFA was employed to obtain the desired selectively acylated compound 5-f (30% yield). Further tritylation of the 5’-OH group resulted in the desired orthogonally protected compound 5-g.
Compound 5-g was treated with POCl3 and triazole to e compound S-h together with de—acylated compound 5-i. Acetylation of these two compounds provided di—acylated, fully protected compound 5-j. Deprotection of compound S-j with acetic acid under heating condition resulted in three products, one of which was compound 5.
To obtain the ponding NTP, a triphosphate on can be conducted (erg, any described herein). Optionally, the NTP can be purified (e. g., using a Sephadex DEAE-A25 column), lyophilized, or evaporated (e.g., from EtOH).
Alternative routes could be applied to obtain compound 5, such as by ing with cytidine as the starting al. In such methods, the 5-position could be reacted with a halogen or a halogenation agent (e.g., any described herein, such as 12/ meta-chloroperoxybenzoic acid), which can be displaced with an alkylating agent. Further, such methods could include the use of one or more N- or 0-protecting groups (e.g., any described herein, such as silylation or acetylation) to protect the amino group of cytidine and/or hydroxyl groups of the sugar moiety.
Exam le 13. S nthesis of 5-TBDMS-0CH -c tidine com ound (HCHO)n n 0 H20,950 3days 6'~a 0 0 H0 NH ‘ 0 N/go TBDMSOfiKNH HO NH A00 N/go POCI; AC0 H l & 0 TBDMS-CI —‘V O 6'-b, 20 9 A00 N o trlazole OAc —, —> AcO OAC HMDS, heat AcO 0A6 ‘ AcO OAc 6'-e 5 g, 78% 6'4: 8+59, N\ B NH2 N TBDMS\O \ mums N \o \N i K I & NTP NHa-dioxane NH3—MeOH H0 N o 4’8 A120 N o O HO OH A00 0A0 compound 6 6'-f ] A 5-hydroxyuracil compound ’-b was glycosylated to obtain compound 6’-d (28% yield), which was silylated to provide compound 6’-e. Activation of the protected uridine ed the desired compound 6 after further amination and deprotection (800 mg of the final compound).
Molecular formula: C16H29N306Si; molecular weight: 387.50 g/mol; ance: white solid; storage conditions: store at 25 OC; HPLC: purity, 97.57%; 1H NMR(CDC13): d 7.81 (s, 1H), 7.40 (bs, 1H), 6.49 (bs, 1H), 5.79 (d, 1H, J: 2.4 Hz), 537532 (m, 1H), 5.007507 (m, 2H), 5 (m, 2H), 3.907394 (m, 2H), 3.807383 (m, 1H), 35073.70 (m, 2H), 0.87 (s, 9H), 0.05 (S, 6H); MS: 388 (M + H)+, 410 (M + Na)+) (FIGS. 7A-7C).
To obtain the ponding NTP, a triphosphate reaction can be conducted (e.g., any described herein). ally, the NTP can be purified (e.g., using a Sephadex DEAE-A25 column), lyophilized, or evaporated (e.g., from EtOH). e 14. Synthesis of luoromethyl cfiidine (compound :21 0 NH2 F 0 F30 1)TPSCI, DMAP, F3C NH 1 HMDS heat NH | & ‘ 2)TMSOTf, N/£0—>2)NH3,MeOHMeCN 2144,, N O roethane O ONLO H (overnig h t) heat 0 OAc AcO 7'A Aco’jj OAc AcO 0Ac HO OH 7—B nd 7 Compound 7-A was glycosylated to provide compound 7-B, which was treated with 2,4,6- triisopropylbenzene sulfonyl chloride (TPSCl) to activate the carbonyl group and to promote reductive amination. Deprotection provided compound 7. Alternative activating agents could be used instead of TPSCl, such as 2,4,6-trimethylbenzene sulfonyl chloride.
To obtain the corresponding NTP, a triphosphate reaction can be conducted (e.g., any described herein). Optionally, the NTP can be purified (e. g., using a Sephadex DEAE—A25 column), lyophilized, or evaporated (e.g., from EtOH).
Exam le 15. S nthesis of 5-trifluorometh luridine com ound8 0 F30 NH F30 F30 - NH 1 HMDS hea l & lN/EOZ)dichloroethane, l k ),TMSOTf A00 §£3mhiilgi?H N 0 HO N 0 heat 8 A_ O OAC ACO OAc ‘ HO OH AGO OAC 88 compound 8 S-Trifluoromethyluracil 8-A was glycosylated with O-acetyl ribo se, and the desired triprotected 5-trifluoromethyluridine 8-B was obtained in good yield. r deprotection gave desired compound 8, which was characterized with NMR, MS and HPLC results. MS: 313 (M + H)+, 335 (M + Na)+; HPLC: purity, 98.87%, ((FIGS 8A-8C).
To obtain the corresponding NTP, a sphate on can be conducted (e.g., any described herein). Optionally, the NTP can be purified (e. g., using a Sephadex DEAE-A25 column), lized, or evaporated (e.g., from EtOH). e 16. Synthesis of 5-]methoxycarbonyllmethyl uridine {compound 9] 0 0 Br 0 O HN )J\ Br 3": | HN ‘ A l Ph N A00 0 | N t3N HO Ac20 0 ammo N AcO CAN 0 A00 CAN H py o Bz-Cl 0 AGO... 930/° 9 50% AGO 0A0 0“ 0” Aco OAc Aco 0AA: 9-c 9—d 9_a 9-b o o o\ o o\ OCH3 o o ‘ NaOMe o 0 H0 02‘»: NTP \ 0 0 j/Bro\l o MeOH, heat k 9 DBU,THF O Dowex—SO (H ) OH OH 50% compound 9 hoxycarbonyl)methyluridine 9'6 (MCM5U) Uridine 9-a was protected to provide compound 9-b (98% yield). This compound was brominated with excess bromine in the presence of acetic anhydride and acetic acid. The 5-bromo analog 9—c was obtained (60% yield) and further benzoylated to provide desired compound 9-d (64% yield). S—Bromo compound 9-d was condensed with dimethyl malonate under basic condition to give the arylated malonate and the fully protected diester 9-e (50% yield). After de-carboxylation and deprotection, compound 9 was obtained verified by NMR (.
To obtain the corresponding NTP, a sphate reaction can be conducted (e.g., any described herein). Optionally, the NTP can be purified (e. g., using a Sephadex DEAE—A25 column), lized, or evaporated (e.g., from EtOH). und 10! O o o 0 :15 Br » HEY “0 N DMAP,Et3N PNT HO 0 B'Z'A°2° N Ac20 AcO 0AM 00 AcO o N 0 O Bz—Cl o AcOH AcO o OH 0 98% Ace o \ Aco Me 0\ \Me \Me Me 1°'c 10'“ 1o-a 10-b o o o\ OCHz \ HN ° ° A ‘ PhAN o 0\ HO 0 0 N \ NaOMe O O ‘ —.NTP Aco 0 o N MeOH, heat DBU, THF 0 Dowex—SO (H+) 0H 0 Aco °\Me 5-(Methoxycarbonyl)methyl-2'-O- -e Methyl uridine (2-OMe-MCM5U) compound 10 r gy to the synthesis of compound 9 above, 2’-O-methyluridine lO-a was acylated and brominated to obtain compound 10-c. Further lation provided 5-bromo analog lO—d, which was condensed with dimethyl malonate provide the desired product 10-e (45% yield).
Decarboxylation and deprotection provided compound 10.
To obtain the corresponding NTP, a triphosphate reaction can be conducted (e.g., any described herein). Optionally, the NTP can be purified (e. g., using a SephadeX DEAE—A25 column), lyophilized, or evaporated (e.g., from EtOH).
Exam le 18. S nthesis of 5-trifluoroace l-aminometh lthiouridine com ound ll o o Aco i I HNJE ><O :0: ,OAc — :1 1) HMDS l + NaOMe A60 5 N N HO 3 s N 2 SnCI4 0 0 or choa TsOH. acetone H A°° °A° or NHa-MeOH DMF 11-a 11'b °’ “OH Aco OAc OH OH 1 1 -c 1 1 -d O 0 If O HNI) HN OH eq Hi 8 N 3 N HO HO S N NaN3, 6 eq (HCHO)n HO 0 0 heat 0 NaOH DMF, heat heat w 0 o 0 0 X X o o 11 -e 11-f 11_g | N3 SN 31%,le <CF3CO 20) acid HNJjANJkCRA 1) PPh3, heat 5 N o o 2) NH4OH 0X0 OX0 OH OH 1 1 -i -TFA-amlnomethyl(hIoundlne_ . . . 11-h compound 11 Glycosylation of 2-thiouracil ll-a provided compound ll-c, which can be deprotected with any useful deprotection reagent. In particular, LiOH provided d product 1 l-d (80-90% yield). Isopropylidene protection provided compound ll-e (90% yield). Further 5- hydroxylmethylation provided compound ll-f. nation, azidation, and further reduction provided methylamine compound ll-i, which was acetylated to provided compound 11.
To obtain the ponding NTP, a triphosphate reaction can be conducted (e.gi, any described ). Optionally, the NTP can be purified (e. g., using a ex DEAE—A25 column), lyophilized, or evaporated (e.g., from EtOH).
Exam le 19. S nthesis of 5-meth meth luridine com mind 12 o o /CH3 (i ) TMSCI HN N HN OH HN CI ‘ Seq..heat ‘ A [ H HCHO —’ o N Uridinefl-L 120(—b N O N excess HO HO O HO ’ O o HCI-dioxane o 12-a :aOH HQNME °><° °><° °><° 12-c 12-a 12-e lzo%AcoH/heat l O 0 0 CH N/ 3 HN CI HN OH H A l A l excess CAN 1 N 4. 0 N 4 0 H0 _ HO 0 o o HZNMe OH OH OH OH OH OH 12-f compound 12 12.9 (ii) 0 O O 0 l I HN OH Br AcO HN ‘ a I 0 ‘ HN/ujAN/CHEH OAc fl 0A” AcO CAN 4. AcO CAN Ho o 0 0 HMDS,heat Mo 0A: % F‘ l? ?‘ AcO OAc 0” 0” AcO OAc 12-h 12-i 12-j nd 12 Compound 12 can be obtained by any useful method (e.g., see schemes (i) and (ii) above).
For example, protected uracil can be glycosylated and subsequently aminated to provide compound 12. Additional protecting, deprotecting, and activating steps can be conducted as needed. To obtain the corresponding NTP, a triphosphate on can be conducted (e.g., any described herein).
Optionally, the NTP can be purified (e. g., using a Sephadex 25 column), lyophilized, or evaporated (e.g., from EtOH).
Exam le 20. S nthesis of 5-TFA-meth laminometh luridine com ound 13 O o HN OH H/l l | '21)] 0%jjfl TMSCI O N Ho 0 N (HCHO)n Seq‘ Ho — —. 0 —.
O 0 NaOH heat heat 0 O OH OH OX0 X 13-a 13-h 13_c o 0 IE: CI I WN/ 3 H 0 N EXCESS HO O N 0 HZNMe 0 0X0 0 0 13-6 13-d \ o O CH CH 3 “N N/ r I oékN I “i HO oékm:3 0 N HO CF3 o O o O OH OH X 13-f compound 13 Uridine l3-a was ted with pylidene to provide compound l3-b and then 5- hydroxymethylated to provide compound l3-c. Chlorination and subsequent ion provided compound l3-e, which can be protected to provided l3-f. Subsequent deprotection provided compound 13.
To obtain the corresponding NTP, a triphosphate reaction can be conducted (e.g., any described herein). Optionally, the NTP can be purified (e. g., using a ex DEAE—A25 column), lyophilized, or evaporated (e.g., from EtOH).
Exam le 21. S nthesis of 5-carbox meth laminometh luridine com ound 14 o o lo ”fro | HN N* N ofifo formaldehyde O HO HOFOO:r: #mw‘ HO Mel pyrrolidine K 0 # W OH OH 0x0 OX0 144: 14-0 ”IN/FOX Griffin/7f % (cacon OHANAwe¥o CF3 14-1 OH OH compound 14 Uridine l4—a was protected with pylidene to provide compound 14-b and then 5- aminoalkylated with the Mannich reaction to provide compound 14-c. Methylation provided quaternary amine l4-d. Subsequent amination and deprotection steps can be used to provide compound 14. To obtain the corresponding NTP, a triphosphate reaction can be ted (e.g., any described herein). ally, the NTP can be purified (e. g., using a Sephadex DEAE—A25 column), lyophilized, or evaporated (e.g., from EtOH). e 22. ative synthesis of 5-methylaminomethyl-Z-uridine (compound 12! and 5- carbox!methylaminomethyl-Z-uridine geompound 141 o O HN Br NH ”N l | CAN \ NBS,AIBN N TBDMSO A H0 N/ko TBDMSO chlorobenzene 00 0 ° TBDMSO OH OH TBDMSO 0TBDMS COTBDMS A B J 0 /Me O HN N HNfiH/TN \6 H TBDMso 09m l TBDMSO O N O TBDMSO OTBDMS TBDMSO OTBDMS l l compound 14 HN N/Me A t H HO 0 N HO OH compound 12 ] In addition to those strategies provided above for compounds 12 and 14, the following strategy can also be implemented. S-Methyluridine A can be silylated to provide compound B.
After radical monobromination, the resultant intermediate bromide C can be used for the preparation ofcompound 12 and nd 14 analogs. Subsequent alkylamination of bromide compound C could provide nds D and E, which can be deprotected to provide nds 14 and 12, respectively. To obtain the corresponding NTP, a triphosphate reaction can be conducted (e.g., any described herein). ally, the NTP can be purified (e. g., using a Sephadex DEAE—A25 column), lyophilized, or evaporated (e.g., from EtOH).
Exam le 23. S nthesis of dimeth [- gNTP of said compound! N 1) P(0)(0Me)s 2) POCl3 fl) (‘3‘ 9‘ ' \ \ -O—P—0—P—O—P—O HO o o 3) TBAPP o 0' 0' 0‘ 4) TEAB 4 EtsNH+ HO OH HO OH dimethyl pseudouridine di-Me-pseudo-UTP compound 1 5 Nucleosides can be phosphorylated by any useful method. For e, as shown above, nucleosides can be reacted with phosphorus oxychloride and subsequently treated with a monophosphate intermediate with bis(tributylammonium)pyrophosphate (TBAPP) to give the triphosphate.
Exam le 24. S nthesis of 2’-C-meth ladenosine com oundl and eth [ATP NTP of said compound! 320 820 320 OBz OBz 320 . . 0BzDess—Mamn penodane 052 O O O /TiCI4 BzCI O —. —> 032 OH OBz O 032 OH 032 032 '3 1&1 16-2 16'4 NHBz NH2 NH2 N \ N N N6-pivach </ f3,“ </ f)“ 1) PY, Proton sponge </ l . / aden-ne N \j‘l N NHEZ N N/ .21IME_._F299I3 Hofifioflf'ofifio N N/ DBU. 320 O MeOH HO 0 3) ylammonlum R R I? o TMS—lriflate, pyrophosphate O O O : _ _ CH3CN 032 {:32 OH 5H '3“an TEAB 0H EH 16-5 compound 16 NTP of compound 16 About 5g of compound 16-2 was ed from 5g of compound 16-] via a Dess—Martin periodane reaction. Compound 16-2 was reacted with MeMgI/TiCl4/—78°C to provide compound 16-3, and crude compound 16-3 (6g) was directly reacted with benzylchloride to prepare compound 16-4. Reaction with the nucleobase and deprotection provided compound 16 (0.56g).
Exam le 25. S nthesis of 2’-C-meth l-c idine isomers com ound 17 and com ound l8 and 2’-C—methyl UTP 1 STP of said compounds! NH2 NHBz NHBz \N NHEz I N’go \ N ‘ \N ‘ k \N Ho 3220, DMF HO N 0 TIDPSCI N 0 1 o —> 2 DMSO, TFAA m, o 0 N’KO DMF I TEA o 0 °“ 0“ SIep 1 OH OH Slepz T'DFs\o OH Step3 TIDp’k 0 o 17-1 17-2 17-3 ‘74 NHBz NHBz [K NHEz NHBz \N \ N \N MeMgl b\N N’go TBAF ‘ N/go + N’go —’ fM’go o o o AcOH I ? HO 0 HO 0 Slep4 TIDPS\ 0” ; TIDPS\ s Step5 ; OH o 0 .
OH 2 = 17-5a OH OH 17-5b 17-6a NHZ 17—sn \N \N NHZ l ‘ 1)Py proton sponge NH? NHyMeoH «so + NAG alwreda (1 H0 OH 0 ulylammonium 71% Vi.OH0 :1 OHo OHo O HO pyrophosphate O\P/O\P,O\P['13 N O + ’O‘P,oOHo 0H0 I”); Ste 5 : 11 : Bu N,TEAB 9 OH : OH OH 3st o 0 god 7 8mm compounaw compound18 OH - OH OH NTP of compound 17 NTP ofcompound 18 About 17.4g of compound 17-3 was prepared from 20g of compound 17-1. Then, 2’- ion and alkylation with MeMgI provided 300mg of compound 17-5a and 80mg ound 17-5b. About 9g of compound 17-5a (about 90% pure) and 2.1g of compound 17-5b (pure) were prepared from 17.4g of compound 17-3 in 2 batches. N— and O-deprotection provided compounds 17 and 18.
Exam le 26. S nthesis of 2’-C-meth l uanosine com ound 19 and 2’-C-meth lGTP TP of said compound) Bzo 520 520 DB: 0B2 B20 oBzDes.s.—Martin periodane 052 0 O MeMgCVTiCL. O B201 0 05129_1OH 052 O 052 OH 19-2 19-3 (:52052 '50149urine c\lN/)‘NH2 DBU. TMS-triflateBZ HCI Ho 04%)‘NH2 1)Py proton sponge2 TMP POCI2 __________ a Ho 9H0 9H0 orb CH CN 3) tnbutytammomum ‘p’ ‘p’ ‘p’ 3 oLN \N)‘NH2 1 ‘1 pyropnosphate 5 ('5 (‘5 0132 0 OH OH BU3NI TEAB '; 19-53 compound 19 OH NTP of compound 19 ] 2’-Oxidation of protected ribose 19-1 and subsequent tion with MeMgCl provided compound 19-3. The resultant compound was further protected to provided compound 19-4, and 1.56g of compound l9-5a was prepared from 3.1 g of compound 19-4. Subsequent oidation and deprotection provided compound 19 (about 90% pure, 50 mg).
To obtain the corresponding NTP, a triphosphate reaction can be conducted (e.g., any described herein). Optionally, the NTP can be purified (e. g., using a Sephadex DEAE-A25 column), lyophilized, or evaporated (e.g., fiom EtOH).
Exam le 27. S nthesis of 2’-C-meth l uridine com ound 20 and 2’-C-meth l UTP NTP of said compound] B20 820 BzO 032 082 820 OBz OBz _ 0 MeMgCI/TICL. O 0 BZCI artin 0 periodane ’ 032 O 052 0H OBZ OH OBZ OBZ -2 20-3 204 -1 0 o ~ NH NH Uracul, DBU, | N/go NH 6kN/go NH I 1) Py, proton sponge TMS-triflatg I & 0H 0H OH CH3CN BzO MEOH AIME-WES o HO 0 HO\ ‘ /O\ t ,Ou ,0 N 0 3) tributylammonium E If" ‘F" o '2 g pyrophosphate o O O OBZ 6B2 0“ OH Bu3N, TEAB OH 5 -5 compound 20 NTP of compound 20 2’-Oxidation of protected ribose 20-1 and uent alkylation with MeMgCl provided nd 20-3. The resultant nd was further protected to provide compound 20—4.
Reaction with uracil and deprotection provided pure compound 20 (50mg).
To obtain the corresponding NTP, a triphosphate on can be conducted (e.g., any described herein). Optionally, the NTP can be purified (e. g., using a Sephadex 25 column), lyophilized, or evaporated (e.g., from EtOH).
Exam le 28. S nthesis of S -2’-C-meth ladenosine com ound 21 and S -2’-C—meth lATP ]NTP of said compound] 3f” MHZ N \N N \ Hon N NA o <N6lN/J Cr03.A(>ZO <Nn lN/J )3 O ° 0 pyridine, DCM OH ‘ TIDPS\ 0 TIDPS\ 21-1 OH21-2a 0 o 21-3a HNJLPh )k HN Ph [i] MeMgl N \ [H] Old —’N \N MeMgl /f\N N <N ‘ \N TBAF a —’ </ I a —— N N/ N N N o (I) O T DP IDPS\ OH I S\O () IDP O 21-3b S\o 21—4a 21.4 N \ N </ I N \ N N </ l N _1_)_Ify,_P_rgt_or]iponge HO 9H0 9H0 9H0 N HO 0 N/) 2)TMP,POCI3 ‘fi‘ \fi“ ‘fi‘ 0 _; OH 3) tributylammonium O O O . OH 0H : osphate OH é coqund 21 Bu3N, TEAB NTP of compound 21 step 7 Compound 21-] (5g) was protected to form compound 21-2a, and chromium ion provided compound 21-3a. Alkylation via route [i] (Seq. MeMgI in ether at -500C) provided compound 21-4. Optionally, yield could be improved via route [ii] by protecting the amino group to provide compound 21-3b and then alkylating at the 2’-C position to e compound 21—4a. nd 21-3a was alkylated to provide crude compound 21-4 (3g, 20% of compound 3a in this crude t), where the product can be optionally purified. Deprotection of compound 21—4 afforded compound 21 (50% yield).
] To obtain the corresponding NTP, a triphosphate reaction can be conducted (e.g., any described herein). Optionally, the NTP can be purified (e. g., using a Sephadex DEAE—A25 column), lyophilized, or evaporated (e.g., from EtOH).
Exam le 29. S nthesis of S -2’-C-meth l uanosine com ound 22 and S -2’-meth IGTP (NTP of said compound) o o o <Nf1;\ N N N / <N ‘NA/NfLNH </ fiNHNANHBZ NH </ l A Ho Me SiCI MeasiO NH Measio HO N 2 N NHBz O NH23—> O BzCI O depmtect o OH OH MeQSiO OSiMea Messio OSiMea OH OH 22-1a 22-2a 22-2 0 o o N N NH NH NH / I A TIDPSCI <’ f: Bess—Martin periodane < l A MeMgl TBA; —’ N <N/ N N/ N NHEz —. N NHBz _, NHBz DMF (I) o TEA <17 0 ‘1) O }/—\f 7;} AcOH . OH TIDP13\0 TIDPS‘0 TIDPS\ s 0H o 0 22-3 224 22-5 0 o O 8 ljf 816£T m 1wy, proton Sponge 8 / NH MeOH | N 3' N NHBz N . N/ [i 02 NH2 .211'ME_RQ§2|3..- O\\O:"O\<‘)E‘O N NANHZ HO HO 3)tributylammonium R fi E O OH osphate O O O : 7:0: _ _ OH 0H 0H : OH 3 BuaN,TEAB OH 5 22'5 compound 22 NTP of compound 22 About 30g of compound 22-1 was silylated to provide nd 22-2 in three steps.
Further protection provided compound 22-3, and Dess—Martin periodane oxidation provided compound 22-4 (1.6g) in two batches. 2’-C alkylation (Seq. MeMgI in ether, -50°C to RT) provided compound 22-5, and further ection steps provided compound 22.
To obtain the corresponding NTP, a triphosphate reaction can be conducted (e.g., any described herein). Optionally, the NTP can be purified (e. g., using a Sephadex DEAE-A25 ), lyophilized, or evaporated (e.g., from EtOH).
Exam le 30. S nthesis of S -2’-C-meth luridine com ound 23 and of S -2’-C—meth lUTP QNTP of said compound! O \ O w Nu, ““ \\ TIPDSCI N 1“ HN1 0H 2 N”go j Z Bess-Martin N O MeMgBr ' /O O periodane TIPDS/O O HO OH TIPDS\O \ OH 0 23-1 23-2 23_3 6w 1 l & I! NH N O 1) Py, proton sponge I N o ect HO 0 o Q5O _g)__TME’_,_Fjog3 HO\\Ol-|O\‘OleJOl-‘O NAG TIPDS/ 3) tributylammonium R R R 0 HO 5 0H pyrophosphate O O O BuaN, TEAB OH : nd 23 23-4 NTP of compound 23 e 23-] (2.0g) was protected with TIPDSClz (l ,3-dichloro-l,l ,3,3- tetraisopropyldisiloxane) to provide compound 23-2. Oxidation provided compound 23-3, and 2’-C alkylation ed compound 23 -4, which can be optionally purified with Prep-HPLC prior to the next step. Then, deprotection provided desired compound 23.
To obtain the corresponding NTP, a triphosphate reaction can be conducted (e.g., any described herein). Optionally, the NTP can be purified (e.g., using a Sephadex DEAE—A25 column), lyophilized, or ated (e.g., from EtOH).
Exam le 31. S nthesis of 4’-C-meth ladenosine com ound 24 and 4’-C-meth [ATP NTP of said compound) H0 H6 24-1 24-2 24_3 o ~do H0 +00w AcOH m" ‘°><. NaIO4 HCHO c —* . —> ~ ”0 HO . "'o 1 : MeOH °\\_<j:>< NaOH BnO 24.4 BnO 24-5 and 24-6 0 MO BnBr HOHow >< 4» tributyltin hydride _ Bno:>Oi,::>< 0 Bn8:><j:l::>< 5 NaH AIBN Bno Bno BO" 248 249 ..\o , , o ..o o Blow ,OXPd’CH2v00X' E, HCI 82OM fi.
Bno BZO 24—12 24—10 2411 BzHN 0 .“0H o .nOBz 4 \ /) 820' 320%; 820 N B ow2 a 6 NHBz Ad- - o NH3 .,,OH OBz —. ___________ > :- “H. M OH BZO DBU e 2443 24-14 TMS—Triflate 032 032 CH3CN 2445 H2N H2N \g 1) Py, proton sponge N \r‘i N QIMEFIQQ'a Q N/ HO 9H0 9H0 9H0 0 N 3)tributylammonium ‘P’ \P’ ‘P’ o “' pyrophosphate 6 5' (‘5 5 OH OH BU3N,TEAB OH OH compound 24 NTP of compound 24 l,2:5,6-Di-O-isopropylidene—(x-D-glucofuranose 24-1 was converted via sequential oxidation, reduction, and protection steps to provide compound 24—4. The first ion step to provide compound 24-2 can be implemented with any useful reagents, such as 0.75 eq. pyridinium mate (PDC) with 1 eq. AcZO or 1.2 eq. of Dess-Martin periodane. Subsequent deprotection, formylation, and reduction provided compound 24-7, which was followed with protection and deoxygenation steps to provide compound 24—10. About 0.4g of compound 24-14 was prepared from lg of nd 24—10 via sequential protection and deprotection steps. Addition ofN6- benzoyladenine and uent deprotection provided compound 24.
To obtain the corresponding NTP, a triphosphate on can be conducted (e.g., any described herein). Optionally, the NTP can be purified (e. g., using a Sephadex DEAE-A25 column), lyophilized, or evaporated (e.g., fiom EtOH).
Exam le 32. S nthesis of 4’-C-meth lc tidine com ound 25 and 4’-C-meth lCTP said compound! o o o P00 ° ° o 1 0 0 Enc' ‘f\d:‘ >< NaBH4 '~o _____. —————+ ~ \ °>< --* o ,0 0 1 .« 0 NaH H0 H5 251 25'2 -3 0x00 .50 Ho 0 AcOH “‘°>< Na|04 0\ 0 -"° +6 HCHO :- HO :- MeOH ’0 : NaOH BnO 25.4 Bno 25-5 and 25-6 \ tributyltin '0 "’0 (S __. :- AIBN Bn BnO 258 3'10 25-9 -7 O 5‘0 o “‘0 OKWC''H2w,, no BzCl ,1 HCI *> Elm/«Jon 4’ Bnd BZG -10 25-11 25-12NHBZ o .1082‘ , o ,, ‘ 0Hu Kim BzCl 4-NHBz-C 320 BZO N/ko BzO 4. 4» NHVMEOH . "OH OB: : 5 DBU \ B (3 820 ' z . 2543 2544 amasctflflate GB: 082 2515 NH2 NH2 \N 1)P rotons on e \N l V P P 9 | H0 (Niko 2)_TMF3 309's. . HO\9%\9L—b-9% N’go O 3) tributylammonium fi f? E 0 w“ pymphosphate O O O 5‘ OH OH Bu3N,TEAB OH OH compound 25 NTP of nd 25 Similar to the gy provided above for compound 24, compound 25-14 was produced with compound 25-1. Addition of cytidine and subsequent deprotection provided compound 25.
] To obtain the corresponding NTP, a triphosphate reaction can be conducted (erg, any described ). Optionally, the NTP can be purified (e. g., using a Sephadex DEAE-A25 column), lyophilized, or evaporated (e.g., from EtOH).
Exam le 33. S nthesis of 4’-C-meth l uanosine com ound 26 and 4’-C-meth lGTP NTP gigflfliaunngunfll o o P00 ° ° i 0 BnCI +00W >< NaBH4 w,0 —> : —> +0OW“¢ —> o‘ , "’0 ”0 NaH 0 HO 26-1 26-2 26-3 0w“::><AcOH How.“0 Na|04 0\ O “‘ +0 HCHO ----- b ,”05 M OHe "’0 0‘ :' NaOH Bno 254 50” BnO 26-6 H831“:NXHHBnO:B>§J2f:BnBr M ano:><j:>< tributyllin hydride.
AIBN Bno ENG 26-7 '><Pd/CH2@OX *>\0 z, 0 .uo ' BzCI HCI BnO 820: .,,2:: 4. 2510 26-11 <’:fl)’: , 0 “0H \AJ BzCI 0nos: NH2 82o o<’:IEINH2 BzO fi’ B OwZ "'OH ”OB: ------------ \_ 325 \w ,Wlifi, . MeOH .13 26-14 TMS—Trlflate 032 032 CH3CN 0 02645 (IN I JN\H 1)Py, proton sponge Ho N QZIMEE’QQ'a o o o NNS:/ N NH2 NH2 0 3) ylammonium v“ pyrophosphate u u H 0 0 0 ,~ OH OH Bu3N,TEAB Ho‘fitb‘fitb‘fitbjg OH OH compound 26 NTP of compound 26 Similar to the strategy provided above for compound 24, compound 26-14 was produced with compound 26-1. Addition of 2-aminochloropurine, subsequent oxidation, and then deprotection provided compound 26.
To obtain the corresponding NTP, a triphosphate on can be conducted (eigi, any bed herein). Optionally, the NTP can be purified (e.g., using a Sephadex DEAE—A25 column), lyophilized, or evaporated (e.g., from EtOH).
Exam le 34. S nthesis of 4’-C-meth luridine com ound 27 and 4’-C-meth lUTP NTP of saidcomound OHIZKPCC_> g’:><NaBH4 jig—Q‘WOK %o no 27—1 27-2 27.3 ‘ MeOH ’0 NaOH BnO 27-4 BnO 27-5 BnO 27-6 ng:N><4>Hanow:>< tributyltin B Bn r lz__ Bno:><j:>< hydride' AlBN BnO BOn6 BO“ 27_3 27-9 27—7 w:XPd/C-”2”@m)< , o ‘0 —.Bz°' B20%; L ”0 no BnO 2 27-10 27-11 27420 0 ,.‘0H o 03z l ., BzCI 52° B Ow uracil N’go NH 820 —’ z 0 . "’OBz : DBU 320 820 MeOH 27-13 27-14 39.36?be 082 GB: 3 27-15 NH 1)Py. proton sponge l NH Ho NKO ngME’fQE'i I 0 3) tnbutylammonlum L,FI,0Fl”OOH orborb ,u Fl” N 0 w“ 0 pyrophosphate a ICI) : Bu3N.TEAB 5 OH OH OH OH compound 27 NTP of compound 27 r to the strategy provided above for compound 24, compound 27-14 was produced with compound 27-1. Addition of uracil and subsequent deprotection provided compound 27.
To obtain the corresponding NTP, a triphosphate reaction can be conducted (e.g., any described herein). Optionally, the NTP can be purified (e. g., using a SephadeX DEAE-A25 column), lized, or evaporated (e.g., from EtOH).
Exam le 35. S nthesis of 2’-0 4’-C-meth lene adenosine com ound 28 and 2’-0 4’-C- meth lene ATP NTP of said com ound o o o 0 \‘ ><pc>c +0 ‘ ""0 —» +6 ”/0 _.0+Na__BH.. om”o__><BnCHI”0: 3+6 v><uo 0 H5 Bn01 23-4 231 23-2 23-3 “o CLO, no AcOH Na|04 o ~"\ \ >< _, HOHCHO H030 MsCl/pyridine MeOH "’0 ; NaOH “ BnO 20'5 and B"° 287 28—6 “5:190 M50 TFA, 30% Mson ”lawman Msojma OBn 01703" OBn OH 30% 0A5 23-3/k 23.9 23-10 0 NAN “”32 BzHN GNVJ' / N gCNS «96 M50 M50 N LiOH/THF/HZO k0? DMF BSA,TMSOTf,MeCN, 33% “.50ij HOAc ‘\ 30% OB" 70% 0 OAc 23-11 BnO 23-12 </:fj' qf/JN 9 9 9 <’ | NH40HIMeNH2 2/C/HCOONH4HO N H07F707?707|‘370 “ """""* ’ HO\O EtOH/H o, 90% Ho/\oo OH OH OH o 2844 compound 28 NTP of compound 28 Similar to the strategy provided above for compound 24, compound 28-7 was produced with compound 28-1. Subsequent mesylation, deprotection, and acetylation provided compound 28- , which was followed by addition of N6-benzoyladenine and subsequent internal atjon.
Various protection and deprotection steps provided compound 28.
Exam le 36. S nthesis of 5-meth l-2’-0 4’-C-meth lenec tidine com mind 29 and 5- methxl-Z’-0,4’-C-methxlene CTP {NTP of said compound! Mso MsO HO M50 ° 0 T: o 0 OAc o M50 u o .MS.°.'!P¥EI91"£.Mso o .Tfétfifl‘tt- M50 0H .C‘F39’??."FJU?.-__ HO OE" 03" ________________________ . can 0"— 03“ 0"— 0“ 30% °A° OTLMeCNfiZ‘M» 29'3 29'4 29-1 29-2 v o o o ‘ NH N’KO MNrrrCI,pyrininsv|MTro IN’go VVVVVVVVVVVVVVV, M504 L? HOAC (#0?) "."'*‘..k0?’ 'Q'C 05" v 0A0 87% 0 0E0?! 51995 Brio/\o 0 29—5 29-6 29-7 N1 29—8 29—9 0 ""310 (N'N "HI NH ‘N | \ I 510 N’koflf’orqaw. 520 I N10 fl. “10 "A” “Q o [:1 1,2,44nazole L?! 2% /o a o 29—10a B"Ozsa-liaa ’\° B"° N—\ 0 0 '“ W) “*0 2 Pd/C H2 \N | *0 I N’KO ,,,,,,51,C,(,)?,,,,,MMTro MeOH 29-12 #0 29-130_ \fii HO—P—O—fi—O—I‘IDI—D N 0 "50?) "0 NTP of compound 29 compound 29 Aldofuranose compound 29-1 was reacted via various protection steps, and then 5— methyluracil was added to provide compound 29-5. Subsequent internal cyclization, deprotection, tion, and amination steps provided compound 29.
Exam le 37. S s of 2’-0 4’-C-meth lene uanosine com ound 30 and 2’-0 4’-C- meth lene GTP NTP of said com ound M50 Mso Ho-JQO 0 o W .WQ'PYE'S‘YP.Mso 1553929.- M50 0“ .53295’2’53‘555, M50 oa'1 OH 05 an0°10— 580%3 " on 03,1 0,!— S‘ew Step2 -1 30-2 30-3 '5” 30-4 CI 0 1,, N N \ \N N NH NH ”\/)\ “A “A H 4 ””2 N N M50 N N NH: NaOEzJDMF Ele N N “’50 H2 N NaOHITHF/HO ------------------------ - o W-> 0 ............._, 0 BSAJn/Isornocam M50 HOAc 90% _ \ 30 5 # 30 7 Sb“ 03“ 55% 0 30'6 ° 0A5 BnO/ Shep?» ano Steps 0 o </N ”H | ”” </ I N <3 <3 a? EtOHIPyridine NANH Ho NAN Ho ‘NANHZFd/CJHCOOHIMeOH 2 ”2 O HO‘E'O'T'O‘E'O , 0 ----------------------- - 777777777 ’ OH OH OH HOAc. 88% ‘ 50% 30_8 /\0 Noreferenoe steps H0 Steps) compound 30 NTP of compound 30 Similar to the strategy provided above for compound 29, aldofuranose compound 30-] was reacted via various tion steps, and then 2-aminochloropurine was added to provide nd 30-5. Subsequent internal cyclization, amination, and deprotection steps provided compound 30.
To obtain the corresponding NTP, a triphosphate reaction can be conducted (e.g., any bed herein). Optionally, the NTP can be purified (e. g., using a Sephadex DEAE—A25 column), lyophilized, or evaporated (e.g., from EtOH).
Exam le 38. S nthesis of 2’-0 4’-C-meth lene uridine com ound 31 and 2’-0 4’-C- meth lene UTP NTP of said com ound 0 0 o _o 0 m0 ° ° ° 3"“ c K PDC #6 "'0 —* °K “a“ °W“>< ”*0 K ov‘ —’ $6 ”no —*7Lo~‘ "no a ""0 HO H6 and 0 314 31-1 31-2 31.3 Ho 0 .mo 0 mo O AcOH w Kfl» 0%.“on yndlne MSO‘MO HCHO 3830 >< HO "7/0 "”0 OBn \» ; . ""0 O \ MeOH ~ C N OH3 BnO 31-5 BnO BnO 31-6 31_7 31-8 / 1: M50 Ms0 0 | 0 0 OAc {km 0 _ _ E N/KO TFA 80% M50 OH A020/pyndlne M50 0 03,. 0Bn OH 80% OAc SOTf,DCE,75% M50 31-9 31.10 OBn 31-11 6NH NH ‘ fi NH N/go N/go ‘ M o N OBz/DMF BZO ’3 NaOMe/THF1H20 5 w a HO N 0 0 w0 \Ho HOAc 1/\ §/\ .. o O \ BnO 3'10 31-13 BnO/ 0 31-14 31-12 0 O NH NH ‘ 0 o o ‘ PdICIHCOOH/MeOH HO N/gO HO,‘F‘,,O,E,O,E,,O N’go E --------- -> O w w w OH OH OH 0 No reference v" Step 8 / 0 Step 9 O 0 compound 31 NTP of compound 31 Similar to the strategy provided above for compound 24, compound 31-7 was produced with compound 31-1. Subsequent mesylation, deprotection, and acetylation provided compound 30- . Addition of uracil and subsequent internal cyclization provided compound 31-12, and various protection and ection steps provided compound 31. A subsequent sphate reaction (e.g., as described herein) provided the NTP of compound 31, which can be optionally purified (eg., with HPLC).
Exam le 39. S s of 2’-chloro adenosine com ound 32 and 2’-chloro ATP NTP of said compound! NH2 N WIN? (/N ‘ \j Tfo<(5an 2/ imidazole ::r\9 o DMAP/Tf20 PK:1 /Si 'Pr/ 1 3 Dichloro1 1 3 3—'P O\Si DCM/70/o0 0 tetraisopropyldisilox SO5' 0H p 2 ane/DMF, 95% ”Dr/ \_ iPr/ \ Step1 'Pr Pr 32—3 32-1 32—2 NH2 NH2 N \ </ N N \N 1 a </ 1 N a o N TBAF/THF HO N N V LICIIDMF P[\s‘i o 0 T”3/6 80% o ‘510 CI Step4 OHCI Stew / \ ‘Pr ipr 32_4 compound 32 Hoipioipioipio --——(----§-——)-2€3—)-->nBu NH P o H (5H 6H 6H 0206“ Step 5 OH CI NTP of compound 32 Arabinoadenosine 32-] was ted via steps 1 and 2 and then chlorinated to provide compound 32-4. Subsequent deprotection provided compound 32, and the triphosphate reaction ed the NTP of compound 32.
Exam le 40. S nthesis of 2’-iodo adenosine com ound 33 and 2’-iodo ATP TP of said compound! NH2 NH: NH2 N WIN? (/N o ‘N’JN Tfo<(5an 2/ imidazole ::r\/:. 0.0 DMAP/Tf20 PK; DCM/70/a0 'Pr/ 1 3 ro1 1 3 3—'P OSi o tetraisopropyldisilox 5'05' 0H p 2 ane/DMF, 95% ”Dr/ \_ iPr/ \ Step1 'Pr Pr 33_3 33—1 33—2 NH2 NH2 N \ N N </ \N I g </ I N J N o N TBAF/THF HO N _ i HMPT,LI| Pr\SIi o ____________________ - o (I) 80% \ .0 I SI Ste 4 OH I Shana /\ I Pr ipr 33_4 compound 33 N \ N o o o </ I J H075707E’704‘3LO N N/ (nBu NH) P o H ) ________§____2€_%__7__2___- (5H 5H (5H 0 Step5 OH I NTP of compound 33 Arabinoadenosine 33-1 was protected via steps 1 and 2 and then iodinated to provide nd 33-4. Subsequent deprotection provided compound 33, and the triphosphate reaction in DMF provided the NTP of compound 33.
Exam le 41. S nthesis of 2’-bromo c tidine com ound 34 and 2’-bromo CTP NTP of said compound} NHZ NH2 HN NH2 (\N \ \ \ N TfO \ i TfO f l *0 N o . o N 0 'm'dm'e, , o o .
‘P'\ HO i. 0.6“ EtaNlDMAPITfQO {Pks‘i prS‘i _’ war/57' —n’ ipr/i Ipr/ l £1: chhloro-‘lldj,§l5,3-. 0‘ DCM/70 /o e slox Si 0‘5'0I o‘SiO opy Ste 2 ””2 9 F, 95% /\ / P ip \. ip/ \ip r '9’ r Step 1 34—1 4- 34—3 3 34_3b O A020 N“ NH (Vi EtaNIDMAP/TfZO 1.HMPT,LiBr 6”l ,g & in] .Pr\o N o o 0 Si %o 'P55'. 0- PR;‘ 2. CH30H/NH3 'Pr/c‘, ?l 'F’r C', 'Pr/ ‘Sio 3’ 'Pr Ippr 34—2a 34—3a TBAFITHF HO N’go 1?? ---------- > A0 (nBuSNHuPzOsz) Slep4 OH Br HO’F’O’P’O’P’O‘WOH OH OH 06:; SlepS compound 34 0H 5' NTP of compound 34 ocytidine 34-] was protected under various conditions and then brominated to provide compound 34-4. Optionally, the reaction can provide compound 34-4 via compound 34—3a under any useful protection reactions, such as (i) 1.5 eq. Et3N, 1 eq. DMAP, 1.2 eq. TfCl, in DCM (lOmL); (ii) 3 eq. DMAP, 1.2 eq. TfCl in DCM (lSmL); or (iii) 15 eq. DMAP, 1.5 eq. Tf20, in DCM (lSmL) at -10°C to 0°C for 2 hour. In particular, 55mg of compound 34-3a was obtained from reaction condition (iii). Subsequent deprotection provided compound 34, and the triphosphate reaction in DMF provided the NTP of compound 34. Crude t 34 could be optionally d prior to phosphorylation.
Exam le 42. S nthesis of 2’-chloro uanosine com (mm! 35 and 2’-chloro GTP NTP of said ‘3 o o N NH N N <’ ‘ A < ‘ A </ | 0 N N N - , , r //J\ NH: o N NH 'm'dm'e 2 N P’\S‘- o 'P'\ ‘.
H0 N NH2 ‘I EISNIDMAP/Tf o2 o 0 iPr/s‘. 1,3 Dichloro—1,1,3,3— 03.0 DOM/64% 0H 0‘ .0 on Ietraisopropyldisilox / { SI Step 2 OHOH ane/DMF, 95% ‘pr iPr iPr/ \iP' step1 35—3 1 352 o 0 NH NH </ l A <, l A HMPT,Na0Ac N N NH2 0 0“ N o NH2 ‘Pr --------------> 'Pr\ ,_ 09 c -»-»--»-»---> \S‘i O- -------------------> 45% .pr/Sfl \pr/cw) SlepS Ste 3 o, Ste P 4 ” \ _0 /S{° R P 'Pr ' ‘Pr 35—5 ‘Pr 35_4 0 O NH NH <’ | A </ l N A NH N < \ 'F’r\9 N N 0 f NHZ ‘Pr\9. N NH? N HO N/)\NH2 /s: 0‘ ------------------ - ---------------- > /s. 0 'Pr (I) step 5 'Pr (5 Step 7 /\S\i0 /~s{o CI .7 OH Cl d 35 Pr 'Pr compoun .p, 35—6 ‘Pr o o o (N l H H H /JN\H ------------------------ > Ho,?,o,fi,o,fi,,o N NH2 (nBuaNHMPZOfi-IZ) OH OH 0H 0 step 8 ()H CI NTP of compound 35 ] Guanosine 35-] was protected under various conditions and then acetylated to provide compound 35-4. The on from compound 35-2 to compound 35-3 was conducted with 2 eq.
DMAP, 2 eq. Et3N, 3 eq. TfZO in 1,2-dichloroethane (10mL) at 40°C for 4 hours. About 55mg of compound 35-3 was ed after the purification.
Desired compound 35 can be obtained by any useful method. For example, as shown above, compound 35-4 can be treated with subsequent protection, chlorination, and deprotection steps to e compound 35. To obtain the corresponding NTP, a triphosphate reaction can be ted (e.g., any described herein). Optionally, the NTP can be purified (e. g., using a Sephadex DEAE—A25 colunm), lized, or evaporated (e. g., from EtOH).
Exam le 43. S nthesis of 2’-iodo uridine com ound 36 and 2’-iodo UTP TP of said compound} HO pyridine 36—2 351' I NH .139 Nal, TsOH, e, (PhSe)2, I & E“ HO N o z NaBH4, EtOH/THF, reflux, 90% :0; OH SePh 3&3 O | r 1)POCI3, PO(OMe)3 9 9 e? (kmA HO-P—O-P—O-P—O N 0 ”0" N 0 2) (nBu3NH)2(P207H2)' 6H (5H (5H 0 OH I NTP of compound 36 OH | compound 36 02,2’-Cyclouridine 36-] was protected to provide compound 36-2. uent iodination, optionally mediated with um, provided compound 36. A triphosphate reaction was conducted to provide the NTP of nd 36. Optionally, the NTP can be purified (e.g., using a Sephadex DEAE—AZS column), lyophilized, or evaporated (e.g., from EtOH).
Exam le 44. S nthesis of 2’-0 4’-C-meth lene adenosine com ound 37 and 2’-0 4’-C- meth lene ATP NTP of said com ound O O o | i 6 pDc a >< NaBH_4_ Ow‘OK?BnCl o '10 -.,o>< NO —H>+O<OW“‘°><_ (”,0 HO H6 B l'l 0/: 3M 37-1 37-2 37-3 HO 0 “0 o ..\\O ..
AcOH Na|O4 MsCI/pyrldlne \ HCHO _.HO ”O #1010 . to g MeOH NaOH ~‘ BnO 37-5 Bnd Bno 37'7 37-6 M50 M50 0 0 0 OAc TFA,BO% Mso OH ACzo/pyndlne M50 M50 0 OB" 09" OBn OH 80% OAc 37-9 37—3 37-10 NHBZ [>)J\HJNI\(KHNJNNAN BzHN NHBz \N N \ \ N \ /) </ l J B o </N Z lN/J M50 (N N N N N HF/HzO M50 NaOBflDMF 0 0 HOAc /\0ko?l BSA,TMSOTf,MeCN, 68% M5029 ’/\ 80% QB" 70% 0 OAc 37-11 BnO 37-12 BnO 37—13 NH2 NH2 </:f“ \ N J <N/ ‘ o o o (/:fNZN NH40HIMeNH2 Ho/\ “0 N/J Pd(OH)2/C/HCOONH4 HO,,LO,‘F‘,,O,‘F‘,,O /\7/—?" l l l ___* EtOH/H20,90% 0H 0H 0H 5# MeOH,80% O HO o o 37-14 37-15 compound 37 ] Similar to the strategy provided above for nd 24, compound 37-7 was produced with compound 37-1. Subsequent mesylation, deprotection, and acetylation provided compound 37- ‘ on of uracil and subsequent internal cyclization provided compound 37-12. Various protection and ection steps provided compound 37.
To obtain the corresponding NTP, a triphosphate reaction can be conducted (erg, any described herein). Optionally, the NTP can be purified (e.g., using a Sephadex DEAE—A25 colunm), lyophilized, or evaporated (e.g., from EtOH).
Exam le 45. S nthesis of c clo entene diolc tidine com ound 38 andc clo entene diol CTP NTP of said com ound OH OH OSMDBT kOZ/‘DH / K 1°” TBDMSCIJmidazoIe 4p0 ,. OH /\/ OSMDBT TsOHiacetone MgBr OH ”OH mAF‘THF 0H2.2-dlmethoxypropane 0 35% O OX0 95% o O 95% 0415959 x 38-3 38_2 x 38-4 OH / / / 0 0 methyllriphenyl OH OH ' “lOH V, x ”OH ”1:333?“ \ u,10H grubbs catalyst g . .
Nalo, pyndmmm a‘e —- — —.oo 0 O X 38—5 97% 0 0 NaH, DMSO 0 O O 0 molecular sieves,AcOH,DCM x X3845 X 38—7 93% X 38—8 36% 3&9 Se Fh J< ° o 0 \0 sodwum tedrahydroborahe, o cerium(|ll) chloride in methanol 777777777 - 0 "r" ,,,,,,,,- O ,,,,,,,,,,,,,,,,, ...........- 7< 0 0 i \ 0H 0 N o o N o o o (H) x/fl 1K1 Tj ammonia in methanol my ””””””””’ cog ””” ””””ma ***************** * °7Q gag; 7§ 0 38—14 7{ 0 38—15 0 slew sel 5 38—13 P 7Q ”Pi 6 OX NH; NH2 o \ N o o o o N o (kN u \N ,g YJ/ \§ l k l ,g OHilf‘leoilI-Loiléio N o N / 0 N 0 N 0 )Vo "T5 , - 0H OH OH OH _ Iliad/[SEE - S'JE’E‘AN'C'JZEIBZ'I'Q 0 sten 9 OH OH 7§ stepa step10 074° 38-17 NTP Of 38—1 6 OH OH 0% com ounp d 38 nd 38 D-ribose was protected and then allylated to provide compound 38-4, which was subsequently cyclized and reduced to provide compound 38-7. Olefin metathesis and subsequent ion provided compound 38-9, and further reduction reactions and addition of N—benzoyluracil provided compound 38-14. onal deprotection and protection reactions provided compound 38, and triphosphate reaction (e. g,. with any useful reaction condition, such as those described herein or in US. Pat. No. 7,893,227, incorporated herein by reference) provided the NTP of compound 38.
Exam le 46. S nthesis of 2’-meth luridine com ound 39 and 2’-meth lUTP NTP of said -’ \‘CZN " . O CAS:1779—49—3 Prii‘Si .
- - C q ~O\ step 3 Pri step 4 Pri step 5 I O O O N/go H H H ,,,,, 7 N O o ---------------------- - HOPOPOPOW6H (5H 6H step6 ; .., Ho‘° QI’CH HO 3 ’CH3 NTP of compound 39 nd 39 Uridine 39-] was protected and then oxidized with 2 eq. of Dess—Martin periodane to provide compound 39-3. Subsequent Wittig reaction, hydrogenation, and deprotection steps provided compound 39.
Exam le 47. S nthesis of 2’-meth lc tidine com ound 40 and 2’-meth lCTP NTP of said \ \ CA8: 177949a . . . c Priilsi oc , CH2 d EH3 step 5 step 3 Pri step 4 P“ 40—4 40-5 NH2 NH2 | N’KO o o o 0 1: HO HoiPioiP‘ioiPo N 0 W o ------------------------------- - OH OH OH 1O step 6 _ ; _ v, Hox‘ ZCH3 HO CH3 compound 40 NTP of compound 40 Cytidine 40-] was protected and then oxidized to provide nd 40-3. Subsequent Wittig reaction, hydrogenation, and deprotection steps provided compound 40.
Exam le 48. S nthesis of N-ace lc tidine com ound 41 and N-ace lCTP NTP of said compound! NHAc NHAc NHAc 01.1’rImeMylphosphale 9 N 0 4.511314 0 0 °. N 0 HO cl—fi—D —> O . Ho_p'_o_l'l=_o_p'_o O Sponge2 Probn 5. Blsmzbutylammomum) Cl OH OH OH pyrophosphate 3 Pocl3 o” OH ' ”'2 "" T“ W” NTP of Egrfigound 41 comEmmy;und 41 c11HnN301sP: Bran Mass: 255.10 Exact Mass: 55.1!) A solution etyl-cytidine (compound 41) (103.0 mg, 0.36 mmol) was added to proton sponge (1 15.72mg, 0.54 mmol, 1.50 equiv) in 1.0 mL trimethylphosphate (TMP) and 1.0 mL of anhydrous tetrahydrofuran (THF). The solution was stirred for 10 s at 0°C. Phosphorous oxychloride (POC13) (67.2 111, 0.72 mmol, 2.0 eqiv.) was added se to the solution before being kept stirring for 2 hours under N2 atmosphere. After 2 hours the on was reacted with a mixture of bistributylammonium pyrophosphate (TBAPP or (n-BugNI-I)2H2P207) (1.28 g, 2.34 mmol, 6.5 eqiv.) and ylamine (350.0 111, 1.45 mmol, 4.0 equiv.) in 2.5 ml of dimethylformamide. After imately 15 minutes, the reaction was quenched with 24.0 ml of 0.2M triethylammonium bicarbonate (TEAB) and the clear solution was stirred at room temperature for an hour. The reaction mixture was lyophilized overnight and the crude reaction mixture was purified by HPLC (Shimadzu, Kyoto Japan, Phenomenex C18 preparative colurrm, 250 x 21.20 mm, 10.0 micron; gradient: 100 % A for 3.0 min, then 1% B/min, A : 100 mM TEAB buffer, B : ACN; flow rate: .0 mL/min; retention time: 16.81-17.80 min). Fractions containing the desired compound were pooled and lyophilized to produce the NTP of compound 41. The sphorylation reactions were carried out in a two-neck flask ried under N2 here. Nucleosides and the protein sponge were dried over P205 under vacuum overnight prior to use. The ion of monophosphates was monitored by LCMS. e 49. Synthesis of 5-methoxy uridine {compound 42) and 5-methoxy UTP g STP of said compound! O O 0 H360 H3120 NH NH | N’ko | N’go Hacofim| 4. Bug” "A0 _ c”) 9 9 c”; 1. Trlmethyl phosphate HO —> 0 clipio O HO-P-O-P-O-P-O 2_ Proton Sponge 6| 5. Bisflributylammonium) 6H 6H 6H —’ pyrophosphate 0” 0” ”Sock 0” 0” 6.0.2MTEAB buffer °H 0” compound 42 NTP of nd 42 7 1s c1nH11N2°1sP3 °1°”““2°' Exact Mass: 513.99 Exact Mass: 274.03 A solution of 5-methoxy uridine (compound 42) (69.0 mg, 0.25 mmol, plus heat to make it soluble) was added to proton sponge (80.36 mg, 0.375 mmol, 1.50 equiv.) in 0.7 mL trimethylphosphate (IMF) and was stirred for 10 minutes at 0°C. Phosphorous oxychloride (POCI3) (46.7 ul, 0.50 mmol, 2.0 equiv.) was added dropwise to the solution before being kept stirring for 2 hours under N2 atmosphere. After 2 hours the solution was reacted with a mixture of bistributylammonium osphate (TBAPP or (n-BugNH)2H2P207) (894.60 mg, 1.63 mmol, 6.50 equiv.) and tributylamine (243.0 111, 1.00 mmol, 4.0 equiv.) in 2.0 ml of dimethylformamide. After approximately 15 minutes, the reaction was quenched with 17.0 ml of 0.2M ylammonium bicarbonate (TEAB) and the clear solution was stirred at room temperature for an hour. The reaction mixture was lyophilized overnight and the crude reaction mixture was purified by HPLC (Shimadzu, Kyoto Japan, Phenomenex C18 preparative column, 250 X 21 .20 mm, 10.0 micron; gradient: 100 % A for 3.0 min, then 1% B/min, A = 100 mM TEAB , B = ACN; flow rate: .0 mL/min; retention time: 16.57-17.51 min). Fractions containing the desired nd were pooled and lyophilized to produce the NTP of nd 42. The triphosphorylation reactions were carried out in a two-neck flask flame-dried under N2 atmosphere. Nucleosides and the protein sponge were dried over P205 under vacuum overnight prior to use. The formation of monophosphates was monitored by LCMS.
Exam le 50. S nthesis of 5-f0rm l c tidine com ound 43 and 5-f0rm l CTP TP of said NHz NH2 NH2 wcfil awkfi'L one?" N ° N ° 4— Bus" "3° . 9 .. .. 9 HO 1.Tr|mathyl phospham —> O Clil'fO O HO-P-O-P-O-Ff-O O 2_ Proton Sponge 5. Bfltrlhutylammomum)_ Cl 6H 6H OH W pyrophosphate 0" 0“ ' 0“ °H 6.0.2MTEAB buffer 0“ 0" compound 43 NTP of compound 43 7 18 7 N3°s 01.,H15N3015P3 Exact Mass: 271.03 5““ "35‘: 51033 A on of 5-formyl cytidine (compound 43) ) (48.4 mg, 0.18 mmol, plus heat to make it soluble) was added to proton sponge (57.86 mg, 0.27 mmol, 1.50 equiv.) in 0.7 mL hylphosphate (TMP) and was stirred for 10 s at 0°C. Phosphorous oxychloride (POC13) (33.6 ul, 0.36 mmol, 2.0 equiv.) was added dropwise to the solution before being kept stirring for 2 hours under N2 atmosphere. After 2 hours the solution was reacted with a mixture of bistributylammonium pyrophosphate (TBAPP or (II-BU3NH)2H2P207) (642.0 mg, 1.17 mmol, 6.50 equiv.) and tributylamine (175.0 111, 0.72 mmol, 4.0 equiv.) in 1.7 ml of dimethylformamide. After approximately 15 minutes, the on was quenched with 12.0 ml of 0.2M triethylammonium bicarbonate (TEAB) and the clear solution was stirred at room temperature for an hour. The reaction mixture was lyophilized overnight and the crude reaction mixture was purified by HPLC (Shimadzu, Kyoto Japan, Phenomenex C18 preparative column, 250 x 21.20 mm, 100 micron; gradient: 100 % A for 3.0 min, then 1% B/min, A : 100 mM TEAB , B : ACN; flow rate: .0 mL/min; retention time: 17.04-17.87 min). Fractions containing the desired compound were pooled and lyophilized to provide the NTP of compound 43. The triphosphorylation reactions were carried out in a two-neck flask flame-dried under N2 atmosphere. Nucleosides and the protein sponge were dried over P205 under vacuum overnight prior to use. The formation of monophosphates was monitored by LCMS.
Exam le 51. S nthesis of 3-meth luridine com ound 44 and 3-meth l UTP NTP of said o o o 31“” fir“ 61”” N o C“) N 0 9 9 9 N 0 4. Bu:N HO 1. Trimethyl phosphate 0 c|_p_° —> O _?_°_?_° 0 2. Proton Sponge ['2' 5- Bbttfibu‘vhmmmiu'") 6H OH OH mm... 0H 0H m...—>- a OH 0. a. 0.. ' 0'2 M TEAB bum" compound 44 NTP of compound 44 cwHuNzos c10H17N2°15P3 Exact Mass: 258.09 Exact Mass: 497.96 A solution of 3-methyl uridine (compound 44) (45.80 mg, 0.18 mmol) was added to proton sponge (57.86 mg, 0.27 mmol, 1.50 equiv.) in 0.5 mL trimethylphosphate (IMF) and was stirred for 10 s at 0°C. Phosphorous oxychloride (POC13) (33.6 111, 0.36 mmol, 2.0 equiv.) was added dropwise to the solution before being kept stirring for 2 hours under N2 atmosphere.
After 2 hours the solution was reacted with a mixture of bistributylammonium pyrophosphate (TBAPP or (n-Bu3NH)2H2P2O7) (652.0 mg, 1.19 mmol, 6.60 equiv.) and tributylamine (175.0 ul, 0.72 mmol, 4.0 equiv.) in 1.3 ml of dimethylformamide. After approximately 15 minutes, the reaction was quenched with 12.0 ml of 0.2M triethylammonium bicarbonate (TEAB) and the clear on was stirred at room ature for an hour. The reaction mixture was lyophilized overnight and the crude reaction mixture was d by HPLC (Shimadzu, Kyoto Japan, Phenomenex C18 preparative column, 250 x 21.20 mm, 10.0 micron; gradient: 100 % A for 3.0 min, then 1% B/min, A : 100 mM TEAB buffer, B : ACN; flow rate: 10.0 mL/min; retention time: 18.52-19.57 min). Fractions containing the desired compound were pooled and lyophilized to provide the NTP of nd 44. The triphosphorylation reactions were carried out in a ck flask flame-dried under N2 here. Nucleosides and the protein sponge were dried over P205 under vacuum overnight prior to use. The formation of monophosphates was monitored by LCMS.
Exam le 52. S nthesis of Nl-meth l seudouridine com ound 45 and Nl-meth l pseudoUTP 1NTP of said compound! HNJLN’CHa 1 it HN “4m, HN We"'3 / / / 0 1. Trimethyl phosphate 0 o 4. BuaN 9 o o H —> I ll HO 2. Proton sponge 0 O o Ho-lf-o-lf-o-If-o O —> El 5. Bisttributyhmmonium) OH OH OH 3. POCI, pyrophosphate OH OH OH OH nu ma compound 45 s. 0.2 M TEAE buffer NTP of compound 45 cmHuNzos cioH17N2°1sP3 Exact Mass: 358.09 Exact Mass: 497.96 A solution ofNl-methyl pseudouridine (compound 45) (96.6 mg, 0.374 mmol, plus heat to make it soluble) was added to proton sponge (120.0 mg, 0.56 mmol, 1.50 equiv.) in 0.8 mL trimethylphosphate (IMF) and was stirred for 10 minutes at 0°C. Phosphorous oxychloride (POC13) (70.0 ul, 0.75 mmol, 2.0 equiv.) was added dropwise to the solution before being kept stirring for 2 hours under N2 atmosphere. After 2 hours the solution was reacted with a mixture of bistributylammonium pyrophosphate (TBAPP or 3NH)2H2P207) (1.36g, 2.47 mmol, 6.60 equiv.) and tributylamine (362.0 111, 1.5 mmol, 4.0 equiv.) in 2.5 ml of dimethylformamide. After approximately 15 minutes, the on was quenched with 17.0 ml of 0.2M triethylammonium bicarbonate (TEAB) and the clear solution was stirred at room temperature for an hour. The reaction mixture was lyophilized overnight and the crude on e was purified by HPLC (Shimadzu, Kyoto Japan, Phenomenex C18 preparative column, 250 x 21.20 mm, 10.0 micron; gradient: 100 % A for 3.0 min, then 1% B/min, A : 100 mM TEAB buffer, B : ACN; flow rate: .0 mL/min; retention time: 15.91-17.01 min). Fractions containing the desired compound were pooled and lized was subjected to a sphorylation reaction to provide the NTP of compound 45. The triphosphorylation reactions were carried out in a two-neck flask ried under N2 atmosphere. Nucleosides and the protein sponge were dried over P205 under vacuum overnight prior to use. The formation of monophosphates was monitored by LCMS.
Exam le 53. S nthesis of 5-methox carbon lethen luridine com ound4 and 5- methox carbon lethen IUTP VTP of said com ound O O O O HN|\OM3 OA‘N r1|\om 9 o N 1. Trimathyl ate 0 Cl—R—O 0 2. Proton Sponga Cl OH OH 3-POCI3 OH OH 4.Bu3N compound 46 5. B's(tributylammonium) 7 22 7 pyrophosphata C13H15N203 Exact Mass: 328.09 5. 0.2 M TEAB buffer 0 0 “We,“ 0 DAN —o—lf—o—l‘l—o 0 OH OH OH OH OH NTP of compound 46 c13H19N2°11P3 Exact Mass: 567.99 A solution of 5-methoxycarbonylethenyl uridine (compound 46) (102.0 mg, 0.31 mmol) was added to proton sponge (99.65 mg, 0.46 mmol, 1.50 equiv.) in 0.8 mL trimethylphosphate (IMF) and was stirred for 10 minutes at 0°C. Phosphorous oxychloride (POClg) (57.8 ul, 0.62 mmol, 2.0 equiv) was added dropwise to the solution before being kept stirring for 2 hours under N2 atmosphere. After 2 hours the solution was reacted with a mixture of bistributylammonium pyrophosphate (TBAPP or (n-BugNH)2H2PZO7) (1.12g, 2.05 mol, 6.60 equiv.) and tributylamine (300.0 ul, 1.24 mmol, 4.0 ) in 2.5 ml of dimethylformamide. After approximately 15 minutes, the reaction was quenched with 20.0 ml of 0.2M triethylammonium bicarbonate (TEAB) and the clear on was stirred at room temperature for an hour. The reaction mixture was lyophilized overnight and the crude reaction e was purified by HPLC (Shimadzu, Kyoto Japan, Phenomenex C18 preparative column, 250 x 21.20 mm, 10.0 micron; gradient: 100 % A for 3.0 min, then 1% B/min, A : 100 mM TEAB buffer, B : ACN; flow rate: 10.0 mL/min; retention time: 2156-23 .21 min). Fractions containing the desired compound were pooled and lyophilized to provide the NTP of compound 46. The triphosphorylation ons were carried out in a two-neck flask flame-dried under N2 atmosphere. sides and the protein sponge were dried over P205 under vacuum overnight prior to use. The formation of monophosphates was monitored by LCMS.
Exam le 54. S nthesis of 5-amino r0 en ne com (mm! 47 and S-amino r0 en IUTP [NTP of said compound! \ HN NHTFA MN NHTFA l A | A o 0 N O N H CI—P-O 0 HO 1. Trimethyl phosphate O (El 2. Proton Sponge OH OH 0H 0H 3J0“: 4.5u,N Protected compound 47 24 . B'sflributylammonium) _ _ c14H1sFJN307 pyrophosphate Exact Mass: 395.09 5. 02 M TEAB buffer \ \ HM NH2 HN NHTFA ‘ | o a 02W 02W Ho—g—o—g—o—b—ou u \ NH4OH H u u o 2 —HO-fi-O-fi-O-If-O 0 0H OH OH OH oH OH OH OH OH OH NTP of compound 47 c H o Exalzl "21:2: 3:391 914H19FaNao1sPa Exact Mass: 53499 5-Aminopropenyl uridine 47 was ted and a on of protected compound 47 (86.0 mg, 0.22 mmol) was added to proton sponge (70.7 mg, 0.33 mmol, 1.50 ) in 0.7 mL trimethylphosphate (IMF) and was stirred for 10 minutes at 0°C. Phosphorous oxychloride (POC13) (41.1 ul, 0.44 mmol, 2.0 equiv.) was added dropwise to the solution before being kept stirring for 2 hours under N2 atmosphere. After 2 hours the solution was reacted with a mixture of bistributylammonium pyrophosphate (TBAPP or (II-BU3NH)2H2P207) (784.6 mg, 1.43 mmol, 6.50 equiv.) and tributylamine (213.0 111, 0.88 mmol, 4.0 ) in 1.6 ml of dimethylformamide. After approximately 15 minutes, the reaction was quenched with 15.0 ml of 0.2M triethylammonium bicarbonate (TEAB) and the clear solution was stirred at room temperature for an hour. 180 ml of concentrated ammonium hydroxide was added to the reaction mixture to remove the trifluoroacetyl group. It was then stored stirring overnight. The reaction mixture was lyophilized overnight and the crude reaction mixture was purified by HPLC (Shimadzu, Kyoto Japan, enex C18 preparative colunm, 250 x 21.20 mm, 10.0 micron; gradient: 100 % A for 3.0 min, then 1% B/min, A : 100 mM TEAB buffer, B : ACN; flow rate: 10.0 mL/min; retention time: 16.14-17.02 min).
Fractions ning the desired compound were pooled and lyophilized to provide the NTP of compound 47. The triphosphorylation ons were carried out in a two-neck flask flame-dried under N2 atmosphere. Nucleosides and the protein sponge were dried over P205 under vacuum overnight prior to use. The formation of monophosphates was monitored by LCMS.
Exam le 55. S nthesis of N-PEG adenosine com ound 48 and N-PEG ATP NTP of said compound} H N/\/\o/\/°\/\ HN/\/\°/\/O\/\ N \ N I) < | H w /J o < A o N ON 1. Trina-thyl ata 9 N 0” H0 0 2. Proton Sponge Cl—E—O o —> 01 NHTFA 3' P°°'3 NHTFA OH on 0H 0H protected compound 48 27 Nsom 7 7 Exact Mass: 654.28 4. augu . Bis(tributylammonium) pyrophosphate S. 0.2 M TEAB buffer HNMONOWO “NMONOWO N \ N H N \ N < i) H No < l A 9 N N ° 0 o o N N 0“”o . .. : . ”Hm“ Ho—g—o—If—o—If—o o H071|$eOe1§eOel$eO 0 OH OH OH 0H 0H OH OH OH NH2 NHTFA on 0H NTP of compound 48 23 OMHastomP: CZSHMFSNS°19P3 Exact Mass: 798.20 Exact Mass: 894.18 N-PEG adenosine 48 was protected and a solution of the protected compound 48 (100.0 mg, 0.15 mmol) was added to proton sponge (49.3 mg, 0.23 mmol, 1.50 equiv.) in 0.65 mL trimethylphosphate (IMF) and was stirred for 10 minutes at 0°C. Phosphorous oxychloride (POC13) (28.0 ul, 0.3 mmol, 2.0 ) was added dropwise to the solution before being kept stirring for 2 hours under N2 here. After 2 hours the solution was reacted with a mixture of bistributylammonium pyrophosphate (TBAPP or (H'BU3NH)2H2P207) (537.7 mg, 0.98 mmol, 6.50 equiv.) and ylamine (146.0 111, 0.6 mmol, 4.0 equiv.) in 1.2 ml of dimethylformamide. Afler approximately 15 minutes, the reaction was quenched with 10.0 ml of 0.2M triethylammonium bicarbonate (TEAB) and the clear on was stirred at room temperature for an hour. 18.0 ml of concentrated ammonium hydroxide was added to the reaction mixture to remove the trifluoroacetyl group. It was then stored stirring overnight. The reaction mixture was lyophilized overnight and the crude reaction mixture was purified by HPLC (Shimadzu, Kyoto Japan, Phenomenex C18 preparative column, 250 x 21.20 mm, 10.0 micron; gradient: 100 % A for 3.0 min, then 1% B/min, A = 100 mM TEAB buffer, B = ACN; flow rate: 10.0 ; retention time: 24.5-25.5 min).
Fractions containing the desired compound were pooled and lyophilized to provide the NTP of nd 48. The triphosphorylation reactions were carried out in a two-neck flask flame-dried under N2 atmosphere. Nucleosides and the protein sponge were dried over P205 under vacuum overnight prior to use. The formation of monophosphates was red by LCMS.
Exam le 56. S nthesis of N-meth [adenosine com ound 49 and N-meth lATP NTP of said compound) NHMe NHME NHMe N \ \ x N N N / ‘ <faN < / <’ f) A N N/ N N N N ”us" 1.1rumethyl phosphate. 9 9 9 9 HO CI—P—o H0_F_°_F_0_F_o 0 o . o 2' Proton swngg e] 5. Ensuributyhmmomum) 6H 6H 6H —> pyrophosphate 0H 01-! “we" 0“ 0“ 6.0.2MTEAB buffer 0“ 0“ compound 49 NTP of compound 49 3° 7 c11H1,N,o., c11H18N5013P3 Exact Mass: 281.11 Exact Mass: 521m A solution ofN-methyl adenosine (compound 49) (70.0 mg, 0.25 mmol) was added to proton sponge (79.29 mg, 0.37 mmol, 1.50 equiv.) in 0.7 mL trimethylphosphate (IMF) and was stirred for 10 minutes at 0°C. Phosphorous oxychloride (POC13) (46.66 111, 0.50 mmol, 2.0 equiv.) was added dropwise to the solution before being kept stirring for 2 hours under N2 atmosphere.
After 2 hours the on was reacted with a mixture of bistributylammonium pyrophosphate (TBAPP or (n-BugNH)2H2P207) (888.85 mg, 1.62 mmol, 6.50 equiv.) and tributylamine (241.0 ul, 1.0 mmol, 4.0 equiv.) in 1.3 ml of dimethylformamide. After approximately 15 minutes, the reaction was quenched with 16.0 ml of 0.2 M triethylammonium bicarbonate (TEAB) and the clear solution was stirred at room temperature for an hour. The on e was lyophilized ght and the crude reaction mixture was purified by HPLC (Shimadzu, Kyoto Japan, Phenomenex C18 preparative column, 250 x 21.20 mm, 10.0 micron; gradient: 100 % A for 3.0 min, then 1% B/min, A : 100 mM TEAB buffer, B : ACN; flow rate: 10.0 mL/min; retention time: 20.14 min). ons containing the desired compound were pooled and lyophilized to provide the NTP of compound 49. The triphosphorylation ons were carried out in a two-neck flask flame-dried under N2 here. Nucleosides and the protein sponge were dried over P205 under vacuum overnight prior to use. The formation of monophosphates was monitored by LCMS.
[NTP of said compound! 0 O O N N N NH NH NH < f: <N ‘ NANM N N NMez 92 <N:(N:‘\NM 9 4_ BU3N 92 9 '0' Ho 1.Trimethyl phosphate Cl—P—O 0 O O-P-O-P-O O 2. Proton Sponge 0| 5. Bisttribulylammonium) 0H 0H 0H pyrophosphate 3 POCI 0" OH ' 3 0H 0" 6.0.2MTEAB buffer 0" 0" compound 50 NTP of nd 50 7 7 C12H11N505 cuflansouP; BuctMass: 311.12 Exact Mass: 551.02 A solution ofN,N-dimethyl guanosine (compound 50) (65.8 mg, 0.21 mmol) was added to proton sponge (68.58 mg, 0.32 mmol, 1.50 equiv) in 0.7 mL trimethylphosphate (TMP) and was stirred for 10 minutes at 0°C. Phosphorous oxychloride (POC13) (39.20 111, 0.42 mmol, 2.0 equiv.) was added dropwise to the solution before being kept stirring for 2 hours under N2 atmosphere.
After 2 hours the solution was d with a mixture of bistributylammonium pyrophosphate (TBAPP or (H'BU3NH)2H2P207) (751.67 mg, 1.37 mmol, 6.50 equiv.) and tributylamine (204.0 ul, 0.84 mmol, 4.0 equiv.) in 1.5 ml of dimethylformamide. After approximately 15 minutes, the reaction was quenched with 14.0 ml of 0.2 M triethylammonium bicarbonate (TEAB) and the clear solution was stirred at room temperature for an hour. The reaction e was lized overnight and the crude reaction e was purified by HPLC dzu, Kyoto Japan, Phenomenex C18 preparative column, 250 x 21.20 mm, 10.0 micron; gradient: 100 % A for 3.0 min, then 1% B/min, A : 100 mM TEAB buffer, B : ACN; flow rate: 10.0 mL/min; retention time: 19.27-19.95 min). Fractions containing the desired compound were pooled and lyophilized to provide the NTP of compound 50. The triphosphorylation reactions were carried out in a ck flask flame-dried under N2 atmosphere. Nucleosides and the protein sponge were dried over P205 under vacuum overnight prior to use. The formation of monophosphates was monitored by LCMS.
Exam le 58. General s for tri hos hate 5 nthesis of NTPS | & f5": N o N 0 —. 9 9 9 ° HOAX—Zo _fi’_°_fi"o/\S_ZOH OH OH HO OH HO OH The side i can be phosphorylated by any useful method to provide a triphosphate compound ii. For e, the nucleoside can be added to proton sponge and trimethylphosphate (TMP) and cooled (e. g., to -40°C). Phosphorous oxychloride (POClg) can be added dropwise before reacting with bistributylammonium pyrophosphate (TBAPP or (n-BugNH)2H2PZO7) and tributylamine. The reaction can then be quickly quenched with triethylammonium bicarbonate (TEAB). Exemplary conditions are provided in US. Pat. No. 7,893,227, which is incorporated herein by reference.
After the phosphorylation reaction, the reaction mixture can be optionally lyophilized, purified (e.g., by change tography and/or HPLC), or converted to a sodium salt (e.g., by dissolving in MeOH and adding sodium perchlorate in acetone).
Example 59: PCR for cDNA Production PCR procedures for the preparation of cDNA are performed using 2x KAPA HIFITM HotStart ReadyMix by Kapa tems (Woburn, MA). This system includes 2x KAPA ReadyMixl2.5 ul; Forward Primer (10 uM) 0.75 ul; Reverse Primer (10 uM) 0.75 ul; Template cDNA 100 ng; and dHZO diluted to 25.0 ul. The reaction conditions are at 95° C for 5 min. and 25 cycles of98° C for 20 sec, then 58° C for 15 sec, then 72° C for 45 sec, then 72° C for 5 min. then 4° C to termination.
The reverse primer of the instant invention incorporates a poly-T120 for a poly—A120 in the mRNA. Other reverse primers with longer or shorter poly-T tracts can be used to adjust the length of the poly-A tail in the mRNA.
The reaction is cleaned up using Invitrogen’s PURELINKTM PCR Micro Kit (Carlsbad, CA) per manufacturer’s instructions (up to 5 ug). Larger reactions will require a cleanup using a product with a larger capacity. Following the cleanup, the cDNA is quantified using the op and analyzed by agarose gel electrophoresis to confirm the cDNA is the ed size. The cDNA is then submitted for sequencing analysis before proceeding to the in vitro transcription reaction.
Example 60. In vitro Transcription [IVT] The in vitro transcription on generates mRNA containing modified nucleotides or modified RNA. The input nucleotide triphosphate (NTP) mix is made se using l and un- natural NTPs.
A typical in virro ription reaction includes the following: Template cDNA 1.0 ug 10x transcription buffer (400 mM Tris-HCl pH 2.0 ul 8.0, 190 mM MgC12, 50 mM DTT, 10 mM Spermidine) Custom NTPs (25mM each 7.2 ul RNase Inhibitor 20 U T7 RNA polymerase 3000 U dH20 Up to 20.0 ul Incubation at 370 C for 3 hr—5 hrs.
The crude IVT mix may be stored at 40 C overnight for cleanup the next day. 1 U of RNase-free DNase is then used to digest the original template. After 15 minutes of incubation at 37° C, the mRNA is purified using Ambion’s MEGACLEARTM Kit (Austin, TX) following the manufacturer’s instructions. This kit can purify up to 500 ug ofRNA. Following the cleanup, the RNA is quantified using the op and analyzed by agarose gel electrophoresis to confirm the RNA is the proper size and that no ation of the RNA has occurred.
The T7 RNA polymerase may be selected from, T7 RNA polymerase, T3 RNA polymerase and mutant polymerases such as, but not limited to, the novel rases able to incorporate modified NTPs as well as those polymerases described by Liu (Esvelt et al. (Nature (2011)472(7344):499-503 and US. Publication No. 20110177495) which recognize alternate promoters, Ellington (Chelliserrykattil and Ellington, Nature Biotechnology (2004) 22(9):]155- 1160) describing a T7 RNA polymerase variant to transcribe 2’-O-methyl RNA and Sousa (Padilla and Sousa, c Acids Research (2002) 30(24): e128) describing a T7 RNA polymerase double mutant; herein incorporated by reference in their ties.
Example 61. Enymatic g of mRNA Capping of the mRNA is performed as s where the mixture includes: IVT RNA 60 ug-l 80ug and ngO up to 72 [11. The mixture is incubated at 650 C for 5 minutes to denature RNA, and then is transferred immediately to ice.
The protocol then involves the mixing of 10x Capping Buffer (0.5 M Tris-HCI (pH 8.0), 60 mM KCI, 12.5 mM MgC12) (10.0 pl); 20 mM GTP (5.0 pl); 20 mM S-Adenosyl Methionine (2.5 pl); RNase Inhibitor (100 U); 2’-O-Methyltransferase (400U); Vaccinia g enzyme (Guanylyl transferase) (40 U); dH20 (Up to 28 pl); and incubation at 37° C for 30 minutes for 60 pg RNA or up to 2 hours for 180 pg ofRNA.
The mRNA is then purified using Ambion’s MEGACLEARTM Kit (Austin, TX) following the manufacturer’s instructions. Following the cleanup, the RNA is quantified using the NANODROPTM (ThermoFisher, Waltham, MA) and analyzed by agarose gel electrophoresis to confirm the RNA is the proper size and that no degradation of the RNA has ed. The RNA product may also be sequenced by running a reverse-transcription-PCR to generate the cDNA for sequencing.
Example 62. Pole Tailing Reaction Without a poly-T in the cDNA, a poly-A tailing reaction must be performed before cleaning the final product. This is done by mixing Capped IVT RNA (100 pl); RNase tor (20 U); 10x Tailing Buffer (0.5 M Tris-HCl (pH 8.0), 2.5 M NaCl, 100 mM MgC12)(12.0 pl); 20 mM ATP (6.0 pl); Poly-A Polymerase (20 U); dH20 up to 123.5 pl and incubation at 370 C for 30 min. If the poly-A tail is already in the transcript, then the tailing reaction may be skipped and proceed ly to cleanup with Ambion’s MEGACLEARTM kit n, TX) (up to 500 pg). Poly— A Polymerase is preferably a recombinant enzyme sed in yeast.
For s performed and described herein, the poly-A tail is encoded in the IVT template to comprise160 nucleotides in length. However, it should be understood that the sivity or integrity of the poly-A tailing reaction may not always result in exactly 160 nucleotides. Hence poly- A tails ofapproximately 160 nucleotides, e.g, about 150-165, 155, 156, 157, 158, 159, 160, 161, 162, 163, 164 or 165 are within the scope of the invention.
Example 63. Method of Screening for Protein Expression A. Electrospray Ionization A biological sample which may contain proteins encoded by modified RNA administered to the subject is prepared and analyzed according to the manufacturer protocol for ospray ionization (ESI) using 1, 2, 3 or 4 mass analyzers. A biologic sample may also be analyzed using a tandem ESI mass ometry system. ns of protein fragments, or whole proteins, are compared to known controls for a given protein and identity is determined by comparison.
B. Matrix-Assisted Laser Desorption/Ionization A biological sample which may contain proteins encoded by modified RNA administered to the subject is prepared and analyzed according to the manufacturer protocol for matrix-assisted laser desorption/ionization (MALDI).
Patterns of protein nts, or whole proteins, are compared to known controls for a given protein and identity is determined by comparison.
C. Liquid Chromatography-Mass ometry-Mass ometry ] A biological sample, which may n proteins encoded by modified RNA, may be treated with a trypsin enzyme to digest the proteins contained within. The resulting es are analyzed by liquid chromatography-mass spectrometry-mass spectrometry (LC/MS/MS). The peptides are fragmented in the mass spectrometer to yield diagnostic patterns that can be matched to n ce databases Via computer algorithms. The digested sample may be diluted to achieve 1 ng or less ng material for a given protein. Biological samples containing a simple buffer background (e. g. water or volatile salts) are amenable to direct in-solution digest; more complex backgrounds (e. g. detergent, non-volatile salts, glycerol) require an additional clean-up step to facilitate the sample analysis.
Patterns of protein nts, or whole proteins, are compared to known controls for a given protein and identity is determined by comparison.
Example 64. e Study: PBMC A. PBMC isolation and Culture 50 mL ofhuman blood from two donors was received from Research Blood Components (lots KP30928 and KP30931) in sodium heparin tubes. For each donor, the blood was pooled and diluted to 70 mL with DPBS (SAFC Bioscience 59331C, lot 08) and split evenly between two 50 mL conical tubes. 10 mL of Ficoll Paque (GE Healthcare 1703, lot 10074400) was gently dispensed below the blood layer. The tubes were centrifuged at 2000 rpm for 30 minutes with low acceleration and braking. The tubes were removed and the buffy coat PBMC layers were gently transferred to a fresh 50 mL conical and washed with DPBS. The tubes were centrifuged at 1450 rpm for 10 minutes.
The supernatant was aspirated and the PBMC s were resuspended and washed in 50 mL ofDPBS. The tubes were centrifuged at 1250 rpm for 10 minutes. This wash step was repeated, and the PBMC pellets were resuspended in 19 mL of Optimem I (Gibco 11058, lot 1072088) and counted. The cell suspensions were adjusted to a concentration of 3.0 X 10“6 cells / mL live cells.
These cells were then plated on five 96 well tissue culture treated round bottom plates (Costar 3799) per donor at 50 uL per well. Within 30 s, ection mixtures were added to each well at a volume of 50 uL per well. After 4 hours post transfection, the media was supplemented with 10 uL of Fetal Bovine Serum (Gibco 10082, lot 1012368) B. Transfection Preparation Modified mRNA encoding human G—CSF (mRNA sequence showu in SEQ ID NO: 1; polyA tail of approximately 160 nucleotides not shown in sequence; 5’cap, Capl) (containing either (1) natural NTPs, (2) 100% substitution with 5-methyl cytidine and pseudouridine, or (3) 100% tution with 5-methyl cytidine and Nl-methyl pseudouridine; mRNA ng luciferase (IVT cDNA sequence shown in SEQ ID NO: 2; mRNA sequence shown in SEQ ID NO: 3, polyA tail of approximately 160 nucleotides not shown in sequence, S’cap, Capl, fully modified with 5- cytosine at each cytosine and pseudouridine replacement at each uridine site) ining either (1) natural NTPs or (2) 100% substitution with 5-methyl cytidine and pseudouridine) and TLR agonist R848 (Invivogen tlrl-r848) were diluted to 38.4 ng / uL in a final volume of 2500 uL Optimem I.
Separately, 110 uL of Lipofectamine 2000 (Invitrogen 11668-027, lot 1070962) was diluted with 6.76 mL Optimem I. In a 96 well plate nine aliquots of 135 uL of each mRNA, positive control (R—848) or negative control (Optimem I) was added to 135 uL of the diluted Lipofectamine 2000. The plate ning the material to be transfected was incubated for 20 minutes. The transfection mixtures were then transferred to each of the human PBMC plates at 50 uL per well.
The plates were then incubated at 37°C. At 2, 4, 8, 20, and 44 hours each plate was removed from the incubator, and the supernatants were frozen.
] After the last plate was removed, the supernatants were assayed using a human G-CSF ELISA kit (Invitrogen KHC2032) and human IFN-alpha ELISA kit (Thermo Scientific 4] 105-2).
Each condition was done in duplicate.
C. n and Innate Immune Response Analysis The ability of unmodified and modified mRNA to produce the encoded protein was ed (G-CSF production) over time as was the ability of the mRNA to trigger innate immune recognition as measured by eron-alpha production. Use of in vitro PBMC cultures is an accepted way to measure the immunostimulatory potential of ucleotides (Robbins et al., Oligonucleotides 2009 19:89-102).
Results were interpolated against the standard curve of each ELISA plate using a four ter logistic curve fit. Shown in Tables 4 and 5 are the average from 3 separate PBMC donors of the G-CSF, eron-alpha (IFN-alpha) and tumor necrosis factor alpha (TN'F-alpha) production over time as measured by c ELISA.
In the G-CSF ELISA, background signal from the Lipofectamine 2000 (LF2000) untreated condition was subtracted at each time point. The data demonstrated specific production of human G- CSF protein by human peripheral blood mononuclear is seen with G-CSF mRNA containing natural NTPs, 100% substitution with y1 cytidine and pseudouridine, or 100% substitution with 5- methyl cytidine and hyl pseudouridine. Production of G-CSF was significantly increased through the use of 5-methy1 cytidine and N1 -methyl uridine modified mRNA relative to 5- methyl cytidine and pseudouridine modified mRNA.
With regards to innate immune recognition, while both modified mRNA chemistries largely prevented IFN—alpha and TNF-alpha production ve to positive ls (R848, p(I)p(C)), significant differences did exist between the chemistries. 5-methyl cytidine and uridine modified mRNA resulted in low but detectable levels of IFN—alpha and TNF-alpha production, while 5-methyl cytidine and N1 -methyl pseudouridine modified mRNA resulted in no detectable IFN—alpha and TNF-alpha production.
Consequently, it has been determined that, in addition to the need to review more than one cytokine marker of the activation of the innate immune response, it has surprisingly been found that combinations of modifications provide differing levels of cellular se in production and immune activation). The modification, Nl-methyl pseudouridine, in this study has been shown to convey added protection over the standard combination of 5-methylcytidjne/pseudouridjne explored by others resulting in twice as much protein and almost 150 fold reduction in immune activation (TNF-alpha).
Given that PBMC contain a large array of innate immune RNA recognition sensors and are also capable of protein translation, it offers a useful system to test the interdependency ofthese two pathways. It is known that mRNA translation can be negatively affected by activation of such innate immune pathways (Kariko et a1. Immunity (2005) 23:165-175; Warren et al. Cell Stem Cell (2010) 72618-630). Using PBMC as an in vitro assay system it is possible to ish a ation between translation (in this case G-CSF n production) and cytokine production (in this case exemplified by IFN-alpha and TNT-alpha protein production). Better protein production is correlated with lower induction of innate immune activation pathway, and new chemistries can be judged favorably based on this ratio (Table 6).
In this study, the PC Ratio for the two al modifications, uridine and N1- methyl pseudouridine, both with 5-methy ne was 4742/141=34 as compared to 9944/ l=9944 for the cytokine IFN-alpha. For the ne, TNF-alpha, the two tries had PC Ratios of 153 and 1243, respectively suggesting that for either cytokine, the Nl-methylpseudouridine is the superior ation. In Tables 4 and 5, “NT” means not tested.
Table 4. G—CSF G—CSF: 3 Donor Average (pg/ml) G-CSF 4742 -methyl cytosine/ pseudouridine G-CSF 9944 -methylcytosine/ N 1 -methylpseudouridine Luciferase 1 8 LF2000 1 6 Table 5. IFN-alpha and TNF-alpha IFN—alpha: 3 Donor TNF—alpha: 3 Donor Average (pg/n11) Average (pg/ml) G—CSF 141 3 1 -methyl cytosine/ pseudouridine G—CSF 1 8 -methylcytosine/ Nl-methylpseudouridine P(I)P(C) 1 104 NT R—848 NT 1477 LF2000 17 25 Table 6. G—CSF to Cytokine Ratios G—CSF/ IFN—alpha (ratio) G—CSF/TNF—alpha (ratio) S-methyl 5-methylcytosine/ 5-methy1 5-mcthylcytosine/ cytosine/ N1- cytosine/ N1- pseudouridine methylpseudouridine pseudouridine methylpseudouridine PC Ratio 34 9944 153 1243 Example 65. Chemical Modification Ranges of Modified mRNA Modified sides such as, but not limited to, the chemical modifications 5- methylcytosine and pseudouridine have been shown to lower the innate immune response and increase expression of RNA in mammalian cells. Surprisingly and not previously known, the effects manifested by these chemical modifications can be ed when the amount of chemical modification of a particular nucleotide is less than 100%. usly, it was believed that the benefit of chemical modification could be derived using less than complete ement of a modified nucleoside and published reports t no loss of benefit until the level of tution with a modified nucleoside is less than 50% (Kariko et al., Immunity (2005) 23:165-175).
However, it has now been shown that the benefits of chemical modification are directly correlated with the degree of chemical modification and must be considered in View of more than a single measure of immune se. Such benefits include enhanced n production or mRNA translation and reduced or avoidance of stimulating the innate innnune response as ed by cytokine profiles and metrics of immune response triggers.
Enhanced mRNA translation and reduced or lack of innate immune stimulation are seen with 100% tution with a modified nucleoside. Lesser tages of substitution result in less mRNA translation and more innate immune stimulation, with unmodified mRNA g the lowest translation and the highest innate immune stimulation.
In Vitro PBMC Studies: Percent modification 480 ng of G—CSF mRNA modified with 5-methylcytosine (5mC) and pseudouridine (pseudoU) or unmodified G—CSF mRNA was transfected with 0.4 uL of Lipofectamine 2000 into peripheral blood mononuclear cells (PBMC) from three normal blood donors (D1, D2, and D3). The G—CSF mRNA (SEQ ID NO: 1; polyA tail of approximately 160 nucleotides not shown in sequence; ’cap, Capl) was completely modified with 5mC and pseudo (100% modification), not modified with 5mC and pseudo (0% modification) or was partially modified with 5mC and pseudoU so the mRNA would contain 75% modification, 50% modification or 25% ation. A control sample of Luciferase (mRNA sequence shown in SEQ ID NO: 3; polyA tail of approximately 160 nucleotides not shown in sequence; 5’cap, Capl; fully modified 5meC and pseudoU) was also analyzed for G—CSF expression. For TNF-alpha and IFN—alpha l samples of Lipofectamine2000, LPS, R—848, Luciferase (mRNA sequence shown in SEQ ID NO: 3; polyA tail of approximately 160 nucleotides not shown in sequence; 5’cap, Capl; fully modified 5mC and pseudo), and P(I)P(C) were also analyzed. The supernatant was harvested and run by ELISA 22 hours after ection to determine the protein expression. The expression of G-CSF is shown in Table 7 and the expression of IFN—alpha and TNF-alpha is shown in Table 8. The expression of IFN-alpha and TNF-alpha may be a secondary effect from the transfection of the G-CSF mRNA.
Tables 7, 8 and show that the amount of chemical modification of G-CSF, interferon alpha (lFN-alpha) and tumor is factor-alpha (TNF-alpha) is titratable when the mRNA is not fully modified and the titratable trend is not the same for each target.
As mentioned above, using PBMC as an in vitro assay system it is possible to establish a correlation between translation (in this case G-CSF protein production) and cytokine production (in this case exemplified by IFN—alpha protein production). Better protein production is correlated with lower induction of innate immune activation pathway, and the percentage modification of a chemistry can be judged favorably based on this ratio (Table 9). As calculated from Tables 7 and 8 and shown in Table 9, full modification with 5-methylcytidine and pseudouridine shows a much better ratio of n/cytokine production than without any modification (natural G—CSF anNA) (lOO-fold for IFN-alpha and d for pha). Partial modification shows a linear onship with increasingly less modification resulting in a lower protein/cytokine ratio.
Table 7. G—CSF Expression G—CSF Expression (pg/ml) D1 D2 D3 100% ation 1968.9 2595.6 2835.7 75% modification 566.7 631.4 659.5 50% ation 188.9 187.2 191.9 % modification 139.3 126.9 102.0 0% modification 194.8 182.0 183.3 Luciferase 90.2 0.0 22.1 Table 8. IFN-alpha and TNF—alpha Expression IFN—alpha Expression (pg/ml) TNF—alpha Expression (pgiml) D1 D2 D3 D1 D2 D3 100% modification 336.5 78.0 46.4 115.0 15.0 11.1 75% modification 339.6 107.6 160.9 107.4 21.7 11.8 50% modification 478.9 261.1 389.7 49.6 24.1 10.4 % modification 564.3 400.4 670.7 85.6 26.6 19.8 0% modification 1421.6 810.5 1260.5 154.6 96.8 45.9 LPS 0.0 0.6 0.0 0.0 12.6 4.3 R—848 0.5 3.0 14.1 655.2 989.9 420.4 P(I)P(C) 130.8 297.1 585.2 765.8 2362.7 1874.4 Lipid only 1952.2 866.6 855.8 248.5 82.0 60.7 Table 9. PC Ratio and Effect of Percentage of Modification % Modification Average e Average G—CSF/ IFN- G-CSFKTNF— G-CSF IFN-a TNF—a alpha alpha ) (pg/ml) (pg/ml) (PC ratio) (PC ratio) 100 2466 153 47 16 52 75 619 202 47 3.1 13 50 189 376 28 0.5 6.8 122 545 44 0.2 2.8 0 186 1164 99 0.16 1.9 Example 66. Modified RNA transfected in PBMC 500 ng of G—CSF mRNA modified with 5-methylcytosine (SmC) and pseudouridine (pseudoU) or unmodified G—CSF mRNA was transfected with 0.4 uL of Lipofectamine 2000 into peripheral blood mononuclear cells (PBMC) from three normal blood donors (D1, D2, and D3). The G-CSF mRNA (SEQ ID NO: 1; polyA tail of approximately 160 nucleotides not shown in sequence; ’cap, Capl) was completely modified with SmC and pseudo (100% modification), not modified with SmC and pseudo (0% modification) or was partially modified with SmC and pseudoU so the mRNA would contain 50% modification, 25% modification, 10% modification, %5 modification, 1% modification or 0.1% modification. A control sample of mCherry (mRNA sequence shown in SEQ ID NO: 6; polyA tail of imately 160 nucleotides not shown in sequence; 5’cap, Cap]; fully modified 5meC and pseudouridine) and G—CSF fully d with ylcytosine and pseudouridine (Control G—CSF) was also analyzed for G—CSF sion. For tumor is factoralpha (TNF-alpha) and interferon-alpha lpha) control samples of Lipofectamine2000, LPS, R- 848, Luciferase (mRNA ce shown in SEQ ID NO: 3; polyA tail of approximately 160 nucleotides not shown in sequence; 5’cap, Cap]; fully modified SmC and pseudo), and P(I)P(C) were also analyzed. The supernatant was harvested 6 hours and 18 hours after transfection and run by ELISA to determine the protein expression. The expression of G—CSF, IFN—alpha, and TNF-alpha for Donor l is shown in Table 10, Donor 2 is shown in Table 11 and Donor 3 is shown in Table 12.
Full 100% modification with 5-methylcytidine and pseudouridine resulted in the most protein translation (G—CSF) and the least amount of cytokine produced across all three human PBMC . Decreasing s of modification results in more cytokine production (IFN-alpha and TNF-alpha), thus further highlighting the importance of fully modification to reduce cytokines and to improve protein translation (as evidenced here by G-CSF production).
Table 10. Donor 1 G—CSF (pg/mL) IFN-alpha (pg/mL) TNF-alpha (pg/mL) 6 hours 18 hours 6 hours 18 hours 6 hours 18 hours 100% Mod 1815 2224 1 13 0 0 75% Mod 591 614 0 89 0 0 50% Mod 172 147 0 193 0 0 % Mod 1 1 1 92 2 219 0 0 % Mod 138 138 7 536 18 0 1% Mod 199 214 9 660 18 3 0.1% Mod 222 208 10 597 0 6 0 % Mod 273 299 10 501 10 0 Control G-CSF 957 1274 3 123 18633 1620 mChen'y 0 0 0 10 0 0 Untreated N/A N/A 0 0 1 1 Table 11. Donor 2 G—CSF (p lmL) IFN—alpha (pg/mL) TNF—alplla ) 6 hours 18 hours 6 hours 18 hours 6 hours 18 hours 100% Mod 2184 2432 0 7 0 11 75% Mod 93 5 958 3 130 0 0 50% Mod 192 253 2 625 7 23 % Mod 153 158 7 464 6 6 % Mod 203 223 25 700 22 39 1% Mod 288 275 27 962 51 66 0.1% Mod 318 288 33 635 28 5 0%Mod 389 413 26 748 1 253 Control G-CSF 1461 1634 1 59 481 814 mCherry 0 7 0 1 0 0 Untreath N/A N/A 1 0 0 0 Table 12. Donor 3 G—CSF (p /mL) IFN—alpha (pg/mL) TNF—alpha (pg/mL) 6 hours 18 hours 6 hours 18 hours 6 hours 18 hours 100% Mod 6086 7549 7 658 11 11 75% Mod 2479 2378 23 752 4 35 50% Mod 667 774 24 896 22 18 % Mod 480 541 57 1557 43 115 % Mod 838 956 159 2755 144 123 1% Mod 1108 1197 235 3415 88 270 011% Mod 1338 1177 191 2873 37 363 0 % Mod 1463 1666 215 3793 74 429 Control G-CSF 3272 3603 16 1557 731 9066 mCherry 0 0 2 645 0 0 Untreated N/A N/A 1 1 0 8 e 67. Microames Reverse Mutation Screen of Modifications Background and Methods The microames screen is a version of the full Ames preincubation assay. It detects both frameshift and base-pair substitution mutations using four Salmonella tester strains (TA97a, TA98, TA100 and TA1535) and one Escherichia coli strain (WP2 uvrA pKMlOl). Strains TA97a and TA98 detect frameshift mutations, and TA100, TA1535 and WP2 uvrA pKMlOl detect base-pair substitution mutations. This scaled-down Ames test uses minimal compound, is conducted with and without metabolic activation (S9 fraction), and uses multiwell plates. This teste is a microbial assay to detect the mutagenic ial of test nds.
The microAmes screen for ylcytidine, Pseudouridine or N’-methylpseudouridine test article was tested in duplicate with strains TA97a, TA98, TA100, TA1535 and WP2 uvrA pKMlOl in the presence and absence of a metabolic activation system (AROCLORTM 1254 induced rat liver S9 microsomal on) at 0.25, 2.5, 12.5, 25, 75, and 250 ug/well. Positive control nds were used at 4 different concentrations to ensure the assay system was sensitive to known mutagenic compounds. DMSO was used as the vehicle control. Positive and vehicle controls yielded the expected results, demonstrating that the microAmes screen is sufficiently sensitive to detect ns.
Results For 5-methylcytosine, precipitates were not observed with any tester strain either with or without metabolic activation. Cytotoxicity (reduction in the background lawn and/or number of revertants) was not observed in any strain either with or without metabolic tion. There was no increase in the number of ant colonies as ed with the vehicle control in any strain with or t metabolic activation. Therefore, 5-Methylcytidine was not nic up to 250 ug/well in strains TA97a, TA98, TA100, TA1535 and WP2 uvrA pKMlOl with or without metabolic tion under the conditions of the microAmes screen.
Precipitates were not observed with any tester strain either with or without metabolic activation for pseudouridine. Cytotoxicity (reduction in the number of revertants) was observed with strain TA100 without metabolic activation. Cytotoxicity (reduction in the background lawn and/or number of revertants) was not observed in any other strain either with or without metabolic activation. There was no increase in the number of revertant colonies as compared with the vehicle control in any strain with or without metabolic activation. ore, uridine was not mutagenic up to 75 ug/well in strain TA100 without metabolic activation and up to 250 [lg/well in strains TA97a, TA98, TA1535 and WP2 uvrA pKMlOl with or without metabolic activation and strain TA100 Without metabolic activation under the conditions of this microAmes screen.
For the modification, Nl-methylpseudouridine precipitates were not observed with any tester strain either with or without metabolic activation. Cytotoxicity (reduction in the background lawn and/or number of revertants) was not observed in any strain either with or without metabolic activation. There was no increase in the number of revertant colonies as compared with the vehicle control in any strain with or without metabolic activation. Nl-methylpseudouridine was not mutagenic up to 250 ug/well in strains TA97a, TA98, TA100, TA1535 and WP2 uvrA pKMlOl with or without metabolic activation under the conditions of this microAmes screen. NI- methylpseudouridine was found less mutagenic than pseudouridine.
The comparison in this microAMES test of 5 methyl cytidine, pseudouridine, and N1- methylpseudouridine reveal them to be generally tagenic. Of particular note, however, was the difference between pseudouridine and N1 -methylpseudouridine, where pseudouridine did show a cytotoxic response in one bacterial strain where N] -methylpseudouridine did not. These microAMES tests are routinely used as part of the pre-clinical assessment of nd safety and highlight an important difference between N] -methylpseudouridine and pseudouridine.
Exam le 68. Toxici of Nucleoside Tri hos hates TPs The cytotoxicity of l and modified side triphosphates (NTPs) alone or in combination with other bases, was analyzed in human embryonic kidney 293 (HEK293) cells in the e of transfection reagent. HEK293 cells were seeded on 96-well plates at a density of 30,000 cells per well having 0.75ul ofRNAiMAX TM (Invitrogen, Carlsbad, CA) per well at a total well volume of 100ul. 10 ul of the NTPs outlined in Table 12 were combined with 10 ul of lipid on and incubated for 30 minutes to form a complex before 80 ul of the HEK293 cell suspension was added to the NTP complex. ] l and modified NTPs were ected at a concentration of 2.1 nM, 21 nM, 210 nM, 2.1 um, 21 uM, 210 um or 2.1 mM. NTPs in combination were transfected at a total tration ofNTPs of 8.4 nM, 84 nM, 840 nM, 8.4 uM, 84 uM, 840 uM and 8.4 mM. As a control modified G-CSF mRNA (SEQ ID NO: 1; polyA tail of approximately 160 tides not shown in sequence; 5’cap, Cap]; fully modified 5-methylcytosine and pseudouridine) was ected in HEK293 cells at a concentration of 8.4 nM. The cytotoxicity of the NTPs and the modified G-CSF mRNA was assayed at 4, 24, 48 and 72 hours post addition to the I-[EK293 cells using a CYTO OTM assay from a (Madison, WI) following the manufacturer protocol except pippeting was used for lysing the cells instead of shaking the plates.
Table 13 and 14 show the t of viable cells for each of the NTPs, NTP combinations and controls tested. There was no toxicity seen with the individual NTPs as compared to the untreated cells. These data demonstrate that introduction of individual NTPs, including 5- methylcytidine, pseudouridine, and Nl-methylpseudoun'dine, into mammalian cells is not toxic at doses 1,000,000 times an ive dose when introduced as a modified mRNA.
Table 13. Cytotoxicity of Individual NTPs Individual NTP Cytotoxicity Dose Time 2.1 210 2.1 210 21 uM 21 nM 2.1 nM mM uM uM nM 4hr 90.03 85.97 91.20 90.23 90.36 93.21 93.48 24hr 88.42 87.31 86.86 86.81 86.94 87.19 86.44 Aden'm’ 48 hr 93.71 90.55 89.94 89.80 89.17 91.13 92.12 72 hr 97.49 94.81 93.83 94.58 92.22 93.88 95.74 4 hr 90.51 89.88 91.41 90.49 88.95 93.11 93.34 24 hr 86.92 86.33 85.72 86.70 86.12 86.16 85.78 cytosine 48 hr 94.23 87.81 87.28 87.73 85.36 88.95 88.99 72 hr 97.15 92.34 92.22 88.93 88.22 91.80 94.22 4 hr 90.96 90.14 91.36 90.60 90.00 92.84 93.33 24 hr 86.37 85.86 85.93 86.13 86.35 85.50 85.41 Guanine 48 hr 93.83 87.05 88.18 87.89 85.31 87.92 89.57 72 hr 97.04 91.41 92.39 92.30 92.19 92.55 93.72 4 hr 90.97 89.60 91.95 90.90 91.05 92.90 93.15 24 hr 87.68 86.48 85.89 86.75 86.52 87.23 87.63 48 hr 94.39 88.98 89.11 89.44 88.33 88.89 91.28 72 hr 96.82 93.45 93.63 94.60 94.50 94.53 95.51 4 hr 92.09 92.37 91.35 92.02 92.84 91.96 92.26 Pseudonridi 24 hr 88.38 86.68 86.05 86.75 85.91 87.59 87.31 ne 48 hr 88.62 87.79 87.73 87.66 87.82 89.03 91.99 72 hr 96.87 89.82 94.23 93.54 92.37 94.26 94.25 4hr 92.01 91.54 91.16 91.31 92.31 91.40 92.23 -methyl 24 hr 87.97 85.76 84.72 85.14 84.71 86.37 86.35 cytosine 48 hr 87.29 85.94 85.74 86.18 86.44 87.10 88.18 72 hr 96.08 88.10 92.26 90.92 89.97 92.10 91.93 4hr 92.45 91.43 91.48 90.41 92.15 91.44 91.89 Ntfle‘h’fih. 24 hr 88.92 86.48 85.17 85.72 85.89 86.85 87.79 I]: " 0“" ‘ 48 hr 89.84 86.02 87.52 85.85 87.38 86.72 87.81 72 hr 96.80 93.03 93.83 92.25 92.40 92.84 92.98 4 hr 92.77 -- -- -- .. .. .— Untreated 24 hr 87.52 __ __ __ __ -- -- 48hr 92.95 -- -- -- -- .- .. 72hr 96.97 -- -- -- .. .- ..
Table 14. Cytotoxicity of NTPs in Combination NTP Combination Cytotoxicity Dose Time 8.4 840 8.4 840 8.4 84 “M 84 “M mM uM uM nM nM Pseudouridine/ 4 hr 92.27 92.04 91.47 90.86 90.87 91.10 91.50 — 24 hr 88.51 86.90 86.43 88.15 88.46 86.28 87.51 methylcytosine 48 hr 88.30 87.36 88.58 88.13 87.39 88.72 90.55 / ““3"”, 72 hr 96.53 94.42 94.31 94.53 94.38 94.36 93.65 Guanine Nl-methyl 4hr 92.31 91.71 91.36 91.15 91.30 90.86 91.38 pseudouridine/ 24 hr 88.19 87.07 86.46 87.70 88.13 85.30 87.21 — 48 hr 87.17 86.53 87.51 85.85 84.69 87.73 86.79 methylcytosine lAdenine/ 72 hr 96.40 94.88 94.40 93.65 94.82 92.72 93.10 Guanine 4 hr na na na na na na 92.63 G—CSF 24 hr na na na na na na 87.53 48 hr na na na na na na 91.70 mRNA 72 hr na na na na na na 96.36 Example 69. Innate Immune Response Study in BJ Fibroblasts Human primary foreskin fibroblasts (BJ fibroblasts) were obtained from American Type Culture tion (ATCC) (catalog # CRL-2522) and grown in Eagle’s m Essential Medium (ATCC, g # 30-2003) supplemented with 10% fetal bovine serum at 37°C, under 5% C021 BJ fibroblasts were seeded on a 24-well plate at a density of 300,000 cells per well in 0.5 ml of culture medium. 250 ng of modified G-CSF mRNA (mRNA sequence shown in SEQ ID NO: 1; polyA tail of approximately 160 tides not shown in sequence; 5’cap, Capl) fully d with 5-methylcytosine and pseudouridine (Genl) or fully modified with 5-methylcytosine and N1- methylpseudouridine (Gen2) having CapO, Capl or no cap was transfected using Lipofectamine 2000 (Invitrogen, catalog # 11668-019), following manufacturer’s protocol. Control samples of poly I:C (PIC), Lipofectamine 2000 , natural luciferase mRNA (mRNA sequence shown in SEQ ID NO: 3; polyA tail of approximately 160 nucleotides not shown in ce; 5’cap, Capl) and natural G-CSF mRNA were also transfected. The cells were harvested after 18 hours, the total RNA was isolated and DNASE® treated using the RNeasy micro kit (catalog #74004) following the manufacturer’s protocol. 100 ng of total RNA was used for cDNA synthesis using High Capacity cDNA Reverse Transcription kit (catalog #4368814) following the manufacturer’s protocol. The cDNA was then analyzed for the expression of innate immune response genes by quantitative real time PCR using Syerreen in a Biorad CFX 384 instrument following manufacturer’s protocol.
Table 15 shows the expression level of innate immune se transcripts relative to house-keeping gene HPRT (hypoxanthine phosphoribosytransferase) and is expressed as fold-induction relative to HPRT. In the table, the panel of standard metrics includes: RIG-I is retinoic acid ble gene 1, IL6 is interleukin-6, OAS-1 is denylate synthetase 1, IFNb is interferon-beta, AIM2 is absent in ma-2, IFIT-l is interferon-induced protein with tetratricopeptide repeats 1, PKR is protein kinase R, TNFa is tumor necrosis factor alpha and IFNa is interferon alpha.
Table 15. Innate Immune Response Transcript Levels Formulation RIG—I IL6 OAS—1 IFNb AIMZ IFIT—l P101 TNFa IFNa Natlm‘l 71.5 20.6 20.778 11.404 0.251 151.218 16.001 0.526 0.067 Luelferase 2;?“ G‘ 73.3 47.1 19.359 13.615 0.264 142.011 11.667 1.185 0.153 PIC 30.0 2.8 8.628 1.523 0.100 71.914 10.326 0.264 0.063 “ Genl' 0.81 0.22 0.080 0.009 0.008 2.220 1.592 0.090 0.027 (as? Genl' 0.54 0.26 0.042 0.005 0.008 1.314 1.568 0.088 0.038 ail“ Genl' 0.58 0.30 0.035 0.007 0.006 1.510 1.371 0.090 0.040 “ Genz' 0.21 0.20 0.002 0.007 0.007 0.603 0.969 0.129 0.005 (Ci-1:31: Genz' 0.23 0.21 0.002 0.0014 0.007 0.648 1.547 0.121 0.035 $ng Genz' 0.27 0.26 0.011 0.004 0.005 0.678 1.557 0.099 0.037 Lipo 0.27 0.53 0.001 0 0.007 0.954 1.536 0.158 0.064 Example 70. In vivo detection of Innate Immune Response In an effort to distringuish the ance of ent chemical modification ofmRNA on in vivo protein production and ne response in viva, female BALB/C mice (n:5) are injected intramuscularly with G—CSF mRNA (GCSF mRNA unmod) (mRNA sequence shown in SEQ ID NO: I; polyA tail of approximately 160 nucleotides not shown in sequence;) with a S’cap of Cap] G-CSF mRNA fully modified with 5-methylcytosine and pseudouridine (GCSF mRNA 5mc/pU), G- CSF mRNA fully modified with 5-methylcytosine and Nl-methylpseudouridine with (GCSF mRNA pU) or Without a 5’ cap (GCSF mRNA 5mc/N1 pU no cap) or a l of either R848 or % e as described in Table 16.
Table 16. Dosing Chart Formulation Route Dose (ug/mouse) Dose (ul) GCSF mRNA unmod I.M. 200 50 GCSF mRNA 5mc/pU I.M. 200 50 GCSF mRNA 200 50 SmofNlpU ' ' GCSF mRNA 200 50 U no cap ' ' R848 I.M. 75 50 % sucrose I.M. - 50 Untreated I.M. - - Blood is collected at 8 hours after dosing. Using ELISA the protein levels of G-CSF, TNF-alpha and IFN-alpha is determined by ELISA. 8 hours after , muscle is collected from the injection site and quantitative real time polymerase chain reaction (QPCR) is used to determine the mRNA levels of RIG-I, PKR, AIM-2, IFIT-l, OAS-2, MDA-S, IFN—beta, TNF—alpha, IL-6, G— CSF, CD45 in the muscle.
Example 71. In vivo detection of Innate Immune Response Study Female BALB/C mice (n:5) were injected intramuscularly with G-CSF mRNA (GCSF mRNA unmod) (mRNA sequence shown in SEQ ID NO: 1; polyA tail of approximately 160 nucleotides not shown in ce;) with a 5’cap of Capl G—CSF mRNA fully modified with 5- methylcytosine and pseudouridine (GCSF mRNA 5mc/pU), G—CSF mRNA fully modified with 5- methylcytosine and Nl-methylpseudouridine with (GCSF mRNA pU) or without a 5’ cap (GCSF mRNA 5mc/Nl pU no cap) or a control of either R848 or 5% sucrose as described in Table 17‘ Blood is collected at 8 hours after dosing and using ELISA the protein levels of G—CSF and interferon-alpha (IFN-alpha) is determined by ELISA and are shown in Table 17.
As shown in Table 17, unmodified, 5mc/pU, and 5mc/N1pU modified G—CSF mRNA resulted in human G—CSF expression in mouse serum. The uncapped 5mC/N1pU modified G-CSF mRNA showed no human G—CSF expression in serum, highlighting the importance of having a 5’ cap structure for protein translation.
As expected, no human G—CSF protein was expressed in the R848, 5% sucrose only, and untreated groups. Importantly, cant differences were seen in cytokine production as measured by mouse IFN-alpha in the serum. As expected, unmodified G-CSF mRNA demonstrated a robust cytokine response in viva er than the R848 ve control). The 5mc/pU modified G-CSF mRNA did show a low but detectable cytokine response in viva, while the 5mc/N lpU modified mRNA showed no detectable IFN-alpha in the serum (and same as vehicle or untreated animals).
] Also, the response of 5mc/NlpU modified mRNA was the same regardless of whether it was capped or not. These in viva results reinforce the conclusion that 1) that unmodified mRNA produce a robust innate immune response, 2) that this is reduced, but not abolished, through 100% incorporation of 5mc/pU modification, and 3) that incorporation of 5mc/N1pU modifications results in no able cytokine response.
Lastly, given that these injections are in 5% sucrose (which has no effect by itself), these result should accurately reflect the immunostimulatory potential of these modifications.
From the data it is evident that NlpU d molecules produce more protein while concomitantly having little or no effect on IFN—alpha expression. It is also evident that capping is required for protein production for this chemical modification. The Protein: Cytokine Ratio of 748 as compared to the PC Ratio for the unmodified mRNA (PC:9) means that this chemical modification is far or as related to the effects or biological implications associated with IFN—alpha.
Table 17. Human G—CSF and Mouse IFN-alpha in serum Formulation Route Dose Dose G—CSF IFN—alpha PC (ug/mouse) (ul) protein expression Ratio (Pg/ml) ) GCSF mRNA unmod I.M. 200 50 605.6 67.01 9 GCSF mRNA 5mc/pU I.M. 200 50 356.5 8.87 40 GCSF mRNASmc/NlpU I.M. 200 50 748.1 0 748 GCSF mRNASmc/NlpU no cap I.M. 200 50 6.5 0 65 R848 I.M. 75 50 3.4 40.97 .08 % sucrose I.M. - 50 0 1.49 0 Untreated I.M. - - 0 0 0 e 72: In Vivo Delivefl Using Lipoplexes A. Human G—CSF d RNA A formulation containing 100 ug of one of two versions of modified human G-CSF mRNA (mRNA sequence shown in SEQ ID NO: 1; polyA tail of approximately 160 nucleotides not shown in sequence; 5’cap, Capl) (G—CSF fully modified with 5-methylcytosine and uridine (G-CSF) or G-CSF fully modified with 5-methylcytosine and N1 -methyl-pseudouridine (G-CSF-Nl) lipoplexed with 30% by volume ofRNAIMAXTM and red in 150 uL intramuscularly (I.M.) and in 225uL intravenously (I.V.) to C57/BL6 mice.
Three control groups were administered either 100 pg of modified luciferase mRNA (IVT cDNA sequence shown in SEQ ID NO: 2; mRNA sequence shown in SEQ ID NO: 3, polyA tail of approximately 160 nucleotides not shown in sequence, 5’cap, Cap], fully modified with 5- methylcytosine at each cytosine and pseudouridine replacement at each uridine site) intramuscularly (Luc-unsp I.M.) or 150 pg of modified luciferase mRNA intravenously (Luc-unsp I.V.) or 150 uL of the formulation buffer intramuscularly (Buffer I.M.). 6 hours after administration of a formulation, serum was collected to measure the amount of human G—CSF protein in the mouse serum by human G-CSF ELISA and the results are shown in Table 18.
These results demonstrate that both 5-methylcytosine/pseudouridine and 5- methylcytosine/N 1 -methylpseudouridine modified human G—CSF mRNA can result in specific human G—CSF protein expression in serum when delivered via I.V. or I.M. route of stration in a lipoplex formulation.
Table 18. Human G—CSF in Serum (I.M. and I.V. Injection Route) Formulation Route G—CSF (pg/ml) G-CSF I.M. 85.6 Nl I.M. 40.1 G-CSF I.V. 3 1 .0 G-CSF-Nl I.V. 6.1 Luc-unsp I.M. 0.0 Luc-unsp I.V. 0.0 Buffer I.M. 0.0 B. Human G—CSF Modified RNA Comparison A formulation containing 100 pg of either modified human G—CSF mRNA lipoplexed with 30% by volume ofRNAIMAXTM with a S-methylcytosine (5mc) and a pseudouridine (\II) modification -Genl-Lipoplex), modified human G—CSF mRNA with a 5mc and \[I modification in saline (G—CSF-Genl-Saline), d human G—CSF mRNA with a N1—5- methylcytosine (N l-Smc) and a w modification lipoplexed with 30% by volume ofRNAIMAXTM (G-CSF-Gen2-Lipoplex), d human G—CSF mRNA with a Nl-Smc and \V modification in saline (G-CSF-GenZ-Saline), d luciferase with a 5mc and w modification lipoplexcd with % by volume ofRNAIMAXTM (Luc-Lipoplex), or luciferase mRNA fully modified with 5mc and w modifications in saline (Luc-Saline) was delivered ir1tramuscularly(I.M.) or subcutaneously (SC) and a control group for each method of stration was giving a dose of 80uL of the formulation buffer (F. Buffer) to C57/BL6 mice. 13 hours post injection serum and tissue from the site of injection were collected from each mouse and analyzed by G-CSF ELISA to compare human G-CSF protein levels. The results of the human G-CSF protein in mouse serum from the intramuscular administration and the aneous administration s are shown in Table 19.
These results trate that 5-methylcytosine/pseudouridine and ylcytosine/N l- methylpseudouridine d human G—CSF mRNA can result in specific human G—CSF protein expression in serum when delivered via I.M. or S.C. route of administration whether in a saline formulation or in a lipoplex formulation. As shown in Table 19, 5-methylcytosine/N1- methylpseudouridine modified human G-CSF mRNA generally demonstrates sed human G- CSF protein production relative to 5-methylcytosine/pseudouridine modified human G-CSF mRNA.
Table 19. Human G—CSF Protein in Mouse Serum G—CSF . /ml) Formulation I.M. Iujgllgion Route S.C. Injection Route G—CSF-Genl -LipopleX 13.988 42.855 GCSF-Genl -saline 9.375 4.614 GCSF-Gen2-lipoplex 75 .572 32.107 GCSF-GenZ-saline 20.190 45.024 Luc lipoplex 0 3.754 Luc saline 0.0748 0 F. Buffer 4.977 2.156 e 73. Multi—Site stration: Intramuscular and Subcutaneous Human G-CSF modified mRNA (mRNA sequence shown in SEQ ID NO: 1; polyA tail of approximately 160 nucleotides not shown in sequence; 5’cap, Capl) modified as either Gen] or Gen2 (5-methylcytosine (5mc) and a pseudouridine (w) modification, G-CSF-Genl; or Nl—5— methylcytosine (N l-5mc) and a w modification, G—CSF-Gen2) and formulated in saline were delivered to mice Via intramuscular (IM) or subcutaneous (SC) injection. Injection of four doses or 2X 50ug (two sites) daily for three days (24 hrs interval) was performed. The fourth dose was administered 6 hrs before blood collection and CBC analysis. Controls included Luciferase (cDNA sequence for IVT shown in SEQ 1]) NO: 2; mRNA sequence shown in SEQ ID NO: 3, polyA tail of imately 160 nucleotides not shown in ce, 5’cap, Capl, fully modified with 5- methylcytosine at each cytosine and pseudouridine replacement at each uridine site) or the formulation buffer (F.Buffer). The mice were bled at 72 hours after the first mRNA injection (6 hours after the last mRNA dose) to determine the effect of mRNA-encoded human G-CSF on the neutrophil count. The dosing regimen is shown in Table 20 as are the resulting neutrophil counts (thousands/uL). In Table 20, an asterisks (*) indicate statistical significance at p<0.05.
For intramuscular administration, the data reveal a four fold increase in neutrophil count above control at day 3 for the Genl G-CSF mRNA and a two fold increase for the Gen2 G-CSF mRNA. For subcutaneous stration, the data reveal a two fold increase in neutrophil count above control at day 3 for the Gen2 G-CSF mRNA.
These data demonstrate that both 5-methylcytidine/pseudouridine and 5- methylcytidine/Nl -methylpseudouridine-modified mRNA can be biologically active, as evidenced by specific increases in blood neutrophil counts.
Table 20. Dosing Regimen Gr. Treatment Route N Dose (pig/mouse) Dose Dosing phil = Vol. Vehicle Thous/uL (”l/mouse) l G—CSF (Genl) I.M 5 2X50ug (four doses) 50 F. buffer 840* 2 G—CSF (Genl) SC 5 2X50ug (four doses) 50 F. buffer 430 3 G—CSF (Gen2) I.M 5 2X50ug (four doses) 50 F. buffer 746* 4 G—CSF (Gen2) SC 5 2X50ug (four doses) 50 F. buffer 683 Luc (Genl) I.M. 5 2X50ug (four doses) 50 F. buffer 201 6 Luc (Genl) SC. 5 2X50ug (four doses) 50 F. buffer 307 7 Luc (Gen2) I.M 5 2X50ug (four doses) 50 F. buffer 336 8 Luc (Gen2) SC 5 2X50ug (four doses) 50 F. buffer 357 9 F. Buffer I.M 4 0 (four doses) 50 F. buffer 245 F. Buffer SC. 4 0 (four doses) 50 F. buffer 509 11 Untreated 312 Example 74. Intravenous Administration 2] Human G-CSF modified mRNA (mRNA ce shown in SEQ ID NO: 1; polyA tail of approximately 160 nucleotides not shown in sequence; S’cap, Capl) d with S-methylcytosine (5mc) and a pseudouridine (w) modification (Genl); or having no modifications and formulated in % lipoplex (RNAIMAXTM) were delivered to mice at a dose of 50 ug RNA and in a volume of 100 ul via intravenous (IV) injection at days 0, 2 and 4. Neutrophils were measured at days 1, 5 and 8. Controls included non-specific ian RNA or the formulation buffer alone (F.Buffer). The mice were bled at days 1, 5 and 8 to determine the effect of mRNA-encoded human G-CSF to increase neutrophil count. The dosing regimen is shown in Table 21 as are the resulting neutrophil counts (thousands/uL; K/uL).
For intravenous administration, the data reveal a four to five fold increase in neutrophil count above control at day 5 with G-CSF modified mRNA but not with unmodified G-CSF mRNA or non-specific controls. Blood count returned to ne four days after the final injection. No other changes in leukocyte populations were observed.
In Table 21, an asterisk (*) indicates statistical cance at p<0.001 compared to buffer.
These data demonstrate that ex-formulated 5-methylcytidine/pseudouridine- modified mRNA can be biologically active, when delivered through an I.V. route of administration as evidenced by specific increases in blood phil counts. No other cell subsets were significantly altered. Unmodified G—CSF mRNA similarly administered showed no cologic effect on neutrophil counts.
Table 21. Dosing Regimen Gr. Treatment N Dose Dosing Neutrophil Vol. Vehicle K/uL (”l/mouse) 1 G-CSF (Genl) Day 1 5 100 10% lipoplex 2.91 2 G-CSF (Genl) Day 5 5 100 10% lipoplex 532* 3 G-CSF (Genl) Day 8 5 100 10% lipoplex 2.06 4 G-CSF (no modification) Day 1 5 100 10% lipoplex 1.88 G-CSF (no ation) Day 5 5 100 10% lipopleX 1.95 6 G-CSF (no modification) Day 8 5 100 10% eX 2.09 7 RNA control Day 1 5 100 10% lipopleX 2.90 8 RNA control Day 5 5 100 10% lipopleX 1.68 9 RNA control Day 8 4 100 10% lipopleX 1.72 F. Buffer Day 1 4 100 10% lipopleX 2.51 11 F. Buffer Day 5 4 100 10% lipopleX 1.31 12 F. Buffer Day 8 4 100 10% lipopleX 1.92 Example 75: Routes of Administration Studies were performed to investigate split dosing using different routes of administration.
Studies utilizing multiple subcutaneous or uscular injection sites at one time point were designed and performed to investigate ways to increase d mRNA drug exposure and improve protein production. In addition to detection of the expressed protein product, an assessment of the physiological function of proteins was also determined through analyzing samples from the animal tested.
Surprisingly, it has been determined that split dosing of modified mRNA produces greater protein production and phenotypic responses than those produced by single unit dosing or osing schemes.
The design of a split dose experiment involved using human erythropoietin (EPO) d mRNA (mRNA sequence shown in SEQ ID NO: 5; polyA tail of approximately 160 nucleotides not shown in sequence; 5’cap, Capl) or luciferase modified mRNA (mRNA ce shown in SEQ ID NO: 3; polyA tail of approximately 160 nucleotides not shown in sequence; 5’cap, Capl) administered in buffer alone or formulated with 30% lipoplex (RNAIMAXTM). The dosing vehicle (buffer) consisted of 150mM NaCl, 2 mM CaClz, 2 mM Na+-phosphate (1.4mM monobasic sodium phosphate; 0.6mM dibasic sodium phosphate), and 0.5 mM EDTA, pH 6.5. The pH was adjusted using sodium hydroxide and the final solution was filter ized. The mRNA was modified with Smethle (SmeC) at each cytosine and pseudouridine replacement at each uridine site. 4 mice per group were dosed intramuscularly (I.M.), intravenously (I.V.) or aneously (S.C.) by the dosing chart outlined in Table 22. Serum was collected 13 hours post ion from all mice, tissue was collected from the site of ion from the intramuscular and subcutaneous group and the spleen, liver and kidneys were collected from the intravenous group.
The results from the intramuscular group and the subcutaneous group results are shown in Table 23.
Table 22. Dosing Chart Group Treatment Route Dose of modified mRNA Total Dosing Dose Vehicle 1 Lipoplex-human EPO I.M. 4 x 100 ug W 30% Lipoplex 4x70 111 ex modified mRNA 2 Lipoplex-human EPO I.M. 4 x 100 ug 4x70 111 Buffer modified mRNA 3 Lipoplex-human EPO SC. 4 x 100 ug W 30% Lipoplex 4x70 111 Lipoplex modified mRNA 4 Lipoplex-human EPO SC. 4 x 100 ug 4x70 111 Buffer modified mRNA Lipoplex-human EPO IV. 200 ug W 30% Lipoplex 140 111 Lipoplex modified mRNA 6 Lipoplexed-Luciferase I.M. 100 ug W 30% Lipoplex 4x70 111 Lipoplex modified mRNA 7 Lipoplexed-Luciferase I.M. 100 ug 4x70 111 Buffer modified mRNA 8 Lipoplexed-Luciferase SC. 100 ug W 30% Lipoplex 4x70 111 Lipoplex modified mRNA 9 Lipoplexed-Luciferase SC. 100 ug 4x70 111 Buffer modified mRNA Lipoplexed-human EPO IV. 200 ug -- 30% Lipoplex 140 ul Lipoplex modified mRNA 1 1 Formulation Buffer I.M. 4x multi dosing 4x70 ul Buffer Table 23. Human EPO Protein in Mouse Serum (I.M. Injection Route) EPO (pg/ml) Formulation.
I.M. Injection Route S.C. Injection Route Epo-Lipoplex 67.1 2.2 Luc-Lipoplex 0 0 Epo-Saline 100.9 11.4 Luo-Saline 0 0 Formulation Buffer 0 0 Example 76: In Vivo Deliveg Using Vaging Lipid Ratios d mRNA was delivered to C57/BL6 mice to evaluate varying lipid ratios and the resulting protein expression. Formulations of 100ug modified human EPO mRNA (mRNA sequence shown in SEQ ID NO: 5; polyA tail of approximately 160 nucleotides not shown in sequence; 5’cap, Cap]; fully modified with 5-methylcytosine and pseudouridine) lipoplexed with %, 30% or 50% RNAIMAXTM, 100pg modified luciferase mRNA (IVT cDNA sequence shown in SEQ ID NO: 2; mRNA sequence shown in SEQ ID NO: 3, polyA tail of approximately 160 tides not shown in sequence, 5 ’cap, Cap] modified with 5-methylcytosine at each , fully cytosine and pseudouridine replacement at each uridine site) exed with 10%, 30% or 50% RNAIMAXTM or a formulation buffer were administered intramuscularly to mice in a single 70 pl dose. Serum was collected 13 hours post injection to undergo a human EPO ELISA to determine the human EPO n level in each mouse. The results of the human EPO ELISA, shown in Table 24, show that modified human EPO expressed in the muscle is secreted into the serum for each ofthe different percentage of RNAIMAXTM.
Table 24. Human EPO Protein in Mouse Serum (IM Injection Route) Formulation EPO ) Epo W 10% RNAiMAX 11.4 Luc W 10% RNAiMAX 0 Epo W 30% RNAiMAX 27.1 Luc W 30% RNAiMAX 0 Epo W 50% RNAiMAX 19.7 Luc W 50% RNAiMAX 0 F. Buffer 0 Example 77: In Vivo Delivefl of Modified RNA in Rats 1] Protein production of modified mRNA was evaluated by delivering d G-CSF mRNA or modified Factor IX mRNA to female Sprague Dawley rats (n=6). Rats were injected with 400 ug in 100 111 of G-CSF mRNA (mRNA sequence shown in SEQ ID NO: 1; polyA tail of approximately 160 nucleotides not shown in sequence; 5’cap, Capl) fully modified with 5- methylcytosine and pseudouridine (G-CSF Genl), G—CSF mRNA fully modified with 5- methylcytosine and Nl-methylpseudouridine (G-CSF Gen2) or Factor IX mRNA (mRNA sequence shown in SEQ ID NO: 6; polyA tail of approximately 160 nucleotides not shown in sequence; 5’cap, Capl) fully modified with 5-methylcytosine and pseudouridine r IX Genl) reconstituted from the lyophilized form in 5% sucrose. Blood was collected 8 hours after ion and the G-CSF protein level in serum was measured by ELISA. Table 25 shows the G—CSF protein levels in serum after 8 hours.
These results demonstrate that both G-CSF Gen 1 and G—CSF Gen 2 modified mRNA can produce human G-CSF protein in a rat following a single intramuscular injection, and that human G- CSF protein production is improved when using Gen 2 chemistry over Gen 1 chemistry.
Table 25. G—CSF Protein in Rat Serum (I.M. Injection Route) Formulation G—CSF protein (pg/ml) G-CSF Genl 19.37 G-CSF GenZ 64.72 Factor IX Gen 1 2.25 Example 78. Chemical Modification: In vitro s A. In vitro Screening in PBMC 500 ng of G—CSF (mRNA sequence shown in SEQ ID NO: 1; polyA tail of approximately 160 nucleotides not shown in sequence; 5’cap, Capl) mRNA fully modified with the al modification outlined Tables 26 and 27 was transfected with 0.4 uL Lipofectamine 2000 into peripheral blood mononuclear cells (PBMC) from three normal blood donors. Control samples ofLPS, R848, P(I)P(C) and mCherry (mRNA sequence shown in SEQ ID NO: 4; polyA tail of approximately 160 nucleotides not shown in sequence, 5’cap, Capl; fully modified with 5— methylcytosine and pseudouridine) were also ed. The supernatant was harvested and stored frozen until analyzed by ELISA to determine the G—CSF protein expression, and the induction of the cytokines interferon-alpha (IFN-ot) and tumor necrosis factor alpha (TNF-(x). The protein expression ofG-CSF is shown in Table 26, the sion of IFN—(x and TNF-(x is shown in Table 27.
The data in Table 26 demonstrates that many, but not all, al modifications can be used to productively produce human G—CSF in PBMC. Of note, 100% Nl-methylpseudouridine substitution demonstrates the highest level of human G-CSF production t lO-fold higher than uridine itself). When Nl-methylpseudouridine is used in combination with 5-methylcytidine a high level of human G-CSF protein is also produced (this is also higher than when pseudouridine is used in combination with 5 methylcytidine).
Given the inverse relationship between protein production and cytokine production in PBMC, a similar trend is also seen in Table 27, where 100% substitution with N1- methylpseudouridine results no ne induction (similar to ection only controls) and pseudouridine shows detectable cytokine induction which is above background.
Other modifications such as N6-methyladenosine and (x-thiocytidine appear to increase cytokine stimulation.
Table 26. Chemical Modifications and G—CSF Protein Expression Chemical Modifications G—CSF Protein Expression (fig/ml) Donor 1 Donor 2 Donor 3 Pseudouridinc 2477 1,909 1,498 -methyluridinc 3 1 8 3 59 345 N1—methylpscudouridinc 2 1 ,495 16,550 12 ,441 2-thiouridinc 93 2 1 ,000 600 4—thiouridinc 5 3 9 1 2 1 8 -mcthoxyuridinc 2,964 1,832 1,800 -mcthylcytosinc and pseudouridinc (1 5‘ set) 2,632 1,955 1,373 -mcthylcytosinc and N1 -mcthylpscudouridinc (1 5‘ set) 10,23 2 7,245 6,214 roguanosine 59 186 177 2’Fluorouridine 1 18 209 191 -mcthylcytosine and pseudouridinc (2nd set) 1 ,682 1 ,3 82 1 ,03 6 -mcthylcytosinc and N1 -mcthylpscudouridinc (2ud set) 9,564 8 ,509 7,141 -bromouridinc 3 14 482 291 -(2-carbomethoxyvinyl)uridinc 77 2 8 6 177 -[3(l -E-propcnylamino)uridinc 541 491 550 a—thiocytidinc 105 264 245 ylcytosinc and pseudouridinc (3"I set) 1 ,595 1 ,43 2 955 Nl-methyladcnosinc 1 82 177 191 N6-methyladcnosinc 1 00 1 68 200 ylcytidinc 291 277 359 N4-acetylcytidinc 50 13 6 36 -formylcytidinc 1 8 205 23 -methylcytosinc and pseudouridinc (4m set) 264 350 1 82 ylcytosinc and N1 -mcthylpscudouridinc (4m set) 9,505 6,927 5,405 LPS 1 ,209 786 636 mCherry 5 1 68 l 64 R848 709 732 636 P(I)P(C) 5 186 182 Table 27. al Modifications and Cytokine Expression Chemical Modifications IFN-a Expression (pg/ml) TNF-a Expression (ngml) Donor 1 Donor 2 Donor 3 Donor 1 Donor 2 Donor 3 Pseudouridine 120 77 171 36 81 126 -methyluridine 245 135 334 94 100 157 N 1 -methylpseudouridine 26 75 13 8 101 106 134 2-thiouridine 100 108 154 133 133 141 4-thiouridine 463 258 659 169 126 254 -methoxyuridine 0 64 13 3 3 9 74 1 1 1 -methylcytosine and 88 94 148 64 89 121 pseudouridine (1St set) -methyloytosine and N1 - 0 60 136 54 79 126 methylpseudouridine (1St set) 2’Fluoroguanosine 107 97 194 91 94 141 2’Fluorou1idine 158 103 178 164 121 156 -methylcytosine and 133 92 167 99 1 1 1 150 pseudouridine (2“‘1 set) -methylcytosine and N1 - 0 66 140 54 97 149 pseudouridine (2“1 set) -bromou1idine 95 86 181 87 106 157 -(2- 0 61 130 40 81 116 cmbomethoxyvinyl)utidine -[3(l—E— 0 58 132 71 90 119 propenylamino)uridine a—thiocytidine 1,13 8 565 695 3 00 2 73 277 -methylcytosine and 8 8 75 150 84 89 130 Jseudouridine (3‘d set) Nl-methyladenosine 322 255 377 256 157 294 N6—methyladenosine 1,935 1,065 1 ,492 1,080 630 857 -methylcytidine 643 3 59 529 176 13 6 193 N4-acetylcytidine 789 593 43 1 263 67 207 -fotmylcytidine 1 80 93 8 8 1 3 6 3 0 40 -methylcytosine and 13 1 2 8 1 8 53 24 29 uridine (4th set) -methylcytosine and N1 - 0 0 0 3 6 14 l 3 methylpseudouridine (4th set) LPS 0 67 146 7,004 3 ,974 4,020 mCherry 100 75 143 67 100 133 R848 674 619 562 11,179 8,546 9,907 P(I)P(C) 470 117 362 249 177 197 B. In vitro ing in HeLa Cells 7] The day before transfection, 20,000 HeLa cells (ATCC no. CCL—2; Manassas, VA) were harvested by ent with Trypsin-EDTA solution (LifeTechnologies, Grand Island, NY) and seeded in a total volume of 100ul EMEM medium (supplemented with 10%FCS and 1x Glutamax) per well in a 96-well cell culture plate (Corning, Manassas, VA). The cells were grown at 370G in % C02 here overnight. Next day, 83 ng of Luciferase modified RNA (mRNA sequence shown in SEQ ID NO: 3; polyA tail of approximately 160 nucleotides not shown in sequence; 5’cap, Capl) with the chemical modification described in Table 28, were d in 10ul final volume of OPTI-MEM (LifeTechnologies, Grand Island, NY). Lipofectamine 2000 (LifeTechnologies, Grand Island, NY) was used as transfection reagent and 0.2 ul were diluted in 10 111 final volume of OPTI- MEM. After 5 minutes of incubation at room temperature, both ons were combined and ted an additional 15 minute at room ature. Then the 20ul combined solution was added to the lOOul cell culture medium containing the HeLa cells and incubated at room temperature.
After 18 to 22 hours of incubation cells expressing luciferase were lysed with 100 ul of Passive Lysis Buffer (Promega, Madison, WI) according to manufacturer instructions. Aliquots of the lysates were transferred to white opaque polystyrene 96-well plates (Coming, Manassas, VA) and combined with 100 ul complete luciferase assay solution ga, Madison, WI). The lysate volumes were adjusted or diluted until no more than 2 mio relative light units (RLU) per well were detected for the est signal producing samples and the RLUs for each chemistry tested are shown in Table 28. The plate reader was a BioTek Synergy H1 (BioTek, Winooski, VT).The background signal of the plates without reagent was about 200 relative light units per well.
These results demonstrate that many, but not all, chemical modifications can be used to productively produce human G—CSF in HeLa cells. Of note, 100% N1 -methylpseudouridjne substitution demonstrates the highest level of human G-CSF production.
Table 28. Relative Light Units of Luciferase Chemical Modification RLU N6-methyladenosine (m6a) 534 S-methylcytidine (m50) 13 8 ,42 8 N4-acetylcytidine (ac4c) 235,412 S-formylcytidine (1'50) 436 S-methylcytosine/pseudouridine, test A1 48,659 S-methylcytosine/N 1 -methylpseudouridine, test A1 190,924 uridine 655 ,63 2 l—methylpseudouridine (m l u) 1 ,5 17,998 2-thiouridine (sZu) 3387 -methoxyuridine (moSu) 253 ,719 ylcytosine/pseudouridine, test B l 3 17,744 -methylcytosine/N l -methylpseudouridine, test B l 265 ,8 7 1 -Bromo-uridine 43 ,2 76 (2 inyl) uridine 531 (3-113 propenyl Amino) uridine 446 -methylcytosine/pseudouridine, test A2 295,824 -methylcytosine/N1-methy1pseudouridine, test A2 233 ,92 1 -methyluridine 50,932 ot-Thio-cytidine 26,358 -methylcytosine/pseudouridine, test B2 481,477 -methylcytosine/N 1 -methylpseudouridine, test B2 271 ,989 -methylcytosine/pseudouridine, test A3 43 8,83 1 -methylcytosine/N1-methylpseudouridine, test A3 277,499 Unmodified Luciferase 2 C. In vitro Screening in Rabbit Reticulocyte Lysates rase mRNA (mRNA sequence shown in SEQ ID NO: 3; polyA tail of approximately 160 nucleotides not shown in sequence; 5’cap, Capl) was modified with the chemical modification listed in Table 29 and were diluted in sterile nuclease-free water to a final amount of 250 ng in 10 ul. The diluted luciferase was added to 40 ul of freshly prepared Rabbit Reticulocyte Lysate and the in vitro translation reaction was done in a standard 1.5 mL polypropylene reaction tube (Thermo Fisher Scientific, Waltham, MA) at 30°C in a dry heating block. The translation assay was done with the Rabbit locyte Lysate (nuclease-treated) kit (Promega, Madison, WI) according to the manufacturer’s instructions. The reaction buffer was supplemented with a one-to- one blend of provided amino acid stock solutions devoid of either Leucine or Methionine resulting in a reaction mix containing sufficient amounts of both amino acids to allow effective in vitro translation.
After 60 minutes of incubation, the reaction was stopped by g the reaction tubes on ice. ts of the in vitro ation reaction containing luciferase modified RNA were transferred to white opaque polystyrene 96-well plates (Corning, Manassas, VA) and combined with 100ul complete luciferase assay solution (Promega, Madison, WI). The s of the in vitro translation reactions were adjusted or diluted until no more than 2 mio relative light units (RLUs) per well were detected for the strongest signal producing s and the RLUs for each chemistry tested are shown in Table 29. The plate reader was a BioTek Synergy H1 (BioTek, ki, VT).The background signal of the plates without reagent was about 200 relative light units per well.
These cell-free translation results very nicely ate with the protein production results in HeLa, with the same modifications generally working or not working in both s. One notable exception is S-formylcytidine modified luciferase mRNA which worked in the cell-free translation system, but not in the HeLa cell-based transfection system A similar difference between the two assays was also seen with 5-formylcytidine modified G-CSF mRNA.
Table 29. ve Light Units of Luciferase Chemical Modification RLU hyladenosine (m6a) 3 98 -methylcytidine (m50) 152,989 N4-acetylcytidine (ac4c) 60,879 -formylcytidine (150) 55,208 -methylcytosine/pseudouridine, test A1 349,398 ylcytosine/N 1 -methylpseudouridine, test A1 205,465 Pseudouridine 587,795 l-methylpseudouridine (ml u) 5 89,75 8 2-thiouridine (sZu) 708 -methoxyuridine (mo5u) 2 8 8 ,647 -methylcytosine/pseudouridine, test B 1 454,662 -methylcytosine/N 1 -methylpseudouridine, test B 1 223 ,73 2 -Bromo-uridine 22 1 ,879 (2 carbovinyl) uridine 225 (3-1E propenyl Amino) uridine 21 1 -methylcytosine/pseudouridine, test A2 558,779 S-methylcytosine/N 1 -methylpseudouridine, test A2 3 3 3 ,082 S-methyluridine 2 14,680 ot-Thio-cytidine 123 ,878 -methylcytosine/pseudouridine, test B2 487,805 S-methylcytosine/N 1 -methylpseudouridine, test B2 154,096 -methylcytosine/pseudouridine, test A3 413 ,535 S-methylcytosine/N 1 -methylpseudouridine, test A3 292,954 Unmodified Luciferase 225,986 Example 79. al Modification: In vivo studies A. In vivo Screening of G—CSF Modified mRNA Balb-C mice (n=4) are intramuscularly ed in each leg with modified G-CSF mRNA (mRNA sequence shown in SEQ ID NO: 1; polyA tail of imately 160 nucleotides not shown in ce; S’cap, Capl), fully modified with the chemical modifications outlined in Table 30, is formulated in leBS. A control of luciferase modified mRNA (mRNA sequence shown in SEQ ID NO: 3; polyA tail of imately 160 nucleotides not shown in sequence; 5 ’cap, Capl; fully modified with pseudouridine and 5-methylcytosine) and a control of PBS are also . After 8 hours serum is collected to ine G-CSF protein levels cytokine levels by ELISA.
Table 30. G—CSF mRNA Chemical Modifications G-CSF Pseudouridine G-CSF 5-methyluridine G-CSF 2-thiouridine G-CSF 4-thiouridine G-CSF 5-methoxyuridine G-CSF 2 ’-fluorouridine G-CSF 5-bromouridine G-CSF 5-[3(1-E-propenylamino)uridine) G-CSF alpha-thio-cytidine G—CSF 5-methylcytidine G-CSF N4-acetylcytidine G—CSF Pseudouridine and 5-methylcytosine G—CSF N1 -methylpseudouridine and 5-methylcytosine Luciferase uridine and 5-methylcytosine PBS None B. In vivo Screening of Luciferase d mRNA Balb-C mice (n:4) were subcutaneously injected with 200ul containing 42 to 103 ug of modified luciferase mRNA (mRNA ce shown in SEQ ID NO: 3; polyA tail of approximately 160 nucleotides not shown in sequence; 5’cap, Capl), fully modified with the chemical modifications outlined in Table 31, was formulated in leBS. A l of PBS was also tested.
The dosages of the modified luciferase mRNA is also outlined in Table 31. 8 hours after dosing the mice were imaged to determine luciferase expression. Twenty minutes prior to imaging, mice were injected intraperitoneally with a D-luciferin solution at 150 mg/kg. Animals were then anesthetized and images were acquired with an IVIS Lumina II g system (Perkin Elmer) Bioluminescence was measured as total flux (photons/second) of the entire mousei As demonstrated in Table 31, all luciferase mRNA modified chemistries demonstrated in viva activity, with the exception of rouridine. In addition l-methylpseudouridine modified mRNA demonstrated very high expression of luciferase (5-fold greater expression than pseudouridine containing mRNA).
Table 31. Luciferase Screening mRNA Chemical Modifications Dose (ug) Dose Luciferase of mRNA volume (ml) expression (photon/second) Luciferase 5-methy1cytidine 83 0 .72 1 .94E+07 Luciferase N4-acetylcytidine 76 0.72 1.1 11307 Luciferase Pseudouridine 95 1.20 1.36E+07 Luciferase l-methylpseudouridine 103 0.72 7.40E+07 rase 5-methoxyuridine 95 1 .22 3 .32+07 Luciferase 5-methyluridine 94 0.86 7.4213706 Luciferase 5-bromouridine 89 1.49 3 .75E"07 Luciferase 2 ’-fluoroguanosine 42 0.72 5.88E7705 Luciferase 2 ’-fluorocytidine 47 0.72 4.2 11337705 Luciferase 2 ’-flurorouridine 59 0. 72 3 47137705 PBS None - 0.72 3.1613705 e 80. In vivo Screening of Combination Luciferase d mRNA Balb-C mice (n:4) were subcutaneously injected with 200ul of 100 ug of modified luciferase mRNA (mRNA sequence shown in SEQ ID NO: 3; polyA tail of imately 160 tides not shown in sequence; S’cap, Capl), fully modified with the chemical modifications outlined in Table 32, was formulated in 1x PBS. A control of PBS was also tested. The dosages of the modified luciferase mRNA is also ed in Table 29. 8 hours after dosing the mice were imaged to determine luciferase expression. Twenty minutes prior to imaging, mice were ed intraperitoneally with a D-luciferin solution at 150 mg/kg. Animals were then anesthetized and images were acquired with an IVIS Lumina II imaging system n Elmer). Bioluminescence was measured as total flux (photons/second) of the entire mouse.
As demonstrated in Table 32, all luciferase mRNA modified chemistries (in combination) demonstrated in viva activity. In addition the presence ofNl-methylpseudouridine in the modified mRNA (with N4-acetylcytidine or 5 methylcytidine) demonstrated higher expression than when the same combinations where tested using with pseudouridine. Taken together, these data trate that Nl-methylpseudouridine containing luciferase mRNA results in improved protein expression in vivo whether used alone (Table 31) or when used in combination with other d nulceotides (Table 32).
Table 32. rase Screening Combinations mRNA Chemical Modifications Luciferase expression (photon/second) rase N4-acetylcytidine/pseudouridine 4. 1 8E+06 Luciferase N4-acetylcytidine/N l -methylpseudouridine 2 . 8 8E+07 Luciferase 5-methy1cytidine/5-methoxyuridine 3 .48E+07 Luciferase 5-methylcytidine/5-methyluridine l .44E+07 Luciferase 5-methylcytidine/where 50% of the uridine is replaced with 2-thiouridine 2.39E+06 Luciferase 5-methylcytidine/pseudouridine -O7 rase 5-methylcytidine/N 1 -methyl-pseudouridine 4. 15E--07 PBS None 3 .59E--05 Example 81. Stabilifl of Modified RNA A. Storage of Modified RNA Stability experiments were conducted to obtain a better understanding of storage conditions to retain the integrity of modified RNA. Unmodified G—CSF mRNA (mRNA sequence shown in SEQ ID NO: 1; polyA tail of approximately 160 nucleotides not shown in ce; 5’cap, Capl), G—CSF mRNA fully modified with 5-methylcytosine and uridine and G-CSF mRNA fully modified with 5-methylcytosine and pseudouridine lipoplexed with 0.75% by volume of RNAIMAXTM was stored at 50°C, 40°C, 37°C, 25°C, 4°C or -20°C. After the mRNA had been stored for 0 hours, 2 hours, 6 hours, 24 hours, 48 hours, 5 days and 14 days, the mRNA was analyzed by gel electrophoresis using a Bio-Rad EXPERIONTM system. The modified, unmodified and lipoplexed G—CSF mRNA was also stored in RNASTABLE® (Biomatrica, Inc. San Diego, CA) at 40°C or water at -80 °C or 40°C for 35 days before being analyzed by gel electrophoresis.
All mRNA samples without stabilizer were stable after 2 weeks after storage at 4°C or - °C. Modified G—CSF mRNA, with or without lipoplex, was more stable than fied G—CSF when stored at 25°C (stable out to 5 days versus 48 hours), 37°C (stable out to 24 hours versus 6 hours) and 50°C (stable out to 6 hours versus 2 hours). fied G—CSF mRNA, modified G- CSF mRNA with or without lipoplex tolerated l2 freeze/thaw cycles. mRNA samples stored in izer at 40°C showed similar stability to the mRNA samples stored in water at -80°C after 35 days whereas the mRNA stored in water at 40°C showed heavy degradation after 18 days.
Example 82. Cell viabilifl in BJ Fibroblasts 2] Human primary foreskin fibroblasts (BJ fibroblasts) were obtained from American Type Culture Collection (ATCC) (catalog #CRL-2522) and grown in Eagle’s Minimum Essential Medium (ATCC, cat# 3) supplemented with 10% fetal bovine serum at 37°C, under 5% C02.
BJ fibroblasts were seeded on a 24-well plate at a density of 130,000 cells per well in 0.5 ml of culture medium. 250 ng of modified G-CSF mRNA (mRNA sequence shown in SEQ ID NO: 1; polyA tail of approximately 160 nucleotides not shown in sequence; 5’cap, Cap 1) fully modified with 5-methylcytosine and pseudouridine (Genl) or fully modified with S-methylcytosine and N]- methylpseudouridine (Gen2) was transfected using Lipofectamine 2000 (Invitrogen, cat# 11668- 019), following manufacturer’s protocol. Control s of Lipofectamine 2000 0) and unmodified G-CSF mRNA were also ected. The modified mRNA or control samples were transfected daily for 4 days. The viability of the cells after transfection was evaluated 6 hours and 24 hours after the first transfection (T1, 6 hours or T1, 24 hours), and 24 hours after the second (T2, 24 hours) and fourth transfection (T4, 24 .
To determine cell Viability, the culture medium was completely removed and the cells were washed once with 600ul of sterile PBS without Ca2+/Mg21L (Gibco/Life Technologies, Manassas, VA) in order to off loosely attached cells. PBS was removed and discarded. The cleaned fibroblasts in each well were treated with 220ul of a diluted CELL TITER GLO® (Promega, catalog #G7570) stock solution (the CELL TITER GLO® stock solution was further diluted 1:1 with an equal amount of sterile PBS). A sterile pipet tip was used to scratch the cells off the plate and accelerate the lysis process.
For two time intervals, T1, 24 hours and T2, 24 hours, an ative protocol was applied. Cells were washed with PBS, as described above, and subsequently trypsinized with Trypsin/EDTA solution (Gibco/Life Technologies, Manassas, VA). Cells were detached and collected in 500ul of medium containing n tor. Cells were harvested by firgation at 1200 rcf for 5 minutes. The cell pellet was resuspended in 500ul PBS. This cell suspension was kept on ice, and 100ul of this was combined with 100ul of undiluted Cell Titer Glo solution.
All of the CELL TITER GLO® lysates were then incubated at room temperature for 20 minutes. 20 ul of the lysates were transferred to a white opaque polystyrene 96-well plate ng, as, VA) and combined with 100 111 diluted CELL TITER GLO® solution. The plate reader used was from BioTek y H1 (BioTek, Winooski, VT) and the absolute values were normalized to signal of the untreated BJ Fibroblasts to 100% cell vitality. The percent viability for the B] asts are shown in Table 33.
Importantly, all of these experiments are conducted in the absence of any interferon or other cytokine inhibitors and thus represent an accurate measure of the cytotoxicity of the different mRNA.
These results demonstrate that repeated transfection of B] asts with fied mRNA results in loss of cell Viability that is nt as early as 24 hrs after the first transfection (Tl , 24 hours) and ues to be nt and more pronounced at subsequent time points.
There is also a loss of viability with repeated transfection of Smethylcytidine and pseudouridine modified mRNA that is apparent 24 hours after the fourth daily transfection (T4, 24 hours). No loss of cell viability over the course of this experiment is seen using Smethylcytidine and Nl-methylpseudouridine modified mRNA. These s demonstrate that lcytidine and N1- methylpseudouridine containing mRNA have improved cell viability when analyzed under repeated transfection. The ability to repeatedly administer modified mRNA is important in most therapeutic applications, and as such the y to do so t cytotoxicity is also important. While not wishing to be bound by theory, it is believed that response genes following a single transfection may lead to a decrease in protein production, cytokjne induction, and eventually loss of cell viability.
These results are consistent with N1-methylpseudouridine—containing mRNA showing an improved profile in this respect relative to both unmodified mRNA and pseudouridine—modified mRNA.
Table 33. Percent Viability T1, 6 hours T1, 24 hours T2, 24 hours T4, 24 hours Gen 1 G—CSF 81 108 91 65 Gen 2 G-CSF 99 102 128 87 Unmodified G-CSF 101 72 74 42 LF2000 99 80 114 106 Untreated 100 100 100 100 Example 83. Innate Immune Response in BJ Fibroblasts Human primary foreskin fibroblasts (BJ fibroblasts) are obtained from American Type Culture Collection (ATCC) (catalog #CRL-2522) and grown in Eagle’s Minimum Essential Medium (ATCC, cat# 30-2003) supplemented with 10% fetal bovine serum at 37°C, under 5% C02.
BJ fibroblasts are seeded on a 24-well plate at a density of 130,000 cells per well in 0.5 ml of e medium. 250 ng ofmodified G—CSF mRNA (mRNA sequence shown in SEQ ID NO: 1; polyA tail of approximately 160 nucleotides not shown in sequence; S’cap, Capl) fully modified with 5- methylcytosine and pseudouridine (Genl) or fully modified with 5-methylcytosine and N1- methylpseudouridine (Gen2) is transfected using Lipofectamine 2000 (Invitrogen, cat# 11668-019), following manufacturer’s protocol. Control samples of ctamine 2000 and unmodified G-CSF mRNA (natural G-CSF) are also transfected. The cells are transfected for five consecutive days. The transfection complexes are removed four hours after each round of transfection.
The culture supernatant is assayed for secreted GCSF (R&D Systems, catalog #DCSSO), tumor necrosis factor-alpha (TNF-alpha) and interferon alpha (IFN-alpha) by ELISA every day after transfection following manufacturer’s ols. The cells are analyzed for viability using CELL TITER GLO® ga, catalog #G7570) 6 hrs and 18 hrs after the first round of transfection and every alternate day following that. At the same time from the harvested cells, total RNA is isolated and treated with DNASE® using the RNAEASY micro kit (catalog #74004) following the manufacturer’s protocol. 100 ng of total RNA is used for cDNA synthesis using the High ty cDNA Reverse Transcription kit (Applied Biosystems, cat #4368814) following the manufacturer’s protocol. The cDNA is then analyzed for the expression of innate immune response genes by quantitative real time PCR using Syerreen in a Biorad CFX 384 instrument following the cturer’s ol.
Example 84. In vitro Transcription with wild-type T7 polymerase Luciferase mRNA (mRNA sequence shown in SEQ ID NO: 3; polyA tail of approximately 160 nucleotides not shown in sequence; S’cap, Capl) and G—CSF mRNA (mRNA sequence shown in SEQ ID NO: 1; polyA tail of approximately 160 nucleotides not shown in sequence; S’cap, Capl) were fully modified with different chemistries and try combinations listed in Tables 34-37 using wild-type T7 polymerase as usly described. 2] The yield of the translation reactions was ined by spectrophometric measurement ) and the yield for Luciferase is shown in Table 34 and G—CSF is shown in Table 36.
The luciferase and G—CSF modified mRNA were also subjected to an enzymatic capping reaction and each modified mRNA g reaction was evaluated for yield by spectrophometic measurement (OD260) and correct size assessed using bioanalyzer. The yield from the capping reaction for rase is shown in Table 35 and G—CSF is shown in Table 37.
Table 34. In vitro transcription chemistry for Luciferase Chemical Modification Yield (mg) N6-methyladenosine 0‘99 -methylcytidine 1 .29 N4-acetylcytidine 1 .0 -formylcytidine 0.55 Pseudouridine 2 .0 N 1 1pseudouridine 1 ‘43 2-thiouridine 1 .56 -methoxyuridine 2.35 yluridine 1 .01 a-Thio-cytidine 0.83 -Br-uridine (5Bru) 1.96 (2 carbomethoxyvinyl) uridine 0.89 (3-1E yl Amino) uridine 2.01 N4-aoetylcytidine/pseudouridine 1 .34 N4-acetylcytidine/N 1 -methylpseudoun'dine 1 .26 -methylcytidine/5-methoxyuridine 1 .3 8 —methylcytidine/5-bromouridine 0. 12 —methylcytidine/5-methylun'dine 2.97 —methylcytidine/ half of the uridines are modified with 2-thiou1idine 1.59 —methylcytidine/2-thi0uridine 0.90 —methylcytidine/pseudoutidine 1.83 —methylcytidine/N 1 methyl pseudouridine 1.33 Table 35. Capping chemistry and yield for Luciferase modified mRNA Chemical Modification Yield (mg) —methylcytidine 1 .02 N4-acetylcytidine 0.93 -formylcytidine 0.55 Pseudouridine 2 .07 Nl-methylpseudoun'dine 1‘27 2-thiouridine 1 ‘44 -methoxyun'dine 2 -methyluridine 0‘8 a—Thio-cytidine 0‘74 -Br-uridine (5Bru) 1‘29 (2 ethoxyvinyl) e 0‘54 (3-1E propenyl Amino) un'dine 1‘39 N4-acetyleytidine/pseudouridine 0‘99 N4-acetyleytidine/N 1 -methy1pseudoun'dine 1 ‘08 yleytidine/5-methoxyuridine 1 1 13 -methyleytidine/5-methyluridine 1 ‘08 -methyleytidine/ half of the uridines are modified with 2-thiouridine 1‘2 -methyleytidine/2 -thiouridine 1‘27 -methylcytidine/pseudouridine 1 . 19 S-methylcytidine/N 1 methyl pseudouridine 1‘04 Table 36. In vitro transcription chemistry and yield for G—CSF modified mRNA Chemical Modification Yield (mg) N6-methy1adenosine 1. 57 -methy1cytidine 2.05 N4-acetylcytidine 3. 1 3 -formylcytidine 1 .4 1 Pseudouridine 4. 1 N l-methylpseudouridine 3 .24 2-thiouridine 3.46 -methoxyuridine 2.57 -methyluridine 4.27 4—thiouridine 1 .45 ridine 0.96 u—Thio-cytidine 2.29 uanosine 0.6 N—l—methyladenosine 0.63 S—Br-uridine (SBru) 1.08 (2 carbomethoxyvinyl) un'dine 1.8 (3-1E propenyl Amino) uridine 2.09 N4—acetylcytidine/pseudouridine 1 .72 tylcytidine/N 1 -methy1pseudouridine 1 .3 7 —methylcytidine/5-methoxyuridine 1 .85 —methylcytidine/5-methy1uridine 1 .56 —methylcytidine/ half of the uridines are modified with 2-thioun'dine 1.84 —methylcytidine/2-thiouridine 2.53 —methylcytidine/pseudouridine 0.63 N4-acetylcytidine/2-thioun'dine 1 .3 N4-acetylcytidine/5-bromouridine 1.37 -methylcytidine/N 1 methyl pseudouridine 1125 N4-acetylcytidine/pseudouridine 2124 Table 37. Capping chemistry and yield for G—CSF modified mRNA al Modification Yield (mg) N6-methyladenosine 1 104 -methylcytidine 1 10 8 N4-acetyleytidine 2173 yleytidine 0195 Pseudouridine 318 8 Nl-methylpseudouridine 215 8 2-thiouridine 215 7 -methoxyuridine 2105 -methyluridine 3156 4-thiouridine 019 1 2'-F—uridine 0. 54 a-Thio-cytidine 1 .79 2'-F-guanosine 0. 14 -Br-uridine (5Bru) 0.79 (2 carbomethoxyvinyl) uridine 1 .28 (3-113 propenyl Amino) uridine 1.78 N4-acetylcytidine/pseudouridine 0.29 N4-acetylcytidine/N 1 -methylpseudouridine 0.3 3 -methy1cytidine/S-methoxyuridine 0.9 1 -methylcytidine/5-methy1uridine 0.6 1 -methylcytidine/ half of the uridines are modified with 2-thiouridine 1.24 -methylcytidine/pseudouridine 1 .08 N4-acetylcytidine/2-thiouridine 1 .3 4 N4-aoetylcytidine/5-bromouridine 1 .22 -methylcytidine/N1 methyl pseudoun'dine 1.56 Example 85. In vitro Transcription with mutant T7 polymerase Luciferase mRNA (mRNA sequence shown in SEQ ID NO: 3; polyA tail of approximately 160 tides not shown in sequence; 5’cap, Capl) and G—CSF mRNA (mRNA ce shown in SEQ ID NO: 1; polyA tail of approximately 160 nucleotides not shown in sequence; S’cap, Capl) were fully d with different chemistries and chemistry combinations listed in Tables 38-41 using a mutant T7 polymerase cribe® T7 Transcription kit (Cat. No.
DS010925) (Epicentre®, Madison, WI).
The yield of the translation reactions was determined by spectrophometric measurement (OD260) and the yield for Luciferase is shown in Table 38 and G—CSF is shown in Table 40.
The luciferase and G—CSF modified mRNA were also subjected to an enzymatic capping reaction and each modified mRNA capping reaction was evaluated for yield by ophometic measurement (OD260) and correct size ed using bioanalyzer. The yield from the capping reaction for luciferase is shown in Table 39 and G—CSF is shown in Table 41.
Table 38. In vitro transcription chemistry and yield for Luciferase modified mRNA Chemical Modification Yield (ug) 2’Fluorocytosine 71.4 rouridine 57.5 -methy1cytosine/pseudouridine, test A 26.4 -methy1cytosine/Nl -methy1pseudouridine, test A 73 .3 tylcytidine/2-fluorouridine 202 .2 -methylcytidine/2 -fluorouridine 13 1 .9 ocytosine/pseudouridine 1 1913 2-fluorocytosine/N l -methy1pseudouridine 107 ‘0 2-fluorocytosine/2-thiouridine 34i7 2-fluorocytosine/5-bromouridine 8 1 .0 2-fluorocytosine/Z-fluorouridine 80.4 2-fluoroguanine/5-methylcytosine 61.2 2-fluoroguanine/5-methylcytosine/pseudouridine 65.0 2-fluoroguanine/5-methylcytidine/N 1 -methy1pseudouridine 41 .2 2-fluoroguanine/pseudouridine 79.1 2-fluoroguanine/N1-methy1pseudouridine 74.6 -methylcytidine/pseudouridine, test B 91.8 -methylcytidine/N1-methy1pseudouridine, test B 72.4 2’fluoroadenosine 190.98 Table 39. Capping chemistry and yield for Luciferase modified mRNA al Modification Yield (ug) 2’Fluorocytosine 19.2 2’Fluorouu'dine 16.7 —methylcytosine/pseudouridine, test A 7.0 —methylcytosine/N 1 -methylpseudoun'dine, test A 2 1 .5 N1—acetylcytidine/2-flu0rouridine 47.5 —methylcytidine/2-flu0routidine 53 .2 2—fluorocytosine/pseudoutidine 5 8 .4 2—fluorocytosine/N 1-methylpseudoun'dine 26.2 ocytosine/2-thiouridine 12 .9 2—fluor0cytosine/5-bromouridine 26.5 2—fluorocytosine/2-fluorouridine 3 5 .7 2—fluor0guanine/5-methylcytosine 24.7 2—fluoroguanine/5-methylcytosine/pseudoun'dine 3 2 .3 2—fluoroguanine/5-methylcytidine/N 1 -methylpseudouridine 3 1 .3 2—fluoroguanine/pseudouridine 20.9 2-fluoroguanine/N1-methylpseudouridine 29.8 -methylcytidine/pseudouridine, test B 5 8 .2 -methylcytidine/N 1 lpseudouridine, test B 44.4 Table 40. In vitro transcription chemistry and yield for G—CSF modified mRNA Chemical Modification Yield (ug) 2’Fluorocytosine 56.5 2’Fluorouridine 79‘4 -methylcytosine/pseudouridine, test A 2 1 ‘2 -methylcytosine/N 1 -methy1pseudouridine, test A 77. 1 N1-acetylcytidine/2-fluorouridine 168.6 ylcytidine/2 -fluorouridine 134.7 2-fluorocytosine/pseudouridine 97. 8 2-fluorocytosine/N l -methy1pseudouridine 103. 1 2-fluorocytosine/2-thiouridine 5 8. 8 2-fluorocytosine/5-bromouridine 8 8. 8 2-fluorocytosine/2-fluorouridine 93 .9 2-fluoroguanine/5-methylcytosine 97.3 2-fluoroguanine/5-methylcytosine/pseudouridine 96.0 2-fluoroguanine/5-methylcytidine/N 1 1pseudouridine 82 .0 2-fluoroguanine/pseudouridine 68.0 2-fluoroguanine/N 1 lpseudouridine 59 .3 -methy1cytidine/pseudouridine, test B 58,7 -methylcytidine/N1-methy1pseudouridine, test B 78.0 Table 41. Capping chemistry and yield for G-CSF modified mRNA Chemical Modification Yield (ug) Z’Fluorocytosine 16.9 roun'dine 17.0 -methylcytosine/pseudouridine, test A 10.6 -methylcytosine/N1 -methylpseudouridine, test A 22.7 N1 -acetylcytidine/Z-flu0rouridine 19.9 —methylcytidine/2-flu0rouridine 2 1 .3 2—fluorocytosine/pseudouridine 65 .2 2—fluorocytosine/N 1 -methylpseudouridine 5 8.9 2—fluorocytosine/2-thiouridine 41 .2 2—fluorocytosine/5-bromouridine 3 5.8 2—fluorocytosine/2-fluorouridine 3 6.7 2—fluoroguanine/5-methylcytosine 3 6.6 2—fluoroguanine/5-methylcytosine/pseudouridine 3 7.3 2—fluoroguanine/5-methylcytidine/N 1 -methy1pseudouridine 3 0.7 2—fluoroguanine/pseudouridine 29.0 2—fluor0guanine/N1-methylpseudouridine 22.7 -methylcytidine/pseudouridine, test B 60.4 -methylcytidine/N 1 -methylpseudouridine , test B 3 3 .0 Example 86. 2’O-methyl and 2’Fluoro compounds 7] rase mRNA (mRNA sequence shown in SEQ ID NO: 3; polyA tail of approximately 160 tides not shown in sequence; 5’cap, Capl) were ed as fully modified ns with the chemistries in Table 42 and transcribed using mutant T7 polymerase (Durascribe® T7 Transcription kit (Cat. No. DSOIO925) (Epicentre®, Madison, WI). 2’ fluorocontaining mRNA were made using Durascribe T7, however, 2’Omethyl-contajning mRNA could not be transcribed using Durascribe T7.
Incorporation of 2’Omethyl modified mRNA might possibly be accomplished using other mutant T7 polymerases (Nat Biotechnol. (2004) 22:1155-1160; Nucleic Acids Res. (2002) 302el38).
Alternatively, 2’OMe modifications could be introduced post-transcriptionally using enzymatic means.
Introduction of ations on the 2’ group of the sugar has many potential advantages. 2’OMe substitutions, like 2’ fluoro substitutions are known to protect against nucleases and also have been shown to abolish innate immune recognition when incorporated into other c acids such as siRNA and anti-sense (incorporated in its ty, Crooke, ed. nse Drug Technology, 2““1 edition; Boca Raton: CRC press).
The 2’Fluoro-modified mRNA were then transfected into HeLa cells to assess protein production in a cell context and the same mRNA were also assessed in a cell-free rabbit reticulocyte system. A l of unmodified luciferase (natural luciferase) was used for both transcription experiments, a control of untreated and mock ected (Lipofeetamine 2000 alone) were also analyzed for the HeLa transfection and a control of no RNA was analyzed for the rabbit reticulysates.
For the HeLa transfection experiments, the day before transfection, 20,000 HeLa cells (ATCC no. CCL—2; Manassas, VA) were harvested by treatment with n-EDTA solution (LifeTechnologies, Grand Island, NY) and seeded in a total volume of 100ul EMEM medium (supplemented with 10%FCS and 1x Glutamax) per well in a 96-well cell culture plate (Corning, Manassas, VA). The cells were grown at 370G in 5% C02 atmosphere ght. Next day, 83 ng of the 2’fluoro-containing luciferase modified RNA (mRNA sequence shown in SEQ ID NO: 3; polyA tail of approximately 160 nucleotides not shown in sequence; 5’cap, Capl) with the chemical modification described in Table 42, were diluted in 10ul final volume of OPTI-MEM (LifeTechnologies, Grand Island, NY). Lipofectamine 2000 (LifeTechnologies, Grand Island, NY) was used as transfection reagent and 0.2 111 were d in 10 111 final volume of OPTI—MEM. After minutes of incubation at room temperature, both solutions were combined and ted an additional 15 minute at room temperature. Then the 20ul combined solution was added to the 100ul cell culture medium containing the HeLa cells and incubated at room ature. After 18 to 22 hours of incubation cells expressing luciferase were lysed with 100 111 of Passive Lysis Buffer (Promega, Madison, WI) according to manufacturer instructions. Aliquots of the lysates were erred to white opaque polystyrene 96-well plates ng, Manassas, VA) and combined with 100 ul complete luciferase assay on (Promega, Madison, WI). The lysate volumes were adjusted or diluted until no more than 2 mio relative light units (RLU) per well were detected for the strongest signal producing samples and the RLUs for each chemistry tested are shown in Table 42.
The plate reader was a BioTek Synergy H1 (BioTek, Winooski, VT).The background signal of the plates without reagent was about 200 relative light units per well.
For the rabbit reticulocyte lysate assay, 2’-fluoro-containing luciferase mRNA were diluted in sterile nuclease-free water to a final amount of 250 ng in 10 111 and added to 40 ul of y prepared Rabbit Reticulocyte Lysate and the in Vitro translation reaction was done in a standard 1.5 mL polypropylene reaction tube (Thermo Fisher Scientific, Waltham, MA) at 30°C in a dry heating block. The translation assay was done with the Rabbit Reticulocyte Lysate (nuclease- treated) kit (Promega, Madison, WI) according to the manufacturer’s ctions. The reaction buffer was supplemented with a one-to-one blend of provided amino acid stock solutions devoid of either Leucine or Methionine resulting in a reaction mix containing ent amounts of both amino acids to allow ive in Vitro translation. After 60 minutes of incubation, the reaction was stopped by placing the reaction tubes on ice. 3] Aliquots of the in Vitro translation reaction containing luciferase modified RNA were transferred to white opaque polystyrene 96-well plates (Corning, Manassas, VA) and combined with lOOul complete luciferase assay solution (Promega, Madison, WI). The volumes of the in Vitro translation reactions were adjusted or diluted until no more than 2 mio relative light units (RLUs) per well were detected for the strongest signal producing samples and the RLUs for each chemistry tested are shown in Table 43. The plate reader was a BioTek Synergy H1 (BioTek, Winooski, VT).
The ound signal of the plates without reagent was about 160 ve light units per well.
As can be seen in Table 42 and 43, le 2’Fluoro-containing compounds are active in Vitro and produce luciferase protein.
Table 42. HeLa Cells Chemical ation Concentration Volume (ul) Yield (ug) RLU (ug/ml) 2’Fluoroadenosine 381.96 500 190.98 388.5 2’Fluorocytosine 654.56 500 327.28 2420 2’Fluoroguanine 541,795 500 270.90 1 1,7055 2’Flurorouridine 944.005 500 472.00 67675 Natural luciferase N/A N/A N/A 133 ,8535 Mock N/A N/A N/A 340 Untreated N/A N/A N/A 23 8 Table 43. Rabbit Reticulysates al Modification RLU 2 ’Fluoroadenosine 162 2 ’Fluorocytosine 208 2 ’Fluoroguanine 371,509 2 ’Flurorouridine 258 l luciferase 2,159,968 No RNA 156 Example 87. Luciferase in HeLa Cells using a combination of modifications To evaluate using of 2’fluoro-modified mRNA in combination with other modification a series of mRNA were transcribed using either wild-type T7 polymerase (non-fluoro-containing compounds) or using mutant T7 polymerases o-containing compounds) as described in Example 86. All d mRNA were tested by in vitro transfection in HeLa cells.
The day before transfection, 20,000 HeLa cells (ATCC no. CCL—2; Manassas, VA) were harvested by treatment with Trypsin-EDTA solution (LifeTechnologies, Grand Island, NY) and seeded in a total volume of 100ul EMEM medium (supplemented with 10%FCS and 1x Glutamax) per well in a l cell culture plate ng, Manassas, VA). The cells were grown at 370G in % C02 atmosphere overnight. Next day, 83 ng of rase modified RNA (mRNA sequence shown in SEQ ID NO: 3; polyA tail of approximately 160 nucleotides not shown in sequence; 5’cap, Capl) with the chemical modification described in Table 44, were diluted in 10ul final volume of OPTI—MEM (LifeTechnologies, Grand Island, NY). Lipofectamine 2000 echnologies, Grand Island, NY) was used as transfection reagent and 0.2 111 were diluted in 10 111 final volume of OPTI— IVIEM. Afier 5 minutes of incubation at room temperature, both solutions were combined and incubated an additional 15 minute at room temperature. Then the 20ul combined solution was added to the 100ul cell culture medium containing the HeLa cells and incubated at room temperature.
After 18 to 22 hours of incubation cells expressing luciferase were lysed with 100 u] of Passive Lysis Buffer (Promega, Madison, WI) ing to manufacturer instructions. Aliquots of the lysates were transferred to white opaque polystyrene 96-well plates ng, Manassas, VA) and combined with 100 111 complete luciferase assay solution (Promega, Madison, WI). The lysate s were adjusted or diluted until no more than 2 mio relative light units (RLU) per well were detected for the strongest signal producing samples and the RLUs for each chemistry tested are shown in Table 44. The plate reader was a BioTek Synergy H1 k, Winooski, e background signal of the plates t reagent was about 200 relative light units per well.
As evidenced in Table 44, most combinations of modifications resulted in mRNA which produced functional luciferase protein, including all the non-flouro containing compounds and many of the combinations containing 2’fluro modifications.
Table 44. Luciferase Chemical Modification RLU N4-acetylcytidine/pseudouridine 1 13,796 tylcytidine/N 1 -methy1pseudouridine 3 16,326 -methylcytidine/5-methoxyuridine 24,948 -methylcytidine/5-methyluridine 43 ,675 -methylcytidine/half of the uridines modified with 50% 2-thiouridine 41,601 -methylcytidine/2-thiouridine 1 102 -methylcytidine/pseudouridine 5 1 ,03 5 -methylcytidine/N 1 methyl pseudouridine 152,151 tyloytidine/Z'Fluoroun'dine triphosphate 288 -methylcytidine/2'Fluorouridine tn'phosphate 269 2’Fluorocytosine triphosphate /pseudouridine 260 2'Fluorocytosine triphosphate /N1-methylpseudouridine 412 2'Fluorocytosine triphosphate/Z-thiouridine 427 2'Fluorocytosine triphosphate/S-bromouridine 253 2'Fluorocytosine triphosphate /2'Fluorouridine triphosphate 184 2'Fluoroguanine triphosphate/S-methylcytidine 3 21 2'Fluoroguanine triphosphate/S-methylcytidine/Pseudouridine 207 2'Fluoroguanine hylcytidine/N l methylpsuedouridine 235 2'Fluoroguanine/pseudouridine 2 l 8 2'Fluoroguanine/N 1 -methylpsuedouridine 247 —methylcytidine/pseudouridine, test A 13 ,833 —methylcytidine/N—methylpseudouridine, test A 598 2'Fluorocytosine triphosphate 201 2'F luorouridine triphosphate 305 —methyleytidine/pseudouridine, test B 1 15,401 -methylcytidine/N—methylpseudouridine, test B Z l ,034 Natural lueiferase 30,80] Untreated 344 Mock 262 Example 88. G-CSF In Vitro ription To assess the activity of all our different chemical modifications in the context of a second open reading frame, we replicated experiments usly conducted using luciferase mRNA, with human G—CSF mRNA. G—CSF mRNA (mRNA sequence shown in SEQ ID NO: 1; polyA tail of approximately 160 nucleotides not shown in sequence; 5’cap, Capl) were fully modified with the chemistries in Tables 45 and 46 using wild-type T7 polymerase (for all non- fluoro-containing compounds) or mutant T7 polymerase (for all fluoro-contajning compounds). The mutant T7 rase was obtained commercially (Durascribe® T7 Transcription kit (Cat. No.
DS010925) (Epicentre®, n, WI).
The modified RNA in Tables 45 and 46 were transfected in vitro in HeLa cells or added to rabbit reticulysates (250ng of modified mRNA) as ted. A control of untreated, mock ected (transfection reagent alone), G—CSF fully modified with S-methylcytosine and N1- methylpseudouridine or luciferase control (mRNA sequence shown in SEQ ID NO: 3; polyA tail of approximately 160 nucleotides not shown in sequence; 5’cap, Capl) fully modified with 5- methylcytosine and Nl-methylpseudouridine were also analyzed. The expression F protein was determined by ELISA and the values are shown in Tables 45 and 46. In Table 45, “NT” means not tested.
As shown in Table 45, many, but not all, chemical modifications resulted in human G- CSF protein production. These results from cell-based and cell-free translation systems correlate very nicely with the same modifications generally g or not working in both systems. One notable exception is 5-formylcytidine modified G—CSF mRNA which worked in the cell-free translation system, but not in the HeLa cell-based transfection system. A similar difference between the two assays was also seen with 5-formylcytidine modified rase mRNA.
As trated in Table 46, many, but not all, G—CSF mRNA modified chemistries (when used in combination) demonstrated in viva activity. In addition the presence ofN1- methylpseudouridine in the modified mRNA (with N4-acetylcytidine or 5 methylcytidine) demonstrated higher expression than when the same combinations where tested using with pseudouiidine. Taken together, these data demonstrate that N1 -methylpseudouridine ning G- CSF mRNA results in improved protein sion in vitro.
Table 45. G—CSF Expression Chemical Modification G—CSF protein G—CSF protein (pg/ml) (pg/ml) HeLa cells Rabbit reticulysates cells Pseudouridine 1,150,909 147,875 -methyluridine 347,045 147,250 2-thiouridine 3 18 ,3 75 Nl-methylpseudouridine NT 230,000 4-thiouridine 107,273 52,375 -methoxyuridine 1,715 ,909 0 -methylcytosine/pseudouridine, Test A 609,545 1 19,750 -methylcytosine/N 1 -methylpseudouridine Test A , 3 18 1 10,500 2'-Fluoro-guanosine 1 1,818 0 2'-Fluoro-uridine 60,455 0 -methylcytosine/pseudouridine, Test B 358 ,182 57,875 -methylcytosine/N1-methy1pseudouridine Test B , 1,568,636 76,750 o-uridine 186,591 72,000 -(2carbomethoxyvinyl) uridine 1,3 64 0 -[3(1-E-propenylamino) uridine 27,955 32,625 a-thio-cytidine 120,455 42,625 -methylcytosine/pseudouridine, Test C 882,500 49,250 N 1 -methyl-adenosine 4,773 0 N6-methyl-adenosine 1,591 0 -methy1-cytidine 646,591 79,375 N4-acetylcytidine 39,545 8 ,000 -formyl-cytidine 0 24,000 -methylcytosine/pseudouridine, Test D 87,045 47,750 -methyloytosine/N 1 lpseudouridine Test D , 1,168,864 97,125 Mock 909 682 Untreated 0 0 -methyloytosine/N 1 -methylpseudouridine Control 1 ,106,591 NT Luciferase l NT 0 Table 46. Combination Chemistries in HeLa cells Chemical Modification G—CSF n (pg/ml) HeLa cells N4-acetylcytidine/pseudouridine 537,273 N4-acetylcytidine/N l -methylpseudoun'dine l ,091 ,8 l 8 —methylcytidine/5-methoxyuridine 5 l 6, l 3 6 -methylcytidine/5-bromouridine 48,864 -methylcytidine/5-methylun'dine 0 -methylcytidine/2-thioun'dine 3 3 ,409 N4-acetylcytidine/5-bromouridine 2 l l ,591 N4-acetylcytidine/2-thiouridine 46,136 -methylcytosine/pseudouridine 3 01 ,3 64 -methylcytosine/N l -methylpseudoun'dine 1 ,017,727 N4-acetylcytidine/2'Fluorouridine tn'phosphate 62,273 -methylcytidine/2'Fluorouridine triphosphate 49,3 18 2'Fluorocytosine tn'phosphate/pseudouridine 7,955 2'Fluorocytosine triphosphate/N l -methylpseudoun'dine l ,3 64 2'Fluorocytosine triphosphate/2-thiouridine 0 rocytosine triphosphate/5-bromoun'dine l ,8 l 8 rocytosine triphosphate/2'Fluorouridine triphosphate 909 2'Fluoroguanine triphosphate/5-methylcytidine 0 2'Fluoroguanine triphosphate/5-methylcytidine/pseudouridine 0 2'Fluoroguanine triphosphate /5-methylcytidine/N l l ,8 l 8 methylpseudouridine 2'Fluoroguanine triphosphate/pseudouridine 1 ,13 6 2'Fluoroguanine triphosphate/2 ’Fluorocytosine 0 sphate/N 1 -methylpseudouridine -methyloytidine/pseudouridine 617,727 yloytidine/N 1 -methylpseudouridine 747,045 -methyloytidine/pseudouridine 475,455 -methylcytidine/N 1 -methylpseudouridine 689,091 -methylcytosine/N1-methylpseudouridine, Control 1 848,409 -methylcytosine/N l -methylpseudouridine, Control 2 58 1 ,8 1 8 Mock 682 Untreated 0 Luciferase Z‘Fluorocytosine triphosphate 0 Luciferase Z'Fluorouridine triphosphate 0 Example 89. Screening of Chemistries The tables listed in below (Tables 47-49) ize much of the in vitro and in vitro screening data with the different compounds ted in the previous es. A good correlation exists between cell-based and cell-free translation assays. The same chemistry substitutions generally show good concordance whether tested in the context of luciferase or G—CSF mRNA.
Lastly, Nl—methylpseudouridine containing mRNA show a very high level of protein sion with little to no detectable cytokine stimulation in vitro and in vivo, and is superior to mRNA containing pseudouridine both in vitro and in viva.
Luciferase mRNA (mRNA sequence shown in SEQ ID NO: 3; polyA tail of approximately 160 nucleotides not shown in sequence; 5’cap, Capl) and G—CSF mRNA (mRNA sequence shown in SEQ ID NO: 1; polyA tail of approximately 160 tides not shown in sequence; 5’cap, Capl) were modified with lly and non-naturally occurring chemistries bed in Tables 47 and 48 or combination tries described in Table 48 and tested using methods described .
In Tables 47 and 48, “*” refers to in vitro transcription reaction using a mutant T7 polymerase (Durascribe® T7 Transcription kit (Cat. No. DS010925) (Epicentre®, Madison, WI); “**” refers to the second result in vitro transcription reaction using a mutant T7 polymerase (Durascribe® T7 Transcription kit (Cat. No. DS010925) (Epicentre®, Madison, WI); “***” refers to production seen in cell free translations (rabbit reticulocyte lysates); the protein production of HeLa is judged by “+,” “**/-“ and “-“; when referring to G—CSF PBMC “+H+” means greater than 6,000 pg/ml G-CSF, «H,» means greater than 3,000 pg/ml G-CSF, “++” means greater than 1,500 pg/ml G-CSF, “+” means greater than 300 pg/ml G-CSF, “+/-“ means 150-300 pg/ml G-CSF and the background was about 110 pg/ml; when referring to cytokine PBMC “++4-|-” means greater than 1,000 pg/ml interferon-alpha (IFN—alpha), “+++” means greater than 600 pg/ml IFN-alpha, “++” means greater than 300 pg/ml pha, “+” means greater than 100 pg/ml IFN-alpha, - means less than 100 pg/ml and the background was about 70 pg/ml; and “NT” means not tested. In Table -3l3- 48, the protein production was evaluated using a mutant T7 polymerase (Durascribe® T? Transcription kit (Cat. No. DSOlO925) (Epicentre®, Madison, WI).
Table 47. Naturally Occurring Common Name IVT IVT n Protein Protein Cytokines In Vivo In Vivo (symbol) (Luc) (G— (Luc; (G— (G-CSF; ; Protein Protein CSF) HeLa) CSF; PBMC) PBMC) (Luc) (G HeLa) CSF) l -methyladenosine Fail Pass NT - /- NT NT (m'A) Ns-methyladenosine Pass Pass - - W/- l : NT NT (mGA) 2'-O- Fail* Not NT NT NT NT NT NT methyladenosine Done (Am) -methylcytidine Pass Pass + + + H + NT (mSC) 2'-O—methylcytidine Fail* Not NT NT NT NT NT NT (Cm) Done 2-thiocytidine (52C) Fail Fail NT NT NT NT NT NT N4-acetylcytidine Pass Pass + + +/- W+F + NT (304C) -fonnylcytidine Pass Pass -*** -*** W - NT NT (15C) 2'-O- Fail* Not NT NT NT NT NT NT methylguanosine Done (Gm) inosine (I) Fail Fail NT NT NT NT NT NT pseudouridine (Y) Pass Pass W W W+ W + NT -methyluridine Pass Pass W W W/- W NT NT (msU) 2'-O-methyluridine Fai1* Not NT NT NT NT NT NT (Um) Done 1- Pass Pass W Not W+F+ - + NT methylpseudouridine Done (le) 2-thiouridine (szU) Pass Pass W W W - NT NT 4-thiouridine (s4U) Fail Pass ,, ”/— 7+ NT NT -methoxyuridine Pass Pass + W 7+ - + NT (mosU) 3-methyluridine Fail Fail NT NT NT NT NT NT (msU) Table 48. turally Occurring Common Name IVT IVT Protein Protein Protein ne In Vivo In Vivo (Luc) (G- (Luc; (G- (G—CSF; s(G— Protein Protein CSF) HeLa) CSF; PBMC) CSF; (Luc) (G- HeLa) PBMC) CSF) 2'-F-ara-guanosine Fail Fail NT NT NT NT NT NT 2'-F—ara-adenosine Fail Fail NT NT NT NT NT NT 2'-F-ara-cytidine Fail Fail NT NT NT NT NT NT 2'-F-ara-uridine Fail Fail NT NT NT NT NT NT Fail/ Pass/F 2'-F-guanosine Pass* +** +/- - + + NT a11**.
Fail/ Sill/Fa. 2‘-F-adenosine Pass* —** NT NT NT NT NT Fail/ . 2'-F-cytidine Pass* iii/Pa ~** NT NT NT + NT Fail/ 2'-F—uridine Pass* Pas:1P 77* * + +/- + - NT ara—guanosine Fail Fail NT NT NT NT NT NT N“ N“ 2'-OH-ara—adenosine NT NT NT NT NT NT Done Done ara—cytidine Fail Fail NT NT NT NT NT NT 2'-OH-ara—utidine Fail Fail NT NT NT NT NT NT -Br—U1idine Pass Pass + + W + + -(2- carbomethoxyvinyl) Pass Pass - - "/- - Uridine -[3-(1-E— Pro en [aminoUrif‘l’iney Pass Pass W _ + _ (aka Cliem N6—(19—Amino- pentaoxanonadecyl) Fail Fail NT NT NT NT NT NT Z'D‘méthylamm" Fail Fail NT NT NT NT NT NT guanosme 6-Aza-cytidine Fail Fail NT NT NT NT NT NT a—Thio-cytidine Pass Pass + + +/- 4—H NT NT Pseudo-isocytidine NT NT NT NT NT NT NT NT -Iodo-uridine NT NT NT NT NT NT NT NT a—Thio-uridine NT NT NT NT NT NT NT NT 6-Au—uridine NT NT NT NT NT NT NT NT Deoxy-thymidine NT NT NT NT NT NT NT NT a—Thio guanosine NT NT NT NT NT NT NT NT 8-Oxo-guanosine NT NT NT NT NT NT NT NT O6-Methyl- NT NT NT NT NT NT NT NT guanosine 7-Deaza-guanosine NT NT NT NT NT NT NT NT ro-purine NT NT NT NT NT NT NT NT a-Thio-adenosine NT NT NT NT NT NT NT NT 7-Deaza-adenosine NT NT NT NT NT NT NT NT -iodo-cytidine NT NT NT NT NT NT NT NT In Table 49, the protein production of HeLa is judged by “+,” “+/-“ and “-“; when referring to G-CSF PBMC “--I-++” means r than 6,000 pg/ml G-CSF, “+++” means greater than 3,000 pgfml G—CSF, “+-»” means greater than 1,500 pg/ml G-CSF, “+” means greater than 300 pg/ml G-CSF, “+/-“ means 150-300 pg/ml G-CSF and the background was about 110 pg/ml; when referring to cytokine PBMC “+4-I-+” means greater than 1,000 pg/ml interferon-alpha (IFN-alpha), “HJr” means greater than 600 pg/ml IFN—alpha, “++” means greater than 300 pg/ml TFN-alpha, “+” cc cc means greater than 100 pg/ml IFN—alpha, - means less than 100 pg/ml and the background was about 70 pg/ml; “WT” refers to the wild type T7 polymerase, “MT” refers to mutant T7 rase (Durascribe® T7 Transcription kit (Cat. No. DSOlO925) (Epicentre®, Madison, WI) and “NT” means not .
Table 49. Combination Chemistry Cytidine Uridine Purin IVT IVT Protein Protein Protein Cytokines In analog analog e Luc (G— (Luc; (G— (G— (G—CSF; Vivo CSF) HeLa) CSF; CSF; PBMC) Protein HeLa) PBMC (Luc) N4- pseudouridin Pass Pass acetylcytidine ’ e WT WT + + NT NT + N1 — Pass Pass pseud A,G WT WT acetylcytidine ouridine + + NT NT + — Pass Pass cytidine nmfiethoxyurldi A,G WT WT + + NT NT + — Pass Pass . . bromouridin A,G WT WT Not methylcytidme e Done + NT NT — Pass Pass methylcytidine renethylurldm A,G WT WT + + NT NT + 50% 2— Pass Pass thiouridine; A,G WT WT methylcytidine 50% e + NT NT NT + — 100% 2— Pass Pass methylcytidine thiouridine ’ WT WT — + NT NT — pseudouridin Pass Pass methylcytidine ’ e WT WT + + ++ + + N1- Pass Pass . . ouridi/n:meth 1 send A,G WT WT methylcytldme + + ++++ _ + N4- Not Pass Not acetylcytidine 2-thiouridine ’ Done WT Done + NT NT NT - Pass bromouridin Not WT Not acety[0ytidine e A,G Done Done + NT NT NT N4- Fluorouriclin acetylcytidine e sphate A,G Pass Pass - + NT NT NT - Fluorouridin methylcytidine e triphosphate A,G Pass Pass — + NT NT NT Escudoundm. .
Fluorocytosine triphosphate A,G Pass Pass — + NT NT NT 2 N1— Fluorocytosine methylpseud sphate ouridine A,G Pass Pass — +/— NT NT NT Fluorocytosine triphosphate 2—thiouridine A,G Pass Pass — — NT NT NT 2 5— Fluorocytosine bromouridin triphosphate e A,G Pass Pass — +/— NT NT NT Fluorouridin Fluorocytosine trlphosphate triphosphate A,G Pass Pass — +/— NT NT NT uridine Fluoro methycytl me1 'd' GTP Pass Pass — — NT NT NT — pseudouridin 11:1):0m methylcytldme e GTP Pass Pass — — NT NT NT N1— A,2 . . methylpseud Fluoro cytldme ouridine GTP Pass Pass — +/— NT NT NT 2 A’2 1;seudouxidin Fluorocytosine Fluoro lIiphosphate GTP Pass Pass — +/— NT NT NT 2 N1— A,2 Fluorocytosine methylpseud Fluoro triphosphate ouridine GTP Pass Pass — — NT NT NT Example 90. 2’Fluoro Chemistries in PBMC The ability of G—CSF modified mRNA (mRNA sequence shown in SEQ ID NO: 1; polyA tail of approximately 160 tides not shown in sequence; S’cap, Capl) to trigger innate an immune response was determined by measuring interferon-alpha (IFN-alpha) and tumor necrosis -alpha (TNT-alpha) production. Use of in vitro PBMC cultures is an accepted way to measure the stimulatory potential of oligonucleotides (Robbins et al., Oligonucleotides 2009 19:89- 102) and transfection methods are described herein. Shown in Table 50 are the average from 2 or 3 separate PBMC donors of the interferon-alpha (IFN—alpha) and tumor necrosis factor alpha (TNF- alpha) production over time as ed by specific ELISA. Controls of R848, P(I)P(C), LPS and Lipofectamine 2000 (L2000) were also analyzed.
With regards to innate immune recognition, while both modified mRNA chemistries largely prevented IFN—alpha and TNF-alpha production ve to positive controls (R848, P(I)P(C)), 2’fluoro compounds reduce IFN—alpha and TNF-alpha production even lower than other ations and N4-acetylcytidine combinations raised the cytokine profile.
Table 50. IFN-alpha and TNF-alpha pha: TNF—alpha: 3 Donor Average 2 Donor (pg/ml) Average (pglml) L2000 1 361 P(I)P(C) 482 544 R848 45 8 ,235 LPS 0 6,889 N4—acetylcytidine/pseudouridine 694 528 N4—acetylcytidine/N l -methylpseudouridine 3 07 2 83 —methylcytidine/5-methoxyuridine 0 4 1 1 —methylcytidine/5-bromouridine 0 270 -methylcytidine/5-methyluridine 456 428 -methylcytidine/2-thiouridine 274 277 N4-acetylcytidine/2-thiouridine 0 2 85 N4-acetylcytidine/5-bromouridine 44 403 ylcytidine/pseudouridine 73 3 3 2 -methylcytidine/N l -methylpseudouridine 3 l 280 tylcytidine/2 ’fluorouridine triphosphate 3 5 3 2 -methylcytodine /2’fluorouridine sphate 24 0 2’fluorocytidine triphosphate/N l— 0 l l methylpseudouridine 2’fluorocytidine triphosphate/Z -thiouridine 0 0 2’fluorocytidine/ triphosphateS-bromouridine l 2 2 2’fluorocytidine triphosphate/2 ’fluorouridine l l 0 lriphosphate 2’fluorocytidine triphosphate/5-methylcytidine 14 23 2’fluorocytidine sphate/S - 6 21 methylcytidine/pseudouridine ocytidine triphosphate/S - 3 15 methylcytidine/N l -methylpseudouridine 2’fluorocytidine triphosphate/pseudouridine 0 4 2’fluorocytidine triphosphate/N l- 6 20 methylpseudouridine -methylcytidine/pseudouridine 8 2 1 8 -methylcytidien/N1-methylpseudouridine 3 5 3 OTHER MENTS It is to be understood that while the present disclosure has been described in conjunction with the detailed description thereof, the foregoing ption is intended to illustrate and not limit the scope of the present disclosure, which is defined by the scope of the appended claims. Other aspects, advantages, and modifications are within the scope of the following claims.

Claims (74)

WHAT IS CLAIMED IS:
1. An isolated mRNA encoding a polypeptide of interest, said isolated mRNA comprising: (a) a sequence of n number of linked nucleosides, (b) a 5’ UTR, (c) a 3’ UTR, and (d) at least one 5′ cap structure, wherein said isolated mRNA is fully modified with 1-methylpseudouridine, wherein said isolated mRNA, when administered to peripheral blood mononuclear cells (PBMCs), provides Protein:Cytokine (P:C) ratios of greater than 100 for TNF alpha and greater than 100 for IFN- alpha after about eighteen or more hours, and wherein said P:C ratios are higher than those of a corresponding mRNA comprising uridine (ψ) in place of 1-methylpseudouridine.
2. The isolated mRNA of claim 1, further comprising a poly-A tail.
3. The isolated mRNA of claim 1 or 2 which is purified.
4. The isolated mRNA of any one of claims 1-3, wherein the at least one 5′ cap structure is selected from the group consisting of Cap0, Cap1, ARCA, inosine, N1-methyl-guanosine, ro-guanosine, 7- deaza-guanosine, 8-oxo-guanosine, o-guanosine, LNA-guanosine, and 2-azido-guanosine.
5. The isolated mRNA of any one of claims 1-4, wherein the ce of n number of linked nucleosides comprises at least one al modification of a nucleoside located in the side base and/or sugar portion of the nucleoside. [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb ionNone set by jessb ation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb
6. The isolated mRNA of claim 5, wherein the at least one chemical modification located in a nucleoside base and the nucleoside base has the formula: XII-a, XII-b, or XII-c: wherein: denotes a single or double bond; X is O or S; U and W are each independently C or N; V is O, S, C or N; wherein when V is C then R1 is H, C1-6 alkyl, C2-6 l, C2-6 alkynyl, halo, or –ORc, wherein C1-20 alkyl, C2-20 alkenyl, C2-20 alkynyl are each optionally substituted with –OH, , - SH, -C(O)Rc, -C(O)ORc, -NHC(O)Rc, or )ORc; and wherein when V is O, S, or N then R1 is absent; R2 is H, -ORc, -SRc, -NRaRb, or halo; or when V is C then R1 and R2 together with the carbon atoms to which they are attached can form a 5- or 6-membered ring optionally substituted with 1-4 substituents selected from halo, -OH, -SH, - NRaRb, C1-20 alkyl, C2-20 alkenyl, C2-20 alkynyl, C1-20 alkoxy, or C1-20 thioalkyl; R3 is H or C1-20 alkyl; R4 is H or C1-20 alkyl; wherein when denotes a double bond then R4 is absent, or NR4 , taken together, forms a positively charged N substituted with C1-20 alkyl; Ra and Rb are each independently H, C1-20 alkyl, C2-20 l, C2-20 alkynyl, or C6-20 aryl; Rc is H, C1-20 alkyl, C2-20 alkenyl, phenyl, benzyl, a polyethylene glycol group, or an amino-polyethylene glycol group. [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb ation] jessb MigrationNone set by jessb [Annotation] jessb ed set by jessb ation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb
7. The isolated mRNA of claim 6, wherein the nucleoside base has the formula: XII-b wherein: R3 is C1-20 alkyl.
8. The ed mRNA of claim 7, wherein R3 is C1-4 alkyl.
9. The isolated mRNA of claim 7, wherein R3 is CH3.
10. The isolated mRNA of any one of claims 1-6, n the sequence of n number of linked nucleosides does not include pseudouridine (ψ) or 5-methyl-cytidine (m5C).
11. A pharmaceutical composition comprising the isolated mRNA of any one of claims 1-10 and a pharmaceutically acceptable excipient.
12. The pharmaceutical composition of claim 11, wherein the excipient is selected from a solvent, s solvent, non-aqueous solvent, dispersion media, diluent, dispersion, suspension aid, surface active agent, isotonic agent, thickening or emulsifying agent, preservative, lipid, lipidoids me, lipid nanoparticle, core-shell nanoparticles, polymer, lipoplex peptide, protein, cell, hyaluronidase, and mixtures thereof.
13. Use of the isolate mRNA of any one of claims 1-10 in the preparation of medicament for expressing a polypeptide of interest in a mammalian subject .
14. The use of claim 13, wherein the mRNA is formulated.
15. The use of claim 13, wherein the medicament is designed to be administered at a total daily dose of between 1 ug and 150 ug.
16. The use of claim 15, n the medicament is designed to be administered by injection. ation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb
17. The use of claim 15, wherein the medicament is designed to be administered intradermally, or subcutaneously, or intramuscularly.
18. The use of claim 13, wherein the medicament is designed to produce levels of the polypeptide of interest in the serum of the mammal of at least 50 pg/mL at least two hours after administration.
19. The use of claim 18, wherein the medicament is designed to produce levels of the polypeptide of st in the serum of the mammal of at least 50 pg/mL for at least 72 hours after administration.
20. The use of claim 19, wherein the medicament is designed to produce levels of the polypeptide of interest in the serum of the mammal of at least 60 pg/mL for at least 72 hours after stration.
21. The use of claim 13, wherein the medicament is designed to be administered in two or more equal or unequal split doses.
22. The use of claim 21, wherein the medicament is designed to produce higher levels of the polypeptide in the mammalian subject by administering split doses than by administering the same total daily dose as a single administration.
23. The use of claim 13, wherein the mammalian subject is a human patient in need of an increased level of the polypeptide of interest.
24. The use of claim 23, n the increased level of the ptide of interest is detectable in a bodily fluid of said patient.
25. The use of claim 24, wherein the bodily fluid is selected from the group consisting of peripheral blood, serum, plasma, ascites, urine, cerebrospinal fluid (CSF), sputum, saliva, bone , synovial fluid, aqueous humor, amniotic fluid, cerumen, breast milk, broncheoalveolar lavage fluid, semen, prostatic fluid, cowper's fluid or pre-ejaculatory fluid, sweat, fecal matter, hair, tears, cyst fluid, pleural and peritoneal fluid, pericardial fluid, lymph, chyme, chyle, bile, interstitial fluid, menses, pus, sebum, vomit, vaginal secretions, mucosal secretion, stool water, atic juice, lavage fluids from sinus cavities, bronchopulmonary aspirates, blastocyl cavity fluid, and umbilical cord blood.
26. The use of claim 23, wherein the medicament is ed to be administered according to a dosing regimen which occurs over the course of hours, days, weeks, months, or years.
27. The use of claim 16, n injection is achieved by using one or more s selected from [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb ation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb ation] jessb None set by jessb [Annotation] jessb ionNone set by jessb ation] jessb Unmarked set by jessb multi-needle injection systems, catheter or lumen systems, and ultrasound, electrical or radiation based systems.
28. The use of claim 21, wherein the amount of mRNA stered in any dose is substantially equal.
29. The use of claim 21, wherein the medicament is designed for administration of a first dose, a second dose or any of a plurality of doses at substantially the same time.
30. The use of claim 13, wherein the ment is designed to be administered as a single unit dose between about 10 mg/kg and about 500 mg/kg.
31. The use of claim 13, wherein the medicament is designed to be administered as a single unit dose between about 1.0 mg/kg and about 10 mg/kg.
32. The use of claim 13, wherein the medicament is designed to be administered as a single unit dose between about 0.001 mg/kg and about 1.0 mg/kg.
33. The isolated mRNA of any one of claims 1-10, wherein the sequence of n number of linked nucleosides comprises at least one chemical modification, and the al modification includes replacing or substituting an atom of a pyrimidine nucleobase with an amine, an SH, a methyl or ethyl, or a chloro or fluoro.
34. The isolated mRNA of any one of claims 1-10, wherein the n number of linked nucleosides comprise the Formula (Ia): Y1 Y5 B R3 R4 R5 R1' R1" R2" m" Y2 R2'm' Y3 P (Ia), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein: U is O, S, N(RU)nu, or C(RU)nu, wherein nu is an integer from 0 to 2 and each RU is, independently, H, halo, or optionally substituted alkyl; is a single or double bond; is a single bond or absent; [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb each of R1’, R2’, R1”, R2”, R1, R2, R3, R4, and R5, if present, is, independently, H, halo, hydroxy, thiol, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted loxy, optionally substituted lkoxy, ally substituted alkoxyalkoxy, optionally substituted hydroxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, ally tuted aminoalkenyl, optionally substituted lkynyl, or absent; wherein the combination of R3 with one or more of R1’, R1”, R2’, R2”, or R5 can join together to form optionally substituted alkylene or optionally substituted heteroalkylene and, taken together with the s to which they are attached, provide an optionally substituted heterocyclyl; wherein the ation of R5 with one or more of R1’, R1”, R2’, or R2” can join together to form optionally substituted alkylene or optionally substituted heteroalkylene and, taken together with the carbons to which they are attached, provide an optionally substituted heterocyclyl; and wherein the combination of R4 and one or more of R1’, R1”, R2’, R2”, R3, or R5 can join er to form optionally substituted alkylene or optionally substituted heteroalkylene and, taken together with the carbons to which they are attached, provide an optionally substituted heterocyclyl; each of m’ and m” is, independently, an integer from 0 to 3; each of Y1, Y2, and Y3, is, independently, O, S, Se, -NRN1-, optionally substituted alkylene, or optionally substituted heteroalkylene, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted l, optionally substituted aryl, or absent; each Y4 is, independently, H, hydroxy, thiol, boranyl, optionally substituted alkyl, optionally substituted alkenyl, ally substituted alkynyl, optionally substituted , optionally substituted alkenyloxy, optionally substituted alkynyloxy, ally substituted thioalkoxy, optionally substituted alkoxyalkoxy, or optionally substituted amino; each Y5 is, independently, O, S, Se, optionally substituted alkylene, or optionally substituted heteroalkylene; n is an integer from 1 to 100,000; and B is a base.
35. The ed mRNA of claim 34, wherein B is not pseudouridine (ψ) or 5-methyl-cytidine (m5C).
36. The ed mRNA of claim 34 or 35, wherein: U is O or C(RU)nu, wherein nu is an integer from 1 to 2 and each RU is, ndently, H, halo, or optionally substituted alkyl; each of R1, R1’, R1”, R2, R2′, and R2”, if present, is, independently, H, halo, hydroxy, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, ally substituted aminoalkoxy, ally substituted alkoxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl; [Annotation] jessb None set by jessb [Annotation] jessb ionNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb each of R3 and R4 is, independently, H, halo, hydroxy, optionally substituted alkyl, or optionally substituted alkoxyalkoxy; each of Y1, Y2, and Y3, is, independently, O, S, Se, -NRN1-, ally substituted alkylene, or optionally substituted heteroalkylene, wherein RN1 is H, optionally substituted alkyl, optionally tuted alkenyl, or ally tuted alkynyl; each Y4 is, independently, H, hydroxy, thiol, boranyl, optionally tuted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, ally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted thioalkoxy, or optionally substituted amino; each Y5 is, independently, O or optionally substituted alkylene; and n is an integer from 10 to 10,000.
37. The isolated mRNA of claim 36, wherein each of R1, R1’, and R1”, if present, is H.
38. The isolated mRNA of claim 37, wherein each of R 2, R2′, and R2”, if present, is, independently, H, halo, hydroxy, optionally substituted alkoxy, or optionally substituted alkoxyalkoxy.
39. The isolated mRNA of claim 38, wherein each of R2, R2′, and R2”, if present, is H.
40. The isolated mRNA of claim 39, wherein each of R 1, R1’, and R1”, if present, is, independently, H, halo, hydroxy, optionally substituted alkoxy, or ally substituted alkoxyalkoxy.
41. The isolated mRNA of claim 34, wherein the sequence of n number of linked nucleotides comprise the Formula (IIa): Y1 Y5 B R5 R1 R3 R4 3 PY (IIa), or a pharmaceutically acceptable salt or isomer thereof.
42. The isolated mRNA of claim 41, wherein the sequence of n number of linked nucleosides comprise the Formula (IIb) or (IIc): [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb Y1 Y5 B B U Y1 Y5 U R5 R1 R5 R1 R4 R3 R4 R2 R2 Y2 Y2 3 PY 3 PY Y4 Y4 (IIb), or (IIc), or a pharmaceutically acceptable salt thereof.
43. The ed mRNA of claim 42, n the sequence of n number of linked nucleosides comprise the Formula (IIb-1), (IIb-2), or (IIc-1)-(IIc-4): Y 1 Y5 B U Y1 Y5 B R2' R2 Y2 Y2 3 PY 3 PY Y4 Y4 (IIb-1), ), Y1 Y5 B U Y1 Y5 B R1 R1 R2 R2 Y2 Y2 3 PY 3 PY Y4 Y4 (IIc-1), (IIc-2), Y1 Y5 B U Y1 Y5 B R3 R3 R2 R2 Y2 Y2 3 PY 3 PY Y4 Y4 (IIc-3), or (IIc-4), or a pharmaceutically acceptable salt thereof.
44. The isolated mRNA of claim 34, wherein the sequence of n number of linked nucleosides se the Formula (IId): [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb ation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb Y1 Y5 B R5 R1 R3 R4 3 PY (IId), or a pharmaceutically acceptable salt or stereoisomer thereof.
45. The isolated mRNA of claim 43, wherein the sequence of n number of linked nucleosides comprise the Formula (IIe) or (IIf): Y1 Y5 B U Y1 Y5 B R5 R1 R5 R1 R3 R4 R3 R4 R2 R2 Y2 Y2 3 PY 3 PY Y4 Y4 (IIe) or (IIf), or a pharmaceutically acceptable salt f.
46. The isolated mRNA of claim 34, wherein each of said linked nucleotides ndently have one of Formulas (IIg)-(IIj): Y1 Y5 B Y1 Y5 B U U R3 R4 R3 R4 R5 R1' R1" R5 R1' R1" R2" R2" Y2 R2' Y2 R2' Y3 P Y3 P Y4 Y4 (IIg), (IIh), Y1 Y5 B3 Y1 Y5 B3 U U R3 Rb3 R3 Rb3 R5 B1 B2 R5 B1 B2 Rb2 Rb2 Y2 Rb1 Y2 Rb1 Y3 P Y3 P Y4 Y4 (IIi), or (IIj),or a pharmaceutically acceptable salt or stereoisomer thereof. [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb ation] jessb None set by jessb [Annotation] jessb ionNone set by jessb [Annotation] jessb Unmarked set by jessb
47. The isolated mRNA of claim 34, wherein the sequence of n number of linked nucleosides comprise the Formula (IIk): Y1 Y5 B R5 R1' R3 R4 Y2 m 3 PY (IIk) , or a pharmaceutically acceptable salt or stereoisomer thereof.
48. The isolated mRNA of claim 47, wherein the sequence of n number of linked nucleosides comprise the Formula (IIl): Y1 Y5 R3 R4 Y3 P (IIl), or a pharmaceutically acceptable salt or stereoisomer thereof.
49. The ed mRNA of claim 47, wherein the sequence of n number of linked sides comprise the Formula (IIm): Y1 Y5 B R3 R4 R5 R1' R1" Y2 R2' Y3 P (IIm), or a ceutically acceptable salt or stereoisomer thereof, wherein: each of R1’, R1”, R2′, and R2” is, independently, H, halo, hydroxy, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, or absent; and wherein the combination of R2′ and R3 or the combination of R2” and R3 can be taken together to form optionally substituted alkylene or optionally substituted heteroalkylene. [Annotation] jessb None set by jessb ation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb
50. The isolated mRNA of any one of claims 37-49, wherein: U is O or C(RU)nu, wherein nu is an integer from 1 to 2 and each RU is, independently, H, halo, or optionally substituted alkyl; each of R1 and R2 is, independently, H, halo, hydroxy, optionally tuted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted amino, azido, ally substituted aryl, optionally tuted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted lkynyl; each of R3 and R4 is, independently, H or optionally substituted alkyl; each of Y1, Y2, and Y3, is, independently, O, S, Se, -NRN1-, ally substituted alkylene, or optionally substituted heteroalkylene, wherein RN1 is H, optionally substituted alkyl, optionally substituted l, or optionally substituted alkynyl; each Y4 is, independently, H, hydroxy, thiol, boranyl, optionally substituted alkyl, optionally tuted alkenyl, ally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, ally substituted alkynyloxy, optionally substituted thioalkoxy, or optionally substituted amino; each Y5 is, independently, O or optionally substituted alkylene; and n is an integer from 10 to .
51. The isolated mRNA of claim 34, wherein the sequence of n number of linked sides comprise the Formula (IIn): Y1 Y5 B R3 R4 3 PY (IIn), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein: U is O or u, wherein nu is an integer from 1 to 2 and each RU is, ndently, H, halo, or optionally substituted alkyl; each of R1 and R4 is, independently, H, halo, hydroxy, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, ally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally substituted amino, azido, optionally substituted aryl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl; [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb ation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb ionNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb R3′ is O, S, or -NRN1-, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, or optionally substituted aryl; R3” is optionally substituted alkylene or optionally substituted heteroalkylene; each of Y1, Y2, and Y3, is, independently, O, S, Se, -NRN1-, ally substituted alkylene, or optionally substituted heteroalkylene, wherein RN1 is H, optionally substituted alkyl, optionally substituted alkenyl, or optionally substituted alkynyl; each Y4 is, independently, H, hydroxy, thiol, boranyl, optionally substituted alkyl, ally substituted alkenyl, ally substituted l, optionally substituted alkoxy, ally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted thioalkoxy, or optionally substituted amino; each Y5 is, ndently, O, S, optionally substituted alkylene (e.g., methylene), or ally substituted heteroalkylene; and n is an integer from 10 to 10,000.
52. The isolated mRNA of claim 34, wherein the sequence of n number of linked nucleosides comprise a (IIn-1) or (II-n2): Y1 Y5 B U Y1 Y5 B R3" R3" R3' O Y2 Y2 3 PY 3 PY Y4 Y4 (IIn-1) or (IIn-2), or a pharmaceutically acceptable salt or stereoisomer thereof.
53. The ed mRNA of any one of claims 34-52, wherein each B independently has a formula selected from Formula (b1)-(b5): T1' T1" R12c R12c R12c R12a R10 R10 V1 N R12a N N N N N T2" N R11 O R11 N T2" N O T2' T2' (b1), (b2), (b3), (b4), or R10 R12c N O (b5), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein: [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb ed set by jessb is a single or double bond; each of T1’, T1”, T2′, and T2” is, independently, H, optionally substituted alkyl, optionally substituted alkoxy, or optionally substituted thioalkoxy, or the ation of T1’ and T1” or the combination of T2′ and T2” join together (e.g., as in T2) to form O (oxo), S (thio), or Se (seleno); each of V1 and V2 is, independently, O, S, N(RVb)nv, or C(RVb)nv, wherein nv is an integer from 0 to 2 and each RVb is, ndently, H, halo, optionally substituted amino acid, optionally substituted alkyl, optionally substituted haloalkyl, optionally substituted l, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, ally substituted alkynyloxy, optionally substituted hydroxyalkyl, optionally substituted yalkenyl, optionally substituted hydroxyalkynyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted acylaminoalkyl, optionally substituted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, or optionally tuted alkoxycarbonylalkoxy; R10 is H, halo, optionally substituted amino acid, hydroxy, optionally substituted alkyl, ally substituted l, ally substituted alkynyl, optionally substituted aminoalkyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, ally tuted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted alkoxy, optionally substituted alkoxycarbonylalkyl, ally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted carboxyalkoxy, optionally substituted carboxyalkyl, or optionally substituted oylalkyl; R11 is H or optionally substituted alkyl; R12a is H, optionally substituted alkyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, ally substituted aminoalkyl, optionally substituted lkenyl, or optionally substituted lkynyl, ally substituted carboxyalkyl, optionally substituted carboxyalkoxy, optionally substituted carboxyaminoalkyl, or optionally substituted carbamoylalkyl; and R12c is H, halo, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted thioalkoxy, optionally substituted amino, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted yalkynyl, optionally substituted aminoalkyl, optionally substituted lkenyl, or optionally substituted aminoalkynyl.
54. The isolated mRNA of any one of claims 34-53, wherein B comprises Formula (b6)-(b9): [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb ionNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb R12c R12c T1' T1" R12c R12a R12a R12b R12a V3 N V3 N R12b N N N N W1 T2'' W1 W2 W2 T2 T2" T2" T2' T2' T2' (b6), (b7), (b8), or (b9), or a pharmaceutically able salt or stereoisomer thereof, wherein: is a single or double bond; each of T1’, T1”, T2′, and T2” is, independently, H, ally substituted alkyl, optionally substituted alkoxy, or ally substituted thioalkoxy, or the combination of T1’ and T1” join together or the combination of T2′ and T2” join together to form O (oxo), S (thio), or Se (seleno), or each T1 and T2 is, independently, O (oxo), S (thio), or Se (seleno); each of W1 and W2 is, independently, N(RWa)nw or C(RWa)nw, n nw is an integer from 0 to 2 and each RWa is, independently, H, optionally substituted alkyl, or optionally substituted alkoxy; each V3 is, independently, O, S, N(RVa)nv, or C(RVa)nv, wherein nv is an integer from 0 to 2 and each RVa is, independently, H, halo, optionally substituted amino acid, optionally substituted alkyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted heterocyclyl, optionally substituted alkheterocyclyl, optionally substituted alkoxy, optionally tuted alkenyloxy, or optionally substituted alkynyloxy, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted acylaminoalkyl, ally substituted carbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylacyl, ally substituted alkoxycarbonylalkoxy, ally substituted carboxyalkyl, optionally substituted carboxyalkoxy, optionally substituted carboxyaminoalkyl, or optionally substituted carbamoylalkyl, and wherein RVa and R12c taken together with the carbon atoms to which they are attached can form optionally substituted cycloalkyl, optionally substituted aryl, or optionally substituted heterocyclyl; R12a is H, ally tuted alkyl, ally substituted yalkyl, optionally tuted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, optionally substituted carboxyalkyl, optionally substituted carboxyalkoxy, optionally tuted carboxyaminoalkyl, optionally substituted carbamoylalkyl, or absent; R12b is H, optionally substituted alkyl, optionally tuted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted yalkynyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, [Annotation] jessb None set by jessb [Annotation] jessb ionNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb optionally substituted aminoalkynyl, optionally substituted l, optionally substituted heterocyclyl, optionally tuted alkheterocyclyl, optionally substituted amino acid, optionally substituted alkoxycarbonylacyl, optionally substituted alkoxycarbonylalkoxy, ally substituted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally substituted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted carboxyalkyl, optionally substituted carboxyalkoxy, optionally substituted carboxyaminoalkyl, or ally tuted carbamoylalkyl, wherein the combination of R12b and T1’ or the combination of R12b and R12c can join together to form optionally tuted cyclyl; and R12c is H, halo, optionally substituted alkyl, ally substituted alkoxy, optionally substituted thioalkoxy, optionally substituted amino, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl.
55. The isolated mRNA of claim 54, wherein R12a, R12b, R12c, or RVa is substituted with - (CH2)s2(OCH2CH2)s1(CH2)s3OR’, wherein s1 is an integer from 1 to 10, each of s2 and s3, independently, is an integer from 0 to 10, and R’ is H or C1-20 alkyl); or -NRN1(CH2)s2(CH2CH2O)s1(CH2)s3NRN1, wherein s1 is an integer from 1 to 10, each of s2 and s3, independently, is an integer from 0 to 10, and each RN1 is, independently, hydrogen or optionally substituted C1-6 alkyl.
56. The isolated mRNA of claim 54, wherein B comprises Formula (b28)-(b31): T1 T1 T1 RVb' R12a RVb' R12a R12b R12a N N N N RVb" N T2 N T2 T2 (b28), (b29), (b30), or RVb' R12a N T2 (b31), or a pharmaceutically acceptable salt or stereoisomer thereof.
57. The ed mRNA of any one of claims 34-56, n B comprises Formula (b10)-(b14): [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb ation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb ation] jessb ionNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb R13a R13b R13b R13a R13b N N N R14 R14 R16 V5 N N N R15 N T3" R15 N T3" R15 N T3" T3' T3' T3' (b10), (b11), (b12), R13a R13b V4 N V4 N R15 T3" R15 N T3" T3' T3' (b13), or (b14), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein: each of T3′ and T3” is, independently, H, optionally tuted alkyl, optionally substituted alkoxy, or optionally substituted thioalkoxy, or the combination of T3′ and T3” join together to form O (oxo), S (thio), or Se (seleno); each V4 is, independently, O, S, N(RVc)nv, or C(RVc)nv, wherein nv is an integer from 0 to 2 and each RVc is, independently, H, halo, optionally substituted amino acid, optionally substituted alkyl, ally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, ally substituted alkenyloxy, optionally substituted heterocyclyl, optionally substituted alkheterocyclyl, or optionally substituted alkynyloxy, wherein the combination of R13b and RVc can be taken together to form optionally substituted cyclyl; each V5 is, independently, N(RVd)nv, or C(RVd)nv, wherein nv is an integer from 0 to 2 and each RVd is, independently, H, halo, optionally tuted amino acid, optionally substituted alkyl, optionally tuted alkenyl, optionally substituted l, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted heterocyclyl, optionally substituted alkheterocyclyl, or optionally substituted alkynyloxy; each of R13a and R13b is, independently, H, ally substituted acyl, optionally substituted acyloxyalkyl, optionally substituted alkyl, or optionally substituted alkoxy, wherein the combination of R13b and R14 can be taken together to form optionally substituted cyclyl; each R14 is, independently, H, halo, hydroxy, thiol, optionally tuted acyl, optionally substituted amino acid, optionally tuted alkyl, optionally substituted haloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted , optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aminoalkoxy, optionally substituted alkoxyalkoxy, optionally tuted yalkyl, optionally substituted amino, azido, optionally substituted aryl, optionally substituted heterocyclyl, optionally [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb ation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb substituted alkheterocyclyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, or optionally substituted aminoalkynyl; and each of R15 and R16 is, independently, H, optionally substituted alkyl, optionally substituted alkenyl, or optionally substituted alkynyl.
58. The isolated mRNA of claim 57, n B comprises Formula (b32)-(b36): R13a R13b R13b N N T1 R14 R14 R16 R14 N N N N R13a R15 N T3 R15 N T3 N (b32), (b33), R13b (b34), R13a R13b R13b N N R14 R14a R15 N R15 N R14b (b35), or (b36) or a pharmaceutically acceptable salt or stereoisomer thereof.
59. The ed mRNA of any one of claims 34-58, n B comprises Formula (b15)-(b17): T4' T4" T5' T5" R23 V5 R18 N N N V6 R21 R24 N R19a N N N N N R19b (b15), R22 (b16), or T5' T5" N R18 N N T6' R22 (b17) or a pharmaceutically acceptable salt or stereoisomer thereof, wherein: each of T4’, T4”, T5′, T5”, T6’, and T6” is, independently, H, optionally substituted alkyl, or optionally substituted alkoxy, and wherein the ation of T4’ and T4” or the combination of T5′ and T5” or the combination of T6’ and T6” join together form O (oxo), S (thio), or Se o); each of V5 and V6 is, independently, O, S, N(RVd)nv, or C(RVd)nv, n nv is an integer from 0 to 2 and each RVd is, independently, H, halo, thiol, optionally substituted amino acid, cyano, amidine, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally tuted aminoalkynyl, optionally substituted alkyl, optionally substituted alkenyl, optionally [Annotation] jessb None set by jessb ation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted koxy, or optionally tuted amino; and each of R17, R18, R19a, R19b, R21, R22, R23, and R24 is, independently, H, halo, thiol, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally tuted thioalkoxy, optionally substituted amino, or optionally tuted amino acid.
60. The isolated mRNA of claim 59, wherein B comprises a (b37)-(b40): T4 T4' T4 N R18 N R18 N N N N R19a N N N R19a N N N R19a N N R19b (b37), R19b (b38), R19b (b39), or N R18 N R19a N N R19b (b40), or a ceutically acceptable salt or stereoisomer thereof.
61. The isolated mRNA of any one of claims 35-59, wherein B comprises Formula (b18)-(b20): R26a R26b R26b N N V7 V7 R28 N N R25 R25 N N R27 N N R27 (b18), (b19), or N N R27 (b20), or a pharmaceutically acceptable salt or stereoisomer thereof wherein: each V7 is, independently, O, S, N(RVe)nv, or nv, wherein nv is an integer from 0 to 2 and each RVe is, ndently, H, halo, optionally substituted amino acid, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted alkenyloxy, or optionally substituted alkynyloxy; [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb ation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb ed set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb ation] jessb Unmarked set by jessb each R25 is, ndently, H, halo, thiol, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally tuted thioalkoxy, or optionally substituted amino; each of R26a and R26b is, independently, H, optionally substituted acyl, optionally substituted amino acid, optionally substituted carbamoylalkyl, optionally substituted alkyl, ally substituted l, optionally tuted alkynyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted hydroxyalkynyl, optionally substituted alkoxy, or polyethylene glycol group, or an amino-polyethylene glycol group; each R27 is, independently, H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted alkoxy, optionally substituted thioalkoxy, or optionally substituted amino; each R28 is, ndently, H, optionally substituted alkyl, ally substituted l, or optionally tuted alkynyl; and each R29 is, independently, H, optionally substituted acyl, optionally substituted amino acid, optionally substituted oylalkyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted hydroxyalkyl, optionally substituted hydroxyalkenyl, optionally substituted alkoxy, or optionally substituted amino.
62. The isolated mRNA of claim 61, wherein B comprises Formula (b41)-(b43): R26a R26b R26a R26b R26a R26b N N N N N N N N N N N R27 N N N N (b41), (b42), or (b43), or a pharmaceutically acceptable salt or stereoisomer thereof.
63. The isolated mRNA of claim 61, wherein R26a, R26b, or R29 is substituted with -(CH2)s2(OCH2CH2)s1(CH2)s3OR’, wherein s1 is an integer from 1 to 10, each of s2 and s3, independently, is an integer from 0 to 10, and R’ is H or C1-20 alkyl); or -NRN1(CH2)s2(CH2CH2O)s1(CH2)s3NRN1, wherein s1 is an integer from 1 to 10, each of s2 and s3, independently, is an integer from 0 to 10, and each RN1 is, independently, hydrogen or optionally substituted C1-6 alkyl.
64. The isolated mRNA of any one of claims 34-63, wherein B ses Formula (b21): [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb ionNone set by jessb [Annotation] jessb Unmarked set by jessb xa X12 R12a N N (b21), or a pharmaceutically acceptable salt or stereoisomer thereof, n X12 is, independently, O, S, optionally substituted alkylene, or optionally substituted heteroalkylene; xa is an integer from 0 to 3; R12a is H, optionally substituted alkyl, optionally substituted aminoalkyl, optionally substituted lkenyl, optionally substituted aminoalkynyl, or absent; and T2 is O, S, or Se.
65. The isolated mRNA of any one of claims 34-64, wherein B comprises Formula (b22): O T1 R10' R12a N N R11 N T2 (b22), or a pharmaceutically acceptable salt or stereoisomer thereof, wherein R10’ is, independently, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, ally substituted aryl, optionally substituted heterocyclyl, optionally substituted aminoalkyl, ally substituted aminoalkenyl, optionally substituted lkynyl, optionally substituted alkoxy, optionally tuted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally substituted carbonylalkynyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted carboxyalkoxy, optionally substituted carboxyalkyl, or optionally tuted carbamoylalkyl; R11 is H or optionally substituted alkyl; R12a is H, optionally substituted alkyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, ally substituted aminoalkynyl, or absent; and each of T1 and T2 is, independently, O, S, or Se.
66. The ed mRNA of any one of claims 34-65, wherein B comprises Formula (b23): R10 R12a R11 N T2 (b23), wherein R10 is optionally substituted heterocyclyl or optionally substituted aryl; R11 is H or optionally substituted alkyl; R12a is H, optionally tuted alkyl, optionally substituted aminoalkyl, optionally substituted aminoalkenyl, optionally substituted aminoalkynyl, or absent; and each of T1 and T2 is, independently, O, S, or Se. [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb Unmarked set by jessb [Annotation] jessb None set by jessb [Annotation] jessb MigrationNone set by jessb [Annotation] jessb ed set by jessb
67. The isolated mRNA of any one of claims 34-66, wherein B comprises Formula (b24) or (b25): R13a R13b R13a R13b O N O N R14' N N R14' N N H H R15 N T3 R15 N T3 (b24) or (b25), wherein: T3 is O, S, or Se; each of R13a and R13b is, ndently, H, ally substituted acyl, optionally substituted alkyl, or ally substituted alkoxy, wherein the combination of R13b and R14 can be taken together to form ally substituted cyclyl; R14’ is, independently, optionally tuted alkyl, optionally substituted alkenyl, ally substituted alkynyl, optionally substituted aryl, optionally substituted heterocyclyl, optionally substituted alkheterocyclyl, optionally substituted alkaryl, optionally substituted aminoalkyl, optionally tuted aminoalkenyl, optionally substituted aminoalkynynyl, optionally substituted alkoxy, optionally substituted alkoxycarbonylalkyl, optionally substituted alkoxycarbonylalkenyl, optionally tuted alkoxycarbonylalkynyl, optionally substituted alkoxycarbonylalkoxy, optionally substituted carboxyalkoxy, optionally substituted carboxyalkyl, or optionally substituted carbamoylalkyl; and each R15 is, independently, H, optionally substituted alkyl, ally substituted alkenyl, or optionally substituted alkynyl.
68. The isolated mRNA of any one of claims 34-67, wherein B comprises Formula (b26) or (b27): (b26) or (b27).
69. The isolated mRNA of any one of claims 34-68, wherein B comprises Formula (b28)-(b43).
70. The isolated mRNA of claim 35, wherein said isolated mRNA is prepared from one or more building blocks selected from BB-1 to BB-274, or a pharmaceutically acceptable salts or stereoisomers thereof.
71. The isolated mRNA of claim 34, n said isolated mRNA is prepared from one or more building blocks selected from compounds 1-50, or a pharmaceutically acceptable salts or stereoisomers thereof.
72. The isolated mRNA of claim 1, substantially as hereinbefore described with reference to any one of the Examples.
73. The pharmaceutical composition of claim 11, substantially as before described with reference to any one of the Examples.
74. The use of claim 13, substantially as hereinbefore described with reference to any one of the Examples. 33 5R '8 ii fix." an: h 53 .5: :3 \‘k Kg (:3 Ks 1? k )\ wk ¢$ M a': \‘ix. c. 2’33? 4 . 4 . 4 . 4 . 1 : ”W“ gamma/«(W :n. N :2) x. r o) u m ct. 1“:- !\ Q) m. r \‘J ((4 “ gx. 3x \x‘ v. m x: m as 5.; .6 in 3E3. 2 =0 SUBSTITUTE SHEET (RULE 26)
NZ623476A 2011-10-03 2012-10-03 Modified nucleosides, nucleotides, and nucleic acids, and uses thereof NZ623476B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201161542533P 2011-10-03 2011-10-03
US61/542,533 2011-10-03
PCT/US2012/058519 WO2013052523A1 (en) 2011-10-03 2012-10-03 Modified nucleosides, nucleotides, and nucleic acids, and uses thereof

Publications (2)

Publication Number Publication Date
NZ623476A NZ623476A (en) 2016-09-30
NZ623476B2 true NZ623476B2 (en) 2017-01-05

Family

ID=

Similar Documents

Publication Publication Date Title
US20210308283A1 (en) Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US10286086B2 (en) Alternative nucleic acid molecules and uses thereof
US20240165267A1 (en) Modified nucleic acid molecules and uses thereof
EP2918275B1 (en) Alternative nucleic acid molecules and uses thereof
WO2015196118A1 (en) Alternative nucleic acid molecules and uses thereof
US20170175129A1 (en) Alternative nucleic acid molecules and uses thereof
NZ623476B2 (en) Modified nucleosides, nucleotides, and nucleic acids, and uses thereof