WO2018232355A1 - Anticorps d'arn - Google Patents

Anticorps d'arn Download PDF

Info

Publication number
WO2018232355A1
WO2018232355A1 PCT/US2018/037918 US2018037918W WO2018232355A1 WO 2018232355 A1 WO2018232355 A1 WO 2018232355A1 US 2018037918 W US2018037918 W US 2018037918W WO 2018232355 A1 WO2018232355 A1 WO 2018232355A1
Authority
WO
WIPO (PCT)
Prior art keywords
composition
mir
antibody
sequence
polynucleotide
Prior art date
Application number
PCT/US2018/037918
Other languages
English (en)
Inventor
Sunny HIMANSU
Elisabeth NARAYANAN
Giuseppe Ciaramella
Original Assignee
Modernatx, 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 Modernatx, Inc. filed Critical Modernatx, Inc.
Publication of WO2018232355A1 publication Critical patent/WO2018232355A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/08Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
    • C07K16/10Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses from RNA viruses
    • C07K16/1036Retroviridae, e.g. leukemia viruses
    • C07K16/1045Lentiviridae, e.g. HIV, FIV, SIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/42Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum viral
    • 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
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/08Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
    • C07K16/10Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses from RNA viruses
    • C07K16/1018Orthomyxoviridae, e.g. influenza virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/525Virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/53DNA (RNA) vaccination
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/569Single domain, e.g. dAb, sdAb, VHH, VNAR or nanobody®
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/64Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising a combination of variable region and constant region components
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance
    • 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
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16111Human Immunodeficiency Virus, HIV concerning HIV env
    • C12N2740/16134Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • 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
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/14011Filoviridae
    • C12N2760/14111Ebolavirus, e.g. Zaire ebolavirus
    • C12N2760/14134Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • 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
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16111Influenzavirus A, i.e. influenza A virus
    • C12N2760/16134Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • Infectious diseases kill over 10 million people per year globally and are the fourth leading cause of death in the United States. They also represent a $68 billion dollar pharmaceutical market. Antibiotics and anti-virals overlook millennia of nature’s learnings by failing to utilize the immune system. And while intravenous immunoglobulin heeds nature’s example, its applications in infectious disease are few and its limitations numerous (e.g., cost, cumbersome manufacturing, risk of infection).
  • RNA compositions relate to compositions and methods of preventing and/or treating conditions, diseases, and disorders related to infectious disease and cancer.
  • a ribonucleic acid (RNA) composition is provided herein that builds on the knowledge that RNA (e.g., messenger RNA (mRNA)) can safely direct the body’s cellular machinery to produce nearly any protein of interest, from native proteins to antibodies and other entirely novel protein constructs that can have therapeutic activity inside and outside of cells.
  • mRNA messenger RNA
  • RNA compositions of the disclosure may be utilized in various settings depending on the prevalence of the infection or the degree or level of unmet medical need.
  • the RNA compositions may be utilized to treat and/or prevent an infection by bacteria or virus of various genotypes, strains, and isolates.
  • the disclosure relates to RNA compositions that may be utilized to provide passive immunization against an infectious disease.
  • the disclosure relates to methods of treating and/or preventing infectious disease or cancer in a subject.
  • the invention is a composition, comprising: a ribonucleic acid (RNA) polynucleotide having an open reading frame encoding an antibody or antigen-binding fragment thereof, wherein the composition when administered to a subject in need thereof as a single intravenous dose is sufficient to (i) maintain antibody levels at a normal
  • composition includes a delivery agent.
  • the present disclosure further provides a method of expressing antibodies in a human subject in need thereof comprising administering to the subject an effective amount of a pharmaceutical composition or a polynucleotide, e.g. an mRNA, described herein, wherein the pharmaceutical composition or polynucleotide is suitable for administrating as a single dose or as a plurality of single unit doses to the subject.
  • the drug may be administered in a clinical setting, e.g., hospital or clinical site, in an IV infusion over a few hours. For instance, it may be administered as a bolus IV injection, or as a procedure carried out in a day for a patient in the clinic/hospital.
  • the single dose may be followed up by subsequent treatments, at a certain frequency, every week, two weeks, three weeks, four weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, every month, two months, three months, four months, five months, six months, or every year.
  • the invention is a pharmaceutical composition, comprising a ribonucleic acid (RNA) polynucleotide having an open reading frame encoding an antibody or antigen-binding fragment thereof and a lipid nanocarrier.
  • RNA ribonucleic acid
  • the polynucleotide is single stranded.
  • the polynucleotide is double stranded.
  • the polynucleotide is RNA.
  • the polynucleotide is mRNA.
  • the polynucleotide comprises at least one chemically modified nucleobase, sugar, backbone, or any combination thereof.
  • the at least one chemically modified nucleobase is selected from the group consisting of pseudouracil ( ⁇ ), N1-methylpseudouracil 2-thiouracil (s2U), 4’-thiouracil, 5-methylcytosine, 5-methyluracil, and any combination thereof.
  • the at least one chemically modified nucleobase is a modified uracil such as 5-methoxyuracil.
  • At least about 25%, at least about 30%, at least about 40%, 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 99%, or 100% of the uracils are 5-methoxyuracils.
  • the polynucleotide further comprises a miRNA binding site.
  • the miRNA binding site binds to miR-142.
  • the miRNA binding site binds to miR-142-3p or miR-142-5p.
  • the miR-142 comprises SEQ ID NO: 104.
  • the polynucleotide further comprises a 5' UTR.
  • the 5' UTR comprises a nucleic acid sequence at least 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99%, or about 100% identical to a sequence selected from the group consisting of SEQ ID NOs:71-88.
  • the polynucleotide further comprises a 3' UTR.
  • the 3' UTR comprises a nucleic acid sequence at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99%, or about 100% identical to a sequence selected from the group consisting of SEQ ID NOs:89-99.
  • the miRNA binding site is located within the 3' UTR.
  • the polynucleotide has a 5' terminal cap selected from a Cap0, Cap1, ARCA, inosine, N1-methyl-guanosine, 2′-fluoro-guanosine, 7-deaza-guanosine, 8-oxo- guanosine, 2-amino-guanosine, LNA-guanosine, 2-azidoguanosine, Cap2, Cap4, 5' methylG cap, or an analog thereof.
  • the polynucleotide further comprises a poly-A region.
  • the poly-A region is at least about 10, at least about 20, at least about 30, at least about 40, at least about 50, at least about 60, at least about 70, at least about 80, or at least about 90 nucleotides in length.
  • the poly-A region has about 10 to about 200, about 20 to about 180, about 50 to about 160, about 70 to about 140, or about 80 to about 120 nucleotides in length.
  • the polynucleotide has:
  • the polynucleotide comprises:
  • the 3'-UTR comprises a miRNA binding site.
  • the present disclosure provides, in certain aspects, a method of producing a polynucleotide as described herein, the method comprising modifying an ORF encoding an antibody by substituting at least one uracil nucleobase with an adenine, guanine, or cytosine nucleobase, or by substituting at least one adenine, guanine, or cytosine nucleobase with a uracil nucleobase, wherein all the substitutions are synonymous substitutions.
  • the method further comprises replacing at least about 90%, at least about 95%, at least about 99%, or about 100% of uracils with 5-methoxyuracils.
  • composition comprising a polynucleotide as described herein;
  • the delivery agent comprises a lipid nanoparticle including an ionizable lipid.
  • the ionizable lipid is compound 18 or compound 25.
  • the delivery agent further comprises a structural lipid.
  • the structural lipid is selected from the group consisting of cholesterol, fecosterol, sitosterol, ergosterol, campesterol, stigmasterol, brassicasterol, tomatidine, ursolic acid, alpha-tocopherol, and any mixtures thereof.
  • the delivery agent further comprises a PEG lipid.
  • the PEG lipid is selected from the group consisting of a PEG- modified phosphatidylethanolamine, a PEG-modified phosphatidic acid, a PEG-modified ceramide, a PEG-modified dialkylamine, a PEG-modified diacylglycerol, a PEG-modified dialkylglycerol, compound 428 and any mixtures thereof.
  • the present disclosure provides a polynucleotide comprising an open reading frame (ORF) encoding an antibody, wherein the uracil or thymine content of the ORF relative to the theoretical minimum uracil or thymine content of a nucleotide sequence encoding the antibody (%U TM or %T TM ) is between about 125% and about 150%.
  • ORF open reading frame
  • the %U TM or %T TM is between about 100% and about 220%, about 134% and about 140%, about 134% and about 145%, about 130% and about 145%, about 120% and about 140%, about 124% and about 130%, about 130% and about 140%, about 114% and about 150%, or about 134% and about 148%.
  • the uracil or thymine content of the ORF relative to the uracil or thymine content of the corresponding wild-type ORF is less than 100%.
  • the %U WT or %T WT is less than about 95%, less than about 90%, less than about 85%, less than about 80%, less than about 75%, or less than 74%.
  • the %U WT or %T WT is between 68% and 74%.
  • the uracil or thymine content in the ORF relative to the total nucleotide content in the ORF is less than about 50%, less than about 40%, less than about 30%, or less than about 21%.
  • the %U TL or %T TL is less than about 21%.
  • the %U TL or %T TL is between about 14% and about 16%.
  • the guanine content of the ORF with respect to the theoretical maximum guanine content of a nucleotide sequence encoding the antibody (%G TMX ) is at least 71%, at least 72%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or about 100%.
  • the %G TMX is between about 72% and about 80%, between about 72% and about 79%, between about 73% and about 78%, or between about 74% and about 77%.
  • the cytosine content of the ORF relative to the theoretical maximum cytosine content of a nucleotide sequence encoding the antibody is at least 63%, at least 64%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, or about 100%.
  • the %C TMX is between about 65% and about 80%, between about 65% and about 79%, between about 65% and about 78%, or between about 72% and about 77%.
  • the guanine and cytosine content (G/C) of the ORF relative to the theoretical maximum G/C content in a nucleotide sequence encoding the antibody (%G/C TMX ) is at least about 81%, at least about 82%, at least about 85%, at least about 90%, at least about 95%, or about 100%. In some embodiments, the %G/C TMX is between about 80% and about 100%, between about 85% and about 99%, between about 90% and about 97%, or between about 90% and about 93%.
  • the G/C content in the ORF relative to the G/C content in the corresponding wild-type ORF is at least 102%, at least 103%, at least 104%, at least 105%, at least 106%, at least 107%, or at least about 110%.
  • the average G/C content in the 3 rd codon position in the ORF is at least 20%, at least 21%, at least 22%, at least 23%, or at least 24% higher than the average G/C content in the 3 rd codon position in the corresponding wild-type ORF.
  • the ORF further comprises at least one low-frequency codon.
  • the composition is formulated for in vivo delivery.
  • the composition is formulated for intramuscular, subcutaneous, or intradermal delivery.
  • the present disclosure provides a composition
  • a composition comprising an RNA (e.g., mRNA) polynucleotide having an open reading frame encoding a single-domain antibody or a fragment thereof having specificity for Ebola virus (e.g., a fragment capable of binding to the Ebola virus).
  • the single-domain antibody or fragment thereof is a variable domain of a heavy-chain (V H H) antibody.
  • the single-domain antibody e.g., a V H H antibody
  • the single-domain antibody or fragment thereof has binding specificity for an Ebola virus (EBOV) protein.
  • the EBOV protein is an EBOV glycoprotein.
  • the EBOV glycoprotein is GP, secreted GP (sGP), small sGP (ssGP), surface GP, or shed GP.
  • the EBOV protein is an EBOV nucleoprotein.
  • the EBOV protein is an EBOV matrix protein.
  • the EBOV matrix protein is VP24, VP30, VP35, or VP30.
  • the methods and compositions of the present application are useful for preventing and/or treating infection by Ebola virus.
  • the Ebola virus is from Ebola virus species Zaire ebolavirus, species Sudan ebolavirus, species
  • Bundibugyo ebolavirus species Ta ⁇ Forest ebolavirus, species Reston ebolavirus, or a combination thereof.
  • the RNA polynucleotide encodes a single-domain antibody having greater than 90% identity to an amino acid sequence of any one of Tables 1 and 2. In some embodiments, the RNA polynucleotide encodes a single-domain antibody having greater than 95% identity to an amino acid sequence of any one of Tables 1 and 2. In some embodiments, the RNA polynucleotide encodes a single-domain antibody having greater than 96% identity to an amino acid sequence of any one of Tables 1 and 2. In some embodiments, the RNA polynucleotide encodes a single-domain antibody having greater than 97% identity to an amino acid sequence of any one of Tables 1 and 2.
  • the RNA polynucleotide encodes a single-domain antibody having greater than 98% identity to an amino acid sequence of any one of Tables 1 and 2. In some embodiments, the RNA polynucleotide encodes a single-domain antibody having greater than 99% identity to an amino acid sequence of any one of Tables 1 and 2. In some embodiments, the RNA polynucleotide encodes a single-domain antibody having greater than 95-99% identity to an amino acid sequence of any one of Tables 1 and 2.
  • the RNA polynucleotide encodes a single-domain antibody having an amino acid sequence of any one of Tables 1 and 2, and wherein the RNA polynucleotide is codon optimized mRNA.
  • RNA e.g., mRNA
  • an Ebola virus treatment that includes at least one RNA (e.g., mRNA) polynucleotide having an open reading frame encoding a single-domain antibody, at least one 5′ terminal cap, and at least one chemical modification.
  • RNA e.g., mRNA
  • the invention is a ribonucleic acid (RNA) polynucleotide having an open reading frame encoding an antibody or single-chain variable fragment (scFv) having specificity for a human immunodeficiency virus (HIV) and a pharmaceutically acceptable carrier or excipient.
  • scFv comprises variable regions of heavy (VH) and light chains (VL) of immunoglobulins, connected with a linker peptide of about 4 to about 30 amino acids.
  • the scFv comprises variable regions of heavy (VH) and light chains (VL) of immunoglobulins, connected with a linker peptide of about 20 amino acids.
  • the antibody or scFv comprises a polypeptide sequence of any of SEQ ID NOs: 182-198; a polypeptide sequence having at least 80% sequence identity to any one of SEQ ID NOs: 182-198; a polypeptide sequence having at least 90% sequence identity to any one of SEQ ID NOs: 182-198; a polypeptide sequence having at least 95% sequence identity to any one of SEQ ID NOs: 182-198; a polypeptide sequence having at least 98% sequence identity to any one of SEQ ID NOs: 182-198; or a polypeptide sequence having at least 99% sequence identity to any one of SEQ ID NOs: 182-198.
  • the ORF comprises a nucleic acid sequence having at least 80%, 85%, 90%, 95%, or 100% sequence identity to any one of SEQ ID NOs: 166-171.
  • the RNA comprises a nucleic acid sequence having at least 80%, 85%, 90%, 95%, or 100% sequence identity to any one of SEQ ID NOs: 174-179.
  • the invention is a composition, of a ribonucleic acid (RNA) polynucleotide having an open reading frame encoding an isolated antibody that specifically binds to human immunodeficiency virus (HIV), wherein the antibody has two or more of the following characteristics: (a) demonstrates neutralization of HIV, with an ID 50 of within 20% of a control neutralization activity, wherein the control is a corresponding protein antibody; (b) is an scFv that demonstrates protection, as measured by decreased plasma viremia relative to baseline prior to administration of the scFv in an animal model of HIV infection when administered either before or after virus challenge; (c) is an scFv having a (G4S) 4 linker; or (d) wherein the antibody or scFv comprises three complementarity determining regions (CDRs) contained within any one of the variable region sequences listed in SEQ ID NO.200- 217 and a pharmaceutically acceptable carrier or excipient.
  • the antibody is a full length antibody or
  • RNA ribonucleic acid
  • ORF open reading frame
  • the first ORF encodes an antibody that specifically interacts with a CD4 binding site. In some embodiments the first ORF encodes an N6 IgG.
  • the composition includes four RNA polynucleotides.
  • two of the four RNA polynucleotides comprise ORFs encoding a light chain and a heavy chain of an N6 IgG.
  • the ORFs encoding a light chain and a heavy chain of an N6 IgG have a nucleic acid sequence of SEQ ID NO.171 and 170 respectively.
  • the ORFs encoding a light chain and a heavy chain of an N6 IgG have a nucleic acid sequence having at least 80%, 85%, 90%, 95%, or 99% sequence identity to SEQ ID NO.171 and 170 respectively.
  • the first ORF encodes a polypeptide having at least 80%, 81%, 82%, 93%, 94%, 95%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to SEQ ID NO.186-187.
  • the second ORF encodes an scFv that specifically interacts with a V1/V2 region of HIV.
  • the second ORF encodes PDGM_1400 (scFV- FC_var7).
  • the second ORF encodes a polypeptide having at least 80%, 81%, 82%, 93%, 94%, 95%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to SEQ ID NO.185, 188-192.
  • the second ORF has at least 80%, 81%, 82%, 93%, 94%, 95%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to SEQ ID NO.169.
  • the RNA polynucleotide having the second ORF has a nucleic acid sequence of SEQ ID NO.177.
  • the RNA polynucleotide having the second ORF has at least 80%, 85%, 90%, 95%, or 99% sequence identity to a nucleic acid sequence of SEQ ID NO.177.
  • the scFv that specifically interacts with a V1/V2 region of HIV demonstrates at least 2 times higher neutralization of HIV than an scFv that specifically interacts with a V1/V2 region of HIV and includes a (GGGS)3 (SEQ ID NO: 199) linker.
  • the third ORF encodes an scFv that specifically interacts with a V3 base region.
  • the third ORF encodes a variant of PGT_121.
  • the third ORF encodes a polypeptide having at least 80%, 81%, 82%, 93%, 94%, 95%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to SEQ ID NO.182-184, 193-194, 197-198.
  • the third ORF has at least 80%, 81%, 82%, 93%, 94%, 95%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to SEQ ID NO.166-168. In some embodiments the third ORF has at least 90%, 95%, 985, or 100% sequence identity to SEQ ID NO.166-168. In other embodiments the RNA
  • polynucleotide having the third ORF has a nucleic acid sequence of SEQ ID NO.174-176.
  • RNA polynucleotide having the third ORF has at least 80%, 85%, 90%, 95%, or 99% sequence identity to a nucleic acid sequence of SEQ ID NO.174-176.
  • the third ORF encodes a functional variant of a polypeptide having at least 80%, 85%, 90%, 95%, 98% or 100% sequence identity to SEQ ID NO.195-196.
  • the scFv that specifically interacts with a V3 region of HIV demonstrates at least 2 times higher neutralization of HIV than an scFv that specifically interacts with a V3 region of HIV and includes a (GGGS)3 (SEQ ID NO: 199) linker.
  • the composition is formulated in a lipid nanoparticle (LNP) comprising an ionizable amino lipid, a structural lipid, a PEG lipid and a non-cationic lipid.
  • LNP lipid nanoparticle
  • each RNA polynucleotide is formulated in a separate LNP.
  • all the RNA polynucleotides are formulated in a separate LNP.
  • the invention is a method of treating an HIV infection in a subject, by administering to a subject any of the compositions described herein in a therapeutically effective amount to treat the HIV infection.
  • the subject is a human.
  • the method of treating is a method of passively immunizing a mammalian subject against an HIV infection by administering to the subject the composition, wherein the subject is at risk of having or being exposed to an influenza virus infection.
  • the invention is some aspect is a method of treating an HIV infection in a subject, comprising administering to a subject the composition as described herein, wherein each of the RNA polynucleotides in the composition is administered separately to the subject or is administered together in a single formulation in a therapeutically effective amount to treat the HIV infection.
  • a 5’ terminal cap is 7mG(5')ppp(5')NlmpNp.
  • the RNA polynucleotide comprises at least one chemical modification is selected from pseudouridine, N1-methylpseudouridine, 2-thiouridine, 4’- thiouridine, 5-methylcytosine, 2-thio-1-methyl-1-deaza-pseudouridine, 2-thio-1-methyl- pseudouridine, 2-thio-5-aza-uridine , 2-thio-dihydropseudouridine, 2-thio-dihydrouridine, 2- thio-pseudouridine, 4-methoxy-2-thio-pseudouridine, 4-methoxy-pseudouridine, 4-thio-1- methyl-pseudouridine, 4-thio-pseudouridine, 5-aza-uridine, dihydropseudouridine, 5- methyluridine,), 5-methoxyuridine and 2’-O-methyl uridine.
  • the RNA polynucleotide is formulated in a lipid nanoparticle (LNP) carrier.
  • the lipid nanoparticle carrier comprises a molar ratio of about 20-60% cationic lipid: 5-25% non-cationic lipid: 25-55% sterol; and 0.5-15% PEG- modified lipid.
  • the cationic lipid is selected from the group consisting of for example, 2,2-dilinoleyl-4-dimethylaminoethyl-[1,3]-dioxolane (DLin-KC2-DMA), dilinoleyl-methyl-4-dimethylaminobutyrate (DLin-MC3-DMA), and di((Z)-non-2-en-1-yl) 9- ((4-(dimethylamino)butanoyl)oxy)heptadecanedioate (L319).
  • DLin-KC2-DMA 2,2-dilinoleyl-4-dimethylaminoethyl-[1,3]-dioxolane
  • DLin-MC3-DMA dilinoleyl-methyl-4-dimethylaminobutyrate
  • a lipid nanoparticle comprises a cationic lipid, a PEG-modified lipid, a sterol and a non-cationic lipid.
  • the cationic lipid is an ionizable cationic lipid and the non- cationic lipid is a neutral lipid, and the sterol is a cholesterol.
  • Some embodiments of the present disclosure provide an Ebola virus treatment that includes at least one RNA polynucleotide having an open reading frame encoding a single- domain antibody, wherein at least 80% of the uracil in the open reading frame have a chemical modification, optionally wherein the Ebola virus treatment is formulated in a lipid nanoparticle.
  • 100% of the uracil in the open reading frame have a chemical modification.
  • a chemical modification is in the 5-position of the uracil.
  • a chemical modification is an N1-methyl pseudouridine.
  • Some embodiments of the present disclosure provide methods of treating an Ebola virus infection in a subject, the method comprising administering to a subject having an Ebola virus infection a composition described herein in a therapeutically effective amount to treat the Ebola virus infection.
  • the composition comprises a polynucleotide that encodes a polypeptide targeted against a viral protein.
  • an Ebola virus treatment is administered to the subject subcutaneously, intraocularly, intravitreally, parenterally, subcutaneously, intravenously, intracerebro-ventricularly, intramuscularly, intrathecally, orally, intraperitoneally, by oral or nasal inhalation, or by direct injection to one or more cells, tissues, or organs.
  • the subject is a mammal. In some embodiments, the subject is a human.
  • the mammalian subject is a human.
  • the subject is a non-human primate.
  • Non-limiting examples of non-human primate subjects include macaques, marmosets, tamarins, spider monkeys, owl monkeys, vervet monkeys, squirrel monkeys, baboons, gorillas, chimpanzees, and orangutans.
  • Other exemplary subjects include domesticated animals such as dogs and cats; livestock such as horses, cattle, pigs, sheep, goats, and chickens; and other animals such as mice, rats, guinea pigs, and hamsters.
  • the present disclosure provides a composition
  • a composition comprising an RNA (e.g., mRNA) polynucleotide having an open reading frame encoding a heavy-chain-only antibody (HCAb) having specificity for an influenza virus (e.g., an HCAb or a fragment thereof capable of binding to the influenza virus) and a pharmaceutically acceptable carrier or excipient.
  • HCAb comprises a fragment crystallizable (Fc) region.
  • the RNA polynucleotide encodes an HCAb comprising a polypeptide sequence of any of SEQ ID NOs: 1-10. In some embodiments, the RNA polynucleotide encodes an HCAb comprising a polypeptide sequence having at least 80% sequence identity to any one of SEQ ID NOs: 1-10. In some embodiments, the RNA polynucleotide encodes an HCAb comprising a polypeptide sequence having at least 90% sequence identity to any one of SEQ ID NOs: 1-10. In some embodiments, the RNA polynucleotide encodes an HCAb comprising a polypeptide sequence having at least 95% sequence identity to any one of SEQ ID NOs: 1-10.
  • the RNA polynucleotide encodes an HCAb comprising a polypeptide sequence having at least 98% sequence identity to any one of SEQ ID NOs: 1-10. In some embodiments, the RNA polynucleotide encodes an HCAb comprising a polypeptide sequence having at least 99% sequence identity to any one of SEQ ID NOs: 1-10.
  • RNA ribonucleic acid
  • HA hemagglutinin subtype
  • the composition is formulated in a lipid nanocarrier.
  • the antibody or antigen-binding fragment thereof comprises a polypeptide sequence of any of SEQ ID NOs: 1-10 or has at least 80%, 85%, 90%, 95%, 97%, 98%, or 99% sequence identity to any one of SEQ ID NOs: 1-10.
  • the antibody or antigen- binding fragment thereof is a broadly neutralizing antibody that cross-protects against influenza strains having different HA subtypes. In other embodiments the antibody cross protects against influenza virus having one or more of H1, H2 or H3. In some embodiments the antibody or antigen-binding fragment thereof binds to an epitope in the HA stem. In some embodiments the antibody or antigen-binding fragment thereof binds to a hydrophobic groove on the HA stem. In other embodiments the antibody or antigen-binding fragment thereof binds to an epitope in HA that prevents the viral fusion process. In yet other embodiments the antibody or antigen-binding fragment thereof binds to an epitope in HA that prevents virus attachment.
  • the epitope is THLKFKYPAL...TGN.
  • the epitope is an H3 epitope having the following amino acid sequence: Xaa1 Xaa2 Leu Xaa3 Xaa4 Lys Tyr Pro Xaa5 Xaa6, wherein Xaa1 is Thr, His, or Tyr; Xaa2 is His, Lys, Asn or Gln; Xaa3 is Lys, Glu or Asn; Xaa4 is Phe or Tyr, Xaa5 is Ala of Glu; and Xaa6 is Leu or Gln.
  • the epitope is an H3 epitope having the following amino acid sequence: Xaa1 Xaa2 Leu Xaa3 Xaa4 Lys Tyr Pro Xaa5 Xaa6 Xaa7 Gly Asn, wherein Xaa1 is Thr, His, or Tyr; Xaa2 is His, Lys, Asn or Gln; Xaa3 is Lys, Glu or Asn; Xaa4 is Phe or Tyr, Xaa5 is Ala of Glu; Xaa6 is Leu or Gln, and Xaa 7 is Thr or Lys.
  • the RNA polynucleotide encodes an HCAb comprising a polypeptide sequence of SEQ ID NO: 11. In some embodiments, the RNA polynucleotide encodes an HCAb comprising a polypeptide sequence of SEQ ID NO: 12. In some embodiments, the RNA polynucleotide encodes an HCAb comprising a polypeptide sequence of SEQ ID NO: 13. In some embodiments, the HCAb binds to a hemagglutinin (HA) protein of the influenza virus. In some embodiments, the HA protein is a type 3 HA (H3). In some embodiments, the HA protein is a type 1 HA (H1).
  • HA hemagglutinin
  • influenza virus is a Type A influenza virus. In some embodiments, the Type A influenza virus is of subtype H3N2. In some embodiments, the Type A influenza virus is of subtype H1N1.
  • composition comprising the RNA (e.g., mRNA) polynucleotide having the open reading frame further comprises an adjuvant.
  • the open reading frame is codon-optimized.
  • the RNA (e.g., mRNA) polynucleotide comprises at least one chemical modification.
  • the chemical modification is selected from pseudouridine, N1-methylpseudouridine, N1-ethylpseudouridine, 2-thiouridine, 4′- thiouridine, 5-methylcytosine, 2-thio-1-methyl-1-deaza-pseudouridine, 2-thio-1-methyl- pseudouridine, 2-thio-5-aza-uridine , 2-thio-dihydropseudouridine, 2-thio-dihydrouridine, 2- thio-pseudouridine, 4-methoxy-2-thio-pseudouridine, 4-methoxy-pseudouridine, 4-thio-1- methyl-pseudouridine, 4-thio-pseudouridine, 5-aza-uridine, dihydropseudouridine, 5- methyluridine,), 5-methoxy
  • the RNA (e.g., mRNA) polynucleotide comprises at least one 5′ terminal cap. In some embodiments, the at least one 5′ terminal cap comprises
  • the composition comprising the RNA (e.g., mRNA) polynucleotide is formulated in a nanoparticle.
  • the nanoparticle has a mean diameter of 50-200 nm.
  • the nanoparticle is a lipid nanoparticle.
  • the lipid nanoparticle comprises a cationic lipid, a PEG-modified lipid, a sterol and a non-cationic lipid.
  • the cationic lipid is an ionizable cationic lipid and the non-cationic lipid is a neutral lipid, and the sterol is a cholesterol.
  • the cationic lipid is selected from 2,2-dilinoleyl-4- dimethylaminoethyl-[1,3]-dioxolane (DLin-KC2-DMA), dilinoleyl-methyl-4- dimethylaminobutyrate (DLin-MC3-DMA), and di((Z)-non-2-en-1-yl) 9-((4- (dimethylamino)butanoyl)oxy)heptadecanedioate (L319).
  • the nanoparticle has a polydispersity value of less than 0.4.
  • the nanoparticle has a net neutral charge at a neutral pH value.
  • the lipid nanoparticle comprises a molar ratio of about 20-60% cationic lipid:5-25% non-cationic lipid:25-55% sterol:0.5-15% PEG-modified lipid.
  • At least 80% of the uracil in the open reading frame have a chemical modification. In some embodiments, 100% of the uracil in the open reading frame have a chemical modification. In some embodiments, the chemical modification is in the 5- position of the uracil. In some embodiments, the chemical modification is a N1-methyl pseudouridine.
  • the invention is a composition of a ribonucleic acid (RNA) polynucleotide having an open reading frame encoding an isolated recombinant antibody or antigen-binding fragment thereof that specifically binds to influenza A hemagglutinin (HA), wherein the antibody has two or more of the following characteristics: (a) binds to influenza HA with a dissociation constant (K D ) of less than 5.5x10 -9 M as measured in a real-time bio- layer interferometer based biosensor (Octet HTX assay); (b) demonstrates neutralization of a single influenza A virus selected from H1N1 and H3N2, with an IC 50 of less than 15 ⁇ g/mL; (c) is a heavy chain variable domain antibody (VHH) that demonstrates protection, as measured by increased survival in an animal model of influenza virus infection when administered either before or after virus challenge in comparison to a multi-domain antibody; or (d) wherein the antibody or antigen-binding fragment thereof comprises
  • the recombinant antibody or antigen-binding fragment thereof binds to influenza HA with a K D of less than 1.3x10 -9 M. In other embodiments the recombinant antibody or antigen-binding fragment thereof demonstrates neutralization of a single influenza A virus selected from H1N1 and H3N2, with an IC 50 of less than 7 ⁇ g/mL.
  • RNA ribonucleic acid
  • HA hemagglutinin
  • the antibody or antigen-binding fragment thereof does not cross neutralize other HA subtypes.
  • the single HA subtype is H1or H3.
  • the invention is a composition of a ribonucleic acid (RNA) polynucleotide having an open reading frame encoding an isolated recombinant antibody or antigen-binding fragment thereof that specifically binds to an influenza A hemagglutinin (HA) subtype, wherein the antibody or antigen-binding fragment thereof does not bind a BNInfAb-1 (Broadly Neutralizing Influenzan antibody-1) epitope or a BNInfAb-2 (Broadly Neutralizing Influenzan antibody-2) epitope, and a pharmaceutically acceptable carrier or excipient.
  • RNA ribonucleic acid
  • the antibody or antigen-binding fragment thereof is a heavy chain variable domain antibody (VHH).
  • VHH heavy chain variable domain antibody
  • RNA ribonucleic acid
  • VHH neutralizing anti-influenza single heavy chain variable domain antibody
  • an isolated polypeptide of at least one single domain antibody that specifically binds to a hemagglutinin (HA) protein of an influenza virus and having a sequence represented by any of SEQ ID NOs: 1 to 18 or to a sequence having at least 70% sequence identity with a sequence of any of SEQ ID NOs: 1 to 18, wherein the polypeptide does not comprise a naturally occurring antibody is provided in other aspects of the invention.
  • the at least one single domain antibody is a VHH domain.
  • RNA ribonucleic acid
  • the disclosure provides methods of treating an influenza virus infection in a subject, comprising administering to a subject having an influenza virus infection a composition provided herein in a therapeutically effective amount to treat the influenza virus infection.
  • the influenza virus infection is a Type A influenza virus infection.
  • the Type A influenza virus infection is of subtype H3N2.
  • the Type A influenza virus infection is of subtype H1N1.
  • the subject is a mammal. In some embodiments, the mammal is a human. In some embodiments, the subject is a non-human primate.
  • Non-limiting examples of non-human primate subjects include macaques, marmosets, tamarins, spider monkeys, owl monkeys, vervet monkeys, squirrel monkeys, baboons, gorillas, chimpanzees, and orangutans.
  • Other exemplary subjects include domesticated animals such as dogs and cats; livestock such as horses, cattle, pigs, sheep, goats, and chickens; and other animals such as mice, rats, guinea pigs, and hamsters.
  • the composition is administered subcutaneously, intraocularly, intravitreally, parenterally, subcutaneously, intravenously, intracerebro-ventricularly, intramuscularly, intrathecally, orally, intraperitoneally, by oral or nasal inhalation, or by direct injection to one or more cells, tissues, or organs.
  • the disclosure provides methods of passively immunizing a mammalian subject against an influenza virus infection comprising administering to the subject a composition provided herein, wherein the subject is at risk of having or being exposed to an influenza virus infection.
  • influenza virus infection is a Type A influenza virus infection.
  • the Type A influenza virus infection is of subtype H3N2.
  • the Type A influenza virus infection is of subtype H1N1.
  • the mammalian subject is a human. In some embodiments, the mammalian subject is a non-human primate.
  • non-human primate subjects include macaques, marmosets, tamarins, spider monkeys, owl monkeys, vervet monkeys, squirrel monkeys, baboons, gorillas, chimpanzees, and orangutans.
  • Other exemplary subjects include domesticated animals such as dogs and cats; livestock such as horses, cattle, pigs, sheep, goats, and chickens; and other animals such as mice, rats, guinea pigs, and hamsters.
  • the composition is administered subcutaneously, intraocularly, intravitreally, parenterally, subcutaneously, intravenously, intracerebro-ventricularly, intramuscularly, intrathecally, orally, intraperitoneally, by oral or nasal inhalation, or by direct injection to one or more cells, tissues, or organs.
  • FIG.1 depicts the results of an exemplary phage ELISA showing nine anti-H3 and one anti-H1 HCAbs selected from a phage display library.
  • FIG.2 shows the results of an exemplary set of binding experiments, in which the binding of non-limiting HCAbs to HA of different strains of H3N2 and H1N1 was assessed.
  • FIG.3 shows exemplary results of epitope binning using non-limiting HCAbs.
  • FIG.4A depicts kinetic traces for non-limiting HCAbs binding to hemagglutinin.
  • FIG.4B reports the kinetic parameters obtained during the exemplary kinetic characterization depicted in FIG.4A.
  • FIG.5A depicts survival data when mice have been treated with mRNA encoding antibody of the invention and challenged with a lethal dose of Influenza H1N1.
  • FIG.5B depicts a graph showing changes in viral load in the same animals as treated in FIG.5A.
  • FIG.6 is a schematic of certain polynucleotide constructs of the present invention illustrating the modular design of the encoding polynucleotides.
  • FIGs.7A-7B depict graphs showing expression of AB 9114 in non-human primates at doses of 0.1 mg/kg (7A) or 0.3 mg/kg (7B).
  • FIGs.8A-8C depict graphs showing tolerability to expression of AB 9114 in non- human primates including body weight (8A), aspartate aminotransferase levels (8B) and alanine aminotransferase levels (8C).
  • FIGs.9A-9C depict graphs showing tolerability to expression of AB 9114 in non- human primates including complement activation (9A), IL6 levels (9B) and MCP-1 levels (9C).
  • FIGs.10A-10B depict bar graphs showing changes in expression levels of AB 9114 formulated in PEG stearate LNPs (10B) versus PEG-DMG LNPs (10A) in non-human primates.
  • FIG.11 is a graph depicting a polyclonal phage ELISA showing that EBOV binders and enriched with each round of panning.
  • FIG.12A-12B is a set of graphs showing multiple screens (12A) and a VHH phage screen yielding EBOV Zaire and Sudan strain cross-binders (12B) .
  • FIG.13 shows the most prevelant CDR3s from Round 1 of Screen 1, which were determined using Next Generation Sequencing (NGS). The sequences, from top to bottom, correspond to SEQ ID NOs: 146-155.
  • NGS Next Generation Sequencing
  • FIG.14A-14B show the sequences and Western blots of two soluble purified VHHs, ZSZ-01 (SEQ ID NO: 156) (14A) and ZSZ-02 (SEQ ID NO: 157) (14B).
  • FIG.15 is a graph depicting hEPO expression (ng/mL) at predose, 2 hours, 6 hours, 12 hours, 24 hours, 48 hours, and 72 hours following once weekly IV administration of hEPO mRNA–LNP formulations.
  • FIG.16 is a graph depicting levels of anti-PEG IgM (U/mL) following once weekly IV administration of the hEPO mRNA–LNP formulations.
  • FIG.17 shows in vitro biophysical characterization of IgG and scFV-FC. Single chains showed heterogeneity. A longer linker reduces dimer peaks.
  • FIG.18 shows that tolerance to conversion to scFV-FC format varied among three bnAbs.
  • FIG.19 shows antibody titers, neutralization data, meting temperature and aggregation properties for PGDM1400 scFv variants in comparison to IgG. Sequences at the bottom correspond from left to right to SEQ ID NOs: 158 and 159.
  • FIG.20 shows antibody titers, neutralization data, meting temperature and aggregation properties for PGT-121 scFv variants in comparison to IgG. Sequences at the bottom correspond from left to right to SEQ ID NOs: 158 and 159.
  • FIG.21 shows antibody titers, neutralization data, meting temperature and aggregation properties for 10-1074 scFv variants in comparison to IgG.
  • the sequence at the bottom corresponds to SEQ ID NO: 158.
  • FIG.22 shows IgG expression. DETAILED DESCRIPTION
  • RNA ribonucleic acid
  • One beneficial outcome is to cause intracellular translation of the nucleic acid and production of at least one encoded peptide or polypeptide of interest.
  • Antibodies also known as immunoglobulins, are glycoproteins produced by B cells. Using a unique and highly evolved system of recognintion, antibodies can recognize a target and tag a target epitope, foreign entity or invading microbe for attack by the immune system thereby neutralizing its effect. The production of antibodies is the main function of the humoral immune system. Antibodies are secreted by a plasma cell which is a type of white blood cell.
  • Antibodies occur in two physical forms, a soluble form that is secreted from the cell, and a membrane-bound form that is attached to the surface of a B cell and is referred to as the B cell receptor (BCR). Soluble antibodies are released into the blood and tissue fluids, as well as many secretions to continue to survey for invading microorganisms.
  • BCR B cell receptor
  • the majority of antibodies comprise two heavy chains and two light chains. There are several different types of antibody heavy chains, and several different kinds of antibodies, which are grouped into different isotypes based on which heavy chain they possess. Five different antibody isotypes isotypes (IgA, IgD, IgE, IgG and IgM) are known in mammals and trigger a different immune response for each different type of foreign object, epitope or microbe they encounter.
  • IgA, IgD, IgE, IgG and IgM Five different antibody isotypes isotypes (IgA, IgD, IgE, IgG and IgM) are known in mammals and trigger a different immune response for each different type of foreign object, epitope or microbe they encounter.
  • the immunoglobulins mediate a variety of these effector functions. These functions include fixation of complement, binding of phagocytic cells, lymphocytes, platelets, mast cells, and basophils which have immunoglobulin receptors. This binding can activate the cells to perform some function. Some antibodies or immunoglobulins also bind to receptors on placental trophoblasts, which results in transfer of the immunoglobulin across the placenta. As a result, the transferred maternal antibodies provide immunity to the fetus and newborn.
  • the invention is a ribonucleic acid (RNA) polynucleotide having an open reading frame encoding an antibody or single-chain variable fragment (scFv) (i.e., one or more antibodies or scFv fragments) having specificity for a human immunodeficiency virus (HIV) and a pharmaceutically acceptable carrier or excipient.
  • scFv single-chain variable fragment
  • the scFv comprises variable regions of heavy (VH) and light chains (VL) of immunoglobulins, connected with a linker peptide of about 4 to about 30 amino acids.
  • compositions including pharmaceutical compositions
  • methods for the design, preparation, manufacture and/or formulation of antibodies where at least one component of the antibody is encoded by a polynucleotide.
  • the present invention is directed, in part, to polynucleotides, specifically IVT polynucleotides, chimeric polynucleotides and/or circular polynucleotides encoding one or more antibodies and/or components thereof.
  • the polynucleotides are preferably modified in a manner as to avoid the deficiencies of or provide improvements over other antibody molecules of the art.
  • polynucleotide(s) and antibody compositions comprising at least one polynucleotide which have been designed to produce a therapeutic outcome and optionally improve one or more of the stability and/or clearance in tissues, receptor uptake and/or kinetics, cellular access, engagement with translational machinery, mRNA half-life, translation efficiency, protein production capacity, secretion efficiency (when applicable), accessibility to circulation, protein half-life and/or modulation of a cell’s status, antibody target affinity and/or specificity, reduction of antibody cross reactivity, increase of antibody purity, increase or alteration of antibody effector function and/or antibody activity.
  • the methods of the present invention are and can be utilized to engineer novel polynucleotides for the in vivo production of antibodies in such a manner as to provide improvements over standard antibody technology.
  • the polynucleotides are designed to produce one or more antibodies, or combinations of antibodies selected from the group consisting of IgA, IgG, IgM, IgE, and IgD.
  • An antibody is an immunoglobulin molecule capable of specific binding to a target, such as a carbohydrate, polynucleotide, lipid, polypeptide, through at least one antigen recognition site, located in the variable region of the immunoglobulin molecule.
  • a target such as a carbohydrate, polynucleotide, lipid, polypeptide
  • the antibodies described herein can be derived from murine, rat, human, or any other origin.
  • antibody encompasses not only intact (i.e., full-length) antibodies, but also antigen-binding fragments thereof (such as Fab, Fab', F(ab')2, Fv), single chain (scFv), mutants thereof, fusion proteins comprising an antibody portion, humanized antibodies, chimeric antibodies, diabodies, nanobodies, linear antibodies, single chain antibodies, multispecific antibodies (e.g., bispecific antibodies), single domain antibodies such as heavy-chain antibodies, and any other modified configuration of the immunoglobulin molecule that comprises an antigen recognition site of the required specificity, including glycosylation variants of antibodies, amino acid sequence variants of antibodies, and covalently modified antibodies.
  • An antibody includes an antibody of any class, such as IgD, IgE, IgG, IgA, or IgM (or sub-class thereof), and the antibody need not be of any particular class.
  • immunoglobulins can be assigned to different classes. There are five major classes of naturally-occurring immunoglobulins: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2.
  • the heavy-chain constant domains that correspond to the different classes of immunoglobulins are called alpha, delta, epsilon, gamma, and mu, respectively.
  • the subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known.
  • An antibody such as anti-viral antibody described herein may comprise a heavy chain variable region (V H ), a light chain variable region (V L ), or a combination thereof.
  • the antibody may further comprise an antibody constant region or a portion thereof (e.g., C H 1, C H 2, C H 3, or a combination thereof).
  • the heavy chain constant region can be of any suitable class as described herein and of any suitable origin, e.g., human, mouse, rat, or rabbit.
  • the heavy chain constant region is derived from a human IgG (a gamma heavy chain).
  • the light chain constant region can be a kappa chain or a lambda chain from a suitable origin.
  • Antibody heavy and light chain constant regions are well known in the art, e.g., those provided in the IMGT database (www.imgt.org) or at www.vbase2.org/vbstat.php., both of which are incorporated by reference herein.
  • the antibody as described herein may comprise a modified constant region.
  • it may comprise a modified constant region that is immunologically inert, e.g., does not trigger complement mediated lysis, or does not stimulate antibody- dependent cell mediated cytotoxicity (ADCC).
  • ADCC activity can be assessed using methods disclosed in U.S. Pat. No.5,500,362.
  • the constant region may be modieifed such that it has an elevated effort activity, for example, enhanced ADCC activity.
  • the constant region can be modified as described in Eur. J. Immunol. (1999) 29:2613-2624; PCT Application No. PCT/GB99/01441; and/or UK Patent Application No.9809951.8.
  • the heavy chain constant region used in the antibodies described herein may comprise mutations (e.g., amino acid residue substitutions) to enhance a desired characteristic of the antibody, for example, increasing the binding activity to the neonatal Fc receptor (FcRn) and thus the serum half-life of the antibodies. It was known that binding to FcRn is critical for maintaining antibody homeostasis and regulating the serum half-life of antibodies.
  • One or more mutations e.g., amino acid residue substitutions
  • the antibodies described herein specifically bind to the corresponding target antigen or an epitope thereof.
  • An antibody that“specifically binds” to an antigen or an epitope is a term well understood in the art. A molecule is said to exhibit “specific binding” if it reacts more frequently, more rapidly, with greater duration and/or with greater affinity with a particular target antigen than it does with alternative targets.
  • An antibody“specifically binds” to a target antigen or epitope if it binds with greater affinity, avidity, more readily, and/or with greater duration than it binds to other substances.
  • an antibody that specifically (or preferentially) binds to an antigen (e.g., HA of a specific influenza virus strain) or an antigenic epitope therein is an antibody that binds this target antigen with greater affinity, avidity, more readily, and/or with greater duration than it binds to other antigens or other epitopes in the same antigen.
  • an antibody that specifically binds to a first target antigen may or may not specifically or preferentially bind to a second target antigen.
  • “specific binding” or“preferential binding” does not necessarily require (although it can include) exclusive binding.
  • an antibody that“specifically binds” to a target antigen or an epitope thereof may not bind to other antigens or other epitopes in the same antigen.
  • an antibody as described herein has a suitable binding affinity for the target antigen or antigenic epitopes thereof.
  • binding affinity refers to the apparent association constant or K A .
  • the K A is the reciprocal of the dissociation constant (K D ).
  • the antibody described herein may have a binding affinity (K D ) of at least 10 -5 , 10 -6 , 10 -7 , 10 -8 , 10 -9 , 10 -10 M, or lower for the target antigen or antigenic epitope.
  • An increased binding affinity corresponds to a decreased K D .
  • Higher affinity binding of an antibody for a first antigen relative to a second antigen can be indicated by a higher K A (or a smaller numerical value K D ) for binding the first antigen than the K A (or numerical value K D ) for binding the second antigen.
  • the antibody has specificity for the first antigen (e.g., a first protein in a first conformation or mimic thereof) relative to the second antigen (e.g., the same first protein in a second conformation or mimic thereof; or a second protein).
  • the anti-influenza virus antibodies described herein have a higher binding affinity (a higher K A or smaller K D ) to a first influenza virus strain or the HA antigen of a first influenza virus strain as compared to the binding affinity to second influenza virus strain or the HA antigen of the second influenza virus strain.
  • Differences in binding affinity can be at least 1.5, 2, 3, 4, 5, 10, 15, 20, 37.5, 50, 70, 80, 91, 100, 500, 1000, 10,000 or 10 5 fold.
  • any of the anti-influenza virus antibodies may be further affinity matured to increase the binding affinity of the antibody to the target antigen or antigenic epitope thereof.
  • Binding affinity can be determined by a variety of methods including equilibrium dialysis, equilibrium binding, gel filtration, ELISA, surface plasmon resonance, or spectroscopy (e.g., using a fluorescence assay).
  • Exemplary conditions for evaluating binding affinity are in HBS-P buffer (10 mM HEPES pH7.4, 150 mM NaCl, 0.005% (v/v) Surfactant P20). These techniques can be used to measure the concentration of bound binding protein as a function of target protein concentration.
  • humanized antibodies refer to forms of non-human (e.g., murine) antibodies that are specific chimeric immunoglobulins, immunoglobulin chains, or antigen-binding fragments thereof that contain minimal sequence derived from non-human immunoglobulin.
  • humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a complementary determining region (CDR) of the recipient are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat, or rabbit having the desired specificity and/or affinity.
  • CDR complementary determining region
  • donor antibody such as mouse, rat, or rabbit having the desired specificity and/or affinity.
  • Fv framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues.
  • the humanized antibody may comprise residues that are found neither in the recipient antibody nor in the imported CDR or framework sequences, but are included to further refine and optimize antibody performance.
  • the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin sequence (e.g., a germline sequence or a consensus sequence).
  • the humanized antibody optimally may also comprise at least a portion of an immunoglobulin constant region or domain (Fc), typically that of a human immunoglobulin.
  • Antibodies may have Fc regions modified as described in WO 99/58572.
  • humanized antibodies have one or more CDRs (one, two, three, four, five, and/or six) which are altered with respect to the original antibody (termed one or more CDRs“derived from” one or more CDRs from the original antibody).
  • Humanized antibodies may also involve optimized antibodies derived from affinity maturation.
  • the antibody as described herein is a chimeric antibody, which can include a heavy constant region and optionally a light constant region from a human antibody.
  • Chimeric antibodies refer to antibodies having a variable region or part of variable region from a first species and a constant region from a second species.
  • the variable region of both light and heavy chains mimics the variable regions of antibodies derived from one species of mammals (e.g., a non-human mammal such as mouse, rabbit, and rat), while the constant portions are homologous to the sequences in antibodies derived from another mammal such as human.
  • amino acid modifications can be made in the variable region and/or the constant region.
  • the antibody described herein can be a single-domain antibody, which interacts with the target antigen via only one single variable domain such as a single heavy chain domain (as opposed to traditional antibodies, which interact with the target antigen via heavy chain and light chain variable domains).
  • a single-domain antibody can be a heavy-chain antibody (VHH) which contains only an antibody heavy chain and is devoid of light chain.
  • VHH heavy-chain antibody
  • a single-domain antibody may further comprise a contant region, for example, C H 1, C H 2, C H 3, C H 4, or a combination thereof.
  • the antibodies and antigen binding fragments thereof comprises a heavy chain variable region having an amino acid sequence sharing at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97% 98%, or 99% identity with any of the amino acid sequences provided herein (e.g. HA in Table 1).
  • the amino acid sequence of the antibody comprises an amino acid sequence provided herein.
  • the antibody binds the same epitope as an antibody comprising any of the VH chains known in the art and/or exemplified herein and/or competes against such an antibody from binding to the antigen.
  • an antibody may comprise the same heavy chain CDRs as those known in the art and/or exemplified herein.
  • An antibody having the same CDR (e.g., CDR3) as a reference antibody means that the two antibodies have the same amino acid sequence in that CDR region as determined by the same methodology (e.g., the Kabat definition, the Chothia definition, the AbM definition, or the contact definition).
  • an antibody described herein may comprise up to 5 (e.g., 4, 3, 2, or 1) amino acid residue variations in one or more of the CDR regions of one of the antibodies known in the art and/or exemplified herein and binds the same epitope of antigen with substantially similar affinity (e.g., having a K D value in the same order).
  • the amino acid residue variations are conservative amino acid residue substitutions.
  • a“conservative amino acid substitution” refers to an amino acid substitution that does not alter the relative charge or size characteristics of the protein in which the amino acid substitution is made.
  • Variants can be prepared according to methods for altering polypeptide sequence known to one of ordinary skill in the art such as are found in references which compile such methods, e.g., Molecular Cloning: A Laboratory Manual, J. Sambrook, et al., eds., Second Edition, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York, 1989, or Current Protocols in Molecular Biology, F.M. Ausubel, et al., eds., John Wiley & Sons, Inc., New York.
  • amino acids include substitutions made amongst amino acids within the following groups: (a) M, I, L, V; (b) F, Y, W; (c) K, R, H; (d) A, G; (e) S, T; (f) Q, N; and (g) E, D.
  • the antibody may be a germlined variant of any of the exemplary antibodies disclosed herein.
  • a germlined variant contains one or more mutations in the framework regions as relative to its parent antibody towards the corresponding germline sequence.
  • the heavy or light chain variable region sequence of the parent antibody or a portion thereof e.g., a framework sequence
  • an antibody germline sequence database e.g., www.bioinfo.org.uk/abs/
  • the antibody is a single chain antibody, which may comprise only one variable region (e.g., V H ) or comprise both a V H and a V L .
  • V H variable region
  • V L variable region
  • Such an antibody can be encoded by a single RNA molecule.
  • the antibody described herein is a multi-chain antibody comprising an independent heavy chain and an independent light chain.
  • Such a multi-chain antibody may be encoded by a single ribonucleic acid (RNA) molecule, which can be a bicistronic molecule encoding two separate polypeptide chains.
  • RNA ribonucleic acid
  • Such an RNA molecule may contain a signal sequence between the two coding sequences such that two separate polypeptides would be produced in the translation process.
  • the RNA molecule may include a sequence coding for a cleavage site (e.g., a protease cleavage site) inbetween the heavy and light chains such that it produces a single precursor polypeptide, which can be processed via cleavage at the cleavage site to produce the two separate heavy and light chains.
  • a cleavage site e.g., a protease cleavage site
  • the heavy and light antibody chains may be encoded by two separate RNA molecules.
  • RNA (e.g., mRNA) treatments of the present disclosure comprise one or more polynucleotides, e.g., polynucleotide constructs, which encode one or more antibodies and antigen binding fragments thereof.
  • polynucleotides of the disclosure e.g., antibody-encoding RNA polynucleotides, may include at least one chemical
  • polynucleotides of the disclosure e.g., antibody- encoding RNA polynucleotides, may be fully modified (e.g., chemically modified) with respect to one or more nucleobases.
  • compositions comprising RNA polynucleotides encoding one or more antibodies and antigen binding fragments thereof.
  • the antibodies and antigen binding fragments thereof encoded by an RNA polynucleotide of the present application comprises a heavy chain antibody comprising a variable (VH) domain.
  • the antibodies and antigen binding fragments thereof encoded by an RNA polynucleotide of the present application comprises a fragment crystallizable (Fc) region.
  • the Fc region is the tail region of an antibodies and antigen binding fragments thereof which contains constant domains (e.g., CH 2 and CH 3 ); the other region of the antibodies and antigen binding fragments thereof being the Fab region which contains a variable domain (e.g., VH) and a constant domain (e.g., CH 1 ), the former of which defines binding specificity.
  • antibodies can comprise a VH domain.
  • the VH domain further comprises one or more constant domains (e.g., CH 2 and/or CH 3 ) of an Fc region and/or one or more constant domains (e.g., CH 1 ) of a Fab region.
  • each of the one or more constant domains e.g., CH 1 , CH 2 , and/or CH 3
  • the constant domain comprises 99% or less, 98% or less, 97% or less, 96% or less, 95% or less, 90% or less, 80% or less, 70% or less, 60% or less, 50% or less, 40% or less, 30% or less, 20% or less, or 10% or less of the corresponding full sequence.
  • polynucleotides, constructs, and/or compositions of the present invention are useful in targeting or binding to polypeptides or proteins.
  • polypeptides or proteins In some embodiments
  • polynucleotides encode one or more antibodies or fragments thereof which bind to an infective agent such as a bacteria, virus or biomolecules thereof, cell surface molecules or cancer antigens.
  • infectious agents which may be targeted or bound by the peptides or proteins encoded by the polynucleotides of the present invention include both bacteria and viruses.
  • adenovirus chikungunya, Herpes simplex, type 1; Herpes simplex, type 2; Varicella-zoster virus; Epstein-barr virus; Human cytomegalovirus; Human herpesvirus, type 8; Human papillomavirus; BK virus; JC virus; Smallpox; Hepatitis B virus; Human bocavirus; Parvovirus B19; Human astrovirus;
  • Norwalk virus coxsackievirus; hepatitis A virus; poliovirus; rhinovirus; Severe acute respiratory syndrome virus; Hepatitis C virus; yellow fever virus; dengue virus; West Nile virus; Rubella virus; Hepatitis E virus; Human immunodeficiency virus (HIV); Influenza virus; Guanarito virus; Junin virus; Lassa virus; Machupo virus; Sabiá virus; Crimean- Congo hemorrhagic fever virus; Ebola virus; Marburg virus; Measles virus; Mumps virus; Parainfluenza virus; Respiratory syncytial virus; Human metapneumovirus; Hendra virus; Nipah virus; Rabies virus; Hepatitis D; Rotavirus; Orbivirus; Coltivirus; or Banna virus.
  • pathogenic bacteria examples include, but are not limited to, Acinetobacter baumannii, Bordetella pertussis, Borrelia burgdorferi, Brucella abortus, Brucella canis, Brucella melitensis, Brucella suis, Campylobacter jejuni, Chlamydia pneumoniae, Chlamydia trachomatis, Chlamydophila psittaci, Clostridium botulinum, Clostridium difficile,
  • Clostridium perfringens Clostridium tetani, Corynebacterium diphtheriae, Enterococcus faecalis, Enterococcus faecium, Escherichia coli, Francisella tularensis, Haemophilus influenzae, Helicobacter pylori, Legionella pneumophila, Leptospira interrogans, Listeria monocytogenes, Mycobacterium leprae, Mycobacterium tuberculosis, Mycobacterium ulcerans, Mycoplasma pneumoniae, Neisseria gonorrhoeae, Neisseria meningitidis,
  • Pseudomonas aeruginosa Rickettsia rickettsii, Salmonella typhi, Salmonella typhimurium, Shigella sonnei, Staphylococcus aureus, Staphylococcus epidermidis, Staphylococcus saprophyticus, Streptococcus agalactiae, Streptococcus pneumoniae, Streptococcus pyogenes, Treponema pallidum, Vibrio cholerae, Yersinia pestis, Yersinia enterocolitica, and/or Yersinia pseudotuberculosis.
  • compositions and methods are useful for the treatment of chikungunya virus infection.
  • Chikungunya virus is a mosquito-borne virus belonging to the Alphavirus genus of the Togaviridae family.
  • Some embodiments of the present disclosure provide RNA encoding an anti-Chikungunya virus (CHIKV) antibody, which comprises one or more RNA polynucleotides having an open reading frame encoding at least one anti- CHIKV antibody, specific for a CHIKV antigenic polypeptide.
  • the RNA polynucleotide encodes two or more anti- CHIKV antibodies.
  • the anti-CHIKV antibody is specific for an antigenic polypeptide which is a CHIKV structural protein or an antigenic fragment thereof.
  • a CHIKV structural protein may be an envelope protein (E), a 6K protein, or a capsid (C) protein.
  • the CHIKV structural protein is an envelope protein selected from E1, E2, and E3.
  • the CHIKV structural protein is E1 or E2.
  • the CHIKV structural protein is a capsid protein.
  • the antigenic polypeptide is a fragment or epitope of a CHIKV structural protein.
  • the antibody is an antibody that binds to an epitope on the E2 protein of CHIKV. In some embodiments, the antibody binds to E2-A162, or an epitope formed by residues E2-G95, E2-A162, E2-A164, E2-E165, E2-E166 and/or E2-I167, or any combination thereof. In some embodiments, the antibody binds to an epitope formed by residues E2-Y69, E2-F84, E2-V113, E2-G114, E2-T116, and/or E2-D117, or any
  • the epitope comprises E2-G95.
  • the antibody is an antibody that binds to at least one of:
  • the antibody binds to Subunit I-E2-E24 and Subunit I-E2-I121 and at least one of: Subunit II-E2-G55, Subunit II-E2-W64, Subunit II-E2-K66, Subunit II-E2-R80.
  • the antibody binds to at least two of Subunit II-E2-G55, Subunit II-E2-W64, Subunit II-E2-K66, Subunit II-E2- R80.
  • the antibody binds to at least three of Subunit II-E2-G55, Subunit II-E2-W64, Subunit II-E2-K66, Subunit II-E2-R80. In some embodiments, the antibody binds to at least three of Subunit II-E2-G55, Subunit II-E2-W64, Subunit II-E2- K66, and Subunit II-E2-R80. In some embodiments, the antibody binds to Subunit II-E2- G55, Subunit II-E2-W64, Subunit II-E2-K66, and Subunit II-E2-R80.
  • the antibody binds to the membrane distal region of a CHIKV E1/E2 trimer. In some embodiments, the antibody binds to the exterior face of the E1/E2 heterocomplex.
  • the exterior face refers to the portion of the E1/E2 heterocomplex that is exposed when the E1/E2 hetero-protein is in its native form on the virion surface, such as in its trimeric form.
  • compositions and methods are useful for the treatment of influenza virus infection.
  • Influenza hemagglutinin (HA) is a glycoprotein found on the surface of influenza viruses. HA is responsible for binding the virus to cells with sialic acid on the membranes, such as cells in the upper respiratory tract or erythrocytes. HA is also responsible for the fusion of the viral envelope with the endosome membrane. As it is the major surface protein of the type A influenza virus and its function essential to the entry process, HA neutralization would provide an effective option for targeted therapeutics.
  • RNA e.g., mRNA
  • antibodies for example, antibodies and antigen binding fragments thereof including heavy-chain-only antibodies (HCAbs), that bind influenza HA.
  • Compositions and methods of treating influenza virus infection, as provided herein, may be used to induce a balanced immune response, comprising both cellular and humoral immunity.
  • Influenza HA has at least eighteen different HA subtypes, classified as subtypes H1 through H18.
  • the first three HA subtypes, H1, H2, and H3, are found in human influenza viruses.
  • the antibodies and antigen binding fragments thereof provided in the present disclosure can be useful in binding and/or neutralizing one or more of H1, H2, and H3.
  • compositions and methods provided herein can be useful in treating influenza subtypes characterized by H1, H2, and/or H3.
  • influenza strains of subtypes H1N1 and/or H3N2 can be targeted using compositions and methods described herein.
  • influenza strains of subtype H1N1 include A/swine/Iowa/15/1930, A/Solomon Island/3/2006, A/South Carolina/1/1918, A/New Caledonia/20/1999, A/Puerto Rico/08/1934, A/Ohio/1983, A/WSN/1933, or A/California/04/2009.
  • influenza strains of subtype H3N2 include A/Moscow/10/1999, A/Sydney/5/1997, A/Hong
  • the VH domain comprises an amino acid sequence consisting of or essentially consisting of an amino acid sequence of Table 1 (SEQ ID NO: 1-13).
  • Table 1 Amino acid VH sequences of HCAbs
  • the antibodies and antigen binding fragments thereof encoded by an RNA polynucleotide of the present application comprises an amino acid sequence of the consensus sequence“CON-1” in Table 1 (SEQ ID NO: 11).
  • CON-1 represents a consensus sequence for SEQ ID NOs: 1-10, where“X” is any amino acid and“Z” is optionally any amino acid or no amino acid at that position.
  • the antibodies and antigen binding fragments thereof encoded by an RNA polynucleotide of the present application comprises an amino acid sequence of the consensus sequence“CON-2” in Table 1 (SEQ ID NO: 12).
  • CON-2 represents a consensus sequence for SEQ ID NOs: 1-2 and 4-8, where“X” is any amino acid and“Z” is optionally any amino acid or no amino acid at that position.
  • the consensus sequence provides a framework of VHH antibodies for an influenza VHH.
  • the antibodies and antigen binding fragments thereof encoded by an RNA polynucleotide of the present application comprises an amino acid sequence of the consensus sequence“CON-3” in Table 1 (SEQ ID NO: 13).
  • CON-3 represents a consensus sequence for SEQ ID NOs: 1 and 4-5, where“X” is any amino acid and“Z” is optionally any amino acid or no amino acid at that position.
  • polypeptide comrpsing any of the following sequences or nucelci acids encoding the peptides is provided.
  • VQLX 1 EX 2 GGG (SEQ ID NO: 56), wherein X 1 is L or V and X 2 is S or T
  • GGSLRLSCAASGF (SEQ ID NO: 57)
  • MNSLRAEDTAVYX 3 CA (SEQ ID NO: 59); wherein X 3 is Y or S
  • compositions may be designed as a single therapeutic that treats all strains of seasonal flu and pandemic flu where the polynucleotides encode IgG against hemaglutinins associated with emerging strains with pandemic potential.
  • polynucleotides or constructs and their associated compositions may be designed to produce a commercially available antibody, a variant or a portion thereof in vivo.
  • the polynucleotide encodes CR6261.
  • CR6261 is a monoclonal antibody that binds to a broad range of the influenza virus including the 1918 "Spanish flu” (SC1918/H1) and to a virus of the H5N1 class of avian influenza that jumped from chickens to a human in Vietnam in 2004 (Viet04/H5).
  • CR6261 is reported to neutralize numerous strains from multiple subtypes.
  • CR6261 recognizes a highly conserved helical region in the membrane-proximal stem of hemagglutinin, the predominant protein on the surface of the influenza virus.
  • the polynucleotides may encode a CR6261 single chain Fv (scFv) antibody fragments fused to the human IgG Fc moiety (scFv-Fc).
  • scFv single chain Fv
  • scFv-Fc human IgG Fc moiety
  • CR6261 variable light and/or heavy chain domains are linked via a suitable linker known in the art and/or described herein, and the CR6261 variable light and/or heavy chain domains are further connected through a linker to a Fc moiety.
  • the CR6261 variable light and/or heavy chain domains within a scFv-Fc construct are variants of the native CR6261 variable light and/or heavy chain domains.
  • the native CR6261 variable light and/or heavy chain domains may comprise one or more single amino acid substitutions in order to optimize the CR6261 variable light and/or heavy chain domains for the scFv format and/or to ensure optimal stability and antigen binding.
  • the CR6261 variable light and/or heavy chain domains in the scFv-Fc construct are arranged so that the variable heavy chain is 5’ to the variable light chain (V H -V L ). In other embodiment, the CR6261 variable light and/or heavy chain domains in the scFv-Fc construct are arranged so that the variable light chain is 5’ to the variable heavy chain (V L -V H ).
  • the CR6261 scFv constructs comprise the Dall’Acqua half-life extending“YTE” Fc substitutions M265Y, S267T, and T269E (using the Kabat numbering).
  • the CR6261 variable light domain may comprise at least one substitution.
  • the CR6261 variable light domain may comprise one, two, three, four, five or more than five substitutions.
  • the CR6261 variable light domain may comprise the two substitutions A13S (where alanine (A) at position 13 is substituted with serine (S)) and L111T (where leucine (L) at position 111 is substituted with
  • the CR6261 variable light may comprise the four substitutions A13S (where alanine (A) at position 13 is substituted with serine (S)), L40K (wherein leucine (L) at position 40 is substituted with lysine (K)), G82E (where glycine (G) at position 82 is substituted with glutamic acid (E)) and L111T (where leucine (L) at position 111 is substituted with threonine (T)).
  • the CR6261 variable heavy domain may comprise at least one substitution.
  • the CR6261 variable heavy domain may comprise one, two, three, four, five or more than five substitutions.
  • the CR6261 variable heavy domain may comprise the two substitutions V11T (where valine (V) at position 11 is substituted with threonine (T)) and M93T (where methionine (M) at position 93 is substituted with threonine (T)).
  • the CR6261 variable heavy may comprise the three substitutions E6Q (where glutamic acid (E) at position 6 is substituted with glutamine (Q)), V11T (where valine (V) at position 11 is substituted with threonine (T) and M93T (where methionine (M) at position 93 is substituted with threonine (T)).
  • the polynucleotides may encode CR9114. In some embodiments, the polynucleotides may encode CR9114.
  • the polynucleotides may encode a CR9114 single chain Fv (scFv) antibody fragments fused to the human IgG Fc moiety (scFv-Fc).
  • CR9114 variable light and/or heavy chain domains are linked via a suitable linker known in the art and/or described herein, and the CR9114 variable light and/or heavy chain domains are further connected through a linker to a Fc moiety.
  • the CR9114 variable light and/or heavy chain domains within a scFv-Fc construct are variants of the native CR9114 variable light and/or heavy chain domains.
  • the native CR9114 variable light and/or heavy chain domains may comprise one or more single amino acid substitutions in order to optimize the CR9114 variable light and/or heavy chain domains for the scFv format and/or to ensure optimal stability and antigen binding.
  • the CR9114 variable light and/or heavy chain domains in the scFv-Fc construct are arranged so that the variable heavy chain is 5’ to the variable light chain (V H -V L ). In other embodiment, the CR9114 variable light and/or heavy chain domains in the scFv-Fc construct are arranged so that the variable light chain is 5’ to the variable heavy chain (V L -V H ).
  • the CR9114 variable light domain may comprise at least one substitution.
  • the CR9114 variable light domain may comprise one, two, three, four, five, six, seven or more than seven substitutions.
  • the CR9114 variable light domain may comprise the four substitutions S1Q (where serine (S) at position 1 is substituted with glutamine (Q)), Y2S (where tyrosine (Y) at position 2 is substituted with serine (S)), V3A (where valine (V) at position 3 is substituted with alanine (A)) and A9S (where alanine (A) at position 9 is substituted with serine (S)).
  • the CR9114 variable light may comprise the seven substitutions S1Q (where serine (S) at position 1 is substituted with glutamine (Q)), Y2S (where tyrosine (Y) at position 2 is substituted with serine (S)), V3A (where valine (V) at position 3 is substituted with alanine (A)), A9S (where alanine (A) at position 9 is substituted with serine (S)), F40K (where phenylalanine (F) at position 40 is substituted with lysine (K)), V58G (where valine (V) at position 58 is substituted with glycine (G)) and L110T (where leucine (L) at position 110 is substituted with threonine (T)).
  • the CR9114 variable heavy domain may comprise at least one substitution.
  • the CR9114 variable heavy domain may comprise one, two, three, four, five or more than five substitutions.
  • the CR9114 variable heavy domain may comprise the three substitutions V11T (where valine (V) at position 11 is substituted with threonine (T)), T87R (where threonine (T) at position 87 is substituted with arginine (R)) and V93T (valine (V) at position 93 is substituted with threonine (T)).
  • the CR9914 variable heavy domain may comprise the five substitutions V11T (where valine (V) at position 11 is substituted with threonine (T)), D46E (where aspartic acid (D) at position 46 is substituted with glutamic acid (E)), N84S (where asparagine (N) at position 84 is substituted with serine (S)), T87R (where threonine (T) at position 87 is substituted with arginine (R)) and V93T (valine (V) at position 93 is substituted with threonine (T)).
  • the polynucleotides of the invention may be used to protect an infant against infection or disease, including but not limited to diseases caused by respiratory syncytial virus (RSV) alone or in combination with B. pertussis, by administering the polynucleotides to a pregnant female with a gestational infant.
  • RSV respiratory syncytial virus
  • the antibody encoded by the polynucleotide of interest may be transferred via the placenta to the gestational infant, protecting the infant against infection or disease.
  • the polynucleotides of the invention may be administered alone or in combination with an immunogenic composition as described in WO2014/024024 and WO2014/024026, the contents of each which is herein incorporated by reference in its entirety.
  • Hepatitis C is a contagious liver disease that results from infection with the hepatitis C virus, one of the most common viral liver infections with approximately 150 million people have chronic infections with risk of liver cirrhosis and/or liver cancer, 3-4 million people infected yearly and about 350,000 deaths every year. It can range in severity from a mild illness lasting a few weeks to a serious, lifelong illness. HCV infection and associated liver cirrhosis is the most common indication for orthotopic liver transplantation among adults and HCV infection remains a problem after transplantation and recurrent hepatic infection is the leading cause of graft failure.
  • Chronic hepatitis C is characterized by a high turnover of infected cells and continuous de novo infection of target cells. Due to the vital role of de novo infection in maintenance of HCV infection, blocking of de novo infection is a potential target for antiviral therapy.
  • the viral envelope glycoproteins, E1 and E2 are the major components of the HCV particle and hence play a pivotal role in the entry process and hypervariable region 1 (HVR1), consisting of the first 27 amino acids of E2 (aa 384–410), is a major target for neutralizing antibodies.
  • Another approach that is currently promoted for the treatment and prevention of HCV infection/re-infection is blocking a pathway preventing the immune system from recognizing and fighting cancer cells and pathogens.
  • Current treatment involves a combination of IFN- ⁇ and ribavirin.
  • the polynucleotides of the invention may be used in the treatment and/or prevention of hepatitis C virus (HCV) infection.
  • HCV hepatitis C virus
  • hepatitis C virus or “HCV” means a viral disease that can lead to swelling of the liver.
  • the polynucleotides of the invention may encode at least one antibody, fragment or variant thereof which may be used for prognosing, diagnosing, and/or treating of HCV in a subject.
  • the polynucleotides of the invention may be used to protect a subject from or inhibit HCV-mediated morbidity or mortality in a subject.
  • the polynucleotides of the current invention may be used in combination with ribavirin, IFN- ⁇ and/or pegylated (peg) IFN- ⁇ to treat and/or prevent HCV.
  • ribavirin IFN- ⁇
  • peg pegylated
  • Rabies is a wide spread viral disease that is transmitted from animals to humans. Around 60,000 people die annually from rabies, and the disease threatens over 3 billion people in rural areas of Asia and Africa where human vaccines and immunoglobulin are not readily available or accessible. Rabies is an RNA virus that belongs to the order
  • RIG rabies virus immunoglobulin
  • HRIG Human RIG
  • ERIG Equine RIG
  • the polynucleotides of the invention may be used in the treatment or prevention of rabies virus infection.
  • rabies virus is a virus normally spread to people from the saliva of infected animals and infects nerve cells.
  • the polynucleotides of the invention may encode at least one antibody, fragment or variant thereof which may be used for the treatment or prevention of rabies.
  • the human immunodeficiency virus is a lentivirus that causes the acquired immunodeficiency syndrome (AIDS). HIV infects cells of the human immune system such as helper cells expressing the CD4 receptor on their surface, macrophages, and dendritic cells, compromising cell-mediated immunity and allowing opportunistic infections and cancers to thrive.
  • AIDS acquired immunodeficiency syndrome
  • the polynucleotides of the invention may be used in the treatment or prevention of human immunodeficiency virus (HIV).
  • HSV human immunodeficiency virus
  • immunodeficiency virus or“HIV” means a variable retrovirus that invades and inactivates helper T cells of the immune system and is a cause of AIDS and AIDS-related complex.
  • the polynucleotides of the invention may encode at least one antibody, fragment or variant thereof which may be used for the treatment or prevention of HIV.
  • the polynucleotides may encode at least one neutralizing HIV antibody, which may target the HIV-1 viral spike.
  • mRNA encoding antibodies can be used in the treatment of HIV infection. Although these complex proteins are difficult to engineer as mRNA therapeutics, optimal compositions, sequences and combinations having a broad spectrum of neutralization activities when expressed in vivo from mRNA have been developed according to the invention. The parameters involved in the creation and
  • HIV antibodies have been developed for the prophylactic treatment of HIV infection.
  • the antibodies are particularly useful in reducing the risk of HIV transmission for vulnerable populations and reducing the burden of disease globally.
  • These antibodies are delivered as proteins.
  • nucleic acid delivery of these antibodies would be desirable a number of challenges exist in the development of mRNA encoding antibodies. For instance, challenges exist in the expression of complex proteins such as antibodies and scFv. A number of factors can effect activity of scFv.
  • optimal combinations of antibodies delivered as proteins don’t necessarily translate to optimal activity when the antibodies are delivered as mRNA.
  • HIV antibodies include PGT12110-1074, PGDM1400, and 3BNC117 NIH 45-46 N6 (Scharf et al. Cell Reports 20147, 785-795). While combinations of these protein antibodies are useful as therapeutics, simply expressing the proteins as mRNAs and co-delivering the mRNAs does not result in an adequate therapeutic composition. For instance, co-expression of these proteins from mRNA as combinations of IgGs produces non-functional pairings. A therapeutically effective combination of these antibodies involves the optimal convergence of the spectrum of neutralizing activity across many HIV strains/clades, optimized level of expression, and protein stability contributing to half-life in vivo.
  • scFv variants designed herein have significnatly higher expression and/or neutralization and/or thermostabiliity properties in comparison to scFv designed according to traditional rules/expections for designing scFv.
  • the invention is a composition of mRNA encoding a combination of HIV antibodies.
  • the composition may include one or more intact intact antibodies such as IgGs, one or more mono- and bispecific scFvFcs, and IgG/sc “hybrids” and one or more antigen binding fragments.
  • the composition may be any combination of mRNA/ lipid nanoparticle (LNP) carrier.
  • LNP lipid nanoparticle
  • multiple antibodies may be included on a single mRNA or separate mRNAs.
  • each peptide chain of antibody is encoded by an individual mRNA.
  • each mRNA may be coformulated individually in an LNP.
  • one or more or all of the mRNAs may be formulated in an LNP.
  • the HIV compositions of the invention may be designed based on known HIV antibodies or newly developed HIV antibodies.
  • the mRNA encodes known therapeutically effective HIV antibodies.
  • the mRNA encodes variants of known HIV antibodies.
  • the mRNA does not encode full intact PGT12110-1074, PGDM1400, and 3BNC117 NIH 45-46 N6.
  • the mRNA does encode variants of one or more of PGT12110-1074, PGDM1400, and 3BNC117 NIH 45-46 N6, including antigen binding fragments thereof and scFv.
  • the composition comprises mRNA encoding a combination of an intact IgG and two scFvFcs.
  • This product in some embodiments includes four mRNAs total, which produce an IgG (two mRNAs encoding light chain and heavy chain) and two scFv-Fcs (each one mRNA).
  • the methods for developing the mRNA encoding the HIV antibodies of the invention involves selecting broadly neutralizing antibodies. Key antigen binding components of such antibodies may be identified and used to build variants.
  • the composition in some embodiments comprises a combination of mRNA encoding at least one antibody that binds CD4 binding site of gp120, at least one antibody that blocks the V1/V2 site of gp120, and/or at least one antibody that blocks the V3 site of gp120.
  • the antibodies may be any combination of intact antibodies and scFv.
  • a composition of antibodies that bind to and block these three regions of the HIV virus have been shown to be highly effective at preventing HIV transmission and are thus useful therapeutically in subjects at risk of infection with HIV as well as subjects infected with HIV, in preventing further infection.
  • compositions of three or more ribonucleic acid (RNA) polynucleotides each having an open reading frame (ORF) encoding an HIV antibody or scFv are encompassed within the invention.
  • RNA ribonucleic acid
  • ORF open reading frame
  • compositions include therapeutically effective antibody combinations. Such combinations may include for instance, a combination of an IgG and two or more scFv.
  • the intact IgG antibody may be one that blocks the CD4 binding site of gp120, e.g., a 3BNC117, NIH45-46, or an N6 IgG.
  • the scFv may be an scFv antibody that blocks the V1/V2 site of gp120, e.g., PDGM_1400 (such as scFV-FC_var7) and/or an scFv that blocks the V3 site of gp120, e.g., PGT_121 or 10-1074.
  • a composition of the invention comprises a nucleotide sequence encoding an the intact IgG antibody comprising the CDRs of N6 IgG, an scFv antibody comprising the CDRs of PDGM1400 and an scFv comprising the CDRs of PGT-121.
  • the antibodies described herein may include modified or variant variable domains from the sequences disclosed herein.
  • the variant or modification may be, for instance, amino acid substitutions, compared to the sequences disclosed herein .
  • Modifications can also include amino acid deletions.
  • one or two amino acids may be deleted from a variant VH and/or VL domain.
  • the deleted amino acids typically may be from the carboxyl or amino terminal ends of the VH and/or VL domains.
  • variable domain of the antibodies described herein comprises three
  • a VH domain may comprise a set of three heavy chain CDRs, HCDR1, HCDR2, and HCDR3.
  • a VL domain may comprise a set of three light chain CDRs, LCDR1, LCDR2, and LCDR3.
  • a set of HCDRs disclosed herein can be provided in a VH domain that is used in combination with a VL domain.
  • a VH domain may be provided with a set of HCDRs as disclosed herein, and if such a VH domain is paired with a VL domain, then the VL domain may be provided with a set of LCDRs disclosed herein.
  • Exemplary CDRs useful in the antibodies are included herein as SEQ ID NOs.200-217).
  • Exemplary PGT-121 CDRs and PGDM-1400 CDRs identified based on two different labeling scheme, Kabat and Chothia are presented below.
  • the HIV antibody is a single chain Fv (scFv).
  • scFv single chain Fv
  • the term“single-chain” refers to a molecule comprising amino acid monomers linearly linked by peptide bonds.
  • An scFv has a variable domain of light chain (VL) connected from its C- terminus to the N-terminal end of a variable domain of heavy chain (VH) by a polypeptide chain.
  • VL variable domain of light chain
  • VH variable domain of heavy chain
  • the scFv comprises of polypeptide chain where in the C-terminal end of the VH is connected to the N-terminal end of VL by a polypeptide chain.
  • the scFv constructs may be oriented in a variety of ways.
  • the order to VH and VL in the construct may vary and alter the expression and/or activity of the scFv.
  • the scFv constructs are oriented, from N to C terminus, VL-linker-VH-linker- CH2-CH3. It was discovered that this orientation may produce optimal results.
  • the scFv fragments which may be attached to one another and to Fc domains by flexible linkers.
  • the variable heavy and variable light chains are covalently attached using flexible linkers.
  • flexible linkers such as those containing glycine and serine are used.
  • the scFV-FC synthesis from a typical antibody format involved the addition of linkers.
  • an ideal linker is considered to be (G 4 S) 3 .
  • longer linkers were more effective in producing highly neutralizing scFv. For instance, it was found that increasing the VL-VH linker length could reduce strain and oligomerization.
  • the (G4S) 3 linker is a shorter linker, which tends to produce more strain.
  • the (G4S) 4 linker (also referred to as Linker20) is a longer linker with less strain. Increasing the length of the linker between the VL and VH domains reduced the strain, causing opening of the VL-VH interface and domain swapping, in particular in the PGT-121. It is desirable for the linker to have greater than 15 amino acids in length. In some embodiments it is desirable to have a linker of 16-30 amino acids. In other embodiments the linker has 16—25, 16-20, 18-30, 18-25, 18-20, 19-30, 19-25, 19-20, 20-30, 20-25, or 20-22 amino acids. In some embodiments the linker is (G 4 S) 4 .
  • FC region is a wild type of FC region. In other embodiments it is a variant of wild type.
  • a wild t pe constant re ion is a wild t pe I G1 constant re ion (e. .,
  • the antibody constructs may be enhanced by modifying glycan stability to enhance the effectivity of the antibody. For instance, as shown in the Examples presented herein adding glycan stability to antibodies such as scFv i.e., PGT-121 variants, improved the activity of the antibody.
  • compositions and combinations of compositions described herein are useful for treating HIV infection, typically by blocking entry of HIV into host cells, and thus blocking infection.
  • Latent reservoirs of HIV-1 infected cells are difficult to treat with traditional HIV medicine.
  • the ability to block entry of HIV provides a significant advance in the treatment and prevention of HIV-1 infection.
  • a latent reservoir is established within days of initial infection and persists for the lifetime of the individual.
  • the combinations of antibodies disclosed herein can be used in preventing the establishment of the reservoir.
  • the antibodies are administered as a bolus IV injection or bolus. This form of delivery can produce high levels of expressed antibody.
  • Staphylococcus is a genus of Gram-positive bacteria. Staphylococci, gram positive bacteria including coagulase-negative staphylococci (CONS) and Staphylococcus aureus, are responsible for between 56 and >75% of hospital-acquired, late-onset neonatal sepsis.
  • CONS coagulase-negative staphylococci
  • Staphylococcus aureus are responsible for between 56 and >75% of hospital-acquired, late-onset neonatal sepsis.
  • the polynucleotides of the invention may be used in the treatment or prevention Staphylococcus infection.
  • Staphylococcus means a bacteria that can cause sepsis.
  • the polynucleotides of the invention may encode at least one antibody, fragment or variant thereof which may be used for the treatment or prevention of sepsis caused by Staphylococcus.
  • Anthrax is a serious infectious disease caused by gram-positive bacteria known as Bacillus anthracis (B. anthracis). Although rare, human can get infected with anthrax if they come in contact with infected animals or contaminated animal products. Bacillus anthracis has also long been considered a potential biological warfare agent.
  • Anthrax toxin is composed of a cell-binding protein, known as protective antigen (PA), and two enzyme components, called edema factor (EF) and lethal factor (LF). These three protein components act together to impart their physiological effects.
  • PA is a protein component of the toxins produced by the bacterium. It initiates the activity of the toxins by attaching to cells in the infected person, and then facilitates the entry of additional destructive factors– LF and EF into the cells.
  • PA comprises a protein having a weight of about 83 kD (PA83) that is cleaved into a protein having a weight of about 63 kD (PA63).
  • the polynucleotides of the invention may be used in the treatment or prevention of Bacillus anthracis (B. anthracis) infection and anthrax.
  • Bacillus anthracis B. anthracis
  • Bacillus anthracis or“B. anthracis” is the bacterium that causes anthrax.
  • the polynucleotides of the invention may encode at least one antibody, fragment or variant thereof which may be used for the treatment or prevention of anthrax.
  • Shiga toxin (Stx)-producing Escherichia coli (STEC) causes hemorrhagic colitis and hemolytic-uremic syndrome (HUS).
  • HUS hemolytic-uremic syndrome
  • Diarrhea-associated HUS is a common cause of acute renal failure and up to 50% of patients with HUS develop some degree of renal impairment.
  • the Shiga toxins produced by E. coli are majorly Stx1 and Stx2. (Thorpe, Clinical Infectious Disease, vol.38(9), 1298-1303 (2004), the contents of which are incorporated herein by reference in their entirety).
  • the polynucleotides of the invention may encode any antibody that targets a Shiga toxin, including, but not limited to the shigamabs antibody, for the prevention or treatment of STEC and HUS.
  • the polynucleotides may be used in combination with antibiotic therapies in the prevention and/or treatment of STEC and HUS.
  • Clostridium difficile (C. difficile) is a bacterium that can cause symptoms ranging from diarrhea to life-threatening inflammation of the colon. C. difficile most commonly affects older adults in hospitals or in long-term care facilities and often occurs after the use of antibiotics.
  • the most well-understood toxins produced by pathogenic C. difficile strains are enterotoxin (Clostridium difficile toxin A) and cytotoxin (Clostridium difficile toxin B), both of which can produce diarrhea and inflammation in infected patients.
  • the polynucleotides of the invention may be used in the treatment or prevention of Clostridium difficile (C. difficile) infection.
  • the polynucleotides of the invention may encode at least one antibody, fragment or variant thereof which may be used for the treatment or prevention of C. difficile infection.
  • the polynucleotides described herein encode a monoclonal antibody that is directed to toxin A and/or toxin B for C. difficile, which may be used for the treatment or prevention of C. difficile infection.
  • a monoclonal antibody that is directed to toxin A and/or toxin B for C. difficile, which may be used for the treatment or prevention of C. difficile infection.
  • Pseudomonas aeruginosa there are other genera of Gram-negative bacteria, such as the Acinetobacter species, that often produce multidrug-resistant and even pan- resistant strains.
  • Acinetobacter baumannii is a Gram-negative bacterium that has been isolated form water and soil samples.
  • A. baumannii affects people with compromised immune systems, and is becoming increasingly more frequent as a hospital-derived infection. Due to its ability to form a biofilm, it can persist on artificial surfaces and infect new patients. It is thought that the ability of A. baumannii to form biofilms is correlated with multi drug resistance (MDR).
  • MDR multi drug resistance
  • A. baumannii also forms protective capsules composed of polysaccharides around each individual cell, further providing additional protection from antibiotics and antibacterial agents.
  • VAP ventilator- associated pneumonia
  • passive immunization approach provides only temporary immunity, it may be sufficient to clear an acute A. baumannii infection, alone or in combination with other antimicrobials. Passive immunization may therefore become an important therapeutic approach, in particular given the high incidence of multi-drug resistant strains.
  • the antibodies encoded by the polynucleotides of the present invention bind or target one or more proteins or peptides of Acinetobacter baumannii.
  • Hepatitis B virus causes an infectious illness of the liver and has caused epidemics in parts of Asia and Africa, and is still endemic in China.
  • the virus is transmitted by exposure to infectious blood or body fluids such as semen and vaginal fluids.
  • Perinatal infection is a major route of infection in developing countries.
  • the acute illness causes liver inflammation, vomiting, jaundice.
  • HBV results in one million deaths annually, primarily due to cirrhosis and liver cancer.
  • the hepatitis B virus is a partially double stranded DNA virus composed of a 27 nm nucleocapsid core (HBcAg), surrounded by an outer lipoprotein coat (envelope) containing the surface antigen (HBsAg).
  • the nucleocapsid has been found to be very immunogenic and a number of antibodies with nucleocapsid epitopes have been described.
  • the nucleocapsid is dimorphic and is comprised of either 90 or 120 dimers arranged such that the four-helix bundles project from the surface as 25 ⁇ -long spike. Together these two capsid forms are known as core antigen (HBcAg).
  • the principal antigenic determinant of HBcAg is located at the apices of the capsid spikes (Watts et al, Non- Canonical Binding Of An Antibody Resembling A Na ⁇ ve B Cell Receptor Immunoglobin To Hepatitis B Virus Capsids J Mol Biol. Jun 20, 2008; 379(5): 1119–1129, the contents of which are herein incorporated by reference in its entirety).
  • the polynucleotides of the invention may encode at least one antibody that binds to the nucleocapsid of HBV.
  • the polynucleotides of the invention may encode at least one antibody that binds to the nucleocapsid of HBV for the prevention, management, or treatment of HBV infections.
  • the polynucleotides of the invention may encode at least one antibody that binds to the nucleocapsid of HBV that can be used in combination with other HBV treatments, including existing HBV vaccines.
  • Cancer is one of the leading causes of death in the United States. Conventional methods of cancer treatment like chemotherapy, surgery or radiation therapy, can be limited in their efficacy since they are often nonspecific to the cancer. In many cases tumors, however, can specifically express genes whose products are required for inducing or maintaining the malignant state. These proteins may serve as antigen markers for the development and establishment of efficient anti-cancer treatments.
  • the polynucleotides of the invention may encode anti-cancer antibodies. Such antibodies may be used to target cancer cells by binding cancer antigens.
  • Cancer antigens can elicit an immune response.
  • These antigens can be either proteins, polysaccharides, lipids, or glycolipids, which can be recognized as foreign by immune cells, such as T cells and B cells. Exposure of immune cells to one or more of these antigens can elicit a rapid cell division and differentiation response resulting in the formation of clones of the exposed T cells and B cells. B cells can differentiate into plasma cells which in turn can produce antibodies which selectively bind to the antigens.
  • tumor antigens there are four general groups of tumor antigens: (i) viral tumor antigens which can be identical for any viral tumor of this type, (ii) carcinogenic tumor antigens which can be specific for patients and for the tumors, (iii) isoantigens of the transplantation type or tumor-specific transplantation antigens which can be different in all individual types of tumor but can be the same in different tumors caused by the same virus; and (iv) embryonic antigens.
  • the polynucleotides of the ivention encode any tyoe of cancer antigen including any of these 4 classes of antigens.
  • an anti-CD40 antibody that is a CD40 agonist can be used to activate dendritic cells to enhance the immune response.
  • antibodies may function as immune checkpoint modulators.
  • the polynucleotides encode an antibody that is a T cell activator such as an immune checkpoint modulator.
  • Immune checkpoint modulators include both stimulatory checkpoint molecules and inhibitory checkpoint molecules i.e., an anti-CTLA4 and anti-PD1 antibody.
  • Stimulatory checkpoint inhibitors function by promoting the checkpoint process.
  • GITR Glucocorticoid-Induced TNFR family Related gene
  • CD27 supports antigen-specific expansion of na ⁇ ve T cells and is involved in the generation of T and B cell memory.
  • Several agonistic anti-CD27 antibodies are in development.
  • CD122 is the Interleukin-2 receptor beta sub-unit.
  • NKTR-214 is a CD122-biased immune-stimulatory cytokine.
  • Inhibitory checkpoint molecules include but are not limited to PD-1, TIM-3, VISTA, A2AR, B7-H3, B7-H4, BTLA, CTLA-4, IDO, KIR and LAG3.
  • CTLA-4, PD-1 and its ligands are members of the CD28-B7 family of co-signaling molecules that play important roles throughout all stages of T-cell function and other cell functions.
  • CTLA-4, Cytotoxic T- Lymphocyte-Associated protein 4 (CD152) is involved in controlling T cell proliferation.
  • the PD-1 receptor is expressed on the surface of activated T cells (and B cells) and, under normal circumstances, binds to its ligands (PD-L1 and PD-L2) that are expressed on the surface of antigen-presenting cells, such as dendritic cells or macrophages. This interaction sends a signal into the T cell and inhibits it.
  • Cancer cells take advantage of this system by driving high levels of expression of PD-L1 on their surface. This allows them to gain control of the PD-1 pathway and switch off T cells expressing PD-1 that may enter the tumor microenvironment, thus suppressing the anticancer immune response.
  • Pembrolizumab (formerly MK-3475 and lambrolizumab, trade name Keytruda) is a human antibody used in cancer immunotherapy.
  • the checkpoint inhibitor is a molecule such as a monoclonal antibody, a humanized antibody, a fully human antibody, a fusion protein or a combination thereof or a small molecule.
  • the checkpoint inhibitor inhibits a checkpoint protein which may be CTLA-4, PDL1, PDL2, PD1, B7-H3, B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4, CD160, CGEN-15049, CHK 1, CHK2, A2aR, B-7 family ligands or a
  • Ligands of checkpoint proteins include but are not limited to CTLA-4, PDL1, PDL2, PD1, B7-H3, B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4, CD160, CGEN-15049, CHK 1, CHK2, A2aR, and B-7 family ligands.
  • the anti-PD-1 antibody is BMS-936558 (nivolumab).
  • the anti-CTLA-4 antibody is ipilimumab (trade name Yervoy, formerly known as MDX-010 and MDX-101).
  • RNA polynucleotide of the ivnetnion may encode an antibody against any cancer antigen.
  • cancer antigens are antigens which can potentially stimulate apparently tumor-specific immune responses. Some of these antigens are encoded, although not necessarily expressed, by normal cells. These antigens can be characterized as those which are normally silent (i.e., not expressed) in normal cells, those that are expressed only at certain stages of differentiation and those that are temporally expressed such as embryonic and fetal antigens.
  • cancer antigens are encoded by mutant cellular genes, such as oncogenes (e.g., activated ras oncogene), suppressor genes (e.g., mutant p53), fusion proteins resulting from internal deletions or chromosomal translocations. Still other cancer antigens can be encoded by viral genes such as those carried on RNA and DNA tumor viruses.
  • an antibody specific for a cell surface antigen of, for example, a cancer cell may promote an immune response resulting in antibody dependent cellular cytotoxicity (ADCC).
  • the antibody may be selected from the group consisting of Ributaxin, Herceptin, Quadramet, Panorex, IDEC-Y2B8, BEC2, C225, Oncolym, SMART M195, ATRAGEN, Ovarex, Bexxar, LDP-03, ior t6, MDX-210, MDX-11, MDX-22, OV103, 3622W94, anti-VEGF, Zenapax, MDX-220, MDX-447, MELIMMUNE-2, MELIMMUNE- 1, CEACIDE, Pretarget, NovoMAb-G2, TNT, Gliomab-H, GNI-250, EMD-72000,
  • LymphoCide CMA 676, Monopharm-C, 4B5, ior egf.r3, ior c5, BABS, anti-FLK-2, MDX- 260, ANA Ab, SMART 1D10 Ab, SMART ABL 364 Ab and ImmuRAIT-CEA.
  • an intrabody construct is a polynucleotide which has been modified for expression inside a target cell and where the expression product binds an intracellular protein.
  • Such constructs may have sub picomolar binding affinities and may be formulated for targeting to particular sites or tissues.
  • intrabody constructs may be formulated in any of the lipid nanoparticle formulations disclosed herein.
  • a bicistronic construct is a polynucleotide encoding a two-protein chain antibody on a single polynucleotide strand.
  • a pseudo- bicistronic construct is a polynucleotide encoding a single chain antibody discontinuously on a single polynucleotide strand.
  • the encoded two strands or two portions/regions and/or domains are separated by at least one nucleotide not encoding the strands or domains.
  • the separation comprises a cleavage signal or site or a non-coding region of nucleotides.
  • uch cleavage sites include, for example, furin cleavage sites encoded as an“RKR” site in the resultant polypeptide.
  • a single domain construct comprises one or two polynucleotides ecoding a single monomeric variable antibody domain. See Figs 3B and 4B for examples.
  • single domain antibodies comprise one variable domain (VH) of a heavy-chain antibody.
  • a single chain Fv constructs is a polynucleotide encoding at least two coding regions and a linker region.
  • the scFv construct may encode a fusion protein of the variable regions of the heavy (VH) and light chains (VL) of
  • linker is usually rich in glycine for flexibility, as well as serine or threonine for solubility, and can either connect the N-terminus of the VH with the C- terminus of the VL, or vice versa.
  • Other linkers include those known in the art and disclosed herein.
  • Single chain antibodies may be camelid antibodies. They may also be human heavy chain only antibodies such as those made by Crescendo Biologics. Bispecific Constructs
  • a bispecific construct is a polynucleotide encoding portions or regions of two different antibodies.
  • Bispecific constructs encode polypeptides which may bind two different antigens. See Fig.4A for an example.
  • Polynucleotides of the present invention may also encode trispecific antibodies having an affinity for three antigens.
  • the polynucleotides can be designed as modular IgA antibodies. These antibodies may be monomers or dimers or multimers.
  • Modular IgA constructs comprise a VHH antigen binding domain and an IgA2 backbone which can provide protection from bacterial IgA1 protease.
  • Multimeric IgA constructs can be designed using an IgJ chain for polymerization and may be encoded on a polycistronic transcript using a 2A peptide or PACE cleavage. Further modification can include a secretory component of for example polyimmunoglobulin receptor (PIGR) for production of secreted IgA.
  • PIGR polyimmunoglobulin receptor
  • the length of a region encoding at least one peptide polypeptide of interest of the polynucleotides present invention is greater than about 30 nucleotides in length (e.g., at least or greater than about 35, 40, 45, 50, 55, 60, 70, 80, 90, 100, 120, 140, 160, 180, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, 900, 1,000, 1,100, 1,200, 1,300, 1,400, 1,500, 1,600, 1,700, 1,800, 1,900, 2,000, 2,500, and 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, 20,000, 30,000, 40,000, 50,000, 60,000, 70,000, 80,000, 90,000 or up to and including 100,000 nucleotides).
  • a region may be referred to as a “coding region” or“region encoding.”
  • the polynucleotides of the present invention is or functions as a messenger RNA (mRNA).
  • mRNA messenger RNA
  • the term“messenger RNA” (mRNA) refers to any polynucleotide which encodes at least one peptide or polypeptide of interest and which is capable of being translated to produce the encoded peptide polypeptide of interest in vitro, in vivo, in situ or ex vivo.
  • Influenza virus treatments comprise at least one (e.g., one or more) RNA (e.g., mRNA) polynucleotide having an open reading frame encoding at least one antibody, antibody domain, antibody portion, and/or antibody fragment thereof that binds to an influenza virus HA protein.
  • RNA e.g., mRNA
  • Polynucleotides may be or may include, for example, RNAs, deoxyribonucleic acids (DNAs), threose nucleic acids (TNAs), glycol nucleic acids (GNAs), peptide nucleic acids (PNAs), locked nucleic acids (LNAs, including LNA having a ⁇ - D-ribo configuration, ⁇ -LNA having an ⁇ -L-ribo configuration (a diastereomer of LNA), 2′-amino-LNA having a 2′-amino functionalization, and 2′-amino- ⁇ - LNA having a 2′-amino functionalization), ethylene nucleic acids (ENA), cyclohexenyl nucleic acids (CeNA) or chimeras or combinations thereof.
  • DNAs deoxyribonucleic acids
  • TAAs threose nucleic acids
  • GNAs glycol nucleic acids
  • PNAs peptide nucleic acids
  • polynucleotides of the present disclosure function as messenger RNA (mRNA).“Messenger RNA” (mRNA) refers to any polynucleotide that encodes at least one polypeptide (a naturally-occurring, non-naturally-occurring, or modified polymer of amino acids) and can be translated to produce the encoded polypeptide in vitro, in vivo, in situ or ex vivo.
  • mRNA messenger RNA
  • the basic components of an mRNA molecule typically include at least one coding region, a 5′ untranslated region (UTR), a 3′ UTR, a 5′ cap, and a poly-A tail.
  • Polynucleotides of the present disclosure may function as mRNA but can be distinguished from wild-type mRNA in their functional and/or structural design features which serve to overcome existing problems of effective polypeptide expression using nucleic-acid based therapeutics.
  • an RNA polynucleotide encodes 2-10, 2-9, 2-8, 2-7, 2-6, 2-5, 2-4, 2-3, 3-10, 3-9, 3-8, 3-7, 3-6, 3-5, 3-4, 4-10, 4-9, 4-8, 4-7, 4-6, 4-5, 5-10, 5-9, 5-8, 5-7, 5- 6, 6-10, 6-9, 6-8, 6-7, 7-10, 7-9, 7-8, 8-10, 8-9 or 9-10 antibodies and antigen binding fragment polypeptides.
  • an RNA polynucleotide encodes at least 10, 20, 30, 40, 50 , 60, 70, 80, 90 or 100 antibodies and antigen binding fragment polypeptides.
  • an RNA polynucleotide encodes at least 100 or at least 200 antibodies and antigen binding fragment polypeptides. In some embodiments, an RNA polynucleotide encodes 1-10, 5-15, 10-20, 15-25, 20-30, 25-35, 30-40, 35-45, 40-50, 1-50, 1-100, 2-50 or 2- 100 antibodies and antigen binding fragment polypeptides. Signal Sequences
  • the polynucleotides e.g., a RNA, e.g., an mRNA
  • One such feature that aids in protein trafficking is the signal sequence, or targeting sequence.
  • the peptides encoded by these signal sequences are known by a variety of names, including targeting peptides, transit peptides, and signal peptides.
  • the polynucleotide (e.g., a RNA, e.g., an mRNA) comprises a nucleotide sequence (e.g., an ORF) that encodes a signal peptide operably linked a nucleotide sequence that encodes an antibody described herein.
  • a nucleotide sequence e.g., an ORF
  • the "signal sequence” or “signal peptide” is a polynucleotide or polypeptide, respectively, which is from about 30-210, e.g., about 45-80 or 15-60nucleotides (3-70 amino acids) in length that, optionally, is incorporated at the 5′ (or N-terminus) of the coding region or the polypeptide, respectively. Addition of these sequences results in trafficking the encoded polypeptide to a desired site, such as the endoplasmic reticulum or the mitochondria through one or more targeting pathways. Some signal peptides are cleaved from the protein, for example by a signal peptidase after the proteins are transported to the desired site.
  • the polynucleotide of the present disclosure comprises a nucleotide sequence encoding an antibody, wherein the nucleotide sequence further comprises a 5' nucleic acid sequence encoding a native signal peptide.
  • the polynucleotide of the present disclosure comprises a nucleotide sequence encoding an antibody, wherein the nucleotide sequence lacks the nucleic acid sequence encoding a native signal peptide.
  • the polynucleotide of the present disclosure comprises a nucleotide sequence encoding an antibody, wherein the nucleotide sequence further comprises a 5' nucleic acid sequence encoding a heterologous signal peptide.
  • the polynucleotide of the present disclosure can comprise more than one nucleic acid sequence (e.g., an ORF) encoding a polypeptide of interest.
  • polynucleotides of the present disclosure comprise a single ORF encoding an antibody, a functional fragment, or a variant thereof.
  • the polynucleotide of the present disclosure can comprise more than one ORF, for example, a first ORF encoding an antibody (a first polypeptide of interest), a functional fragment, or a variant thereof, and a second ORF expressing a second polypeptide of interest.
  • two or morepolypeptides of interest can be genetically fused, i.e., two or more polypeptides can be encoded by the same ORF.
  • the polynucleotide can comprise a nucleic acid sequence encoding a linker (e.g., a (G 4 S) n peptide linker (wherein n is 2, 3, 4, 5, 6, 7, 8, 9 or 10) or another linker known in the art) between two or more polypeptides of interest.
  • a linker e.g., a (G 4 S) n peptide linker (wherein n is 2, 3, 4, 5, 6, 7, 8, 9 or 10) or another linker known in the art
  • a polynucleotide of the present disclosure e.g., a RNA, e.g., an mRNA
  • a polynucleotide of the present disclosure can comprise two, three, four, or more ORFs, each expressing a polypeptide of interest.
  • the polynucleotide of the present disclosure can comprise a first nucleic acid sequence (e.g., a first ORF) encoding an antibody and a second nucleic acid sequence (e.g., a second ORF) encoding a second polypeptide of interest such as an antibody Fc domain.
  • a first nucleic acid sequence e.g., a first ORF
  • a second nucleic acid sequence e.g., a second ORF
  • the polynucleotide (e.g., a RNA, e.g., an mRNA) of the present disclosure is sequence optimized.
  • the polynucleotide (e.g., a RNA, e.g., an mRNA) of the present disclosure comprises a nucleotide sequence (e.g., an ORF) encoding an antibody, optionally, a nucleotide sequence (e.g, an ORF) encoding another polypeptide of interest, a 5'-UTR, a 3'-UTR, the 5’ UTR or 3’ UTR optionally comprising at least one microRNA binding site, optionally a nucleotide sequence encoding a linker, a polyA tail, or any combination thereof), in which the ORF(s) that are sequence optimized.
  • a sequence-optimized nucleotide sequence e.g., an codon-optimized mRNA sequence encoding an antibody, is a sequence comprising at least one synonymous nucleobase substitution with respect to a reference sequence (e.g., a wild type nucleotide sequence encoding an antibody).
  • a sequence-optimized nucleotide sequence can be partially or completely different in sequence from the reference sequence.
  • a reference sequence encoding polyserine uniformly encoded by TCT codons can be sequence-optimized by having 100% of its nucleobases substituted (for each codon, T in position 1 replaced by A, C in position 2 replaced by G, and T in position 3 replaced by C) to yield a sequence encoding polyserine which would be uniformly encoded by AGC codons.
  • the percentage of sequence identity obtained from a global pairwise alignment between the reference polyserine nucleic acid sequence and the sequence-optimized polyserine nucleic acid sequence would be 0%.
  • sequence optimization also sometimes referred to codon optimization
  • results can include, e.g., matching codon frequencies in certain tissue targets and/or host organisms to ensure proper folding; biasing G/C content to increase mRNA stability or reduce secondary structures; minimizing tandem repeat codons or base runs that can impair gene construction or expression; customizing transcriptional and translational control regions; inserting or removing protein trafficking sequences; removing/adding post translation modification sites in an encoded protein (e.g., glycosylation sites); adding, removing or shuffling protein domains; inserting or deleting restriction sites; modifying ribosome binding sites and mRNA degradation sites; adjusting translational rates to allow the various domains of the protein to fold properly; and/or reducing or eliminating problem secondary structures within the polynucleotide.
  • Sequence optimization tools, algorithms and services are known in the art, non-limiting examples include services from GeneArt (Life Technologies), DNA2.0 (
  • a polynucleotide e.g., a RNA, e.g., an mRNA
  • a sequence-optimized nucleotide sequence e.g., an ORF
  • the antibody, functional fragment, or a variant thereof encoded by the sequence-optimized nucleotide sequence has improved properties (e.g., compared to an antibody, functional fragment, or a variant thereof encoded by a reference nucleotide sequence that is not sequence optimized), e.g., improved properties related to expression efficacy after administration in vivo.
  • Such properties include, but are not limited to, improving nucleic acid stability (e.g., mRNA stability), increasing translation efficacy in the target tissue, reducing the number of truncated proteins expressed, improving the folding or prevent misfolding of the expressed proteins, reducing toxicity of the expressed products, reducing cell death caused by the expressed products, increasing and/or decreasing protein aggregation.
  • nucleic acid stability e.g., mRNA stability
  • increasing translation efficacy in the target tissue reducing the number of truncated proteins expressed, improving the folding or prevent misfolding of the expressed proteins, reducing toxicity of the expressed products, reducing cell death caused by the expressed products, increasing and/or decreasing protein aggregation.
  • sequence-optimized nucleotide sequence (e.g., an ORF) is codon optimized for expression in human subjects, having structural and/or chemical features that avoid one or more of the problems in the art, for example, features which are useful for optimizing formulation and delivery of nucleic acid-based therapeutics while retaining structural and functional integrity; overcoming a threshold of expression; improving expression rates; half-life and/or protein concentrations; optimizing protein localization; and avoiding deleterious bio-responses such as the immune response and/or degradation pathways.
  • an ORF codon optimized for expression in human subjects, having structural and/or chemical features that avoid one or more of the problems in the art, for example, features which are useful for optimizing formulation and delivery of nucleic acid-based therapeutics while retaining structural and functional integrity; overcoming a threshold of expression; improving expression rates; half-life and/or protein concentrations; optimizing protein localization; and avoiding deleterious bio-responses such as the immune response and/or degradation pathways.
  • the polynucleotides of the present disclosure comprise a nucleotide sequence (e.g., a nucleotide sequence (e.g, an ORF) encoding an antibody, a 5'- UTR, a 3'-UTR, a microRNA, a nucleic acid sequence encoding a linker, or any combination thereof) that is sequence-optimized according to a method comprising:
  • the sequence-optimized nucleotide sequence (e.g., an ORF encoding an antibody) has at least one improved property with respect to the reference nucleotide sequence.
  • the sequence optimization method is multiparametric and comprises one, two, three, four, or more methods disclosed herein and/or other optimization methods known in the art.
  • regions of the polynucleotide can be encoded by or within regions of the polynucleotide and such regions can be upstream (5') to, downstream (3') to, or within the region that encodes the antibody. These regions can be incorporated into the polynucleotide before and/or after sequence-optimization of the protein encoding region or open reading frame (ORF). Examples of such features include, but are not limited to, untranslated regions (UTRs), microRNA sequences, Kozak sequences, oligo(dT) sequences, poly-A tail, and detectable tags and can include multiple cloning sites that can have XbaI recognition.
  • UTRs untranslated regions
  • microRNA sequences microRNA sequences
  • Kozak sequences oligo(dT) sequences
  • poly-A tail poly-A tail
  • detectable tags can include multiple cloning sites that can have XbaI recognition.
  • the polynucleotide of the present disclosure comprises a 5′ UTR. a 3′ UTR and/or a miRNA binding site. In some embodiments, the polynucleotide comprises two or more 5′ UTRs and/or 3′ UTRs, which can be the same or different sequences. In some embodiments, the polynucleotide comprises two or more miRNA binding site, which can be the same or different sequences. Any portion of the 5’ UTR, 3’ UTR, and/or miRNA binding site, including none, can be sequence-optimized and can be sequence-optimized and can be
  • the polynucleotide is reconstituted and transformed into a vector such as, but not limited to, plasmids, viruses, cosmids, and artificial chromosomes.
  • a vector such as, but not limited to, plasmids, viruses, cosmids, and artificial chromosomes.
  • the optimized polynucleotide can be reconstituted and transformed into chemically competent E. coli, yeast, neurospora, maize, drosophila, etc. where high copy plasmid-like or chromosome structures occur by methods described herein.
  • the polynucleotide of the present disclosure comprises a sequence-optimized nucleotide sequence encoding an antibody disclosed herein. In some embodiments, the polynucleotide of the present disclosure comprises an open reading frame (ORF) encoding an antibody, wherein the ORF has been sequence optimized.
  • ORF open reading frame
  • sequence-optimized nucleotide sequences disclosed herein are distinct from the corresponding wild type nucleotide acid sequences and from other known sequence- optimized nucleotide sequences, e.g., these sequence-optimized nucleic acids have unique compositional characteristics.
  • the percentage of uracil or thymine nucleobases in a sequence- optimized nucleotide sequence is modified (e.g,. reduced) with respect to the percentage of uracil or thymine nucleobases in the reference wild-type nucleotide sequence.
  • a sequence is referred to as a uracil-modified or thymine-modified sequence.
  • the percentage of uracil or thymine content in a nucleotide sequence can be determined by dividing the number of uracils or thymines in a sequence by the total number of nucleotides and multiplying by 100.
  • the sequence-optimized nucleotide sequence has a lower uracil or thymine content than the uracil or thymine content in the reference wild-type sequence.
  • the uracil or thymine content in a sequence-optimized nucleotide sequence of the present disclosure is greater than the uracil or thymine content in the reference wild-type sequence and still maintain beneficial effects, e.g., increased expression and/or reduced Toll-Like Receptor (TLR) response when compared to the reference wild-type sequence.
  • TLR Toll-Like Receptor
  • the polynucleotide e.g., a RNA, e.g., an mRNA
  • a sequence optimized nucleic acid disclosed herein encoding an antibody can be can be tested to determine whether at least one nucleic acid sequence property (e.g., stability when exposed to nucleases) or expression property has been improved with respect to the non-sequence optimized nucleic acid.
  • expression property refers to a property of a nucleic acid sequence either in vivo (e.g., translation efficacy of a synthetic mRNA after administration to a subject in need thereof) or in vitro (e.g., translation efficacy of a synthetic mRNA tested in an in vitro model system).
  • Expression properties include but are not limited to the amount of protein produced by an mRNA encoding an antibody after administration, and the amount of soluble or otherwise functional protein produced.
  • sequence optimized nucleic acids disclosed herein can be evaluated according to the viability of the cells expressing a protein encoded by a sequence optimized nucleic acid sequence (e.g., a RNA, e.g., an mRNA) encoding an antibody disclosed herein.
  • a sequence optimized nucleic acid sequence e.g., a RNA, e.g., an mRNA
  • a plurality of sequence optimized nucleic acids disclosed herein e.g., a RNA, e.g., an mRNA
  • a property of interest for example an expression property in an in vitro model system, or in vivo in a target tissue or cell.
  • the desired property of the polynucleotide is an intrinsic property of the nucleic acid sequence.
  • the nucleotide sequence e.g., a RNA, e.g., an mRNA
  • the nucleotide sequence can be sequence optimized for in vivo or in vitro stability.
  • the nucleotide sequence can be sequence optimized for expression in a particular target tissue or cell.
  • the nucleic acid sequence is sequence optimized to increase its plasma half by preventing its degradation by endo and exonucleases.
  • the nucleic acid sequence is sequence optimized to increase its resistance to hydrolysis in solution, for example, to lengthen the time that the sequence optimized nucleic acid or a pharmaceutical composition comprising the sequence optimized nucleic acid can be stored under aqueous conditions with minimal degradation.
  • sequence optimized nucleic acid can be optimized to increase its resistance to hydrolysis in dry storage conditions, for example, to lengthen the time that the sequence optimized nucleic acid can be stored after lyophilization with minimal degradation.
  • the desired property of the polynucleotide is the level of expression of an antibody encoded by a sequence optimized sequence disclosed herein.
  • Protein expression levels can be measured using one or more expression systems.
  • expression can be measured in cell culture systems, e.g., CHO cells or HEK293 cells.
  • expression can be measured using in vitro expression systems prepared from extracts of living cells, e.g., rabbit reticulocyte lysates, or in vitro expression systems prepared by assembly of purified individual components.
  • the protein expression is measured in an in vivo system, e.g., mouse, rabbit, monkey, etc.
  • protein expression in solution form can be desirable.
  • a reference sequence can be sequence optimized to yield a sequence optimized nucleic acid sequence having optimized levels of expressed proteins in soluble form.
  • Levels of protein expression and other properties such as solubility, levels of aggregation, and the presence of truncation products (i.e., fragments due to proteolysis, hydrolysis, or defective translation) can be measured according to methods known in the art, for example, using electrophoresis (e.g., native or SDS-PAGE) or chromatographic methods (e.g., HPLC, size exclusion chromatography, etc.).
  • electrophoresis e.g., native or SDS-PAGE
  • chromatographic methods e.g., HPLC, size exclusion chromatography, etc.
  • heterologous therapeutic proteins encoded by a nucleic acid sequence can have deleterious effects in the target tissue or cell, reducing protein yield, or reducing the quality of the expressed product (e.g., due to the presence of protein fragments or precipitation of the expressed protein in inclusion bodies), or causing toxicity.
  • sequence optimization of a nucleic acid sequence disclosed herein can be used to increase the viability of target cells expressing the protein encoded by the sequence optimized nucleic acid.
  • Heterologous protein expression can also be deleterious to cells transfected with a nucleic acid sequence for autologous or heterologous transplantation. Accordingly, in some embodiments of the present disclosure the sequence optimization of a nucleic acid sequence disclosed herein can be used to increase the viability of target cells expressing the protein encoded by the sequence optimized nucleic acid sequence. Changes in cell or tissue viability, toxicity, and other physiological reaction can be measured according to methods known in the art. d. Reduction of Immune and/or Inflammatory Response
  • the administration of a sequence optimized nucleic acid encoding antibody or a functional fragment thereof can trigger an immune response, which could be caused by (i) the therapeutic agent (e.g., an mRNA encoding an antibody), or (ii) the expression product of such therapeutic agent (e.g., the antibody encoded by the mRNA), or (iv) a combination thereof.
  • the sequence optimization of nucleic acid sequence e.g., an mRNA
  • the sequence optimization of nucleic acid sequence can be used to decrease an immune or inflammatory response (other than coagulation pathway activation) triggered by the administration of a nucleic acid encoding an antibody or by the expression product of antibody encoded by such nucleic acid.
  • an inflammatory response can be measured by detecting increased levels of one or more inflammatory cytokines using methods known in the art, e.g., ELISA.
  • inflammatory cytokine refers to cytokines that are elevated in an inflammatory response. Examples of inflammatory cytokines include interleukin-6 (IL-6), CXCL1
  • chemokine (C-X-C motif) ligand 1 also known as GRO ⁇ , interferon- ⁇ (IFN ⁇ ), tumor necrosis factor ⁇ (TNF ⁇ ), interferon ⁇ -induced protein 10 (IP-10), or granulocyte-colony stimulating factor (G-CSF).
  • inflammatory cytokines includes also other cytokines associated with inflammatory responses known in the art, e.g., interleukin-1 (IL-1), interleukin-8 (IL-8), interleukin-12 (IL-12), interleukin-13 (Il-13), interferon ⁇ (IFN- ⁇ ), etc.
  • the polynucleotide (e.g., a RNA, e.g., an mRNA) of the present disclosure comprises a chemically modified nucleobase, , for example, a chemically modified uracil, e.g., pseudouracil, 1-methylpseuodouracil, 5-methoxyuracil, or the like.
  • a chemically modified uracil e.g., pseudouracil, 1-methylpseuodouracil, 5-methoxyuracil, or the like.
  • the mRNA is a uracil-modified sequence comprising an ORF encoding an antibody, wherein the mRNA comprises a chemically modified nucleobase, for example, a chemically modified uracil, e.g., pseudouracil, 1-methylpseuodouracil, or 5-methoxyuracil.
  • a chemically modified uracil e.g., pseudouracil, 1-methylpseuodouracil, or 5-methoxyuracil.
  • modified uracil base when the modified uracil base is connected to a ribose sugar, as it is in polynucleotides, the resulting modified nucleoside or nucleotide is refered to as modified uradine.
  • polynucleotide is at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least 90%, at least 95%, at least 99%, or about 100% modified uracil. In one embodiment, uracil in the polynucleotide is at least 95% modified uracil. In another embodiment, uracil in the polynucleotide is 100% modified uracil.
  • a binding peptide e.g., antibody, such as an influenza virus binding polypeptide
  • polypeptides include gene products, naturally occurring polypeptides, synthetic polypeptides, homologs, orthologs, paralogs, fragments and other equivalents, variants, and analogs of the foregoing.
  • a polypeptide may be a single molecule or may be a multi-molecular complex such as a dimer, trimer or tetramer.
  • Polypeptides may also comprise single chain or multichain polypeptides such as antibodies and may be associated or linked. Most commonly, disulfide linkages are found in multichain polypeptides.
  • the term polypeptide may also apply to amino acid polymers in which at least one amino acid residue is an artificial chemical analogue of a corresponding naturally-occurring amino acid.
  • polypeptide variant refers to molecules which differ in their amino acid sequence from a native or reference sequence.
  • the amino acid sequence variants may possess substitutions, deletions, and/or insertions at certain positions within the amino acid sequence, as compared to a native or reference sequence.
  • variants possess at least 50% identity to a native or reference sequence.
  • variants share at least 80%, or at least 90% identity with a native or reference sequence.
  • variant mimics are provided.
  • the term “variant mimic” is one which contains at least one amino acid that would mimic an activated sequence.
  • glutamate may serve as a mimic for phosphoro-threonine and/or phosphoro-serine.
  • variant mimics may result in deactivation or in an inactivated product containing the mimic, for example, phenylalanine may act as an inactivating substitution for tyrosine; or alanine may act as an inactivating substitution for serine.
  • orthologs refers to genes in different species that evolved from a common ancestral gene by speciation. Normally, orthologs retain the same function in the course of evolution. Identification of orthologs is critical for reliable prediction of gene function in newly sequenced genomes.
  • Analogs is meant to include polypeptide variants which differ by one or more amino acid alterations, for example, substitutions, additions or deletions of amino acid residues that still maintain one or more of the properties of the parent or starting polypeptide.
  • compositions that are polynucleotide or polypeptide based, including variants and derivatives. These include, for example, substitutional, insertional, deletion and covalent variants and derivatives.
  • derivative is used synonymously with the term“variant” but generally refers to a molecule that has been modified and/or changed in any way relative to a reference molecule or starting molecule.
  • polypeptide sequences or polypeptides containing substitutions, insertions and/or additions, deletions and covalent modifications with respect to reference sequences, in particular the polypeptide sequences disclosed herein are included within the scope of this disclosure.
  • sequence tags or amino acids, such as one or more lysines can be added to peptide sequences (e.g., at the N-terminal or C-terminal ends).
  • Sequence tags can be used for peptide detection, purification or localization.
  • Lysines can be used to increase peptide solubility or to allow for biotinylation.
  • amino acid residues located at the carboxy and amino terminal regions of the amino acid sequence of a peptide or protein may optionally be deleted providing for truncated sequences.
  • Certain amino acids e.g., C-terminal or N-terminal residues may alternatively be deleted depending on the use of the sequence, as for example, expression of the sequence as part of a larger sequence which is soluble, or linked to a solid support.
  • substitutional variants when referring to polypeptides are those that have at least one amino acid residue in a native or starting sequence removed and a different amino acid inserted in its place at the same position. Substitutions may be single, where only one amino acid in the molecule has been substituted, or they may be multiple, where two or more amino acids have been substituted in the same molecule.
  • conservative amino acid substitution refers to the substitution of an amino acid that is normally present in the sequence with a different amino acid of similar size, charge, or polarity.
  • conservative substitutions include the substitution of a non-polar (hydrophobic) residue such as isoleucine, valine, and leucine for another non-polar residue.
  • conservative substitutions include the substitution of one polar (hydrophilic) residue for another such as between arginine and lysine, between glutamine and asparagine, and between glycine and serine.
  • substitution of a basic residue such as lysine, arginine or histidine for another, or the substitution of one acidic residue such as aspartic acid or glutamic acid for another acidic residue are additional examples of conservative substitutions.
  • non-conservative substitutions include the substitution of a non-polar (hydrophobic) amino acid residue such as isoleucine, valine, leucine, alanine, methionine for a polar (hydrophilic) residue such as cysteine, glutamine, glutamic acid or lysine and/or a polar residue for a non-polar residue.
  • Features when referring to polypeptide or polynucleotide are defined as distinct amino acid sequence-based or nucleotide-based components of a molecule respectively.
  • Features of the polypeptides encoded by the polynucleotides include surface manifestations, local conformational shape, folds, loops, half-loops, domains, half-domains, sites, termini, or any combination thereof.
  • domain refers to a motif of a polypeptide having one or more identifiable structural or functional characteristics or properties (e.g., binding capacity, serving as a site for protein-protein interactions).
  • the terms“site” as it pertains to amino acid based embodiments is used synonymously with“amino acid residue” and“amino acid side chain.”
  • the terms“site” as it pertains to nucleotide based embodiments is used synonymously with“nucleotide.”
  • a site represents a position within a peptide or polypeptide or polynucleotide that may be modified, manipulated, altered, derivatized or varied within the polypeptide or polynucleotide based molecules.
  • polypeptides or polynucleotides refers to an extremity of a polypeptide or polynucleotide, respectively. Such extremity is not limited only to the first or final site of the polypeptide or polynucleotide, but may include additional amino acids or nucleotides in the terminal regions.
  • Polypeptide-based molecules may be characterized as having both an N-terminus (terminated by an amino acid with a free amino group (NH 2 )) and a C-terminus (terminated by an amino acid with a free carboxyl group (COOH)).
  • Proteins are in some cases made up of multiple polypeptide chains brought together by disulfide bonds or by non-covalent forces (e.g., multimers, oligomers). These proteins have multiple N- and C-termini. Alternatively, the termini of the polypeptides may be modified such that they begin or end, as the case may be, with a non- polypeptide based moiety such as an organic conjugate.
  • protein fragments, functional protein domains, and homologous proteins are also considered to be within the scope of polypeptides of interest.
  • any protein fragment meaning a polypeptide sequence at least one amino acid residue shorter than a reference polypeptide sequence but otherwise identical
  • a reference protein 10 20, 30, 40, 50, 60, 70, 80, 90, 100 or greater than 100 amino acids in length.
  • any protein that includes a stretch of 20, 30, 40, 50, or 100 amino acids which are 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 100% identical to any of the sequences described herein can be utilized in accordance with the disclosure.
  • a polypeptide includes 2, 3, 4, 5, 6, 7, 8, 9, 10, or more mutations as shown in any of the sequences provided or referenced herein.
  • any protein that includes a stretch of 20, 30, 40, 50, or 100 amino acids that are greater than 80%, 90%, 95%, or 100% identical to any of the sequences described herein, wherein the protein has a stretch of 5, 10, 15, 20, 25, or 30 amino acids that are less than 80%, 75%, 70%, 65% or 60% identical to any of the sequences described herein can be utilized in accordance with the disclosure.
  • Polypeptide or polynucleotide molecules of the present disclosure may share a certain degree of sequence similarity or identity with the reference molecules (e.g., reference polypeptides or reference polynucleotides), for example, with art-described molecules (e.g., engineered or designed molecules or wild-type molecules).
  • the term“identity” as known in the art refers to a relationship between the sequences of two or more polypeptides or polynucleotides, as determined by comparing the sequences. In the art, identity also means the degree of sequence relatedness between them as determined by the number of matches between strings of two or more amino acid residues or nucleic acid residues.
  • Identity measures the percent of identical matches between the smaller of two or more sequences with gap alignments (if any) addressed by a particular mathematical model or computer program (e.g.,“algorithms”). Identity of related peptides can be readily calculated by known methods. “% identity” as it applies to polypeptide or polynucleotide sequences is defined as the percentage of residues (amino acid residues or nucleic acid residues) in the candidate amino acid or nucleic acid sequence that are identical with the residues in the amino acid sequence or nucleic acid sequence of a second sequence after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent identity. Methods and computer programs for the alignment are well known in the art.
  • variants of a particular polynucleotide or polypeptide have at least 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% but less than 100% sequence identity to that particular reference polynucleotide or polypeptide as determined by sequence alignment programs and parameters described herein and known to those skilled in the art.
  • tools for alignment include those of the BLAST suite (Stephen F.
  • FGSAA Fast Optimal Global Sequence Alignment Algorithm
  • homologous refers to the overall relatedness between polymeric molecules, e.g. between nucleic acid molecules (e.g., DNA molecules and/or RNA molecules) and/or between polypeptide molecules.
  • Polymeric molecules e.g., nucleic acid molecules (e.g., DNA molecules and/or RNA molecules) and/or polypeptide molecules
  • homologous e.g., nucleic acid molecules (e.g., DNA molecules and/or RNA molecules) and/or polypeptide molecules
  • homologous e.g., nucleic acid molecules (e.g., DNA molecules and/or RNA molecules) and/or polypeptide molecules) that share a threshold level of similarity or identity determined by alignment of matching residues are termed homologous.
  • Homology is a qualitative term that describes a relationship between molecules and can be based upon the quantitative similarity or identity. Similarity or identity is a quantitative term that defines the degree of sequence match between two compared sequences.
  • polymeric molecules are considered to be“homologous” to one another 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 (polynucleotide or polypeptide sequences). Two polynucleotide sequences are considered homologous if the polypeptides they encode are at least 50%, 60%, 70%, 80%, 90%, 95%, or even 99% for at least one stretch of at least 20 amino acids. In some embodiments, homologous
  • polynucleotide 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. Two protein sequences are considered homologous if the proteins are at least 50%, 60%, 70%, 80%, or 90% identical for at least one stretch of at least 20 amino acids.
  • homolog refers to a first amino acid sequence or nucleic acid sequence (e.g., gene (DNA or RNA) or protein sequence) that is related to a second amino acid sequence or nucleic acid sequence by descent from a common ancestral sequence.
  • the term“homolog” may apply to the relationship between genes and/or proteins separated by the event of speciation or to the relationship between genes and/or proteins separated by the event of genetic duplication.
  • “Orthologs” are genes (or proteins) in different species that evolved from a common ancestral gene (or protein) by speciation.
  • orthologs typically retain the same function in the course of evolution.“Paralogs” are genes (or proteins) related by duplication within a genome. Orthologs retain the same function in the course of evolution, whereas paralogs evolve new functions, even if these are related to the original one.
  • identity refers to the overall relatedness between polymeric molecules, for example, between polynucleotide molecules (e.g. DNA molecules and/or RNA molecules) and/or between polypeptide molecules. Calculation of the percent identity of two polynucleic acid 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).
  • 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 position.
  • 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 comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm.
  • the percent identity between two nucleic acid sequences can be determined using methods such as those described in Computational Molecular Biology, Lesk, A. M., ed., Oxford University Press, New York, 1988;
  • the percent identity between two nucleic acid sequences can be determined using the algorithm of Meyers and Miller (CABIOS, 1989, 4:11-17), which has been incorporated 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 identity between two nucleic acid sequences can, alternatively, be determined using the GAP program in the GCG software package 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 programs.
  • Exemplary computer software to determine homology between two sequences include, but are not limited to, GCG program package, Devereux, J., et al., Nucleic Acids Research, 12(1), 387 (1984)), BLASTP, BLASTN, and FASTA Altschul, S. F. et al., J.
  • RNA (e.g., mRNA) treatments of the present disclosure comprise at least one ribonucleic acid (RNA) polynucleotide having an open reading frame encoding at least one binding polypeptide that comprises at least one chemical modification.
  • RNA ribonucleic acid
  • compositions of the present disclosure comprise, in some embodiments, at least one nucleic acid (e.g., RNA) having an open reading frame encoding at least one antibody, wherein the nucleic acid comprises nucleotides and/or nucleosides that can be standard (unmodified) or modified as is known in the art.
  • nucleotides and nucleosides of the present disclosure comprise modified nucleotides or nucleosides.
  • modified nucleotides and nucleosides can be naturally-occurring modified nucleotides and nucleosides or non-naturally occurring modified nucleotides and nucleosides.
  • modifications can include those at the sugar, backbone, or nucleobase portion of the nucleotide and/or nucleoside as are recognized in the art.
  • a naturally-occurring modified nucleotide or nucleotide of the disclosure is one as is generally known or recognized in the art.
  • Non-limiting examples of such naturally occurring modified nucleotides and nucleotides can be found, inter alia, in the widely recognized MODOMICS database.
  • a non-naturally occurring modified nucleotide or nucleoside of the disclosure is one as is generally known or recognized in the art.
  • Non-limiting examples of such non-naturally occurring modified nucleotides and nucleosides can be found, inter alia, in published US application Nos. PCT/US2012/058519; PCT/US2013/075177;
  • RNA e.g., mRNA
  • nucleotides and nucleosides of the present disclosure comprise standard nucleoside residues such as those present in transcribed RNA (e.g. A, G, C, or U).
  • nucleotides and nucleosides of the present disclosure comprise standard deoxyribonucleosides such as those present in DNA (e.g. dA, dG, dC, or dT).
  • nucleic acids of the disclosure can comprise standard nucleotides and nucleosides, naturally-occurring nucleotides and nucleosides, non-naturally-occurring nucleotides and nucleosides, or any combination thereof.
  • Nucleic acids of the disclosure e.g., DNA nucleic acids and RNA nucleic acids, such as mRNA nucleic acids
  • nucleic acids of the disclosure in some embodiments, comprise various (more than one) different types of standard and/or modified nucleotides and nucleosides.
  • a particular region of a nucleic acid contains one, two or more (optionally different) types of standard and/or modified nucleotides and nucleosides.
  • a modified RNA nucleic acid e.g., a modified mRNA nucleic acid
  • introduced to a cell or organism exhibits reduced degradation in the cell or organism, respectively, relative to an unmodified nucleic acid comprising standard nucleotides and nucleosides.
  • a modified RNA nucleic acid (e.g., a modified mRNA nucleic acid), introduced into a cell or organism, may exhibit reduced immunogenicity in the cell or organism, respectively (e.g., a reduced innate response) relative to an unmodified nucleic acid comprising standard nucleotides and nucleosides.
  • Nucleic acids e.g., RNA nucleic acids, such as mRNA nucleic acids
  • Nucleic acids in some embodiments, comprise non-natural modified nucleotides that are introduced during synthesis or post-synthesis of the nucleic acids to achieve desired functions or properties.
  • the modifications may be present on internucleotide linkages, purine or pyrimidine bases, or sugars.
  • the modification may be introduced with chemical synthesis or with a polymerase enzyme at the terminal of a chain or anywhere else in the chain. Any of the regions of a nucleic acid may be chemically modified.
  • nucleic acid e.g., RNA nucleic acids, such as mRNA nucleic acids.
  • A“nucleoside” refers to a compound containing a sugar molecule (e.g., a pentose or ribose) or a derivative thereof in combination with an organic base (e.g., a purine or pyrimidine) or a derivative thereof (also referred to herein as“nucleobase”).
  • A“nucleotide” refers to a nucleoside, including a phosphate group.
  • Modified nucleotides may by synthesized by any useful method, such as, for example, chemically, enzymatically, or recombinantly, to include one or more modified or non-natural nucleosides.
  • Nucleic acids can comprise a region or regions of linked nucleosides. Such regions may have variable backbone linkages. The linkages can be standard phosphodiester linkages, in which case the nucleic acids would comprise regions of nucleotides.
  • Modified nucleotide base pairing encompasses not only the standard adenosine- thymine, adenosine-uracil, or guanosine-cytosine base pairs, but also base pairs formed between nucleotides and/or modified nucleotides comprising non-standard or modified bases, wherein the arrangement of hydrogen bond donors and hydrogen bond acceptors permits hydrogen bonding between a non-standard base and a standard base or between two complementary non-standard base structures, such as, for example, in those nucleic acids having at least one chemical modification.
  • non-standard base pairing is the base pairing between the modified nucleotide inosine and adenine, cytosine or uracil. Any combination of base/sugar or linker may be incorporated into nucleic acids of the present disclosure.
  • modified nucleobases in nucleic acids comprise 1-methyl-pseudouridine (m1 ⁇ ), 1-ethyl- pseudouridine (e1 ⁇ ), 5-methoxy-uridine (mo5U), 5-methyl-cytidine (m5C), and/or pseudouridine ( ⁇ ).
  • modified nucleobases in nucleic acids e.g., RNA nucleic acids, such as mRNA nucleic acids
  • the polyribonucleotide includes a combination of at least two (e.g., 2, 3, 4 or more) of any of the aforementioned modified nucleobases, including but not limited to chemical modifications.
  • a RNA nucleic acid of the disclosure comprises 1-methyl- pseudouridine substitutions at one or more or all uridine positions of the nucleic acid.
  • a RNA nucleic acid of the disclosure comprises 1-methyl- pseudouridine substitutions at one or more or all uridine positions of the nucleic acid and 5-methyl cytidine substitutions at one or more or all cytidine positions of the nucleic acid.
  • a RNA nucleic acid of the disclosure comprises pseudouridine ( ⁇ ) substitutions at one or more or all uridine positions of the nucleic acid.
  • a RNA nucleic acid of the disclosure comprises pseudouridine ( ⁇ ) substitutions at one or more or all uridine positions of the nucleic acid and 5-methyl cytidine substitutions at one or more or all cytidine positions of the nucleic acid.
  • a RNA nucleic acid of the disclosure comprises uridine at one or more or all uridine positions of the nucleic acid.
  • nucleic acids e.g., RNA nucleic acids, such as mRNA nucleic acids
  • RNA nucleic acids are uniformly modified (e.g., fully modified, modified throughout the entire sequence) for a particular modification.
  • a nucleic acid can be uniformly modified with 1- methyl-pseudouridine, meaning that all uridine residues in the mRNA sequence are replaced with 1-methyl-pseudouridine.
  • a nucleic acid can be uniformly modified for any type of nucleoside residue present in the sequence by replacement with a modified residue such as those set forth above.
  • nucleic acids of the present disclosure may be partially or fully modified along the entire length of the molecule.
  • one or more or all or a given type of nucleotide e.g., purine or pyrimidine, or any one or more or all of A, G, U, C
  • nucleotides X in a nucleic acid of the present disclosure are modified nucleotides, wherein X may be any one of nucleotides A, G, U, C, or any one of the combinations A+G, A+U, A+C, G+U, G+C, U+C, A+G+U, A+G+C, G+U+C or A+G+C.
  • the nucleic acid may contain from about 1% to about 100% modified nucleotides (either in relation 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 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
  • the nucleic acids may contain at a minimum 1% 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 nucleotides, or at least 90% modified nucleotides.
  • the nucleic acids may contain a modified pyrimidine such as a modified uracil or cytosine.
  • 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 replaced with a modified uracil (e.g., a 5-substituted uracil).
  • the modified 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).
  • cytosine in the nucleic acid is replaced with a modified cytosine (e.g., a 5-substituted 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).
  • Antibodies and antigen binding fragments thereof of the present disclosure comprise at least one RNA polynucleotide, such as an mRNA (e.g., modified mRNA).
  • mRNA e.g., modified mRNA
  • mRNA is transcribed in vitro from template DNA, referred to as an“in vitro transcription template.”
  • an in vitro transcription template encodes a 5′ untranslated (UTR) region, contains an open reading frame, and encodes a 3′ UTR and a polyA tail.
  • UTRs Untranslated Regions
  • Translation of a polynucleotide comprising an open reading frame encoding a polypeptide can be controlled and regulated by a variety of mechanisms that are provided by various cis-acting nucleic acid structures.
  • cis-acting RNA elements that form hairpins or other higher-order (e.g., pseudoknot) intramolecular mRNA secondary structures can provide a translational regulatory activity to a polynucleotide, wherein the RNA element influences or modulates the initiation of polynucleotide translation, particularly when the RNA element is positioned in the 5′ UTR close to the 5’-cap structure (Pelletier and Sonenberg (1985) Cell 40(3):515-526; Kozak (1986) Proc Natl Acad Sci 83:2850-2854).
  • Untranslated regions are nucleic acid sections of a polynucleotide before a start codon (5' UTR) and after a stop codon (3' UTR) that are not translated.
  • a polynucleotide e.g., a ribonucleic acid (RNA), e.g., a messenger RNA (mRNA)
  • RNA e.g., a messenger RNA (mRNA)
  • ORF open reading frame
  • UTR e.g., a 5′UTR or functional fragment thereof, a 3′UTR or functional fragment thereof, or a combination thereof.
  • Cis-acting RNA elements can also affect translation elongation, being involved in numerous frameshifting events (Namy et al., (2004) Mol Cell 13(2):157-168).
  • Internal ribosome entry sequences represent another type of cis-acting RNA element that are typically located in 5′ UTRs, but have also been reported to be found within the coding region of naturally-occurring mRNAs (Holcik et al. (2000) Trends Genet 16(10):469-473).
  • IRES In cellular mRNAs, IRES often coexist with the 5′-cap structure and provide mRNAs with the functional capacity to be translated under conditions in which cap-dependent translation is compromised (Gebauer et al., (2012) Cold Spring Harb Perspect Biol 4(7):a012245).
  • Another type of naturally-occurring cis-acting RNA element comprises upstream open reading frames (uORFs).
  • uORFs Naturally-occurring uORFs occur singularly or multiply within the 5′ UTRs of numerous mRNAs and influence the translation of the downstream major ORF, usually negatively (with the notable exception of GCN4 mRNA in yeast and ATF4 mRNA in mammals, where uORFs serve to promote the translation of the downstream major ORF under conditions of increased eIF2 phosphorylation (Hinnebusch (2005) Annu Rev Microbiol 59:407-450)). Additional exemplary translational regulatory activities provided by components, structures, elements, motifs, and/or specific sequences comprising
  • polynucleotides include, but are not limited to, mRNA stabilization or destabilization (Baker & Parker (2004) Curr Opin Cell Biol 16(3):293-299), translational activation (Villalba et al., (2011) Curr Opin Genet Dev 21(4):452-457), and translational repression (Blumer et al., (2002) Mech Dev 110(1-2):97-112). Studies have shown that naturally-occurring, cis-acting RNA elements can confer their respective functions when used to modify, by incorporation into, heterologous polynucleotides (Goldberg-Cohen et al., (2002) J Biol Chem 277(16):13635-13640).
  • a UTR can be homologous or heterologous to the coding region in a polynucleotide.
  • the UTR is homologous to the ORF encoding the antibody.
  • the UTR is heterologous to the ORF encoding the antibody.
  • the polynucleotide comprises two or more 5′UTRs or functional fragments thereof, each of which have the same or different nucleotide sequences.
  • the polynucleotide comprises two or more 3′UTRs or functional fragments thereof, each of which have the same or different nucleotide sequences.
  • the 5′UTR or functional fragment thereof, 3′ UTR or functional fragment thereof, or any combination thereof is sequence optimized.
  • the 5′UTR or functional fragment thereof, 3′ UTR or functional fragment thereof, or any combination thereof comprises at least one chemically modified nucleobase, e.g., 5-methoxyuracil.
  • UTRs can have features that provide a regulatory role, e.g., increased or decreased stability, localization and/or translation efficiency.
  • a polynucleotide comprising a UTR can be administered to a cell, tissue, or organism, and one or more regulatory features can be measured using routine methods.
  • a functional fragment of a 5'UTR or 3'UTR comprises one or more regulatory features of a full length 5' or 3' UTR, respectively.
  • Natural 5′UTRs bear features that play roles in translation initiation. They harbor signatures like Kozak sequences that are commonly known to be involved in the process by which the ribosome initiates translation of many genes.
  • Kozak sequences have the consensus CCR(A/G)CCAUGG, where R is a purine (adenine or guanine) three bases upstream of the start codon (AUG), which is followed by another 'G'.5′UTRs also have been known to form secondary structures that are involved in elongation factor binding.
  • liver-expressed mRNA such as albumin, serum amyloid A, Apolipoprotein A/B/E, transferrin, alpha fetoprotein, erythropoietin, or antibody, can enhance expression of polynucleotides in hepatic cell lines or liver.
  • 5′UTR from other tissue-specific mRNA to improve expression in that tissue is possible for muscle (e.g., MyoD, Myosin, Myoglobin, Myogenin, Herculin), for endothelial cells (e.g., Tie-1, CD36), for myeloid cells (e.g., C/EBP, AML1, G-CSF, GM-CSF, CD11b, MSR, Fr-1, i- NOS), for leukocytes (e.g., CD45, CD18), for adipose tissue (e.g., CD36, GLUT4, ACRP30, adiponectin) and for lung epithelial cells (e.g., SP-A/B/C/D).
  • muscle e.g., MyoD, Myosin, Myoglobin, Myogenin, Herculin
  • endothelial cells e.g., Tie-1, CD36
  • myeloid cells e.g., C/E
  • UTRs are selected from a family of transcripts whose proteins share a common function, structure, feature or property.
  • an encoded polypeptide can belong to a family of proteins (i.e., that share at least one function, structure, feature, localization, origin, or expression pattern), which are expressed in a particular cell, tissue or at some time during development.
  • the UTRs from any of the genes or mRNA can be swapped for any other UTR of the same or different family of proteins to create a new polynucleotide.
  • the 5’UTR and the 3’UTR can be heterologous.
  • the 5'UTR can be derived from a different species than the 3'UTR.
  • the 3'UTR can be derived from a different species than the 5'UTR.
  • Exemplary UTRs of the application include, but are not limited to, one or more 5′UTR and/or 3′UTR derived from the nucleic acid sequence of: a globin, such as an ⁇ - or ⁇ - globin (e.g., a Xenopus, mouse, rabbit, or human globin); a strong Kozak translational initiation signal; a CYBA (e.g., human cytochrome b-245 ⁇ polypeptide); an albumin (e.g., human albumin7); a HSD17B4 (hydroxysteroid (17- ⁇ ) dehydrogenase); a virus (e.g., a tobacco etch virus (TEV), a Venezuelan equine encephalitis virus (VEEV), a Dengue virus, a cytomegalovirus (CMV) (e.g., CMV immediate early 1 (IE1)), a hepatitis virus (e.g., hepatitis B virus), a Sindbis virus
  • Col6A1 a ribophorin (e.g., ribophorin I (RPNI)); a low density lipoprotein receptor-related protein (e.g., LRP1); a cardiotrophin-like cytokine factor (e.g., Nnt1); calreticulin (Calr); a procollagen-lysine, 2-oxoglutarate 5-dioxygenase 1 (Plod1); and a nucleobindin (e.g., Nucb1).
  • RPNI ribophorin I
  • LRP1 low density lipoprotein receptor-related protein
  • LRP1 low density lipoprotein receptor-related protein
  • a cardiotrophin-like cytokine factor e.g., Nnt1
  • Calr calreticulin
  • Plod1 2-oxoglutarate 5-dioxygenase 1
  • Nucb1 nucleobindin
  • the 5'UTR is selected from the group consisting of a ⁇ -globin 5’UTR; a 5′UTR containing a strong Kozak translational initiation signal; a cytochrome b- 245 ⁇ polypeptide (CYBA) 5'UTR; a hydroxysteroid (17- ⁇ ) dehydrogenase (HSD17B4) 5'UTR; a Tobacco etch virus (TEV) 5'UTR; a Vietnamese etch virus (TEV) 5'UTR; a decielen equine encephalitis virus (TEEV) 5'UTR; a 5' proximal open reading frame of rubella virus (RV) RNA encoding nonstructural proteins; a Dengue virus (DEN) 5'UTR; a heat shock protein 70 (Hsp70) 5'UTR; a eIF4G 5'UTR; a GLUT15'UTR; functional fragments thereof and any combination thereof.
  • CYBA cytochrome b-
  • the 3'UTR is selected from the group consisting of a ⁇ -globin 3’UTR; a CYBA 3'UTR; an albumin 3'UTR; a growth hormone (GH) 3'UTR; a VEEV 3'UTR; a hepatitis B virus (HBV) 3'UTR; ⁇ -globin 3′UTR; a DEN 3'UTR; a PAV barley yellow dwarf virus (BYDV-PAV) 3'UTR; an elongation factor 1 ⁇ 1 (EEF1A1) 3'UTR; a manganese superoxide dismutase (MnSOD) 3'UTR; a ⁇ subunit of mitochondrial H(+)-ATP synthase ( ⁇ -mRNA) 3'UTR; a GLUT13'UTR; a MEF2A 3'UTR; a ⁇ -F1-ATPase 3'UTR; functional fragments thereof and combinations thereof.
  • Wild-type UTRs derived from any gene or mRNA can be incorporated into the polynucleotides of the present disclosure.
  • a UTR can be altered relative to a wild type or native UTR to produce a variant UTR, e.g., by changing the orientation or location of the UTR relative to the ORF; or by inclusion of additional nucleotides, deletion of nucleotides, swapping or transposition of nucleotides.
  • variants of 5′ or 3′ UTRs can be utilized, for example, mutants of wild type UTRs, or variants wherein one or more nucleotides are added to or removed from a terminus of the UTR.
  • one or more synthetic UTRs can be used in combination with one or more non-synthetic UTRs.
  • UTRs or portions thereof can be placed in the same orientation as in the transcript from which they were selected or can be altered in orientation or location. Hence, a 5′ and/or 3′ UTR can be inverted, shortened, lengthened, or combined with one or more other 5′ UTRs or 3′ UTRs.
  • the polynucleotide comprises multiple UTRs, e.g., a double, a triple or a quadruple 5’UTR or 3’UTR.
  • a double UTR comprises two copies of the same UTR either in series or substantially in series.
  • a double beta-globin 3′UTR can be used (see US2010/0129877, the contents of which are incorporated herein by reference in its entirety).
  • the polynucleotides of the present disclosure comprise a 5'UTR and/or a 3'UTR selected from any of the UTRs disclosed herein.
  • the 5'UTR and/or 3'UTR sequence of the present disclosure comprises a nucleotide sequence at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99%, or about 100% identical to a sequence selected from the group consisting of 5'UTR sequences comprising any of SEQ ID NOs: 71-88, 160, and 162 and/or 3'UTR sequences comprises any of SEQ ID NOs: 89-99, 161 and 163, and any combination thereof.
  • the polynucleotides of the present disclosure can comprise combinations of features.
  • the ORF can be flanked by a 5′UTR that comprises a strong Kozak translational initiation signal and/or a 3′UTR comprising an oligo(dT) sequence for templated addition of a poly-A tail.
  • a 5′UTR can comprise a first polynucleotide fragment and a second
  • polynucleotide fragment from the same and/or different UTRs (see, e.g., US2010/0293625, herein incorporated by reference in its entirety).
  • polynucleotides of the present disclosure For example, introns or portions of intron sequences can be incorporated into the polynucleotides of the present disclosure.
  • the polynucleotide of the present disclosure comprises an internal ribosome entry site (IRES) instead of or in addition to a UTR (see, e.g., Yakubov et al., Biochem. Biophys. Res. Commun.2010394(1):189-193, the contents of which are incorporated herein by reference in their entirety).
  • IRES internal ribosome entry site
  • the polynucleotide comprises an IRES instead of a 5’UTR sequence.
  • the polynucleotide comprises an ORF and a viral capsid sequence.
  • the polynucleotide comprises a synthetic 5'UTR in combination with a non-synthetic 3'UTR.
  • the UTR can also include at least one translation enhancer polynucleotide, translation enhancer element, or translational enhancer elements
  • TEE refers to nucleic acid sequences that increase the amount of polypeptide or protein produced from a polynucleotide.
  • the TEE can be located between the transcription promoter and the start codon.
  • the 5'UTR comprises a TEE.
  • a 5′UTR and/or 3'UTR comprising at least one TEE described herein can be incorporated in a monocistronic sequence such as, but not limited to, a vector system or a nucleic acid vector.
  • a 5′UTR and/or 3′UTR of a polynucleotide of the present disclosure comprises a TEE or portion thereof described herein.
  • the TEEs in the 3′UTR can be the same and/or different from the TEE located in the 5′UTR.
  • a 5'UTR and/or 3'UTR of a polynucleotide of the present disclosure can include at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18 at least 19, at least 20, at least 21, at least 22, at least 23, at least 24, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, at least 55 or more than 60 TEE sequences.
  • the 5′UTR of a polynucleotide of the present disclosure can include 1-60, 1-55, 1-50, 1-45, 1-40, 1-35, 1-30, 1-25, 1-20, 1-15, 1- 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 TEE sequences.
  • the TEE sequences in the 5′UTR of the polynucleotide of the present disclosure can be the same or different TEE sequences.
  • a combination of different TEE sequences in the 5′UTR of the polynucleotide of the present disclosure can include combinations in which more than one copy of any of the different TEE sequences are incorporated.
  • the 5′UTR and/or 3'UTR comprises a spacer to separate two TEE sequences.
  • the spacer can be a 15 nucleotide spacer and/or other spacers known in the art.
  • the 5′UTR and/or 3'UTR comprises a TEE sequence-spacer module repeated at least once, at least twice, at least 3 times, at least 4 times, at least 5 times, at least 6 times, at least 7 times, at least 8 times, at least 9 times, at least 10 times, or more than 10 times in the 5′UTR and/or 3'UTR, respectively.
  • the 5′UTR and/or 3'UTR comprises a TEE sequence- spacer module repeated 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 times.
  • the spacer separating two TEE sequences can include other sequences known in the art that can regulate the translation of the polynucleotide of the present disclosure, e.g., miR binding site sequences described herein (e.g., miR binding sites and miR seeds).
  • miR binding site sequences described herein e.g., miR binding sites and miR seeds.
  • each spacer used to separate two TEE sequences can include a different miR binding site sequence or component of a miR sequence (e.g., miR seed sequence).
  • the present disclosure provides synthetic polynucleotides comprising a modification (e.g., an RNA element), wherein the modification provides a desired translational regulatory activity.
  • the disclosure provides a polynucleotide comprising a 5’ untranslated region (UTR), an initiation codon, a full open reading frame encoding a polypeptide, a 3’ UTR, and at least one modification, wherein the at least one modification provides a desired translational regulatory activity, for example, a modification that promotes and/or enhances the translational fidelity of mRNA translation.
  • the desired translational regulatory activity is a cis-acting regulatory activity.
  • the desired translational regulatory activity is an increase in the residence time of the 43S pre-initiation complex (PIC) or ribosome at, or proximal to, the initiation codon. In some embodiments, the desired translational regulatory activity is an increase in the initiation of polypeptide synthesis at or from the initiation codon. In some embodiments, the desired translational regulatory activity is an increase in the amount of polypeptide translated from the full open reading frame. In some embodiments, the desired translational regulatory activity is an increase in the fidelity of initiation codon decoding by the PIC or ribosome. In some embodiments, the desired translational regulatory activity is inhibition or reduction of leaky scanning by the PIC or ribosome.
  • the desired translational regulatory activity is a decrease in the rate of decoding the initiation codon by the PIC or ribosome. In some embodiments, the desired translational regulatory activity is inhibition or reduction in the initiation of polypeptide synthesis at any codon within the mRNA other than the initiation codon. In some embodiments, the desired translational regulatory activity is inhibition or reduction of the amount of polypeptide translated from any open reading frame within the mRNA other than the full open reading frame. In some embodiments, the desired translational regulatory activity is inhibition or reduction in the production of aberrant translation products. In some embodiments, the desired translational regulatory activity is a combination of one or more of the foregoing translational regulatory activities.
  • the present disclosure provides a polynucleotide, e.g., an mRNA, comprising an RNA element that comprises a sequence and/or an RNA secondary structure(s) that provides a desired translational regulatory activity as described herein.
  • the mRNA comprises an RNA element that comprises a sequence and/or an RNA secondary structure(s) that promotes and/or enhances the translational fidelity of mRNA translation.
  • the mRNA comprises an RNA element that comprises a sequence and/or an RNA secondary structure(s) that provides a desired translational regulatory activity, such as inhibiting and/or reducing leaky scanning.
  • the disclosure provides an mRNA that comprises an RNA element that comprises a sequence and/or an RNA secondary structure(s) that inhibits and/or reduces leaky scanning thereby promoting the translational fidelity of the mRNA.
  • the RNA element comprises natural and/or modified nucleotides. In some embodiments, the RNA element comprises of a sequence of linked nucleotides, or derivatives or analogs thereof, that provides a desired translational regulatory activity as described herein. In some embodiments, the RNA element comprises a sequence of linked nucleotides, or derivatives or analogs thereof, that forms or folds into a stable RNA secondary structure, wherein the RNA secondary structure provides a desired translational regulatory activity as described herein.
  • RNA elements can be identified and/or characterized based on the primary sequence of the element (e.g., GC-rich element), by RNA secondary structure formed by the element (e.g.
  • RNA molecules e.g., located within the 5’ UTR of an mRNA
  • biological function and/or activity of the element e.g.,“translational enhancer element”
  • the disclosure provides an mRNA having one or more structural modifications that inhibits leaky scanning and/or promotes the translational fidelity of mRNA translation, wherein at least one of the structural modifications is a GC-rich RNA element. In some aspects, the disclosure provides a modified mRNA comprising at least one
  • the GC-rich RNA element comprising a sequence of linked nucleotides, or derivatives or analogs thereof, preceding a Kozak consensus sequence in a 5’ UTR of the mRNA.
  • the GC-rich RNA element is located about 30, about 25, about 20, about 15, about 10, about 5, about 4, about 3, about 2, or about 1 nucleotide(s) upstream of a Kozak consensus sequence in the 5’ UTR of the mRNA.
  • the GC-rich RNA element is located 15-30, 15-20, 15- 25, 10-15, or 5-10 nucleotides upstream of a Kozak consensus sequence.
  • the GC-rich RNA element is located immediately adjacent to a Kozak consensus sequence in the 5’ UTR of the mRNA.
  • the disclosure provides a GC-rich RNA element which comprises a sequence of 3-30, 5-25, 10-20, 15-20, about 20, about 15, about 12, about 10, about 7, about 6 or about 3 nucleotides, derivatives or analogs thereof, linked in any order, wherein the sequence composition is 70-80% cytosine, 60-70% cytosine, 50%- 60% cytosine, 40-50% cytosine, 30-40% cytosine bases.
  • the disclosure provides a GC-rich RNA element which comprises a sequence of 3- 30, 5-25, 10-20, 15-20, about 20, about 15, about 12, about 10, about 7, about 6 or about 3 nucleotides, derivatives or analogs thereof, linked in any order, wherein the sequence composition is about 80% cytosine, about 70% cytosine, about 60% cytosine, about 50% cytosine, about 40% cytosine, or about 30% cytosine.
  • the disclosure provides a GC-rich RNA element which comprises a sequence of 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, or 3 nucleotides, or derivatives or analogs thereof, linked in any order, wherein the sequence composition is 70-80% cytosine, 60-70% cytosine, 50%-60% cytosine, 40-50% cytosine, or 30-40% cytosine.
  • the disclosure provides a GC-rich RNA element which comprises a sequence of 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, or 3 nucleotides, or derivatives or analogs thereof, linked in any order, wherein the sequence composition is about 80% cytosine, about 70% cytosine, about 60% cytosine, about 50% cytosine, about 40% cytosine, or about 30% cytosine.
  • the disclosure provides a modified mRNA comprising at least one modification, wherein at least one modification is a GC-rich RNA element comprising a sequence of linked nucleotides, or derivatives or analogs thereof, preceding a Kozak consensus sequence in a 5’ UTR of the mRNA, wherein the GC-rich RNA element is located about 30, about 25, about 20, about 15, about 10, about 5, about 4, about 3, about 2, or about 1 nucleotide(s) upstream of a Kozak consensus sequence in the 5’ UTR of the mRNA, and wherein the GC-rich RNA element comprises a sequence of 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 nucleotides, or derivatives or analogs thereof, linked in any order, wherein the sequence composition is >50% cytosine.
  • at least one modification is a GC-rich RNA element comprising a sequence of linked nucleotides, or derivatives or analogs thereof, preceding a Kozak consensus sequence in
  • the sequence composition is >55% cytosine, >60% cytosine, >65% cytosine, >70% cytosine, >75% cytosine, >80% cytosine, >85% cytosine, or >90% cytosine.
  • the disclosure provides a modified mRNA comprising at least one modification, wherein at least one modification is a GC-rich RNA element comprising a sequence of linked nucleotides, or derivatives or analogs thereof, preceding a Kozak consensus sequence in a 5’ UTR of the mRNA, wherein the GC-rich RNA element comprises any one of the sequences set forth in Table 5.
  • the GC-rich RNA element is located about 30, about 25, about 20, about 15, about 10, about 5, about 4, about 3, about 2, or about 1 nucleotide(s) upstream of a Kozak consensus sequence in the 5’ UTR of the mRNA.
  • the GC-rich RNA element is located about 15-30, 15-20, 15-25, 10-15, or 5-10 nucleotides upstream of a Kozak consensus sequence. In another embodiment, the GC-rich RNA element is located immediately adjacent to a Kozak consensus sequence in the 5’ UTR of the mRNA.
  • the disclosure provides a modified mRNA comprising at least one modification, wherein at least one modification is a GC-rich RNA element comprising the sequence V1 [CCCCGGCGCC] (SEQ ID NO: 100) as set forth in Table 5, or derivatives or analogs thereof, preceding a Kozak consensus sequence in the 5’ UTR of the mRNA.
  • the GC-rich element comprises the sequence V1 as set forth in Table 5 located immediately adjacent to and upstream of the Kozak consensus sequence in the 5’ UTR of the mRNA.
  • the GC-rich element comprises the sequence V1 as set forth in Table 5 located 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 bases upstream of the Kozak consensus sequence in the 5’ UTR of the mRNA. In other embodiments, the GC-rich element comprises the sequence V1 as set forth in Table 5 located 1-3, 3-5, 5-7, 7-9, 9-12, or 12-15 bases upstream of the Kozak consensus sequence in the 5’ UTR of the mRNA.
  • the disclosure provides a modified mRNA comprising at least one modification, wherein at least one modification is a GC-rich RNA element comprising the sequence V2 [CCCCGGC] as set forth in Table 5, or derivatives or analogs thereof, preceding a Kozak consensus sequence in the 5’ UTR of the mRNA.
  • the GC-rich element comprises the sequence V2 as set forth in Table 5 located immediately adjacent to and upstream of the Kozak consensus sequence in the 5’ UTR of the mRNA.
  • the GC-rich element comprises the sequence V2 as set forth in Table 5 located 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 bases upstream of the Kozak consensus sequence in the 5’ UTR of the mRNA.
  • the GC-rich element comprises the sequence V2 as set forth in Table 5 located 1-3, 3-5, 5-7, 7-9, 9-12, or 12-15 bases upstream of the Kozak consensus sequence in the 5’ UTR of the mRNA.
  • the disclosure provides a modified mRNA comprising at least one modification, wherein at least one modification is a GC-rich RNA element comprising the sequence EK [GCCGCC] as set forth in Table 5, or derivatives or analogs thereof, preceding a Kozak consensus sequence in the 5’ UTR of the mRNA.
  • the GC- rich element comprises the sequence EK as set forth in Table 5 located immediately adjacent to and upstream of the Kozak consensus sequence in the 5’ UTR of the mRNA.
  • the GC-rich element comprises the sequence EK as set forth in Table 5 located 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 bases upstream of the Kozak consensus sequence in the 5’ UTR of the mRNA.
  • the GC-rich element comprises the sequence EK as set forth in Table 5 located 1-3, 3-5, 5-7, 7-9, 9-12, or 12-15 bases upstream of the Kozak consensus sequence in the 5’ UTR of the mRNA.
  • the disclosure provides a modified mRNA comprising at least one modification, wherein at least one modification is a GC-rich RNA element comprising the sequence V1 [CCCCGGCGCC] (SEQ ID NO: 100) as set forth in Table 5, or derivatives or analogs thereof, preceding a Kozak consensus sequence in the 5’ UTR of the mRNA, wherein the 5’ UTR comprises the following sequence shown in Table 5:
  • the GC-rich element comprises the sequence V1 as set forth in Table 5 located immediately adjacent to and upstream of the Kozak consensus sequence in the 5’ UTR sequence shown in Table 5. In some embodiments, the GC-rich element comprises the sequence V1 as set forth in Table 5 located 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 bases upstream of the Kozak consensus sequence in the 5’ UTR of the mRNA, wherein the 5’ UTR comprises the following sequence shown in Table 5:
  • the GC-rich element comprises the sequence V1 as set forth in Table 5 located 1-3, 3-5, 5-7, 7-9, 9-12, or 12-15 bases upstream of the Kozak consensus sequence in the 5’ UTR of the mRNA, wherein the 5’ UTR comprises the following sequence shown in Table 5:
  • the 5’ UTR comprises the following sequence set forth in Table 5:
  • the disclosure provides a modified mRNA comprising at least one modification, wherein at least one modification is a GC-rich RNA element comprising a stable RNA secondary structure comprising a sequence of nucleotides, or derivatives or analogs thereof, linked in an order which forms a hairpin or a stem-loop.
  • the stable RNA secondary structure is upstream of the Kozak consensus sequence.
  • the stable RNA secondary structure is located about 30, about 25, about 20, about 15, about 10, or about 5 nucleotides upstream of the Kozak consensus sequence.
  • the stable RNA secondary structure is located about 20, about 15, about 10 or about 5 nucleotides upstream of the Kozak consensus sequence.
  • the stable RNA secondary structure is located about 5, about 4, about 3, about 2, about 1 nucleotides upstream of the Kozak consensus sequence. In another embodiment, the stable RNA secondary structure is located about 15-30, about 15-20, about 15-25, about 10-15, or about 5-10 nucleotides upstream of the Kozak consensus sequence. In another embodiment, the stable RNA secondary structure is located 12-15 nucleotides upstream of the Kozak consensus sequence. In another embodiment, the stable RNA secondary structure has a deltaG of about -30 kcal/mol, about -20 to -30 kcal/mol, about -20 kcal/mol, about -10 to -20 kcal/mol, about -10 kcal/mol, about -5 to -10 kcal/mol.
  • the modification is operably linked to an open reading frame encoding a polypeptide and wherein the modification and the open reading frame are heterologous.
  • the sequence of the GC-rich RNA element is comprised exclusively of guanine (G) and cytosine (C) nucleobases.
  • RNA elements that provide a desired translational regulatory activity as described herein can be identified and characterized using known techniques, such as ribosome profiling .
  • Ribosome profiling is a technique that allows the determination of the positions of PICs and/or ribosomes bound to mRNAs (see e.g., Ingolia et al., (2009) Science 324(5924):218-23, incorporated herein by reference). The technique is based on protecting a region or segment of mRNA, by the PIC and/or ribosome, from nuclease digestion. Protection results in the generation of a 30-bp fragment of RNA termed a‘footprint’.
  • RNA footprints can be analyzed by methods known in the art (e.g., RNA-seq).
  • the footprint is roughly centered on the A-site of the ribosome. If the PIC or ribosome dwells at a particular position or location along an mRNA, footprints generated at these position would be relatively common. Studies have shown that more footprints are generated at positions where the PIC and/or ribosome exhibits decreased processivity and fewer footprints where the PIC and/or ribosome exhibits increased processivity (Gardin et al., (2014) eLife 3:e03735).
  • residence time or the time of occupancy of a the PIC or ribosome at a discrete position or location along an polynucleotide comprising any one or more of the RNA elements described herein is determined by ribosome profiling.
  • a polynucleotide of the present disclosure comprises a miR and/or TEE sequence.
  • the incorporation of a miR sequence and/or a TEE sequence into a polynucleotide of the present disclosure can change the shape of the stem loop region, which can increase and/or decrease translation. See e.g., Kedde et al., Nature Cell Biology 201012(10):1014-20, herein incorporated by reference in its entirety).
  • Polynucleotides of the present disclosure can include regulatory elements, for example, microRNA (miRNA) binding sites, transcription factor binding sites, structured mRNA sequences and/or motifs, artificial binding sites engineered to act as pseudo-receptors for endogenous nucleic acid binding molecules, and combinations thereof.
  • regulatory elements for example, microRNA (miRNA) binding sites, transcription factor binding sites, structured mRNA sequences and/or motifs, artificial binding sites engineered to act as pseudo-receptors for endogenous nucleic acid binding molecules, and combinations thereof.
  • polynucleotides including such regulatory elements are referred to as including “sensor sequences”.
  • a polynucleotide e.g., a ribonucleic acid (RNA), e.g., a messenger RNA (mRNA)
  • RNA ribonucleic acid
  • mRNA messenger RNA
  • ORF open reading frame
  • miRNA binding site(s) provides for regulation of polynucleotides of the present disclosure, and in turn, of the polypeptides encoded therefrom, based on tissue-specific and/or cell-type specific expression of naturally-occurring miRNAs.
  • a miRNA e.g., a natural-occurring miRNA
  • a miRNA sequence comprises a “seed” region, i.e., a sequence in the region of positions 2-8 of the mature miRNA.
  • a miRNA seed can comprise positions 2-8 or 2-7 of the mature miRNA.
  • microRNA (miRNA or miR) binding site refers to a sequence within a polynucleotide, e.g., within a DNA or within an RNA transcript, including in the 5′UTR and/or 3′UTR, that has sufficient complementarity to all or a region of a miRNA to interact with, associate with or bind to the miRNA.
  • a polynucleotide of the present disclosure comprising an ORF encoding a polypeptide of interest and further comprises one or more miRNA binding site(s).
  • a 5'UTR and/or 3'UTR of the polynucleotide e.g., a ribonucleic acid (RNA), e.g., a messenger RNA (mRNA)
  • RNA ribonucleic acid
  • mRNA messenger RNA
  • a miRNA binding site having sufficient complementarity to a miRNA refers to a degree of complementarity sufficient to facilitate miRNA-mediated regulation of a polynucleotide, e.g., miRNA-mediated translational repression or degradation of the polynucleotide.
  • a miRNA binding site having sufficient complementarity to the miRNA refers to a degree of complementarity sufficient to facilitate miRNA-mediated degradation of the polynucleotide, e.g., miRNA-guided RNA- induced silencing complex (RISC)-mediated cleavage of mRNA.
  • miRNA-guided RNA- induced silencing complex RISC
  • the miRNA binding site can have complementarity to, for example, a 19-25 nucleotide miRNA sequence, to a 19-23 nucleotide miRNA sequence, or to a 22 nucleotide miRNA sequence.
  • a miRNA binding site can be complementary to only a portion of a miRNA, e.g., to a portion less than 1, 2, 3, or 4 nucleotides of the full length of a naturally-occurring miRNA sequence.
  • Full or complete complementarity e.g., full complementarity or complete complementarity over all or a significant portion of the length of a naturally-occurring miRNA is preferred when the desired regulation is mRNA degradation.
  • a miRNA binding site includes a sequence that has
  • the miRNA binding site includes a sequence that has complete complementarity with a miRNA seed sequence.
  • a miRNA binding site includes a sequence that has complementarity (e.g., partial or complete complementarity) with an miRNA sequence.
  • the miRNA binding site includes a sequence that has complete complementarity with a miRNA sequence.
  • a miRNA binding site has complete complementarity with a miRNA sequence but for 1, 2, or 3 nucleotide substitutions, terminal additions, and/or truncations.
  • the miRNA binding site is the same length as the
  • the miRNA binding site is one, two, three, four, five, six, seven, eight, nine, ten, eleven or twelve nucleotide(s) shorter than the corresponding miRNA at the 5' terminus, the 3' terminus, or both.
  • the microRNA binding site is two nucleotides shorter than the corresponding microRNA at the 5' terminus, the 3' terminus, or both. The miRNA binding sites that are shorter than the corresponding miRNAs are still capable of degrading the mRNA incorporating one or more of the miRNA binding sites or preventing the mRNA from translation.
  • the miRNA binding site binds the corresponding mature miRNA that is part of an active RISC containing Dicer. In another embodiment, binding of the miRNA binding site to the corresponding miRNA in RISC degrades the mRNA containing the miRNA binding site or prevents the mRNA from being translated. In some embodiments, the miRNA binding site has sufficient complementarity to miRNA so that a RISC complex comprising the miRNA cleaves the polynucleotide comprising the miRNA binding site. In other embodiments, the miRNA binding site has imperfect complementarity so that a RISC complex comprising the miRNA induces instability in the polynucleotide comprising the miRNA binding site. In another embodiment, the miRNA binding site has imperfect complementarity so that a RISC complex comprising the miRNA represses transcription of the polynucleotide comprising the miRNA binding site.
  • the miRNA binding site has one, two, three, four, five, six, seven, eight, nine, ten, eleven or twelve mismatch(es) from the corresponding miRNA.
  • the miRNA binding site has at least about ten, at least about eleven, at least about twelve, at least about thirteen, at least about fourteen, at least about fifteen, at least about sixteen, at least about seventeen, at least about eighteen, at least about nineteen, at least about twenty, or at least about twenty-one contiguous nucleotides complementary to at least about ten, at least about eleven, at least about twelve, at least about thirteen, at least about fourteen, at least about fifteen, at least about sixteen, at least about seventeen, at least about eighteen, at least about nineteen, at least about twenty, or at least about twenty-one, respectively, contiguous nucleotides of the corresponding miRNA.
  • the polynucleotide By engineering one or more miRNA binding sites into a polynucleotide of the present disclosure, the polynucleotide can be targeted for degradation or reduced translation, provided the miRNA in question is available. This can reduce off-target effects upon delivery of the polynucleotide. For example, if a polynucleotide of the present disclosure is not intended to be delivered to a tissue or cell but ends up is said tissue or cell, then a miRNA abundant in the tissue or cell can inhibit the expression of the gene of interest if one or multiple binding sites of the miRNA are engineered into the 5′UTR and/or 3′UTR of the polynucleotide.
  • miRNA binding sites can be removed from polynucleotide sequences in which they naturally occur in order to increase protein expression in specific tissues.
  • a binding site for a specific miRNA can be removed from a polynucleotide to improve protein expression in tissues or cells containing the miRNA.
  • a polynucleotide of the present disclosure can include at least one miRNA-binding site in the 5'UTR and/or 3′UTR in order to regulate cytotoxic or
  • cytoprotective mRNA therapeutics to specific cells such as, but not limited to, normal and/or cancerous cells.
  • a polynucleotide of the present disclosure can include two, three, four, five, six, seven, eight, nine, ten, or more miRNA-binding sites in the 5'-UTR and/or 3′-UTR in order to regulate cytotoxic or cytoprotective mRNA therapeutics to specific cells such as, but not limited to, normal and/or cancerous cells.
  • Regulation of expression in multiple tissues can be accomplished through introduction or removal of one or more miRNA binding sites, e.g., one or more distinct miRNA binding sites.
  • the decision whether to remove or insert a miRNA binding site can be made based on miRNA expression patterns and/or their profilings in tissues and/or cells in development and/or disease. Identification of miRNAs, miRNA binding sites, and their expression patterns and role in biology have been reported (e.g., Bonauer et al., Curr Drug Targets 2010 11:943-949; Anand and Cheresh Curr Opin Hematol 201118:171-176; Contreras and Rao Leukemia 201226:404-413 (2011 Dec 20. doi: 10.1038/leu.2011.356); Bartel Cell 2009 136:215-233; Landgraf et al, Cell, 2007129:1401-1414; Gentner and Naldini, Tissue
  • tissues where miRNA are known to regulate mRNA, and thereby protein expression include, but are not limited to, liver (miR-122), muscle (miR-133, miR-206, miR- 208), endothelial cells (miR-17-92, miR-126), myeloid cells (miR-142-3p, miR-142-5p, miR- 16, miR-21, miR-223, miR-24, miR-27), adipose tissue (let-7, miR-30c), heart (miR-1d, miR- 149), kidney (miR-192, miR-194, miR-204), and lung epithelial cells (let-7, miR-133, miR- 126).
  • liver miR-122
  • muscle miR-133, miR-206, miR- 208
  • endothelial cells miR-17-92, miR-126
  • myeloid cells miR-142-3p, miR-142-5p, miR- 16, miR-21, miR
  • miRNAs are known to be differentially expressed in immune cells (also called hematopoietic cells), such as antigen presenting cells (APCs) (e.g., dendritic cells and macrophages), macrophages, monocytes, B lymphocytes, T lymphocytes, granulocytes, natural killer cells, etc.
  • APCs antigen presenting cells
  • Immune cell specific miRNAs are involved in immunogenicity, autoimmunity, the immune-response to infection, inflammation, as well as unwanted immune response after gene therapy and tissue/organ transplantation. Immune cells specific miRNAs also regulate many aspects of development, proliferation, differentiation and apoptosis of hematopoietic cells (immune cells).
  • miR-142 and miR-146 are exclusively expressed in immune cells, particularly abundant in myeloid dendritic cells. It has been demonstrated that the immune response to a polynucleotide can be shut-off by adding miR- 142 binding sites to the 3′-UTR of the polynucleotide, enabling more stable gene transfer in tissues and cells.
  • miR-142 efficiently degrades exogenous polynucleotides in antigen presenting cells and suppresses cytotoxic elimination of transduced cells (e.g., Annoni A et al., blood, 2009, 114, 5152-5161; Brown BD, et al., Nat med.2006, 12(5), 585-591; Brown BD, et al., blood, 2007, 110(13): 4144-4152, each of which is incorporated herein by reference in its entirety).
  • An antigen-mediated immune response can refer to an immune response triggered by foreign antigens, which, when entering an organism, are processed by the antigen presenting cells and displayed on the surface of the antigen presenting cells. T cells can recognize the presented antigen and induce a cytotoxic elimination of cells that express the antigen.
  • Introducing a miR-142 binding site into the 5'UTR and/or 3′UTR of a polynucleotide of the present disclosure can selectively repress gene expression in antigen presenting cells through miR-142 mediated degradation, limiting antigen presentation in antigen presenting cells (e.g., dendritic cells) and thereby preventing antigen-mediated immune response after the delivery of the polynucleotide.
  • the polynucleotide is then stably expressed in target tissues or cells without triggering cytotoxic elimination.
  • binding sites for miRNAs that are known to be expressed in immune cells can be engineered into a polynucleotide of the present disclosure to suppress the expression of the polynucleotide in antigen presenting cells through miRNA mediated RNA degradation, subduing the antigen-mediated immune response. Expression of the polynucleotide is maintained in non-immune cells where the immune cell specific miRNAs are not expressed.
  • any miR- 122 binding site can be removed and a miR-142 (and/or mirR-146) binding site can be engineered into the 5'UTR and/or 3'UTR of a polynucleotide of the present disclosure.
  • a polynucleotide of the present disclosure can include a further negative regulatory element in the 5'UTR and/or 3'UTR, either alone or in combination with miR-142 and/or miR-146 binding sites.
  • the further negative regulatory element is a Constitutive Decay Element (CDE).
  • Immune cell specific miRNAs include, but are not limited to, hsa-let-7a-2-3p, hsa-let- 7a-3p, hsa-7a-5p, hsa-let-7c, hsa-let-7e-3p, hsa-let-7e-5p, hsa-let-7g-3p, hsa-let-7g-5p, hsa- let-7i-3p, hsa-let-7i-5p, miR-10a-3p, miR-10a-5p, miR-1184, hsa-let-7f-1--3p, hsa-let-7f-2--- 5p, hsa-let-7f-5p, miR-125b-1-3p, miR-125b-2-3p, miR-125b-5p, miR-1279, miR-130a-3p, miR-130a-5p, miR-132-3p, miR-132-5p, miR-142-3p, miR-142-5
  • novel miRNAs can be identified in immune cell through micro-array hybridization and microtome analysis (e.g., Jima DD et al, Blood, 2010, 116:e118-e127; Vaz C et al., BMC Genomics, 2010, 11,288, the content of each of which is incorporated herein by reference in its entirety.)
  • miRNAs that are known to be expressed in the liver include, but are not limited to, miR-107, miR-122-3p, miR-122-5p, miR-1228-3p, miR-1228-5p, miR-1249, miR-129-5p, miR-1303, miR-151a-3p, miR-151a-5p, miR-152, miR-194-3p, miR-194-5p, miR-199a-3p, miR-199a-5p, miR-199b-3p, miR-199b-5p, miR-296-5p, miR-557, miR-581, miR-939-3p, and miR-939-5p.
  • MiRNA binding sites from any liver specific miRNA can be introduced to or removed from a polynucleotide of the present disclosure to regulate expression of the polynucleotide in the liver.
  • Liver specific miRNA binding sites can be engineered alone or further in combination with immune cell (e.g., APC) miRNA binding sites in a
  • miRNAs that are known to be expressed in the lung include, but are not limited to, let- 7a-2-3p, let-7a-3p, let-7a-5p, miR-126-3p, miR-126-5p, miR-127-3p, miR-127-5p, miR- 130a-3p, miR-130a-5p, miR-130b-3p, miR-130b-5p, miR-133a, miR-133b, miR-134, miR- 18a-3p, miR-18a-5p, miR-18b-3p, miR-18b-5p, miR-24-1-5p, miR-24-2-5p, miR-24-3p, miR-296-3p, miR-296-5p, miR-32-3p, miR-337-3p, miR-337-5p, miR-381-3p, and miR-381- 5p.
  • miRNA binding sites from any lung specific miRNA can be introduced to or removed from a polynucleotide of the present disclosure to regulate expression of the polynucleotide in the lung.
  • Lung specific miRNA binding sites can be engineered alone or further in combination with immune cell (e.g., APC) miRNA binding sites in a polynucleotide of the present disclosure.
  • miRNAs that are known to be expressed in the heart include, but are not limited to, miR-1, miR-133a, miR-133b, miR-149-3p, miR-149-5p, miR-186-3p, miR-186-5p, miR- 208a, miR-208b, miR-210, miR-296-3p, miR-320, miR-451a, miR-451b, miR-499a-3p, miR- 499a-5p, miR-499b-3p, miR-499b-5p, miR-744-3p, miR-744-5p, miR-92b-3p, and miR-92b- 5p.
  • mMiRNA binding sites from any heart specific microRNA can be introduced to or removed from a polynucleotide of the present disclosure to regulate expression of the polynucleotide in the heart.
  • Heart specific miRNA binding sites can be engineered alone or further in combination with immune cell (e.g., APC) miRNA binding sites in a
  • miRNAs that are known to be expressed in the nervous system include, but are not limited to, miR-124-5p, miR-125a-3p, miR-125a-5p, miR-125b-1-3p, miR-125b-2-3p, miR- 125b-5p,miR-1271-3p, miR-1271-5p, miR-128, miR-132-5p, miR-135a-3p, miR-135a-5p, miR-135b-3p, miR-135b-5p, miR-137, miR-139-5p, miR-139-3p, miR-149-3p, miR-149-5p, miR-153, miR-181c-3p, miR-181c-5p, miR-183-3p, miR-183-5p, miR-190a, miR-190b, miR- 212-3p, miR-212-5p, miR-219-1-3p, miR-219-2-3p, miR-23a-3p, miR-23a-5p,
  • miRNAs enriched in the nervous system further include those specifically expressed in neurons, including, but not limited to, miR-132-3p, miR-132-3p, miR-148b-3p, miR-148b-5p, miR-151a-3p, miR-151a-5p, miR-212-3p, miR- 212-5p, miR-320b, miR-320e, miR-323a-3p, miR-323a-5p, miR-324-5p, miR-325, miR-326, miR-328, miR-922 and those specifically expressed in glial cells, including, but not limited to, miR-1250, miR-219-1-3p, miR-219-2-3p, miR-219-5p, miR-23a-3p, miR-23a-5p, miR- 3065-3p, miR-3065-5p, miR-30e-3p, miR-30e-5p, miR-32-5p, miR-338-5p, and miR-657.
  • miRNA binding sites from any CNS specific miRNA can be introduced to or removed from a polynucleotide of the present disclosure to regulate expression of the polynucleotide in the nervous system.
  • Nervous system specific miRNA binding sites can be engineered alone or further in combination with immune cell (e.g., APC) miRNA binding sites in a
  • miRNAs that are known to be expressed in the pancreas include, but are not limited to, miR-105-3p, miR-105-5p, miR-184, miR-195-3p, miR-195-5p, miR-196a-3p, miR-196a- 5p, miR-214-3p, miR-214-5p, miR-216a-3p, miR-216a-5p, miR-30a-3p, miR-33a-3p, miR- 33a-5p, miR-375, miR-7-1-3p, miR-7-2-3p, miR-493-3p, miR-493-5p, and miR-944.
  • MiRNA binding sites from any pancreas specific miRNA can be introduced to or removed from a polynucleotide of the present disclosure to regulate expression of the polynucleotide in the pancreas.
  • Pancreas specific miRNA binding sites can be engineered alone or further in combination with immune cell (e.g. APC) miRNA binding sites in a polynucleotide of the present disclosure.
  • miRNAs that are known to be expressed in the kidney include, but are not limited to, miR-122-3p, miR-145-5p, miR-17-5p, miR-192-3p, miR-192-5p, miR-194-3p, miR-194-5p, miR-20a-3p, miR-20a-5p, miR-204-3p, miR-204-5p, miR-210, miR-216a-3p, miR-216a-5p, miR-296-3p, miR-30a-3p, miR-30a-5p, miR-30b-3p, miR-30b-5p, miR-30c-1-3p, miR-30c- 2-3p, miR30c-5p, miR-324-3p, miR-335-3p, miR-335-5p, miR-363-3p, miR-363-5p, and miR-562.
  • kidney specific miRNA binding sites from any kidney specific miRNA can be introduced to or removed from a polynucleotide of the present disclosure to regulate expression of the polynucleotide in the kidney.
  • Kidney specific miRNA binding sites can be engineered alone or further in combination with immune cell (e.g., APC) miRNA binding sites in a
  • miRNAs that are known to be expressed in the muscle include, but are not limited to, let-7g-3p, let-7g-5p, miR-1, miR-1286, miR-133a, miR-133b, miR-140-3p, miR-143-3p, miR-143-5p, miR-145-3p, miR-145-5p, miR-188-3p, miR-188-5p, miR-206, miR-208a, miR- 208b, miR-25-3p, and miR-25-5p.
  • MiRNA binding sites from any muscle specific miRNA can be introduced to or removed from a polynucleotide of the present disclosure to regulate expression of the polynucleotide in the muscle.
  • Muscle specific miRNA binding sites can be engineered alone or further in combination with immune cell (e.g., APC) miRNA binding sites in a polynucleotide of the present disclosure.
  • miRNAs are also differentially expressed in different types of cells, such as, but not limited to, endothelial cells, epithelial cells, and adipocytes.
  • miRNAs that are known to be expressed in endothelial cells include, but are not limited to, let-7b-3p, let-7b-5p, miR-100-3p, miR-100-5p, miR-101-3p, miR-101-5p, miR- 126-3p, miR-126-5p, miR-1236-3p, miR-1236-5p, miR-130a-3p, miR-130a-5p, miR-17-5p, miR-17-3p, miR-18a-3p, miR-18a-5p, miR-19a-3p, miR-19a-5p, miR-19b-1-5p, miR-19b-2- 5p, miR-19b-3p, miR-20a-3p, miR-20a-5p, miR-217, miR-210, miR-21-3p, miR-21-5p, miR- 221-3p, miR-221-5p, miR-222-3p, miR-222-5p, miR-23a-3p, miR-23a-5p, miR-2
  • miRNA binding sites from any endothelial cell specific miRNA can be introduced to or removed from a polynucleotide of the present disclosure to regulate expression of the polynucleotide in the endothelial cells.
  • miRNAs that are known to be expressed in epithelial cells include, but are not limited to, let-7b-3p, let-7b-5p, miR-1246, miR-200a-3p, miR-200a-5p, miR-200b-3p, miR-200b-5p, miR-200c-3p, miR-200c-5p, miR-338-3p, miR-429, miR-451a, miR-451b, miR-494, miR- 802 and miR-34a, miR-34b-5p, miR-34c-5p, miR-449a, miR-449b-3p, miR-449b-5p specific in respiratory ciliated epithelial cells, let-7 family, miR-133a, miR-133b, miR-126 specific in lung epithelial cells, miR-382-3p, miR-382-5p specific in renal epithelial cells, and miR-762 specific in corneal epithelial cells. miRNA binding sites from any epithelial
  • a large group of miRNAs are enriched in embryonic stem cells, controlling stem cell self-renewal as well as the development and/or differentiation of various cell lineages, such as neural cells, cardiac, hematopoietic cells, skin cells, osteogenic cells and muscle cells (e.g., Kuppusamy KT et al., Curr.
  • MiRNAs abundant in embryonic stem cells include, but are not limited to, let-7a-2- 3p, let-a-3p, let-7a-5p, let7d-3p, let-7d-5p, miR-103a-2-3p, miR-103a-5p, miR-106b-3p, miR-106b-5p, miR-1246, miR-1275, miR-138-1-3p, miR-138-2-3p, miR-138-5p, miR-154- 3p, miR-154-5p, miR-200c-3p, miR-200c-5p, miR-290, miR-301a-3p, miR-301a-5p, miR- 302a-3p, miR-302a-5p, miR-302b-3p, miR-302b-5p, miR-302c-3p, miR-302c-5p, miR-302d- 3p, miR-302d-5p, miR-302e, miR-367-3p, miR-367-5p, miR
  • the binding sites of embryonic stem cell specific miRNAs can be included in or removed from the 3'UTR of a polynucleotide of the present disclosure to modulate the development and/or differentiation of embryonic stem cells, to inhibit the senescence of stem cells in a degenerative condition (e.g. degenerative diseases), or to stimulate the senescence and apoptosis of stem cells in a disease condition (e.g. cancer stem cells).
  • a degenerative condition e.g. degenerative diseases
  • apoptosis of stem cells e.g. cancer stem cells
  • miRNA binding sites for miRNAs that are over-expressed in certain cancer and/or tumor cells can be removed from the 3'UTR of a polynucleotide of the present disclosure, restoring the expression suppressed by the over-expressed miRNAs in cancer cells, thus ameliorating the corresponsive biological function, for instance, transcription stimulation and/or repression, cell cycle arrest, apoptosis and cell death.
  • miRNA can also regulate complex biological processes such as angiogenesis (e.g., miR-132) (Anand and Cheresh Curr Opin Hematol 201118:171-176).
  • angiogenesis e.g., miR-132
  • miRNA binding sites that are involved in such processes can be removed or introduced, in order to tailor the expression of the polynucleotides to biologically relevant cell types or relevant biological processes.
  • the polynucleotides of the present disclosure are defined as auxotrophic polynucleotides.
  • a polynucleotide of the present disclosure comprises a miRNA binding site, wherein the miRNA binding site comprises one or more nucleotide sequences selected from Table 6, including one or more copies of any one or more of the miRNA binding site sequences.
  • a polynucleotide of the present disclosure further comprises at least one, two, three, four, five, six, seven, eight, nine, ten, or more of the same or different miRNA binding sites selected from Table 6, including any combination thereof.
  • the miRNA binding site binds to miR-142 or is
  • the miR-142 comprises SEQ ID NO:113.
  • the miRNA binding site binds to miR-142-3p or miR-142-5p.
  • the miR-142-3p binding site comprises SEQ ID NO:106.
  • the miR-142-5p binding site comprises SEQ ID NO:108. In some embodiments, the miR-142-5p binding site comprises SEQ ID NO:108.
  • the miRNA binding site comprises a nucleotide sequence at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to SEQ ID NO:106 or SEQ ID NO:108.
  • a miRNA binding site is inserted in the polynucleotide of the present disclosure in any position of the polynucleotide (e.g., the 5'UTR and/or 3'UTR).
  • the 5'UTR comprises a miRNA binding site.
  • the 3'UTR comprises a miRNA binding site.
  • the 5'UTR and the 3'UTR comprise a miRNA binding site.
  • the insertion site in the polynucleotide can be anywhere in the polynucleotide as long as the insertion of the miRNA binding site in the polynucleotide does not interfere with the translation of a functional polypeptide in the absence of the corresponding miRNA; and in the presence of the miRNA, the insertion of the miRNA binding site in the polynucleotide and the binding of the miRNA binding site to the corresponding miRNA are capable of degrading the polynucleotide or preventing the translation of the polynucleotide.
  • a miRNA binding site is inserted in at least about 30 nucleotides downstream from the stop codon of an ORF in a polynucleotide of the present disclosure comprising the ORF. In some embodiments, a miRNA binding site is inserted in at least about 10 nucleotides, at least about 15 nucleotides, at least about 20 nucleotides, at least about 25 nucleotides, at least about 30 nucleotides, at least about 35 nucleotides, at least about 40 nucleotides, at least about 45 nucleotides, at least about 50 nucleotides, at least about 55 nucleotides, at least about 60 nucleotides, at least about 65 nucleotides, at least about 70 nucleotides, at least about 75 nucleotides, at least about 80 nucleotides, at least about 85 nucleotides, at least about 90 nucleotides, at least about 95 nucleotides, or at least about 100 nucleotides
  • a miRNA binding site is inserted in about 10 nucleotides to about 100 nucleotides, about 20 nucleotides to about 90 nucleotides, about 30 nucleotides to about 80 nucleotides, about 40 nucleotides to about 70 nucleotides, about 50 nucleotides to about 60 nucleotides, about 45 nucleotides to about 65 nucleotides downstream from the stop codon of an ORF in a polynucleotide of the present disclosure.
  • miRNA gene regulation can be influenced by the sequence surrounding the miRNA such as, but not limited to, the species of the surrounding sequence, the type of sequence (e.g., heterologous, homologous, exogenous, endogenous, or artificial), regulatory elements in the surrounding sequence and/or structural elements in the surrounding sequence.
  • the miRNA can be influenced by the 5′UTR and/or 3′UTR.
  • a non- human 3′UTR can increase the regulatory effect of the miRNA sequence on the expression of a polypeptide of interest compared to a human 3′UTR of the same sequence type.
  • other regulatory elements and/or structural elements of the 5′UTR can influence miRNA mediated gene regulation.
  • a regulatory element and/or structural element is a structured IRES (Internal Ribosome Entry Site) in the 5′UTR, which is necessary for the binding of translational elongation factors to initiate protein translation. EIF4A2 binding to this secondarily structured element in the 5′-UTR is necessary for miRNA mediated gene expression (Meijer HA et al., Science, 2013, 340, 82- 85, herein incorporated by reference in its entirety).
  • the polynucleotides of the present disclosure can further include this structured 5′UTR in order to enhance microRNA mediated gene regulation.
  • At least one miRNA binding site can be engineered into the 3′UTR of a polynucleotide of the present disclosure.
  • at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, or more miRNA binding sites can be engineered into a 3′UTR of a polynucleotide of the present disclosure.
  • 1 to 10, 1 to 9, 1 to 8, 1 to 7, 1 to 6, 1 to 5, 1 to 4, 1 to 3, 2, or 1 miRNA binding sites can be engineered into the 3′UTR of a polynucleotide of the present disclosure.
  • miRNA binding sites incorporated into a polynucleotide of the present disclosure can be the same or can be different miRNA sites.
  • a combination of different miRNA binding sites incorporated into a polynucleotide of the present disclosure can include combinations in which more than one copy of any of the different miRNA sites are incorporated.
  • miRNA binding sites incorporated into a polynucleotide of the present disclosure can target the same or different tissues in the body.
  • tissue-, cell-type-, or disease-specific miRNA binding sites in the 3′-UTR of a polynucleotide of the present disclosure can be reduced.
  • specific cell types e.g., hepatocytes, myeloid cells, endothelial cells, cancer cells, etc.
  • a miRNA binding site can be engineered near the 5′ terminus of the 3′UTR, about halfway between the 5′ terminus and 3′ terminus of the 3′UTR and/or near the 3′ terminus of the 3′UTR in a polynucleotide of the present disclosure.
  • a miRNA binding site can be engineered near the 5′ terminus of the 3′UTR and about halfway between the 5′ terminus and 3′ terminus of the 3′UTR.
  • a miRNA binding site can be engineered near the 3′ terminus of the 3′UTR and about halfway between the 5′ terminus and 3′ terminus of the 3′UTR.
  • a miRNA binding site can be engineered near the 5′ terminus of the 3′UTR and near the 3′ terminus of the 3′UTR.
  • a 3′UTR can comprise 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 miRNA binding sites.
  • the miRNA binding sites can be complementary to a miRNA, miRNA seed sequence, and/or miRNA sequences flanking the seed sequence.
  • a polynucleotide of the present disclosure can be engineered to include more than one miRNA site expressed in different tissues or different cell types of a subject.
  • a polynucleotide of the present disclosure can be engineered to include miR-192 and miR-122 to regulate expression of the polynucleotide in the liver and kidneys of a subject.
  • a polynucleotide of the present disclosure can be engineered to include more than one miRNA site for the same tissue.
  • the expression of a polynucleotide of the present disclosure can be controlled by incorporating at least one miR binding site in the polynucleotide and formulating the polynucleotide for administration.
  • a miR binding site in the polynucleotide and formulating the polynucleotide for administration.
  • polynucleotide of the present disclosure can be targeted to a tissue or cell by incorporating a miRNA binding site and formulating the polynucleotide in a lipid nanoparticle comprising an ionizable e.g., an ionizable amino lipid, sometimes referred to in the prior art as an“ionizable cationic lipid”, including any of the lipids described herein.
  • an ionizable e.g., an ionizable amino lipid sometimes referred to in the prior art as an“ionizable cationic lipid”, including any of the lipids described herein.
  • a polynucleotide of the present disclosure can be engineered for more targeted expression in specific tissues, cell types, or biological conditions based on the expression patterns of miRNAs in the different tissues, cell types, or biological conditions.
  • tissue-specific miRNA binding sites Through introduction of tissue-specific miRNA binding sites, a polynucleotide of the present disclosure can be designed for optimal protein expression in a tissue or cell, or in the context of a biological condition.
  • a polynucleotide of the present disclosure can be designed to incorporate miRNA binding sites that either have 100% identity to known miRNA seed sequences or have less than 100% identity to miRNA seed sequences.
  • a polynucleotide of the present disclosure can be designed to incorporate miRNA binding sites that have at least: 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identity to known miRNA seed sequences.
  • the miRNA seed sequence can be partially mutated to decrease miRNA binding affinity and as such result in reduced downmodulation of the polynucleotide.
  • the degree of match or mis-match between the miRNA binding site and the miRNA seed can act as a rheostat to more finely tune the ability of the miRNA to modulate protein expression.
  • mutation in the non-seed region of a miRNA binding site can also impact the ability of a miRNA to modulate protein expression.
  • a miRNA sequence can be incorporated into the loop of a stem loop.
  • a miRNA seed sequence can be incorporated in the loop of a stem loop and a miRNA binding site can be incorporated into the 5′ or 3′ stem of the stem loop.
  • the miRNA sequence in the 5′UTR can be used to stabilize a polynucleotide of the present disclosure described herein.
  • a miRNA sequence in the 5′UTR of a polynucleotide of the present disclosure can be used to decrease the accessibility of the site of translation initiation such as, but not limited to a start codon.
  • a site of translation initiation such as, but not limited to a start codon.
  • LNA antisense locked nucleic acid
  • EJCs exon-junction complexes
  • a polynucleotide of the present disclosure can comprise a miRNA sequence, instead of the LNA or EJC sequence described by Matsuda et al, near the site of translation initiation in order to decrease the accessibility to the site of translation initiation.
  • the site of translation initiation can be prior to, after or within the miRNA sequence.
  • the site of translation initiation can be located within a miRNA sequence such as a seed sequence or binding site.
  • the site of translation initiation can be located within a miR-122 sequence such as the seed sequence or the mir-122 binding site.
  • a polynucleotide of the present disclosure can include at least one miRNA in order to dampen the antigen presentation by antigen presenting cells.
  • the miRNA can be the complete miRNA sequence, the miRNA seed sequence, the miRNA sequence without the seed, or a combination thereof.
  • a miRNA incorporated into a polynucleotide of the present disclosure can be specific to the
  • a miRNA incorporated into a polynucleotide of the present disclosure to dampen antigen presentation is miR-142-3p.
  • a polynucleotide of the present disclosure can include at least one miRNA in order to dampen expression of the encoded polypeptide in a tissue or cell of interest.
  • a polynucleotide of the present disclosure can include at least one miR-122 binding site in order to dampen expression of an encoded polypeptide of interest in the liver.
  • a polynucleotide of the present disclosure can include at least one miR-142-3p binding site, miR-142-3p seed sequence, miR- 142-3p binding site without the seed, miR-142-5p binding site, miR-142-5p seed sequence, miR-142-5p binding site without the seed, miR-146 binding site, miR-146 seed sequence and/or miR-146 binding site without the seed sequence.
  • a polynucleotide of the present disclosure can comprise at least one miRNA binding site in the 3′UTR in order to selectively degrade mRNA therapeutics in the immune cells to subdue unwanted immunogenic reactions caused by therapeutic delivery.
  • the miRNA binding site can make a polynucleotide of the present disclosure more unstable in antigen presenting cells.
  • Non-limiting examples of these miRNAs include mir-142-5p, mir-142-3p, mir-146a-5p, and mir-146-3p.
  • a polynucleotide of the present disclosure comprises at least one miRNA sequence in a region of the polynucleotide that can interact with a RNA binding protein.
  • the polynucleotide of the present disclosure e.g., a RNA, e.g., an mRNA
  • a RNA e.g., an mRNA
  • a sequence-optimized nucleotide sequence e.g., an ORF
  • an antibody e.g., the wild-type sequence, functional fragment, or variant thereof
  • a miRNA binding site e.g., a miRNA binding site that binds to miR-142
  • the polynucleotide of the present disclosure (e.g., a RNA, e.g., an mRNA) comprises a sequence-optimized nucleotide sequence (e.g., an ORF) encoding an antibody (e.g., full length antibody, scFv, functional fragment, or variant thereof), wherein the polynucleotide comprises 1-methylpseudouridines.
  • the polynucleotide further comprises a 5’ UTR having SEQ ID NO.160 or 162 and a 3’UTR having SEQ ID NO.161 and 163.
  • the polynucleotide disclosed herein is formulated with a delivery agent, e.g., a lipid nanoparticle comprised of an ionizable lipid of compound 18 or 25, a neutral lipid, a structural lipid and a PEG lipid.
  • a delivery agent e.g., a lipid nanoparticle comprised of an ionizable lipid of compound 18 or 25, a neutral lipid, a structural lipid and a PEG lipid.
  • PEG lipid comprising Formula VI, or an ionizable cationic lipid of
  • a polynucleotide of the present disclosure e.g., a
  • polynucleotide comprising a nucleotide sequence encoding an antibody of the present disclosure) further comprises a 3' UTR.
  • 3'-UTR is the section of mRNA that immediately follows the translation termination codon and often contains regulatory regions that post-transcriptionally influence gene expression. Regulatory regions within the 3'-UTR can influence polyadenylation, translation efficiency, localization, and stability of the mRNA.
  • the 3'-UTR useful for the present disclosure comprises a binding site for regulatory proteins or microRNAs. Regions having a 5′ Cap
  • the present disclosure also includes a polynucleotide that comprises both a 5′ Cap and a polynucleotide of the present disclosure (e.g., a polynucleotide comprising a nucleotide sequence encoding an antibody ).
  • the 5′ cap structure of a natural mRNA is involved in nuclear export, increasing mRNA stability and binds the mRNA Cap Binding Protein (CBP), which is responsible for mRNA stability in the cell and translation competency through the association of CBP with poly(A) binding protein to form the mature cyclic mRNA species.
  • CBP mRNA Cap Binding Protein
  • the cap further assists the removal of 5′ proximal introns during mRNA splicing.
  • Endogenous mRNA molecules can be 5′-end capped generating a 5′-ppp-5′- triphosphate linkage between a terminal guanosine cap residue and the 5′-terminal transcribed sense nucleotide of the mRNA molecule.
  • This 5′-guanylate cap can then be methylated to generate an N7-methyl-guanylate residue.
  • the ribose sugars of the terminal and/or anteterminal transcribed nucleotides of the 5′ end of the mRNA can optionally also be 2′-O- methylated.5′-decapping through hydrolysis and cleavage of the guanylate cap structure can target a nucleic acid molecule, such as an mRNA molecule, for degradation.
  • the polynucleotides of the present disclosure e.g., a polynucleotide comprising a nucleotide sequence encoding an antibody
  • incorporate a cap moiety e.g., a poly
  • polynucleotides of the present disclosure e.g., a
  • polynucleotide comprising a nucleotide sequence encoding an antibody
  • polynucleotide sequence encoding an antibody
  • modified nucleotides can be used during the capping reaction.
  • a Vaccinia Capping Enzyme from New England Biolabs (Ipswich, MA) can be used with ⁇ -thio- guanosine nucleotides according to the manufacturer's instructions to create a
  • Additional modified guanosine nucleotides can be used such as ⁇ -methyl-phosphonate and seleno-phosphate nucleotides.
  • Additional modifications include, but are not limited to, 2′-O-methylation of the ribose sugars of 5′-terminal and/or 5′-anteterminal nucleotides of the polynucleotide (as mentioned above) on the 2′-hydroxyl group of the sugar ring.
  • Multiple distinct 5′-cap structures can be used to generate the 5′-cap of a nucleic acid molecule, such as a
  • Cap analogs which herein are also referred to as synthetic cap analogs, chemical caps, chemical cap analogs, or structural or functional cap analogs, differ from natural (i.e., endogenous, wild-type or physiological) 5′- caps in their chemical structure, while retaining cap function. Cap analogs can be chemically (i.e., non-enzymatically) or enzymatically synthesized and/or linked to the polynucleotides of the present disclosure.
  • the Anti-Reverse Cap Analog (ARCA) cap contains two guanines linked by a 5′-5′-triphosphate group, wherein one guanine contains an N7 methyl group as well as a 3′-O-methyl group (i.e., N7,3′-O-dimethyl-guanosine-5′-triphosphate-5′-guanosine (m 7 G- 3′mppp-G; which can equivalently be designated 3′ O-Me-m7G(5')ppp(5')G).
  • the 3′-O atom of the other, unmodified, guanine becomes linked to the 5′-terminal nucleotide of the capped polynucleotide.
  • the N7- and 3′-O-methlyated guanine provides the terminal moiety of the capped polynucleotide.
  • mCAP is similar to ARCA but has a 2′-O-methyl group on guanosine (i.e., N7,2′-O-dimethyl-guanosine-5′-triphosphate-5′-guanosine, m 7 Gm- ppp-G).
  • the cap is a dinucleotide cap analog.
  • the dinucleotide cap analog can be modified at different phosphate positions with a boranophosphate group or a phophoroselenoate group such as the dinucleotide cap analogs described in U.S. Patent No. US 8,519,110, the contents of which are herein incorporated by reference in its entirety.
  • the cap is a cap analog is a N7-(4-chlorophenoxyethyl) substituted dicucleotide form of a cap analog known in the art and/or described herein.
  • Non- limiting examples of a N7-(4-chlorophenoxyethyl) substituted dicucleotide form of a cap analog include a N7-(4-chlorophenoxyethyl)-G(5')ppp(5')G and a N7-(4- chlorophenoxyethyl)-m 3'-O G(5')ppp(5')G cap analog (See, e.g., the various cap analogs and the methods of synthesizing cap analogs described in Kore et al. Bioorganic & Medicinal Chemistry 201321:4570-4574; the contents of which are herein incorporated by reference in its entirety).
  • a cap analog of the present disclosure is a 4- chloro/bromophenoxye
  • cap analogs allow for the concomitant capping of a polynucleotide or a region thereof, in an in vitro transcription reaction, up to 20% of transcripts can remain uncapped. This, as well as the structural differences of a cap analog from an endogenous 5′-cap structures of nucleic acids produced by the endogenous, cellular transcription machinery, can lead to reduced translational competency and reduced cellular stability.
  • Polynucleotides of the present disclosure can also be capped post-manufacture (whether IVT or chemical synthesis), using enzymes, in order to generate more authentic 5′-cap structures.
  • the phrase "more authentic” refers to a feature that closely mirrors or mimics, either structurally or functionally, an endogenous or wild type feature.
  • a "more authentic" feature is better representative of an endogenous, wild-type, natural or physiological cellular function and/or structure as compared to synthetic features or analogs, etc., of the prior art, or which outperforms the corresponding endogenous, wild-type, natural or physiological feature in one or more respects.
  • Non-limiting examples of more authentic 5′cap structures of the present disclosure are those that, among other things, have enhanced binding of cap binding proteins, increased half-life, reduced susceptibility to 5′ endonucleases and/or reduced 5′decapping, as compared to synthetic 5′cap structures known in the art (or to a wild-type, natural or physiological 5′cap structure).
  • recombinant Vaccinia Virus Capping Enzyme and recombinant 2′-O-methyltransferase enzyme can create a canonical 5′-5′-triphosphate linkage between the 5′-terminal nucleotide of a polynucleotide and a guanine cap nucleotide wherein the cap guanine contains an N7 methylation and the 5′- terminal nucleotide of the mRNA contains a 2′-O-methyl.
  • Cap1 structure is termed the Cap1 structure.
  • Cap structures include, but are not limited to,
  • capping chimeric polynucleotides post-manufacture can be more efficient as nearly 100% of the chimeric polynucleotides can be capped. This is in contrast to ⁇ 80% when a cap analog is linked to a chimeric polynucleotide in the course of an in vitro transcription reaction.
  • 5′ terminal caps can include endogenous caps or cap analogs.
  • a 5′ terminal cap can comprise a guanine analog.
  • Useful guanine analogs include, but are not limited to, inosine, N1-methyl-guanosine, 2′fluoro-guanosine, 7-deaza-guanosine, 8-oxo-guanosine, 2-amino-guanosine, LNA- guanosine, and 2-azido-guanosine.
  • Poly-A Tails include, but are not limited to, inosine, N1-methyl-guanosine, 2′fluoro-guanosine, 7-deaza-guanosine, 8-oxo-guanosine, 2-amino-guanosine, LNA- guanosine, and 2-azido-guanosine.
  • the polynucleotides of the present disclosure (e.g., a polynucleotide comprising a nucleotide sequence encoding an antibody) further comprise a poly-A tail.
  • terminal groups on the poly-A tail can be incorporated for stabilization.
  • a poly-A tail comprises des-3' hydroxyl tails.
  • a long chain of adenine nucleotides can be added to a polynucleotide such as an mRNA molecule in order to increase stability.
  • poly-A polymerase adds a chain of adenine nucleotides to the RNA.
  • the process called polyadenylation, adds a poly-A tail that can be between, for example, approximately 80 to approximately 250 residues long, including approximately 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240 or 250 residues long.
  • PolyA tails can also be added after the construct is exported from the nucleus.
  • terminal groups on the poly A tail can be incorporated for stabilization.
  • Polynucleotides of the present disclosure can include des-3' hydroxyl tails. They can also include structural moieties or 2'-Omethyl modifications as taught by Junjie Li, et al. (Current Biology, Vol.15, 1501–1507, August 23, 2005, the contents of which are incorporated herein by reference in its entirety).
  • the polynucleotides of the present disclosure can be designed to encode transcripts with alternative polyA tail structures including histone mRNA. According to Norbury, "Terminal uridylation has also been detected on human replication-dependent histone mRNAs. The turnover of these mRNAs is thought to be important for the prevention of potentially toxic histone accumulation following the completion or inhibition of
  • mRNAs are distinguished by their lack of a 3 ⁇ poly(A) tail, the function of which is instead assumed by a stable stem–loop structure and its cognate stem–loop binding protein (SLBP); the latter carries out the same functions as those of PABP on polyadenylated mRNAs" (Norbury, "Cytoplasmic RNA: a case of the tail wagging the dog," Nature Reviews Molecular Cell Biology; AOP, published online 29 August 2013; doi:10.1038/nrm3645) the contents of which are incorporated herein by reference in its entirety.
  • SLBP stem–loop binding protein
  • the length of a poly-A tail when present, is greater than 30 nucleotides in length. In another embodiment, the poly-A tail is greater than 35 nucleotides in length (e.g., at least or greater than about 35, 40, 45, 50, 55, 60, 70, 80, 90, 100, 120, 140, 160, 180, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, 900, 1,000, 1,100, 1,200, 1,300, 1,400, 1,500, 1,600, 1,700, 1,800, 1,900, 2,000, 2,500, and 3,000 nucleotides).
  • the poly-A tail is greater than 35 nucleotides in length (e.g., at least or greater than about 35, 40, 45, 50, 55, 60, 70, 80, 90, 100, 120, 140, 160, 180, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, 900, 1,000, 1,100, 1,200, 1,300, 1,400, 1,500, 1,600,
  • the polynucleotide or region thereof includes from about 30 to about 3,000 nucleotides (e.g., from 30 to 50, from 30 to 100, from 30 to 250, from 30 to 500, from 30 to 750, from 30 to 1,000, from 30 to 1,500, from 30 to 2,000, from 30 to 2,500, from 50 to 100, from 50 to 250, from 50 to 500, from 50 to 750, from 50 to 1,000, from 50 to 1,500, from 50 to 2,000, from 50 to 2,500, from 50 to 3,000, from 100 to 500, from 100 to 750, from 100 to 1,000, from 100 to 1,500, from 100 to 2,000, from 100 to 2,500, from 100 to 3,000, from 500 to 750, from 500 to 1,000, from 500 to 1,500, from 500 to 2,000, from 500 to 2,500, from 500 to 3,000, from 1,000 to 1,500, from 1,000 to 2,000, from 1,000 to 2,500, from 1,000 to 3,000, from 1,500 to 2,000, from 1,500 to 2,500, from 1,500 to 3,000, from from about 30 to
  • the poly-A tail is designed relative to the length of the overall polynucleotide or the length of a particular region of the polynucleotide. This design can be based on the length of a coding region, the length of a particular feature or region or based on the length of the ultimate product expressed from the polynucleotides.
  • the poly-A tail can be 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100% greater in length than the polynucleotide or feature thereof.
  • the poly-A tail can also be designed as a fraction of the polynucleotides to which it belongs.
  • the poly-A tail can be 10, 20, 30, 40, 50, 60, 70, 80, or 90% or more of the total length of the construct, a construct region or the total length of the construct minus the poly-A tail.
  • engineered binding sites and conjugation of polynucleotides for Poly-A binding protein can enhance expression.
  • multiple distinct polynucleotides can be linked together via the PABP (Poly-A binding protein) through the 3′-end using modified nucleotides at the 3′-terminus of the poly-A tail.
  • Transfection experiments can be conducted in relevant cell lines at and protein production can be assayed by ELISA at 12hr, 24hr, 48hr, 72hr and day 7 post- transfection.
  • the polynucleotides of the present disclosure are designed to include a polyA-G quartet region.
  • the G-quartet is a cyclic hydrogen bonded array of four guanine nucleotides that can be formed by G-rich sequences in both DNA and RNA.
  • the G-quartet is incorporated at the end of the poly-A tail.
  • the resultant polynucleotide is assayed for stability, protein production and other parameters including half-life at various time points. It has been discovered that the polyA-G quartet results in protein production from an mRNA equivalent to at least 75% of that seen using a poly-A tail of 120 nucleotides alone. Start codon region
  • the present disclosure also includes a polynucleotide that comprises both a start codon region and the polynucleotide described herein (e.g., a polynucleotide comprising a nucleotide sequence encoding an antibody).
  • the polynucleotides of the present disclosure can have regions that are analogous to or function like a start codon region.
  • the translation of a polynucleotide can initiate on a codon that is not the start codon AUG.
  • Translation of the polynucleotide can initiate on an alternative start codon such as, but not limited to, ACG, AGG, AAG, CTG/CUG, GTG/GUG,
  • the translation of a polynucleotide begins on the alternative start codon ACG.
  • polynucleotide translation begins on the alternative start codon CTG or CUG.
  • the translation of a polynucleotide begins on the alternative start codon GTG or GUG.
  • Nucleotides flanking a codon that initiates translation such as, but not limited to, a start codon or an alternative start codon, are known to affect the translation efficiency, the length and/or the structure of the polynucleotide. (See, e.g., Matsuda and Mauro PLoS ONE, 20105:11; the contents of which are herein incorporated by reference in its entirety).
  • Masking any of the nucleotides flanking a codon that initiates translation can be used to alter the position of translation initiation, translation efficiency, length and/or structure of a polynucleotide.
  • a masking agent can be used near the start codon or alternative start codon in order to mask or hide the codon to reduce the probability of translation initiation at the masked start codon or alternative start codon.
  • masking agents include antisense locked nucleic acids (LNA) polynucleotides and exon- junction complexes (EJCs) (See, e.g., Matsuda and Mauro describing masking agents LNA polynucleotides and EJCs (PLoS ONE, 20105:11); the contents of which are herein incorporated by reference in its entirety).
  • a masking agent can be used to mask a start codon of a polynucleotide in order to increase the likelihood that translation will initiate on an alternative start codon.
  • a masking agent can be used to mask a first start codon or alternative start codon in order to increase the chance that translation will initiate on a start codon or alternative start codon downstream to the masked start codon or alternative start codon.
  • a start codon or alternative start codon can be located within a perfect complement for a miR binding site.
  • the perfect complement of a miR binding site can help control the translation, length and/or structure of the polynucleotide similar to a masking agent.
  • the start codon or alternative start codon can be located in the middle of a perfect complement for a miRNA binding site.
  • the start codon or alternative start codon can be located after the first nucleotide, second nucleotide, third nucleotide, fourth nucleotide, fifth nucleotide, sixth nucleotide, seventh nucleotide, eighth nucleotide, ninth nucleotide, tenth nucleotide, eleventh nucleotide, twelfth nucleotide, thirteenth nucleotide, fourteenth nucleotide, fifteenth nucleotide, sixteenth nucleotide, seventeenth nucleotide, eighteenth nucleotide, nineteenth nucleotide, twentieth nucleotide or twenty-first nucleotide.
  • the start codon of a polynucleotide can be removed from the polynucleotide sequence in order to have the translation of the polynucleotide begin on a codon that is not the start codon.
  • Translation of the polynucleotide can begin on the codon following the removed start codon or on a downstream start codon or an alternative start codon.
  • the start codon ATG or AUG is removed as the first 3 nucleotides of the polynucleotide sequence in order to have translation initiate on a downstream start codon or alternative start codon.
  • the polynucleotide sequence where the start codon was removed can further comprise at least one masking agent for the downstream start codon and/or alternative start codons in order to control or attempt to control the initiation of translation, the length of the polynucleotide and/or the structure of the
  • the present disclosure also includes a polynucleotide that comprises both a stop codon region and the polynucleotide described herein (e.g., a polynucleotide comprising a nucleotide sequence encoding an antibody).
  • the polynucleotides of the present disclosure can include at least two stop codons before the 3' untranslated region (UTR).
  • the stop codon can be selected from TGA, TAA and TAG in the case of DNA, or from UGA, UAA and UAG in the case of RNA.
  • the polynucleotides of the present disclosure include the stop codon TGA in the case or DNA, or the stop codon UGA in the case of RNA, and one additional stop codon.
  • the addition stop codon can be TAA or UAA.
  • the polynucleotides of the present disclosure include three consecutive stop codons, four stop codons, or more.
  • the mRNAs of the disclosure encode more than one peptide, referred to herein as multimer constructs.
  • the mRNA further encodes a linker located between each domain.
  • the linker can be, for example, a cleavable linker or protease-sensitive linker.
  • the linker is selected from the group consisting of F2A linker, P2A linker, T2A linker, E2A linker, and combinations thereof.
  • This family of self-cleaving peptide linkers, referred to as 2A peptides has been described in the art (see for example, Kim, J.H. et al. (2011) PLoS ONE6:e18556).
  • the linker is an F2A linker. In certain embodiments, the linker is a GGGS linker. In certain embodiments, the multimer construct contains three domains with intervening linkers, having the structure: domain-linker-domain-linker-domain. The skilled artisan will likewise appreciate that other multicistronic constructs may be suitable for use in the invention. In exemplary embodiments, the construct design yields approximately equimolar amounts of intrabody and/or domain thereof encoded by the constructs of the invention. In one embodiment, the self-cleaving peptide may be, but is not limited to, a 2A peptide.
  • 2A peptides are known and available in the art and may be used, including e.g., the foot and mouth disease virus (FMDV) 2A peptide, the equine rhinitis A virus 2A peptide, the Thosea asigna virus 2A peptide, and the porcine teschovirus-12A peptide.
  • FMDV foot and mouth disease virus
  • 2A peptides are used by several viruses to generate two proteins from one transcript by ribosome-skipping, such that a normal peptide bond is impaired at the 2A peptide sequence, resulting in two discontinuous proteins being produced from one translation event.
  • the 2A peptide may have the protein sequence:
  • the 2A peptide cleaves between the last glycine and last proline.
  • the polynucleotide sequence of the 2A peptide may be modified or codon optimized by the methods described herein and/or are known in the art.
  • a polynucleotide of the present disclosure for example a polynucleotide comprising an mRNA nucleotide sequence encoding an antibody, comprises from 5’ to 3’ end:
  • an open reading frame encoding an antibody e.g., a sequence optimized nucleic acid sequence encoding an antibody disclosed herein;
  • the polynucleotide further comprises a miRNA binding site, e.g, a miRNA binding site that binds to miRNA-142.
  • the 5’UTR comprises the miRNA binding site.
  • a polynucleotide of the present disclosure comprises a nucleotide sequence encoding a polypeptide sequence at least 70%, at least 80%, at least 81%, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96% , at least 97%, at least 98%, at least 99%, or 100% identical to the protein sequence of an antibody described herein.
  • the present disclosure also provides methods for making a polynucleotide of the present disclosure (e.g., a polynucleotide comprising a nucleotide sequence encoding an antibody) or a complement thereof.
  • a polynucleotide of the present disclosure e.g., a polynucleotide comprising a nucleotide sequence encoding an antibody
  • a polynucleotide e.g., a RNA, e.g., an mRNA
  • a polynucleotide e.g., a RNA, e.g., an mRNA
  • a polynucleotide e.g., a RNA, e.g., an mRNA
  • encoding an antibody can be constructed by chemical synthesis using an oligonucleotide synthesizer.
  • a polynucleotide e.g., a RNA, e.g., an mRNA
  • encoding an antibody is made by using a host cell.
  • a polynucleotide e.g., a RNA, e.g., an mRNA
  • encoding an antibody is made by one or more combination of the IVT, chemical synthesis, host cell expression, or any other methods known in the art.
  • Naturally occurring nucleosides non-naturally occurring nucleosides, or
  • RNA e.g., an mRNA
  • the resultant polynucleotides e.g., mRNAs, can then be examined for their ability to produce protein and/or produce a therapeutic outcome.
  • the polynucleotides of the present disclosure disclosed herein can be transcribed using an in vitro transcription (IVT) system.
  • the system typically comprises a transcription buffer, nucleotide triphosphates (NTPs), an RNase inhibitor and a polymerase.
  • NTPs can be selected from, but are not limited to, those described herein including natural and unnatural (modified) NTPs.
  • the polymerase can be selected from, but is not limited to, T7 RNA polymerase, T3 RNA polymerase and mutant polymerases such as, but not limited to, polymerases able to incorporate polynucleotides disclosed herein. See U.S. Publ. No. US20130259923, which is herein incorporated by reference in its entirety.
  • RNA polymerases can be modified by inserting or deleting amino acids of the RNA polymerase sequence.
  • the RNA polymerase can be modified to exhibit an increased ability to incorporate a 2 ⁇ -modified nucleotide triphosphate compared to an unmodified RNA polymerase (see International Publication WO2008078180 and U.S. Patent 8,101,385; herein incorporated by reference in their entireties).
  • Variants can be obtained by evolving an RNA polymerase, optimizing the RNA polymerase amino acid and/or nucleic acid sequence and/or by using other methods known in the art.
  • T7 RNA polymerase variants can be evolved using the continuous directed evolution system set out by Esvelt et al.
  • T7 RNA polymerase can encode at least one mutation such as, but not limited to, lysine at position 93 substituted for threonine (K93T), I4M, A7T, E63V, V64D, A65E, D66Y, T76N, C125R, S128R, A136T, N165S, G175R, H176L, Y178H, F182L, L196F, G198V, D208Y, E222K, S228A, Q239R, T243N, G259D, M267I, G280C, H300R, D351A, A354S, E356D, L360P, A383V, Y385C, D388Y, S397R, M401T, N410S, K450R, P451T, G452V, E484A, H523L, H
  • T7 RNA polymerase variants can encode at least mutation as described in U.S. Pub. Nos.20100120024 and 20070117112; herein incorporated by reference in their entireties.
  • Variants of RNA polymerase can also include, but are not limited to, substitutional variants, conservative amino acid substitution, insertional variants, deletional variants and/or covalent derivatives.
  • the polynucleotide can be designed to be recognized by the wild type or variant RNA polymerases. In doing so, the polynucleotide can be modified to contain sites or regions of sequence changes from the wild type or parent chimeric polynucleotide.
  • Polynucleotide or nucleic acid synthesis reactions can be carried out by enzymatic methods utilizing polymerases.
  • Polymerases catalyze the creation of phosphodiester bonds between nucleotides in a polynucleotide or nucleic acid chain.
  • DNA polymerases can be divided into different families based on amino acid sequence comparison and crystal structure analysis.
  • DNA polymerase I poly I
  • a polymerase family including the Klenow fragments of E. coli, Bacillus DNA polymerase I, Thermus aquaticus (Taq) DNA polymerases, and the T7 RNA and DNA polymerases, is among the best studied of these families.
  • DNA polymerase ⁇ (pol ⁇ ) or B polymerase family, including all eukaryotic replicating DNA polymerases and polymerases from phages T4 and RB69. Although they employ similar catalytic mechanism, these families of polymerases differ in substrate specificity, substrate analog-incorporating efficiency, degree and rate for primer extension, mode of DNA synthesis, exonuclease activity, and sensitivity against inhibitors.
  • DNA polymerases are also selected based on the optimum reaction conditions they require, such as reaction temperature, pH, and template and primer concentrations.
  • RNA polymerases from bacteriophage T3, T7, and SP6 have been widely used to prepare RNAs for biochemical and biophysical studies. RNA polymerases, capping enzymes, and poly-A polymerases are disclosed in the co-pending International Publication No. WO2014028429, the contents of which are incorporated herein by reference in their entirety.
  • the RNA polymerase which can be used in the synthesis of the polynucleotides of the present disclosure is a Syn5 RNA polymerase.
  • the Syn5 RNA polymerase was recently characterized from marine cyanophage Syn5 by Zhu et al. where they also identified the promoter sequence (see Zhu et al. Nucleic Acids Research 2013, the contents of which is herein incorporated by reference in its entirety). Zhu et al.
  • Syn5 RNA polymerase catalyzed RNA synthesis over a wider range of temperatures and salinity as compared to T7 RNA polymerase. Additionally, the requirement for the initiating nucleotide at the promoter was found to be less stringent for Syn5 RNA polymerase as compared to the T7 RNA polymerase making Syn5 RNA polymerase promising for RNA synthesis.
  • a Syn5 RNA polymerase can be used in the synthesis of the
  • RNA polymerase can be used in the synthesis of the polynucleotide requiring a precise 3 ⁇ -terminus.
  • a Syn5 promoter can be used in the synthesis of the polynucleotides.
  • the Syn5 promoter can be 5 ⁇ -ATTGGGCACCCGTAAGGG-3 ⁇ (SEQ ID NO: 110).
  • a Syn5 RNA polymerase can be used in the synthesis of polynucleotides comprising at least one chemical modification described herein and/or known in the art (see e.g., the incorporation of pseudo-UTP and 5Me-CTP.
  • the polynucleotides described herein can be synthesized using a Syn5 RNA polymerase which has been purified using modified and improved purification procedure described by Zhu et al. (Nucleic Acids Research 2013).
  • PCR polymerase chain reaction
  • SDA strand displacement amplification
  • NASBA nucleic acid sequence-based amplification
  • TMA transcription mediated amplification
  • RCA rolling-circle amplification
  • DNA or RNA ligases promote intermolecular ligation of the 5 ⁇ and 3 ⁇ ends of polynucleotide chains through the formation of a phosphodiester bond.
  • Standard methods can be applied to synthesize an isolated polynucleotide sequence encoding an isolated polypeptide of interest, such as a polynucleotide of the present disclosure (e.g., a polynucleotide comprising a nucleotide sequence encoding an antibody).
  • a polynucleotide of the present disclosure e.g., a polynucleotide comprising a nucleotide sequence encoding an antibody.
  • a single DNA or RNA oligomer containing a codon-optimized nucleotide sequence coding for the particular isolated polypeptide can be synthesized.
  • several small oligonucleotides coding for portions of the desired polypeptide can be synthesized and then ligated.
  • the individual oligonucleotides typically contain 5' or 3' overhangs for complementary assembly.
  • a polynucleotide disclosed herein e.g., a RNA, e.g., an mRNA
  • a RNA e.g., an mRNA
  • a polynucleotide disclosed herein can be chemically synthesized using chemical synthesis methods and potential nucleobase substitutions known in the art. See, for example, International Publication Nos. WO2014/093924,
  • Purification of the polynucleotides described herein can include, but is not limited to, polynucleotide clean-up, quality assurance and quality control.
  • Clean-up can be performed by methods known in the arts such as, but not limited to, AGENCOURT® beads (Beckman Coulter Genomics, Danvers, MA), poly-T beads, LNA TM oligo-T capture probes (EXIQON® Inc., Vedbaek, Denmark) or HPLC based purification methods such as, but not limited to, strong anion exchange HPLC, weak anion exchange HPLC, reverse phase HPLC (RP-HPLC), and hydrophobic interaction HPLC (HIC-HPLC).
  • AGENCOURT® beads Beckman Coulter Genomics, Danvers, MA
  • poly-T beads poly-T beads
  • LNA TM oligo-T capture probes EXIQON® Inc., Vedbaek, Denmark
  • HPLC based purification methods such as, but not limited to, strong anion exchange HPLC, weak anion exchange HPLC, reverse phase HPLC (RP-HPLC), and hydrophobic interaction HPLC (HIC-HPLC).
  • purified when used in relation to a polynucleotide such as a “purified polynucleotide” refers to one that is separated from at least one contaminant.
  • a "contaminant” is any substance that makes another unfit, impure or inferior.
  • a purified polynucleotide e.g., DNA and RNA
  • purification of a polynucleotide of the present disclosure removes impurities that can reduce or remove an unwanted immune response, e.g., reducing cytokine activity.
  • the polynucleotide of the present disclosure e.g., a
  • polynucleotide comprising a nucleotide sequence encoding an antibody is purified prior to administration using column chromatography (e.g., strong anion exchange HPLC, weak anion exchange HPLC, reverse phase HPLC (RP-HPLC), and hydrophobic interaction HPLC (HIC-HPLC), or (LCMS)).
  • column chromatography e.g., strong anion exchange HPLC, weak anion exchange HPLC, reverse phase HPLC (RP-HPLC), and hydrophobic interaction HPLC (HIC-HPLC), or (LCMS)
  • the polynucleotide of the present disclosure e.g., a
  • polynucleotide comprising a nucleotide sequence an antibody
  • column chromatography e.g., strong anion exchange HPLC, weak anion exchange HPLC, reverse phase HPLC (RP-HPLC, hydrophobic interaction HPLC (HIC-HPLC), or (LCMS)
  • RP-HPLC reverse phase HPLC
  • HIC-HPLC hydrophobic interaction HPLC
  • LCMS hydrophobic interaction HPLC
  • a column chromatography e.g., strong anion exchange HPLC, weak anion exchange HPLC, reverse phase HPLC (RP-HPLC), hydrophobic interaction HPLC (HIC-HPLC), or (LCMS)
  • purified polynucleotide comprises a nucleotide sequence encoding an antibody comprising one or more of the point mutations known in the art.
  • the use of RP-HPLC purified polynucleotide increases antibody expression levels in cells when introduced into those cells, e.g., by 10-100%, i.e., at least about 10%, at least about 20%, at least about 25%, 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 90%, at least about 95%, or at least about 100% with respect to the expression levels of antibody in the cells before the RP-HPLC purified polynucleotide was introduced in the cells, or after a non-RP-HPLC purified polynucleotide was introduced in the cells.
  • the use of RP-HPLC purified polynucleotide increases functional antibody expression levels in cells when introduced into those cells, e.g., by 10- 100%, i.e., at least about 10%, at least about 20%, at least about 25%, 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 90%, at least about 95%, or at least about 100% with respect to the functional expression levels of antibody in the cells before the RP-HPLC purified
  • polynucleotide was introduced in the cells, or after a non-RP-HPLC purified polynucleotide was introduced in the cells.
  • the use of RP-HPLC purified polynucleotide increases detectable antibody activity in cells when introduced into those cells, e.g., by 10-100%, i.e., at least about 10%, at least about 20%, at least about 25%, 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 90%, at least about 95%, or at least about 100% with respect to the activity levels of functional antibody in the cells before the RP-HPLC purified polynucleotide was introduced in the cells, or after a non-RP-HPLC purified polynucleotide was introduced in the cells.
  • the purified polynucleotide is at least about 80% pure, at least about 85% pure, at least about 90% pure, at least about 95% pure, at least about 96% pure, at least about 97% pure, at least about 98% pure, at least about 99% pure, or about 100% pure.
  • a quality assurance and/or quality control check can be conducted using methods such as, but not limited to, gel electrophoresis, UV absorbance, or analytical HPLC.
  • the polynucleotide can be sequenced by methods including, but not limited to reverse-transcriptase-PCR. d. Quantification of Expressed Polynucleotides Encoding Antibody
  • the polynucleotides of the present disclosure e.g., a polynucleotide comprising a nucleotide sequence encoding an antibody
  • their expression products, as well as degradation products and metabolites can be quantified according to methods known in the art.
  • the polynucleotides of the present disclosure can be quantified in exosomes or when derived from one or more bodily fluid.
  • bodily fluids include 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, 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,
  • exosomes can be retrieved from an organ selected from the group consisting of lung, heart, pancreas, stomach, intestine, bladder, kidney, ovary, testis, skin, colon, breast, prostate, brain, esophagus, liver, and placenta.
  • exosome quantification method a sample of not more than 2mL is obtained from the subject and the exosomes isolated by size exclusion chromatography, density gradient centrifugation, differential centrifugation, nanomembrane ultrafiltration,
  • the level or concentration of a polynucleotide can be an expression level, presence, absence, truncation or alteration of the administered construct. It is advantageous to correlate the level with one or more clinical phenotypes or with an assay for a human disease biomarker.
  • the assay can be performed using construct specific probes, cytometry, qRT-PCR, real-time PCR, PCR, flow cytometry, electrophoresis, mass spectrometry, or combinations thereof while the exosomes can be isolated using immunohistochemical methods such as enzyme linked immunosorbent assay (ELISA) methods. Exosomes can also be isolated by size exclusion chromatography, density gradient centrifugation, differential centrifugation, nanomembrane ultrafiltration, immunoabsorbent capture, affinity purification, microfluidic separation, or combinations thereof.
  • immunohistochemical methods such as enzyme linked immunosorbent assay (ELISA) methods.
  • Exosomes can also be isolated by size exclusion chromatography, density gradient centrifugation, differential centrifugation, nanomembrane ultrafiltration, immunoabsorbent capture, affinity purification, microfluidic separation, or combinations thereof.
  • the polynucleotide can be quantified using methods such as, but not limited to, ultraviolet visible spectroscopy (UV/Vis).
  • UV/Vis ultraviolet visible spectroscopy
  • a non-limiting example of a UV/Vis spectrometer is a NANODROP® spectrometer (ThermoFisher, Waltham, MA).
  • the quantified polynucleotide can be analyzed in order to determine if the polynucleotide can be of proper size, check that no degradation of the polynucleotide has occurred.
  • Degradation of the polynucleotide can be checked by methods such as, but not limited to, agarose gel electrophoresis, HPLC based purification methods such as, but not limited to, strong anion exchange HPLC, weak anion exchange HPLC, reverse phase HPLC (RP-HPLC), and hydrophobic interaction HPLC (HIC-HPLC), liquid chromatography-mass spectrometry (LCMS), capillary electrophoresis (CE) and capillary gel electrophoresis (CGE).
  • HPLC based purification methods such as, but not limited to, strong anion exchange HPLC, weak anion exchange HPLC, reverse phase HPLC (RP-HPLC), and hydrophobic interaction HPLC (HIC-HPLC), liquid chromatography-mass spectrometry (LCMS), capillary electrophoresis (CE) and capillary gel electrophoresis (CGE).
  • compositions e.g., pharmaceutical compositions
  • methods, kits and reagents for prevention and/or treatment of disease in humans and other mammals.
  • the antibodies can be used as therapeutic or prophylactic agents.
  • the antibody is an anti-influenza virus antibody
  • the RNA encoding such an antibody is used to provide prophylactic or therapeutic protection from influenza virus infection.
  • Prophylactic protection from influenza virus infection can be achieved following administration of a composition (e.g., a composition comprising one or more polynucleotides encoding one or more HCAbs) of the present disclosure.
  • the influenza virus is a type A influenza virus.
  • influenza virus is any influenza virus comprising HA proteins of subtype H1 and/or H3 (e.g., H1N1, H3N2).
  • Compositions can be administered once, twice, three times, four times or more.
  • the compositions can be administered to an infected individual to achieve a therapeutic response. Dosing may need to be adjusted accordingly.
  • RNA (mRNA) therapeutic treatments are particularly amenable to combination vaccination approaches due to a number of factors including, but not limited to, speed of manufacture, ability to rapidly tailor treatments to accommodate perceived geographical threat, and the like.
  • a combination treatment can be administered that includes RNA encoding at least one polypeptide (or portion thereof) of a first influenza HA-binding HCAb and further includes RNA encoding at least one polypeptide (or portion thereof) of a second influenza-HA binding HCAb.
  • RNAs can be co-formulated, for example, in a single lipid nanoparticle (LNP) or can be formulated in separate LNPs destined for co- administration.
  • the influenza virus is a type A influenza virus.
  • the influenza virus is any influenza virus comprising HA proteins of subtype H1 and/or H3 (e.g., H1N1, H3N2).
  • a prophylactically effective dose is a therapeutically effective dose that prevents infection with the virus at a clinically acceptable level.
  • the virus at a clinically acceptable level.
  • a therapeutically effective dose is a dose listed in a package insert for the treatment.
  • a prophylactic therapy as used herein refers to a therapy that prevents, to some extent, the infection from increasing. The infection may be prevented completely or partially.
  • the methods of the inention involve, in some aspects, passively immunizing a mammalian subject against an influenza virus infection.
  • the method involves administering to the subject a composition comprising at least one RNA polynucleotide having an open reading frame encoding at least one HCAb that targets (e.g., binds to) an influenza virus protein.
  • methods of the present disclosure provide prophylactic treatments against an influenza virus infection.
  • the influenza virus is a type A influenza virus.
  • the influenza virus is any influenza virus comprising HA proteins of subtype H1 and/or H3 (e.g., H1N1, H3N2).
  • Therapeutic methods of treatment are also included within the invention.
  • Methods of treating an influenza virus infection in a subject are provided in aspects of the disclosure.
  • the method involves administering to the subject having an influenza virus infection a composition comprising at least one RNA polynucleotide having an open reading frame encoding at least one HCAb that targets (e.g., binds to) an influenza virus protein.
  • the influenza virus protein is an HA protein.
  • the influenza virus is a type A influenza virus.
  • the influenza virus is any influenza virus comprising HA proteins of subtype H1 and/or H3 (e.g., H1N1, H3N2).
  • the polynucleotide encodes an amino acid sequence of an antibody that binds Ebola virus (EBOV) protein and comprises at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97% 98%, or 99% identity with any of the amino acid sequences provided in the Tables disclosing ebola antibodies.
  • EBOV Ebola virus
  • compositions comprising RNA polynucleotides encoding single-domain antibodies.
  • the single domain antibody encoded by an RNA polynucleotide of the present application is a heavy chain antibody such as found in camelidae (e.g., camels and llamas).
  • the binding elements of such heavy chain antibodies consist of a single polypeptide domain, known as the variable domain of heavy chain antibodies (V H H). These antibody fragments are naturally devoid of light-chains, with the V H H forming the entirety of the antigen-binding site.
  • V H H domains are the preferred types of molecules for immuno-affinity purification due to their high stability and ability to refold efficiently after complete denaturation, which frequently occurs during elution of antigen. Additionally, the smaller size and single domain make V H H domains optimal for cellular transformation.
  • Exemplary polynucleotides include antibody- encoding mRNA polynucleotides.
  • the RNA treatment of the disclosure is a polynucleotide encoding an antibody that binds to Ebola virus (EBOV) protein.
  • Ebola virus There are five Ebola viruses within the genus Ebolavirus. Four of the five known ebolaviruses cause a severe and often fatal hemorrhagic fever in humans and other mammals, known as Ebola virus disease (EVD).
  • Ebola glycoprotein (GP) is the only virally expressed protein on the virion surface, where it is essential for the attachment to host cells and catalyzes membrane fusion.
  • the Ebola GP is a critical component of vaccines, as well as a target of neutralizing antibodies and inhibitors of attachment and fusion.
  • Pre-GP is cleaved by furin at a multi-basic motif into two subunits, GP1 and GP2, which remain associated through a disulfide linkage between Cys53 of GP1 and Cys609 of GP2.
  • the heterodimer (GP1 and GP2) then assembles into a 450-kDa trimer (3 GP1 and 3 GP2) at the surface of nascent virions, where it exerts its functions.
  • the single-domain antibody encoded by the nucleic acid composition targets (e.g., binds to) an EBOV glycoprotein (GP). In some embodiments, the single-domain antibody targets (e.g., binds to) surface GP. In some embodiments, the single- domain antibody targets (e.g., binds to) secreted GP (sGP). In some embodiments, the single-domain antibody targets (e.g., binds to) small sGP (ssGP). In some embodiments, the single-domain antibody targets (e.g., binds to) shed GP.
  • GP EBOV glycoprotein
  • the single- domain antibody encoded by the nucleic acid composition targets (e.g., binds to) an EBOV nucleoprotein. In some embodiments, the single-domain antibody encoded by the nucleic acid composition targets (e.g., binds to) an EBOV matrix protein.
  • the terms treat, treated, or treating when used with respect to a disorder such as a viral infection refers to a treatment which increases the resistance of a subject to development of the disease or, in other words, decreases the likelihood that the subject will develop the disease in response to infection with the virus as well as a treatment after the subject has developed the disease in order to fight the infection or prevent the infection from becoming worse.
  • An“effective amount” of an antibody RNA treatment is provided based, at least in part, on the target tissue, target cell type, means of administration, physical characteristics of the polynucleotide (e.g., size, and extent of modified nucleosides), and other components of the RNA treatment, and other determinants.
  • Increased antibody production may be demonstrated by increased cell transfection (the percentage of cells transfected with the RNA treatment), increased protein translation from the polynucleotide, decreased nucleic acid degradation (as demonstrated, for example, by increased duration of protein translation from a modified polynucleotide), or altered response of the host cell.
  • RNA treatments in accordance with the present disclosure may be used for treatment of the disease.
  • RNA treatments may be administered prophylactically or therapeutically as part of an active immunization scheme to healthy individuals or early in infection during the incubation phase or during active infection after onset of symptoms.
  • the amount of RNA treatments of the present disclosure provided to a cell, a tissue or a subject may be an amount effective for immune prophylaxis.
  • RNA treatments may be administered with other prophylactic or therapeutic compounds.
  • a prophylactic or therapeutic compound may be a vaccine containing an virus treatment with or without an adjuvant or a booster.
  • the term “booster” refers to an extra administration of the prophylactic composition.
  • a booster (or booster vaccine) may be given after an earlier administration of the prophylactic composition.
  • the time of administration between the initial administration of the prophylactic composition and the booster may be, but is not limited to, 1 minute, 2 minutes, 3 minutes, 4 minutes, 5 minutes, 6 minutes, 7 minutes, 8 minutes, 9 minutes, 10 minutes, 15 minutes, 20 minutes 35 minutes, 40 minutes, 45 minutes, 50 minutes, 55 minutes, 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours, 12 hours, 13 hours, 14 hours, 15 hours, 16 hours, 17 hours, 18 hours, 19 hours, 20 hours, 21 hours, 22 hours, 23 hours, 1 day, 36 hours, 2 days, 3 days, 4 days, 5 days, 6 days, 1 week, 10 days, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 1 year, 18 months, 2 years, 3 years, 4 years, 5 years, 6 years, 7 years, 8 years, 9 years, 10 years, 11 years, 12 years, 13 years, 14
  • RNA treatments may be administered subcutaneously, intraocularly, intravitreally, parenterally, subcutaneously, intravenously, intracerebro- ventricularly, intramuscularly, intrathecally, orally, intraperitoneally, by oral or nasal inhalation, or by direct injection to one or more cells, tissues, or organs.
  • compositions including RNA treatments and RNA compositions and/or complexes optionally in combination with one or more
  • RNA treatments may be formulated or administered in combination with one or more pharmaceutically-acceptable excipients.
  • compositions comprise at least one additional active substances, such as, for example, a therapeutically-active substance, a prophylactically-active substance, or a combination of both.
  • Treatment compositions may be sterile, pyrogen-free or both sterile and pyrogen-free. General considerations in the formulation and/or manufacture of pharmaceutical agents, such as treatment compositions, may be found, for example, in Remington: The Science and Practice of Pharmacy 21st ed., Lippincott Williams & Wilkins, 2005 (incorporated herein by reference in its entirety).
  • RNA treatments are administered to humans, human patients, or subjects.
  • the phrase“active ingredient” generally refers to the RNA treatments or the polynucleotides contained therein, for example, RNA polynucleotides (e.g., mRNA polynucleotides) encoding HCAb polypeptides.
  • Formulations of the compositions described herein may be prepared by any method known or hereafter developed in the art of pharmacology.
  • preparatory methods include the step of bringing the active ingredient (e.g., mRNA polynucleotide) into association with an excipient and/or one or more other accessory ingredients, and then, if necessary and/or desirable, dividing, shaping and/or packaging the product into a desired single- or multi-dose unit.
  • active ingredient e.g., mRNA polynucleotide
  • compositions in accordance with the 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.
  • the composition may comprise between 0.1% and 100%, e.g., between 0.5 and 50%, between 1-30%, between 5-80%, at least 80% (w/w) active ingredient.
  • RNA treatments can be formulated using one or more excipients to: (1) increase stability; (2) increase cell transfection; (3) permit the sustained or delayed release (e.g., from a depot formulation); (4) alter the biodistribution (e.g., target to specific tissues or cell types); (5) increase the translation of encoded protein in vivo; and/or (6) alter the release profile of encoded protein (e.g., HCAb) in vivo.
  • excipients to: (1) increase stability; (2) increase cell transfection; (3) permit the sustained or delayed release (e.g., from a depot formulation); (4) alter the biodistribution (e.g., target to specific tissues or cell types); (5) increase the translation of encoded protein in vivo; and/or (6) alter the release profile of encoded protein (e.g., HCAb) in vivo.
  • excipients can include, without limitation, lipidoids, liposomes, lipid nanoparticles, polymers, lipoplexes, core-shell nanoparticles, peptides, proteins, cells transfected with RNA treatments (e.g., for transplantation into a subject), hyaluronidase, nanoparticle mimics, and
  • Naturally-occurring eukaryotic mRNA molecules have been found to contain stabilizing elements, including, but not limited to untranslated regions (UTR) at their 5′-end (5′UTR) and/or at their 3′-end (3′UTR), in addition to other structural features, such as a 5′- cap structure or a 3′-poly(A) tail.
  • UTR untranslated regions
  • 3′UTR 3′-end
  • Both the 5′UTR and the 3′UTR are typically transcribed from the genomic DNA and are elements of the premature mRNA.
  • Characteristic structural features of mature mRNA, such as the 5′-cap and the 3′-poly(A) tail are usually added to the transcribed (premature) mRNA during mRNA processing.
  • the 3′-poly(A) tail is typically a stretch of adenine nucleotides added to the 3′-end of the transcribed mRNA. It can comprise up to about 400 adenine nucleotides. In some embodiments the length of the 3′-poly(A) tail may be an essential element with respect to the stability of the individual mRNA.
  • the RNA treatment may include one or more stabilizing elements.
  • Stabilizing elements may include, for instance, a histone stem-loop.
  • a stem-loop binding protein (SLBP) a 32 kDa protein has been identified. It is associated with the histone stem-loop at the 3′-end of the histone messages in both the nucleus and the cytoplasm. Its expression level is regulated by the cell cycle; it is peaks during the S-phase, when histone mRNA levels are also elevated. The protein has been shown to be essential for efficient 3′-end processing of histone pre-mRNA by the U7 snRNP.
  • SLBP continues to be associated with the stem-loop after processing, and then stimulates the translation of mature histone mRNAs into histone proteins in the cytoplasm.
  • the RNA binding domain of SLBP is conserved through metazoa and protozoa; its binding to the histone stem-loop depends on the structure of the loop.
  • the minimum binding site includes at least three nucleotides 5′ and two nucleotides 3′ relative to the stem-loop.
  • the RNA treatments include a coding region, at least one histone stem-loop, and optionally, a poly(A) sequence or polyadenylation signal.
  • the poly(A) sequence or polyadenylation signal generally should enhance the expression level of the encoded protein.
  • the encoded protein in some embodiments, is not a histone protein, a reporter protein (e.g. Luciferase, GFP, EGFP, ⁇ -Galactosidase, EGFP), or a marker or selection protein (e.g. alpha-Globin, Galactokinase and Xanthine:guanine phosphoribosyl transferase (GPT)).
  • a reporter protein e.g. Luciferase, GFP, EGFP, ⁇ -Galactosidase, EGFP
  • a marker or selection protein e.g. alpha-Globin, Galactokinase and Xanthine:guanine phospho
  • the combination of a poly(A) sequence or polyadenylation signal and at least one histone stem-loop acts synergistically to increase the protein expression beyond the level observed with either of the individual elements. It has been found that the synergistic effect of the combination of poly(A) and at least one histone stem-loop does not depend on the order of the elements or the length of the poly(A) sequence.
  • the invention provides compounds, compositions and methods of use thereof for reducing the effect of ABC on a repeatedly administered active agent such as a biologically active agent.
  • a repeatedly administered active agent such as a biologically active agent.
  • reducing or eliminating altogether the effect of ABC on an administered active agent effectively increases its half-life and thus its efficacy.
  • the term reducing ABC refers to any reduction in ABC in comparison to a positive reference control ABC inducing LNP such as an MC3 LNP.
  • ABC inducing LNPs cause a reduction in circulating levels of an active agent upon a second or subsequent administration within a given time frame.
  • a reduction in ABC refers to less clearance of circulating agent upon a second or subsequent dose of agent, relative to a standard LNP.
  • the reduction may be, for instance, at least 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, or 100%.
  • the reduction is 10-100%, 10-50%, 20-100%, 20-50%, 30-100%, 30-50%, 40%-100%, 40-80%, 50-90%, or 50-100%.
  • the reduction in ABC may be characterized as at least a detectable level of circulating agent following a second or subsequent administration or at least a 2 fold, 3 fold, 4 fold, 5 fold increase in circulating agent relative to circulating agent following administration of a standard LNP.
  • the reduction is a 2-100 fold, 2-50 fold, 3-100 fold, 3-50 fold, 3-20 fold, 4-100 fold, 4-50 fold, 4-40 fold, 4-30 fold, 4-25 fold, 4-20 fold, 4-15 fold, 4-10 fold, 4-5 fold, 5 - 100 fold, 5-50 fold, 5-40 fold, 5-30 fold, 5-25 fold, 5-20 fold, 5-15 fold, 5-10 fold, 6-100 fold, 6-50 fold, 6-40 fold, 6-30 fold, 6-25 fold, 6-20 fold, 6-15 fold, 6-10 fold, 8-100 fold, 8- 50 fold, 8-40 fold, 8-30 fold, 8-25 fold, 8-20 fold, 8-15 fold, 8-10 fold, 10-100 fold, 10-50 fold, 10-40 fold, 10-30 fold, 10-25 fold, 10-20 fold, 10-15 fold, 20-100 fold, 20-50 fold, 20- 40 fold, 20-30 fold, or 20-25 fold.
  • the disclosure provides lipid-comprising compounds and compositions that are less susceptible to clearance and thus have a longer half-life in vivo. This is particularly the case where the compositions are intended for repeated including chronic administration, and even more particularly where such repeated administration occurs within days or weeks.
  • ABC accelerated blood clearance
  • This disclosure provides compounds and compositions that are less susceptible, if at all susceptible, to ABC.
  • such compounds and compositions are lipid-comprising compounds or compositions.
  • the lipid-containing compounds or compositions of this disclosure surprisingly, do not experience ABC upon second and subsequent administration in vivo.
  • This resistance to ABC renders these compounds and compositions particularly suitable for repeated use in vivo, including for repeated use within short periods of time, including days or 1-2 weeks.
  • This enhanced stability and/or half-life is due, in part, to the inability of these compositions to activate B1a and/or B1b cells and/or conventional B cells, pDCs and/or platelets.
  • ABC accelerated blood clearance
  • lipidated agents or lipid- comprising formulations such as lipid nanoparticles administered in vivo trigger and are subject to ABC.
  • sensors one or more cells involved in generating an innate immune response
  • effectors a cascade of immune factors that promote ABC and toxicity.
  • B1a and B1b cells may bind to LNP, become activated (alone or in the presence of other sensors such as pDC and/or effectors such as IL6) and secrete natural IgM that binds to the LNP.
  • Pre-existing natural IgM in the subject may also recognize and bind to the LNP, thereby triggering complement fixation.
  • the production of natural IgM begins within 1-2 hours of administration of the LNP. Typically by about 2-3 weeks the natural IgM is cleared from the system due to the natural half-life of IgM.
  • Natural IgG is produced beginning around 96 hours after administration of the LNP.
  • the agent when administered in a na ⁇ ve setting, can exert its biological effects relatively unencumbered by the natural IgM produced post-activation of the B1a cells or B1b cells or natural IgG.
  • the natural IgM and natural IgG are non-specific and thus are distinct from anti-PEG IgM and anti-PEG IgG.
  • LNPs trigger ABC and/or toxicity through the following mechanisms. It is believed that when an LNP is administered to a subject the LNP is rapidly transported through the blood to the spleen. The LNPs may encounter immune cells in the blood and/or the spleen. A rapid innate immune response is triggered in response to the presence of the LNP within the blood and/or spleen. Applicant has shown herein that within hours of administration of an LNP several immune sensors have reacted to the presence of the LNP. These sensors include but are not limited to immune cells involved in generating an immune response, such as B cells, pDC, and platelets.
  • the sensors may be present in the spleen, such as in the marginal zone of the spleen and/or in the blood.
  • the LNP may physically interact with one or more sensors, which may interact with other sensors. In such a case the LNP is directly or indirectly interacting with the sensors.
  • the sensors may interact directly with one another in response to recognition of the LNP. For instance many sensors are located in the spleen and can easily interact with one another. Alternatively one or more of the sensors may interact with LNP in the blood and become activated. The activated sensor may then interact directly with other sensors or indirectly (e.g., through the stimulation or production of a messenger such as a cytokine e.g., IL6).
  • a messenger such as a cytokine e.g., IL6
  • the LNP may interact directly with and activate each of the following sensors: pDC, B1a cells, B1b cells, and platelets. These cells may then interact directly or indirectly with one another to initiate the production of effectors which ultimately lead to the ABC and/or toxicity associated with repeated doses of LNP.
  • pDC pDC
  • B1a cells B1a cells
  • B1b cells platelets
  • platelets pDC cells
  • LNP has been found to interact with the surface of platelets and B cells relatively quickly. Blocking the activation of any one or combination of these sensors in response to LNP is useful for dampening the immune response that would ordinarily occur. This dampening of the immune response results in the avoidance of ABC and/or toxicity.
  • An effector is an immune molecule produced by an immune cell, such as a B cell.
  • Effectors include but are not limited to immunoglobulin such as natural IgM and natural IgG and cytokines such as IL6.
  • B1a and B1b cells stimulate the production of natural IgMs within 2-6 hours following administration of an LNP.
  • Natural IgG can be detected within 96 hours.
  • IL6 levels are increased within several hours.
  • the natural IgM and IgG circulate in the body for several days to several weeks. During this time the circulating effectors can interact with newly administered LNPs, triggering those LNPs for clearance by the body. For instance, an effector may recognize and bind to an LNP.
  • the Fc region of the effector may be recognized by and trigger uptake of the decorated LNP by macrophage.
  • the macrophage are then transported to the spleen.
  • the production of effectors by immune sensors is a transient response that correlates with the timing observed for ABC.
  • the administered dose is the second or subsequent administered dose, and if such second or subsequent dose is administered before the previously induced natural IgM and/or IgG is cleared from the system (e.g., before the 2-3 window time period), then such second or subsequent dose is targeted by the circulating natural IgM and/or natural IgG or Fc which trigger alternative complement pathway activation and is itself rapidly cleared.
  • LNP are administered after the effectors have cleared from the body or are reduced in number, ABC is not observed.
  • LNP is designed to limit or block interaction of the LNP with a sensor.
  • the LNP may have an altered PC and/or PEG to prevent interactions with sensors.
  • an agent that inhibits immune responses induced by LNPs may be used to achieve any one or more of these effects.
  • conventional B cells are also implicated in ABC. Specifically, upon first administration of an agent, conventional B cells, referred to herein as CD19(+), bind to and react against the agent. Unlike B1a and B1b cells though, conventional B cells are able to mount first an IgM response (beginning around 96 hours after
  • IgG immunoglobulin G
  • conventional B cells react against the administered agent and contribute to IgM (and eventually IgG) that mediates ABC.
  • the IgM and IgG are typically anti-PEG IgM and anti-PEG IgG.
  • the majority of the ABC response is mediated through B1a cells and B1a-mediated immune responses. It is further contemplated that in some instances, the ABC response is mediated by both IgM and IgG, with both conventional B cells and B1a cells mediating such effects. In yet still other instances, the ABC response is mediated by natural IgM molecules, some of which are capable of binding to natural IgM, which may be produced by activated B1a cells.
  • the natural IgMs may bind to one or more components of the LNPs, e.g., binding to a phospholipid component of the LNPs (such as binding to the PC moiety of the phospholipid) and/or binding to a PEG-lipid component of the LNPs (such as binding to PEG-DMG, in particular, binding to the PEG moiety of PEG-DMG).
  • B1a expresses CD36, to which phosphatidylcholine is a ligand, it is contemplated that the CD36 receptor may mediate the activation of B1a cells and thus production of natural IgM.
  • the ABC response is mediated primarily by conventional B cells.
  • the ABC phenomenon can be reduced or abrogated, at least in part, through the use of compounds and compositions (such as agents, delivery vehicles, and formulations) that do not activate B1a cells.
  • compounds and compositions such as agents, delivery vehicles, and formulations
  • B1a inert compounds and compositions Compounds and compositions that do not activate B1a cells
  • the ABC phenomenon can be reduced or abrogated, at least in part, through the use of compounds and compositions that do not activate conventional B cells.
  • Compounds and compositions that do not activate conventional B cells may in some embodiments be referred to herein as CD19-inert compounds and compositions.
  • the compounds and compositions do not activate B1a cells and they do not activate conventional B cells.
  • Compounds and compositions that do not activate B1a cells and conventional B cells may in some embodiments be referred to herein as B1a/CD19-inert compounds and compositions.
  • this disclosure provides compounds and compositions that do not promote ABC. These may be further characterized as not capable of activating B1a and/or B1b cells, platelets and/or pDC, and optionally conventional B cells also.
  • These compounds e.g., agents, including biologically active agents such as prophylactic agents, therapeutic agents and diagnostic agents, delivery vehicles, including liposomes, lipid nanoparticles, and other lipid-based encapsulating structures, etc.
  • compositions e.g., formulations, etc.
  • the agent is a nucleic acid based therapeutic that is provided to a subject at regular, closely-spaced intervals.
  • the findings provided herein may be applied to these and other agents that are similarly administered and/or that are subject to ABC.
  • lipid-comprising compounds lipid-comprising particles
  • lipid-comprising compositions as these are known to be susceptible to ABC.
  • Such lipid- comprising compounds particles, and compositions have been used extensively as
  • compositions that do not stimulate or boost an acute phase response (ARP) associated with repeat dose administration of one or more biologically active agents.
  • ARP acute phase response
  • the composition in some instances, may not bind to IgM, including but not limited to natural IgM.
  • composition in some instances, may not bind to an acute phase protein such as but not limited to C-reactive protein.
  • composition in some instances, may not trigger a CD5(+) mediated immune response.
  • a CD5(+) mediated immune response is an immune response that is mediated by B1a and/or B1b cells. Such a response may include an ABC response, an acute phase response, induction of natural IgM and/or IgG, and the like.
  • composition in some instances, may not trigger a CD19(+) mediated immune response.
  • a CD19(+) mediated immune response is an immune response that is mediated by conventional CD19(+), CD5(-) B cells.
  • Such a response may include induction of IgM, induction of IgG, induction of memory B cells, an ABC response, an anti- drug antibody (ADA) response including an anti-protein response where the protein may be encapsulated within an LNP, and the like.
  • B1a cells are a subset of B cells involved in innate immunity. These cells are the source of circulating IgM, referred to as natural antibody or natural serum antibody. Natural IgM antibodies are characterized as having weak affinity for a number of antigens, and therefore they are referred to as“poly-specific” or“poly-reactive”, indicating their ability to bind to more than one antigen. B1a cells are not able to produce IgG. Additionally, they do not develop into memory cells and thus do not contribute to an adaptive immune response. However, they are able to secrete IgM upon activation. The secreted IgM is typically cleared within about 2-3 weeks, at which point the immune system is rendered relatively na ⁇ ve to the previously administered antigen.
  • the antigen is not rapidly cleared. However, significantly, if the antigen is presented within that time period (e.g., within 2 weeks, including within 1 week, or within days), then the antigen is rapidly cleared. This delay between consecutive doses has rendered certain lipid-containing therapeutic or diagnostic agents unsuitable for use.
  • B1a cells are CD19(+), CD20(+), CD27(+), CD43(+), CD70(-) and CD5(+).
  • B1a cells are CD19(+), CD5(+), and CD45 B cell isoform B220(+). It is the expression of CD5 which typically distinguishes B1a cells from other convention B cells. B1a cells may express high levels of CD5, and on this basis may be distinguished from other B-1 cells such as B-1b cells which express low or undetectable levels of CD5.
  • CD5 is a pan- T cell surface glycoprotein.
  • B1a cells also express CD36, also known as fatty acid translocase.
  • CD36 is a member of the class B scavenger receptor family. CD36 can bind many ligands, including oxidized low density lipoproteins, native lipoproteins, oxidized phospholipids, and long-chain fatty acids.
  • B1b cells are another subset of B cells involved in innate immunity. These cells are another source of circulating natural IgM.
  • antigens including PS, are capable of inducing T cell independent immunity through B1b activation.
  • CD27 is typically upregulated on B1b cells in response to antigen activation.
  • the B1b cells are typically located in specific body locations such as the spleen and peritoneal cavity and are in very low abundance in the blood.
  • the B1b secreted natural IgM is typically cleared within about 2-3 weeks, at which point the immune system is rendered relatively na ⁇ ve to the previously administered antigen. If the same antigen is presented after this time period (e.g., at about 3 weeks after the initial exposure), the antigen is not rapidly cleared.
  • the antigen is presented within that time period (e.g., within 2 weeks, including within 1 week, or within days), then the antigen is rapidly cleared. This delay between consecutive doses has rendered certain lipid-containing therapeutic or diagnostic agents unsuitable for use.
  • B1a and/or B1b cell activation it is desirable to block B1a and/or B1b cell activation.
  • One strategy for blocking B1a and/or B1b cell activation involves determining which components of a lipid nanoparticle promote B cell activation and neutralizing those components. It has been discovered herein that at least PEG and phosphatidylcholine (PC) contribute to B1a and B1b cell interaction with other cells and/or activation. PEG may play a role in promoting aggregation between B1 cells and platelets, which may lead to activation.
  • PC a helper lipid in LNPs
  • PEG-lipid alternatives e.g. oleic acid or analogs thereof
  • PC replacement lipids e.g. oleic acid or analogs thereof
  • Applicant has established that replacement of one or more of these components within an LNP that otherwise would promote ABC upon repeat administration, is useful in preventing ABC by reducing the production of natural IgM and/or B cell activation.
  • the invention encompasses LNPs that have reduced ABC as a result of a design which eliminates the inclusion of B cell triggers.
  • Another strategy for blocking B1a and/or B1b cell activation involves using an agent that inhibits immune responses induced by LNPs.
  • agents block the interaction between B1a/B1b cells and the LNP or platelets or pDC.
  • the agent may be an antibody or other binding agent that physically blocks the interaction.
  • An example of this is an antibody that binds to CD36 or CD6.
  • the agent may also be a compound that prevents or disables the B1a/B1b cell from signaling once activated or prior to activation.
  • the agent may act one or more effectors produced by the B1a/B1b cells following activation. These effectors include for instance, natural IgM and cytokines.
  • pDC cell activation may be blocked by agents that interfere with the interaction between pDC and LNP and/or B cells/platelets.
  • agents that act on the pDC to block its ability to get activated or on its effectors can be used together with the LNP to avoid ABC.
  • Platelets may also play an important role in ABC and toxicity. Very quickly after a first dose of LNP is administered to a subject platelets associate with the LNP, aggregate and are activated. In some embodiments it is desirable to block platelet aggregation and/or activation.
  • One strategy for blocking platelet aggregation and/or activation involves determining which components of a lipid nanoparticle promote platelet aggregation and/or activation and neutralizing those components. It has been discovered herein that at least PEG contribute to platelet aggregation, activation and/or interaction with other cells. Numerous particles have PEG-lipid alternatives and PEG-less have been designed and tested.
  • the invention encompasses LNPs that have reduced ABC as a result of a design which eliminates the inclusion of platelet triggers.
  • agents that act on the platelets to block its activity once it is activated or on its effectors can be used together with the LNP to avoid ABC. Measuring ABC Activity and related activities
  • LNPs do not promote ABC activity upon administration in vivo.
  • LNPs may be characterized and/or identified through any of a number of assays, such as but not limited to those described below, as well as any of the assays disclosed in the Examples section, include the methods subsection of the Examples.
  • the methods involve administering an LNP without producing an immune response that promotes ABC.
  • An immune response that promotes ABC involves activation of one or more sensors, such as B1 cells, pDC, or platelets, and one or more effectors, such as natural IgM, natural IgG or cytokines such as IL6.
  • administration of an LNP without producing an immune response that promotes ABC at a minimum involves administration of an LNP without significant activation of one or more sensors and significant production of one or more effectors.
  • Significant used in this context refers to an amount that would lead to the physiological consequence of accelerated blood clearance of all or part of a second dose with respect to the level of blood clearance expected for a second dose of an ABC triggering LNP.
  • the immune response should be dampened such that the ABC observed after the second dose is lower than would have been expected for an ABC triggering LNP.
  • B cells such as B1a or B1b cells (CD19+ CD5+) and/or conventional B cells (CD19+ CD5-).
  • Activation of B1a cells, B1b cells, or conventional B cells may be determined in a number of ways, some of which are provided below.
  • B cell population may be provided as fractionated B cell populations or unfractionated populations of splenocytes or peripheral blood mononuclear cells (PBMC). If the latter, the cell population may be incubated with the LNP of choice for a period of time, and then harvested for further analysis. Alternatively, the supernatant may be harvested and analyzed. Upregulation of activation marker cell surface expression
  • B1a cells, B1b cells, or conventional B cells may be demonstrated as increased expression of B cell activation markers including late activation markers such as CD86.
  • B cell activation markers including late activation markers such as CD86.
  • unfractionated B cells are provided as a splenocyte population or as a PBMC population, incubated with an LNP of choice for a particular period of time, and then stained for a standard B cell marker such as CD19 and for an activation marker such as CD86, and analyzed using for example flow cytometry.
  • a suitable negative control involves incubating the same population with medium, and then performing the same staining and visualization steps. An increase in CD86 expression in the test population compared to the negative control indicates B cell activation.
  • Pro-inflammatory cytokine release indicates B cell activation.
  • B cell activation may also be assessed by cytokine release assay.
  • activation may be assessed through the production and/or secretion of cytokines such as IL-6 and/or TNF-alpha upon exposure with LNPs of interest.
  • Such assays may be performed using routine cytokine secretion assays well known in the art.
  • An increase in cytokine secretion is indicative of B cell activation. LNP binding/association to and/or uptake by B cells
  • LNP association or binding to B cells may also be used to assess an LNP of interest and to further characterize such LNP.
  • Association/binding and/or uptake/internalization may be assessed using a detectably labeled, such as fluorescently labeled, LNP and tracking the location of such LNP in or on B cells following various periods of incubation.
  • compositions provided herein may be capable of evading recognition or detection and optionally binding by downstream mediators of ABC such as circulating IgM and/or acute phase response mediators such as acute phase proteins (e.g., C-reactive protein (CRP).
  • downstream mediators of ABC such as circulating IgM and/or acute phase response mediators such as acute phase proteins (e.g., C-reactive protein (CRP).
  • acute phase proteins e.g., C-reactive protein (CRP).
  • LNPs which may encapsulate an agent such as a therapeutic agent, to a subject without promoting ABC.
  • the method comprises administering any of the LNPs described herein, which do not promote ABC, for example, do not induce production of natural IgM binding to the LNPs, do not activate B1a and/or B1b cells.
  • an LNP that“does not promote ABC” refers to an LNP that induces no immune responses that would lead to substantial ABC or a substantially low level of immune responses that is not sufficient to lead to substantial ABC.
  • An LNP that does not induce the production of natural IgMs binding to the LNP refers to LNPs that induce either no natural IgM binding to the LNPs or a substantially low level of the natural IgM molecules, which is insufficient to lead to substantial ABC.
  • An LNP that does not activate B1a and/or B1b cells refer to LNPs that induce no response of B1a and/or B1b cells to produce natural IgM binding to the LNPs or a substantially low level of B1a and/or B1b responses, which is insufficient to lead to substantial ABC.
  • the terms do not activate and do not induce production are a relative reduction to a reference value or condition.
  • the reference value or condition is the amount of activation or induction of production of a molecule such as IgM by a standard LNP such as an MC3 LNP.
  • the relative reduction is a reduction of at least 30%, for example at least 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%.
  • the terms do not activate cells such as B cells and do not induce production of a protein such as IgM may refer to an undetectable amount of the active cells or the specific protein. Platelet effects and toxicity
  • the invention is further premised in part on the elucidation of the mechanism underlying dose-limiting toxicity associated with LNP administration.
  • toxicity may involve coagulopathy, disseminated intravascular coagulation (DIC, also referred to as consumptive coagulopathy), whether acute or chronic, and/or vascular thrombosis.
  • DIC disseminated intravascular coagulation
  • the dose-limiting toxicity associated with LNPs is acute phase response (APR) or complement activation-related psudoallergy (CARPA).
  • coagulopathy refers to increased coagulation (blood clotting) in vivo.
  • the findings reported in this disclosure are consistent with such increased coagulation and significantly provide insight on the underlying mechanism.
  • Coagulation is a process that involves a number of different factors and cell types, and heretofore the relationship between and interaction of LNPs and platelets has not been understood in this regard.
  • This disclosure provides evidence of such interaction and also provides compounds and compositions that are modified to have reduced platelet effect, including reduced platelet association, reduced platelet aggregation, and/or reduced platelet aggregation.
  • the ability to modulate, including preferably down-modulate, such platelet effects can reduce the incidence and/or severity of coagulopathy post-LNP administration. This in turn will reduce toxicity relating to such LNP, thereby allowing higher doses of LNPs and importantly their cargo to be administered to patients in need thereof.
  • CARPA is a class of acute immune toxicity manifested in hypersensitivity reactions (HSRs), which may be triggered by nanomedicines and biologicals. Unlike allergic reactions, CARPA typically does not involve IgE but arises as a consequence of activation of the complement system, which is part of the innate immune system that enhances the body’s abilities to clear pathogens.
  • HSRs hypersensitivity reactions
  • CARPA typically does not involve IgE but arises as a consequence of activation of the complement system, which is part of the innate immune system that enhances the body’s abilities to clear pathogens.
  • CARPA complement pathway
  • AP alternative pathway
  • LP lectin pathway
  • the classical pathway is triggered by activation of the C1-complex, which contains. C1q, C1r, C1s, or C1qr2s2.
  • Activation of the C1-complex occurs when C1q binds to IgM or IgG complexed with antigens, or when C1q binds directly to the surface of the pathogen.
  • Such binding leads to conformational changes in the C1q molecule, which leads to the activation of C1r, which in turn, cleave C1s.
  • the C1r2s2 component now splits C4 and then C2, producing C4a, C4b, C2a, and C2b.
  • C4b and C2b bind to form the classical pathway C3- convertase (C4b2b complex), which promotes cleavage of C3 into C3a and C3b.
  • C3b then binds the C3 convertase to from the C5 convertase (C4b2b3b complex).
  • the alternative pathway is continuously activated as a result of spontaneous C3 hydrolysis.
  • APR Acute phase response
  • lipid nanoparticles are provided.
  • a lipid nanoparticle comprises lipids including an ionizable lipid, a structural lipid, a phospholipid, and mRNA.
  • Each of the LNPs described herein may be used as a formulation for the mRNA described herein.
  • a lipid nanoparticle comprises an ionizable lipid, a structural lipid, a phospholipid, and mRNA.
  • the LNP comprises an ionizable lipid, a PEG-modified lipid, a phospholipid and a structural lipid.
  • the LNP has a molar ratio of about 20-60% ionizable lipid: about 5-25% phospholipid: about 25-55% structural lipid; and about 0.5-15% PEG-modified lipid. In some embodiments, the LNP comprises a molar ratio of about 50% ionizable lipid, about 1.5% PEG-modified lipid, about 38.5% structural lipid and about 10% phospholipid. In some embodiments, the LNP comprises a molar ratio of about 55% ionizable lipid, about 2.5% PEG lipid, about 32.5% structural lipid and about 10%
  • the ionizable lipid is an ionizable amino or cationic lipid and the phospholipid is a neutral lipid, and the structural lipid is a cholesterol.
  • the LNP has a molar ratio of 50:38.5:10:1.5 of ionizable lipid:
  • the present disclosure provides pharmaceutical compositions with advantageous properties.
  • the lipids described herein e.g. those having any of Formula (I), (IA), (II), (IIa), (IIb), (IIc), (IId), (IIe), (III), (IV), (V), or (VI) may be advantageously used in lipid nanoparticle compositions for the delivery of therapeutic and/or prophylactic agents to mammalian cells or organs.
  • the lipids described herein have little or no immunogenicity.
  • the lipid compounds disclosed herein have a lower immunogenicity as compared to a reference lipid (e.g., MC3, KC2, or DLinDMA).
  • a formulation comprising a lipid disclosed herein and a therapeutic or prophylactic agent has an increased therapeutic index as compared to a corresponding formulation which comprises a reference lipid (e.g., MC3, KC2, or DLinDMA) and the same therapeutic or prophylactic agent.
  • a reference lipid e.g., MC3, KC2, or DLinDMA
  • the present application provides pharmaceutical
  • compositions comprising:
  • nucleic acids of the invention are formulated in a lipid nanoparticle (LNP).
  • LNP lipid nanoparticle
  • Lipid nanoparticles typically comprise ionizable cationic lipid, non-cationic lipid, sterol and PEG lipid components along with the nucleic acid cargo of interest.
  • the lipid nanoparticles of the invention can be generated using components, compositions, and methods as are generally known in the art, see for example PCT/US2016/052352; PCT/US2016/068300; PCT/US2017/037551; PCT/US2015/027400; PCT/US2016/047406; PCT/US2016000129; PCT/US2016/014280; PCT/US2016/014280; PCT/US2017/038426; PCT/US2014/027077; PCT/US2014/055394; PCT/US2016/52117; PCT/US2012/069610; PCT/US2017/027492; PCT/US2016/059575 and
  • Nucleic acids of the present disclosure are typically formulated in lipid nanoparticle.
  • the lipid nanoparticle comprises at least one ionizable cationic lipid, at least one non-cationic lipid, at least one sterol, and/or at least one polyethylene glycol (PEG)-modified lipid.
  • PEG polyethylene glycol
  • the lipid nanoparticle comprises a molar ratio of 20-60% ionizable cationic lipid.
  • the lipid nanoparticle may comprise a molar ratio of 20-50%, 20-40%, 20-30%, 30-60%, 30-50%, 30-40%, 40-60%, 40-50%, or 50-60% ionizable cationic lipid.
  • the lipid nanoparticle comprises a molar ratio of 20%, 30%, 40%, 50, or 60% ionizable cationic lipid.
  • the lipid nanoparticle comprises a molar ratio of 5-25% non- cationic lipid.
  • the lipid nanoparticle may comprise a molar ratio of 5-20%, 5- 15%, 5-10%, 10-25%, 10-20%, 10-25%, 15-25%, 15-20%, or 20-25% non-cationic lipid.
  • the lipid nanoparticle comprises a molar ratio of 5%, 10%, 15%, 20%, or25% non-cationic lipid.
  • the lipid nanoparticle comprises a molar ratio of 25-55% sterol.
  • the lipid nanoparticle may comprise a molar ratio of 25-50%, 25-45%, 25-40%, 25-35%, 25-30%, 30-55%, 30-50%, 30-45%, 30-40%, 30-35%, 35-55%, 35-50%, 35-45%, 35-40%, 40-55%, 40-50%, 40-45%, 45-55%, 45-50%, or 50-55% sterol.
  • the lipid nanoparticle comprises a molar ratio of 25%, 30%, 35%, 40%, 45%, 50%, or 55% sterol.
  • the lipid nanoparticle comprises a molar ratio of 0.5-15% PEG-modified lipid.
  • the lipid nanoparticle may comprise a molar ratio of 0.5- 10%, 0.5-5%, 1-15%, 1-10%, 1-5%, 2-15%, 2-10%, 2-5%, 5-15%, 5-10%, or 10-15%.
  • the lipid nanoparticle comprises a molar ratio of 0.5%, 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, or 15% PEG-modified lipid.
  • the lipid nanoparticle comprises a molar ratio of 20-60% ionizable cationic lipid, 5-25% non-cationic lipid, 25-55% sterol, and 0.5-15% PEG-modified lipid.
  • the ionizable lipids of the present disclosure may be one or more of compounds of Formula (I):
  • R 1 is selected from the group consisting of C 5-30 alkyl, C 5-20 alkenyl, -R*YR”, -YR”, and -R”M’R’;
  • R 2 and R 3 are independently selected from the group consisting of H, C 1-14 alkyl, C 2-14 alkenyl, -R*YR”, -YR”, and -R*OR”, or R 2 and R 3 , together with the atom to which they are attached, form a heterocycle or carbocycle;
  • R 4 is selected from the group consisting of hydrogen, a C 3-6 carbocycle, -(CH 2 ) n Q, -(CH 2 ) n CHQR, -CHQR, -CQ(R) 2 , and unsubstituted C 1-6 alkyl, where Q is selected from a carbocycle, heterocycle, -OR, -O(CH 2 ) n N(R) 2 , -C(O)OR, -OC(O)R, -CX 3 , -CX 2 H, -CXH 2 , -CN, -N(R) 2 , -C(O)N(R) 2 , -N(R)C(O)R, -N(R)S(O) 2 R, -N(R)C(O)N(R) 2 , -N(R)C(S)N(R) 2 , -N(R)C(S)N(R) 2 , -N(
  • each R 5 is independently selected from the group consisting of C 1-3 alkyl, C 2-3 alkenyl, and H;
  • each R 6 is independently selected from the group consisting of C 1-3 alkyl, C 2-3 alkenyl, and H;
  • M and M’ are independently selected from -C(O)O-, -OC(O)-, -OC(O)-M”-C(O)O-, -C(O)N(R’)-, -N(R’)C(O)-, -C(O)-, -C(S)-, -C(S)S-, -SC(S)-, -CH(OH)-, -P(O)(OR’)O-, -S(O) 2 -, -S-S-, an aryl group, and a heteroaryl group, in which M” is a bond, C 1-13 alkyl or C 2-13 alkenyl;
  • R 7 is selected from the group consisting of C 1-3 alkyl, C 2-3 alkenyl, and H;
  • R 8 is selected from the group consisting of C 3-6 carbocycle and heterocycle
  • R 9 is selected from the group consisting of H, CN, NO 2 , C 1-6 alkyl, -OR, -S(O) 2 R, -S(O) 2 N(R) 2 , C 2-6 alkenyl, C 3-6 carbocycle and heterocycle; each R is independently selected from the group consisting of C 1-3 alkyl, C 2-3 alkenyl, and H;
  • each R’ is independently selected from the group consisting of C 1-18 alkyl, C 2-18 alkenyl, -R*YR”, -YR”, and H;
  • each R is independently selected from the group consisting of C 3-15 alkyl and C 3-15 alkenyl
  • each R* is independently selected from the group consisting of C 1-12 alkyl and C 2-12 alkenyl;
  • each Y is independently a C 3-6 carbocycle
  • each X is independently selected from the group consisting of F, Cl, Br, and I; and m is selected from 5, 6, 7, 8, 9, 10, 11, 12, and 13; and wherein when R 4
  • Q is -(CH 2 ) n Q, -(CH 2 ) n CHQR,–CHQR, or -CQ(R) 2 , then (i) Q is not -N(R) 2 when n is 1, 2, 3, 4 or 5, or (ii) Q is not 5, 6, or 7-membered heterocycloalkyl when n is 1 or 2.
  • a subset of compounds of Formula (I) includes those of Formula (IA):
  • M and M’ are independently selected from -C(O)O-, -OC(O)-, -OC(O)-M”-C(O)O-, -C(O)N(R’)-, -P(O)(OR’)O-, -S-S-, an aryl group, and a heteroaryl group,; and R 2 and R 3 are independently selected from the group consisting of H, C 1-14 alkyl, and C 2-14 alkenyl. For example, m is 5, 7, or 9.
  • Q is OH, -NHC(S)N(R) 2 , or -NHC(O)N(R) 2 .
  • Q is -N(R)C(O)R, or -N(R)S(O) 2 R.
  • a subset of compounds of Formula (I) includes those of Formula (IB): (IB), or its N-oxide, or a salt or isomer thereof in which all variables are as defined herein.
  • m is selected from 5, 6, 7, 8, and 9;
  • R 4 is hydrogen, unsubstituted C 1-3 alkyl, or -(CH 2 ) n Q, in which Q is OH, -NHC(S)N(R) 2 ,
  • M and M’ are independently selected from -C(O)O-, -OC(O)-, -OC(O)-M”-C(O)O-, -C(O)N(R’)-, -P(O)(OR’)O-, -S-S-, an aryl group, and a heteroaryl group; and R 2 and R 3 are independently selected from the group consisting of H, C 1-14 alkyl, and C 2-14 alkenyl. For example, m is 5, 7, or 9.
  • Q is OH, -NHC(S)N(R) 2 , or -NHC(O)N(R) 2 .
  • Q is -N(R)C(O)R, or -N(R)S(O) 2 R.
  • a subset of compounds of Formula (I) includes those of Formula (II):
  • R 2 and R 3 are independently selected from the group consisting of H, C 1-14 alkyl, and C 2-14 alkenyl.
  • the compounds of Formula (I) are of Formula (IIa),
  • the compounds of Formula (I) are of Formula (IIb),
  • the compounds of Formula (I) are of Formula (IIc) or (IIe):
  • the compounds of Formula (I) are of Formula (IIf):
  • M is -C(O)O- or–OC(O)-
  • M is C 1-6 alkyl or C 2-6 alkenyl
  • R 2 and R 3 are independently selected from the group consisting of C 5-14 alkyl and C 5-14 alkenyl
  • n is selected from 2, 3, and 4.
  • the compounds of Formula (I) are of Formula (IId),
  • each of R 2 and R 3 may be independently selected from the group consisting of C 5-14 alkyl and C 5-14 alkenyl.
  • the compounds of Formula (I) are of Formula (IIg),
  • M is C 1-6 alkyl (e.g., C 1-4 alkyl) or C 2-6 alkenyl (e.g. C 2-4 alkenyl).
  • R 2 and R 3 are independently selected from the group consisting of C 5-14 alkyl and C 5-14 alkenyl.
  • the ionizable lipids are one or more of the compounds described in U.S. Application Nos.62/220,091, 62/252,316, 62/253,433, 62/266,460, 62/333,557, 62/382,740, 62/393,940, 62/471,937, 62/471,949, 62/475,140, and 62/475,166, and PCT Application No. PCT/US2016/052352.
  • the ionizable lipids are selected from Compounds 1-280 described in U.S. Application No.62/475,166.
  • the ionizable lipid is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N
  • the ionizable lipid is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N
  • the ionizable lipid is or a salt thereof. In some embodiments, the ionizable lipid is
  • a lipid may have a positive or partial positive charge at physiological pH.
  • Such lipids may be referred to as cationic or ionizable (amino)lipids.
  • Lipids may also be zwitterionic, i.e., neutral molecules having both a positive and a negative charge.
  • the ionizable lipids of the present disclosure may be one or more of compounds of formula (III),
  • t 1 or 2;
  • a 1 and A 2 are each independently selected from CH or N;
  • Z is CH 2 or absent wherein when Z is CH 2 , the dashed lines (1) and (2) each represent a single bond; and when Z is absent, the dashed lines (1) and (2) are both absent;
  • R 1 , R 2 , R 3 , R 4 , and R 5 are independently selected from the group consisting of C 5-20 alkyl, C 5-20 alkenyl, -R”MR’, -R*YR”, -YR”, and -R*OR”;
  • R X1 and R X2 are each independently H or C 1 - 3 alkyl
  • each M is independently selected from the group consisting of
  • M* is C 1 -C 6 alkyl
  • W 1 and W 2 are each independently selected from the group consisting of
  • each R 6 is independently selected from the group consisting of H and C 1-5 alkyl
  • X 1 , X 2 , and X 3 are independently selected from the group consisting of a bond, -CH 2 -, -(CH 2 ) 2 -, -CHR-, -CHY-, -C(O)-, -C(O)O-, -OC(O)-, -(CH 2 ) n -C(O)-, -C(O)-(CH 2 ) n -,
  • each Y is independently a C 3-6 carbocycle
  • each R* is independently selected from the group consisting of C 1-12 alkyl and C 2-12 alkenyl;
  • each R is independently selected from the group consisting of C 1-3 alkyl and a C 3-6 carbocycle;
  • each R’ is independently selected from the group consisting of C 1-12 alkyl, C 2-12 alkenyl, and H;
  • each R is independently selected from the group consisting of C 3-12 alkyl, C 3-12 alkenyl and -R*MR’;
  • n is an integer from 1-6;
  • R 1 , R 2 , R 3 , R 4 , and R 5 is -R”MR’.
  • the compound is of any of formulae (IIIa1)-(IIIa8):
  • the ionizable lipids are one or more of the compounds described in U.S. Application Nos.62/271,146, 62/338,474, 62/413,345, and 62/519,826, and PCT Application No. PCT/US2016/068300.
  • the ionizable lipids are selected from Compounds 1-156 described in U.S. Application No.62/519,826.
  • the ionizable lipids are selected from Compounds 1-16, 42-66, 68-76, and 78-156 described in U.S. Application No.62/519,826.
  • a lipid may have a positive or partial positive charge at physiological pH.
  • Such lipids may be referred to as cationic or ionizable (amino)lipids.
  • Lipids may also be zwitterionic, i.e., neutral molecules having both a positive and a negative charge.
  • the lipid composition of the lipid nanoparticle composition disclosed herein can comprise one or more phospholipids, for example, one or more saturated or (poly)unsaturated phospholipids or a combination thereof.
  • phospholipids comprise a phospholipid moiety and one or more fatty acid moieties.
  • a phospholipid moiety can be selected, for example, from the non-limiting group consisting of phosphatidyl choline, phosphatidyl ethanolamine, phosphatidyl glycerol, phosphatidyl serine, phosphatidic acid, 2-lysophosphatidyl choline, and a sphingomyelin.
  • a fatty acid moiety can be selected, for example, from the non-limiting group consisting of lauric acid, myristic acid, myristoleic acid, palmitic acid, palmitoleic acid, stearic acid, oleic acid, linoleic acid, alpha-linolenic acid, erucic acid, phytanoic acid, arachidic acid, arachidonic acid, eicosapentaenoic acid, behenic acid, docosapentaenoic acid, and docosahexaenoic acid.
  • Particular phospholipids can facilitate fusion to a membrane.
  • a cationic phospholipid can interact with one or more negatively charged phospholipids of a membrane (e.g., a cellular or intracellular membrane). Fusion of a phospholipid to a membrane can allow one or more elements (e.g., a therapeutic agent) of a lipid-containing composition (e.g., LNPs) to pass through the membrane permitting, e.g., delivery of the one or more elements to a target tissue.
  • a membrane e.g., a cellular or intracellular membrane.
  • Non-natural phospholipid species including natural species with modifications and substitutions including branching, oxidation, cyclization, and alkynes are also contemplated.
  • a phospholipid can be functionalized with or cross-linked to one or more alkynes (e.g., an alkenyl group in which one or more double bonds is replaced with a triple bond).
  • an alkyne group can undergo a copper- catalyzed cycloaddition upon exposure to an azide.
  • Such reactions can be useful in functionalizing a lipid bilayer of a nanoparticle composition to facilitate membrane permeation or cellular recognition or in conjugating a nanoparticle composition to a useful component such as a targeting or imaging moiety (e.g., a dye).
  • Phospholipids include, but are not limited to, glycerophospholipids such as phosphatidylcholines, phosphatidylethanolamines, phosphatidylserines,
  • Phospholipids also include phosphosphingolipid, such as sphingomyelin.
  • a phospholipid of the invention comprises 1,2-distearoyl-sn- glycero-3-phosphocholine (DSPC), 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), 1,2-dilinoleoyl-sn-glycero-3-phosphocholine (DLPC), 1,2-dimyristoyl-sn-gly cero- phosphocholine (DMPC), 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), l,2- dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), 1,2-diundecanoyl-sn-glycero- phosphocholine (DUPC), 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC), 1,2-di- O-octadecenyl-sn-glycero-3-phosphocholine (
  • cholesterylhemisuccinoyl-sn-glycero-3-phosphocholine OChemsPC
  • 1-hexadecyl-sn- glycero-3-phosphocholine C16 Lyso PC
  • 1,2-dilinolenoyl-sn-glycero-3-phosphocholine 1,2- diarachidonoyl-sn-glycero-3-phosphocholine, 1,2-didocosahexaenoyl-sn-glycero-3- phosphocholine
  • 1,2-diphytanoyl-sn-glycero-3-phosphoethanolamine ME 16.0 PE
  • 1,2- distearoyl-sn-glycero-3-phosphoethanolamine 1,2-dilinoleoyl-sn-glycero-3- phosphoethanolamine
  • 1,2-dilinolenoyl-sn-glycero-3-phosphoethanolamine 1,2- diarachidonoyl-sn-glycero
  • a phospholipid useful or potentially useful in the present invention is an analog or variant of DSPC. In certain embodiments, a phospholipid useful or potentially useful in the present invention is a compound of Formula (IV):
  • each R 1 is independently optionally substituted alkyl; or optionally two R 1 are joined together with the intervening atoms to form optionally substituted monocyclic carbocyclyl or optionally substituted monocyclic heterocyclyl; or optionally three R 1 are joined together with the intervening atoms to form optionally substituted bicyclic carbocyclyl or optionally substitute bicyclic heterocyclyl;
  • n 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10;
  • n 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10;
  • A is of the formula: ;
  • each instance of L 2 is independently a bond or optionally substituted C 1-6 alkylene, wherein one methylene unit of the optionally substituted C 1-6 alkylene is optionally replaced with O, N(R N ), S, C(O), C(O)N(R N ), NR N C(O), C(O)O, OC(O), OC(O)O, OC(O)N(R N ), - NR N C(O)O, or NR N C(O)N(R N );
  • each instance of R 2 is independently optionally substituted C 1-30 alkyl, optionally substituted C 1-30 alkenyl, or optionally substituted C 1-30 alkynyl; optionally wherein one or more methylene units of R 2 are independently replaced with optionally substituted carbocyclylene, optionally substituted heterocyclylene, optionally substituted arylene, optionally substituted heteroarylene, N(R N ), O, S, C(O), C(O)N(R N ), NR N C(O),
  • Ring B is optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl;
  • p 1 or 2;
  • R 2 is independently unsubstituted alkyl, unsubstituted alkenyl, or unsubstituted alkynyl.
  • the phospholipids may be one or more of the phospholipids described in U.S. Application No.62/520,530. i) Phospholipid Head Modifications
  • a phospholipid useful or potentially useful in the present invention comprises a modified phospholipid head (e.g., a modified choline group).
  • a phospholipid with a modified head is DSPC, or analog thereof, with a modified quaternary amine.
  • at least one of R 1 is not methyl.
  • at least one of R 1 is not hydrogen or methyl.
  • the com ound of Formula IV is of one of the followin formulae:
  • each t is independently 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10;
  • each u is independently 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10;
  • each v is independently 1, 2, or 3.

Abstract

Des aspects de l'invention concernent des compositions et des méthodes pour le traitement et/ou la prévention d'une maladie par administration d'anticorps à un sujet. Les compositions et les traitements décrits ici comprennent un ou plusieurs polynucléotides d'ARN présentant un cadre de lecture ouvert codant pour un ou plusieurs anticorps ou fragments de ceux-ci. L'invention concerne également des procédés de préparation et d'utilisation de ces traitements.
PCT/US2018/037918 2017-06-15 2018-06-15 Anticorps d'arn WO2018232355A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201762520474P 2017-06-15 2017-06-15
US62/520,474 2017-06-15
US201762593790P 2017-12-01 2017-12-01
US62/593,790 2017-12-01

Publications (1)

Publication Number Publication Date
WO2018232355A1 true WO2018232355A1 (fr) 2018-12-20

Family

ID=64661024

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2018/037918 WO2018232355A1 (fr) 2017-06-15 2018-06-15 Anticorps d'arn

Country Status (1)

Country Link
WO (1) WO2018232355A1 (fr)

Cited By (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10383937B2 (en) 2015-10-22 2019-08-20 Modernatx, Inc. Human cytomegalovirus RNA vaccines
US10449244B2 (en) 2015-07-21 2019-10-22 Modernatx, Inc. Zika RNA vaccines
US10485885B2 (en) 2015-12-10 2019-11-26 Modernatx, Inc. Compositions and methods for delivery of agents
US10493143B2 (en) 2015-10-22 2019-12-03 Modernatx, Inc. Sexually transmitted disease vaccines
US10517940B2 (en) 2015-10-22 2019-12-31 Modernatx, Inc. Zika virus RNA vaccines
US10526629B2 (en) 2017-08-18 2020-01-07 Modernatx, Inc. RNA polymerase variants
US10543269B2 (en) 2015-10-22 2020-01-28 Modernatx, Inc. hMPV RNA vaccines
US10653712B2 (en) 2016-09-14 2020-05-19 Modernatx, Inc. High purity RNA compositions and methods for preparation thereof
US10653767B2 (en) 2017-09-14 2020-05-19 Modernatx, Inc. Zika virus MRNA vaccines
US10695419B2 (en) 2016-10-21 2020-06-30 Modernatx, Inc. Human cytomegalovirus vaccine
US10709779B2 (en) 2014-04-23 2020-07-14 Modernatx, Inc. Nucleic acid vaccines
US10751386B2 (en) 2011-09-12 2020-08-25 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
WO2020190750A1 (fr) * 2019-03-15 2020-09-24 Modernatx, Inc. Vaccins à base d'arn contre le vih
US10925958B2 (en) 2016-11-11 2021-02-23 Modernatx, Inc. Influenza vaccine
WO2021079002A2 (fr) 2019-10-24 2021-04-29 Novago Therapeutics Ag Nouveaux anticorps anti-nogo-a
US11045540B2 (en) 2017-03-15 2021-06-29 Modernatx, Inc. Varicella zoster virus (VZV) vaccine
US11103578B2 (en) 2016-12-08 2021-08-31 Modernatx, Inc. Respiratory virus nucleic acid vaccines
WO2021245184A1 (fr) 2020-06-02 2021-12-09 Neurimmune Ag Anticorps humains contre le coronavirus 2 du syndrome respiratoire aigu sévère (sars-cov-2)
US20220144960A1 (en) * 2018-12-26 2022-05-12 Nanjing Legend Biotech Co., Ltd. Cd30-binding moieties, chimeric antigen receptors, and uses thereof
CN114469996A (zh) * 2021-12-23 2022-05-13 中国医学科学院医学生物学研究所 一种包含miR-135b-5p的外泌体及在抗轮状病毒感染中的应用
US11351242B1 (en) 2019-02-12 2022-06-07 Modernatx, Inc. HMPV/hPIV3 mRNA vaccine composition
US11364292B2 (en) 2015-07-21 2022-06-21 Modernatx, Inc. CHIKV RNA vaccines
US11384352B2 (en) 2016-12-13 2022-07-12 Modernatx, Inc. RNA affinity purification
US11406703B2 (en) 2020-08-25 2022-08-09 Modernatx, Inc. Human cytomegalovirus vaccine
WO2022212191A1 (fr) * 2021-04-01 2022-10-06 Modernatx, Inc. Expression mucosale de structures et isotypes d'anticorps par arnm
US11464848B2 (en) 2017-03-15 2022-10-11 Modernatx, Inc. Respiratory syncytial virus vaccine
US11485960B2 (en) 2019-02-20 2022-11-01 Modernatx, Inc. RNA polymerase variants for co-transcriptional capping
US11497807B2 (en) 2017-03-17 2022-11-15 Modernatx, Inc. Zoonotic disease RNA vaccines
US11524023B2 (en) 2021-02-19 2022-12-13 Modernatx, Inc. Lipid nanoparticle compositions and methods of formulating the same
US11547673B1 (en) 2020-04-22 2023-01-10 BioNTech SE Coronavirus vaccine
US11564893B2 (en) 2015-08-17 2023-01-31 Modernatx, Inc. Methods for preparing particles and related compositions
WO2023015261A1 (fr) * 2021-08-04 2023-02-09 Modernatx, Inc. Arnm codant pour des polypeptides chimériques de reprogrammation métabolique et leurs utilisations
US11576961B2 (en) 2017-03-15 2023-02-14 Modernatx, Inc. Broad spectrum influenza virus vaccine
US11643441B1 (en) 2015-10-22 2023-05-09 Modernatx, Inc. Nucleic acid vaccines for varicella zoster virus (VZV)
US11744801B2 (en) 2017-08-31 2023-09-05 Modernatx, Inc. Methods of making lipid nanoparticles
US11752206B2 (en) 2017-03-15 2023-09-12 Modernatx, Inc. Herpes simplex virus vaccine
US11786607B2 (en) 2017-06-15 2023-10-17 Modernatx, Inc. RNA formulations
WO2023218431A1 (fr) * 2022-05-13 2023-11-16 BioNTech SE Compositions d'arn ciblant le vih
US11851694B1 (en) 2019-02-20 2023-12-26 Modernatx, Inc. High fidelity in vitro transcription
US11866696B2 (en) 2017-08-18 2024-01-09 Modernatx, Inc. Analytical HPLC methods
US11878055B1 (en) 2022-06-26 2024-01-23 BioNTech SE Coronavirus vaccine
US11905525B2 (en) 2017-04-05 2024-02-20 Modernatx, Inc. Reduction of elimination of immune responses to non-intravenous, e.g., subcutaneously administered therapeutic proteins
US11912982B2 (en) 2017-08-18 2024-02-27 Modernatx, Inc. Methods for HPLC analysis
US11911453B2 (en) 2018-01-29 2024-02-27 Modernatx, Inc. RSV RNA vaccines

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014160243A1 (fr) * 2013-03-14 2014-10-02 The Trustees Of The University Of Pennsylvania Purification et évaluation de la pureté de molécules d'arn synthétisées comprenant des nucléosides modifiés
WO2016066708A1 (fr) * 2014-10-29 2016-05-06 Novartis Ag Expression directe d'anticorps
WO2016176330A1 (fr) * 2015-04-27 2016-11-03 The Trustees Of The University Of Pennsylvania Arn à nucléoside modifié destiné à induire une réponse immunitaire adaptative
WO2017070626A2 (fr) * 2015-10-22 2017-04-27 Modernatx, Inc. Vaccins contre les virus respiratoires
WO2017186928A1 (fr) * 2016-04-29 2017-11-02 Curevac Ag Arn codant pour un anticorps

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014160243A1 (fr) * 2013-03-14 2014-10-02 The Trustees Of The University Of Pennsylvania Purification et évaluation de la pureté de molécules d'arn synthétisées comprenant des nucléosides modifiés
WO2016066708A1 (fr) * 2014-10-29 2016-05-06 Novartis Ag Expression directe d'anticorps
WO2016176330A1 (fr) * 2015-04-27 2016-11-03 The Trustees Of The University Of Pennsylvania Arn à nucléoside modifié destiné à induire une réponse immunitaire adaptative
WO2017070626A2 (fr) * 2015-10-22 2017-04-27 Modernatx, Inc. Vaccins contre les virus respiratoires
WO2017186928A1 (fr) * 2016-04-29 2017-11-02 Curevac Ag Arn codant pour un anticorps

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
HOLLEVOET K. ET AL.: "State of play and clinical prospects of antibody gene transfer", JOURNAL OF TRANSLATIONAL MEDICINE, vol. 15, no. 1, 7 June 2017 (2017-06-07), pages 131, XP055557656 *
PARDI N. ET AL.: "Administration of nucleoside-modified mRNA encoding broadly neutralizing antibody protects humanized mice from HIV-1 challenge", NATURE COMMUNICATIONS, vol. 8, 2 March 2017 (2017-03-02), pages 14630, XP055557652, Retrieved from the Internet <URL:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5337964/pdf/ncomms14630.pdf> [retrieved on 20181101] *
THRAN, M. ET AL.: "mRNA mediates passive vaccination against infectious agents, toxins, and tumors", EMBO MOLECULAR MEDICINE, vol. 9, no. 10, 9 August 2017 (2017-08-09), pages 1434 - 1447, XP055505303 *

Cited By (70)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10751386B2 (en) 2011-09-12 2020-08-25 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US10709779B2 (en) 2014-04-23 2020-07-14 Modernatx, Inc. Nucleic acid vaccines
US11364292B2 (en) 2015-07-21 2022-06-21 Modernatx, Inc. CHIKV RNA vaccines
US10449244B2 (en) 2015-07-21 2019-10-22 Modernatx, Inc. Zika RNA vaccines
US11007260B2 (en) 2015-07-21 2021-05-18 Modernatx, Inc. Infectious disease vaccines
US10702597B2 (en) 2015-07-21 2020-07-07 Modernatx, Inc. CHIKV RNA vaccines
US11564893B2 (en) 2015-08-17 2023-01-31 Modernatx, Inc. Methods for preparing particles and related compositions
US11872278B2 (en) 2015-10-22 2024-01-16 Modernatx, Inc. Combination HMPV/RSV RNA vaccines
US10702600B1 (en) 2015-10-22 2020-07-07 Modernatx, Inc. Betacoronavirus mRNA vaccine
US10383937B2 (en) 2015-10-22 2019-08-20 Modernatx, Inc. Human cytomegalovirus RNA vaccines
US10675342B2 (en) 2015-10-22 2020-06-09 Modernatx, Inc. Chikungunya virus RNA vaccines
US11484590B2 (en) 2015-10-22 2022-11-01 Modernatx, Inc. Human cytomegalovirus RNA vaccines
US10702599B2 (en) 2015-10-22 2020-07-07 Modernatx, Inc. HPIV3 RNA vaccines
US10543269B2 (en) 2015-10-22 2020-01-28 Modernatx, Inc. hMPV RNA vaccines
US11235052B2 (en) 2015-10-22 2022-02-01 Modernatx, Inc. Chikungunya virus RNA vaccines
US10517940B2 (en) 2015-10-22 2019-12-31 Modernatx, Inc. Zika virus RNA vaccines
US10716846B2 (en) 2015-10-22 2020-07-21 Modernatx, Inc. Human cytomegalovirus RNA vaccines
US10493143B2 (en) 2015-10-22 2019-12-03 Modernatx, Inc. Sexually transmitted disease vaccines
US11643441B1 (en) 2015-10-22 2023-05-09 Modernatx, Inc. Nucleic acid vaccines for varicella zoster virus (VZV)
US11278611B2 (en) 2015-10-22 2022-03-22 Modernatx, Inc. Zika virus RNA vaccines
US10933127B2 (en) 2015-10-22 2021-03-02 Modernatx, Inc. Betacoronavirus mRNA vaccine
US10556018B2 (en) 2015-12-10 2020-02-11 Modernatx, Inc. Compositions and methods for delivery of agents
US11285222B2 (en) 2015-12-10 2022-03-29 Modernatx, Inc. Compositions and methods for delivery of agents
US10485885B2 (en) 2015-12-10 2019-11-26 Modernatx, Inc. Compositions and methods for delivery of agents
US10653712B2 (en) 2016-09-14 2020-05-19 Modernatx, Inc. High purity RNA compositions and methods for preparation thereof
US11202793B2 (en) 2016-09-14 2021-12-21 Modernatx, Inc. High purity RNA compositions and methods for preparation thereof
US11541113B2 (en) 2016-10-21 2023-01-03 Modernatx, Inc. Human cytomegalovirus vaccine
US10695419B2 (en) 2016-10-21 2020-06-30 Modernatx, Inc. Human cytomegalovirus vaccine
US11197927B2 (en) 2016-10-21 2021-12-14 Modernatx, Inc. Human cytomegalovirus vaccine
US11696946B2 (en) 2016-11-11 2023-07-11 Modernatx, Inc. Influenza vaccine
US10925958B2 (en) 2016-11-11 2021-02-23 Modernatx, Inc. Influenza vaccine
US11103578B2 (en) 2016-12-08 2021-08-31 Modernatx, Inc. Respiratory virus nucleic acid vaccines
US11384352B2 (en) 2016-12-13 2022-07-12 Modernatx, Inc. RNA affinity purification
US11045540B2 (en) 2017-03-15 2021-06-29 Modernatx, Inc. Varicella zoster virus (VZV) vaccine
US11918644B2 (en) 2017-03-15 2024-03-05 Modernatx, Inc. Varicella zoster virus (VZV) vaccine
US11752206B2 (en) 2017-03-15 2023-09-12 Modernatx, Inc. Herpes simplex virus vaccine
US11576961B2 (en) 2017-03-15 2023-02-14 Modernatx, Inc. Broad spectrum influenza virus vaccine
US11464848B2 (en) 2017-03-15 2022-10-11 Modernatx, Inc. Respiratory syncytial virus vaccine
US11497807B2 (en) 2017-03-17 2022-11-15 Modernatx, Inc. Zoonotic disease RNA vaccines
US11905525B2 (en) 2017-04-05 2024-02-20 Modernatx, Inc. Reduction of elimination of immune responses to non-intravenous, e.g., subcutaneously administered therapeutic proteins
US11786607B2 (en) 2017-06-15 2023-10-17 Modernatx, Inc. RNA formulations
US11912982B2 (en) 2017-08-18 2024-02-27 Modernatx, Inc. Methods for HPLC analysis
US10526629B2 (en) 2017-08-18 2020-01-07 Modernatx, Inc. RNA polymerase variants
US11066686B2 (en) 2017-08-18 2021-07-20 Modernatx, Inc. RNA polymerase variants
US11866696B2 (en) 2017-08-18 2024-01-09 Modernatx, Inc. Analytical HPLC methods
US11767548B2 (en) 2017-08-18 2023-09-26 Modernatx, Inc. RNA polymerase variants
US11744801B2 (en) 2017-08-31 2023-09-05 Modernatx, Inc. Methods of making lipid nanoparticles
US11207398B2 (en) 2017-09-14 2021-12-28 Modernatx, Inc. Zika virus mRNA vaccines
US10653767B2 (en) 2017-09-14 2020-05-19 Modernatx, Inc. Zika virus MRNA vaccines
US11911453B2 (en) 2018-01-29 2024-02-27 Modernatx, Inc. RSV RNA vaccines
US20220144960A1 (en) * 2018-12-26 2022-05-12 Nanjing Legend Biotech Co., Ltd. Cd30-binding moieties, chimeric antigen receptors, and uses thereof
US11351242B1 (en) 2019-02-12 2022-06-07 Modernatx, Inc. HMPV/hPIV3 mRNA vaccine composition
US11851694B1 (en) 2019-02-20 2023-12-26 Modernatx, Inc. High fidelity in vitro transcription
US11485960B2 (en) 2019-02-20 2022-11-01 Modernatx, Inc. RNA polymerase variants for co-transcriptional capping
WO2020190750A1 (fr) * 2019-03-15 2020-09-24 Modernatx, Inc. Vaccins à base d'arn contre le vih
WO2021079002A2 (fr) 2019-10-24 2021-04-29 Novago Therapeutics Ag Nouveaux anticorps anti-nogo-a
US11951185B2 (en) 2020-04-22 2024-04-09 BioNTech SE RNA constructs and uses thereof
US11925694B2 (en) 2020-04-22 2024-03-12 BioNTech SE Coronavirus vaccine
US11779659B2 (en) 2020-04-22 2023-10-10 BioNTech SE RNA constructs and uses thereof
US11547673B1 (en) 2020-04-22 2023-01-10 BioNTech SE Coronavirus vaccine
WO2021245184A1 (fr) 2020-06-02 2021-12-09 Neurimmune Ag Anticorps humains contre le coronavirus 2 du syndrome respiratoire aigu sévère (sars-cov-2)
US11406703B2 (en) 2020-08-25 2022-08-09 Modernatx, Inc. Human cytomegalovirus vaccine
US11524023B2 (en) 2021-02-19 2022-12-13 Modernatx, Inc. Lipid nanoparticle compositions and methods of formulating the same
US11622972B2 (en) 2021-02-19 2023-04-11 Modernatx, Inc. Lipid nanoparticle compositions and methods of formulating the same
WO2022212191A1 (fr) * 2021-04-01 2022-10-06 Modernatx, Inc. Expression mucosale de structures et isotypes d'anticorps par arnm
WO2023015261A1 (fr) * 2021-08-04 2023-02-09 Modernatx, Inc. Arnm codant pour des polypeptides chimériques de reprogrammation métabolique et leurs utilisations
CN114469996A (zh) * 2021-12-23 2022-05-13 中国医学科学院医学生物学研究所 一种包含miR-135b-5p的外泌体及在抗轮状病毒感染中的应用
CN114469996B (zh) * 2021-12-23 2023-10-20 中国医学科学院医学生物学研究所 一种包含miR-135b-5p的外泌体及在抗轮状病毒感染中的应用
WO2023218431A1 (fr) * 2022-05-13 2023-11-16 BioNTech SE Compositions d'arn ciblant le vih
US11878055B1 (en) 2022-06-26 2024-01-23 BioNTech SE Coronavirus vaccine

Similar Documents

Publication Publication Date Title
WO2018232355A1 (fr) Anticorps d&#39;arn
US20210128721A1 (en) Immunomodulatory therapeutic mrna compositions encoding activating oncogene mutation peptides
RU2768829C2 (ru) Противораковые рнк-вакцины
US20240042015A1 (en) Circular rna compositions and methods
JP7395483B2 (ja) mRNAの細胞内送達のためのペプチドおよびナノ粒子
CN113271926A (zh) 脂质纳米颗粒的制备及其施用方法
WO2022212191A1 (fr) Expression mucosale de structures et isotypes d&#39;anticorps par arnm
JP2020520640A (ja) 連結したインターロイキン−12(il12)ポリペプチドをコードするポリヌクレオチド及びその使用
CN116113419A (zh) 环状rna组合物和方法
CN116194151A (zh) 包含具有延长的半衰期的mRNA治疗剂的LNP组合物
EP4228601A1 (fr) Ciblage in vivo de la fibrose par le ciblage de l&#39;anti-cd5 dans l&#39;arnm-lnp fap-car-t
US11802146B2 (en) Polynucleotides encoding anti-chikungunya virus antibodies
EP4228602A1 (fr) Ciblage in vivo de lymphocytes t pour la thérapeutique fondée sur l&#39;arnm
CN116546991A (zh) mRNA治疗剂对CD4+-T细胞的体内靶向
EP4153244A1 (fr) Méthodes et compositions d&#39;arn circulaire
EP3576780A1 (fr) Compositions thérapeutiques immunomodulatrices d&#39;arnm codant pour des peptides de mutation d&#39;activation d&#39;oncogènes

Legal Events

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

Ref document number: 18817650

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18817650

Country of ref document: EP

Kind code of ref document: A1