WO2022152869A1 - Use of oligonucleotides for individuals with hepatic impairment - Google Patents

Use of oligonucleotides for individuals with hepatic impairment Download PDF

Info

Publication number
WO2022152869A1
WO2022152869A1 PCT/EP2022/050776 EP2022050776W WO2022152869A1 WO 2022152869 A1 WO2022152869 A1 WO 2022152869A1 EP 2022050776 W EP2022050776 W EP 2022050776W WO 2022152869 A1 WO2022152869 A1 WO 2022152869A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
rnai
rnai agent
administered
subject
Prior art date
Application number
PCT/EP2022/050776
Other languages
French (fr)
Inventor
Thomas Naoki Kakuda
Original Assignee
Janssen Sciences Ireland Unlimited Company
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 Janssen Sciences Ireland Unlimited Company filed Critical Janssen Sciences Ireland Unlimited Company
Publication of WO2022152869A1 publication Critical patent/WO2022152869A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1131Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against viruses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/315Phosphorothioates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3212'-O-R Modification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3222'-R Modification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/33Chemical structure of the base
    • C12N2310/332Abasic residue
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/34Spatial arrangement of the modifications
    • C12N2310/345Spatial arrangement of the modifications having at least two different backbone modifications
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/34Spatial arrangement of the modifications
    • C12N2310/346Spatial arrangement of the modifications having a combination of backbone and sugar modifications
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/351Conjugate
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • C12N2320/31Combination therapy

Definitions

  • HCC cirrhosis and hepatocellular carcinoma
  • the subject has a Child-Pugh score of 7 to 9 (Class B), indicating significant functional hepatic compromise. In some embodiments, the subject has a Child-Pugh score of 10 to 15 (Class C), indicating decompensated disease. In some embodiments where the subject has a Child score of 7-9 or 10-15, the subject also suffers from renal impairment. In some embodiments, the renally impaired subject has a globular filtration rate [GFR] of ⁇ 90 mL/min. In other embodiments, the renally impaired subject is on dialysis.
  • GFR globular filtration rate
  • RNAi agent comprising: an antisense strand comprising a nucleotide sequence of SEQ ID NO:8 and a complementary sense strand (e.g., a sense strand comprising a nucleotide sequence of SEQ ID NO: 16); or
  • a first RNAi agent comprising: an antisense strand comprising a nucleotide sequence that is at least 70% homologous to SEQ ID NO:2 and a complementary sense strand comprising a nucleotide sequence that is at least 80% homologous to SEQ ID NO:11; and
  • the disclosure provides methods of treating HBV in a subject suffering from cirrhosis of the liver, said method comprising subcutaneously administering to the subject a pharmaceutical composition comprising about 200 mg of an RNAi component comprising
  • RNAi agent comprising: an antisense strand comprising a nucleotide sequence of SEQ ID NO:8 and a complementary sense strand (e.g., a sense strand nucleotide sequence of SEQ ID NO:16); or
  • a first RNAi agent comprising: an antisense strand comprising a nucleotide sequence that is at least 70% homologous to SEQ ID NO:2 and a complementary sense strand comprising a nucleotide sequence that is at least 70% homologous to SEQ ID NO:11;
  • the disclosure provides methods of treating HBV in a hepatically impaired subject (e.g., a subject suffering from liver cirrhosis), said method comprising subcutaneously administering to the subject a pharmaceutical composition comprising an RNAi component comprising (i) a first RNAi agent comprising: an antisense strand comprising a nucleotide sequence of SEQ ID NO:2 and a complementary sense strand (e.g., a sense strand comprising a nucleotide sequence of SEQ ID NO: 11); and
  • a second RNAi agent comprising: an antisense strand comprising a nucleotide sequence that is at least 70% homologous to a sequence of SEQ ID NO: 8 and a complementary sense strand comprising a nucleotide sequence that is at least 70% homologous to SEQ ID NO: 16, wherein the molar ratio of the first RNAi agent to the second RNAi agent is about 2:1, and wherein the AUG / of the first component is from about 20,000 ng.h/mL to about 50,000 ng.h/mL and the AUG , of the second component is from about 3,000 ng.h/mL to about 12,000 ng.h/mL.
  • the combination or kit further comprises another agent for treating infection caused by HBV.
  • RNAi component and optionally another agent for treating infection caused by HBV such as a nucleoside analog or a NAP, in the manufacture of a medicament for treating a HBV infection in a subject, enhancing an immune response in a subject with a HBV infection, decreasing viral replication in a subject with a HBV infection, decreasing the expression of one or more HBV polypeptide(s), more particularly of one or more polypeptide(s) from HBsAg and HBeAg, and/or increasing the targeted killing of hepatocytes comprising integrated viral DNA or extrachromosomal DNA in a subject with a HBV infection.
  • another agent for treating infection caused by HBV such as a nucleoside analog or a NAP
  • the RNAi component is administered subcutaneously or intravenously, preferably subcutaneously, at an amount of about 40-1000 mg per dose, more particularly about 40-250 mg per dose, such as about 100-200 mg per dose, more particularly about 200 mg per dose, and it is administered weekly, every two weeks, every 4 weeks, monthly, every 2 months, or every 3 months, preferably every 4 weeks or monthly.
  • a subject has achieved at least one of the following features a)-e), more particularly more than one of the following features a)-e), more particularly at least features a), b) and c), more particularly all of features a)-d), during or after the treatment with a combination according to an embodiment of the application: a) decreased HBV replication as measured by serum HBV DNA level, preferably undetectable serum HBV DNA level; b) decreased expression of one or more HBV polypeptide(s), preferably decreased expression of HBsAg as measured by serum HBsAg level, preferably undetectable serum HBsAg level; c) enhanced HBV-specific T cell responses; d) loss of HBeAg or serocoversion for HBeAg, if the subject is HBeAg positive before the treatment; and e) seroconversion for HBsAg.
  • an embodiment of the application is for use in treating a subject coinfected with CHB and another chronic infection with at least one of: hepatitis D virus (HDV); hepatitis C virus (HCV); or human immunodeficiency virus (HIV).
  • the combination can be used in a method of decreasing the serum levels of HDV RNA in a subject chronically co-infected with both HBV and HDV; a method of normalizing alanine aminotransferase (ALT) level in a subject chronically coinfected with HBV and HDV; or a method of eradicating HDV infection in a subject chronically coinfected with HBV and HDV.
  • the first and the second RNAi agents independently comprise a duplex selected from the group consisting of: an antisense strand comprising SEQ ID NO: 1 and a sense strand comprising SEQ ID NO: 10; an antisense strand comprising SEQ ID NO:2 and a sense strand comprising SEQ ID NO: 11; an antisense strand comprising SEQ ID NO: 3 and a sense strand comprising SEQ ID NO: 11; an antisense strand comprising SEQ ID NO: 4 and a sense strand comprising SEQ ID NO: 12; an antisense strand comprising SEQ ID NO: 8 and a sense strand comprising SEQ ID NO: 16; an antisense strand comprising SEQ ID NO: 8 and a sense strand comprising SEQ ID NO: 17; an antisense strand comprising SEQ ID NO:2 and a sense strand comprising SEQ ID NO: 13; and an antisense strand comprising SEQ ID NO: 8 and a sense strand comprising SEQ ID NO:
  • the molar ratio of the first RNAi agent to the second RNAi agent is in the range of about 1:2 to about 5:1. In another variation, the molar ratio of the first RNAi agent to the second RNAi agent is about 2:1.
  • the first and the second RNAi agents are each independently conjugated to (NAG37)s, the first RNAi agent comprises an antisense strand comprising SEQ ID NO: 2 and a sense strand comprising SEQ ID NO: 11, and the second RNAi agent comprises an antisense strand comprising SEQ ID NO: 8 and a sense strand comprising SEQ ID NO: 16.
  • FIG. 1 illustrates the mean linear (standard deviation) plasma concentration-time profile of an RNAi component of the disclosure after subcutaneous (SC) injection of the RNAi component in participants with liver cirrhosis with moderate hepatic impairment and in participants with normal liver function with no liver cirrhosis.
  • the Y-axis shows mean plasma concentration (ng/mL) of the first RNAi agent.
  • treatment naive refers to a patient, individual or subject not having previously received treatment with a drug, investigational or approved, for HBV infection, in particular a nucleoside or nucleotide analog drug or interferon product.
  • an individual “in need thereof’ may be an individual who has been diagnosed with or previously treated for the condition to be treated.
  • the method further contemplates a step of identifying an individual or subject in need of the particular treatment to be administered or having the particular condition to be treated.
  • the term “pharmaceutically acceptable” refers to a material, such as a carrier or diluent, which does not abrogate the biological activity or properties of the compound, and is relatively non-toxic, i.e., the material can be administered to an individual without causing undesirable biological effects or interacting in a deleterious manner with any of the components of the composition in which it is contained.
  • composition refers to a mixture of at least one compound provided herein with a pharmaceutically acceptable carrier.
  • the pharmaceutical composition facilitates administration of the compound to a patient or subject. Multiple techniques of administering a compound exist in the art including, but not limited to, intravenous, oral, aerosol, parenteral, ophthalmic, pulmonary and topical administration.
  • the term “pharmaceutically acceptable carrier” means a pharmaceutically acceptable material, composition or carrier, such as a liquid or solid filler, stabilizer, dispersing agent, suspending agent, diluent, excipient, thickening agent, solvent or encapsulating material, involved in carrying or transporting a compound provided herein within or to the patient such that it can perform its intended function.
  • a pharmaceutically acceptable material, composition or carrier such as a liquid or solid filler, stabilizer, dispersing agent, suspending agent, diluent, excipient, thickening agent, solvent or encapsulating material, involved in carrying or transporting a compound provided herein within or to the patient such that it can perform its intended function.
  • Such constructs are carried or transported from one organ, or portion of the body, to another organ, or portion of the body.
  • Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation, including the compound provided herein, and not injurious to the patient.
  • materials that can serve as pharmaceutically acceptable carriers include: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol; polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar; buffering agents, such as magnesium hydroxide and aluminum hydroxide; surface active agents; alginic acid; pyrogen-free water; isotonic saline
  • amino refers to a functional group having the formulae -NHz, - NH(alkyl), and -N(alkyl)2, wherein alkyl is as defined herein.
  • Hepatic impairment is a condition wherein normal functioning of the liver reduced. Hepatic impairment can be acute, with rapid onset, or chronic. Chronic hepatic impairment, or cirrhosis, can occur from many causes, such as excessive consumption of alcohol, hepatitis, autoimmune disease, heredity, or metabolism, or can be idiopathic. Liver damage is generally irreversible, and treatment consists of prevention of progression and treatment of symptoms. In severe cases, liver transplant is the only option.
  • the RNAi component comprises a combination or cocktail of a first and a second RNAi agent having different nucleotide sequences.
  • the first and the second RNAi agents are each separately and independently linked to targeting groups.
  • the first and the second RNAi agents are each linked to targeting groups comprised of N-acetyl-galactosamines.
  • each of the RNAi agents is linked to the same targeting group.
  • each of the RNAi agents is linked to different targeting groups, such as targeting groups having different chemical structures.
  • the four phosphorothioate internucleoside linkages are between the nucleotides at positions 1-3 from the 5' end of the sense strand and between the nucleotides at positions 19-21, 20-22, 21-23, 22-24, 23-25, or 24-26 from the 5' end.
  • the first or the second RNAi agent contains at least two phosphorothioate internucleoside linkages in the sense strand and three or four phosphorothioate internucleoside linkages in the antisense strand.
  • G guanosine-3 '-phosphate
  • NfANA 2'-F-Arabino nucleotide
  • TMs 2'-methoxyethylthymidine-3'-phosphorothioate
  • the sense strands in Tables 4 and 5 include a targeting group (NAG25, NAG25s, NAG 37, or NAG37s) at the 5' end.
  • a targeting group NAG25, NAG25s, NAG 37, or NAG37s
  • the disclosure also includes sense strands that have sequences displayed in Tables 4 and 5 but without the targeting group on the 5' end or with targeting groups other than NAG25, NAG25s, NAG37, or NAG37s, as disclosed herein.
  • both the antisense and/or sense strands displayed in Tables 3 and 4 can be modified either at the 5' end or 3' end with a targeting group, as disclosed herein.
  • the first RNAi agent comprises SEQ ID NO: 5 and SEQ ID NO: 14. In some embodiments, the first RNAi agent comprises SEQ ID NO: 6 and SEQ ID NO: 14. In some embodiments, the first RNAi agent comprises SEQ ID NO: 7 and SEQ ID NO: 15. In some embodiments, the first RNAi agent comprises SEQ ID NO: 1 and SEQ ID NO: 10, 11 or 13. In some embodiments, the first RNAi agent comprises SEQ ID NO: 2 and SEQ ID NO: 10, 11 or 13. In some embodiments, the first RNAi agent comprises SEQ ID NO: 3 and SEQ ID NO: 10, 11, or 13. In some embodiments, the first RNAi agent comprises SEQ ID NO: 4 and SEQ ID NO: 12. In some embodiments, the second RNAi agent comprises SEQ ID NO: 9 and SEQ ID NO: 19. In some embodiments, the second RNAi agent comprises SEQ ID NO: 8 and SEQ ID NO: 16, 17 or 18.
  • the RNAi component comprises a first RNAi agent comprising SEQ ID NO: 5 and SEQ ID NO: 14 and a second RNAi agent comprising SEQ ID NO: 9 and SEQ ID NO: 19. In some embodiments, the RNAi component comprises a first RNAi agent comprising SEQ ID NO: 6 and SEQ ID NO: 14 and a second RNAi agent comprising SEQ ID NO: 9 and SEQ ID NO: 19. In some embodiments, the RNAi component comprises a first RNAi agent comprising SEQ ID NO: 7 and SEQ ID NO: 15 and a second RNAi agent comprising SEQ ID NO: 9 and SEQ ID NO: 19.
  • the RNAi component comprises a first RNAi agent comprising SEQ ID NO: 1 and SEQ ID NO: 10, 11 or 13 and a second RNAi agent comprising SEQ ID NO: 8 and SEQ ID NO: 16, 17 or 18.
  • the RNAi component comprises a first RNAi agent comprising SEQ ID NO: 2 and SEQ ID NO: 10, 11 or 13 and a second RNAi agent comprising SEQ ID NO: 8 and SEQ ID NO: 16, 17 or 18.
  • the RNAi component comprises a first RNAi agent comprising SEQ ID NO: 3 and SEQ ID NO: 10, 11 or 13 and a second RNAi agent comprising SEQ ID NO: 8 and SEQ ID NO: 16, 17 or 18.
  • the RNAi component comprises a first RNAi agent comprising SEQ ID NO: 4 and SEQ ID NO: 12 and a second RNAi agent comprising SEQ ID NO: 8 and SEQ ID NO: 16, 17 or 18.
  • the RNAi component comprises a first and a second RNAi agent in a molar ratio of about 1:1, 2:1, 3:1, 4:1 or 5:1.
  • the two HBV RNAi agents are administered in a molar ratio of about 2:1.
  • an HBV RNAi agent comprises an antisense strand and a sense strand having the modified nucleotide sequences of any of the antisense strand and/or sense strand nucleotide sequences of any of the duplexes of Table 6.
  • an HBV RNAi agent comprises an antisense strand and a sense strand having the modified nucleotide sequences of any of the antisense strand and/or sense strand nucleotide sequences of any of the duplexes of Table 6, and further comprises an asialoglycoprotein receptor ligand targeting group.
  • an HBV RNAi agent comprises any of the duplexes of Table 6.
  • an HBV RNAi agent consists of any of the duplexes of Table 6.
  • RNAi agents comprising the RNAi componented by the following structure, shown as the sodium salt:
  • one of the RNAi agents comprising the RNAi component is represented by the following structure, shown as the sodium salt:
  • Injectable solutions may be prepared in which the carrier comprises saline solution, glucose solution or a mixture of saline and glucose solution.
  • Injectable suspensions may also be prepared in which case appropriate liquid carriers, suspending agents and the like may be employed. Also included are solid form preparations intended to be converted, shortly before use, to liquid form preparations.
  • the carrier optionally comprises a penetration enhancing agent and/or a suitable wetting agent, optionally combined with suitable additives of any nature in minor proportions, which additives do not introduce a significant deleterious effect on the skin.
  • the compounds of the present invention may also be administered via oral inhalation or insufflation in the form of a solution, a suspension or a dry powder using any art-known delivery system.
  • Unit dosage form refers to physically discrete units suitable as unitary dosages, each unit containing a predetermined quantity of active ingredient calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • unit dosage forms are tablets (including scored or coated tablets), capsules, pills, suppositories, powder packets, wafers, injectable solutions or suspensions and the like, and segregated multiples thereof.
  • the first and second RNAi agents of an RNAi component that targets or reduces HBsAg can be formulated in the same or separate pharmaceutical compositions.
  • the HBV RNAi agents in the same or separate compositions can be formulated with the same or different excipients and carriers.
  • the HBV RNAi agents in the same or separate compositions can be administered through same or different administration routes.
  • any suitable pharmaceutical composition comprising the first and/or second RNAi agents of the RNAi component and a pharmaceutically acceptable carrier can be used in the present invention in view of the present disclosure.
  • the pharmaceutical composition can comprise any RNAi component described herein or otherwise known in the art.
  • One or more pharmaceutically acceptable excipients can be mixed with the first and/or second RNAi agents of the RNAi component, thereby forming a pharmaceutical formulation suitable for in vivo delivery to a human.
  • compositions including an HBV RNAi agent described herein can be delivered to a cell, group of cells, tumor, tissue, or subject using oligonucleotide delivery technologies known in the art.
  • any suitable method recognized in the art for delivering a nucleic acid molecule in vitro or in vivo
  • a pharmaceutical composition comprising at least one of the first and second RNAi agents of an RNAi component described herein, can be delivered by systemic administration via a parenteral route, including subcutaneous, intravenous, intraperitoneal, and intramuscular administration.
  • the compositions are administered by subcutaneous or intravenous infusion or injection.
  • a pharmaceutical composition or medicament includes a pharmacologically effective amount of at least one of the described therapeutic compounds and a pharmaceutically acceptable carrier.
  • the pharmaceutically acceptable carrier can comprise one or more pharmaceutically acceptable excipients.
  • Pharmaceutically acceptable excipients also referred to herein as “excipients”, are substances other than the active pharmaceutical ingredient that are intentionally included in the drug delivery system. Excipients do not exert or are not intended to exert a therapeutic effect at the intended dosage.
  • compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion.
  • suitable carriers include physiological saline, bacteriostatic water, Cremophor ELTM (BASF, Parsippany, NJ) or phosphate buffered saline. It should be stable under the conditions of manufacture and storage and should be preserved against the contaminating action of microorganisms such as bacteria and fungi.
  • Sterile injectable solutions can be prepared by incorporating the active compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filter sterilization.
  • dispersions are prepared by incorporating the active compound into a sterile vehicle which contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • methods of preparation include vacuum drying and freeze-drying which yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • the kit further comprises a package insert including, without limitation, appropriate instructions for preparation and administration of the formulation, side effects of the formulation, and any other relevant information.
  • the instructions can be in any suitable format, including, but not limited to, printed matter, videotape, computer readable disk, optical disc or directions to internet-based instructions.
  • an effective amount of an RNAi component in the range of about 25-600 mg per dose is administered to the subject.
  • the effective amount of the RNAi component is in the range of about 25-50 mg, about 50-75 mg, about 75-100 mg, about 100-150 mg, about 150-200 mg, about 200-250 mg, about 250-300 mg, about 300-400 mg, about 400-500 mg or about 500-600 mg per dose.
  • the effective amount of the RNAi component can be administered once per month (Q1M), per 4 weeks (Q4W), bimonthly, or any time period in between.
  • the dose of an RNAi component or agent refers to the dose of the RNAi component or agent itself, and not to the dose of the composition that can contain the RNAi component or agent.
  • the dose of the RNAi component or agent refers to the amount of the RNAi component or agent of the conjugate.
  • the dose of an RNAi component refers to the combined amount of the first and second RNAi agents of the RNAi component.
  • the first and second HBV RNAi agents of an RNAi component are administered in a molar ratio of about 1:1, 2:1, 3:1, 4:1 or 5:1. In some embodiments, the first and second HBV RNAi agents of an RNAi component are administered to a subject in a molar ratio of about 2:1.
  • the two HBV RNAi agents are administered in a combined amount of about 35 mg per dose administration and in the molar ratio of about 2: 1. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 40 mg per dose administration and in the molar ratio of about 2:1. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 50 mg per dose administration and in the molar ratio of about 2:1. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 75 mg per dose administration and in the molar ratio of about 2:1. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 200 mg per dose administration and in the molar ratio of about 2:1.
  • the second RNAi agent is administered in an amount of about 20-125 mg per dose administration. In some embodiments, the second RNAi agent is administered in an amount of about 25- 50 mg per dose administration. In some embodiments, the second RNAi agent is administered in an amount of about 5-40 mg per dose administration. In some embodiments, the first RNAi agent is administered in an amount of about 17 mg per dose administration, and the second RNAi agent is administered in an amount of about 8 mg per dose administration. In some embodiments, the first RNAi agent is administered in an amount of about 23 mg per dose administration, and the second RNAi agent is administered in an amount of about 12 mg per dose administration.
  • the first RNAi agent is administered in an amount of about 27 mg per dose administration, and the second RNAi agent is administered in an amount of about 13 mg per dose administration. In some embodiments, the first RNAi agent is administered in an amount of about 33 mg per dose administration, and the second RNAi agent is administered in an amount of about 17 mg per dose administration. In some embodiments, the first RNAi agent is administered in an amount of about 67 mg per dose administration, and the second RNAi agent is administered in an amount of about 33 mg per dose administration.
  • two RNAi agents are administered at a combined dose of 25-400 mg per dose, and the first RNAi agent is administered with the second RNAi agent at a molar ratio of 2:1.
  • the dose of the first RNAi agent is in an amount of about 16 mg, and the dose of the second RNAi agent is in an amount of about 8 mg for a combined dose of about 25 mg.
  • the dose of the first RNAi agent is in an amount of about 24 mg, and the dose of the second RNAi agent is in an amount of about 12 mg for a combined dose of about 35 mg.
  • two RNAi agents are administered at a combined dose of 25-400 mg per dose, the first RNAi agent is administered with the second RNAi agent at a molar ratio of 3:1.
  • the dose of the first RNAi agent is in an amount of about 18 mg, and the dose of the second RNAi agent is in an amount of about 6 mg for a combined dose of about 25 mg.
  • the dose of the first RNAi agent is in an amount of about 27 mg, and the dose of the second RNAi agent is in an amount of about 9 mg for a combined dose of about 35 mg.
  • the first RNAi agent and the second RNAi agent are administered in a combined amount of about 25-400 mg per dose administration. In some embodiments, the first RNAi agent and the second RNAi agent are administered in a combined amount of about 25-50 mg, 50-75 mg, 75-100 mg, 100-125 mg, 125-150 mg, 150-175 mg, 175-200 mg, 200-225 mg, 225-250 mg, 250-275 mg, 275-300 mg, 300-325 mg, 325-350 mg, 350-375 mg, 375-400 mg, 25-75 mg, 50-100 mg, 100-150 mg, 150-200 mg, 200-250 mg, 250-300 mg, 300-350 mg, 350-400 mg, 25-100 mg, 50-150 mg, 100-200 mg, 150-250 mg, 200-300 mg, 300-400 mg, 25-200 mg, or 200-400 mg per dose administration.
  • a combination of the application further comprises one or more other HBV antiviral agents, such as, an HBV polymerase inhibitor (e.g., entecavir and tenofovir); Immunomodulators; Toll-like receptor 7 modulators; Toll-like receptor 8 modulators; Toll-like receptor 3 modulators; Hyaluronidase inhibitors; Modulators of IL-10; HBsAg inhibitors; Toll like receptor 9 modulators; Cyclophilin inhibitors; HBV Prophylactic vaccines; HBV Therapeutic vaccines; HBV viral entry inhibitors; antisense oligonucleotides targeting viral mRNA, more particularly anti-HB V antisense oligonucleotides; short interfering RNAs (siRNA), more particularly anti-HBV siRNA; endonuclease modulators; inhibitors of ribonucleotide reductase; HBV E antigen inhibitors; HBV antibodies targeting the surface antigens of the hepatitis B virus;
  • the mean (SD) unbound fraction was 0.110 (0.039) for the first RNAi agent and 0.126 (0.036) for the second RNAi agent.
  • the mean (standard deviation) unbound fraction in plasma was 0.091 (0.037) for the first RNAi agent and 0.132 for the second RNAi agent.
  • TEAE treatment-emergent AE
  • siRNA Compound a single 200 mg dose of siRNA Compound was safe and well tolerated in participants without and with moderate hepatic impairment. Higher exposures of the siRNA agents were seen in participants with moderate hepatic impairment, but this increase was not considered to be a clinically relevant safety concern. The fraction of siRNA unbound in plasma was not significantly different between healthy participants and Child-Pugh Class B participants.
  • Child-Pugh Score a score based on five clinical measures of hepatic impairment, including levels of total bilirubin, serum albumin, PT INR, ascites, and hepatic encephalopathy. Each measure is given a ranking of 1, 2, or 3, and the sum of the five rankings is the Child-Pugh Score.
  • the Child-Pugh Score can be used to classify hepatic impairment by placing subjects in a Child-Pugh Group

Abstract

The present disclosure relates to methods for treatment of hepatitis B in a subject comprising an RNAi component, wherein the subject has a level of hepatic impairment.

Description

USE OF OLIGONUCLEOTIDES FOR INDIVIDUALS WITH HEPATIC IMPAIRMENT
FIELD OF THE INVENTION
The present disclosure relates to methods for treatment of hepatitis B in a subject, wherein the subject has a level of hepatic impairment.
SUBMISSION OF SEQUENCE LISTING ON ASCII TEXT FILE
The content of the following submission on ASCII text file is incorporated herein by reference in its entirety: a computer readable form (CRF) of the Sequence Listing (file name: 165002001240SEQLIST.TXT, date recorded: January 13, 2022, size: 70,465 bytes).
BACKGROUND
Hepatitis B virus (HBV), a member of the Hepadnaviridae family, is a noncytopathic hepatic DNA virus that only infects the liver of human and great apes (e.g., chimpanzee, orangutan, bonobo, gorilla). The primary infection of adult humans with HBV causes an acute hepatitis with symptoms of organ inflammation, fever, jaundice and increased liver transaminases in blood. About 10-20% of adult patients are not able to overcome the virus infection and suffer a chronic disease progression over many years with increased risk of developing cirrhotic liver or liver cancer through the development of chronic hepatitis B virus (CHB) infection. Perinatal vertical transmission from mothers with CHB to newborns also leads to chronic hepatitis in about 80% of cases. All patients with CHB are at increased risk of progression to cirrhosis and hepatocellular carcinoma (HCC), depending on host and viral factors (Lampertico et al., J Hepatol., 2017, 67(2):370-398). Hepatically impaired patients are at higher risk of progression to end-stage disease in persons with chronic HBV infection.
Unfortunately, treatment options for HBV infections are limited in hepatically impaired patients, such as patients suffering from liver cirrhosis. Available drugs to treat HBV are often contraindicated in patients with advanced liver disease. There exists a need for safely treating individuals who are affected by hepatitis B viral infection, including individuals with hepatic impairment, and bringing functional cure (FC), whereby the patient (still) has undetectable serum HBsAg and HBV DNA at 6 months (or more) after the end of treatment. The FC essentially puts the CHB patient into a similar state as those who recover from acute HBV infection, a viral latency state maintained by HBV-specific T cells and has been shown to improve survival and health-related quality of life by preventing disease progression, including advanced decompensated cirrhosis and development of HCC. SUMMARY
It has been discovered that subcutaneous administration of particular RNAi duplexes can safely be used to treat Hepatitis B viral (HBV) infections in patients suffering from advanced liver disease, particularly patients that are hepatically impaired. It will be understood that the subjects referred to in the disclosure had liver disease or liver impairment prior to be HBV infection for which they are undergoing treatment.
Provided herein is a method of treating a Hepatitis B viral (HBV) infection in a subject, enhancing an immune response in a subject with a Hepatitis B viral (HBV) infection, decreasing viral replication in a subject with a Hepatitis B viral (HBV) infection, decreasing expression of one or more Hepatitis B Virus (HBV) polypeptide(s), more particularly of one or more polypeptide(s) from HBsAg and HBeAg, in a subject in need thereof, and/or increasing the targeted killing of hepatocytes comprising integrated viral DNA or extrachromosomal DNA in a subject with Hepatitis B viral (HBV) infection, wherein the method comprises administering to the subject an effective amount of a pharmaceutical composition comprising an RNAi component having:
(i) a first RNAi agent comprising: an antisense strand comprising a nucleotide sequence of any one of the following: SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, and SEQ ID NO:7 and a complementary sense strand. In some embodiments, the complementary sense strand comprises a nucleotide sequence of any one of the following: SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, and SEQ ID NO: 15; and
(ii) a second RNAi agent comprising: an antisense strand comprising a nucleotide sequence of any one of the following: SEQ ID NO: 8 and SEQ ID NO:9, and a complementary sense strand. In some embodiments, the complementary sense strand comprises a nucleotide sequence of any one of the following: SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, and SEQ ID NO: 19.
In particular, the disclosure provides methods of treating a Hepatitis B viral (HBV) infection in a hepatically impaired subject, enhancing an immune response in a hepatically impaired subject with a Hepatitis B viral (HBV) infection, decreasing viral replication in a hepatically impaired subject with a Hepatitis B viral (HBV) infection, decreasing expression of one or more Hepatitis B Virus (HBV) polypeptide(s), more particularly of one or more polypeptide(s) from HBsAg and HBeAg, in a hepatically impaired subject in need thereof, and/or increasing the targeted killing of hepatocytes comprising integrated viral DNA or extrachromosomal DNA in a hepatically impaired subject with Hepatitis B viral (HBV) infection, comprising administering an RNAi component as set forth in the disclosure. In some such embodiments, the hepatically impaired subject is also renally impaired.
In some embodiments, the subject has a Child-Pugh score of 7 to 9 (Class B), indicating significant functional hepatic compromise. In some embodiments, the subject has a Child-Pugh score of 10 to 15 (Class C), indicating decompensated disease. In some embodiments where the subject has a Child score of 7-9 or 10-15, the subject also suffers from renal impairment. In some embodiments, the renally impaired subject has a globular filtration rate [GFR] of <90 mL/min. In other embodiments, the renally impaired subject is on dialysis.
In some embodiments, the subject may be suffering from compensated cirrhosis of the liver. In other embodiments, the patient may be suffering from decompensated cirrhosis of the liver.
In some embodiments, the disclosure provides methods of treating HBV in a hepatically impaired subject, said method comprising subcutaneously administering to the subject a pharmaceutical composition comprising about 200 mg of an RNAi component comprising
(i) a first RNAi agent comprising: an antisense strand comprising a nucleotide sequence of SEQ ID NO:2 and a complementary sense strand (e.g., a sense strand comprising a nucleotide sequence of SEQ ID NO: 11); and
(ii) a second RNAi agent comprising: an antisense strand comprising a nucleotide sequence of SEQ ID NO:8 and a complementary sense strand (e.g., a sense strand comprising a nucleotide sequence of SEQ ID NO: 16); or
(i) a first RNAi agent comprising: an antisense strand comprising a nucleotide sequence that is at least 70% homologous to SEQ ID NO:2 and a complementary sense strand comprising a nucleotide sequence that is at least 80% homologous to SEQ ID NO:11; and
(ii) a second RNAi agent comprising: an antisense strand comprising a nucleotide sequence that is at least 70% homologous to a sequence of SEQ ID NO: 8 and a complementary sense strand comprising a nucleotide sequence that is at least 80% homologous to SEQ ID NO: 16, wherein the molar ratio of the first RNAi agent to the second RNAi agent is about 2:1. In some such embodiments, the subject also is renally impaired.
In some embodiments, the disclosure provides methods of treating HBV in a subject suffering from cirrhosis of the liver, said method comprising subcutaneously administering to the subject a pharmaceutical composition comprising about 200 mg of an RNAi component comprising
(i) a first RNAi agent comprising: an antisense strand comprising a nucleotide sequence of SEQ ID NO:2 and a complementary sense strand (e.g., a sense strand comprising a nucleotide sequence of SEQ ID NO: 11); and
(ii) a second RNAi agent comprising: an antisense strand comprising a nucleotide sequence of SEQ ID NO:8 and a complementary sense strand (e.g., a sense strand comprising a nucleotide sequence of SEQ ID NO: 16); or (i) a first RNAi agent comprising: an antisense strand comprising a nucleotide sequence that is at least 70% homologous to SEQ ID NO:2 and a complementary sense strand comprising a nucleotide sequence that is at least 70% homologous to SEQ ID NO:11; and
(ii) a second RNAi agent comprising: an antisense strand comprising a nucleotide sequence that is at least 70% homologous to a nucleotide sequence of SEQ ID NO:8 and a complementary sense strand comprising a nucleotide sequence that is at least 70% homologous to SEQ ID NO: 16, wherein the molar ratio of the first RNAi agent to the second RNAi agent is about 2:1. In some such embodiments, the subject also is renally impaired.
In some embodiments, the disclosure provides methods of treating HBV in a hepatically impaired subject (e.g., a subject suffering from liver cirrhosis), said method comprising subcutaneously administering to the subject a pharmaceutical composition comprising an RNAi component comprising
(i) a first RNAi agent comprising: an antisense strand comprising a nucleotide sequence of SEQ ID NO:2 and a complementary sense strand (e.g., a sense strand comprising a nucleotide sequence of SEQ ID NO: 11); and
(ii) a second RNAi agent comprising: an antisense strand comprising a nucleotide sequence of SEQ ID NO:8 and a complementary sense strand (e.g., a sense strand nucleotide sequence of SEQ ID NO:16); or
(i) a first RNAi agent comprising: an antisense strand comprising a nucleotide sequence that is at least 70% homologous to SEQ ID NO:2 and a complementary sense strand comprising a nucleotide sequence that is at least 70% homologous to SEQ ID NO:11; and
(ii) a second RNAi agent comprising: an antisense strand comprising a nucleotide sequence that is at least 70% homologous to a sequence of SEQ ID NO: 8 and a complementary sense strand comprising a nucleotide sequence that is at least 70% homologous to SEQ ID NO: 16, wherein the molar ratio of the first RNAi agent to the second RNAi agent is about 2:1, and wherein the Cmax of the first component is from about 1,500 ng/mL to about 4,000 ng/mL and the Cmax of the second component is from about 200 ng/mL to about 1,000 ng/mL. In some embodiments, the Cmax of the first component is from about 2,000 ng/mL to about 3,000 ng/mL and the Cmax of the second component is from about 400 ng/mL to about 800 ng/mL. In some such embodiments, the subject also is renally impaired.
In some embodiments, the disclosure provides methods of treating HBV in a hepatically impaired subject (e.g., a subject suffering from liver cirrhosis), said method comprising subcutaneously administering to the subject a pharmaceutical composition comprising an RNAi component comprising (i) a first RNAi agent comprising: an antisense strand comprising a nucleotide sequence of SEQ ID NO:2 and a complementary sense strand (e.g., a sense strand comprising a nucleotide sequence of SEQ ID NO: 11); and
(ii) a second RNAi agent comprising: an antisense strand comprising a nucleotide sequence of SEQ ID NO:8 and a sense strand comprising a complementary nucleotide sequence (e.g., a sense strand comprising a sequence of SEQ ID NO: 16); or
(i) a first RNAi agent comprising: an antisense strand comprising a nucleotide sequence that is at least 70% homologous to SEQ ID NO:2 and a complementary sense strand comprising a nucleotide sequence that is at least 70% homologous to SEQ ID NO:11; and
(ii) a second RNAi agent comprising: an antisense strand comprising a nucleotide sequence that is at least 70% homologous to a sequence of SEQ ID NO: 8 and a complementary sense strand comprising a nucleotide sequence that is at least 70% homologous to SEQ ID NO: 16, wherein the molar ratio of the first RNAi agent to the second RNAi agent is about 2:1, and wherein the AUG / of the first component is from about 20,000 ng.h/mL to about 50,000 ng.h/mL and the AUG , of the second component is from about 3,000 ng.h/mL to about 12,000 ng.h/mL. In some embodiments, the A UGG of the first component is from about 30,000 ng.h/mL to about 40,000 ng.h/mL and the AUCoo of the second component is from about 5,000 ng.h/mL to about 10,000 ng.h/mL. In some such embodiments, the subject also is renally impaired.
In some embodiments disclosed above, the first RNAi comprises an antisense strand comprising a nucleotide sequence that is at least 80% homologous to SEQ ID NO:2 and a complementary sense strand comprising a nucleotide sequence that is at least 80% homologous to SEQ ID NO: 11. In some embodiments, the first RNAi comprises an antisense strand comprising a nucleotide sequence that is at least 85% homologous to SEQ ID NO:2 and a complementary sense strand comprising a nucleotide sequence that is at least 85% homologous to SEQ ID NO: 11. In some embodiments, the first RNAi comprises an antisense strand comprising a nucleotide sequence that is at least 90% homologous to SEQ ID NO:2 and a complementary sense strand comprising a nucleotide sequence that is at least 90% homologous to SEQ ID NO: 11. In some embodiments, the first RNAi comprises an antisense strand comprising a nucleotide sequence that is at least 95% homologous to SEQ ID NO:2 and a complementary sense strand comprising a nucleotide sequence that is at least 95% homologous to SEQ ID NO:11.
In some embodiments disclosed above, the second RNAi comprises an antisense strand comprising a nucleotide sequence that is at least 80% homologous to SEQ ID NO:8 and a complementary sense strand comprising a nucleotide sequence that is at least 80% homologous to SEQ ID NO: 16. In some embodiments, the first RNAi comprises an antisense strand comprising a nucleotide sequence that is at least 85% homologous to SEQ ID NO: 8 and a complementary sense strand comprising a nucleotide sequence that is at least 85% homologous to SEQ ID NO: 16. In some embodiments, the first RNAi comprises an antisense strand comprising a nucleotide sequence that is at least 90% homologous to SEQ ID NO: 8 and a complementary sense strand comprising a nucleotide sequence that is at least 90% homologous to SEQ ID NO: 16. In some embodiments, the first RNAi comprises an antisense strand comprising a nucleotide sequence that is at least 95% homologous to SEQ ID NO:8 and a complementary sense strand comprising a nucleotide sequence that is at least 95% homologous to SEQ ID NO: 16.
In some embodiments disclosed above, the first RNAi comprises an antisense strand comprising a nucleotide sequence that is at least 80% homologous to SEQ ID NO:2 and a complementary sense strand comprising a nucleotide sequence that is at least 80% homologous to SEQ ID NO:11, and the second RNAi comprises an antisense strand comprising a nucleotide sequence that is at least 80% homologous to SEQ ID NO:8 and a complementary sense strand comprising a nucleotide sequence that is at least 80% homologous to SEQ ID NO: 16. In some embodiments disclosed above, the first RNAi comprises an antisense strand comprising a nucleotide sequence that is at least 90% homologous to SEQ ID NO:2 and a complementary sense strand comprising a nucleotide sequence that is at least 90% homologous to SEQ ID NO: 11, and the second RNAi comprises an antisense strand comprising a nucleotide sequence that is at least 90% homologous to SEQ ID NO:8 and a complementary sense strand comprising a nucleotide sequence that is at least 90% homologous to SEQ ID NO: 16. In some embodiments disclosed above, the first RNAi comprises an antisense strand comprising a nucleotide sequence that is at least 95% homologous to SEQ ID NO:2 and a complementary sense strand comprising a nucleotide sequence that is at least 95% homologous to SEQ ID NO:11, and the second RNAi comprises an antisense strand comprising a nucleotide sequence that is at least 95% homologous to SEQ ID NO:8 and a complementary sense strand comprising a nucleotide sequence that is at least 95% homologous to SEQ ID NO: 16.
In some embodiments, the disclosed methods further comprise administering to the subject another agent for treating infection caused by HB V. In some embodiments, the second therapeutic agent is a nucleoside analog. In some such embodiments, the nucleoside analog is entecavir (ETV), tenofovir disoproxil fumarate (TDF), or tenofovir alafenamide. In some embodiments, the nucleoside analog is TDF. In particular embodiments, the nucleoside analog is dosed orally under a dosing regimen that provides an effective amount of the drug to the subject. For instance, the nucleoside analog can be dosed once daily to the subject. Another general aspect of the application relates to a combination or a kit for use in treating a HBV infection, such as a chronic HBV infection (CHB), with or without viral co-infection, e.g., with or without co-infection with HDV and/or HCV and/or HIV, more particularly with or without co-infection with at least HDV, and/or for treating chronic HDV infection (CHD) in a subject in need thereof, comprising:
(1) a pharmaceutical composition comprising an RNAi component having: (i) a first RNAi agent comprising: an antisense strand comprising a nucleotide sequence of any one of the following: SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, and SEQ ID NO:7 and a complementary sense strand (e.g., a sense strand comprising a nucleotide sequence of any one of the following: SEQ ID NO:10, SEQ ID NO:11, SEQ ID NO:12, SEQ ID NO:13, SEQ ID NO:14, and SEQ ID NO:15); and
(ii) a second RNAi agent comprising: an antisense strand comprising a nucleotide sequence of any one of the following: SEQ ID NO: 8 and SEQ ID NO:9, and a sense strand comprising a complementary nucleotide sequence (e.g., a sense strand comprising a nucleotide sequence of any one of the following: SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, and SEQ ID NO: 19).
In some embodiments, the combination or kit is for use in enhancing an immune response, decreasing viral replication, and/or decreasing the expression of one or more Hepatitis B Virus (HBV) polypeptides, more particularly of one or more polypeptide(s) selected from HBsAg and HBeAg, in a subject with a Hepatitis B Virus (HBV) infection, more particularly a chronic HBV infection (CHB) with or without viral co-infection.
In some embodiments, the combination or kit further comprises another agent for treating infection caused by HBV.
Also provided herein is an effective amount of an RNAi component and optionally another agent for treating infection caused by HBV such as a nucleoside analog or a NAP, in the manufacture of a medicament for treating a HBV infection in a subject, enhancing an immune response in a subject with a HBV infection, decreasing viral replication in a subject with a HBV infection, decreasing the expression of one or more HBV polypeptide(s), more particularly of one or more polypeptide(s) from HBsAg and HBeAg, and/or increasing the targeted killing of hepatocytes comprising integrated viral DNA or extrachromosomal DNA in a subject with a HBV infection.
In some embodiments, the RNAi component are administered to the subject over the same treatment period for up to 2 years, up to 1 year, up to 6 months, or up to any time of 1 month to 2 years.
In other embodiments, the treatment comprises a first phase conducted before a second phase, and a) the first phase comprises administering the RNAi component to the subject to thereby decrease the HBsAg to a level low enough to allow recovery of T cell function, preferably to a serum HBsAg level of less than 1000, 100, 10, or 1 lU/mL; and b) the second phase comprises administering an additional compound or drug effective for treating hepatitis infection.
In certain embodiments, the second phase does not comprise administering the RNAi component to the subject. In other embodiments, the second phase further comprises administering the RNAi component to the subject. The first phase of the treatment can last about 1-24 months, such as 1- 12 months, 1-3 months, 4-6 months, 7-9 months, 10-12 months, or any period of time in between. The second phase of the treatment can last about 1-24 months, such as 1-12 months, 4-6 months, 7-9 months, 10-12 months, 13-18 months, 19-24 months, or any period of time in between.
In some embodiments, the RNAi component is administered subcutaneously or intravenously, preferably subcutaneously, at an amount of about 40-1000 mg per dose, more particularly about 40-250 mg per dose, such as about 100-200 mg per dose, more particularly about 200 mg per dose, and it is administered weekly, every two weeks, every 4 weeks, monthly, every 2 months, or every 3 months, preferably every 4 weeks or monthly.
In some embodiments, a subject has achieved at least one of the following features a)-e), more particularly more than one of the following features a)-e), more particularly at least features a), b) and c), more particularly all of features a)-d), during or after the treatment with a combination according to an embodiment of the application: a) decreased HBV replication as measured by serum HBV DNA level, preferably undetectable serum HBV DNA level; b) decreased expression of one or more HBV polypeptide(s), preferably decreased expression of HBsAg as measured by serum HBsAg level, preferably undetectable serum HBsAg level; c) enhanced HBV-specific T cell responses; d) loss of HBeAg or serocoversion for HBeAg, if the subject is HBeAg positive before the treatment; and e) seroconversion for HBsAg.
In some embodiments, an embodiment of the application is for use in treating a subject coinfected with CHB and another chronic infection with at least one of: hepatitis D virus (HDV); hepatitis C virus (HCV); or human immunodeficiency virus (HIV). The combination can be used in a method of decreasing the serum levels of HDV RNA in a subject chronically co-infected with both HBV and HDV; a method of normalizing alanine aminotransferase (ALT) level in a subject chronically coinfected with HBV and HDV; or a method of eradicating HDV infection in a subject chronically coinfected with HBV and HDV.
In any of the methods, the RNAi agent comprises at least one modified nucleotide or at least one modified internucleoside linkage. In another variation, at least 90% or substantially all of the nucleotides in the first and the second RNAi agents are modified nucleotides. In a further variation, the first or the second RNAi agent further comprises a targeting ligand that is conjugated to the first or the second RNAi agent. In one aspect, the targeting ligand comprises N-acetyl-galactosamine. In a particular aspect, the targeting ligand is selected from the group consisting of (NAGI 3), (NAG13)s, (NAGI 8), (NAG18)s, (NAG24), (NAG24)s, (NAG25), (NAG25)s, (NAG26), (NAG26)s, (NAG27), (NAG27)s, (NAG28), (NAG28)s, (NAG29), (NAG29)s, (NAG30), (NAG30)s, (NAG31), (NAG31)s, (NAG32), (NAG32)s, (NAG33), (NAG33)s, (NAG34), (NAG34)s, (NAG35), (NAG35)s, (NAG36), (NAG36)s, (NAG37), (NAG37)s, (NAG38), (NAG38)s, (NAG39), and (NAG39)s. In one variation, the targeting ligand is (NAG25), (NAG25)s, (NAG31), (NAG31)s, (NAG37), or (NAG37)s. In another variation, the targeting ligand is conjugated to the sense strand of the first or the second RNAi agent. In another variation, the targeting ligand is conjugated to the 5’ terminus of the sense stand of the first or the second RNAi agent. In still another variation, the first and the second RNAi agents independently comprise a duplex selected from the group consisting of: an antisense strand comprising SEQ ID NO: 1 and a sense strand comprising SEQ ID NO: 10; an antisense strand comprising SEQ ID NO:2 and a sense strand comprising SEQ ID NO: 11; an antisense strand comprising SEQ ID NO: 3 and a sense strand comprising SEQ ID NO: 11; an antisense strand comprising SEQ ID NO: 4 and a sense strand comprising SEQ ID NO: 12; an antisense strand comprising SEQ ID NO: 8 and a sense strand comprising SEQ ID NO: 16; an antisense strand comprising SEQ ID NO: 8 and a sense strand comprising SEQ ID NO: 17; an antisense strand comprising SEQ ID NO:2 and a sense strand comprising SEQ ID NO: 13; and an antisense strand comprising SEQ ID NO: 8 and a sense strand comprising SEQ ID NO: 18. In a particular variation, the first and the second RNAi agents are each independently conjugated to a targeting ligand comprising N-acetyl-galactosamine, and the first and the second RNAi agents independently comprise a duplex selected from the group consisting of: an antisense strand comprising SEQ ID NO:2 and a sense strand comprising SEQ ID NO: 11; an antisense strand comprising SEQ ID NO: 4 and a sense strand comprising SEQ ID NO: 12; an antisense strand comprising SEQ ID NO: 8 and a sense strand comprising SEQ ID NO: 16; an antisense strand comprising SEQ ID NO:2 and a sense strand comprising SEQ ID NO: 13; and an antisense strand comprising SEQ ID NO: 8 and a sense strand comprising SEQ ID NO: 18. In still another variation, the molar ratio of the first RNAi agent to the second RNAi agent is in the range of about 1:2 to about 5:1. In another variation, the molar ratio of the first RNAi agent to the second RNAi agent is about 2:1. In certain aspects, the first and the second RNAi agents are each independently conjugated to (NAG37)s, the first RNAi agent comprises an antisense strand comprising SEQ ID NO: 2 and a sense strand comprising SEQ ID NO: 11, and the second RNAi agent comprises an antisense strand comprising SEQ ID NO: 8 and a sense strand comprising SEQ ID NO: 16.
In certain embodiments, a method or use according to an embodiment of the application further comprises one or more additional agents for treating HBV, particularly CHB. The other agent can be, for example, a nucleoside analog. In some embodiments, the nucleoside analog is entecavir, tenofovir disoproxil fumarate, tenofovir alafenamide, lamivudine, telbivudine, or a combination thereof. The other agent can also be, for example, a NAP, including, but are not limited to, REP2006, REP2031, REP2055, STOPSTM (S-antigen transport-inhibiting oligonucleotide polymers), and those disclosed in Patent Application Publication Nos. WO200424919; WO201221985; and WO202097342 and U.S. Patent Nos. 7,358,068; 8,008,269; 8,008,270; and 8,067,385, or REP2006, REP2031, REP2055, STOPSTM (S-antigen transport-inhibiting oligonucleotide polymers), and those disclosed in Patent Application Publication Nos. WO200424919; WO201221985; and WO202097342 and U.S. Patent Nos. 7,358,068; 8,008,269; 8,008,270; and 8,067,385. The other agent can also be one or more Capsid Assembly Modulator (CAM). A method or a combination for use according to an embodiment of the application can further comprise one or more of interferons, such as interferon alpha or lambda, preferably a pegylated interferon, more preferably a pegylated interferon alpha-2a or pegylated interferon lambda-la.
Other aspects, features and advantages of the invention will be apparent from the following disclosure, including the detailed description of the invention and its preferred embodiments and the appended claims.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1 illustrates the mean linear (standard deviation) plasma concentration-time profile of an RNAi component of the disclosure after subcutaneous (SC) injection of the RNAi component in participants with liver cirrhosis with moderate hepatic impairment and in participants with normal liver function with no liver cirrhosis. The Y-axis shows mean plasma concentration (ng/mL) of the first RNAi agent.
FIG. 2 illustrates the mean linear (standard deviation) plasma concentration-time profile of an RNAi component of the disclosure after subcutaneous (SC) injection of 200 mg the RNAi component in participants with liver cirrhosis with moderate hepatic impairment and in participants with normal liver function with no liver cirrhosis. The Y-axis shows mean plasma concentration (ng/mL) of the second RNAi agent.
DETAILED DESCRIPTION OF THE INVENTION
DEFINITIONS
Listed below are definitions of various terms used herein. These definitions apply to the terms as they are used throughout this specification and claims, unless otherwise limited in specific instances, either individually or as part of a larger group.
Unless defined otherwise, all technical and scientific terms used herein generally have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Generally, the nomenclature used herein and the laboratory procedures in cell culture, molecular genetics, organic chemistry, and peptide chemistry are those well-known and commonly employed in the art.
As used herein, the articles “a” and “an” refer to one or to more than one (i.e., to at least one) of the grammatical object of the article. By way of example, “an element” means one element or more than one element. Furthermore, use of the term “including” as well as other forms, such as “include,” “includes,” and “included,” is not limiting. As used in the specification and in the claims, the term “comprising” can include the embodiments “consisting of’ and “consisting essentially of.” The terms “comprise(s),” “include(s),” “having,” “has,” “can,” “contain(s),” and variants thereof, as used herein, are intended to be open-ended transitional phrases, terms, or words that require the presence of the named ingredients/steps and permit the presence of other ingredients/steps. However, such description should be construed as also describing compositions or processes as “consisting of’ and “consisting essentially of’ the enumerated compounds, which allows the presence of only the named compounds, along with any pharmaceutically acceptable carriers, and excludes other compounds.
As used herein, approximating language can be applied to modify any quantitative representation that can vary without resulting in a change in the basic function to which it is related. Accordingly, a value modified by a term or terms, such as “substantially,” cannot be limited to the precise value specified, in some cases. In at least some instances, the approximating language can correspond to the precision of an instrument for measuring the value.
As used herein, the term “treatment” or “treating,” is defined as the application or administration of a therapeutic agent, i.e., a compound provided herein (alone or in combination with another pharmaceutical agent), to a patient, or application or administration of a therapeutic agent to an isolated tissue or cell line from a patient (e.g., for diagnosis or ex vivo applications), who has HBV infection, in particular chronic HBV infection, a symptom of HBV infection or the potential to develop HBV infection, with the purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve or affect HBV infection, the symptoms of HBV infection or the potential to develop HBV infection. Such treatments can be specifically tailored or modified, based on knowledge obtained from the field of pharmacogenomics .
As used herein, the term “patient,” “individual” or “subject” refers to a human or a non-human mammal. Non-human mammals include, for example, livestock and pets, such as ovine, bovine, porcine, canine, feline and murine mammals. Preferably, the patient, subject, or individual is human.
As used herein, “treatment naive” refers to a patient, individual or subject not having previously received treatment with a drug, investigational or approved, for HBV infection, in particular a nucleoside or nucleotide analog drug or interferon product.
Alternatively, patients, individuals or subjects treated according to the methods of the disclosure can be “treatment experienced.” As used herein, “treatment experienced” refers to a patient, individual or subject who has had at least one previous course of an HBV antiviral therapy, in particular a nucleoside or nucleotide. Particular nucleosides or nucleotides include entecavir or a pharmaceutically acceptable salt or solvate thereof, such as entecavir monohydrate, or tenofovir or a salt or a prodrug thereof, such as tenofovir alafenamide or tenofovir disoproxil fumarate.
When used with respect to methods of treatment and the use of the compounds and pharmaceutical compositions thereof described herein, an individual “in need thereof’ may be an individual who has been diagnosed with or previously treated for the condition to be treated. Typically, when a step of administering a compound provided herein, the method further contemplates a step of identifying an individual or subject in need of the particular treatment to be administered or having the particular condition to be treated.
As used herein, the term “pharmaceutically acceptable” refers to a material, such as a carrier or diluent, which does not abrogate the biological activity or properties of the compound, and is relatively non-toxic, i.e., the material can be administered to an individual without causing undesirable biological effects or interacting in a deleterious manner with any of the components of the composition in which it is contained.
As used herein, the term “pharmaceutically acceptable salt” refers to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or base moiety to its salt form. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. The pharmaceutically acceptable salts provided herein include the conventional non-toxic salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. The pharmaceutically acceptable salts provided herein can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., 1985, p. 1418 and Journal of Pharmaceutical Science, 66, 2 (1977), each of which is incorporated herein by reference in its entirety.
As used herein, the term “composition” or “pharmaceutical composition” refers to a mixture of at least one compound provided herein with a pharmaceutically acceptable carrier. The pharmaceutical composition facilitates administration of the compound to a patient or subject. Multiple techniques of administering a compound exist in the art including, but not limited to, intravenous, oral, aerosol, parenteral, ophthalmic, pulmonary and topical administration.
As used herein, the term “pharmaceutically acceptable carrier” means a pharmaceutically acceptable material, composition or carrier, such as a liquid or solid filler, stabilizer, dispersing agent, suspending agent, diluent, excipient, thickening agent, solvent or encapsulating material, involved in carrying or transporting a compound provided herein within or to the patient such that it can perform its intended function. Typically, such constructs are carried or transported from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation, including the compound provided herein, and not injurious to the patient. Some examples of materials that can serve as pharmaceutically acceptable carriers include: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol; polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar; buffering agents, such as magnesium hydroxide and aluminum hydroxide; surface active agents; alginic acid; pyrogen-free water; isotonic saline; Ringer’s solution; ethyl alcohol; phosphate buffer solutions; and other non-toxic compatible substances employed in pharmaceutical formulations. As used herein, “pharmaceutically acceptable carrier” also includes any and all coatings, antibacterial and antifungal agents, and absorption delaying agents, and the like that are compatible with the activity of the compound provided herein, and are physiologically acceptable to the patient. Supplementary active compounds can also be incorporated into the compositions. The “pharmaceutically acceptable carrier” can further include a pharmaceutically acceptable salt of the compound provided herein. Other additional ingredients that can be included in the pharmaceutical compositions provided herein are known in the art and described, for example in Remington's Pharmaceutical Sciences (Genaro, Ed., Mack Publishing Co., 1985, Easton, PA), which is incorporated herein by reference.
The term “tablet,” as used herein, denotes an orally administrable, single-dose, solid dosage form that can be produced by compressing a drug substance or a pharmaceutically acceptable salt thereof, with suitable excipients (e.g., fillers, disintegrants, lubricants, glidants, and/or surfactants) by conventional tableting processes. The tablet can be produced using conventional granulation methods, for example, wet or dry granulation, with optional comminution of the granules with subsequent compression and optional coating. The tablet can also be produced by spray-drying.
As used herein, the terms “effective amount,” “pharmaceutically effective amount,” and “therapeutically effective amount” refer to a nontoxic but sufficient amount of an agent to provide the desired biological result. That result may be reduction or alleviation of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system. An appropriate therapeutic amount in any individual case may be determined by one of ordinary skill in the art using routine experimentation.
The term “combination”, “therapeutic combination”, “pharmaceutical combination”, or “combination product” as used herein refer to a non-fixed combination or a kit of parts for the combined administration where two or more therapeutic agents can be administered independently, at the same time or separately within time intervals, especially where these time intervals allow that the combination partners show a cooperative, e.g., synergistic, effect.
As used herein “Adverse Event (AE)” is any untoward medical event that occurs in a subject administered with an investigational product, e.g. an RNAi component disclosed herein or a pharmaceutical formulation thereof, and it does not necessarily indicate only events with clear causal relationship with the relevant investigational product. As used herein, the term “alkyl,” by itself or as part of another substituent means, unless otherwise stated, a straight or branched chain hydrocarbon having the number of carbon atoms designated (i.e., Ci-Ce-alkyl means an alkyl having one to six carbon atoms) and includes straight and branched chains. In an embodiment, Ci-Ce alkyl groups are provided herein. Examples include methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, pentyl, neopentyl, and hexyl. Other examples of Ci-Ce-alkyl include ethyl, methyl, isopropyl, isobutyl, n-pentyl, and n-hexyl.
As used herein, the term “alkoxy” refers to an alkyl (carbon and hydrogen chain) group singular bonded to oxygenlike for instance a methoxy group or ethoxy group.
As used herein, the term “amino” refers to a functional group having the formulae -NHz, - NH(alkyl), and -N(alkyl)2, wherein alkyl is as defined herein.
As used herein, the term “amide” refers to a functional group having the formulae -C(O)N(R)2 or -N(R)C(O)alkyl, wherein the carbon atom is doubly bound to the oxygen atom and R is independently at each occurrence hydrogen or alkyl.
As used herein, the term “ester” refers to a functional group having the formulae -C(O)alkoxy, COzalkyl, -OC(O)alkyl, wherein the carbon atom is doubly bound to one oxygen atom and singly bound to an alkoxy group as defined herein.
As used herein, the term “halo” or “halogen” alone or as part of another substituent means, unless otherwise stated, a fluorine, chlorine, bromine, or iodine atom, preferably, fluorine, chlorine, or bromine, more preferably, fluorine or chlorine.
As used herein, the term “nitrile” refers to the functional group -CN, where carbon is triply bound to nitrogen.
As used herein, the term “heterocycle” refers to molecules that are saturated or partially saturated an include tetrahydrofuran, oxetane, dioxane or other cyclic ethers. Heterocycle also includes bicyclic structures that may be bridged or spirocyclic in nature with each individual ring within the bicycle varying from 3-8 atoms, and containing 0, 1, or 2 N, O, or S atoms. The term “heterocyclyl” includes cyclic esters (i.e., lactones) and cyclic amides (i.e., lactams) and also specifically includes, but is not limited to, epoxidyl, oxetanyl, tetrahydrofuranyl, tetrahydropyranyl (i.e., oxanyl), pyranyl, dioxanyl, aziridinyl, azetidinyl, pyrrolidinyl, 2,5-dihydro-lH-pyrrolyl, oxazolidinyl, thiazolidinyl, piperidinyl, morpholinyl, piperazinyl, thiomorpholinyl, 1,3-oxazinanyl, 1,3-thiazinanyl, 2- azabicyclo[2.1.1]hexanyl, 5-azabicyclo[2.1.1]hexanyl, 6-azabicyclo[3.1.1] heptanyl, 2- azabicyclo[2.2.1]heptanyl, 3-azabicyclo[3.1.1]heptanyl, 2-azabicyclo[3.1.1]heptanyl, 3- azabicyclo[3.1.0]hexanyl, 2-azabicyclo[3.1.0]hexanyl, 3-azabicyclo[3.2.1]octanyl, 8- azabicyclo[3.2.1]octanyl, 3-oxa-7-azabicyclo[3.3.1]nonanyl, 3-oxa-9-azabicyclo[3.3.1]nonanyl, 2-oxa- 5-azabicyclo[2.2. l]heptanyl, 6-oxa-3-azabicyclo[3.1. l]heptanyl, 2-azaspiro[3.3]heptanyl, 2-oxa-6- azaspiro[3.3]heptanyl, 2-oxaspiro[3.3]heptanyl, 2-oxaspiro[3.5]nonanyl, 3-oxaspiro[5.3]nonanyl, and 8-oxabicyclo[3.2. l]octanyl.
It will be understood that when a carbon is in the (/^-configuration, this implies that said carbon is an asymmetric carbon. As the skilled person will acknowledge, symmetric carbons, not being stereocenters, cannot be in the (7?)- or (Sj-configuration. Only asymmetric carbons can be in said configurations. Thus, it will be understood that the carbon of R3 bonded to the amine in the 4-position of the quinazoline is an asymmetric carbon.
Whenever sustituents are represented by chemical structure, ” represents the bond of attachment to the remainder of the molecule. Lines (such as
Figure imgf000017_0001
drawn into a particular ring of a ring system indicate that the bond may be attached to any of the suitable ring atoms.
HBV infections that may be treated according to the disclosed methods include HBV genotype A, B, C, and/or D infections. However, in an embodiment, the methods disclosed may treat any HBV genotype (“pan-genotypic treatment”). HBV genotyping may be performed using methods known in the art, for example, INNO-LIPA® HBV Genotyping, Innogenetics N.V., Ghent, Belgium).
Hepatic impairment is a condition wherein normal functioning of the liver reduced. Hepatic impairment can be acute, with rapid onset, or chronic. Chronic hepatic impairment, or cirrhosis, can occur from many causes, such as excessive consumption of alcohol, hepatitis, autoimmune disease, heredity, or metabolism, or can be idiopathic. Liver damage is generally irreversible, and treatment consists of prevention of progression and treatment of symptoms. In severe cases, liver transplant is the only option. Hepatic impairment can exhibit no significant symptoms, or may be characterized by such symptoms as reduced ability for the blood to clot (coagulopathy) and brain dysfunction (encephalopathy), fluid retention in the abdominal cavity, increased infection risk, hypogonadism, change in liver size, jaundice, and increased sensitivity to medication. The Child- Pugh score is a system for assessing the prognosis of hepatic impairment — including the required strength of treatment and necessity of liver transplant — of chronic liver disease, primarily cirrhosis. It provides a forecast of the increasing severity of the liver disease.
Child-Pugh Group, Child-Pugh Class, and the like: a ranking of level of hepatic impairment based on the Child-Pugh Score. Child-Pugh Scores of 5-6 are classified as Child-Pugh Class A (mild hepatic impairment) and have an expected 2 year survival rate of 85%. Child-Pugh Scores of 7-9 are classified as Child-Pugh Class B (moderate hepatic impairment) and have an expected 2 year survival rate of 57%. Child-Pugh Scores of 10-15 are classified as Child-Pugh Class C (severe hepatic impairment) and have an expected 2 year survival rate of 35%.
Child-Pugh Score: a score based on five clinical measures of hepatic impairment, including levels of total bilirubin, serum albumin, PT INR, ascites, and hepatic encephalopathy. Each measure is given a ranking of 1, 2, or 3, and the sum of the five rankings is the Child-Pugh Score. The Child-Pugh Score can be used to classify hepatic impairment by placing subjects in a Child-Pugh Group.
The levels of hepatic sufficiency of an individual can include, for example, no hepatic impairment (i.e., normal hepatic function), mild hepatic impairment, moderate hepatic impairment, severe hepatic impairment. In some embodiments, the level of hepatic sufficiency is not specified. The methods herein can comprise prescribing or administering different remedies such as RNAi component Compound 1 or a pharmaceutically acceptable salt or a solvate thereof.
As used herein, unless otherwise noted, the term “isolated form” means that the compound is present in a form which is separate from any biological environment (e.g. plasma, blood, gastric fluids, urine, cerebrospinal fluid, and the like).
In particular embodiments of the application, a therapeutically effective amount refers to the amount of a composition or therapeutic combination which is sufficient to achieve one, two, three, four, or more of the following effects: (i) reduce or ameliorate the severity of an HBV infection or a symptom associated therewith; (ii) reduce the duration of an HBV infection or symptom associated therewith; (iii) prevent the progression of an HBV infection or symptom associated therewith; (iv) cause regression of an HBV infection or symptom associated therewith; (v) prevent the development or onset of an HBV infection, or symptom associated therewith; (vi) treat or retreat a chronic HBV infection that recurs due to relapse after functional cure is achieved or symptom associated therewith; (vii) prevent the recurrence of an HBV infection or symptom associated therewith; (viii) reduce hospitalization of a subject having an HBV infection; (ix) reduce hospitalization length of a subject having an HBV infection; (x) increase the survival of a subject with an HBV infection; (xi) eliminate an HBV infection in a subject; (xii) inhibit or reduce HBV replication in a subject; and/or (xiii) enhance or improve the prophylactic or therapeutic effect(s) of another therapy.
A therapeutically effective amount can also be an amount of the compound sufficient to reduce HBsAg levels consistent with evolution to clinical seroconversion; achieve sustained HBsAg clearance associated with reduction of infected hepatocytes by a subject’s immune system; induce HBV-antigen specific activated T-cell populations; and/or achieve persistent loss of HBsAg during or after treatment that then preferably persists at 6 months or more after the end of treatment, most preferably for life.
As used herein, the terms and phrases “in combination,” “in combination with,” “co-delivery,” and “administered together with” in the context of the administration of two or more therapies or components to a subject refers to simultaneous administration or subsequent administration of two or more therapies or components, such as two vectors, e.g., DNA plasmids, peptides, or a therapeutic combination and an adjuvant. “Simultaneous administration” or “simultaneously administered” refers to administration of the two or more therapies or components within the same treatment period, e.g., at least within the same day. When two components are “administered together with,” “administered in combination with,” or “administered within the same treatment period,” they can be administered in separate compositions sequentially within a short time period. “Overlapping administration” refers to administration of the two or more therapies or components not within the same overall treatment period, but with at least one overlapping treatment period. “Subsequent administration” can be administration of the two or more therapies or components during different treatment periods, one after the other. The use of the term “in combination with” does not restrict the order in which therapies or components are administered to a subject. For example, a first therapy or component (e.g. an RNAi component) can be administered prior to (e.g., 5 minutes to one hour before), concomitantly with or simultaneously with, or subsequent to (e.g., 5 minutes to one hour after) the administration of a second therapy or component. In other embodiments, a first therapy or component (e.g. an RNAi component) and a second therapy or component or a stereoisomer or a tautomeric form thereof), a pharmaceutically acceptable salt or a solvate thereof are administered in separate compositions, such as two separate compositions.
RNAi Component
In one aspect, the RNAi component comprises one or more RNAi agents. Each RNAi agent disclosed herein includes at least a sense strand and an antisense strand. The sense strand and the antisense strand can be partially, substantially, or fully complementary to each other. The length of the RNAi agent sense and antisense strands described herein each can be 16 to 30 nucleotides in length. In some embodiments, the sense and antisense strands are independently 17 to 26 nucleotides in length. In some embodiments, the sense and antisense strands are independently 19 to 26 nucleotides in length. In some embodiments, the sense and antisense strands are independently 21 to 26 nucleotides in length. In some embodiments, the sense and antisense strands are independently 21 to 24 nucleotides in length. The sense and antisense strands can be either the same length or different lengths. The HBV RNAi agents disclosed herein have been designed to include antisense strand sequences that are at least partially complementary to a sequence in the HBV genome that is conserved across the majority of known serotypes of HBV. The RNAi agents described herein, upon delivery to a cell expressing HBV, inhibit the expression of one or more HBV genes in vivo or in vitro.
An RNAi agent includes a sense strand (also referred to as a passenger strand) that includes a first sequence, and an antisense strand (also referred to as a guide strand) that includes a second sequence. A sense strand of the HBV RNAi agents described herein includes a core stretch having at least about 85% identity to a nucleotide sequence of at least 16 consecutive nucleotides in an HBV mRNA. In some embodiments, the sense strand core nucleotide stretch having at least about 85% identity to a sequence in an HBV mRNA is 16, 17, 18, 19, 20, 21, 22, or 23 nucleotides in length. An antisense strand of an HBV RNAi agent comprises a nucleotide sequence having at least about 85% complementary over a core stretch of at least 16 consecutive nucleotides to a sequence in an HBV mRNA and the corresponding sense strand. In some embodiments, the antisense strand core nucleotide sequence having at least about 85% complementarity to a sequence in an HBV mRNA or the corresponding sense strand is 16, 17, 18, 19, 20, 21, 22, or 23 nucleotides in length.
In some embodiments, the RNAi component comprises a first RNAi agent comprising an antisense strand comprising a nucleotide sequence of any one of the following: SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO:6, and SEQ ID NO:7, and a complementary sense strand (e.g., a sense strand comprising a nucleotide sequence of any one of the following: SEQ ID NO:10, SEQ ID NO:11, SEQ ID NO:12, SEQ ID NO:13, SEQ ID NO:14, and SEQ ID NO: 15), or a second RNAi agent comprising an antisense strand comprising a nucleotide sequence of any one of the following: SEQ ID NO:8 and SEQ ID NO:9, and a complementary sense strand (e.g., a sense comprising a nucleotide sequence of any one of the following: SEQ ID NO:16, SEQ ID NO:17, SEQ ID NO:18, and SEQ ID NO:19). In some embodiments, the RNAi component comprises a first RNAi agent comprising an antisense strand comprising a nucleotide sequence of any one of the following: SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO:6, and SEQ ID NO:7, and a complementary sense strand (e.g., a sense strand comprising a nucleotide sequence of any one of the following: SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO:14, and SEQ ID NO:15), and a second RNAi agent comprising an antisense strand comprising a nucleotide sequence of any one of the following: SEQ ID NO: 8 and SEQ ID NO:9, and a complementary sense strand (e.g., a sense strand comprising a nucleotide sequence of any one of the following: SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, and SEQ ID NO: 19).
In some embodiments, the first and the second RNAi agents disclosed herein comprise any of the sequences in Table 1.
Table 1. Exemplary sequences for first and second RNAi agents
Figure imgf000020_0001
Targeting Group
In some embodiments, the RNAi agents are delivered to target cells or tissues using any oligonucleotide delivery technology known in the art. Nucleic acid delivery methods include, but are not limited to, by encapsulation in liposomes, by iontophoresis, or by incorporation into other vehicles, such as hydrogels, cyclodextrins, biodegradable nanocapsules, and bioadhesive microspheres, proteinaceous vectors or Dynamic Polyconjugates (DPCs) (see, for example WO 2000/053722, WO 2008/0022309, WO 2011/104169, and WO 2012/083185, each of which is incorporated herein by reference). In some embodiments, an HBV RNAi agent is delivered to target cells or tissues by covalently linking the RNAi agent to a targeting group. In some embodiments, the targeting group can include a cell receptor ligand, such as an asialoglycoprotein receptor (ASGPr) ligand. In some embodiments, an ASGPr ligand includes or consists of a galactose derivative cluster. In some embodiments, a galactose derivative cluster includes an N-acetyl-galactosamine trimer or an N-acetyl- galactosamine tetramer. In some embodiments, a galactose derivative cluster is an N-acetyl- galactosamine trimer or an N-acetyl-galactosamine tetramer.
A targeting group can be linked to the 3' or 5' end of a sense strand or an antisense strand of an HBV RNAi agent. In some embodiments, a targeting group is linked to the 3' or 5' end of the sense strand. In some embodiments, a targeting group is linked to the 5’ end of the sense strand. In some embodiments, a targeting group is linked to the RNAi agent via a linker.
In some embodiments, the RNAi component comprises a combination or cocktail of a first and a second RNAi agent having different nucleotide sequences. In some embodiments, the first and the second RNAi agents are each separately and independently linked to targeting groups. In some embodiments, the first and the second RNAi agents are each linked to targeting groups comprised of N-acetyl-galactosamines. In some embodiments, when first and the second RNAi agents are included in a composition, each of the RNAi agents is linked to the same targeting group. In some embodiments, when first and the second RNAi agents are included in a composition, each of the RNAi agents is linked to different targeting groups, such as targeting groups having different chemical structures.
In some embodiments, targeting groups are linked to the first and the second RNAi agents without the use of an additional linker. In some embodiments, the targeting group is designed having a linker readily present to facilitate the linkage to the first or the second RNAi agent. In some embodiments, when the first and the second RNAi agents are included in a composition, the first and the second RNAi agents may be linked to the targeting groups using the same linkers. In some embodiments, when the first and the second RNAi agents are included in a composition, the first and the second RNAi agents are linked to the targeting groups using different linkers.
Examples of targeting groups and linking groups are provided in Table 2. The non-nucleotide group can be covalently linked to the 3' and/or 5' end of either the sense strand and/or the antisense strand. In some embodiments, the first or second RNAi agent contains a non-nucleotide group linked to the 3' and/or 5' end of the sense strand. In some embodiments, a non-nucleotide group is linked to the 5 ' end of the first or second RNAi agent sense strand. A non-nucleotide group may be linked directly or indirectly to the first or second RNAi agent via a linker/linking group. In some embodiments, a non- nucleotide group is linked to the first or second RNAi agent via a labile, cleavable, or reversible bond or linker. Targeting groups and linking groups include the following, for which their chemical structures are provided below in Table 2: (PAZ), (NAG13), (NAG13)s, (NAG18), (NAG18)s, (NAG24), (NAG24)s, (NAG25), (NAG25)s, (NAG26), (NAG26)s, (NAG27), (NAG27)s, (NAG28), (NAG28)s, (NAG29), (NAG29)s, (NAG30), (NAG30)s, (NAG31), (NAG31)s, (NAG32), (NAG32)s, (NAG33), (NAG33)s, (NAG34), (NAG34)s, (NAG35), (NAG35)s, (NAG36), (NAG36)s, (NAG37), (NAG37)s,
(NAG38), (NAG38)s, (NAG39), (NAG39)s. Each sense strand and/or antisense strand can have any targeting groups or linking groups listed above, as well as other targeting or linking groups, conjugated to the 5' and/or 3' end of the sequence.
Table 2. Structures Representing Various Modified Nucleotides, Targeting Groups, And
Figure imgf000023_0001
Figure imgf000024_0001
Figure imgf000025_0001
Figure imgf000026_0001
Figure imgf000027_0001
Figure imgf000028_0001
Figure imgf000029_0001
Figure imgf000030_0001
Figure imgf000031_0001
Figure imgf000032_0001
Figure imgf000033_0001
Figure imgf000034_0001
Figure imgf000035_0001
Figure imgf000036_0001
Modified Nucleotides
In some embodiments, the first or the second RNAi agent contains one or more modified nucleotides. As used herein, a “modified nucleotide” is a nucleotide other than a ribonucleotide (2'- hydroxyl nucleotide). In some embodiments, at least 50% (e.g., at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 97%, at least 98%, at least 99%, or 100%) of the nucleotides are modified nucleotides. As used herein, modified nucleotides include, but are not limited to, deoxyribonucleotides, nucleotide mimics, abasic nucleotides (represented herein as Ab), 2'-modified nucleotides, 3' to 3' linkages (inverted) nucleotides (represented herein as invdN, invN, invn, invAb), non-natural base-comprising nucleotides, bridged nucleotides, peptide nucleic acids (PNAs), 2', 3 '-seco nucleotide mimics (unlocked nucleobase analogs, represented herein as NUNA or NUNA), locked nucleotides (represented herein as NLNA or NLNA), 3'-O-methoxy (2' intemucleoside linked) nucleotides (represented herein as 3'-OMen), 2'-F-Arabino nucleotides (represented herein as NfANA or NfANA), 5'-Me, 2'-fluoro nucleotide (represented herein as 5Me-Nf), morpholino nucleotides, vinyl phosphonate deoxyribonucleotides (represented herein as vpdN), vinyl phosphonate containing nucleotides, and cyclopropyl phosphonate containing nucleotides (cPrpN). 2'-modified nucleotides (i.e. a nucleotide with a group other than a hydroxyl group at the 2' position of the five-membered sugar ring) include, but are not limited to, 2'-O-methyl nucleotides (represented herein as a lower case letter 'n' in a nucleotide sequence), 2'-deoxy-2'-fluoro nucleotides (represented herein as Nf, also represented herein as 2'-fluoro nucleotide), 2'-deoxy nucleotides (represented herein as dN), 2 '-methoxy ethyl (2'- O-2-methoxylethyl) nucleotides (represented herein as NM or 2'-M0E), 2'-amino nucleotides, and 2'- alkyl nucleotides. It is not necessary for all positions in a given compound to be uniformly modified. Conversely, more than one modification can be incorporated in the first or second RNAi agent or even in a single nucleotide thereof. The RNAi agent sense strands and antisense strands may be synthesized and/or modified by methods known in the art. Modification at one nucleotide is independent of modification at another nucleotide. Modified nucleobases include synthetic and natural nucleobases, such as 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and 0-6 substituted purines, (e.g., 2-aminopropyladenine, 5-propynyluracil, or 5-propynylcytosine), 5 -methylcytosine (5-me-C), 5 -hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-alkyl (e.g., 6-methyl, 6-ethyl, 6-isopropyl, or 6-n-butyl) derivatives of adenine and guanine, 2-alkyl (e.g., 2-methyl, 2-ethyl, 2-isopropyl, or 2-n-butyl) and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine, 2-thiocytosine, 5-halouracil, cytosine, 5-propynyl uracil, 5-propynyl cytosine, 6-azo uracil, 6-azo cytosine, 6-azo thymine, 5-uracil (pseudouracil), 4-thiouracil, 8-halo, 8-amino, 8-sulfhydryl, 8-thioalkyl, 8-hydroxyl and other 8- substituted adenines and guanines, 5-halo (e.g., 5-bromo), 5-trifluoromethyl, and other 5-substituted uracils and cytosines, 7-methylguanine and 7-methyladenine, 8-azaguanine and 8-azaadenine, 7-deazaguanine, 7-deazaadenine, 3-deazaguanine, and 3-deazaadenine.
In some embodiments, all or at least 90% of the nucleotides of the first or the second RNAi agent are modified nucleotides. As used herein, an RNAi agent wherein at least 90% of the nucleotides present are modified nucleotides is an RNAi agent having four or fewer (i.e., 0, 1, 2, 3, or 4) nucleotides in both the sense strand and the antisense strand being ribonucleotides. As used herein, a sense strand, wherein at least 90% of the nucleotides present are modified nucleotides, is a sense strand having two or fewer (i.e., 0, 1, or 2) nucleotides in the sense strand being ribonucleotides. As used herein, an antisense sense strand, wherein at least 90% of the nucleotides present are modified nucleotides, is an antisense strand having two or fewer (i.e., 0, 1, or 2) nucleotides in the sense strand being ribonucleotides. In some embodiments, one or more nucleotides of an RNAi agent is a ribonucleotide.
Modified Intemucleoside Linkages
In some embodiments, one or more nucleotides of the first or the second RNAi agent are linked by non-standard linkages or backbones i.e., modified internucleoside linkages or modified backbones). In some embodiments, a modified internucleoside linkage is a non-phosphate-containing covalent internucleoside linkage. Modified internucleoside linkages or backbones include, but are not limited to, 5’-phosphorothioate groups (represented herein as a lower case “s”), chiral phosphorothioates, thiophosphates, phosphorodi thioates, phosphotriesters, aminoalkyl-phosphotriesters, alkyl phosphonates (e.g., methyl phosphonates or 3'-alkylene phosphonates), chiral phosphonates, phosphinates, phosphoramidates (e.g., 3'-amino phosphoramidate, aminoalkylphosphoramidates, or thionophosphoramidates), thionoalkyl-phosphonates, thionoalkylphosphotriesters, morpholino linkages, boranophosphates having normal 3'-5' linkages, 2'-5' linked analogs of boranophosphates, or boranophosphates having inverted polarity wherein the adjacent pairs of nucleoside units are linked 3'- 5' to 5'-3' or 2'-5' to 5 '-2'. In some embodiments, a modified internucleoside linkage or backbone lacks a phosphorus atom. Modified internucleoside linkages lacking a phosphorus atom include, but are not limited to, short chain alkyl or cycloalkyl inter-sugar linkages, mixed heteroatom and alkyl or cycloalkyl inter-sugar linkages, or one or more short chain heteroatomic or heterocyclic inter-sugar linkages. In some embodiments, modified internucleoside backbones include, but are not limited to, siloxane backbones, sulfide backbones, sulfoxide backbones, sulfone backbones, formacetyl and thioformacetyl backbones, methylene formacetyl and thioformacetyl backbones, alkene-containing backbones, sulfamate backbones, methyleneimino and methylenehydrazino backbones, sulfonate and sulfonamide backbones, amide backbones, and other backbones having mixed N, O, S, and CH2 components.
In some embodiments, a sense strand of the first or the second RNAi agent can contain 1, 2, 3, 4, 5, or 6 phosphorothioate linkages, an antisense strand of the first or the second RNAi agent can contain 1, 2, 3, 4, 5, or 6 phosphorothioate linkages, or both the sense strand and the antisense strand independently can contain 1, 2, 3, 4, 5, or 6 phosphorothioate linkages. In some embodiments, a sense strand of the first or the second RNAi agent can contain 1, 2, 3, or 4 phosphorothioate linkages, an antisense strand of the first or the second RNAi agent can contain 1 , 2, 3, or 4 phosphorothioate linkages, or both the sense strand and the antisense strand independently can contain 1, 2, 3, or 4 phosphorothioate linkages.
In some embodiments, the first or the second RNAi agent sense strand contains at least two phosphorothioate internucleoside linkages. In some embodiments, the at least two phosphorothioate internucleoside linkages are between the nucleotides at positions 1-3 from the 3' end of the sense strand. In some embodiments, the at least two phosphorothioate internucleoside linkages are between the nucleotides at positions 1-3, 2-4, 3-5, 4-6, 4-5, or 6-8 from the 5' end of the sense strand. In some embodiments, the first or the second RNAi agent antisense strand contains four phosphorothioate internucleoside linkages. In some embodiments, the four phosphorothioate internucleoside linkages are between the nucleotides at positions 1-3 from the 5' end of the sense strand and between the nucleotides at positions 19-21, 20-22, 21-23, 22-24, 23-25, or 24-26 from the 5' end. In some embodiments, the first or the second RNAi agent contains at least two phosphorothioate internucleoside linkages in the sense strand and three or four phosphorothioate internucleoside linkages in the antisense strand.
In some embodiments, the first or the second RNAi agent contains one or more modified nucleotides and one or more modified internucleoside linkages. In some embodiments, a 2'-modified nucleoside is combined with modified internucleoside linkage.
In some embodiments, the modified antisense strand sequences comprising the RNAi component has one of the sequences shown in Table 3. Table 3 shows the modified sequence of the antisense strands as well as their underlying unmodified sequences. In some embodiments, the modified sense strand sequences comprising the RNAi component has one of the sequences shown in Table 4. Table 4 shows the modified sequence of the sense strands as well as their underlying unmodified sequences. Table 3. Antisense Strand Sequences.
Figure imgf000039_0001
Figure imgf000040_0001
Figure imgf000041_0001
Figure imgf000042_0001
Figure imgf000043_0001
Figure imgf000044_0001
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000048_0001
Figure imgf000049_0001
In some embodiments, the first and the second RNAi agents disclosed herein comprise any of the modified sequences in Table 5.
Table 5. Exemplary modified sequences for first and second RNAi agents
Figure imgf000050_0001
A = adenosine-3'-phosphate;
C = cytidine-3 '-phosphate;
G = guanosine-3 '-phosphate;
U = uridine-3 '-phosphate n = any 2'-0Me modified nucleotide a = 2'-O-methyladenosine-3 '-phosphate as = 2'-O-methyladenosine-3'-phosphorothioate c = 2'-O-methylcytidine-3 '-phosphate cs = 2'-O-methylcytidine-3'-phosphorothioate g = 2'-O-methylguanosine-3 '-phosphate gs = 2'-O-methylguanosine-3'-phosphorothioate t = 2'-O-methyl-5-methyluridine-3'-phosphate ts = 2'-O-methyl-5-methyluridine-3'-phosphorothioate u = 2'-O-methyluridine-3'-phosphate us = 2'-O-methyluridine-3'-phosphorothioate
Nf = any 2'-fluoro modified nucleotide
Af = 2'-fluoroadenosine-3 '-phosphate
Afs = 2'-fluoroadenosine-3'-phosporothioate
Cf = 2'-fluorocytidine-3 '-phosphate
Cfs = 2'-fluorocytidine-3'-phosphorothioate
Gf = 2'-fluoroguanosine-3 '-phosphate
Gfs = 2'-fluoroguanosine-3'-phosphorothioate
Tf = 2'-fluoro-5'-methyluridine-3'-phosphate
Tfs = 2'-fluoro-5'-methyluridine-3'-phosphorothioate
Uf = 2'-fluorouridine-3 '-phosphate
Ufs = 2'-fluorouridine-3'-phosphorothioate dN = any 2'-deoxyribonucleotide dT = 2'-deoxythymidine-3 '-phosphate
NUNA = 2', 3 '-seco nucleotide mimics (unlocked nucleobase analogs)
NLNA = locked nucleotide
NfANA = 2'-F-Arabino nucleotide
NM = 2 '-methoxy ethyl nucleotide
AM = 2'-methoxyethyladenosine-3 '-phosphate
AMs = 2'-methoxyethyladenosine-3'-phosphorothioate
TM = 2'-methoxyethylthymidine-3 '-phosphate
TMs = 2'-methoxyethylthymidine-3'-phosphorothioate
R = ribitol
(invdN)= any inverted deoxyribonucleotide (3 '-3' linked nucleotide)
(invAb)= inverted (3 '-3' linked) abasic deoxyribonucleotide, see Table 7
(invAb)s = inverted (3 '-3' linked) abasic deoxyribonucleotide-5 '- phosphorothioate, see Table 7
(invn) = any inverted 2'-0Me nucleotide (3'-3' linked nucleotide) s = phosphorothioate linkage
Notably, the sense strands in Tables 4 and 5 include a targeting group (NAG25, NAG25s, NAG 37, or NAG37s) at the 5' end. It will be understood that the disclosure also includes sense strands that have sequences displayed in Tables 4 and 5 but without the targeting group on the 5' end or with targeting groups other than NAG25, NAG25s, NAG37, or NAG37s, as disclosed herein. It will be further understood that both the antisense and/or sense strands displayed in Tables 3 and 4 can be modified either at the 5' end or 3' end with a targeting group, as disclosed herein.
In some embodiments, the first RNAi agent comprises SEQ ID NO: 5 and SEQ ID NO: 14. In some embodiments, the first RNAi agent comprises SEQ ID NO: 6 and SEQ ID NO: 14. In some embodiments, the first RNAi agent comprises SEQ ID NO: 7 and SEQ ID NO: 15. In some embodiments, the first RNAi agent comprises SEQ ID NO: 1 and SEQ ID NO: 10, 11 or 13. In some embodiments, the first RNAi agent comprises SEQ ID NO: 2 and SEQ ID NO: 10, 11 or 13. In some embodiments, the first RNAi agent comprises SEQ ID NO: 3 and SEQ ID NO: 10, 11, or 13. In some embodiments, the first RNAi agent comprises SEQ ID NO: 4 and SEQ ID NO: 12. In some embodiments, the second RNAi agent comprises SEQ ID NO: 9 and SEQ ID NO: 19. In some embodiments, the second RNAi agent comprises SEQ ID NO: 8 and SEQ ID NO: 16, 17 or 18.
In some embodiments, the RNAi component comprises a first RNAi agent comprising SEQ ID NO: 5 and SEQ ID NO: 14 and a second RNAi agent comprising SEQ ID NO: 9 and SEQ ID NO: 19. In some embodiments, the RNAi component comprises a first RNAi agent comprising SEQ ID NO: 6 and SEQ ID NO: 14 and a second RNAi agent comprising SEQ ID NO: 9 and SEQ ID NO: 19. In some embodiments, the RNAi component comprises a first RNAi agent comprising SEQ ID NO: 7 and SEQ ID NO: 15 and a second RNAi agent comprising SEQ ID NO: 9 and SEQ ID NO: 19.
In some embodiments, the RNAi component comprises a first RNAi agent comprising SEQ ID NO: 1 and SEQ ID NO: 10, 11 or 13 and a second RNAi agent comprising SEQ ID NO: 8 and SEQ ID NO: 16, 17 or 18. In some embodiments, the RNAi component comprises a first RNAi agent comprising SEQ ID NO: 2 and SEQ ID NO: 10, 11 or 13 and a second RNAi agent comprising SEQ ID NO: 8 and SEQ ID NO: 16, 17 or 18. In some embodiments, the RNAi component comprises a first RNAi agent comprising SEQ ID NO: 3 and SEQ ID NO: 10, 11 or 13 and a second RNAi agent comprising SEQ ID NO: 8 and SEQ ID NO: 16, 17 or 18. In some embodiments, the RNAi component comprises a first RNAi agent comprising SEQ ID NO: 4 and SEQ ID NO: 12 and a second RNAi agent comprising SEQ ID NO: 8 and SEQ ID NO: 16, 17 or 18. In some embodiments, the RNAi component comprises a first and a second RNAi agent in a molar ratio of about 1:1, 2:1, 3:1, 4:1 or 5:1. In some embodiments, the two HBV RNAi agents are administered in a molar ratio of about 2:1.
In some embodiments, the first and the second RNAi agents are each independently conjugated to (NAG37)s, the first RNAi agent comprises an antisense strand comprising SEQ ID NO: 2 and a sense strand comprising SEQ ID NO: 11, the second RNAi agent comprises an antisense strand comprising SEQ ID NO: 8 and a sense strand comprising SEQ ID NO: 16.
In some embodiments, an HBV RNAi agent comprises an antisense strand and a sense strand having the modified nucleotide sequences of any of the antisense strand and/or sense strand nucleotide sequences of any of the duplexes of Table 6.
In some embodiments, an HBV RNAi agent comprises an antisense strand and a sense strand having the modified nucleotide sequences of any of the antisense strand and/or sense strand nucleotide sequences of any of the duplexes of Table 6, and further comprises an asialoglycoprotein receptor ligand targeting group.
In some embodiments, an HBV RNAi agent comprises any of the duplexes of Table 6.
In some embodiments, an HBV RNAi agent consists of any of the duplexes of Table 6.
Table 6. Examples of HBV RNAi agent duplexes.
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
In some embodiments, one of the RNAi agents comprising the RNAi componented by the following structure, shown as the sodium salt:
Figure imgf000056_0001
In some embodiments, one of the RNAi agents comprising the RNAi componented by the following structure, shown as the sodium salt:
Figure imgf000057_0001
5 In some embodiments, one of the RNAi agents comprising the RNAi component is represented by the following structure, shown as the free acid:
Figure imgf000058_0001
In some embodiments, one of the RNAi agents comprising the RNAi component is represented by the following structure, shown as the sodium salt:
Figure imgf000059_0001
In some embodiments, one of the RNAi agents comprising the RNAi componented by the following structure, shown as the sodium salt:
Figure imgf000060_0001
In some embodiments, one of the RNAi agents comprising the RNAi component is represented by the following structure, shown as the free acid:
Figure imgf000061_0001
In some embodiments, one of the RNAi agents comprising the RNAi component is represented by the following structure, shown as the free acid:
Figure imgf000062_0001
In some embodiments, one of the RNAi agents comprising the RNAi component is represented by the following structure, shown as the free acid:
Figure imgf000063_0001
The combinations described herein can be used in any methods or kits described below.
Compositions and Administrations
Also provided herein is one or more compositions comprising an RNAi component.
As appropriate compositions there can be cited all compositions usually employed for systemically administering drugs. To prepare the pharmaceutical compositions of this invention, an effective amount of the particular compound, optionally in addition salt form, as the active ingredient is combined in intimate admixture with a pharmaceutically acceptable carrier, which carrier may take a wide variety of forms depending on the form of preparation desired for administration. These pharmaceutical compositions are desirable in unitary dosage form suitable, particularly, for administration orally, rectally, percutaneously, or by parenteral injection. For example, in preparing the compositions in oral dosage form, any of the usual pharmaceutical media may be employed such as, for example, water, glycols, oils, alcohols and the like in the case of oral liquid preparations such as suspensions, syrups, elixirs, emulsions and solutions; or solid carriers such as starches, sugars, kaolin, lubricants, binders, disintegrating agents and the like in the case of powders, pills, capsules, and tablets. Because of their ease in administration, tablets and capsules represent the most advantageous oral dosage unit forms, in which case solid pharmaceutical carriers are employed. For parenteral compositions, the carrier will usually comprise sterile water, at least in large part, though other ingredients, for example, to aid solubility, may be included. Injectable solutions, for example, may be prepared in which the carrier comprises saline solution, glucose solution or a mixture of saline and glucose solution. Injectable suspensions may also be prepared in which case appropriate liquid carriers, suspending agents and the like may be employed. Also included are solid form preparations intended to be converted, shortly before use, to liquid form preparations. In the compositions suitable for percutaneous administration, the carrier optionally comprises a penetration enhancing agent and/or a suitable wetting agent, optionally combined with suitable additives of any nature in minor proportions, which additives do not introduce a significant deleterious effect on the skin. The compounds of the present invention may also be administered via oral inhalation or insufflation in the form of a solution, a suspension or a dry powder using any art-known delivery system.
It is especially advantageous to formulate the aforementioned pharmaceutical compositions in unit dosage form for ease of administration and uniformity of dosage. Unit dosage form as used herein refers to physically discrete units suitable as unitary dosages, each unit containing a predetermined quantity of active ingredient calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. Examples of such unit dosage forms are tablets (including scored or coated tablets), capsules, pills, suppositories, powder packets, wafers, injectable solutions or suspensions and the like, and segregated multiples thereof. The first and second RNAi agents of an RNAi component that targets or reduces HBsAg can be formulated in the same or separate pharmaceutical compositions. The HBV RNAi agents in the same or separate compositions can be formulated with the same or different excipients and carriers. The HBV RNAi agents in the same or separate compositions can be administered through same or different administration routes.
Any suitable pharmaceutical composition comprising the first and/or second RNAi agents of the RNAi component and a pharmaceutically acceptable carrier can be used in the present invention in view of the present disclosure. The pharmaceutical composition can comprise any RNAi component described herein or otherwise known in the art. One or more pharmaceutically acceptable excipients (including vehicles, carriers, diluents, and/or delivery polymers) can be mixed with the first and/or second RNAi agents of the RNAi component, thereby forming a pharmaceutical formulation suitable for in vivo delivery to a human.
The HBV RNAi agents disclosed herein can be administered via any suitable parenteral route in a pharmaceutical composition appropriately tailored to the particular route. Thus, herein described pharmaceutical compositions can be administered by injection, for example, intravenously, intramuscularly, subcutaneously, or intraperitoneally. In some embodiments, there herein described pharmaceutical compositions are preferably via subcutaneous injection.
The pharmaceutical compositions including an HBV RNAi agent described herein can be delivered to a cell, group of cells, tumor, tissue, or subject using oligonucleotide delivery technologies known in the art. In general, any suitable method recognized in the art for delivering a nucleic acid molecule (in vitro or in vivo) can be adapted for use with a composition described herein. For example, a pharmaceutical composition comprising at least one of the first and second RNAi agents of an RNAi component described herein, can be delivered by systemic administration via a parenteral route, including subcutaneous, intravenous, intraperitoneal, and intramuscular administration. In certain embodiments, the compositions are administered by subcutaneous or intravenous infusion or injection.
As used herein, a pharmaceutical composition or medicament includes a pharmacologically effective amount of at least one of the described therapeutic compounds and a pharmaceutically acceptable carrier. The pharmaceutically acceptable carrier can comprise one or more pharmaceutically acceptable excipients. Pharmaceutically acceptable excipients, also referred to herein as “excipients”, are substances other than the active pharmaceutical ingredient that are intentionally included in the drug delivery system. Excipients do not exert or are not intended to exert a therapeutic effect at the intended dosage. Excipients can act to a) aid in processing of the drug delivery system during manufacture, b) protect, support or enhance stability, bioavailability or patient acceptability of the API, c) assist in product identification, and/or d) enhance any other attribute of the overall safety, effectiveness, of delivery of the API during storage or use. A pharmaceutically acceptable excipient may or may not be an inert substance.
Excipients include, but are not limited to: absorption enhancers, anti-adherents, anti-foaming agents, anti-oxidants, binders, buffering agents, carriers, coating agents, colors, delivery enhancers, delivery polymers, dextran, dextrose, diluents, emulsifiers, extenders, humectants, oils, polymers, preservatives, saline, salts, solvents, sugars, suspending agents, sustained release matrices, tonicity agents, and vehicles.
Pharmaceutical compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. For intravenous administration, suitable carriers include physiological saline, bacteriostatic water, Cremophor ELTM (BASF, Parsippany, NJ) or phosphate buffered saline. It should be stable under the conditions of manufacture and storage and should be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, and sodium chloride in the composition. Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate and gelatin.
Sterile injectable solutions can be prepared by incorporating the active compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filter sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle which contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, methods of preparation include vacuum drying and freeze-drying which yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
The active compounds can be prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, poly anhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art. Liposomal suspensions can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Patent No. 4,522,811.
The TLR8 agonist and/or HBV RNAi agents can be formulated in compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the disclosure are dictated by and directly dependent on the unique characteristics of the active compound and the therapeutic effect to be achieved, and the limitations inherent in the art of compounding such an active compound for the treatment of individuals.
A pharmaceutical composition can contain other additional components commonly found in pharmaceutical compositions. Such additional components include, but are not limited to: anti-pruritics, astringents, local anesthetics, or anti-inflammatory agents (e.g., antihistamine, diphenhydramine, etc.). It is also envisioned that cells, tissues or isolated organs that express or comprise the herein defined RNAi agents can be used as “pharmaceutical compositions.” As used herein, “pharmacologically effective amount,” “therapeutically effective amount,” or simply “effective amount” refers to that amount of a RNAi agent to produce a pharmacological, therapeutic or preventive result.
The amount administered will likely depend on such variables as the overall health status of the patient, the relative biological efficacy of the compound delivered, the formulation of the drug, the presence and types of excipients in the formulation, and the route of administration. Also, it is to be understood that the initial dosage administered can be increased beyond the above upper level in order to rapidly achieve the desired blood-level or tissue level, or the initial dosage can be smaller than the optimum.
For treatment of disease or for formation of a medicament or composition for treatment of a disease, the pharmaceutical compositions described herein including a HBV RNAi agent can be combined with an excipient or with a second therapeutic agent or treatment including, but not limited to: a second or other RNAi agent, a small molecule drug, an antibody, an antibody fragment, and/or a vaccine.
The described HBV RNAi agents, when added to pharmaceutically acceptable excipients and/or adjuvants, can be packaged into kits, containers, packs, or dispensers. The pharmaceutical compositions described herein can be packaged in pre-filled syringes or vials.
The application also provides methods of making compositions and therapeutic combinations of the application. In another aspect, the kit further comprises a package insert including, without limitation, appropriate instructions for preparation and administration of the formulation, side effects of the formulation, and any other relevant information. The instructions can be in any suitable format, including, but not limited to, printed matter, videotape, computer readable disk, optical disc or directions to internet-based instructions.
In another aspect, kits for treating an individual who suffers from or is susceptible to the conditions described herein are provided, comprising a first container comprising a dosage amount of a composition or formulation as disclosed herein, and a package insert for use. The container can be any of those known in the art and appropriate for storage and delivery of intravenous formulation. In certain embodiments, the kit further comprises a second container comprising a pharmaceutically acceptable carrier, diluent, adjuvant, etc. for preparation of the formulation to be administered to the individual.
In another aspect, kits can also be provided that contain sufficient dosages of the compositions described herein (including pharmaceutical compositions thereof) to provide effective treatment for an individual for an extended period, such as 1-3 days, 1-5 days, a week, 2 weeks, 3, weeks, 4 weeks, 6 weeks, 8 weeks, 1 cycle, 2 cycles, 3 cycles, 4 cycles, 5 cycles, 6 cycles, 7 cycles, 8 cycles or more. In some embodiments, one cycle of treatment is about 1 month, about 2 months, about 3 months, about 4 months, about 5 months, about 6 months, about 7 months, about 8 months, about 9 months, about 10 months, about 11 months, about 12 months, about 15 months, about 18 months, about 21 months or about 24 months.
In some embodiments, the kits can also include multiple doses and may be packaged in quantities sufficient for storage and use in pharmacies, for example, hospital pharmacies and compounding pharmacies. In certain embodiments the kits may include a dosage amount of at least one composition as disclosed herein.
Treatment Regimen
Any of the compositions and therapeutic combinations of the application described herein can be used in a combination treatment regimen or a treatment method of the application.
In some embodiments of the application, an effective amount of an RNAi component in the range of about 25-600 mg per dose is administered to the subject. In some embodiments, the effective amount of the RNAi component is in the range of about 25-50 mg, about 50-75 mg, about 75-100 mg, about 100-150 mg, about 150-200 mg, about 200-250 mg, about 250-300 mg, about 300-400 mg, about 400-500 mg or about 500-600 mg per dose. In some embodiments, an effective amount of the RNAi component is about 25 mg, about 30 mg, about 35 mg, about 40 mg, about 45 mg, about 50 mg, about 55 mg, about 60 mg, about 65 mg, about 70 mg, about 75 mg, about 80 mg, about 85 mg, about 90 mg, about 95 mg, about 100 mg, about 125 mg, about 150 mg, about 175 mg, about 200 mg, about 250 mg, about 300 mg, about 350 mg, about 400 mg, about 450 mg, about 500 mg, about 550 mg or about 600 mg per dose. In some embodiments, an effective amount of the RNAi component is about 25 mg, about 35mg, about 40 mg, about 50 mg, about 100 mg or about 200 mg per dose. The effective amount of the RNAi component can be administered once per month (Q1M), per 4 weeks (Q4W), bimonthly, or any time period in between. As used herein, unless otherwise specified, the dose of an RNAi component or agent refers to the dose of the RNAi component or agent itself, and not to the dose of the composition that can contain the RNAi component or agent. For example, if an RNAi conjugate, such as an RNAi- (NAG37)s conjugate is being administered, the dose of the RNAi component or agent refers to the amount of the RNAi component or agent of the conjugate. The dose of an RNAi component refers to the combined amount of the first and second RNAi agents of the RNAi component.
In some embodiments, the first and second HBV RNAi agents of an RNAi component are administered in a molar ratio of about 1:1, 2:1, 3:1, 4:1 or 5:1. In some embodiments, the first and second HBV RNAi agents of an RNAi component are administered to a subject in a molar ratio of about 2:1.
In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 25-75 mg per dose administration and in the molar ratio of about 2:1, about 3:1, about 1:1, about 4:1, about 5:1 or about 1:2. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 35-40 mg per dose administration and in the molar ratio of about 2:1, about 3:1, about 1:1, about 4:1, about 5:1 or about 1:2. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 50-125 mg per dose administration and in the molar ratio of about 2:1, about 3:1, about 1:1, about 4:1, about 5:1 or about 1:2. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 75-150 mg per dose administration and in the molar ratio of about2:l, about 3:1, about 1:1, about 4:1, about 5:1 or about 1:2. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 100-200 mg per dose administration and in the molar ratio of about 2:1, about 3:1, about 1:1, about 4:1, about 5:1 or about 1:2. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 150- 250 mg per dose administration and in the molar ratio ofabout 2:l, about 3:l, about 1:1, about 4:1, about 5:1 or about 1:2. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 200-300 mg per dose administration and in the molar ratio of about 2:1, about 3:1, about 1:1, about 4:1, about 5:1 or about 1:2. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 300-400 mg per dose administration and in the molar ratio of about 2:1, about 3:1, about 1:1, about 4:1, about 5:1 or about 1:2. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 50-100 mg per dose administration and in the molar ratio of about 2:1, about 3:1, about 1:1, about 4:1, about 5:1 or about 1:2. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 25-400 mg per dose administration and in the molar ratio of about 2:1. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 25-75 mg per dose administration and in the molar ratio of about 2:1. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 35-40 mg per dose administration and in the molar ratio of about 2:1. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 50-125 mg per dose administration and in the molar ratio of about 2:1. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 75-150 mg per dose administration and in the molar ratio of about 2:1. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 100- 200 mg per dose administration and in the molar ratio of about 2: 1. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 125-225 mg per dose administration and in the molar ratio of about 2:1. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 150-250 mg per dose administration and in the molar ratio of about 2:1. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 200- 300 mg per dose administration and in the molar ratio of about 2: 1. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 300-400 mg per dose administration and in the molar ratio of about 2:1. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 100 mg per dose administration and in the molar ratio of about 2:1. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 25 mg per dose administration and in the molar ratio of about 2: 1. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 35 mg per dose administration and in the molar ratio of about 2: 1. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 40 mg per dose administration and in the molar ratio of about 2:1. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 50 mg per dose administration and in the molar ratio of about 2:1. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 75 mg per dose administration and in the molar ratio of about 2:1. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 200 mg per dose administration and in the molar ratio of about 2:1.
In some embodiments, the first RNAi agent is administered in an amount of about 3-650 mg per dose administration, and the second RNAi agent is administered in an amount of about 2-325 mg per dose administration. In some embodiments, the first RNAi agent is administered in an amount of about 15-150 mg per dose administration, and the second RNAi agent is administered in an amount of about 5-75 mg per dose administration. In some embodiments, the first RNAi agent is administered in an amount of about 35-265 mg per dose administration. In some embodiments, the first RNAi agent is administered in an amount of about 50-75 mg per dose administration. In some embodiments, the first RNAi agent is administered in an amount of about 15-75 mg per dose administration. In some embodiments, the second RNAi agent is administered in an amount of about 20-125 mg per dose administration. In some embodiments, the second RNAi agent is administered in an amount of about 25- 50 mg per dose administration. In some embodiments, the second RNAi agent is administered in an amount of about 5-40 mg per dose administration. In some embodiments, the first RNAi agent is administered in an amount of about 17 mg per dose administration, and the second RNAi agent is administered in an amount of about 8 mg per dose administration. In some embodiments, the first RNAi agent is administered in an amount of about 23 mg per dose administration, and the second RNAi agent is administered in an amount of about 12 mg per dose administration. In some embodiments, the first RNAi agent is administered in an amount of about 27 mg per dose administration, and the second RNAi agent is administered in an amount of about 13 mg per dose administration. In some embodiments, the first RNAi agent is administered in an amount of about 33 mg per dose administration, and the second RNAi agent is administered in an amount of about 17 mg per dose administration. In some embodiments, the first RNAi agent is administered in an amount of about 67 mg per dose administration, and the second RNAi agent is administered in an amount of about 33 mg per dose administration.
In some embodiments, two RNAi agents are administered at a combined dose of 25-400 mg per dose administration. In an embodiment, two RNAi agents are administered at a combined dose of 25- 400 mg, and the first RNAi agent is administered with the second RNAi agent at a molar ratio of 1:1. In an embodiment, the dose of each of the first and second RNAi agents is in an amount of about 12 mg for a combined dose of about 25 mg. In an embodiment, the dose of each of the first and second RNAi agents is in an amount of about 17 mg for a combined dose of about 35 mg. In an embodiment, the dose of each of the first and second RNAi agents is in an amount of about 20 mg for a combined dose of about 40 mg. In an embodiment, the dose of each of the first and second RNAi agents is in an amount of about 25 mg for a combined dose of about 50 mg. In an embodiment, the dose of each of the first and second RNAi agents is in an amount of about 50 mg for a combined dose of about 100 mg. In an embodiment, the dose of each of the first and second RNAi agents is in an amount of about 100 mg for a combined dose of about 200 mg. In an embodiment, the dose of each of the first and second RNAi agents is in an amount of about 150 mg for a combined dose of about 300 mg. In an embodiment, the dose of each of the first and second RNAi agents is in an amount of about 200 mg for a combined dose of about 400 mg.
In an embodiment, two RNAi agents are administered at a combined dose of 25-400 mg per dose, and the first RNAi agent is administered with the second RNAi agent at a molar ratio of 2:1. In an embodiment, the dose of the first RNAi agent is in an amount of about 16 mg, and the dose of the second RNAi agent is in an amount of about 8 mg for a combined dose of about 25 mg. In an embodiment, the dose of the first RNAi agent is in an amount of about 24 mg, and the dose of the second RNAi agent is in an amount of about 12 mg for a combined dose of about 35 mg. In an embodiment, the dose of the first RNAi agent is in an amount of about 27 mg, and the dose of the second RNAi agent is in an amount of about 13 mg for a combined dose of about 40 mg. In an embodiment, the dose of the first RNAi agent is in an amount of about 33 mg, and the dose of the second RNAi agent is in an amount of about 17 mg for a combined dose of about 50 mg. In an embodiment, the dose of the first RNAi agent is in an amount of about 65 mg, and the dose of the second RNAi agent is in an amount of about 35 mg for a combined dose of about 100 mg. In an embodiment, the dose of the first RNAi agent is in an amount of about 133 mg, and the dose of the second RNAi agent is in an amount of about 67 mg for a combined dose of about 200 mg. In an embodiment, the dose of the first RNAi agent is in an amount of about 200 mg, and the dose of the second RNAi agent is in an amount of about 100 mg for a combined dose of about 300 mg. In an embodiment, the dose of the first RNAi agent is in an amount of about 270 mg, and the dose of the second RNAi agent is in an amount of about 135 mg for a combined dose of about 400 mg.
In an embodiment, two RNAi agents are administered at a combined dose of 25-400 mg per dose, the first RNAi agent is administered with the second RNAi agent at a molar ratio of 3:1. In an embodiment, the dose of the first RNAi agent is in an amount of about 18 mg, and the dose of the second RNAi agent is in an amount of about 6 mg for a combined dose of about 25 mg. In an embodiment, the dose of the first RNAi agent is in an amount of about 27 mg, and the dose of the second RNAi agent is in an amount of about 9 mg for a combined dose of about 35 mg. In an embodiment, the dose of the first RNAi agent is in an amount of about 30 mg, and the dose of the second RNAi agent is in an amount of about 10 mg for a combined dose of about 40 mg. In an embodiment, the dose of the first RNAi agent is in an amount of about 36 mg, and the dose of the second RNAi agent is in an amount of about 12 mg for a combined dose of about 50 mg. In an embodiment, the dose of the first RNAi agent is in an amount of about 75 mg, and the dose of the second RNAi agent is in an amount of about 25 mg for a combined dose of about 100 mg. In an embodiment, the dose of the first RNAi agent is in an amount of about 150 mg, and the dose of the second RNAi agent is in an amount of about 50 mg for a combined dose of about 200 mg. In an embodiment, the dose of the first RNAi agent is in an amount of about 225 mg, and the dose of the second RNAi agent is in an amount of about 75 mg for a combined dose of about 300 mg. In an embodiment, the dose of the first RNAi agent is in an amount of about 300 mg, and the dose of the second RNAi agent is in an amount of about 100 mg for a combined dose of about 400 mg.
In some embodiments, the first RNAi agent and the second RNAi agent are administered in a combined amount of about 25-400 mg per dose administration. In some embodiments, the first RNAi agent and the second RNAi agent are administered in a combined amount of about 25-50 mg, 50-75 mg, 75-100 mg, 100-125 mg, 125-150 mg, 150-175 mg, 175-200 mg, 200-225 mg, 225-250 mg, 250-275 mg, 275-300 mg, 300-325 mg, 325-350 mg, 350-375 mg, 375-400 mg, 25-75 mg, 50-100 mg, 100-150 mg, 150-200 mg, 200-250 mg, 250-300 mg, 300-350 mg, 350-400 mg, 25-100 mg, 50-150 mg, 100-200 mg, 150-250 mg, 200-300 mg, 300-400 mg, 25-200 mg, or 200-400 mg per dose administration. In some embodiments, the first RNAi agent to the second RNAi agent are administered in a combined amount of about 25 mg, about 50 mg, about 100 mg, about 125 mg, about 150 mg, about 175 mg, about 200 mg, about 225 mg, about 250 mg, about 275 mg, about 300 mg, about 325 mg, about 350 mg, about 375 mg, or about 400 mg per dose administration. In some embodiments, the first RNAi agent and the second RNAi agent are administered in a combined amount of about 50 mg, about 75 mg, about 100 mg, or about 125 mg per dose administration. In some embodiments, the first RNAi agent and the second RNAi agent are administered in a combined amount of about 25 mg, about 35 mg, about 40 mg, or about 200 mg per dose administration.
In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 1-10 mg/kg per dose administration. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 1-5 mg/kg per dose administration. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 1-1.5 mg/kg, about 1.5-2.0 mg/kg, about 2.0-2.5 mg/kg, about 2.5-3.0 mg/kg, about 3.0-3.5 mg/kg, about 3.5-4.0 mg/kg, about 4.0- 4.5 mg/kg, about 4.5-5.0 mg/kg, about 5.0-5.5 mg/kg, about 5.5-6.0 mg/kg, about 6.0-6.5 mg/kg, about 6.5-7.0 mg/kg, about 7.0-7.5 mg/kg, about 7.5-8.0 mg/kg, about 8.0-8.5 mg/kg, about 8.5-9.0 mg/kg, about 9.0-9.5 mg/kg, about 9.5-10 mg/kg, about 1-2.5 mg/kg, about 2.5-5.0 mg/kg, about 5.0-7.5 mg/kg, about 7.5-10 mg/kg, about 1-5.0 mg/kg, or about 5.0-10 mg/kg per dose administration.
In some embodiments, the first RNAi agent is administered in an amount of about 0.6-7 mg/kg per dose administration, and the second RNAi agent is administered in an amount of about 0.3-5 mg/kg per dose administration. In some embodiments, the second RNAi agent is administered in an amount of about 0.5-2.5 mg/kg per dose administration. In some embodiments, the second RNAi agent is administered in an amount of about 0.3-1.5 mg/kg per dose administration. In some embodiments, the first RNAi agent is administered in an amount of about 0.6-5 mg/kg per dose administration. In some embodiments, the first RNAi agent is administered in an amount of about 1-2.5 mg/kg per dose administration.
In some embodiments, the two RNAi agents are administered in about 1-18 week intervals. In some embodiments, the two RNAi agents are administered in about 1-week intervals, about 2-week intervals, about 3-week intervals, about 4-week intervals, about 5-week intervals, about 6-week intervals, about 7-week intervals, about 8-week intervals, about 9-week intervals, about 10-week intervals, about 11 -week intervals, about 12-week intervals, about 13-week intervals, about 14-week intervals, about 15- week intervals, about 16-week intervals, about 17-week intervals, or about 18-week intervals. In some embodiments, the two RNAi agents are administered in about 1-6 month intervals. In some embodiments, the two RNAi agents are administered in about 1 -month intervals, about 2-month intervals, about 3-month intervals, about 4-month intervals, about 5-month intervals, or about 6-month intervals. In some embodiments, the two RNAi agents are administered in about 4-week intervals or 1- month intervals. In some embodiments, the two RNAi agents are administered once per month.
In some embodiments, the first RNAi agent and the second RNAi agent are administered for a duration of about 1-12 months. In some embodiments, the first RNAi agent and the second RNAi agent are administered for a duration of at least about 1 month, at least about 2 months, at least about 3 months, at least about 4 months, at least about 5 months, at least about 6 months, at least about 7 months, at least about 8 months, at least about 9 months, at least about 10 months, at least about 11 months or at least about 12 months. In some embodiments, the first RNAi agent and the second RNAi agent are administered for a duration of about 1-18 weeks. In some embodiments, the first RNAi agent and the second RNAi agent are administered for a duration of at least about 1 week, at least about 5 weeks, at least about 10 weeks, at least about 15 weeks, at least about 20 weeks, at least about 25 weeks, at least about 30 weeks, at least about 35 weeks, at least about 40 weeks, at least about 45 weeks, at least about 50 weeks, at least about 55 weeks, at least about 60 weeks, at least about 65 weeks, at least about 70 weeks, at least about 75 weeks, at least about 80 weeks, at least about 90 weeks, or at least 96 weeks.
In some embodiments, the first RNAi agent and the second RNAi agent are administered at a combined dose of 25-600mg, more particularly 25-400 mg per dose administration. In an embodiment, the first RNAi agent and the second RNAi agent are administered at a combined dose of 25-600mg, more particularly 25-400 mg, and the first RNAi agent is administered with the second RNAi agent at a molar ratio of 1:1. In an embodiment, the dose of the first RNAi agent is administered with the second RNAi agent is in an amount of about 12 mg for a combined dose of about 25 mg. In an embodiment, the dose of each of the first RNAi agent and the second RNAi agent is in an amount of about 17 mg for a combined dose of about 35 mg. In an embodiment, the dose of each of the first RNAi agent and the second RNAi agent is in an amount of about 20 mg for a combined dose of about 40 mg. In an embodiment, the dose of each of the first RNAi agent and the second RNAi agent is in an amount of about 25 mg for a combined dose of about 50 mg. In an embodiment, the dose of each of the first RNAi agent and the second RNAi agent is in an amount of about 50 mg for a combined dose of about 100 mg. In an embodiment, the dose of each of the first RNAi agent and the second RNAi agent is in an amount of about 100 mg for a combined dose of about 200 mg. In an embodiment, the dose of each of the first RNAi agent and the second RNAi agent is in an amount of about 150 mg for a combined dose of about 300 mg. In an embodiment, the dose of each of the first RNAi agent and the second RNAi agent is in an amount of about 200 mg for a combined dose of about 400 mg.
In an embodiment, the first RNAi agent and the second RNAi agent are administered at a combined dose of 25-600 mg, more particularly 25-400 mg per dose, and the second RNAi agent is administered with the first RNAi agent at a molar ratio of 1:2. In an embodiment, the dose of the first RNAi agent is in an amount of about 16 mg, and the dose of the second RNAi agent is in an amount of about 8 mg for a combined dose of about 25 mg. In an embodiment, the dose of the second RNAi agent is in an amount of about 12 mg, and the dose of the first RNAi agent is in an amount of about 24 mg for a combined dose of about 35 mg. In an embodiment, the dose of the first RNAi agent is in an amount of about 27 mg, and the dose of the second RNAi agent is in an amount of about 13 mg for a combined dose of about 40 mg. In an embodiment, the dose of the first RNAi agent is in an amount of about 33 mg, and the dose of the second RNAi agent is in an amount of about 17 mg for a combined dose of about 50 mg. In an embodiment, the dose of the second RNAi agent is in an amount of about 35 mg, and the dose of the first RNAi agent is in an amount of about 65 mg for a combined dose of about 100 mg. In an embodiment, the dose of the second RNAi agent is in an amount of about 67 mg, and the dose of the first RNAi agent is in an amount of about 133 mg for a combined dose of about 200 mg. In an embodiment, the dose of the second RNAi agent is in an amount of about 100 mg, and the dose of the first RNAi agent is in an amount of about 200 mg for a combined dose of about 300 mg. In an embodiment, the dose of the second RNAi agent is in an amount of about 135 mg, and the dose of the first RNAi agent is in an amount of about 270 mg for a combined dose of about 400 mg.
In an embodiment, the first RNAi agent and the second RNAi agent are administered at a combined dose of 25-600 mg, more particularly 25-400 mg per dose, the second RNAi agent is administered with the first RNAi agent at a molar ratio of 1:3. In an embodiment, the dose of the first RNAi agent is in an amount of about 18 mg, and the dose of the second RNAi agent is in an amount of about 6 mg for a combined dose of about 25 mg. In an embodiment, the dose of the second RNAi agent is in an amount of about 9 mg, and the dose of the first RNAi agent is in an amount of about 27 mg for a combined dose of about 35 mg. In an embodiment, the dose of the first RNAi agent is in an amount of about 30 mg, and the dose of the second RNAi agent is in an amount of about 10 mg for a combined dose of about 40 mg. In an embodiment, the dose of the first RNAi agent is in an amount of about 36 mg, and the dose of the second RNAi agent is in an amount of about 12 mg for a combined dose of about 50 mg. In an embodiment, the dose of the second RNAi agent is in an amount of about 25 mg, and the dose of the first RNAi agent is in an amount of about 75 mg for a combined dose of about 100 mg. In an embodiment, the dose of the second RNAi agent is in an amount of about 50 mg, and the dose of the first RNAi agent is in an amount of about 150 mg for a combined dose of about 200 mg. In an embodiment, the dose of the second RNAi agent is in an amount of about 75 mg, and the dose of the first RNAi agent is in an amount of about 225 mg for a combined dose of about 300 mg. In an embodiment, the dose of the second RNAi agent is in an amount of about 100 mg, and the dose of the first RNAi agent is in an amount of about 300 mg for a combined dose of about 400 mg.
In some embodiments, about 1 mg/kg (mpk) of the first RNAi agent and about 1 mg/kg of the second RNAi agent are administered to a subject in need thereof. In some embodiments, about 1.5 mg/kg of the first RNAi agent and about 1.5 mg/kg of the second RNAi agent are administered to a subject in need thereof. In some embodiments, about 2.0 mg/kg of the first RNAi agent and about 1.0 mg/kg of the second RNAi agent are administered to a subject in need thereof. In some embodiments, about 3.0 mg/kg of the first RNAi agent and about 1.0 mg/kg of the second RNAi agent are administered to a subject in need thereof. In some embodiments, about 3.2 mg/kg of the first RNAi agent and about 0.8 mg/kg of the second RNAi agent are administered to a subject in need thereof. In some embodiments, about 2.7 mg/kg of the first RNAi agent and about 1.3 mg/kg of the second RNAi agent are administered to a subject in need thereof. In some embodiments, about 4.0 mg/kg of the first RNAi agent and about 1.0 mg/kg of the second RNAi agent are administered to a subject in need thereof. In some embodiments, about 3.3 mg/kg of the first RNAi agent and about 1.7 mg/kg of the second RNAi agent are administered to a subject in need thereof. In some embodiments, between about 0.05 and about 5 mg/kg of the first RNAi agent and between about 0.05 and about 5 mg/kg of the second RNAi agent are administered to a subject in need thereof. In some embodiments, about the first RNAi agent and about the second RNAi agent are administered separately (e.g., in separate injections). In some embodiments, the respective dose of the first RNAi agent and the respective dose of the second RNAi agent are administered together (e.g., in the same injection). In some embodiments, the respective dose of the first RNAi agent and the respective dose of the second RNAi agent are prepared in a single pharmaceutical composition.
In some embodiments, the RNAi component is administered to the subject once monthly in a dose of about 40-350 mg, such as about 40-250 mg, more particularly 40-200 mg, more particularly 100 mg or 200 mg, more particularly 200 mg.
In some embodiments, the first and the second RNAi agents are each independently conjugated to (NAG37)s, the first RNAi agent comprises an antisense strand comprising SEQ ID NO: 2 and a sense strand comprising SEQ ID NO: 11 , and the second RNAi agent comprises an antisense strand comprising SEQ ID NO: 8 and a sense strand comprising SEQ ID NO: 16.
In certain embodiment, a combination of the application is used for decreasing viral replication, as measured by serum HBV DNA, in the subject with CHB, wherein the subject is not already NUC suppressed. In certain embodiment, a combination of the application is used for decreasing the expression of one or more HBV polypeptides in the subject with CHB, such as HBsAg in serum of the subject. In another embodiment, a combination of the application is used for bringing about an enhanced HBV-specific T cell response, which can be enhanced in a quantitative and/or qualitative manner, in the subject with CHB.
Methods according to embodiments of the application further comprises administering to the subject in need thereof another immunogenic agent (such as another innate immune modulator or a therapeutic vaccine) or another antiviral agent against HBV (such as a nucleoside analog or other direct antiviral compound) in combination with a composition of the application.
In some embodiments, the method further comprises administering to the subject a nucleoside analog. In some embodiments, the nucleoside analog is entecavir, tenofovir disoproxil fumarate, tenofovir alafenamide, lamivudine, telbivudine, or a combination thereof. In some embodiments, the nucleoside analog is entecavir and it is administered in the amount of about 0.01-5 mg, about 0.01-0.05 mg, about 0.05-0.1 mg, about 0.1-0.5 mg, about 0.5-1 mg, about 1-2 mg, about 2-3 mg, about 3-4 mg or about 4-5 mg. In some embodiments, the nucleoside analog is entecavir and it is administered in the amount of about 0.5 mg. In some embodiments, the nucleoside analog is tenofovir disoproxil fumarate and it is administered in the amount of about 100-500 mg, about 100-150 mg, about 150-200 mg, about 200-250 mg, about 250-300 mg, 300-400 mg, about 400-500 mg. In some embodiments, the nucleoside analog is tenofovir disoproxil fumarate and it is administered in the amount of about 300 mg In some embodiments, the nucleoside analog is tenofovir alafenamide and it is administered in the amount of about 5-100 mg, about 5-25 mg, about 25-50 mg, about 50-75 or about 75-100 mg. In some embodiments, the nucleoside analog is tenofovir alafenamide and it is administered in the amount of about 25 mg. In some embodiments, the nucleoside analog is lamivudine and it is administered in the amount of about 50-600 mg, about 100-500 mg, about 150-400 mg, about 200-350, or about 250-300 mg. In some embodiments, the nucleoside analog is lamivudine and it is administered in the amount of 150 mg or 300 mg. In some embodiments, the nucleoside analog is telbivudine and it is administered in the amount of about 300-600 mg, about 300-400 mg, about 400-500 mg, or about 500-600 mg. In some embodiments, the nucleoside analog is telbivudine and it is administered in the amount of 600 mg. In some embodiments, the patients have been exposed to the nucleoside analog prior to the combination therapy. In some embodiments, the patients have been administered the nucleoside analog for at least 1 month, at least 3 months, at least 6 months, or at least 1 year prior to receiving the combination therapy.
In some embodiments, the method further comprises administering to the subject one or more HBV-specific therapeutic vaccines selected from the group consisting of: a subunit vaccine comprising one or more HBV-derived peptide, polypeptide or protein optionally conjugated to a carrier molecule, wherein the subunit vaccine optionally further comprises one or more adjuvants and/or one or more delivery systems, which can also provide adjuvant activity, a recombinant viral vaccine comprising one or more viral vectors encoding the one or more HBV-derived peptide, polypeptide or protein, and optionally further encoding one or more cytokines to provide adjuvant activity, a nucleic acid based vaccine comprising one or more DNA molecules, such as DNA plasmids, encoding the one or more HBV-derived peptide, polypeptide or protein, and optionally further encoding the one or more cytokines to provide adjuvant activity, wherein the DNA molecules are administered by intramuscular injection using a delivery system, such as liposomes and nanoparticles, or are administered to the subject with an electroporation device, or are coated onto gold particles and administered into the dermis by a ballistic device. a nucleic acid based vaccine comprising one or more RNA molecules, such as mRNA or a selfamplifying mRNA or RNA replicon, encoding the one or more HBV-derived peptide, polypeptide or protein, and optionally further encoding the one or more cytokines to provide adjuvant activity, where the RNA molecules are administered by, for example, intramuscular delivery in a lipid nanoparticle delivery system.
In some embodiments, a combination of the application further comprises an HBV-derived peptide, polypeptide or protein comprising one or more, preferably all, of HBV core, pol and surface antigens, or a nucleic acid molecule encoding the HBV-derived peptide, polypeptide or protein. Preferably, the HBV surface antigens comprise one or more, preferably all, of small (S), medium (M) and large (L) envelope proteins. Examples of HB V-derived peptide, polypeptide or protein or its coding sequences useful for a method or combination of the present application include, but are not limited to those described in U.S. patent application publication no. US2019/0185828, the entire content of which is hereby incorporated by reference.
In some other embodiments, a combination of the application further comprises at least one other active ingredient, such as one or more from among antiviral agents, immunomodulatory agents, and Capsid Assembly Modulators (CAMs), e.g., in the form of small molecule(s), antibody(ies), polypeptide(s), protein(s) or nucleic acid(s), including, but not limited to, one or more from among immune checkpoint inhibitors (e.g., anti-PD-1, anti-TIM-3, etc.), other toll-like receptor agonists, RIG- I agonists, IL-15 superagonists (Aitor Bioscience), mutant IRF3 and IRF7 genetic adjuvants, STING agonists (Aduro), FLT3L genetic adjuvant, IL-12 genetic adjuvant, IL-7-hyFc; CAR-T which bind HBV env (S-CAR cells); CAM; cccDNA inhibitors, Interferon alpha receptor ligands.
In some other embodiments, a combination of the application further comprises one or more other HBV antiviral agents, such as, an HBV polymerase inhibitor (e.g., entecavir and tenofovir); Immunomodulators; Toll-like receptor 7 modulators; Toll-like receptor 8 modulators; Toll-like receptor 3 modulators; Hyaluronidase inhibitors; Modulators of IL-10; HBsAg inhibitors; Toll like receptor 9 modulators; Cyclophilin inhibitors; HBV Prophylactic vaccines; HBV Therapeutic vaccines; HBV viral entry inhibitors; antisense oligonucleotides targeting viral mRNA, more particularly anti-HB V antisense oligonucleotides; short interfering RNAs (siRNA), more particularly anti-HBV siRNA; endonuclease modulators; inhibitors of ribonucleotide reductase; HBV E antigen inhibitors; HBV antibodies targeting the surface antigens of the hepatitis B virus; HBV antibodies; CCR2 chemokine antagonists; Thymosin agonists; cytokines, such as IL-12; Capsid Assembly Modulators (CAM), nucleoprotein inhibitors (HBV core or capsid protein inhibitors); nucleic acid polymers (NAPs); stimulators of retinoic acid-inducible gene 1; stimulators of NOD2; HBV replication inhibitors; PI3K inhibitors; cccDNA inhibitors; immune checkpoint inhibitors, such as PD-L1 inhibitors, PD-1 inhibitors, TIM-3 inhibitors, TIGIT inhibitors, Lag3 inhibitors, and CTLA-4 inhibitors; Agonists of co-stimulatory receptors that are expressed on immune cells (more particularly T cells), such as CD27, CD28; BTK inhibitors; Other drugs for treating HBV; IDO inhibitors; Arginase inhibitors; and KDM5 inhibitors.
Method
In some embodiments, the RNAi component comprises: (i) a first RNAi agent comprising: an antisense strand comprising a nucleotide sequence of any one of the following: SEQ ID NO:1, SEQ ID NO:2, SEQ ID NOG, SEQ ID NO:4, SEQ ID NOG, SEQ ID NOG, and SEQ ID NOG, and a sense strand comprising a nucleotide sequence of any one of the following: SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO:12, SEQ ID NO:13, SEQ ID NO:14, and SEQ ID NO:15; and (ii) a second RNAi agent comprising: an antisense strand comprising a nucleotide sequence of any one of the following: SEQ ID NO:8 and SEQ ID NO:9, and a sense strand comprising a nucleotide sequence of any one of the following: SEQ ID NO:16, SEQ ID NO:17, SEQ ID NO:18, and SEQ ID NO:19.
In some embodiments, the first RNAi agent comprises SEQ ID NO: 5 and SEQ ID NO: 14. In some embodiments, the first RNAi agent comprises SEQ ID NO: 6 and SEQ ID NO: 14. In some embodiments, the first RNAi agent comprises SEQ ID NO: 7 and SEQ ID NO: 15. In some embodiments, the first RNAi agent comprises SEQ ID NO: 1 and SEQ ID NO: 10, 11 or 13. In some embodiments, the first RNAi agent comprises SEQ ID NO: 2 and SEQ ID NO: 10, 11 or 13. In some embodiments, the first RNAi agent comprises SEQ ID NO: 3 and SEQ ID NO: 10, 11, or 13. In some embodiments, the first RNAi agent comprises SEQ ID NO: 4 and SEQ ID NO: 12. In some embodiments, the second RNAi agent comprises SEQ ID NO: 9 and SEQ ID NO: 19. In some embodiments, the second RNAi agent comprises SEQ ID NO: 8 and SEQ ID NO: 16, 17 or 18.
In some embodiments, the RNAi component comprises a first RNAi agent comprising SEQ ID NO: 5 and SEQ ID NO: 14 and a second RNAi agent comprising SEQ ID NO: 9 and SEQ ID NO: 19. In some embodiments, the RNAi component comprises a first RNAi agent comprising SEQ ID NO: 6 and SEQ ID NO: 14 and a second RNAi agent comprising SEQ ID NO: 9 and SEQ ID NO: 19. In some embodiments, the RNAi component comprises a first RNAi agent comprising SEQ ID NO: 7 and SEQ ID NO: 15 and a second RNAi agent comprising SEQ ID NO: 9 and SEQ ID NO: 19.
In some embodiments, the RNAi component comprises a first RNAi agent comprising SEQ ID NO: 1 and SEQ ID NO: 10, 11 or 13 and a second RNAi agent comprising SEQ ID NO: 8 and SEQ ID NO: 16, 17 or 18. In some embodiments, the RNAi component comprises a first RNAi agent comprising SEQ ID NO: 2 and SEQ ID NO: 10, 11 or 13 and a second RNAi agent comprising SEQ ID NO: 8 and SEQ ID NO: 16, 17 or 18. In some embodiments, the RNAi component comprises a first RNAi agent comprising SEQ ID NO: 3 and SEQ ID NO: 10, 11 or 13 and a second RNAi agent comprising SEQ ID NO: 8 and SEQ ID NO: 16, 17 or 18. In some embodiments, the RNAi component comprises a first RNAi agent comprising SEQ ID NO: 4 and SEQ ID NO: 12and a second RNAi agent comprising SEQ ID NO: 8 and SEQ ID NO: 16, 17 or 18.
In some embodiments, the RNAi component comprises a first RNAi agent comprising SEQ ID NO: 2 and SEQ ID NO: 11 and the second RNAi agent comprising SEQ ID NO: 16 and SEQ ID NO: 8.
In some embodiments, the two HBV RNAi agents are administered in a ratio of about 1:1, 2:1, 3:1, 4:1 or 5:1. In some embodiments, the two HBV RNAi agents are administered in a ratio of about 2:1.
In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 25-75 mg per dose administration and in the ratio of about 2:1, about 3:1, about 1:1, about 4:1, about 5 : 1 or about 1:2. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 35-40 mg per dose administration and in the ratio of about 2:1, about 3:1, about 1:1, about 4:1, about 5:1 or about 1:2. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 50-125 mg per dose administration and in the ratio of about 2:1, about 3:1, about 1:1, about 4:1, about 5:1 or about 1:2. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 75-150 mg per dose administration and in the ratio of about 2:1, about 3:1, about 1:1, about 4:1, about 5:1 or about 1:2. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 100-200 mg per dose administration and in the ratio of about 2:1, about 3:1, about 1:1, about 4:1, about 5:1 or about 1:2. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 150-250 mg per dose administration and in the ratio of about 2:1, about 3:1, about 1:1, about 4:1, about 5:1 or about 1:2. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 200-300 mg per dose administration and in the ratio of about 2:1, about 3:1, about 1:1, about 4:1, about 5:1 or about 1:2. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 300-400 mg per dose administration and in the ratio of about 2:1, about 3:1, about 1:1, about 4:1, about 5:1 or about 1:2. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 50-100 mg per dose administration and in the ratio of about 2:1, about 3:1, about 1:1, about 4:1, about 5:1 or about 1:2. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 25-400 mg per dose administration and in the ratio of about 2:1. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 25-75 mg per dose administration and in the ratio of about 2:1. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 35-40 mg per dose administration and in the ratio of about 2:1. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 50-125 mg per dose administration and in the ratio of about 2:1. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 75-150 mg per dose administration and in the ratio of about 2:1. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 100-200 mg per dose administration and in the ratio of about 2: 1. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 125-225 mg per dose administration and in the ratio of about 2:1. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 150-250 mg per dose administration and in the ratio of about 2:1. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 200-300 mg per dose administration and in the ratio of about 2:1. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 300-400 mg per dose administration and in the ratio of about 2:1. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 100 mg per dose administration and in the ratio of about 2:1. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 25 mg per dose administration and in the ratio of about 2:1. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 35 mg per dose administration and in the ratio of about 2:1. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 40 mg per dose administration and in the ratio of about 2:1. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 50 mg per dose administration and in the ratio of about 2:1. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 75 mg per dose administration and in the ratio of about 2:1. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 200 mg per dose administration and in the ratio of about 2:1.
In some embodiments, the first RNAi agent is administered in an amount of about 3-650 mg per dose administration, and the second RNAi agent is administered in an amount of about 2-325 mg per dose administration. In some embodiments, the first RNAi agent is administered in an amount of about 15-150 mg per dose administration, and the second RNAi agent is administered in an amount of about 5-75 mg per dose administration. In some embodiments, the first RNAi agent is administered in an amount of about 35-265 mg per dose administration. In some embodiments, the first RNAi agent is administered in an amount of about 50-75 mg per dose administration. In some embodiments, the first RNAi agent is administered in an amount of about 15-75 mg per dose administration. In some embodiments, the second RNAi agent is administered in an amount of about 20-125 mg per dose administration. In some embodiments, the second RNAi agent is administered in an amount of about 25-50 mg per dose administration. In some embodiments, the second RNAi agent is administered in an amount of about 5-40 mg per dose administration. In some embodiments, the first RNAi agent is administered in an amount of about 17 mg per dose administration, and the second RNAi agent is administered in an amount of about 8 mg per dose administration. In some embodiments, the first RNAi agent is administered in an amount of about 23 mg per dose administration, and the second RNAi agent is administered in an amount of about 12 mg per dose administration. In some embodiments, the first RNAi agent is administered in an amount of about 27 mg per dose administration, and the second RNAi agent is administered in an amount of about 13 mg per dose administration. In some embodiments, the first RNAi agent is administered in an amount of about 33 mg per dose administration, and the second RNAi agent is administered in an amount of about 17 mg per dose administration. In some embodiments, the first RNAi agent is administered in an amount of about 67 mg per dose administration, and the second RNAi agent is administered in an amount of about 33 mg per dose administration.
In some embodiments, two RNAi agents are administered at a combined dose of 25-400 mg per dose administration. In an embodiment, two RNAi agents are administered at a combined dose of 25- 400 mg, and the first RNAi agent is administered with the second RNAi agent at a ratio of 1 : 1. In an embodiment, the dose of each of the first and second RNAi agents is in an amount of about 12 mg for a combined dose of about 25 mg. In an embodiment, the dose of each of the first and second RNAi agents is in an amount of about 17 mg for a combined dose of about 35 mg. In an embodiment, the dose of each of the first and second RNAi agents is in an amount of about 20 mg for a combined dose of about 40 mg. In an embodiment, the dose of each of the first and second RNAi agents is in an amount of about 25 mg for a combined dose of about 50 mg. In an embodiment, the dose of each of the first and second RNAi agents is in an amount of about 50 mg for a combined dose of about 100 mg. In an embodiment, the dose of each of the first and second RNAi agents is in an amount of about 100 mg for a combined dose of about 200 mg. In an embodiment, the dose of each of the first and second RNAi agents is in an amount of about 150 mg for a combined dose of about 300 mg. In an embodiment, the dose of each of the first and second RNAi agents is in an amount of about 200 mg for a combined dose of about 400 mg.
In an embodiment, two RNAi agents are administered at a combined dose of 25-400 mg per dose, and the first RNAi agent is administered with the second RNAi agent at a ratio of 2:1. In an embodiment, the dose of the first RNAi agent is in an amount of about 16 mg, and the dose of the second RNAi agent is in an amount of about 8 mg for a combined dose of about 25 mg. In an embodiment, the dose of the first RNAi agent is in an amount of about 24 mg, and the dose of the second RNAi agent is in an amount of about 12 mg for a combined dose of about 35 mg. In an embodiment, the dose of the first RNAi agent is in an amount of about 27 mg, and the dose of the second RNAi agent is in an amount of about 13 mg for a combined dose of about 40 mg. In an embodiment, the dose of the first RNAi agent is in an amount of about 33 mg, and the dose of the second RNAi agent is in an amount of about 17 mg for a combined dose of about 50 mg. In an embodiment, the dose of the first RNAi agent is in an amount of about 65 mg, and the dose of the second RNAi agent is in an amount of about 35 mg for a combined dose of about 100 mg. In an embodiment, the dose of the first RNAi agent is in an amount of about 133 mg, and the dose of the second RNAi agent is in an amount of about 67 mg for a combined dose of about 200 mg. In an embodiment, the dose of the first RNAi agent is in an amount of about 200 mg, and the dose of the second RNAi agent is in an amount of about 100 mg for a combined dose of about 300 mg. In an embodiment, the dose of the first RNAi agent is in an amount of about 270 mg, and the dose of the second RNAi agent is in an amount of about 135 mg for a combined dose of about 400 mg.
In an embodiment, two RNAi agents are administered at a combined dose of 25-400 mg per dose, the first RNAi agent is administered with the second RNAi agent at a ratio of 3:1. In an embodiment, the dose of the first RNAi agent is in an amount of about 18 mg, and the dose of the second RNAi agent is in an amount of about 6 mg for a combined dose of about 25 mg. In an embodiment, the dose of the first RNAi agent is in an amount of about 27 mg, and the dose of the second RNAi agent is in an amount of about 9 mg for a combined dose of about 35 mg. In an embodiment, the dose of the first RNAi agent is in an amount of about 30 mg, and the dose of the second RNAi agent is in an amount of about 10 mg for a combined dose of about 40 mg. In an embodiment, the dose of the first RNAi agent is in an amount of about 36 mg, and the dose of the second RNAi agent is in an amount of about 12 mg for a combined dose of about 50 mg. In an embodiment, the dose of the first RNAi agent is in an amount of about 75 mg, and the dose of the second RNAi agent is in an amount of about 25 mg for a combined dose of about 100 mg. In an embodiment, the dose of the first RNAi agent is in an amount of about 150 mg, and the dose of the second RNAi agent is in an amount of about 50 mg for a combined dose of about 200 mg. In an embodiment, the dose of the first RNAi agent is in an amount of about 225 mg, and the dose of the second RNAi agent is in an amount of about 75 mg for a combined dose of about 300 mg. In an embodiment, the dose of the first RNAi agent is in an amount of about 300 mg, and the dose of the second RNAi agent is in an amount of about 100 mg for a combined dose of about 400 mg.
In some embodiments, the first RNAi agent and the second RNAi agent are administered in a combined amount of about 25-400 mg per dose administration. In some embodiments, the first RNAi agent and the second RNAi agent are administered in a combined amount of about 25-50 mg, 50-75 mg, 75-100 mg, 100-125 mg, 125-150 mg, 150-175 mg, 175-200 mg, 200-225 mg, 225-250 mg, 250-275 mg, 275-300 mg, 300-325 mg, 325-350 mg, 350-375 mg, 375-400 mg, 25-75 mg, 50-100 mg, 100-150 mg, 150-200 mg, 200-250 mg, 250-300 mg, 300-350 mg, 350-400 mg, 25-100 mg, 50-150 mg, 100-200 mg, 150-250 mg, 200-300 mg, 300-400 mg, 25-200 mg, or 200-400 mg per dose administration. In some embodiments, the first RNAi agent to the second RNAi agent are administered in a combined amount of about 25 mg, about 50 mg, about 100 mg, about 125 mg, about 150 mg, about 175 mg, about 200 mg, about 225 mg, about 250 mg, about 275 mg, about 300 mg, about 325 mg, about 350 mg, about 375 mg, or about 400 mg per dose administration. In some embodiments, the first RNAi agent and the second RNAi agent are administered in a combined amount of about 50 mg, about 75 mg, about 100 mg, or about 125 mg per dose administration. In some embodiments, the first RNAi agent and the second RNAi agent are administered in a combined amount of about 25 mg, about 35 mg, about 40 mg, or about 200 mg per dose administration.
In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 1-10 mg/kg per dose administration. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 1-5 mg/kg per dose administration. In some embodiments, the two HBV RNAi agents are administered in a combined amount of about 1-1.5 mg/kg, about 1.5-2.0 mg/kg, about 2.0-2.5 mg/kg, about 2.5-3.0 mg/kg, about 3.0-3.5 mg/kg, about 3.5-4.0 mg/kg, about 4.0- 4.5 mg/kg, about 4.5-5.0 mg/kg, about 5.0-5.5 mg/kg, about 5.5-6.0 mg/kg, about 6.0-6.5 mg/kg, about 6.5-7.0 mg/kg, about 7.0-7.5 mg/kg, about 7.5-8.0 mg/kg, about 8.0-8.5 mg/kg, about 8.5-9.0 mg/kg, about 9.0-9.5 mg/kg, about 9.5-10 mg/kg, about 1-2.5 mg/kg, about 2.5-5.0 mg/kg, about 5.0-7.5 mg/kg, about 7.5-10 mg/kg, about 1-5.0 mg/kg, or about 5.0-10 mg/kg per dose administration.
In some embodiments, the first RNAi agent is administered in an amount of about 0.6-7 mg/kg per dose administration, and the second RNAi agent is administered in an amount of about 0.3-5 mg/kg per dose administration. In some embodiments, the second RNAi agent is administered in an amount of about 0.5-2.5 mg/kg per dose administration. In some embodiments, the second RNAi agent is administered in an amount of about 0.3-1.5 mg/kg per dose administration. In some embodiments, the first RNAi agent is administered in an amount of about 0.6-5 mg/kg per dose administration. In some embodiments, the first RNAi agent is administered in an amount of about 1-2.5 mg/kg per dose administration. In some embodiments, the two RNAi agents are administered in about 1-18 week intervals. In some embodiments, the two RNAi agents are administered in about 1-week intervals, about 2-week intervals, about 3-week intervals, about 4-week intervals, about 5-week intervals, about 6-week intervals, about 7-week intervals, about 8-week intervals, about 9-week intervals, about 10-week intervals, about 11 -week intervals, about 12-week intervals, about 13-week intervals, about 14-week intervals, about 15- week intervals, about 16-week intervals, about 17-week intervals, or about 18-week intervals. In some embodiments, the two RNAi agents are administered in about 1-6 month intervals. In some embodiments, the two RNAi agents are administered in about 1 -month intervals, about 2-month intervals, about 3-month intervals, about 4-month intervals, about 5-month intervals, or about 6-month intervals. In some embodiments, the two RNAi agents are administered in about 4-week intervals or 1- month intervals. In some embodiments, the two RNAi agents are administered once per month.
In some embodiments, the first RNAi agent and the second RNAi agent are administered for a duration of about 1-12 months. In some embodiments, the first RNAi agent and the second RNAi agent are administered for a duration of at least about 1 month, at least about 2 months, at least about 3 months, at least about 4 months, at least about 5 months, at least about 6 months, at least about 7 months, at least about 8 months, at least about 9 months, at least about 10 months, at least about 11 months or at least about 12 months. In some embodiments, the first RNAi agent and the second RNAi agent are administered for a duration of about 1-18 weeks. In some embodiments, the first RNAi agent and the second RNAi agent are administered for a duration of at least about 1 week, at least about 5 weeks, at least about 10 weeks, at least about 15 weeks, at least about 20 weeks, at least about 25 weeks, at least about 30 weeks, at least about 35 weeks, at least about 40 weeks, at least about 45 weeks, at least about 50 weeks, at least about 55 weeks, at least about 60 weeks, at least about 65 weeks, at least about 70 weeks, at least about 75 weeks, at least about 80 weeks, at least about 90 weeks, or at least 96 weeks.
In some embodiments, the first RNAi agent and the second RNAi agent are administered at a combined dose of 25-400 mg per dose administration. In an embodiment, the first RNAi agent and the second RNAi agent are administered at a combined dose of 25-400 mg, and the first RNAi agent is administered with the second RNAi agent at a ratio of 1 : 1. In an embodiment, the dose of the first RNAi agent is administered with the second RNAi agent is in an amount of about 12 mg for a combined dose of about 25 mg. In an embodiment, the dose of each of the first RNAi agent and the second RNAi agent is in an amount of about 17 mg for a combined dose of about 35 mg. In an embodiment, the dose of each of the first RNAi agent and the second RNAi agent is in an amount of about 20 mg for a combined dose of about 40 mg. In an embodiment, the dose of each of the first RNAi agent and the second RNAi agent is in an amount of about 25 mg for a combined dose of about 50 mg. In an embodiment, the dose of each of the first RNAi agent and the second RNAi agent is in an amount of about 50 mg for a combined dose of about 100 mg. In an embodiment, the dose of each of the first RNAi agent and the second RNAi agent is in an amount of about 100 mg for a combined dose of about 200 mg. In an embodiment, the dose of each of the first RNAi agent and the second RNAi agent is in an amount of about 150 mg for a combined dose of about 300 mg. In an embodiment, the dose of each of the first RNAi agent and the second RNAi agent is in an amount of about 200 mg for a combined dose of about 400 mg.
In an embodiment, the first RNAi agent and the second RNAi agent are administered at a combined dose of 25-400 mg per dose, and the second RNAi agent is administered with the first RNAi agent at a ratio of 1:2. In an embodiment, the dose of the first RNAi agent is in an amount of about 16 mg, and the dose of the second RNAi agent is in an amount of about 8 mg for a combined dose of about 25 mg. In an embodiment, the dose of the second RNAi agent is in an amount of about 12 mg, and the dose of the first RNAi agent is in an amount of about 24 mg for a combined dose of about 35 mg. In an embodiment, the dose of the first RNAi agent is in an amount of about 27 mg, and the dose of the second RNAi agent is in an amount of about 13 mg for a combined dose of about 40 mg. In an embodiment, the dose of the first RNAi agent is in an amount of about 33 mg, and the dose of the second RNAi agent is in an amount of about 17 mg for a combined dose of about 50 mg. In an embodiment, the dose of the second RNAi agent is in an amount of about 35 mg, and the dose of the first RNAi agent is in an amount of about 65 mg for a combined dose of about 100 mg. In an embodiment, the dose of the second RNAi agent is in an amount of about 67 mg, and the dose of the first RNAi agent is in an amount of about 133 mg for a combined dose of about 200 mg. In an embodiment, the dose of the second RNAi agent is in an amount of about 100 mg, and the dose of the first RNAi agent is in an amount of about 200 mg for a combined dose of about 300 mg. In an embodiment, the dose of the second RNAi agent is in an amount of about 135 mg, and the dose of the first RNAi agent is in an amount of about 270 mg for a combined dose of about 400 mg.
In an embodiment, the first RNAi agent and the second RNAi agent are administered at a combined dose of 25-400 mg per dose, the second RNAi agent is administered with the first RNAi agent at a ratio of 1:3. In an embodiment, the dose of the first RNAi agent is in an amount of about 18 mg, and the dose of the second RNAi agent is in an amount of about 6 mg for a combined dose of about 25 mg. In an embodiment, the dose of the second RNAi agent is in an amount of about 9 mg, and the dose of the first RNAi agent is in an amount of about 27 mg for a combined dose of about 35 mg. In an embodiment, the dose of the first RNAi agent is in an amount of about 30 mg, and the dose of the second RNAi agent is in an amount of about 10 mg for a combined dose of about 40 mg. In an embodiment, the dose of the first RNAi agent is in an amount of about 36 mg, and the dose of the second RNAi agent is in an amount of about 12 mg for a combined dose of about 50 mg. In an embodiment, the dose of the second RNAi agent is in an amount of about 25 mg, and the dose of the first RNAi agent is in an amount of about 75 mg for a combined dose of about 100 mg. In an embodiment, the dose of the second RNAi agent is in an amount of about 50 mg, and the dose of the first RNAi agent is in an amount of about 150 mg for a combined dose of about 200 mg. In an embodiment, the dose of the second RNAi agent is in an amount of about 75 mg, and the dose of the first RNAi agent is in an amount of about 225 mg for a combined dose of about 300 mg. In an embodiment, the dose of the second RNAi agent is in an amount of about 100 mg, and the dose of the first RNAi agent is in an amount of about 300 mg for a combined dose of about 400 mg.
In some embodiments, about 1 mg/kg (mpk) of the first RNAi agent and about 1 mg/kg of the second RNAi agent are administered to a subject in need thereof. In some embodiments, about 1.5 mg/kg of the first RNAi agent and about 1.5 mg/kg of the second RNAi agent are administered to a subject in need thereof. In some embodiments, about 2.0 mg/kg of the first RNAi agent and about 1.0 mg/kg of the second RNAi agent are administered to a subject in need thereof. In some embodiments, about 3.0 mg/kg of the first RNAi agent and about 1.0 mg/kg of the second RNAi agent are administered to a subject in need thereof. In some embodiments, about 3.2 mg/kg of the first RNAi agent and about 0.8 mg/kg of the second RNAi agent are administered to a subject in need thereof. In some embodiments, about 2.7 mg/kg of the first RNAi agent and about 1.3 mg/kg of the second RNAi agent are administered to a subject in need thereof. In some embodiments, about 4.0 mg/kg of the first RNAi agent and about 1.0 mg/kg of the second RNAi agent are administered to a subject in need thereof. In some embodiments, about 3.3 mg/kg of the first RNAi agent and about 1.7 mg/kg of the second RNAi agent are administered to a subject in need thereof. In some embodiments, between about 0.05 and about 5 mg/kg of the first RNAi agent and between about 0.05 and about 5 mg/kg of the second RNAi agent are administered to a subject in need thereof. In some embodiments, about the first RNAi agent and about the second RNAi agent are administered separately (e.g., in separate injections). In some embodiments, the respective dose of the first RNAi agent and the respective dose of the second RNAi agent are administered together (e.g., in the same injection). In some embodiments, the respective dose of the first RNAi agent and the respective dose of the second RNAi agent are prepared in a single pharmaceutical composition.
In some embodiments, the RNAi component is administered to the subject once monthly in a dose of about 40-350 mg, such as about 40-250 mg, more particularly 40-200 mg, more particularly 100 mg or 200 mg, more particularly 200 mg.
In some embodiments, the method further comprises administering a nucleoside analog. In some embodiments, the nucleoside analog is entecavir, tenofovir disoproxil fumarate, tenofovir alafenamide, lamivudine, telbivudine, or a combination thereof. In some embodiments, the nucleoside analog is entecavir and it is administered in a daily dose in the amount of about 0.01-5 mg, about 0.01- 0.05 mg, about 0.05-0.1 mg, about 0.1-0.5 mg, about 0.5-1 mg, about 1-2 mg, about 2-3 mg, about 3-4 mg or about 4-5 mg. In some embodiments, the nucleoside analog is entecavir and it is administered in a daily dose in the amount of about 0.5 mg. In some embodiments, the nucleoside analog is tenofovir disoproxil fumarate and it is administered in a daily dose in the amount of about 100-500 mg, about
100-150 mg, about 150-200 mg, about 200-250 mg, about 250-300 mg, 300-400 mg, about 400-500 mg. In some embodiments, the nucleoside analog is tenofovir disoproxil fumarate and it is administered in a daily dose in the amount of about 300 mg. In some embodiments, the nucleoside analog is tenofovir alafenamide and it is administered in a daily dose in the amount of about 5-100 mg, about 5-25 mg, about 25-50 mg, about 50-75 or about 75-100 mg. In some embodiments, the nucleoside analog is tenofovir alafenamide and it is administered in a daily dose in the amount of about 25 mg. In some embodiments, the nucleoside analog is lamivudine and it is administered in a daily dose in the amount of about 50-600 mg, about 50-300 mg, about 100-300 mg, about 100-500 mg, about 150-400 mg, about 200-350, or about 250-300 mg. In some embodiments, the nucleoside analog is lamivudine and it is administered in a daily dose in the amount of 100 mg, 150 mg, or 300 mg. In some embodiments, the nucleoside analog is telbivudine and it is administered in a daily dose in the amount of about 300-800 mg, about 400-700 mg, about 300-600 mg, about 300-400 mg, about 400-500 mg, or about 500-600 mg. In some embodiments, the nucleoside analog is telbivudine and it is administered in a daily dose in the amount of 600 mg. In some embodiments, the patients have been exposed to the nucleoside analog prior to the combination therapy. In some embodiments, the patients have been administered the nucleoside analog for at least 1 month, at least 3 months, at least 6 months, or at least 1 year prior to receiving the combination therapy.
In embodiments, the method further comprises administering a nucleic acid polymer (NAP). The NAP can, for example, be selected from REP2139 or REP2165. REP2139 has a sequence of (A,5’MeC)2o with each linkage being phosphorothioated and every ribose being 2’0 methylated (which is disclosed as SEQ ID NO: 10 in WO2016/04525, the content of which is incorporated herein by reference in its entirety). REP2165 has a sequence of (A,5’MeC)2o with each linkage being phosphorothioated, every ribose being 2’0 methylated except adenosines at positions 11, 21, and 31, where riboses are 2’OH (which is disclosed as SEQ ID NO:13 in WO2016/04525).
The NAP can also be other exemplary nucleic acid polymers, which include, but are not limited to, REP2006, REP2031, REP2055, STOPS™ (S-antigen transport-inhibiting oligonucleotide polymers), and those disclosed in Patent Application Publication Nos. WO200424919; WO201221985; and WO202097342 and U.S. Patent Nos. 7,358,068; 8,008,269; 8,008,270; and 8,067,385, the content of each is incorporated herein by reference in its entirety.
In some embodiments, the patients are screened for HBeAg status prior to administration of the combination therapy. In some embodiments, the patients are HBeAg positive. In some embodiments, the patients are HBeAg negative. In some embodiment, the patients are screened for immune tolerance prior to administration of the combination therapy.
In some embodiments, the HBsAg level in the patient is reduced by at least about logw 0.5, about logw0.75, about logw 1, about logw 1-25, about logw 1-5, about logw 1.75, about logw2 or about logw 2.5 from base line on Day 1. In some embodiments, the HBeAg level in the patient is reduced by at least about logw 0.5, about logw0.75, about logw 1, about logw 1.25, about logw 1.5, about logw 1.75, about logio 2 or about logic 2.5 from base line on Day 1. In some embodiments, the HBcrAg level in the patient is reduced by at least about logic 0.5, about logio0.75, about logic 1, about logic 1.25, about logw 1-5, about log 1.75, about logic 2 or about logic 2.5 from base line on Day 1. In some embodiments, the HBV DNA level in the patient is reduced by at least about logw 0.5, about logw 1, about logw 1.5, about logw 2, about logw3, about logw 4, about logw5 or about logw 7.5 from base line on Day 1. In some embodiments, the HBV RNA level in the patient is reduced by at least about logw 0.5, about logw 0.75, about logw 1, about logw 1.25, about logw 1.5, about logw 1.75, about logw2 or about logw 2.5 from base line on Day 1.
In embodiments of the methods of treating HBV infection provided herein, the patient, individual or subject in need thereof is a chronically HBV-infected patient, with or without evidence of underlying liver inflammation. In some embodiments, the patient has a chronic HBV infection. In other embodiments, the patient is suffering from an HBV-induced disease. In some embodiments, the HBV- induced disease is cirrhosis, liver failure or hepatocellular carcinoma. In other embodiments, the patient is a treatment-naive patient. More in particular, the patient is a chronically HBV-infected treatment- naive patient. In a further embodiment, the patient is HBeAg-positive. In still a further embodiment, the patient is treatment-naive and HBeAg-positive.
In an embodiment, the methods further comprise administering to the subject at least one additional therapeutic agent selected from a nucleoside analog, in particular, tenofovir, or a pharmaceutically acceptable salt or prodrug thereof, or entecavir, or a pharmaceutically acceptable salt or solvate thereof. In an embodiment of the method, the nucleoside analog is selected from the group consisting of entecavir monohydrate, tenofovir disoproxil fumarate and tenofovir alafenamide. In an embodiment of the method, the nucleoside analog is entecavir monohydrate. In an embodiment of the method, the nucleoside analog is tenofovir disoproxil fumarate. In a further embodiment of the method, the nucleoside analog is tenofovir alafenamide.
In an embodiment of the method, the tenofovir disoproxil fumarate is administered in an amount of 60-600 mg. In another embodiment of the method, the tenofovir disoproxil fumarate is administered in an amount of 300 mg. In yet another embodiment of the method, the entecavir monohydrate is administered in an amount of 0.1-1 mg. In still another embodiment of the method, the entecavir monohydrate is administered in an amount of 0.5 mg. In another embodiment of the method, the tenofovir alafenamide is administered in an amount of 25 mg.
In an embodiment, the methods further comprise administering to the subject at least one additional therapeutic agent selected from the group consisting of HBV combination drugs, HBV vaccines, HBV DNA polymerase inhibitors, immunomodulators, toll-like receptor (TLR) modulators, interferon alpha receptor ligands, hyaluronidase inhibitors, hepatitis b surface antigen (HBsAg) inhibitors, cytotoxic T-lymphocyte-associated protein 4 (ipi4) inhibitors, cyclophilin inhibitors, HBV viral entry inhibitors, antisense oligonucleotide targeting viral mRNA, short interfering RNAs (siRNA) and ddRNAi endonuclease modulators, ribonucleotide reductase inhibitors, HBV E antigen inhibitors, covalently closed circular DNA (cccDNA) inhibitors, famesoid X receptor agonists, HBV antibodies, CCR2 chemokine antagonists, thymosin agonists, cytokines, nucleoprotein modulators, retinoic acidinducible gene 1 simulators, N0D2 stimulators, phosphatidylinositol 3-kinase (PI3K) inhibitors, indoleamine-2, 3-dioxygenase (IDO) pathway inhibitors, PD-1 inhibitors, PD-L1 inhibitors, recombinant thymosin alpha-1, bruton’s tyrosine kinase (BTK) inhibitors, KDM inhibitors, HBV replication inhibitors, arginase inhibitors, and other HBV drugs.
EXAMPLES
ABBREVIATIONS
Figure imgf000089_0001
EXAMPLE 1 siRNA compounds in Table 5 comprising hepatitis B virus (HBV)- specific, liver-targeted N- galactosamine-conjugated small interfering RNA (siRNA) triggers, the first siRNA agent A and the second siRNA agent B (e.g., the first and the second RNAi agents are each independently conjugated to (NAG37)s or (NAG25)s, the first RNAi agent comprises an antisense strand comprising SEQ ID NO: 2 and a sense strand comprising SEQ ID NO: 11 the second RNAi agent comprises an antisense strand comprising SEQ ID NO: 8 and a sense strand comprising SEQ ID NO: 16; or the first RNAi agent comprises an antisense strand comprising SEQ ID NO: 1 and a sense strand comprising SEQ ID NO: 11 the second RNAi agent comprises an antisense strand comprising SEQ ID NO: 8 and a sense strand comprising SEQ ID NO: 17; or the first RNAi agent comprises an antisense strand comprising SEQ ID NO: 2 and a sense strand comprising SEQ ID NO: 10 the second RNAi agent comprises an antisense strand comprising SEQ ID NO: 8 and a sense strand comprising SEQ ID NO: 18), which silence HBV RNA transcripts from host-integrated HBV DNA and episomal DNA. The siRNA administered 100- 400 mg subcutaneously (SC) every 28 days, with a nucleos(t)ide analog daily, demonstrated HBV surface antigen decline >1 logw lU/mL after 3 doses in chronic HBV patients, and this effect was sustained for up to 48 weeks off treatment.
EXAMPLE 2
Pharmacokinetics (PK), safety and tolerability of an RNAi component comprising a first RNAi agent comprising an antisense strand comprising SEQ ID NO: 2 and a sense strand comprising SEQ ID NO: 11 and a second RNAi agent comprising an antisense strand comprising SEQ ID NO: 8 and a sense strand comprising SEQ ID NO: 16; in participants with moderate hepatic impairment (Child-Pugh B) were evaluated and compared to healthy participants with normal liver function. .Eight (8) participants with Child-Pugh B and 8 healthy participants matching for sex, age (±10 years), and body weight (+10 kg) were enrolled. Participants received a single 200 mg SC dose of siRNA Compound (with the first siRNA and the second siRNA present in a molar ratio of 2:1) in the abdomen on Day 1 of the study, following an overnight fast of >10 hours. Plasma and urine concentrations of the first and second siRNA agents were collected over 72 hours and analyzed using liquid chromatography coupled to a fluorescence detector. Plasma samples were collected pre-dose and at 0.25, 0.5, 1, 2, 3, 4, 5, 6, 8, 10, 12, 24, 36, 48, and 72 hours post-dose. Urine concentrations were measured at 0-6, 6-12, 12-24, 24-36, 36-48, and 48-72 hours post-dose. PK parameters were estimated using non-compartmental analysis (WinNonlin, Certara, Princeton NJ). Safety and tolerability were assessed throughout. Plasma protein binding was determined with ultrafiltration in pre-dosed plasma samples spiked with 5,000 ng/mL RNAi component and plasma samples collected 6 hours after dosing of 200 mg RNAi component.
PK parameters are summarized in Table 7 below and depicted graphically in FIG. 1 (first siRNA agent) and FIG. 2 (second siRNA agent). Cmax and AUC of the first siRNA agent were 1.4- and 1.3-fold higher, respectively, with Child-Pugh B versus case controls; for the second siRNA agent, Cmax and AUC were 2.2- and 2.0-fold higher, respectively. Half-life and amount of drug excreted were slightly higher for both analytes with Child-Pugh Class B participants.
Table 7: PK Parameters
Figure imgf000090_0001
Figure imgf000091_0001
Cmax, maximum plasma concentration; tmax, time to reach Cmax; AUC, area under the plasma concentration-time curve from time 0 to the time of the last measured concentration (AUCiast) or extrapolated to infinity (AUC«>); t^, half-life; CL/F, apparent clearance; Vd/F, apparent volume of distribution, AE, amount of drug excreted.; CI, confidence interval. All data expressed as mean (standard deviation) except for tmax median (range).
Plasma protein binding of the first and second RNAi agents were measured following a single SC dose of the RNAi component. In pre-dose samples of healthy participants, the mean (standard deviation) unbound fraction in plasma was 0.086 (0.024) for the first RNAi agent and 0.115 (0.025) for the second RNAi agent. In plasma samples 6 hours after dosing, the mean (standard deviation) unbound fraction in plasma was 0.142 (0.070) for the first RNAi agent and 0.090 (0.061) for the second RNAi agent.
In comparison, in pre-dose samples of Child-Pugh Class B participants, the mean (SD) unbound fraction was 0.110 (0.039) for the first RNAi agent and 0.126 (0.036) for the second RNAi agent. In plasma samples 6 hours after dosing, the mean (standard deviation) unbound fraction in plasma was 0.091 (0.037) for the first RNAi agent and 0.132 for the second RNAi agent.
There were no deaths, serious adverse events (AEs), or discontinuations. Overall, 2 participants, 1 in each group, reported at least 1 treatment-emergent AE (TEAE). TEAEs were mild or moderate and not related to RNAi agent. All TE graded laboratory abnormalities were mild or moderate, except for 3 with Child-Pugh B who experienced transient: Grade 3 decreases in lymphocytes levels (n=l), and in platelets (n=2). There were no relevant cardiovascular findings.
Therefore, a single 200 mg dose of siRNA Compound was safe and well tolerated in participants without and with moderate hepatic impairment. Higher exposures of the siRNA agents were seen in participants with moderate hepatic impairment, but this increase was not considered to be a clinically relevant safety concern. The fraction of siRNA unbound in plasma was not significantly different between healthy participants and Child-Pugh Class B participants.
Child-Pugh Group, Child-Pugh Class, and the like: a ranking of level of hepatic impairment based on the Child-Pugh Score. Child-Pugh Scores of 5-6 are classified as Child-Pugh Class A (mild hepatic impairment) and have an expected 2 year survival rate of 85%. Child-Pugh Scores of 7-9 are classified as Child-Pugh Class B (moderate hepatic impairment) and have an expected 2 year survival rate of 57%. Child-Pugh Scores of 10-15 are classified as Child-Pugh Class C (severe hepatic impairment) and have an expected 2 year survival rate of 35%.
Child-Pugh Score: a score based on five clinical measures of hepatic impairment, including levels of total bilirubin, serum albumin, PT INR, ascites, and hepatic encephalopathy. Each measure is given a ranking of 1, 2, or 3, and the sum of the five rankings is the Child-Pugh Score. The Child-Pugh Score can be used to classify hepatic impairment by placing subjects in a Child-Pugh Group

Claims

CLAIMS WHAT IS CLAIMED IS
1. A method of treating hepatitis infection, said method comprising administering to a subject in need thereof an effective amount of a pharmaceutical composition comprising an RNAi component having:
(i) a first RNAi agent comprising: an antisense strand comprising a nucleotide sequence of any one of the following: SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, and SEQ ID NO:7 and a sense strand comprising a nucleotide sequence of any one of the following: SEQ ID NO:10, SEQ ID NO:11, SEQ ID NO:12, SEQ ID NO:13, SEQ ID NO:14, and SEQ ID NO:15; and
(ii) a second RNAi agent comprising: an antisense strand comprising a nucleotide sequence of any one of the following: SEQ ID NO: 8 and SEQ ID NO:9, and a sense strand comprising a nucleotide sequence of any one of the following: SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, and SEQ ID NO: 19; wherein the subject has previously been determined to have a level of hepatic impairment.
2. A method of treating hepatitis infection, said method comprising administering to a subject in need thereof an effective amount of a pharmaceutical composition comprising an RNAi component having:
(i ) the first RNAi agent comprising: an antisense strand comprising a nucleotide sequence of any one of the following: SEQ ID NO:5, SEQ ID NO:6, and SEQ ID NO:7 and a sense strand comprising a nucleotide sequence of any one of the following: SEQ ID NO: 14, and SEQ ID NO: 15; and
(ii) a second RNAi agent comprising: an antisense strand comprising a nucleotide sequence of SEQ ID NO:9, and a sense strand comprising a nucleotide sequence of SEQ ID NO:19; wherein at least 90% of the nucleotides in the first and the second RNAi agents are modified nucleotides.
3. A method of treating hepatitis infection, said method comprising administering to a subject in need thereof an effective amount of a pharmaceutical composition comprising an RNAi component having:
(i ) the first RNAi agent comprising: an antisense strand comprising a nucleotide sequence of any one of the following: SEQ ID NO:5, SEQ ID NO:6, and SEQ ID NO:7 and a sense strand comprising a nucleotide sequence of any one of the following: SEQ ID NO: 14, and SEQ ID NO: 15; and
(ii) a second RNAi agent comprising: an antisense strand comprising a nucleotide sequence of SEQ ID NO:9, and a sense strand comprising a nucleotide sequence of SEQ ID NO:19; wherein the first or the second RNAi agent comprises at least one modified nucleotide and/or at least one modified intemucleoside linkage.
4. The method according to any one of claims 1 to 3, wherein the subject is affected with hepatic impairment.
5. The method according to any one of claims 1 to 4, wherein the subject is affected with chronic hepatitis B viral infection.
6. The method according to any one of claims 1 to 5, wherein the subject is a treatment naive patient.
7. The method according to claim 6, wherein the subject is a treatment naive HBeAg+ patient.
8. The method according to any one of claims 1 to 7, wherein the subject is a treatment- experienced patient.
9. The method according to any one of the proceeding claims, wherein the hepatitis infection is a hepatitis B virus infection with hepatitis D virus co-infection.
10. The method according to any one of the proceeding claims, wherein the hepatitis infection is a hepatitis B virus infection without hepatitis D virus co-infection.
11. The method according to any one of the proceeding claims, further comprising administering a nucleoside or nucleotide analog.
12. The method according to claim 11, wherein the nucleoside or nucleotide analog is selected from the group consisting of tenofovir, or a pharmaceutically acceptable salt or prodrug thereof, and entecavir, or a pharmaceutically acceptable salt thereof.
13. The method according to claim 12, wherein the nucleoside or nucleotide analog is tenofovir or a produg thereof, in particular, tenofovir alafenamide, or tenofovir disoproxil fumarate.
14. The method according to any one of claims 1 to 13, further comprising administering at least one additional therapeutic agent selected from the group consisting of HBV combination drugs, HBV vaccines, HBV DNA polymerase inhibitors, immunomodulators, toll-like receptor (TLR) modulators, interferon alpha receptor ligands, hyaluronidase inhibitors, hepatitis b surface antigen (HBsAg) inhibitors, cytotoxic T-lymphocyte-associated protein 4 (ipi4) inhibitors, cyclophilin inhibitors, HBV viral entry inhibitors, antisense oligonucleotide targeting viral mRNA, short interfering RNAs (siRNA) and ddRNAi endonuclease modulators, ribonucleotide reductase inhibitors, HBV E antigen inhibitors, covalently closed circular DNA (cccDNA) inhibitors, famesoid X receptor agonists, HBV antibodies, CCR2 chemokine antagonists, thymosin agonists, cytokines, nucleoprotein modulators, retinoic acid-inducible gene 1 simulators, NOD2 stimulators, phosphatidylinositol 3-kinase (PI3K) inhibitors, indoleamine-2, 3-dioxygenase (IDO) pathway inhibitors, PD-1 inhibitors, PD-L1 inhibitors, recombinant thymosin alpha-1, bruton’s tyrosine kinase (BTK) inhibitors, KDM inhibitors, HBV replication inhibitors, arginase inhibitors, and other HBV drugs.
15. The method according to any of the proceeding claims, further comprising administering an effective amount of a second agent.
16. The method according to any of the proceeding claims, wherein the subject has a Child- Pugh class A hepatic impairment.
17. The method according to any one of claims 1 tol6, wherein the subject has a Child-Pugh class B hepatic impairment.
18. The method according to any one of claims 1 to 16, wherein the subject has a Child-Pugh class C hepatic impairment.
19. The method according to any one of claims 1 to 16, wherein the subject has a Child-Pugh score of below 5.
20. The method according to any one of claims 1 to 16, wherein the subject has a Child-Pugh score of 5 or 6.
21. The method according to any one of claims 1 to 16, wherein the subject the subject has a Child-Pugh score of 7, 8, or 9.
22. The method of any one of claims 1 to 21, wherein the first or the second RNAi agent further comprises a targeting ligand that is conjugated to the first or the second RNAi agent.
23. The method of claim 22, wherein the targeting ligand comprises N-acetyl- galactosamine.
24. The method of claim 23, wherein the targeting ligand is selected from the group consisting of (NAG13), (NAG13)s, (NAGI 8), (NAG18)s, (NAG24), (NAG24)s, (NAG25), (NAG25)s, (NAG26), (NAG26)s, (NAG27), (NAG27)s, (NAG28), (NAG28)s, (NAG29), (NAG29)s, (NAG30), (NAG30)s, (NAG31), (NAG31)s, (NAG32), (NAG32)s, (NAG33), (NAG33)s, (NAG34), (NAG34)s, (NAG35), (NAG35)s, (NAG36), (NAG36)s, (NAG37), (NAG37)s, (NAG38), (NAG38)s, (NAG39), and (NAG39)s.
25. The method of claim 24, wherein the targeting ligand is (NAG25), (NAG25)s, (NAG31), (NAG31)s, (NAG37), or (NAG37)s.
26. The method of any one of claims 22 to 25, wherein the targeting ligand is conjugated to the sense strand of the first or the second RNAi agent.
27. The method of claim 26, wherein the targeting ligand is conjugated to the 5’ terminus of the sense stand of the first or the second RNAi agent.
28. The method of any one of claims 1 to 27, wherein the first and the second RNAi agents independently comprise a duplex selected from the group consisting of:
(a) an antisense strand comprising SEQ ID NO: 1 and a sense strand comprising SEQ ID NO: 10;
(b) an antisense strand comprising SEQ ID NO: 2 and a sense strand comprising SEQ ID NO: 11;
(c) an antisense strand comprising SEQ ID NO: 3 and a sense strand comprising SEQ ID NO: 11;
(d) an antisense strand comprising SEQ ID NO: 4 and a sense strand comprising SEQ ID NO: 12;
(e) an antisense strand comprising SEQ ID NO: 8 and a sense strand comprising SEQ ID NO: 16;
(f) an antisense strand comprising SEQ ID NO: 8 and a sense strand comprising SEQ ID NO: 17;
(g) an antisense strand comprising SEQ ID NO: 2 and a sense strand comprising SEQ ID NO: 12; and
(h) an antisense strand comprising SEQ ID NO: 8 and a sense strand comprising SEQ ID NO: 18.
29. The method of any one of claims 1 to 28, wherein the first and the second RNAi agents are each independently conjugated to a targeting ligand comprising N-acetyl-galactosamine, and the first and the second RNAi agents independently comprise a duplex selected from the group consisting of:
(a) an antisense strand comprising SEQ ID NO: 2 and a sense strand comprising SEQ ID NO: 11;
(b) an antisense strand comprising SEQ ID NO: 4 and a sense strand comprising SEQ
ID NO: 12; (c) an antisense strand comprising SEQ ID NO: 8 and a sense strand comprising SEQ ID NO: 16;
(d) an antisense strand comprising SEQ ID NO: 2 and a sense strand comprising SEQ ID NO: 13; and
(e) an antisense strand comprising SEQ ID NO: 8 and a sense strand comprising SEQ ID NO: 18.
30. The method of any one of claims 1-29, wherein the molar ratio of the first RNAi agent to the second RNAi agent by weight is in the range of about 1:2 to about 5:1.
31. The method of any one of claims 1 to 30, wherein the RNAi component is administered to the subject in a dose of about 40-1000 mg, more particularly about 40-250 mg, more particularly 40- 200 mg, more particularly 100 mg or 200 mg, more particularly 200 mg.
32. The method of any one of claims 1 to 31 , wherein the RNAi component is administered to the subject once monthly (i.e., Q4W) or at longer time intervals, such as once every 8 weeks (Q8W) or once every 12 weeks (Q12W).
33. The method of any one of claims 1 to 32, wherein the RNAi component is administered to the subject for a time period of 1 to 12 months, more particularly for 12 to 48 weeks.
34. The method of any one of claims 1 to 33, wherein the RNAi component is administered via subcutaneous injection.
35. The method of any one of claims 1 to 34, wherein the second agent is administered to the subject once a day, every other day, twice a week or weekly, more particularly weekly.
36. The method of any one of claims 1 to 35, wherein the second agent is administered to the subject for a time period of 1 to 12 months, more particularly for 6 to 48 weeks, more particularly for 12 to 24 weeks.
37. The method of any one of claims 1 to 36, wherein the RNAi component is administered to the subject via intravenous or subcutaneous injection.
38. The method of any one of claims 1 to 37, wherein the second agent is administered orally.
39. The method of any claim 38, wherein the RNAi component is administered simultaneously or sequentially with the second agent.
40. The method of claim 38, wherein the RNAi component is administered separately from the second agent.
41. The method of claim 36, wherein the second agent is administered to the subject about six (6) months after the administration of the RNAi component has started.
42. The method of claim 36 or claim 37, wherein second agent is administered to the subject about six (6) months after the administration of the RNAi component has started, and wherein the second agent is administered to the subject for about six (6) months in total.
43. The method of any one of the proceeding claims, further comprising administering to the subject a nucleoside analog or a nucleotide analog.
44. The method of claim 43, wherein the nucleoside analog is entecavir, tenofovir disoproxil fumarate, tenofovir alafenamide, lamivudine, telbivudine, or a combination thereof.
45. The method of claim 44, wherein entecavir is administered to the subject in a daily dose of about 0.1-5 mg.
46. The method of claim 44, wherein tenofovir is administered to the subject in a daily dose of about 5-50 mg of tenofovir alafenamide or about 200-500 mg of tenofovir disoproxil fumarate.
47. The method of claim 44, wherein lamivudine is administered to the subject in a daily dose of about 100 mg, about 150 mg or about 300 mg.
48. The method of claim 44, wherein telbivudine is administered to the subject in a daily dose of about 600 mg.
49. The method of any one of claims 1 to 48, wherein the effective amount the pharmaceutical composition comprising the RNAi component and the effective amount of the pharmaceutical composition comprising the second agent is administered to the subject for 10-96 weeks, more particularly 12-72 weeks, more particularly 12-60 weeks, more particularly 12-52 weeks, more particularly 48 weeks.
50. The method of claim 49, further comprising administration of a nucleoside analog or a nucleotide analog, such as entecavir, tenofovir disoproxil fumarate, tenofovir alafenamide, lamivudine, telbivudine, or a combination thereof to the subject, and wherein the administration of the nucleoside or nucleotide analog is optionally being continued once the administration of the effective amount the pharmaceutical composition comprising the RNAi component.
51. An RNAi component for use in the treatment of a Hepatitis B Virus infection, more particularly a chronic HBV infection (CHB) with or without a viral co-infection, and/or in the treatment of a chronic Hepatitis D Virus (HDV) infection, wherein the RNAi component is as defined in any one of claims 1-50.
PCT/EP2022/050776 2021-01-15 2022-01-14 Use of oligonucleotides for individuals with hepatic impairment WO2022152869A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202163138322P 2021-01-15 2021-01-15
US63/138,322 2021-01-15
US202163194148P 2021-05-27 2021-05-27
US63/194,148 2021-05-27

Publications (1)

Publication Number Publication Date
WO2022152869A1 true WO2022152869A1 (en) 2022-07-21

Family

ID=80119695

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2022/050776 WO2022152869A1 (en) 2021-01-15 2022-01-14 Use of oligonucleotides for individuals with hepatic impairment

Country Status (1)

Country Link
WO (1) WO2022152869A1 (en)

Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4522811A (en) 1982-07-08 1985-06-11 Syntex (U.S.A.) Inc. Serial injection of muramyldipeptides and liposomes enhances the anti-infective activity of muramyldipeptides
WO2000053722A2 (en) 1999-03-10 2000-09-14 Phogen Limited Delivery of nucleic acids and proteins to cells
WO2004024919A1 (en) 2002-09-13 2004-03-25 Replicor, Inc. Non-sequence complementary antiviral oligonucleotides
WO2008022309A2 (en) 2006-08-18 2008-02-21 F. Hoffmann-La Roche Ag Polyconjugates for in vivo delivery of polynucleotides
WO2011104169A1 (en) 2010-02-24 2011-09-01 F. Hoffmann-La Roche Ag Compositions for targeted delivery of sirna
WO2012021985A1 (en) 2010-08-20 2012-02-23 Replicor Inc. Oligonucleotide chelate complexes
WO2012083185A2 (en) 2010-12-17 2012-06-21 Arrowhead Research Corporations Peptide-based in vivo sirna delivery system
WO2016004525A1 (en) 2014-07-10 2016-01-14 Replicor Inc. Methods for the treatment of hepatitis b and hepatitis d virus infections
WO2018027106A2 (en) * 2016-08-04 2018-02-08 Arrowhead Pharmaceuticals, Inc. RNAi AGENTS FOR HEPATITIS B VIRUS INFECTION
US20190185828A1 (en) 2017-12-19 2019-06-20 Janssen Sciences Ireland Unlimited Company Hepatitis b virus (hbv) vaccines and uses thereof
WO2020097342A1 (en) 2018-11-08 2020-05-14 Aligos Therapeutics, Inc. S-antigen transport inhibiting oligonucleotide polymers and methods
WO2020163747A1 (en) * 2019-02-07 2020-08-13 Arrowhead Pharmaceuticals, Inc. Rnai agents for hepatitis b virus infection
WO2020214976A1 (en) * 2019-04-18 2020-10-22 Janssen Pharmaceuticals, Inc. Combination therapy for treating hepatitis b virus infection

Patent Citations (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4522811A (en) 1982-07-08 1985-06-11 Syntex (U.S.A.) Inc. Serial injection of muramyldipeptides and liposomes enhances the anti-infective activity of muramyldipeptides
WO2000053722A2 (en) 1999-03-10 2000-09-14 Phogen Limited Delivery of nucleic acids and proteins to cells
US8067385B2 (en) 2002-09-13 2011-11-29 Replicor, Inc. Antiviral oligonucleotides targeting HBV
US7358068B2 (en) 2002-09-13 2008-04-15 Replicor, Inc. Antiviral oligonucleotides
US8008270B2 (en) 2002-09-13 2011-08-30 Replicor Inc. Antiviral oligonucleotides targeting viral families
US8008269B2 (en) 2002-09-13 2011-08-30 Replicor Inc. Antiviral oligonucleotides
WO2004024919A1 (en) 2002-09-13 2004-03-25 Replicor, Inc. Non-sequence complementary antiviral oligonucleotides
WO2008022309A2 (en) 2006-08-18 2008-02-21 F. Hoffmann-La Roche Ag Polyconjugates for in vivo delivery of polynucleotides
WO2011104169A1 (en) 2010-02-24 2011-09-01 F. Hoffmann-La Roche Ag Compositions for targeted delivery of sirna
WO2012021985A1 (en) 2010-08-20 2012-02-23 Replicor Inc. Oligonucleotide chelate complexes
WO2012083185A2 (en) 2010-12-17 2012-06-21 Arrowhead Research Corporations Peptide-based in vivo sirna delivery system
WO2016004525A1 (en) 2014-07-10 2016-01-14 Replicor Inc. Methods for the treatment of hepatitis b and hepatitis d virus infections
WO2018027106A2 (en) * 2016-08-04 2018-02-08 Arrowhead Pharmaceuticals, Inc. RNAi AGENTS FOR HEPATITIS B VIRUS INFECTION
US20190185828A1 (en) 2017-12-19 2019-06-20 Janssen Sciences Ireland Unlimited Company Hepatitis b virus (hbv) vaccines and uses thereof
WO2020097342A1 (en) 2018-11-08 2020-05-14 Aligos Therapeutics, Inc. S-antigen transport inhibiting oligonucleotide polymers and methods
WO2020163747A1 (en) * 2019-02-07 2020-08-13 Arrowhead Pharmaceuticals, Inc. Rnai agents for hepatitis b virus infection
WO2020214976A1 (en) * 2019-04-18 2020-10-22 Janssen Pharmaceuticals, Inc. Combination therapy for treating hepatitis b virus infection

Non-Patent Citations (10)

* Cited by examiner, † Cited by third party
Title
"Remington's Pharmaceutical Sciences", 1985, MACK PUBLISHING COMPANY, pages: 1418
ANONYMOUS: "Arrowhead presents Clinical Data on JNJ-3989 (ARO-HBV) at The International Liver Congress?", 12 April 2019 (2019-04-12), XP055691678, Retrieved from the Internet <URL:http://ir.arrowheadpharma.com/node/14511/pdf> [retrieved on 20200505] *
ANONYMOUS: "History of Changes for Study: NCT03365947 Study of ARO-HBV in Normal Adult Volunteers and Patients With Hepatitis B Virus (HBV)", CLINICALTRIALS.GOV ARCHIVE, 25 September 2020 (2020-09-25), XP055906727, Retrieved from the Internet <URL:https://clinicaltrials.gov/ct2/history/NCT03365947?A=11&B=11&C=merged#StudyPageTop> [retrieved on 20220329] *
ANONYMOUS: "To (Hep)B or not to (Hep)B? - Oligonucleotide Therapeutics Society", OLIGONUCLEOTIDE THERAPEUTICS SOCIETY, 23 May 2018 (2018-05-23), XP055691740, Retrieved from the Internet <URL:https://www.oligotherapeutics.org/to-hepb-or-not-to-hepb/> [retrieved on 20200505] *
C. WOODDELL ET AL: "Development of subcutaneously administered RNAi therapeutic ARO-HBV for chronic hepatitis B virus infection", JOURNAL OF HEPATOLOGY, vol. 68, April 2018 (2018-04-01), AMSTERDAM, NL, pages S18 - S19, XP055691743, ISSN: 0168-8278, DOI: 10.1016/S0168-8278(18)30255-1 *
E. GANE: "First results with RNA interference (RNAi) in chronic hepatitis B (CHB) using ARO-HBV", CONFERENCE REPORTS FOR NATAP, THE LIVER MEETING, SAN FRANCISCO NOV 2018, 13 November 2018 (2018-11-13), XP055691686, Retrieved from the Internet <URL:http://www.natap.org/2018/AASLD/AASLD_249.htm> [retrieved on 20200505] *
JOURNAL OF PHARMACEUTICAL SCIENCE, vol. 66, 1977, pages 2
LAMPERTICO ET AL., J HEPATOL., vol. 67, no. 2, 2017, pages 370 - 398
PENG YING ET AL: "Child-Pugh Versus MELD Score for the Assessment of Prognosis in Liver Cirrhosis : A Systematic Review and Meta-Analysis of Observational Studies", MEDICINE, vol. 95, no. 8, February 2016 (2016-02-01), US, pages e2877, XP055906925, ISSN: 0025-7974, DOI: 10.1097/MD.0000000000002877 *
SORIANO VICENTE ET AL: "Advances in hepatitis B therapeutics", THERAPEUTIC ADVANCES IN INFECTIOUS DISEASE JUN 2016, vol. 7, 15 October 2020 (2020-10-15), XP055905796, ISSN: 2049-9361, Retrieved from the Internet <URL:http://journals.sagepub.com/doi/full-xml/10.1177/2049936120965027> DOI: 10.1177/2049936120965027 *

Similar Documents

Publication Publication Date Title
CN109843902B (en) RNAi agents for hepatitis B virus infection
US20210395745A1 (en) Rnai agents for hepatitis b virus infection
JP6922030B2 (en) Methods for the treatment of hepatitis B and hepatitis D virus infections
CN104837501A (en) Methods for the treatment of hepatitis B and hepatitis D infections
US20180179542A1 (en) OLIGONUCLEOTIDE TARGETING STRATEGY FOR cccDNA
WO2020214976A1 (en) Combination therapy for treating hepatitis b virus infection
WO2020214974A1 (en) Combination therapy for treating hepatitis b virus infection
US20230357773A1 (en) Oligonucleotide treatment of hepatitis b patients
WO2022152869A1 (en) Use of oligonucleotides for individuals with hepatic impairment
EP4230209A1 (en) Pharmaceutical composition, pharmaceutical combined formulation, and combined formulation kit for prevention or treatment of chronic hepatitis b, each comprising, as active ingredient, oral antiviral agent and therapeutic vaccine including lipopeptide and poly(i:c) adjuvant
WO2023281434A1 (en) Use of oligonucleotides for individuals with renal impairment
EP4168547A1 (en) Compositions and methods for treatment of hepatitis d virus infection
BR112021015361A2 (en) RNAI AGENTS FOR HEPATITIS B VIRUS INFECTION
KR20220121793A (en) Use of a compound in the prophylaxis or treatment of graft-versus-host disease
TW202220675A (en) Methods for the reduction of z-aat protein levels

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: 22701919

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: 22701919

Country of ref document: EP

Kind code of ref document: A1