WO2020255013A1 - Combination of hepatitis b virus (hbv) vaccines and capsid assembly modulators being amide derivatives - Google Patents

Combination of hepatitis b virus (hbv) vaccines and capsid assembly modulators being amide derivatives Download PDF

Info

Publication number
WO2020255013A1
WO2020255013A1 PCT/IB2020/055704 IB2020055704W WO2020255013A1 WO 2020255013 A1 WO2020255013 A1 WO 2020255013A1 IB 2020055704 W IB2020055704 W IB 2020055704W WO 2020255013 A1 WO2020255013 A1 WO 2020255013A1
Authority
WO
WIPO (PCT)
Prior art keywords
optionally substituted
independently
alkyl
membered
heteroatoms
Prior art date
Application number
PCT/IB2020/055704
Other languages
French (fr)
Inventor
Helen Horton
Jan Martin Berke
Frederik PAUWELS
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 WO2020255013A1 publication Critical patent/WO2020255013A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • A61K31/167Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide having the nitrogen of a carboxamide group directly attached to the aromatic ring, e.g. lidocaine, paracetamol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/18Sulfonamides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/407Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with other heterocyclic ring systems, e.g. ketorolac, physostigmine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41921,2,3-Triazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4453Non condensed piperidines, e.g. piperocaine only substituted in position 1, e.g. propipocaine, diperodon
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/45Non condensed piperidines, e.g. piperocaine having oxo groups directly attached to the heterocyclic ring, e.g. cycloheximide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/53DNA (RNA) vaccination
    • CCHEMISTRY; METALLURGY
    • 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
    • C12N2730/00Reverse transcribing DNA viruses
    • C12N2730/00011Details
    • C12N2730/10011Hepadnaviridae
    • C12N2730/10111Orthohepadnavirus, e.g. hepatitis B virus
    • C12N2730/10134Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein

Definitions

  • This application contains a sequence listing, which is submitted electronically via EFS- Web as an ASCII formatted sequence listing with a file name“065814_36WOl Sequence Listing” and a creation date of June 4, 2020 and having a size of 46 kb.
  • the sequence listing submitted via EFS-Web is part of the specification and is herein incorporated by reference in its entirety.
  • Hepatitis B virus is a small 3.2-kb hepatotropic DNA virus that encodes four open reading frames and seven proteins. Approximately 240 million people have chronic hepatitis B infection (chronic HBV), characterized by persistent virus and subvirus particles in the blood for more than 6 months (Cohen et al. J. Viral Hepat. (2011) 18(6), 377-83). Persistent HBV infection leads to T-cell exhaustion in circulating and intrahepatic HBV-specific CD4+ and CD8+ T-cells through chronic stimulation of HBV-specific T-cell receptors with viral peptides and circulating antigens. As a result, T-cell polyfunctionality is decreased (i.e., decreased levels of IL-2, tumor necrosis factor (TNF)-a, IFN-g, and lack of proliferation).
  • TNF tumor necrosis factor
  • a safe and effective prophylactic vaccine against HBV infection has been available since the 1980s and is the mainstay of hepatitis B prevention (World Health Organization, Hepatitis B: Fact sheet No. 204 [Internet] 2015 March.).
  • the World Health Organization recommends vaccination of all infants, and, in countries where there is low or intermediate hepatitis B endemicity, vaccination of all children and adolescents ( ⁇ 18 years of age), and of people of certain at risk population categories. Due to vaccination, worldwide infection rates have dropped dramatically. However, prophylactic vaccines do not cure established HBV infection.
  • Chronic HBV is currently treated with IFN-a and nucleoside or nucleotide analogs, but there is no ultimate cure due to the persistence in infected hepatocytes of an intracellular viral replication intermediate called covalently closed circular DNA (cccDNA), which plays a fundamental role as a template for viral RNAs, and thus new virions. It is thought that induced virus-specific T-cell and B-cell responses can effectively eliminate cccDNA-carrying hepatocytes.
  • Current therapies targeting the HBV polymerase suppress viremia, but offer limited effect on cccDNA that resides in the nucleus and related production of circulating antigen.
  • HBV surface antigens HBsAg
  • pegylated interferon (pegIFN)-a has proven better in comparison to nucleoside or nucleotide therapy in terms of sustained off-treatment response with a finite treatment course.
  • IFN-a is reported to exert epigenetic suppression of cccDNA in cell culture and humanized mice, which leads to reduction of virion productivity and transcripts (Belloni et al. J. Clin. Invest. (2012) 122(2), 529-537).
  • this therapy is still fraught with side-effects and overall responses are rather low, in part because IFN-a has only poor modulatory influences on HBV-specific T-cells. In particular, cure rates are low ( ⁇ 10%) and toxicity is high.
  • HBV antivirals namely the HBV polymerase inhibitors entecavir and tenofovir
  • HBV polymerase inhibitors entecavir and tenofovir are effective as monotherapy in inducing viral suppression with a high genetic barrier to emergence of drug resistant mutants and consecutive prevention of liver disease progression.
  • cure of chronic hepatitis B defined by HBsAg loss or seroconversion, is rarely achieved with such HBV polymerase inhibitors. Therefore, these antivirals in theory need to be administered indefinitely to prevent reoccurrence of liver disease, similar to antiretroviral therapy for human immunodeficiency virus (HIV).
  • hepatitis B virus particularly chronic HBV
  • the invention satisfies this need by providing therapeutic combinations or compositions and methods for inducing an immune response against hepatitis B viruses (HBV) infection.
  • the immunogenic compositions/combinations and methods of the invention can be used to provide therapeutic immunity to a subject, such as a subject having chronic HBV infection.
  • the application relates to therapeutic combinations or compositions comprising one or more HBV antigens, or one or more polynucleotides encoding the HBV antigens, and a capsid assembly modulator, for use in treating an HBV infection in a subject in need thereof.
  • the therapeutic combination comprises:
  • a truncated HBV core antigen consisting of an amino acid sequence that is at least 95%, such as at least 95%, 96%, 97%, 98%, 99% or 100%, identical to SEQ ID NO: 2, b) a first non-naturally occurring nucleic acid molecule comprising a first polynucleotide sequence encoding the truncated HBV core antigen;
  • an HBV polymerase antigen having an amino acid sequence that is at least 90%, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%, identical to SEQ ID NO: 7, wherein the HBV polymerase antigen does not have reverse transcriptase activity and RNase H activity, and
  • a capsid assembly modulator such as those described herein.
  • the truncated HBV core antigen consists of the amino acid sequence of SEQ ID NO: 2 or SEQ ID NO: 4, and the HBV polymerase antigen comprises the amino acid sequence of SEQ ID NO: 7.
  • the therapeutic combination comprises at least one of the HBV polymerase antigen and the truncated HBV core antigen. In certain embodiments, the therapeutic combination comprises the HBV polymerase antigen and the truncated HBV core antigen. In one embodiment, the therapeutic combination comprises at least one of the first non- naturally occurring nucleic acid molecule comprising the first polynucleotide sequence encoding the truncated HBV core antigen, and the second non-naturally occurring nucleic acid molecule comprising the second polynucleotide sequence encoding the HBV polymerase antigen.
  • the first non-naturally occurring nucleic acid molecule further comprises a polynucleotide sequence encoding a signal sequence operably linked to the N-terminus of the truncated HBV core antigen
  • the second non-naturally occurring nucleic acid molecule further comprises a polynucleotide sequence encoding a signal sequence operably linked to the N-terminus of the HBV polymerase antigen
  • the signal sequence independently comprises the amino acid sequence of SEQ ID NO: 9 or SEQ ID NO: 15, more preferably, the signal sequence is encoded by the polynucleotide sequence of SEQ ID NO: 8 or SEQ ID NO: 14, respectively.
  • the first polynucleotide sequence comprises the polynucleotide sequence having at least 90%, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%, sequence identity to SEQ ID NO: 1 or SEQ ID NO: 3.
  • the second polynucleotide sequence comprises a polynucleotide sequence having at least 90%, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%, sequence identity to SEQ ID NO: 5 or SEQ ID NO: 6.
  • capsid modulators useful for the invention are described in International Patent Application Publications WO 2014106019, WO 2014106019, WO 2014106019, WO 2014106019, WO 2014106019, WO 2014106019, WO 2014106019, WO 2014106019, WO 2014106019, WO 2014106019, WO 2014106019, WO 2014106019, WO 2014106019, WO 2014106019, WO
  • a therapeutic combination comprises:
  • polynucleotide sequence encoding a truncated HBV core antigen consisting of an amino acid sequence that is at least 95%, such as at least 95%, 96%, 97%, 98%, 99% or 100%, identical to SEQ ID NO: 2;
  • a capsid assembly modulator selected from the group consisting of:
  • diastereomer or pharmaceutically accepted salt, hydrate, or solvate thereof including a hydrate, a solvate, a pharmaceutically acceptable salt, a prodrug, or a complex thereof described herein;
  • AVII enantiomer, diastereomer or pharmaceutically accepted salt, hydrate, or solvate thereof, including a hydrate, a solvate, a pharmaceutically acceptable salt, a prodrug, or a complex thereof described herein; and iii. a compound of formula of formula (B-I), or a pharmaceutical salt described herein.
  • the therapeutic combination comprises a) a first non-naturally occurring nucleic acid molecule comprising a first polynucleotide sequence encoding an truncated HBV core antigen consisting of the amino acid sequence of SEQ ID NO: 2 or SEQ ID NO: 4; b) a second non-naturally occurring nucleic acid molecule comprising a second polynucleotide sequence encoding an HBV polymerase antigen having the amino acid sequence of SEQ ID NO: 7, and (c) a compound selected from the group consisting of the exemplified compounds, or an enantiomer, diastereomer or pharmaceutically accepted salt, hydrate, or solvate thereof, including a hydrate, a solvate, a pharmaceutically acceptable salt, a prodrug, or a complex thereof.
  • the therapeutic combination comprises a first non-naturally occurring nucleic acid molecule comprising a polynucleotide sequence having at least 90%, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%, sequence identity to SEQ ID NO: 1 or SEQ ID NO: 3, and a second non-naturally occurring nucleic acid molecule comprising the polynucleotide sequence having at least 90%, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%, sequence identity to SEQ ID NO: 5 or SEQ ID NO: 6.
  • the therapeutic combination comprises a) a first non-naturally occurring nucleic acid molecule comprising a first polynucleotide sequence of SEQ ID NO: 1 or SEQ ID NO: 3; b) a second non-naturally occurring nucleic acid molecule comprising a second polynucleotide sequence of SEQ ID NO: 5 or 6; and c) a compound selected from the group consisting of:
  • each of the first and the second non-naturally occurring nucleic acid molecules is a DNA molecule, preferably the DNA molecule is present on a plasmid or a viral vector.
  • each of the first and the second non-naturally occurring nucleic acid molecules is an RNA molecule, preferably an mRNA or a self-replicating RNA molecule. In some embodiments, each of the first and the second non-naturally occurring nucleic acid molecules is independently formulated with a lipid nanoparticle (LNP).
  • LNP lipid nanoparticle
  • the application relates to a kit comprising a therapeutic combination of the application.
  • the application also relates to a therapeutic combination or kit of the application for use in inducing an immune response against hepatitis B virus (HBV); and use of a therapeutic combination, composition or kit of the application in the manufacture of a medicament for inducing an immune response against hepatitis B virus (HBV).
  • the use can further comprise a combination with another immunogenic or therapeutic agent, preferably another HBV antigen or another HBV therapy.
  • the subject has chronic HBV infection.
  • the application further relates to a therapeutic combination or kit of the application for use in treating an HBV-induced disease in a subject in need thereof; and use of therapeutic combination or kit of the application in the manufacture of a medicament for treating an HBV- induced disease in a subject in need thereof.
  • the use can further comprise a combination with another therapeutic agent, preferably another anti-HBV antigen.
  • the subject has chronic HBV infection, and the HBV-induced disease is selected from the group consisting of advanced fibrosis, cirrhosis, and hepatocellular carcinoma (HCC).
  • the application also relates to a method of inducing an immune response against an HBV or a method of treating an HBV infection or an HBV-induced disease, comprising administering to a subject in need thereof a therapeutic combination according to embodiments of the application.
  • FIG.1A and FIG.1B show schematic representations of DNA plasmids according to embodiments of the application;
  • FIG.1A shows a DNA plasmid encoding an HBV core antigen according to an embodiment of the application;
  • FIG.1B shows a DNA plasmid encoding an HBV polymerase (pol) antigen according to an embodiment of the application;
  • the HBV core and pol antigens are expressed under control of a CMV promoter with an N-terminal cystatin S signal peptide that is cleaved from the expressed antigen upon secretion from the cell;
  • transcriptional regulatory elements of the plasmid include an enhancer sequence located between the CMV promoter and the polynucleotide sequence encoding the HBV antigen and a bGH polyadeny
  • FIG.2A and FIG.2B show the schematic representations of the expression cassettes in adenoviral vectors according to embodiments of the application
  • FIG.2A shows the expression cassette for a truncated HBV core antigen, which contains a CMV promoter, an intron (a fragment derived from the human ApoAI gene - GenBank accession X01038 base pairs 295– 523, harboring the ApoAI second intron), a human immunoglobulin secretion signal, followed by a coding sequence for a truncated HBV core antigen and a SV40 polyadenylation signal
  • FIG.2B shows the expression cassette for a fusion protein of a truncated HBV core antigen operably linked to an HBV polymerase antigen, which is otherwise identical to the expression cassette for the truncated HBV core antigen except the HBV antigen.
  • FIG.3 shows ELISPOT responses of Balb/c mice immunized with different DNA plasmids expressing HBV core antigen or HBV pol antigen, as described in Example 3; peptide pools used to stimulate splenocytes isolated from the various vaccinated animal groups are indicated in gray scale; the number of responsive T-cells are indicated on the y-axis expressed as spot forming cells (SFC) per 10 6 splenocytes; DETAILED DESCRIPTION OF THE INVENTION
  • the conjunctive term“and/or” between multiple recited elements is understood as encompassing both individual and combined options. For instance, where two elements are conjoined by“and/or,” a first option refers to the applicability of the first element without the second. A second option refers to the applicability of the second element without the first. A third option refers to the applicability of the first and second elements together. Any one of these options is understood to fall within the meaning, and therefore satisfy the requirement of the term“and/or” as used herein.
  • any numerical value such as a concentration or a concentration range described herein, are to be understood as being modified in all instances by the term “about.”
  • a numerical value typically includes ⁇ 10% of the recited value.
  • a concentration of 1 mg/mL includes 0.9 mg/mL to 1.1 mg/mL.
  • a concentration range of 1 mg/mL to 10 mg/mL includes 0.9 mg/mL to 11 mg/mL.
  • the use of a numerical range expressly includes all possible subranges, all individual numerical values within that range, including integers within such ranges and fractions of the values unless the context clearly indicates otherwise.
  • phrases“percent (%) sequence identity” or“% identity” or“% identical to” when used with reference to an amino acid sequence describe the number of matches (“hits”) of identical amino acids of two or more aligned amino acid sequences as compared to the number of amino acid residues making up the overall length of the amino acid sequences.
  • hits the number of matches
  • the percentage of amino acid residues that are the same e.g.90%, 91%, 92%, 93%, 94%, 95%, 97%, 98%, 99%, or 100% identity over the full- length of the amino acid sequences
  • sequences which are compared to determine sequence identity may thus differ by substitution(s), addition(s) or deletion(s) of amino acids.
  • Suitable programs for aligning protein sequences are known to the skilled person.
  • the percentage sequence identity of protein sequences can, for example, be determined with programs such as CLUSTALW, Clustal Omega, FASTA or BLAST, e.g. using the NCBI BLAST algorithm (Altschul SF, et al (1997), Nucleic Acids Res.25:3389-3402).
  • 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” can be administration of the two or more therapies or components at least within the same day.
  • two components are“administered together with” or“administered in combination with,” they can be administered in separate compositions sequentially within a short time period, such as 24, 20, 16, 12, 8 or 4 hours, or within 1 hour, or they can be administered in a single composition at the same time.“Subsequent administration” can be administration of the two or more therapies or components in the same day or on separate days.
  • a first therapy or component e.g.
  • first DNA plasmid encoding an HBV antigen 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 (e.g., second DNA plasmid encoding an HBV antigen), and/or a third therapy or component (e.g.,capsid assembly modulator).
  • a first therapy or component e.g.
  • first DNA plasmid encoding an HBV antigen a second therapy or component (e.g., second DNA plasmid encoding an HBV antigen), and a third therapy or component (e.g., capsid assembly modulator) are administered in the same composition.
  • a first therapy or component e.g. first DNA plasmid encoding an HBV antigen
  • a second therapy or component e.g., second DNA plasmid encoding an HBV antigen
  • a third therapy or component e.g., capsid assembly modulator
  • a“non-naturally occurring” nucleic acid or polypeptide refers to a nucleic acid or polypeptide that does not occur in nature.
  • A“non-naturally occurring” nucleic acid or polypeptide can be synthesized, treated, fabricated, and/or otherwise manipulated in a laboratory and/or manufacturing setting.
  • a non-naturally occurring nucleic acid or polypeptide can comprise a naturally-occurring nucleic acid or polypeptide that is treated, processed, or manipulated to exhibit properties that were not present in the naturally-occurring nucleic acid or polypeptide, prior to treatment.
  • a“non-naturally occurring” nucleic acid or polypeptide can be a nucleic acid or polypeptide isolated or separated from the natural source in which it was discovered, and it lacks covalent bonds to sequences with which it was associated in the natural source.
  • A“non-naturally occurring” nucleic acid or polypeptide can be made recombinantly or via other methods, such as chemical synthesis.
  • “subject” means any animal, preferably a mammal, most preferably a human, to whom will be or has been treated by a method according to an embodiment of the application.
  • the term“mammal” as used herein, encompasses any mammal.
  • mammals include, but are not limited to, cows, horses, sheep, pigs, cats, dogs, mice, rats, rabbits, guinea pigs, non-human primates (NHPs) such as monkeys or apes, humans, etc., more preferably a human.
  • NHPs non-human primates
  • operably linked refers to a linkage or a juxtaposition wherein the components so described are in a relationship permitting them to function in their intended manner.
  • a regulatory sequence operably linked to a nucleic acid sequence of interest is capable of directing the transcription of the nucleic acid sequence of interest, or a signal sequence operably linked to an amino acid sequence of interest is capable of secreting or translocating the amino acid sequence of interest over a membrane.
  • HBV vectors of the application may contain particular components, including, but not limited to, certain promoter sequences, enhancer or regulatory sequences, signal peptides, coding sequence of an HBV antigen, polyadenylation signal sequences, etc. arranged in a particular order, those having ordinary skill in the art will appreciate that the concepts disclosed herein may equally apply to other components arranged in other orders that can be used in HBV vectors of the application.
  • the application contemplates use of any of the applicable components in any combination having any sequence that can be used in HBV vectors of the application, whether or not a particular combination is expressly described.
  • the invention generally relates to a therapeutic combination comprising one or more HBV antigens and at least one capsid assembly modulator.
  • HBV Hepatitis B Virus
  • hepatitis B virus or“HBV” refers to a virus of the hepadnaviridae family.
  • HBV is a small (e.g., 3.2 kb) hepatotropic DNA virus that encodes four open reading frames and seven proteins.
  • the seven proteins encoded by HBV include small (S), medium (M), and large (L) surface antigen (HBsAg) or envelope (Env) proteins, pre-Core protein, core protein, viral polymerase (Pol), and HBx protein.
  • HBV expresses three surface antigens, or envelope proteins, L, M, and S, with S being the smallest and L being the largest.
  • the extra domains in the M and L proteins are named Pre-S2 and Pre-S1, respectively.
  • Core protein is the subunit of the viral nucleocapsid. Pol is needed for synthesis of viral DNA (reverse transcriptase, RNaseH, and primer), which takes place in nucleocapsids localized to the cytoplasm of infected hepatocytes.
  • PreCore is the core protein with an N-terminal signal peptide and is proteolytically processed at its N and C termini before secretion from infected cells, as the so-called hepatitis B e-antigen (HBeAg). HBx protein is required for efficient transcription of covalently closed circular DNA (cccDNA). HBx is not a viral structural protein. All viral proteins of HBV have their own mRNA except for core and polymerase, which share an mRNA. With the exception of the protein pre-Core, none of the HBV viral proteins are subject to post-translational proteolytic processing.
  • the HBV virion contains a viral envelope, nucleocapsid, and single copy of the partially double-stranded DNA genome.
  • the nucleocapsid comprises 120 dimers of core protein and is covered by a capsid membrane embedded with the S, M, and L viral envelope or surface antigen proteins.
  • rcDNA relaxed circular DNA
  • phosphorylation of the core protein induces structural changes, exposing a nuclear localization signal enabling interaction of the capsid with so-called importins.
  • importins mediate binding of the core protein to nuclear pore complexes upon which the capsid
  • rcDNA covalently closed circular DNA genome from which overlapping transcripts encode for HBeAg, HBsAg, Core protein, viral polymerase and HBx protein.
  • Core protein, viral polymerase, and pre-genomic RNA (pgRNA) associate in the cytoplasm and self- assemble into immature pgRNA-containing capsid particles, which further convert into mature rcDNA-capsids and function as a common intermediate that is either enveloped and secreted as infectious virus particles or transported back to the nucleus to replenish and maintain a stable cccDNA pool.
  • pgRNA pre-genomic RNA
  • HBV is divided into four serotypes (adr, adw, ayr, ayw) based on antigenic epitopes present on the envelope proteins, and into eight genotypes (A, B, C, D, E, F, G, and H) based on the sequence of the viral genome.
  • the HBV genotypes are distributed over different geographic regions. For example, the most prevalent genotypes in Asia are genotypes B and C. Genotype D is dominant in Africa, the Middle East, and India, whereas genotype A is widespread in Northern Europe, sub-Saharan Africa, and West Africa.
  • the terms“HBV antigen,”“antigenic polypeptide of HBV,”“HBV antigenic polypeptide,”“HBV antigenic protein,”“HBV immunogenic polypeptide,” and“HBV immunogen” all refer to a polypeptide capable of inducing an immune response, e.g., a humoral and/or cellular mediated response, against an HBV in a subject.
  • the HBV antigen can be a polypeptide of HBV, a fragment or epitope thereof, or a combination of multiple HBV polypeptides, portions or derivatives thereof.
  • an HBV antigen is capable of raising in a host a protective immune response, e.g., inducing an immune response against a viral disease or infection, and/or producing an immunity (i.e., vaccinates) in a subject against a viral disease or infection, that protects the subject against the viral disease or infection.
  • an HBV antigen can comprise a polypeptide or immunogenic fragment(s) thereof from any HBV protein, such as HBeAg, pre-core protein, HBsAg (S, M, or L proteins), core protein, viral polymerase, or HBx protein derived from any HBV genotype, e.g., genotype A, B, C, D, E, F, G, and/or H, or combination thereof.
  • each of the terms“HBV core antigen,”“HBc” and“core antigen” refers to an HBV antigen capable of inducing an immune response, e.g., a humoral and/or cellular mediated response, against an HBV core protein in a subject.
  • Each of the terms“core,”“core polypeptide,” and“core protein” refers to the HBV viral core protein.
  • Full-length core antigen is typically 183 amino acids in length and includes an assembly domain (amino acids 1 to 149) and a nucleic acid binding domain (amino acids 150 to 183).
  • the 34-residue nucleic acid binding domain is required for pre-genomic RNA encapsidation. This domain also functions as a nuclear import signal.
  • HBV core protein is dimeric in solution, with the dimers self-assembling into icosahedral capsids. Each dimer of core protein has four a-helix bundles flanked by an a-helix domain on either side. Truncated HBV core proteins lacking the nucleic acid binding domain are also capable of forming capsids.
  • an HBV antigen is a truncated HBV core antigen.
  • a“truncated HBV core antigen” refers to an HBV antigen that does not contain the entire length of an HBV core protein, but is capable of inducing an immune response against the HBV core protein in a subject.
  • an HBV core antigen can be modified to delete one or more amino acids of the highly positively charged (arginine rich) C-terminal nucleic acid binding domain of the core antigen, which typically contains seventeen arginine (R) residues.
  • a truncated HBV core antigen of the application is preferably a C-terminally truncated HBV core protein which does not comprise the HBV core nuclear import signal and/or a truncated HBV core protein from which the C-terminal HBV core nuclear import signal has been deleted.
  • a truncated HBV core antigen comprises a deletion in the C-terminal nucleic acid binding domain, such as a deletion of 1 to 34 amino acid residues of the C-terminal nucleic acid binding domain, e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, or 34 amino acid residues, preferably a deletion of all 34 amino acid residues.
  • a truncated HBV core antigen comprises a deletion in the C-terminal nucleic acid binding domain, preferably a deletion of all 34 amino acid residues.
  • An HBV core antigen of the application can be a consensus sequence derived from multiple HBV genotypes (e.g., genotypes A, B, C, D, E, F, G, and H).
  • Consensus sequence means an artificial sequence of amino acids based on an alignment of amino acid sequences of homologous proteins, e.g., as determined by an alignment (e.g., using Clustal Omega) of amino acid sequences of homologous proteins. It can be the calculated order of most frequent amino acid residues, found at each position in a sequence alignment, based upon sequences of HBV antigens (e.g., core, pol, etc.) from at least 100 natural HBV isolates.
  • a consensus sequence can be non-naturally occurring and different from the native viral sequences.
  • Consensus sequences can be designed by aligning multiple HBV antigen sequences from different sources using a multiple sequence alignment tool, and at variable alignment positions, selecting the most frequent amino acid.
  • a consensus sequence of an HBV antigen is derived from HBV genotypes B, C, and D.
  • the term“consensus antigen” is used to refer to an antigen having a consensus sequence.
  • An exemplary truncated HBV core antigen lacks the nucleic acid binding function, and is capable of inducing an immune response in a mammal against at least two HBV genotypes.
  • a truncated HBV core antigen is capable of inducing a T cell response in a mammal against at least HBV genotypes B, C and D.
  • a truncated HBV core antigen is capable of inducing a CD8 T cell response in a human subject against at least HBV genotypes A, B, C and D.
  • an HBV core antigen of the application is a consensus antigen, preferably a consensus antigen derived from HBV genotypes B, C, and D, more preferably a truncated consensus antigen derived from HBV genotypes B, C, and D.
  • An exemplary truncated HBV core consensus antigen consists of an amino acid sequence that is at least 90% identical to SEQ ID NO: 2 or SEQ ID NO: 4, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9%, or 100% identical to SEQ ID NO: 2 or SEQ ID NO: 4.
  • SEQ ID NO: 2 and SEQ ID NO: 4 are core consensus antigens derived from HBV genotypes B, C, and D.
  • SEQ ID NO: 2 and SEQ ID NO: 4 each contain a 34-amino acid C-terminal deletion of the highly positively charged (arginine rich) nucleic acid binding domain of the native core antigen.
  • an HBV core antigen is a truncated HBV antigen consisting of the amino acid sequence of SEQ ID NO: 2.
  • an HBV core antigen is a truncated HBV antigen consisting of the amino acid sequence of SEQ ID NO: 4.
  • an HBV core antigen further contains a signal sequence operably linked to the N-terminus of a mature HBV core antigen sequence, such as the amino acid sequence of SEQ ID NO: 2 or SEQ ID NO: 4.
  • the signal sequence has the amino acid sequence of SEQ ID NO: 9 or SEQ ID NO: 15.
  • the term“HBV polymerase antigen,”“HBV Pol antigen” or“HBV pol antigen” refers to an HBV antigen capable of inducing an immune response, e.g., a humoral and/or cellular mediated response, against an HBV polymerase in a subject.
  • Each of the terms “polymerase,”“polymerase polypeptide,”“Pol” and“pol” refers to the HBV viral DNA polymerase.
  • the HBV viral DNA polymerase has four domains, including, from the N terminus to the C terminus, a terminal protein (TP) domain, which acts as a primer for minus-strand DNA synthesis; a spacer that is nonessential for the polymerase functions; a reverse transcriptase (RT) domain for transcription; and a RNase H domain.
  • TP terminal protein
  • RT reverse transcriptase
  • an HBV antigen comprises an HBV Pol antigen, or any immunogenic fragment or combination thereof.
  • An HBV Pol antigen can contain further modifications to improve immunogenicity of the antigen, such as by introducing mutations into the active sites of the polymerase and/or RNase domains to decrease or substantially eliminate certain enzymatic activities.
  • an HBV Pol antigen of the application does not have reverse transcriptase activity and RNase H activity, and is capable of inducing an immune response in a mammal against at least two HBV genotypes.
  • an HBV Pol antigen is capable of inducing a T cell response in a mammal against at least HBV genotypes B, C and D. More preferably, an HBV Pol antigen is capable of inducing a CD8 T cell response in a human subject against at least HBV genotypes A, B, C and D.
  • an HBV Pol antigen is an inactivated Pol antigen.
  • an inactivated HBV Pol antigen comprises one or more amino acid mutations in the active site of the polymerase domain.
  • an inactivated HBV Pol antigen comprises one or more amino acid mutations in the active site of the RNaseH domain.
  • an inactivated HBV pol antigen comprises one or more amino acid mutations in the active site of both the polymerase domain and the RNaseH domain.
  • the“YXDD” motif in the polymerase domain of an HBV pol antigen that can be required for nucleotide/metal ion binding can be mutated, e.g., by replacing one or more of the aspartate residues (D) with asparagine residues (N), eliminating or reducing metal coordination function, thereby decreasing or substantially eliminating reverse transcriptase function.
  • the“DEDD” motif in the RNaseH domain of an HBV pol antigen required for Mg2+ coordination can be mutated, e.g., by replacing one or more aspartate residues (D) with asparagine residues (N) and/or replacing the glutamate residue (E) with glutamine (Q), thereby decreasing or substantially eliminating RNaseH function.
  • an HBV pol antigen is modified by (1) mutating the aspartate residues (D) to asparagine residues (N) in the“YXDD” motif of the polymerase domain; and (2) mutating the first aspartate residue (D) to an asparagine residue (N) and the first glutamate residue (E) to a glutamine residue (N) in the“DEDD” motif of the RNaseH domain, thereby decreasing or substantially eliminating both the reverse transcriptase and RNaseH functions of the pol antigen.
  • an HBV pol antigen is a consensus antigen, preferably a consensus antigen derived from HBV genotypes B, C, and D, more preferably an inactivated consensus antigen derived from HBV genotypes B, C, and D.
  • An exemplary HBV pol consensus antigen according to the application comprises an amino acid sequence that is at least 90% identical to SEQ ID NO: 7, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100% identical to SEQ ID NO: 7, preferably at least 98% identical to SEQ ID NO: 7, such as at least 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100% identical to SEQ ID NO: 7.
  • SEQ ID NO: 7 is a pol consensus antigen derived from HBV genotypes B, C, and D comprising four mutations located in the active sites of the polymerase and RNaseH domains.
  • the four mutations include mutation of the aspartic acid residues (D) to asparagine residues (N) in the“YXDD” motif of the polymerase domain; and mutation of the first aspartate residue (D) to an asparagine residue (N) and mutation of the glutamate residue (E) to a glutamine residue (Q) in the“DEDD” motif of the RNaseH domain.
  • an HBV pol antigen comprises the amino acid sequence of SEQ ID NO: 7.
  • an HBV pol antigen consists of the amino acid sequence of SEQ ID NO: 7.
  • an HBV pol antigen further contains a signal sequence operably linked to the N-terminus of a mature HBV pol antigen sequence, such as the amino acid sequence of SEQ ID NO: 7.
  • the signal sequence has the amino acid sequence of SEQ ID NO: 9 or SEQ ID NO: 15.
  • an HBV antigen comprises a fusion protein comprising a truncated HBV core antigen operably linked to an HBV Pol antigen, or an HBV Pol antigen operably linked to a truncated HBV core antigen, preferably via a linker.
  • a linker serves primarily as a spacer between the first and second polypeptides.
  • a linker is made up of amino acids linked together by peptide bonds, preferably from 1 to 20 amino acids linked by peptide bonds, wherein the amino acids are selected from the 20 naturally occurring amino acids.
  • the 1 to 20 amino acids are selected from glycine, alanine, proline, asparagine, glutamine, and lysine.
  • a linker is made up of a majority of amino acids that are sterically unhindered, such as glycine and alanine.
  • Exemplary linkers are polyglycines, particularly (Gly)5, (Gly)8; poly(Gly-Ala), and polyalanines.
  • One exemplary suitable linker as shown in the Examples below is (AlaGly)n, wherein n is an integer of 2 to 5.
  • a fusion protein of the application is capable of inducing an immune response in a mammal against HBV core and HBV Pol of at least two HBV genotypes.
  • a fusion protein is capable of inducing a T cell response in a mammal against at least HBV genotypes B, C and D. More preferably, the fusion protein is capable of inducing a CD8 T cell response in a human subject against at least HBV genotypes A, B, C and D.
  • a fusion protein comprises a truncated HBV core antigen having an amino acid sequence at least 90%, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9%, or 100% identical to SEQ ID NO: 2 or SEQ ID NO: 4, a linker, and an HBV Pol antigen having an amino acid sequence at least 90%, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9%, or 100%, identical to SEQ ID NO: 2 or
  • a fusion protein comprises a truncated HBV core antigen consisting of the amino acid sequence of SEQ ID NO: 2 or SEQ ID NO: 4, a linker comprising (AlaGly)n, wherein n is an integer of 2 to 5, and an HBV Pol antigen having the amino acid sequence of SEQ ID NO: 7. More preferably, a fusion protein according to an embodiment of the application comprises the amino acid sequence of SEQ ID NO: 16. In one embodiment of the application, a fusion protein further comprises a signal sequence operably linked to the N-terminus of the fusion protein. Preferably, the signal sequence has the amino acid sequence of SEQ ID NO: 9 or SEQ ID NO: 15. In one embodiment, a fusion protein comprises the amino acid sequence of SEQ ID NO: 17.
  • HBV vaccines that can be used for the present invention are described in U.S. Patent Application No: 16/223,251, filed December 18, 2018, the contents of the application, more preferably the examples of the application, are hereby incorporated by reference in their entireties.
  • the application provides a non-naturally occurring nucleic acid molecule encoding an HBV antigen useful for an invention according to embodiments of the application, and vectors comprising the non-naturally occurring nucleic acid.
  • a first or second non-naturally occurring nucleic acid molecule can comprise any polynucleotide sequence encoding an HBV antigen useful for the application, which can be made using methods known in the art in view of the present disclosure.
  • a first or second polynucleotide encodes at least one of a truncated HBV core antigen and an HBV polymerase antigen of the application.
  • a polynucleotide can be in the form of RNA or in the form of DNA obtained by recombinant techniques (e.g., cloning) or produced synthetically (e.g., chemical synthesis).
  • the DNA can be single-stranded or double-stranded, or can contain portions of both double-stranded and single- stranded sequence.
  • the DNA can, for example, comprise genomic DNA, cDNA, or
  • the polynucleotide can also be a DNA/RNA hybrid.
  • the polynucleotides and vectors of the application can be used for recombinant protein production, expression of the protein in host cell, or the production of viral particles.
  • a polynucleotide is DNA.
  • a first non-naturally occurring nucleic acid molecule comprises a first polynucleotide sequence encoding a truncated HBV core antigen consisting of an amino acid sequence that is at least 90% identical to SEQ ID NO: 2 or SEQ ID NO: 4, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100% identical to SEQ ID NO: 2, preferably 98%, 99% or 100% identical to SEQ ID NO: 2 or SEQ ID NO: 4.
  • a first non-naturally occurring nucleic acid molecule comprises a first polynucleotide sequence encoding a truncated HBV core antigen consisting the amino acid sequence of SEQ ID NO: 2 or SEQ ID NO: 4.
  • polynucleotide sequences of the application encoding a truncated HBV core antigen consisting of the amino acid sequence of SEQ ID NO: 2 or SEQ ID NO: 4 include, but are not limited to, a polynucleotide sequence at least 90% identical to SEQ ID NO: 1 or SEQ ID NO: 3, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100% identical to SEQ ID NO: 1 or SEQ ID NO: 3, preferably 98%, 99% or 100% identical to SEQ ID NO: 1 or SEQ ID NO: 3.
  • Exemplary non-naturally occurring nucleic acid molecules encoding a truncated HBV core antigen have the polynucleotide sequence of SEQ
  • a first non-naturally occurring nucleic acid molecule further comprises a coding sequence for a signal sequence that is operably linked to the N-terminus of the HBV core antigen sequence.
  • the signal sequence has the amino acid sequence of SEQ ID NO: 9 or SEQ ID NO: 15.
  • the coding sequence for a signal sequence comprises the polynucleotide sequence of SEQ ID NO: 8 or SEQ ID NO: 14.
  • a second non-naturally occurring nucleic acid molecule comprises a second polynucleotide sequence encoding an HBV polymerase antigen comprising an amino acid sequence that is at least 90% identical to SEQ ID NO: 7, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100% identical to SEQ ID NO: 7, preferably 100% identical to SEQ ID NO: 7.
  • a second non-naturally occurring nucleic acid molecule comprises a second polynucleotide sequence encoding an HBV polymerase antigen consisting of the amino acid sequence of SEQ ID NO: 7.
  • polynucleotide sequences of the application encoding an HBV Pol antigen comprising the amino acid sequence of at least 90% identical to SEQ ID NO: 7 include, but are not limited to, a polynucleotide sequence at least 90% identical to SEQ ID NO: 5 or SEQ ID NO: 6, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100% identical to SEQ ID NO: 5 or SEQ ID NO: 6, preferably 98%, 99% or 100% identical to SEQ ID NO: 5 or SEQ ID NO: 6.
  • Exemplary non-naturally occurring nucleic acid molecules encoding an HBV pol antigen have the polynucleotide sequence of SEQ ID NOs: 5 or 6.
  • a second non-naturally occurring nucleic acid molecule further comprises a coding sequence for a signal sequence that is operably linked to the N-terminus of the HBV pol antigen sequence, such as the amino acid sequence of SEQ ID NO: 7.
  • the signal sequence has the amino acid sequence of SEQ ID NO: 9 or SEQ ID NO: 15.
  • the coding sequence for a signal sequence comprises the polynucleotide sequence of SEQ ID NO: 8 or SEQ ID NO: 14.
  • a non-naturally occurring nucleic acid molecule encodes an HBV antigen fusion protein comprising a truncated HBV core antigen operably linked to an HBV Pol antigen, or an HBV Pol antigen operably linked to a truncated HBV core antigen.
  • a non-naturally occurring nucleic acid molecule of the application encodes a truncated HBV core antigen consisting of an amino acid sequence that is at least 90% identical to SEQ ID NO: 2 or SEQ ID NO: 4, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100% identical to SEQ ID NO: 2 or SEQ ID NO: 4, preferably 100% identical to SEQ ID NO: 2 or SEQ ID NO: 4, more preferably 100% identical to SEQ ID NO: 2 or SEQ ID NO:4; a linker; and an HBV polymerase antigen comprising an amino acid sequence that is at least 90% identical to SEQ ID NO: 7, such as at least 90%, 91%, 92%, 93%, 94%
  • a non-naturally occurring nucleic acid molecule encodes a fusion protein comprising a truncated HBV core antigen consisting of the amino acid sequence of SEQ ID NO: 2 or SEQ ID NO: 4, a linker comprising (AlaGly)n, wherein n is an integer of 2 to 5; and an HBV Pol antigen comprising the amino acid sequence of SEQ ID NO: 7.
  • a non-naturally occurring nucleic acid molecule encodes an HBV antigen fusion protein comprising the amino acid sequence of SEQ ID NO: 16.
  • polynucleotide sequences of the application encoding an HBV antigen fusion protein include, but are not limited to, a polynucleotide sequence at least 90% identical to SEQ ID NO: 1 or SEQ ID NO: 3, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100% identical to SEQ ID NO: 1 or SEQ ID NO: 3, preferably 98%, 99% or 100% identical to SEQ ID NO: 1 or SEQ ID NO: 3, operably linked to a linker coding sequence at least 90% identical to SEQ ID NO: 11, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%,
  • a non-naturally occurring nucleic acid molecule encoding an HBV antigen fusion protein comprises SEQ ID NO: 1 or SEQ ID NO: 3, operably linked to SEQ ID NO: 11, which is further operably linked to SEQ ID NO: 5 or SEQ ID NO: 6.
  • a non-naturally occurring nucleic acid molecule encoding an HBV fusion further comprises a coding sequence for a signal sequence that is operably linked to the N-terminus of the HBV fusion sequence, such as the amino acid sequence of SEQ ID NO: 16.
  • the signal sequence has the amino acid sequence of SEQ ID NO: 9 or SEQ ID NO: 15. More preferably, the coding sequence for a signal sequence comprises the
  • the encoded fusion protein with the signal sequence comprises the amino acid sequence of SEQ ID NO: 17.
  • a“vector” is a nucleic acid molecule used to carry genetic material into another cell, where it can be replicated and/or expressed. Any vector known to those skilled in the art in view of the present disclosure can be used. Examples of vectors include, but are not limited to, plasmids, viral vectors (bacteriophage, animal viruses, and plant viruses), cosmids, and artificial chromosomes (e.g., YACs).
  • a vector is a DNA plasmid.
  • a vector can be a DNA vector or an RNA vector.
  • a vector of the application can be an expression vector.
  • expression vector refers to any type of genetic construct comprising a nucleic acid coding for an RNA capable of being transcribed.
  • Expression vectors include, but are not limited to, vectors for recombinant protein expression, such as a DNA plasmid or a viral vector, and vectors for delivery of nucleic acid into a subject for expression in a tissue of the subject, such as a DNA plasmid or a viral vector. It will be appreciated by those skilled in the art that the design of the expression vector can depend on such factors as the choice of the host cell to be transformed, the level of expression of protein desired, etc.
  • Vectors of the application can contain a variety of regulatory sequences.
  • regulatory sequence refers to any sequence that allows, contributes or modulates the functional regulation of the nucleic acid molecule, including replication, duplication, transcription, splicing, translation, stability and/or transport of the nucleic acid or one of its derivative (i.e. mRNA) into the host cell or organism.
  • this term encompasses promoters, enhancers and other expression control elements (e.g., polyadenylation signals and elements that affect mRNA stability).
  • a vector is a non-viral vector.
  • non- viral vectors include, but are not limited to, DNA plasmids, bacterial artificial chromosomes, yeast artificial chromosomes, bacteriophages, etc.
  • non-viral vectors include, but are not limited to, RNA replicon, mRNA replicon, modified mRNA replicon or self-amplifying mRNA, closed linear deoxyribonucleic acid, e.g. a linear covalently closed DNA such as linear covalently closed double stranded DNA molecule.
  • a non-viral vector is a DNA plasmid.
  • DNA plasmids used for expression of an encoded polynucleotide typically comprise an origin of replication, a multiple cloning site, and a selectable marker, which for example, can be an antibiotic resistance gene.
  • DNA plasmids suitable that can be used include, but are not limited to, commercially available expression vectors for use in well-known expression systems (including both prokaryotic and eukaryotic systems), such as pSE420 (Invitrogen, San Diego, Calif.), which can be used for production and/or expression of protein in Escherichia coli; pYES2 (Invitrogen, Thermo Fisher Scientific), which can be used for production and/or expression in Saccharomyces cerevisiae strains of yeast; MAXBAC® complete baculovirus expression system (Thermo Fisher Scientific), which can be used for production and/or expression in insect cells; pcDNATM or pcDNA3TM (Life Technologies, Thermo Fisher Scientific), which can be used for high level constitutive protein expression in mammalian cells; and pVAX or pVAX-1 (Life Technologies, Thermo Fisher Scientific), which can be used for high-level transient expression of a protein of interest in most mammalian cells.
  • the backbone of any commercially available DNA plasmid can be modified to optimize protein expression in the host cell, such as to reverse the orientation of certain elements (e.g., origin of replication and/or antibiotic resistance cassette), replace a promoter endogenous to the plasmid (e.g., the promoter in the antibiotic resistance cassette), and/or replace the polynucleotide sequence encoding transcribed proteins (e.g., the coding sequence of the antibiotic resistance gene), by using routine techniques and readily available starting materials. (See e.g., Sambrook et al., Molecular Cloning a
  • a DNA plasmid is an expression vector suitable for protein expression in mammalian host cells.
  • Expression vectors suitable for protein expression in mammalian host cells include, but are not limited to, pcDNATM, pcDNA3TM, pVAX, pVAX-1, ADVAX, NTC8454, etc.
  • an expression vector is based on pVAX-1, which can be further modified to optimize protein expression in mammalian cells.
  • pVAX-1 is commonly used plasmid in DNA vaccines, and contains a strong human intermediate early cytomegalovirus (CMV-IE) promoter followed by the bovine growth hormone (bGH)-derived polyadenylation sequence (pA).
  • pVAX-1 further contains a pUC origin of replication and kanamycin resistance gene driven by a small prokaryotic promoter that allows for bacterial plasmid propagation.
  • a vector of the application can also be a viral vector.
  • viral vectors are genetically engineered viruses carrying modified viral DNA or RNA that has been rendered non- infectious, but still contains viral promoters and transgenes, thus allowing for translation of the transgene through a viral promoter. Because viral vectors are frequently lacking infectious sequences, they require helper viruses or packaging lines for large-scale transfection. Examples of viral vectors that can be used include, but are not limited to, adenoviral vectors, adeno- associated virus vectors, pox virus vectors, enteric virus vectors, Venezuelan Equine Encephalitis virus vectors, Semliki Forest Virus vectors, Tobacco Mosaic Virus vectors, lentiviral vectors, etc.
  • viral vectors examples include, but are not limited to, arenavirus viral vectors, replication-deficient arenavirus viral vectors or replication-competent arenavirus viral vectors, bi-segmented or tri-segmented arenavirus, infectious arenavirus viral vectors, nucleic acids which comprise an arenavirus genomic segment wherein one open reading frame of the genomic segment is deleted or functionally inactivated (and replaced by a nucleic acid encoding an HBV antigen as described herein), arenavirus such as lymphocytic choriomeningitidis virus (LCMV), e.g., clone 13 strain or MP strain, and arenavirus such as Junin virus e.g., Candid #1 strain.
  • the vector can also be a non-viral vector.
  • a viral vector is an adenovirus vector, e.g., a recombinant adenovirus vector.
  • a recombinant adenovirus vector can for instance be derived from a human adenovirus (HAdV, or AdHu), or a simian adenovirus such as chimpanzee or gorilla adenovirus (ChAd, AdCh, or SAdV) or rhesus adenovirus (rhAd).
  • an adenovirus vector is a recombinant human adenovirus vector, for instance a recombinant human adenovirus serotype 26, or any one of recombinant human adenovirus serotype 5, 4, 35, 7, 48, etc.
  • an adenovirus vector is a rhAd vector, e.g. rhAd51, rhAd52 or rhAd53.
  • a recombinant viral vector useful for the application can be prepared using methods known in the art in view of the present disclosure. For example, in view of the degeneracy of the genetic code, several nucleic acid sequences can be designed that encode the same polypeptide.
  • a polynucleotide encoding an HBV antigen of the application can optionally be codon-optimized to ensure proper expression in the host cell (e.g., bacterial or mammalian cells). Codon-optimization is a technology widely applied in the art, and methods for obtaining codon-optimized polynucleotides will be well known to those skilled in the art in view of the present disclosure.
  • a vector of the application e.g., a DNA plasmid or a viral vector (particularly an adenoviral vector), can comprise any regulatory elements to establish conventional function(s) of the vector, including but not limited to replication and expression of the HBV antigen(s) encoded by the polynucleotide sequence of the vector. Regulatory elements include, but are not limited to, a promoter, an enhancer, a polyadenylation signal, translation stop codon, a ribosome binding element, a transcription terminator, selection markers, origin of replication, etc.
  • a vector can comprise one or more expression cassettes.
  • An“expression cassette” is part of a vector that directs the cellular machinery to make RNA and protein.
  • An expression cassette typically comprises three components: a promoter sequence, an open reading frame, and a 3’-untranslated region (UTR) optionally comprising a polyadenylation signal.
  • An open reading frame is a reading frame that contains a coding sequence of a protein of interest (e.g., HBV antigen) from a start codon to a stop codon.
  • Regulatory elements of the expression cassette can be operably linked to a polynucleotide sequence encoding an HBV antigen of interest.
  • the term“operably linked” is to be taken in its broadest reasonable context, and refers to a linkage of polynucleotide elements in a functional relationship.
  • a polynucleotide is“operably linked” when it is placed into a functional relationship with another polynucleotide.
  • a promoter is operably linked to a coding sequence if it affects the transcription of the coding sequence.
  • Any components suitable for use in an expression cassette described herein can be used in any combination and in any order to prepare vectors of the application.
  • a vector can comprise a promoter sequence, preferably within an expression cassette, to control expression of an HBV antigen of interest.
  • the term“promoter” is used in its conventional sense, and refers to a nucleotide sequence that initiates the transcription of an operably linked nucleotide sequence.
  • a promoter is located on the same strand near the nucleotide sequence it transcribes. Promoters can be a constitutive, inducible, or repressible. Promoters can be naturally occurring or synthetic.
  • a promoter can be derived from sources including viral, bacterial, fungal, plants, insects, and animals.
  • a promoter can be a homologous promoter (i.e., derived from the same genetic source as the vector) or a heterologous promoter (i.e., derived from a different vector or genetic source).
  • the promoter can be endogenous to the plasmid (homologous) or derived from other sources (heterologous).
  • the promoter is located upstream of the polynucleotide encoding an HBV antigen within an expression cassette.
  • promoters examples include, but are not limited to, a promoter from simian virus 40 (SV40), a mouse mammary tumor virus (MMTV) promoter, a human immunodeficiency virus (HIV) promoter such as the bovine immunodeficiency virus (BIV) long terminal repeat (LTR) promoter, a Moloney virus promoter, an avian leukosis virus (ALV) promoter, a cytomegalovirus (CMV) promoter such as the CMV immediate early promoter (CMV-IE), Epstein Barr virus (EBV) promoter, or a Rous sarcoma virus (RSV) promoter.
  • SV40 simian virus 40
  • MMTV mouse mammary tumor virus
  • HAV human immunodeficiency virus
  • HSV human immunodeficiency virus
  • BIV bovine immunodeficiency virus
  • LTR long terminal repeat
  • AMV avian leukosis virus
  • CMV cytomegalovirus
  • a promoter can also be a promoter from a human gene such as human actin, human myosin, human hemoglobin, human muscle creatine, or human metalothionein.
  • a promoter can also be a tissue specific promoter, such as a muscle or skin specific promoter, natural or synthetic.
  • a promoter is a strong eukaryotic promoter, preferably a cytomegalovirus immediate early (CMV-IE) promoter.
  • CMV-IE cytomegalovirus immediate early
  • a vector can comprise additional polynucleotide sequences that stabilize the expressed transcript, enhance nuclear export of the RNA transcript, and/or improve transcriptional- translational coupling.
  • sequences include polyadenylation signals and enhancer sequences.
  • a polyadenylation signal is typically located downstream of the coding sequence for a protein of interest (e.g., an HBV antigen) within an expression cassette of the vector.
  • Enhancer sequences are regulatory DNA sequences that, when bound by transcription factors, enhance the transcription of an associated gene.
  • An enhancer sequence is preferably located upstream of the polynucleotide sequence encoding an HBV antigen, but downstream of a promoter sequence within an expression cassette of the vector.
  • the polyadenylation signal can be a SV40 polyadenylation signal, LTR polyadenylation signal, bovine growth hormone (bGH) polyadenylation signal, human growth hormone (hGH) polyadenylation signal, or human b-globin polyadenylation signal.
  • a polyadenylation signal is a bovine growth hormone (bGH) polyadenylation signal or a SV40 polyadenylation signal.
  • a nucleotide sequence of an exemplary bGH polyadenylation signal is shown in SEQ ID NO: 20.
  • a nucleotide sequence of an exemplary SV40 polyadenylation signal is shown in SEQ ID NO: 13.
  • an enhancer sequence can be human actin, human myosin, human hemoglobin, human muscle creatine, or a viral enhancer, such as one from CMV, HA, RSV, or EBV.
  • a viral enhancer such as one from CMV, HA, RSV, or EBV.
  • WPRE Woodchuck HBV Post- transcriptional regulatory element
  • ApoAI intron/exon sequence derived from human apolipoprotein A1 precursor
  • HTLV-1) long terminal repeat (LTR) untranslated R-U5 domain of the human T-cell leukemia virus type 1 (HTLV-1) long terminal repeat (LTR), a splicing enhancer, a synthetic rabbit b- globin intron, or any combination thereof.
  • an enhancer sequence is a composite sequence of three consecutive elements of the untranslated R-U5 domain of HTLV-1 LTR, rabbit b-globin intron, and a splicing enhancer, which is referred to herein as“a triple enhancer sequence.”
  • a nucleotide sequence of an exemplary triple enhancer sequence is shown in SEQ ID NO: 10.
  • Another exemplary enhancer sequence is an ApoAI gene fragment shown in SEQ ID NO: 12.
  • a vector can comprise a polynucleotide sequence encoding a signal peptide sequence.
  • the polynucleotide sequence encoding the signal peptide sequence is located upstream of the polynucleotide sequence encoding an HBV antigen.
  • Signal peptides typically direct localization of a protein, facilitate secretion of the protein from the cell in which it is produced, and/or improve antigen expression and cross-presentation to antigen-presenting cells.
  • a signal peptide can be present at the N-terminus of an HBV antigen when expressed from the vector, but is cleaved off by signal peptidase, e.g., upon secretion from the cell.
  • a signal peptide can be a cystatin S signal peptide; an immunoglobulin (Ig) secretion signal, such as the Ig heavy chain gamma signal peptide SPIgG or the Ig heavy chain epsilon signal peptide SPIgE.
  • Ig immunoglobulin
  • a signal peptide sequence is a cystatin S signal peptide.
  • Exemplary nucleic acid and amino acid sequences of a cystatin S signal peptide are shown in SEQ ID NOs: 8 and 9, respectively.
  • Exemplary nucleic acid and amino acid sequences of an immunoglobulin secretion signal are shown in SEQ ID NOs: 14 and 15, respectively.
  • a vector such as a DNA plasmid
  • Bacterial origins of replication and antibiotic resistance cassettes can be located in a vector in the same orientation as the expression cassette encoding an HBV antigen, or in the opposite (reverse) orientation.
  • An origin of replication (ORI) is a sequence at which replication is initiated, enabling a plasmid to reproduce and survive within cells. Examples of ORIs suitable for use in the application include, but are not limited to ColE1, pMB1, pUC, pSC101, R6K, and 15A, preferably pUC.
  • An exemplary nucleotide sequence of a pUC ORI is shown in SEQ ID NO: 21.
  • Expression cassettes for selection and maintenance in bacterial cells typically include a promoter sequence operably linked to an antibiotic resistance gene.
  • the promoter sequence operably linked to an antibiotic resistance gene differs from the promoter sequence operably linked to a polynucleotide sequence encoding a protein of interest, e.g., HBV antigen.
  • the antibiotic resistance gene can be codon optimized, and the sequence composition of the antibiotic resistance gene is normally adjusted to bacterial, e.g., E. coli, codon usage.
  • Any antibiotic resistance gene known to those skilled in the art in view of the present disclosure can be used, including, but not limited to, kanamycin resistance gene (Kanr), ampicillin resistance gene (Ampr), and tetracycline resistance gene (Tetr), as well as genes conferring resistance to chloramphenicol, bleomycin, spectinomycin, carbenicillin, etc.
  • Kanr kanamycin resistance gene
  • Amr ampicillin resistance gene
  • Tetr tetracycline resistance gene
  • an antibiotic resistance gene in the antibiotic expression cassette of a vector is a kanamycin resistance gene (Kanr).
  • the sequence of Kanr gene is shown in SEQ ID NO: 22.
  • the Kanr gene is codon optimized.
  • An exemplary nucleic acid sequence of a codon optimized Kanr gene is shown in SEQ ID NO: 23.
  • the Kanr can be operably linked to its native promoter, or the Kanr gene can be linked to a heterologous promoter.
  • the Kanr gene is operably linked to the ampicillin resistance gene (Ampr) promoter, known as the bla promoter.
  • An exemplary nucleotide sequence of a bla promoter is shown in SEQ ID NO: 24.
  • a vector is a DNA plasmid comprising an expression cassette including a polynucleotide encoding at least one of an HBV antigen selected from the group consisting of an HBV pol antigen comprising an amino acid sequence at least 90%, such as 90%, 91%, 92%, 93%, 94%, 95%, 96, 97%, preferably at least 98%, such as at least 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100%, identical to SEQ ID NO: 7, and a truncated HBV core antigen consisting of the amino acid sequence at least 95%, such as 95%, 96, 97%, preferably at least 98%, such as at least 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100%, identical to S
  • Such vector further comprises an antibiotic resistance expression cassette including a polynucleotide encoding an antibiotic resistance gene, preferably a Kanr gene, more preferably a codon optimized Kanr gene of at least 90% identical to SEQ ID NO: 23, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100% identical to SEQ ID NO: 23, preferably 100% identical to SEQ ID NO: 23, operably linked to an Ampr (bla) promoter of SEQ ID NO: 24, upstream of and operably linked to the polynucleotide encoding the antibiotic resistance gene; and an origin of replication, preferably a pUC ori of SEQ ID NO: 21.
  • a vector is a viral vector, preferably an adenoviral vector, more preferably an Ad26 or Ad35 vector, comprising an expression cassette including a polynucleotide encoding at least one of an HBV antigen selected from the group consisting of an HBV pol antigen comprising an amino acid sequence at least 90%, such as 90%, 91%, 92%, 93%, 94%, 95%, 96, 97%, preferably at least 98%, such as at least 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100%, identical to SEQ ID NO: 7, and a truncated HBV core antigen consisting of the amino acid sequence at least 95%, such as 95%, 96, 97%, preferably at least 98%, such as at least 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%
  • a vector such as a plasmid DNA vector or a viral vector (preferably an adenoviral vector, more preferably an Ad26 or Ad35 vector), encodes an HBV Pol antigen having the amino acid sequence of SEQ ID NO: 7.
  • the vector comprises a coding sequence for the HBV Pol antigen that is at least 90% identical to the polynucleotide sequence of SEQ ID NO: 5 or 6, such as 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100% identical to SEQ ID NO: 5 or 6, preferably 100% identical to SEQ ID NO: 5 or 6.
  • a vector such as a plasmid DNA vector or a viral vector (preferably an adenoviral vector, more preferably an Ad26 or Ad35 vector), encodes a truncated HBV core antigen consisting of the amino acid sequence of SEQ ID NO: 2 or SEQ ID NO: 4.
  • the vector comprises a coding sequence for the truncated HBV core antigen that is at least 90% identical to the polynucleotide sequence of SEQ ID NO: 1 or SEQ ID NO: 3, such as 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100% identical to SEQ ID NO: 1 or SEQ ID NO: 3, preferably 100% identical to SEQ ID NO: 1 or SEQ ID NO: 3.
  • a vector such as a plasmid DNA vector or a viral vector (preferably an adenoviral vector, more preferably an Ad26 or Ad35 vector), encodes a fusion protein comprising an HBV Pol antigen having the amino acid sequence of SEQ ID NO: 7 and a truncated HBV core antigen consisting of the amino acid sequence of SEQ ID NO: 1 or SEQ ID NO: 3.
  • the vector comprises a coding sequence for the fusion, which contains a coding sequence for the truncated HBV core antigen at least 90% identical to SEQ ID NO: 1 or SEQ ID NO: 3, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100% identical to SEQ ID NO: 1 or SEQ ID NO: 3, preferably 98%, 99% or 100% identical to SEQ ID NO: 1 or SEQ ID NO: 3, more preferably SEQ ID NO: 1 or SEQ ID NO: 3, operably linked to a coding sequence for the HBV Pol antigen at least 90% identical to SEQ ID NO: 5 or SEQ ID NO: 6, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 95
  • the coding sequence for the truncated HBV core antigen is operably linked to the coding sequence for the HBV Pol antigen via a coding sequence for a linker at least 90% identical to SEQ ID NO: 11, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100% identical to SEQ ID NO: 11, preferably 98%, 99% or 100% identical to SEQ ID NO: 11.
  • a vector comprises a coding sequence for the fusion having SEQ ID NO: 1 or SEQ ID NO: 3 operably linked to SEQ ID NO: 11, which is further operably linked to SEQ ID NO: 5 or SEQ ID NO: 6.
  • polynucleotides and expression vectors encoding the HBV antigens of the application can be made by any method known in the art in view of the present disclosure.
  • a polynucleotide encoding an HBV antigen can be introduced or“cloned” into an expression vector using standard molecular biology techniques, e.g., polymerase chain reaction (PCR), etc., which are well known to those skilled in the art.
  • PCR polymerase chain reaction
  • the application also provides cells, preferably isolated cells, comprising any of the polynucleotides and vectors described herein.
  • the cells can, for instance, be used for recombinant protein production, or for the production of viral particles.
  • Embodiments of the application thus also relate to a method of making an HBV antigen of the application.
  • the method comprises transfecting a host cell with an expression vector comprising a polynucleotide encoding an HBV antigen of the application operably linked to a promoter, growing the transfected cell under conditions suitable for expression of the HBV antigen, and optionally purifying or isolating the HBV antigen expressed in the cell.
  • the HBV antigen can be isolated or collected from the cell by any method known in the art including affinity chromatography, size exclusion chromatography, etc. Techniques used for recombinant protein expression will be well known to one of ordinary skill in the art in view of the present disclosure.
  • the expressed HBV antigens can also be studied without purifying or isolating the expressed protein, e.g., by analyzing the supernatant of cells transfected with an expression vector encoding the HBV antigen and grown under conditions suitable for expression of the HBV antigen.
  • non-naturally occurring or recombinant polypeptides comprising an amino acid sequence that is at least 90% identical to the amino acid sequence of SEQ ID NO: 2, SEQ ID NO: 4, or SEQ ID NO: 7.
  • isolated nucleic acid molecules encoding these sequences, vectors comprising these sequences operably linked to a promoter, and compositions comprising the polypeptide, polynucleotide, or vector are also contemplated by the application.
  • a recombinant polypeptide comprises an amino acid sequence that is at least 90% identical to the amino acid sequence of SEQ ID NO: 2, such as 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100% identical to SEQ ID NO: 2.
  • a non-naturally occurring or recombinant polypeptide consists of SEQ ID NO: 2.
  • a non-naturally occurring or recombinant polypeptide comprises an amino acid sequence that is at least 90% identical to the amino acid sequence of SEQ ID NO: 4, such as 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100% identical to SEQ ID NO: 4.
  • recombinant polypeptide comprises SEQ ID NO: 4.
  • a non-naturally occurring or recombinant polypeptide comprises an amino acid sequence that is at least 90% identical to the amino acid sequence of SEQ ID NO: 7, such as 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100% identical to SEQ ID NO: 7.
  • recombinant polypeptide consists of SEQ ID NO: 7.
  • antibodies or antigen binding fragments thereof that specifically bind to a non-naturally occurring polypeptide of the application.
  • an antibody specific to a non-naturally HBV antigen of the application does not bind specifically to another HBV antigen.
  • an antibody of the application that binds specifically to an HBV Pol antigen having the amino acid sequence of SEQ ID NO: 7 will not bind specifically to an HBV Pol antigen not having the amino acid sequence of SEQ ID NO: 7.
  • the term“antibody” includes polyclonal, monoclonal, chimeric, humanized, Fv, Fab and F(ab ⁇ )2; bifunctional hybrid (e.g., Lanzavecchia et al., Eur. J. Immunol. 17:105, 1987), single-chain (Huston et al., Proc. Natl. Acad. Sci. USA 85:5879, 1988; Bird et al., Science 242:423, 1988); and antibodies with altered constant regions (e.g., U.S. Pat. No.
  • an antibody that“specifically binds to” an antigen refers to an antibody that binds to the antigen with a KD of 1 ⁇ 10-7 M or less.
  • an antibody that “specifically binds to” an antigen refers to an antibody that binds to the antigen with a KD of 1 ⁇ 10-7 M or less.
  • KD refers to the dissociation constant, which is obtained from the ratio of Kd to Ka (i.e., Kd/Ka) and is expressed as a molar concentration (M). KD values for antibodies can be determined using methods in the art in view of the present disclosure.
  • the KD of an antibody can be determined by using surface plasmon resonance, such as by using a biosensor system, e.g., a Biacore® system, or by using bio-layer interferometry technology, such as an Octet RED96 system.
  • a biosensor system e.g., a Biacore® system
  • bio-layer interferometry technology such as an Octet RED96 system.
  • capsid assembly modulators refers to a compound that disrupts or accelerates or inhibits or hinders or delays or reduces or modifies normal capsid assembly (e.g., during maturation) or normal capsid disassembly (e.g., during infectivity) or perturbs capsid stability, thereby inducing aberrant capsid morphology and function.
  • capsid assembly modulators refers to a compound that disrupts or accelerates or inhibits or hinders or delays or reduces or modifies normal capsid assembly (e.g., during maturation) or normal capsid disassembly (e.g., during infectivity) or perturbs capsid stability, thereby inducing aberrant capsid morphology and function.
  • these compounds described in more detail below, are selected from (a) specific compounds of formulae (I), (XI), (XVIII), and (XXI), (b) specific compounds of formula (A-I) to formula (A-VII), and (c) specific compounds of
  • Examples of compounds of formulae (I), (XI), (XVIII), and (XXI) (or enantiomeric and diastereomeric forms, hydrates, solvates, pharmaceutically acceptable salts, prodrugs and complexes thereof), synthesis, biologic activities, uses or other related information thereof are described in International Patent Application Publication WO 2014106019 and U.S. Patent Application Publication US 20160024004, the contents of which are hereby incorporated by reference in their entireties.
  • the compounds for use in the invention are those having the formula (I) or enantiomeric and diastereomeric forms, hydrates, solvates, pharmaceutically acceptable salts, prodrugs and complexes thereof:
  • A is selected from a group consisting of SO 2 and CO;
  • R 1 is selected from a group consisting of optionally substituted C 1-6 linear alkyl, optionally substituted C 1-6 branched alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted benzyl; R 1 may also alternatively or additionally optionally include optionally substituted C 1-6 haloalkyl, optionally substituted 3-7 membered cycloheteroalkyl, optionally substituted C 2-8 alkenyl, optionally substituted C 2-8 alkynyl, optionally substituted C 1-6 alkoxy, optionally substituted amine, optionally substituted amidine, optionally substituted carboxyamine, optionally substituted carboxy-C 1-6 -alkoxide, -SO 2 -C 1- 6 alkyl, optionally substituted heterocyclic, or optionally substituted heteroaryl;
  • R 2 is selected from a group consisting of hydrogen and optionally substituted C 1-6 linear alkyl; R 2 may also alternatively or additionally optionally include optionally substituted cycloalkyl or optionally substituted heterocyclic; or
  • R 1 and R 2 are taken together with the atoms to which they are bound to form an optionally substituted heterocycle (including bicyclic or adamantyl structures) with 3 to 10 atoms; and
  • R 3 is selected from a group consisting of optionally substituted aryl, optionally substituted benzyl, optionally substituted alkylaryl, optionally substituted heteroaryl, and optionally substituted alkylheteroaryl ; R 3 may also optionally comprise an optionally substituted C 1-6 linear alkyl;
  • R 4 is selected from a group consisting of hydrogen and optionally substituted C 1-6 linear alkyl
  • R 5 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C 1-6 branched alkyl, optionally substituted C 1-6 haloalkyl, and OR 9
  • R 5 may also alternatively or additionally optionally include cyano or N(R 9 ) 2 ;
  • R 6 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C 1-6 branched alkyl, optionally substituted C 1-6 haloalkyl, and OR 9 ; in some embodiments, R 6 may also alternatively or additionally optionally include cyano or N(R 9 ) 2 ; R 6 may also alternatively or additionally optionally include cyano or N(R 9 ) 2 ; or
  • R 4 and R 6 are taken together with the atoms to which they are bound to form an optionally substituted carbocyclic or heterocyclic ring with 5 to 6 atoms, optionally containing a carbonyl, optionally containing two carbonyls;
  • R 7 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C 1-6 branched alkyl, optionally substituted C 1-6 haloalkyl, and OR 9 ; R 7 may also alternatively or additionally optionally include cyano or N(R 9 ) 2 ;
  • R 8 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C 1-6 branched alkyl, optionally substituted C 1-6 haloalkyL-and OR 9 ; R 8 may also alternatively or additionally optionally include cyano or N( R 9 ) 2 ; or
  • R 2 and R 8 are taken together with the atoms to which they are bound to form an optionally substituted carbocyclic or heterocyclic ring with 5 to 6 atoms;
  • R 9 is independently at each occurrence selected from a group consisting of hydrogen, optionally substituted C 1-6 linear alkyl, optionally substituted C 1-6 branched alkyl, and optionally substituted C 3-7 cycloalkyl; R 9 may also alternatively or additionally optionally include independently at each occurrence optionally substituted aryl, optionally substituted benzyl, optionally substituted heterocyclyl, or optionally substituted heteroaryl;
  • R 3 is an optionally substituted phenyl and R 1 or R 2 , either individually or when taken together, contain a hydroxyl group, or (b) R 3 is an optionally substituted alkyl or phenyl, and N(R 1 )(R 2 ) is an optionally substituted piperazine or
  • R 3 is optionally substituted alkyl, aryl, or alkaryl and N(R 1 )(R 2 ) is
  • R or R is an unsubstituted or monosubstituted aryl, or an unsubstituted or monosubstituted aralkyl, or unsubstituted or monosubstituted heteroaryl and R 1 and R 2 are taken together with the atoms to which they are bound to form an optionally substituted heterocyclic ring structure with 6 to 12 atoms; or
  • the compound is not 3- ⁇ [(dicyclopropylmethyl)amino]sulfonyl ⁇ -N-(4- isopropoxyphenyl)benzamide; or 3-( ⁇ [2-(lH-benzimidazol-2-yl)propyl]amino ⁇ sulfonyl)-N-(4- isopropoxyphenyl)benzamide; or 3-[(cyclohexylamino)sulfonyl]-N-(4- isopropylphenyl)benzamide; or 3-(anilinosulfonyl)-N-(4-isopropylphenyl)benzamide; or 5- ⁇ [(3- ⁇ [(4-methoxyphenyl)amino]carbonyl ⁇ phenyl)sulfonyl]amino ⁇ pentanoic acid; or 3-[(tert- butylamino)sulfonyl]-N-(4-methoxyphenyl)benzamide;
  • the compounds for use in the invention are those having the formula (XI) or enantiomeric and diastereomeric forms, hydrates, solvates, pharmaceutically acceptable salts, prodrugs and complexes thereof:
  • A is selected from a group consisting of SO 2 and CO;
  • R 1 is selected from a group consisting of optionally substituted C 1-6 linear alkyl, optionally substituted C 1-6 branched alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted benzyl;
  • R 3 is selected from a group consisting of optionally substituted aryl, optionally substituted benzyl, optionally substituted alkylaryl, optionally substituted heteroaryl, and optionally substituted alkylheteroaryl; in some embodiments, R 3 may also comprise an optionally substituted C 1-6 linear alkyl;
  • R 4 is selected from a group consisting of hydrogen and optionally substituted C 1-6 linear alkyl
  • R 5 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C 1-6 branched alkyl, optionally substituted C 1-6 haloalkyl, and OR 9 ; R 5 may also alternatively or additionally optionally include cyano or N(R 9 ) 2 ;
  • R 6 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C 1-6 branched alkyl, optionally substituted C 1-6 haloalkyl, and OR 9 ; R 6 may also alternatively or additionally optionally include cyano or N( or
  • R 4 and R 6 are taken together with the atoms to which they are bound to form an optionally substituted carbocyclic or heterocyclic ring with 5 to 6 atoms, optionally containing a carbonyl, optionally containing two carbonyls;
  • R 7 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C 1-6 branched alkyl, optionally substituted C 1-6 haloalkyl, and OR 9 ; R 7 may also alternatively or additionally optionally include cyano or N(R 9 ) 2 ; R 8 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C 1-6 branched alkyl, optionally substituted C 1-6 haloalkyl, and OR 9 ;, R 8 may also alternatively or additionally optionally include cyano or N(R 9 ) 2 ;
  • R 9 is independently at each occurrence selected from a group consisting of hydrogen, optionally substituted C1-6 linear alkyl, optionally substituted C 1-6 branched alkyl, and optionally substituted cycloalkyl, and optionally substituted C 1-6 haloalkyl; R 9 may also alternatively or additionally optionally include independently at each occurrence optionally substituted aryl, optionally substituted benzyl, optionally substituted heterocyclyl, or optionally substituted heteroaryl;and R 11 is selected from a group consisting of hydrogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, and optionally substituted cycloalkyl.
  • the compounds for use in the invention are those having the formula (XVIII) or enantiomeric and diastereomeric forms, hydrates, solvates, pharmaceutically acceptable salts, prodrugs and complexes thereof:
  • A is selected from a group consisting of SO 2 and CO;
  • R 1 is selected from a group consisting of optionally substituted C 1-6 linear alkyl, optionally substituted C 1-6 branched alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted benzyl;
  • R 5 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C 1-6 branched alkyl, optionally substituted C 1-6 haloalkyl, and OR 9 ; R 5 may also alternatively or additionally optionally include cyano or N(R 9 ) 2 ;
  • R 6 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C 1-6 branched alkyl, optionally substituted C 1-6 haloalkyl, and OR 9 ; R 6 may also alternatively or additionally optionally include cyano or N(R 9 ) 2 ;
  • R 7 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C 1-6 branched alkyl, optionally substituted C 1-6 haloalkyl, and OR 9 ; R 7 may also alternatively or additionally optionally include cyano or N(R 9 ) 2 ;
  • R 8 is selected from a group consisting of hydrogen, halogen, optionally substituted C 1-6 linear alkyl, optionally substituted C 1-6 branched alkyl, optionally substituted C 1-6 haloalkyl, and OR 9 ; R 8 may also alternatively or additionally optionally include cyano or N(R 9 ) 2 ; R 9 is independently at each occurrence selected from a group consisting of hydrogen, optionally substituted C1-6 linear alkyl, optionally substituted C 1-6 branched alkyl, and optionally substituted cycloalkyl; R 9 may also alternatively or additionally optionally include independently at each occurrence optionally substituted aryl, optionally substituted benzyl, optionally substituted heterocyclyl, or optionally substituted heteroaryl;
  • R 11 is selected from a group consisting of hydrogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, and optionally substituted cycloalkyl;
  • R 12 is selected from a group consisting of hydrogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, and optionally substituted cycloalkyl; and R 13 is selected from a group consisting of optionally substituted aryl, optionally substituted benzyl, optionally substituted alkylaryl, optionally substituted heteroaryl, and optionally substituted alkylheteroaryl.
  • the compounds for use in the invention are those having the formula (XXI) or enantiomeric and diastereomeric forms, hydrates, solvates, pharmaceutically
  • R 1 is selected from a group consisting of optionally substituted C 1-6 linear alkyl, optionally substituted C 1-6 branched alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted benzyl; R 1 may also alternatively or additionally optionally include optionally substituted C 1-6 haloalkyl, optionally substituted 3-7 membered cycloheteroalkyl, optionally substituted C 2-8 alkenyl, optionally substituted C 2-8 alkynyl, optionally substituted C 1-6 alkoxy, optionally substituted amine, optionally substituted amidine, optionally substituted carboxyamine, optionally substituted carboxy-C 1-6 -alkoxide, -S0 2 -C 1- 6 alkyl, optionally substituted heterocyclic, or optionally substituted heteroaryl;
  • R 2 is selected from a group consisting of hydrogen and optionally substituted C 1-6 linear alkyl; R 2 may also alternatively or additionally optionally include optionally substituted cycloalkyl or optionally substituted heterocyclic; or
  • R 1 and R 2 are taken together with the atoms to which they are bound to form an optionally substituted heterocycle (including cyclic and heterocyclic structures) with 3 to 10 atoms;
  • R 5 is selected from a group consisting of hydrogen, halogen, optionally substituted C 1-6 linear alkyl, optionally substituted C 1-6 branched alkyl, optionally substituted C 1-6 haloalkyl, and OR 9 ; R 5 may also alternatively or additionally optionally include cyano or N(R 9 ) 2 ;
  • R 6 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C 1-6 branched alkyl, optionally substituted C 1-6 haloalkyl, and OR 9 ; R 6 may also alternatively or additionally optionally include cyano or N(R 9 ) 2 ;
  • R 7 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C 1-6 branched alkyl, optionally substituted C 1-6 haloalkyl, OR 9 ; R 7 may also alternatively or additionally optionally include cyano or N(R 9 ) 2 ;
  • R 8 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C 1-6 branched alkyl, optionally substituted C 1-6 haloalkyl, and OR 9 ; R 8 may also alternatively or additionally optionally include cyano or N(R 9 ) 2 ;
  • R 9 is independently at each occurrence selected from a group consisting of hydrogen, optionally substituted C1-6 linear alkyl, optionally substituted C 1-6 branched alkyl, and optionally substituted cycloalkyl, and optionally substituted C1-6 haloalkyl; R 9 may also alternatively or additionally optionally include, independently at each occurrence, optionally substituted aryl, optionally substituted benzyl, optionally substituted heterocyclyl, or optionally substituted heteroaryl;
  • R 12 is selected from a group consisting of hydrogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, and optionally substituted cycloalkyl; and R 13 is selected from a group consisting of optionally substituted aryl, optionally substituted benzyl, optionally substituted alkylaryl, optionally substituted heteroaryl, and optionally substituted alkylheteroaryl.
  • R 12 is selected from a group consisting of hydrogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, and optionally substituted cycloalkyl
  • R 13 is selected from a group consisting of optionally substituted aryl, optionally substituted benzyl, optionally substituted alkylaryl, optionally substituted heteroaryl, and optionally substituted alkylheteroaryl.
  • Exemplary compounds having formulae (I), (XI), (XVIII), or (XXI) have the following structures, where the substitu
  • exemplary embodiments include compounds having the formula (XXIX) or a pharmaceutically acceptable salt form thereof:
  • Exemplary embodiments include compounds having the formula (XXX) or a pharmaceutically acceptable salt form thereof:
  • Exemplary embodiments include compounds having the formula (XXXI) or a pharmaceutically acceptable salt form thereof:
  • Exemplary embodiments include compounds having the formula (XXXII) or a pharmaceutically acceptable salt form thereof:
  • Exemplary embodiments include compounds having the formula (XXXIII) or a pharmaceutically acceptable salt form thereof:
  • Exemplary embodiments include compounds having the formula (XXXIV) or a pharmaceutically acceptable salt form thereof:
  • Exemplary embodiments include compounds having the formula (XXXV) or a pharmaceutically acceptable salt form thereof:
  • Exemplary embodiments include compounds having the formula (XXXVI) or a pharmaceutically acceptable salt form thereof:
  • Exemplary embodiments include compounds having the formula (XXXVII) or a pharmaceutically acceptable salt form thereof:
  • Exemplary embodiments include compounds having the formula (XXXVIII) or a pharmaceutically acceptable salt form thereof:
  • Exemplary embodiments include compounds having the formula (XXXIX) or a pharmaceutically acceptable salt form thereof:
  • Exemplary embodiments include compounds having the formula (XXXX) or a pharmaceutically acceptable salt form thereof:
  • R 5 , R 6 , R 7 , and R 8 are defined herein below in Table 12.
  • Exemplary embodiments include compounds having the formula (XXXXI-A or XXXXI- B) or a pharmaceutically acceptable salt form thereof:
  • R 5 , R 6 , R 7 , and R 8 are defined herein below in Table 13.
  • Exemplary embodiments include compounds having the formula (XXXXII) or a pharmaceutically acceptable salt form thereof:
  • Exemplary embodiments include compounds having the formula (XXXXIII) or a pharmaceutically acceptable salt form thereof:
  • Exemplary embodiments include compounds having the formula (XXXXIV) or a pharmaceutically acceptable salt form thereof:
  • Exemplary embodiments include compounds having the formula (XXXXV) or a pharmaceutically acceptable salt form thereof:
  • Exemplary embodiments include compounds having the formula (XXXXVI) or a pharmaceutically acceptable salt form thereof:
  • Exemplary embodiments include compounds having the formula (XXXXVII) or a pharmaceutically acceptable salt form thereof:
  • Exemplary embodiments include compounds having the formula (XXXXVIII) or a pharmaceutically acceptable salt form thereof: wherein non- limiting examples of R 1 , and R 13 are defined herein below in Table 20.
  • Table 20 Exemplary embodiments of compounds of the formula (XXXXVIII): Specific compounds having the formulae (I), (XI), (XVIII), and (XXI) which are appropriate for use in embodiments according to the invention include, but are not limited to:
  • the effective amount of the compound having formula (I), (XI), (XVIII), or (XXI), or a pharmaceutically acceptable salt, an enantiomer, or a diastereomer thereof may be determined by routine experimentation, but can be in range of about 0.001 to 1000 mg, preferably from about 0.01 to 100 mg, more preferably from about 0.1 to 10 mg.
  • Suitable processes for synthesizing these compounds can be synthesized by any method or synthetic scheme known in the art or to be discovered.
  • the compounds can be prepared according to Scheme 1, shown below. Examples of specific synthetic compounds are described in WO 2014106019 and US 20160024004.
  • the compounds for use in the invention are those having formula (A- I):
  • A is phenyl, a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms, a five-membered heteroaromatic ring containing one, two, or three heteroatoms which are, independently, N, O, or S; a C 4-14 bicyclic ring, alkylheteroaryl, or alkylaryl; B is a six or seven-membered ring or a six or seven-membered bridged or spiro-fused ring containing zero, one, or two heteroatoms, which are, independently, N, O, or S, a five- membered ring containing zero, one, or two heteroatoms, which are, independently, N, O, or S; a four-membered ring containing zero, one, or two heteroatoms, which are, independently, N, O, or S, or a C 5-14 bicyclic ring,
  • R 1 and R 1 ’ are attached to a carbon, they are, independently, hydrogen, halogen (including F, Cl, Br, and I), SF 5 , CF 3 , hydroxy, N(R’)S(O) 2 R’, S(O) 2 R’, S(O) 2 N(R’) 2 , C 1-6 alkoxy, C 1-6 haloalkoxy, C 2-6 alkenyl, cyano, C 2-6 alkynyl, C 3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C 1-6 alkyl, arylalkoxycarbonyl, carboxy, C 1-6 haloalkyl, heterocyclylalkyl, or C 1-6 hydroxyalkyl;
  • R 1 and R 1 ’ are attached to a nitrogen, they are, independently, hydrogen, C2-6 alkoxy, C3-6 alkoxyalkyl, C2-6 alkenyl, alkoxycarbonyl, carbonylalkyl, carbonyl aryl, C 1-6 alkyl, heterocyclylalkyl, C 2-6 hydroxyalkyl, or S(O) 2 R’;
  • Each R’ is independently H, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, aryl, heteroaryl, alkylaryl, or arylalkyl, or if two R’ reside on the same nitrogen atom, they can come together to form a C 3-6 ring optionally containing a N, O, or S heteroatom;
  • the R’ groups can optionally be substituted with one or more substituents, which substituents are, independently, halo, C 1-6 haloalkyl, C 1-6 hydroxyalkyl, hydroxyl, carboxyl, acyl, aryl, acyloxy, amino, amido, carboxyl derivatives, alkylamino, dialkylamino, arylamino, alkoxy, alkoxyalkyl, aryloxy, nitro, cyano, sulfonic acid, thiol, imine, sulfonyl, sulfanyl, sulfinyl, sulfamonyl, ester, carboxylic acid, amide, phosphonyl, phosphinyl, phosphoryl, phosphine, thioester, thioether, acid halide, anhydride, oxime, hydrozine, carbamate, phosphonic acid, or phosphonate, either unprotected, or protected as
  • u and v are independently 0, 1, 2, 3, 4 or 5;
  • R 3 is H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, or C 2-6 alkynyl;
  • R 2 is C 1-6 alkyl, C 1-6 haloalkyl, C 2-8 alkoxyalkyl, C 2-6 alkenyl, C 2-6 alkynyl, aryl, such as phenyl, heteroaryl, including six-membered heteroaromatic rings containing one, two, or three nitrogen atoms and five-membered heteroaromatic rings containing one, two, or three heteroatoms, which, independently, are N, O, or S, alkylaryl, arylalkyl, a six-membered ring or a six-membered bridged or spiro-fused ring containing zero, one, or two heteroatoms which are, independently, N, O, or S, a seven-membered bridged or spiro-fused ring containing zero, one, or two hetero
  • R 2 is optionally substituted with one or more substituents, which each, independently, are halogen (including F, Cl, Br, and I), CF 3 , SF 5 , hydroxy, N(R’)S(O) 2 R’, S(O) 2 R’, S(O) 2 N(R’) 2 , C 1-6 alkoxy, C 1-6 haloalkoxy, cyano, azido, C 2-6 alkynyl, C 3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C 1-6 alkyl, arylalkoxycarbonyl, carboxy, C 1-6 haloalkyl, heterocyclylalkyl, or C 1-6 hydroxyalkyl; or is optionally substituted with aryl, substituted aryl, heteroaryl, or substituted heteroaryl, where substituents on the substituted aryl and substituted heteroaryl are selected from the group consisting of halogen, SF 5 ,
  • R 2 and R 3 can come together with the nitrogen to which they are attached to form a 6-10 membered bicyclic or bridged ring, a 3 to 8 saturated ring, or a 5 membered unsaturated ring; such bicyclic, bridged, saturated and unsaturated rings optionally containing one or more additional heteroatoms, where each is, independently, O, S or N, and optionally being substituted with one or more substituents, wherein each, independently, is halogen (including F, Cl, Br, I), CF 3 , hydroxy, N(R’)S(O) 2 R’, S(O) 2 R’, S(O) 2 N(R’) 2 , C 1-6 alkoxy, cyano, azido, C 2-6 alkynyl, C 3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C 1-6 alkyl, arylalkoxycarbonyl, carboxy, C 1-6 haloalkyl, heterocycly
  • the compounds for use in the invention are those having formula (A-II): or an enantiomer, diastereomer or pharmaceutically accepted salt, hydrate, or solvate thereof, including a hydrate, a solvate, a pharmaceutically acceptable salt, a prodrug, or a complex thereof,
  • R 1 and R 1 ’ are as defined with respect to formula (A-I);
  • u and v are independently 0, 1, 2, 3, 4 or 5;
  • C is phenyl, a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms, a five-membered heteroaromatic ring containing one, two, or three heteroatoms which are, independently, N, O, or S; a C 4-14 bicyclic ring, alkylaryl, or alkylheteroaryl;
  • D is phenyl, a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms, a five-membered heteroaromatic ring containing one, two, or three heteroatoms, which are, independently, N, O, or S, or a C 5-14 bicyclic ring,
  • R 4 is H or C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl;
  • R 5 is not alkylaryl, alkenyl, or a six-membered bridged ring;
  • R 5 is alkylaryl, arylalkyl, C 2-6 alkenyl, C 2-6 alkynyl, aryl, such as phenyl, heteroaryl, including six-membered heteroaromatic rings containing one, two, or three nitrogen atoms and five-membered heteroaromatic rings containing one, two, or three heteroatoms, which, independently, are N, O, or S; and a six-membered bridged or spiro-fused ring containing zero, one, or two heteroatoms which are, independently, N, O, or S;
  • R 5 is optionally substituted with one or more substituents, each of which is,
  • halogen including F, Cl, Br, and I
  • CF 3 SF 5 , hydroxy, N(R’)S(O) 2 R’, S(O) 2 R’, S(O) 2 N(R’) 2
  • R 4 and R 5 together with the nitrogen to which they are attached form a 3 to 4 membered ring optionally substituted with one or more substituents, each of which is, independently, halogen (including F, Cl, Br, I), CF 3 , hydroxy, N(R’)S(O) 2 R’, S(O) 2 R’, S(O) 2 N(R’) 2 , C 1-6 alkoxy, cyano, azido, C 2-6 alkynyl, C 3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C 1-6 alkyl, arylalkoxycarbonyl, carboxy, C 1-6 haloalkyl, heterocyclylalkyl, or C 1-6 hydroxyalkyl.
  • substituents each of which is, independently, halogen (including F, Cl, Br, I), CF 3 , hydroxy, N(R’)S(O) 2 R’, S(O) 2 R’, S(
  • D is , where R 6 is H, Cl, F or Br, and R 7 is H, methyl, F or Cl.
  • C is a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms, a five-membered
  • heteroaromatic ring containing one, two, or three heteroatoms which are, independently, N, O, or S; a C 4-14 bicyclic ring, alkylaryl, or alkylheteroaryl.
  • D is a C 4-14 bicyclic ring.
  • R 5 is arylalkyl, C 2-6 alkynyl, aryl, such as phenyl, heteroaryl, including six-membered heteroaromatic rings containing one, two, or three nitrogen atoms and five-membered heteroaromatic rings containing one, two, or three heteroatoms, which, independently, are N, O, or S; and a six-membered spiro-fused ring containing zero, one, or two heteroatoms which are, independently, N, O, or S.
  • the compounds for use in the invention are those having formula (A-III):
  • R 1 and R 1 ’ are as defined with respect to formula (A-I);
  • u and v are independently 0, 1, 2, 3, 4 or 5;
  • E is a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms, a five-membered heteroaromatic ring containing one, two, or three heteroatoms, where each is, independently, N, O, or S; a C 4-14 bicyclic ring, alkylheteroaryl, or alkylaryl;
  • F is a five-membered heteroaromatic ring containing one, two, or three heteroatoms which are, independently, N, O, or S, or a C 4-14 bicyclic ring;
  • R 8 is H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, or C 2-6 alkynyl;
  • R 9 is C 1-6 alkyl, C 1-6 haloalkyl, C 2-8 alkoxyalkyl, a six-membered ring or a six- membered bridged or spiro-fused ring containing zero, one, or two heteroatoms, which are independently N, O, or S, a seven-membered bridged or spiro-fused ring containing zero, one, or two heteroatoms, which are, independently, N, O, or S, a five- membered ring containing zero, one, or two heteroatoms, which are, independently, N, O, or S; a four-membered ring containing zero, one, or two heteroatoms, which are, independently, N, O, or S, or a three membered ring;
  • R 9 is optionally substituted with one or more substituents, each of which is independently halogen (including F, Cl, Br, and I), CF 3 , SF 5 , hydroxy, N(R’)S(O) 2 R’, S(O) 2 R’, S(O) 2 N(R’) 2 , C 1-6 alkoxy, C 1-6 haloalkoxy, cyano, azido, C 2-6 alkynyl, C 3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C 1-6 alkyl, cycloalkyl, arylalkoxycarbonyl, carboxy, C 1-6 haloalkyl, heterocyclylalkyl, or C 1-6 hydroxyalkyl; or is substituted with aryl, substituted aryl, heteroaryl, or substituted heteroaryl, where substituents on the substituted aryl and substituted heteroaryl are selected from the group consisting of halogen, SF
  • R 8 and R 9 can come together with the nitrogen to which they are attached to form a 6-10 membered bicyclic or bridged ring or a 3 to 8 saturated ring; such bicyclic, bridged and saturated ring moiety optionally containing one or more additional heteroatoms which, independently, are O, S or N and optionally being substituted with one or more substituents, each, independently, is halogen (including F, Cl, Br, and I), CF 3 , hydroxy, N(R’)S(O) 2 R’, S(O) 2 R’, S(O) 2 N(R’) 2 , C 1-6 alkoxy, cyano, azido, C 2-6 alkynyl, C 3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C 1-6 alkyl, arylalkoxycarbonyl, carboxy, C 1-6 haloalkyl, heterocyclylalkyl, or C 1-6 hydroxyalkyl.
  • the compounds for use in the invention are those having formula (A-IV): or an enantiomer, diastereomer or pharmaceutically accepted salt, hydrate, or solvate thereof, including a hydrate, a solvate, a pharmaceutically acceptable salt, a prodrug, or a complex thereof,
  • G is phenyl, a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms, a five-membered heteroaromatic ring containing one, two, or three heteroatoms, which are, independently, N, O, or S; a C 4-14 bicyclic ring, alkylheteroaryl, or alkylaryl;
  • H is phenyl, a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms, a six-membered non-aromatic ring optionally containing one, two, or three heteroatoms, which are, independently, N, O, or S; or a C 4-14 bicyclic ring;
  • R 1 and R 1 ’ are attached to a carbon they are, independently, hydrogen, halogen (including F, Cl, Br, and I), CF 3 , hydroxy, N(R’)S(O) 2 R’, S(O) 2 R’, S(O) 2 N(R’) 2 , C 1-6 alkoxy, cyano, C 2-6 alkynyl, C 3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C 1-6 alkyl, arylalkoxycarbonyl, carboxy, C 1-6 haloalkyl, heterocyclylalkyl, or C 1-6 hydroxyalkyl;
  • R 1 and R 1 ’ are attached to a nitrogen they are, independently, hydrogen, C 1-6 alkoxy, C 3-6 alkoxyalkyl, alkoxycarbonyl, carbonylalkyl, carbonyl aryl, C 1-6 alkyl, C 2-6 alkynyl, C 2-6 alkenyl, heterocyclylalkyl, C 1-6 hydroxyalkyl, or S(O) 2 R’;
  • Each R’ is independently H, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, aryl, heteroaryl, alkylaryl, or arylalkyl, or if two R’ reside on the same nitrogen atom they can come together to form a C 3-6 alkyl ring optionally containing a N, O, or S; wherein the R’ groups can be substituted with one or more substituents as defined above, for example, C 1-6 hydroxyalkyl, aminoalkyl, and alkoxyalkyl;
  • u and v are independently 0, 1, 2, 3, 4 or 5;
  • R 10 is H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, or C 2-6 alkynyl;
  • R 11 is C 1-6 alkyl, C 1-6 haloalkyl, C 2-8 alkoxyalkyl, C 2-6 alkenyl, C 2-6 alkynyl, aryl, heteroaryl, alkylaryl, arylalkyl, phenyl, a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms, a six-membered ring or a six-membered bridged or spiro-fused ring containing zero, one, or two heteroatoms, which are, independently, N, O, or S, a seven- membered bridged or spiro-fused ring containing zero, one, or two heteroatoms, which are, independently, N, O, or S, a five-membered heteroaromatic ring containing one, two
  • R 11 is optionally substituted with one or more substituents selected from the group consisting of halogen (including F, Cl, Br, and I), SF 5 , CF 3 , hydroxy, N(R’)S(O) 2 R’, S(O) 2 R’, S(O) 2 N(R’) 2 , C 1-6 alkoxy, C 1-6 haloalkoxy, C 2-6 alkenyl, cyano, C 2-6 alkynyl, C 3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C 1-6 alkyl, arylalkoxycarbonyl, carboxy, C 1-6 haloalkyl, heterocyclylalkyl, C 1-6 hydroxyalkyl, aryl, substituted aryl, heteroaryl, and substituted heteroaryl, where substituents on the substituted aryl and substituted heteroaryl are selected from the group consisting of halogen, SF 5 , CF 3 ,
  • alkoxycarbonylalkyl and C 1-6 alkyl
  • R 10 and R 11 can come together with the nitrogen to which they are attached to form a 6-10 membered bicyclic or bridged ring or a 3 to 8 saturated ring; such bicyclic, bridged or saturated ring moiety optionally containing one or more additional heteroatoms, which are each, independently, O, S or N, and optionally substituted with one or more substituents, each of which is, independently, halogen (including F, Cl, Br, and I), CF 3 , SF 5 , hydroxy, N(R’)S(O) 2 R’, S(O) 2 R’, S(O) 2 N(R’) 2 , C 1-6 alkoxy, cyano, azido, C 2-6 alkynyl, C 3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C 1- 6 alkyl, arylalkoxycarbonyl, carboxy, C 1-6 haloalkyl, heterocyclylalkyl, or C 1-6
  • R 1 and R 1 ’ are as defined with respect to formula (A-I);
  • u and v are independently 0, 1, 2, 3, 4 or 5;
  • I is phenyl, a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms, a five-membered heteroaromatic ring containing one, two, or three heteroatoms, which are, independently, N, O, or S, a C 4-14 bicyclic ring; alkylheteroaryl, or alkylaryl;
  • J is a five-membered heteroaromatic ring containing one, two, or three heteroatoms, which are, independently, N, O, or S, a six or seven-membered ring or a six or seven-membered bridged or spiro-fused ring containing zero, one, or two heteroatoms which are, independently, N, O, or S, a five-membered ring containing zero, one, or two heteroatoms which are, independently, N, O, or S; or a four- membered ring containing zero, one, or two heteroatoms which are, independently, N, O, or S,
  • R 12 is H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, or C 2-6 alkynyl;
  • R 13 is C 2-6 alkenyl, C 2-6 alkynyl, aryl, including phenyl, heteroaryl, including six- membered heteroaromatic rings containing one, two, or three nitrogen atoms and five-membered heteroaromatic rings containing one, two, or three heteroatoms, which are, independently, N, O, or S; alkylaryl, arylalkyl, a C 4-14 bicyclic ring; a six- membered bridged or spiro-fused ring containing zero, one, or two heteroatoms which are, independently, N, O, or S,
  • R 13 is optionally substituted with one or more substituents each independently selected from the group consisting of hydrogen, halogen (F, Cl, Br, I), CF 3 , SF 5 , hydroxy, N(R’)S(O) 2 R’, S(O) 2 R’, S(O) 2 N(R’) 2 , C(O)R’, C 1-6 alkoxy, C 1-6 haloalkoxy, cyano, azido, C 2-6 alkynyl, C 3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C 1- 6 alkyl, cycloalkyl, arylalkoxycarbonyl, carboxyl, haloalkyl, heterocyclylalkyl, or C 1-6 hydroxyalkyl; or is optionally substituted with aryl, substituted aryl, heteroaryl, and substituted heteroaryl, where substituents on the substituted aryl and substituted heteroaryl are selected from the
  • R 12 and R 13 together with the nitrogen to which they are attached form a 3 to 4 membered ring optionally substituted with one or more substituents each independently selected from the group consisting of hydrogen, halogen (F, Cl, Br, I), CF 3 , hydroxy, N(R’)S(O) 2 R’, S(O) 2 R’, S(O) 2 N(R’) 2 , C 1-6 alkoxy, cyano, azido, C 2-6 alkynyl, C 3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C 1-6 alkyl, arylalkoxycarbonyl, carboxy, C 1-6 haloalkyl, heterocyclylalkyl, and C 1-6 hydroxyalkyl;
  • the compounds for use in the invention are those having formula (A-VI):
  • R 1 and R 1 ’ are as defined with respect to formula (A-I);
  • u and v are independently 0, 1, 2, 3, 4 or 5;
  • K is a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms, a five-membered heteroaromatic ring containing one, two, or three heteroatoms which are, independently, N, O, or S; a C 4-14 bicyclic ring, alkylheteroaryl, or alkylaryl;
  • L is a five-membered heteroaromatic ring containing one, two, or three heteroatoms which are, independently, N, O, or S, a six or seven -membered ring or a six or seven-membered bridged or spiro-fused ring containing zero, one, or two heteroatoms which are, independently, N, O, or S, a five-membered ring containing zero, one, or two heteroatoms which are, independently, N, O, or S; a four-membered ring containing zero, one, or two heteroatoms which are, independently, N, O, or S, or a C 4-14 bicyclic ring;
  • R 14 is H, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, or C 2-6 alkynyl;
  • R 15 is C 1-6 alkyl, C 1-6 haloalkyl, C 2-8 alkoxyalkyl, a six-membered ring or a six- membered bridged or spiro-fused ring containing zero, one, or two heteroatoms which are, independently, N, O, or S, a seven-membered bridged or spiro-fused ring containing zero, one, or two heteroatoms which are, independently, N, O, or S, a five- membered ring containing zero, one, or two heteroatoms which are, independently, N, O, or S; a four-membered ring containing zero, one, or two heteroatoms which are, independently, N, O, or S;
  • R 15 is optionally substituted with one or more substituents which are, independently, halogen (F, Cl, Br, I), SF5, CF3, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O) 2 N(R’) 2 , C 1-6 alkoxy, C 1-6 haloalkoxy, cyano, azido, C 2-6 alkynyl, C 3-6 alkoxyalkyl, alkoxycarbonyl,
  • substituents which are, independently, halogen (F, Cl, Br, I), SF5, CF3, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O) 2 N(R’) 2 , C 1-6 alkoxy, C 1-6 haloalkoxy, cyano, azido, C 2-6 alkynyl, C 3-6 alkoxyalkyl, alkoxycarbonyl,
  • alkoxycarbonylalkyl C 1-6 alkyl, cycloalkyl, arylalkoxycarbonyl, carboxy, C 1-6 haloalkyl, heterocyclylalkyl, or C 1-6 hydroxyalkyl; or is optionally substituted with aryl, substituted aryl, heteroaryl, or substituted heteroaryl, where substituents on the substituted aryl and substituted heteroaryl are selected from the group consisting of halogen, SF 5 , CF 3 , hydroxy, N(R’)S(O) 2 R’, S(O) 2 R’, S(O) 2 N(R’) 2 , C(O)R’, C 1-6 alkoxy, cyano, azido, C 2-6 alkynyl, C 3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, and C 1-6 alkyl;
  • R 14 and R 15 can come together with the nitrogen to which they are attached to form a 6-10 membered bicyclic or bridged ring or a 3 to 8 saturated ring; such bicyclic, bridged and saturated ring moiety optionally containing one or more additional heteroatoms which are, independently, O, S or N, and optionally being substituted with one or more substituents each independently selected from the group consisting of halogen (including F, Cl, Br, and I), SF 5 , CF 3 , hydroxy, N(R’)S(O) 2 R’, S(O) 2 R’, S(O) 2 N(R’) 2 , C 1-6 alkoxy, cyano, azido, C 2-6 alkynyl, C 3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C 1-6 alkyl, arylalkoxycarbonyl, carboxy, C 1-6 haloalkyl, heterocyclylalkyl, and C 1-6
  • R 1 and R 1 ’ are as defined with respect to formula (A-I);
  • u and v are independently 0, 1, 2, 3, 4 or 5;
  • M is phenyl, a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms, a five-membered heteroaromatic ring containing one, two, or three heteroatoms which are, independently, N, O, or S, a C 4-14 bicyclic ring, alkylheteroaryl, or alkylaryl;
  • N is phenyl, a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms, a five-membered heteroaromatic ring containing one, two, or three heteroatoms independently N, O, or S, a six or seven-membered ring or a six or seven- membered bridged or spiro-fused ring containing zero, one, or two heteroatoms which are, independently, N, O, or S, a five-membered ring containing zero, one, or two heteroatoms which are, independently, N, O, or S; a four-membered ring containing zero, one, or two heteroatoms which are, independently, N, O, or S; or a C 4-14 bicyclic ring,
  • R 16 is C 1-6 alkyl, C 1-6 haloalkyl, C 2-8 alkoxyalkyl, C 2-6 alkenyl, C 2-6 alkynyl, aryl, such as phenyl, heteroaryl, such as a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms or a five-membered heteroaromatic ring containing one, two, or three heteroatoms which are, independently, N, O, or S; a six- membered ring or a six-membered bridged or spiro-fused ring containing zero, one, or two heteroatoms which are, independently, N, O, or S, a five-membered ring containing zero, one, or two heteroatoms which are, independently, N, O, or S; a four- membered ring containing zero, one, or two heteroatoms which are, independently, N, O, or S; alkylaryl, arylalkyl,
  • R 16 is optionally substituted with one or more substituents selected from the group consisting of halogen (including F, Cl, Br, and I), SF 5 , CF 3 , hydroxy, N(R’)S(O) 2 R’, S(O) 2 R’, S(O) 2 N(R’) 2 , C 1-6 alkoxy, C 1-6 haloalkoxy, C 2-6 alkenyl, cyano, C 2-6 alkynyl, C 3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C 1-6 alkyl, arylalkoxycarbonyl, carboxy, C1-6 haloalkyl, heterocyclylalkyl, C1-6 hydroxyalkyl, aryl, substituted aryl, heteroaryl, and substituted heteroaryl, where substituents on the substituted aryl and substituted heteroaryl are selected from the group consisting of halogen, SF5, CF3, hydroxy, N(
  • Particularly preferred compounds include:
  • a particularly preferred compound has the formula:
  • pharmaceutically acceptable salt, an enantiomer, or a diastereomer thereof may be determined by routine experimentation, but can be in range of 7 to 600 mg, preferably from 70 to 600 mg per unit dosage form.
  • Suitable processes for synthesizing these compounds can be synthesized by any method or synthetic scheme known in the art or to be discovered.
  • the compounds can be prepared according to Scheme 1, shown below. Examples of specific synthetic compounds are described in WO 2017156255.
  • the compounds for use in the invention are those having formula (B- I):
  • R 1 is C 1-6 alkyl optionally substituted with 1 to 3 R 1A , C 3-8 cycloalkyl optionally substituted with 1 to 4 R 1B , or 3 to 8 membered monocyclic or bicyclic heterocyclyl having 1 to 3 heteroatoms selected from N, O, and S, optionally substituted with 1 to 3 R 1C ;
  • each R 1A is independently halogen, -OH, -CN, -C 1-2 haloalkyl, -C(O)NR X R Y , C 6-10 aryl optionally substituted with 1 to 3 R 1D , or a 5 to 8 membered heteroaryl having 1 to 3 heteroatoms selected from N, O, and S, optionally substituted with 1 to 3 R 1D , provided no more than 1 R 1A is C 6-10 aryl optionally substituted with 1 to 3 R 1D or 5 to 8 membered heteroaryl having 1 to 3 heteroatoms selected from N, O, and S optionally substituted with 1 to 3 R 1D ;
  • each R 1B is independently -CN, halogen, C 1-6 alkyl optionally substituted with 1 to 3 -OH or -NR a R b , C 2-4 alkynyl, C 1-4 alkoxy, C 1-2 haloalkyl, C 3-6 cycloalkyl, -C(O)NR X R Y , or a 5 to 8 membered heteroaryl having 1 to 3 heteroatoms selected from N, O, and S optionally substituted with 1 to 3 R 1D , provided no more than 1 R 1B is C 3-6 cycloalkyl or 5 to 8 membered heteroaryl having 1 to 3 heteroatoms selected from N, O, and S optionally substituted with 1 to 3 R 1D ; each R 1C is independently C 1-6 alkyl, oxo, C 1-4 haloalkyl, -C(O)H, -C(O)C 1-4 alkyl, - C(O)OC 1-4 alkyl, or a 5
  • each R Y is independently -H or C 1 alkyl optionally substituted with 1 to 3 R Z ;
  • R X and R Y are taken together to form a 3 to 8 membered monocyclic or bicyclic heterocyclyl having 1 to 3 heteroatoms selected from N, O, and S, optionally substituted with 1 to 3 R Z ;
  • each R Z is independently halogen, methyl, ethyl, oxo, -OH, -S(O) 2 C 1-3 alkyl, or 3 to 8 membered monocyclic or bicyclic heterocyclyl having 1 to 3 heteroatoms selected from N, O, and S;
  • each R a is -H, C 1-3 alkyl, or a 3 to 8 membered monocyclic or bicyclic heterocyclyl having 1 to 3 heteroatoms selected from N, O, and S, optionally substituted with 1 to 3 R Z ;
  • each R b is -H or C 1-3 alkyl
  • the moiety is a pyrrolidine or a 5-7 membered bicyclic heterocycle having one nitrogen, optionally substituted with 1 to 6 R 2 groups; wherein each R 2 is independently halogen, C 1-3 alkyl, -OH, or -OC 1-3 alkyl;
  • R 3 is -H, halogen, or C 1-4 alkyl
  • R 4 is C 6-10 aryl optionally substituted with 1 to 5 R 4A , or 5 to 12 membered heteroaryl having 1 to 3 heteroatoms selected from N, O, and S, optionally substituted with 1 to 4 R 4B ; and each R 1D , R 4A , and R 4B are independently -CN, halogen, C 1-4 alkyl, -OC 1-4 alkyl, -OC 1- 4 haloalkyl, or C 1-4 haloalkyl.
  • R 4 is phenyl optionally substituted with 1 to 5 R 4A or pyridinyl optionally substituted with 1 to 4 R 4B .
  • R 4 is phenyl optionally substituted with 1 to 3 R 4A .
  • each R 4A is independently CI, F, -CF 3 , -CHF 2 ,-CH 3 , -OCF 3 , - OCF 2 H, or - CN.
  • R 3 is -Cl or -CH 3 , preferably -CH 3 .
  • Exemplary compounds having formula (B-I) have the following structures, where the substituents are as described previously or in WO 2018039531 and US 20180161307:
  • the effective amount of the compound having formula (B-I) or a pharmaceutically acceptable salt thereof may be determined by routine experimentation, but can be in range of from about 0.00001 mg/kg body weight per day to about 10 mg/kg body weight per day, such as from about 0.0001 mg/kg body weight per day to about 10 mg/kg body weight per day, or such as from about 0.001 mg/kg body weight per day to about 1 mg/kg body weight per day, or such as from about 0.01 mg/kg body weight per day to about 1 mg/kg body weight per day, or such as from about 0.05 mg/kg body weight per day to about 0.5 mg/kg body weight per day, or such as from about 0.3 mg to about 30 mg per day, or such as from about 30 mg to about 300 mg per day.
  • Suitable processes for synthesizing these compounds can be synthesized by any method or synthetic scheme known in the art or to be discovered.
  • the compounds can be prepared according to Schemes 1 and 2, shown below. Examples of specific synthetic compounds are described in WO 2018039531 and US 20180161307.
  • Al can be converted to A2 by treatment with an appropriate alkyl halide such as (3- bromopropoxy)(tert-butyl)diphenylsilane in the presence of a suitable catalyst such as palladium dichloride bis(acetonitrile.
  • A2 can be converted to A3 through a deprotection with a suitable reagent such as tetrabutylammonium fluoride and further halogenated to A4 with a suitable reagents such as carbon tetrabromide and triphenylphosphine.
  • Cyclization to A5 can be effected with an appropriate radical initiator such as 2,2'-azobis(2-methylpropionitrile) in the presence other reagents such as tributyl tin hydride.
  • Ester hydrolysis with a suitable reagent such as lithium hydroxide followed by amide formation via treatment with an appropriate coupling reagent such as HATU and the appropriate aniline or by conversion to the acid chloride with a reagent such a thionyl chloride or oxalyl chloride followed by treatment with the appropriate aniline gives A6.
  • the aniline may be varied based on the R 4 groups disclosued herein.
  • Formation of A7 can be effected by treatment with a suitable reagent such as oxalyl chloride or ethyl 2-chloro-2-oxoacetate and may or may not require the addition of catalyst such as aluminum chloride.
  • Hydrolysis to A8 via a suitable reagent such as lithium hydroxide is followed by preparation of A9 by the coupling of the appropriate amine such as l,l,l- trifluoropropan-2-amine in the presence of a coupling reagent such as HATU.
  • the amine may be varied for particular R 1 groups disclosed herein.
  • Bl can be converted to B2 by treatment with methyl 2-chloro-2-oxoacetate. Conversion to B3 can be carried out with an appropriate hydroxide reagent such as lithium hydroxide or in some cases with hydrogen gas and a suitable catalyst if the ester is a benzyl ester. B3 can be converted to B4 by treatment with a suitable reagent such as N,N'-diisopropylcarbodiimide and alkyne reagent B3B at elevated temperatures. Alternatively, B3 can be treated with oxalyl chloride, worked up and treated with B3B and a suitable base such as 2,6-di-tertbutylpyridine to give B4.
  • a suitable reagent such as N,N'-diisopropylcarbodiimide and alkyne reagent B3B at elevated temperatures.
  • B3 can be treated with oxalyl chloride, worked up and treated with B3B and a suitable base such as 2,6-
  • Hydrolysis to B5 can be effected with a suitable reagent such as lithium hydroxide.
  • Conversion to B6 occurs by addition of an appropriate amine (e.g. R 1 - NH2 ) and an amide coupling reagent such as HATU.
  • B6 can be converted to desired B7 by treatment with a suitable reagent such as lithium hydroxide at elevated temperatures followed a second amide coupling with an appropriate aniline or heteroaryl amine (e.g R 4 -NH2) and a coupling reagent such as HATU.
  • B5 can be treated with an aniline or heteroaryl amine in the presence of a reagent such as Lithium bis(trimethylsilyl)amide to give B6B followed by treatment with an appropriate amine and a coupling reagent such as HATU to give desired B7.
  • a reagent such as Lithium bis(trimethylsilyl)amide
  • the application also relates to compositions, therapeutic combinations, more particularly kits, and vaccines comprising one or more HBV antigens, polynucleotides, and/or vectors encoding one or more HBV antigens according to the application.
  • HBV antigens any of the HBV antigens, polynucleotides (including RNA and DNA), and/or vectors of the application described herein can be used in the compositions, therapeutic combinations or kits, and vaccines of the application.
  • a composition comprises an isolated or non-naturally occurring nucleic acid molecule (DNA or RNA) comprising polynucleotide sequence encoding a truncated HBV core antigen consisting of an amino acid sequence that is at least 90% identical to SEQ ID NO: 2 or SEQ ID NO: 4, or an HBV polymerase antigen comprising an amino acid sequence that is at least 90% identical to SEQ ID NO: 7, a vector comprising the isolated or non- naturally occurring nucleic acid molecule, and/or an isolated or non-naturally occurring polypeptide encoded by the isolated or non-naturally occurring nucleic acid molecule.
  • DNA or RNA isolated or non-naturally occurring nucleic acid molecule
  • polynucleotide sequence encoding a truncated HBV core antigen consisting of an amino acid sequence that is at least 90% identical to SEQ ID NO: 2 or SEQ ID NO: 4, or an HBV polymerase antigen comprising an amino acid sequence that is at least 90% identical to SEQ ID NO: 7,
  • a composition comprises an isolated or non-naturally occurring nucleic acid molecule (DNA or RNA) comprising a polynucleotide sequence encoding an HBV Pol antigen comprising an amino acid sequence that is at least 90% identical to SEQ ID NO: 7, preferably 100% identical to SEQ ID NO: 7.
  • a composition comprises an isolated or non-naturally occurring nucleic acid molecule (DNA or RNA) encoding a truncated HBV core antigen consisting of an amino acid sequence that is at least 90% identical to SEQ ID NO: 2 or SEQ ID NO: 4, preferably 100% identical to SEQ ID NO: 2 or SEQ ID NO: 4.
  • a composition comprises an isolated or non-naturally occurring nucleic acid molecule (DNA or RNA) comprising a polynucleotide sequence encoding a truncated HBV core antigen consisting of an amino acid sequence that is at least 90% identical to SEQ ID NO: 2 or SEQ ID NO: 4, preferably 100% identical to SEQ ID NO: 2 or SEQ ID NO: 4; and an isolated or non-naturally occurring nucleic acid molecule (DNA or RNA) comprising a polynucleotide sequence encoding an HBV Pol antigen comprising an amino acid sequence that is at least 90% identical to SEQ ID NO: 7, preferably 100% identical to SEQ ID NO: 7.
  • DNA or RNA isolated or non-naturally occurring nucleic acid molecule
  • the coding sequences for the truncated HBV core antigen and the HBV Pol antigen can be present in the same isolated or non-naturally occurring nucleic acid molecule (DNA or RNA), or in two different isolated or non-naturally occurring nucleic acid molecules (DNA or RNA).
  • a composition comprises a vector, preferably a DNA plasmid or a viral vector (such as an adenoviral vector) comprising a polynucleotide encoding a truncated HBV core antigen consisting of an amino acid sequence that is at least 90% identical to SEQ ID NO: 2 or SEQ ID NO: 4, preferably 100% identical to SEQ ID NO: 2 or SEQ ID NO: 4.
  • a vector preferably a DNA plasmid or a viral vector (such as an adenoviral vector) comprising a polynucleotide encoding a truncated HBV core antigen consisting of an amino acid sequence that is at least 90% identical to SEQ ID NO: 2 or SEQ ID NO: 4, preferably 100% identical to SEQ ID NO: 2 or SEQ ID NO: 4.
  • a composition comprises a vector, preferably a DNA plasmid or a viral vector (such as an adenoviral vector), comprising a polynucleotide encoding an HBV Pol antigen comprising an amino acid sequence that is at least 90% identical to SEQ ID NO: 7, preferably 100% identical to SEQ ID NO: 7.
  • a composition comprises a vector, preferably a DNA plasmid or a viral vector (such as an adenoviral vector), comprising a polynucleotide encoding a truncated HBV core antigen consisting of an amino acid sequence that is at least 90% identical to SEQ ID NO: 2 or SEQ ID NO: 4, preferably 100% identical to SEQ ID NO: 2 or SEQ ID NO: 4; and a vector, preferably a DNA plasmid or a viral vector (such as an adenoviral vector), comprising a polynucleotide encoding an HBV Pol antigen comprising an amino acid sequence that is at least 90% identical to SEQ ID NO: 7, preferably 100% identical to SEQ ID NO: 7.
  • the vector comprising the coding sequence for the truncated HBV core antigen and the vector comprising the coding sequence for the HBV Pol antigen can be the same vector, or two different vectors.
  • a composition comprises a vector, preferably a DNA plasmid or a viral vector (such as an adenoviral vector), comprising a polynucleotide encoding a fusion protein comprising a truncated HBV core antigen consisting of an amino acid sequence that is at least 90% identical to SEQ ID NO: 2 or SEQ ID NO: 4, preferably 100% identical to SEQ ID NO: 2 or SEQ ID NO: 4, operably linked to an HBV Pol antigen comprising an amino acid sequence that is at least 90% identical to SEQ ID NO: 7, preferably 100% identical to SEQ ID NO: 7, or vice versa.
  • a vector preferably a DNA plasmid or a viral vector (such as an adenoviral vector), comprising a polynucleotide encoding a fusion protein comprising a truncated HBV core antigen consisting of an amino acid sequence that is at least 90% identical to SEQ ID NO: 2 or SEQ ID NO: 4, preferably 100% identical to SEQ
  • the fusion protein further comprises a linker that operably links the truncated HBV core antigen to the HBV Pol antigen, or vice versa.
  • the linker has the amino acid sequence of (AlaGly)n, wherein n is an integer of 2 to 5.
  • a composition comprises an isolated or non-naturally occurring truncated HBV core antigen consisting of an amino acid sequence that is at least 90% identical to SEQ ID NO: 2 or SEQ ID NO: 4, preferably 100% identical to SEQ ID NO: 2 or SEQ ID NO: 4.
  • a composition comprises an isolated or non-naturally occurring HBV Pol antigen comprising an amino acid sequence that is at least 90% identical to SEQ ID NO: 7, preferably 100% identical to SEQ ID NO: 7.
  • a composition comprises an isolated or non-naturally occurring truncated HBV core antigen consisting of an amino acid sequence that is at least 90% identical to SEQ ID NO: 2 or SEQ ID NO: 4, preferably 100% identical to SEQ ID NO: 2 or SEQ ID NO: 4; and an isolated or non-naturally occurring HBV Pol antigen comprising an amino acid sequence that is at least 90% identical to SEQ ID NO: 7, preferably 100% identical to SEQ ID NO: 7.
  • a composition comprises an isolated or non-naturally occurring fusion protein comprising a truncated HBV core antigen consisting of an amino acid sequence that is at least 90% identical to SEQ ID NO: 2 or SEQ ID NO: 14, preferably 100% identical to SEQ ID NO: 2 or SEQ ID NO: 4, operably linked to an HBV Pol antigen comprising an amino acid sequence that is at least 90% identical to SEQ ID NO: 7, preferably 100% identical to SEQ ID NO: 7, or vice versa.
  • the fusion protein further comprises a linker that operably links the truncated HBV core antigen to the HBV Pol antigen, or vice versa.
  • the linker has the amino acid sequence of (AlaGly)n, wherein n is an integer of 2 to 5.
  • the application also relates to a therapeutic combination or a kit comprising
  • polynucleotides expressing a truncated HBV core antigen and an HBV pol antigen according to embodiments of the application. Any polynucleotides and/or vectors encoding HBV core and pol antigens of the application described herein can be used in the therapeutic combinations or kits of the application.
  • a therapeutic combination or kit for use in treating an HBV infection in a subject in need thereof comprises:
  • a truncated HBV core antigen consisting of an amino acid sequence that is at least 95% identical to SEQ ID NO: 2, and
  • a first non-naturally occurring nucleic acid molecule comprising a first polynucleotide sequence encoding the truncated HBV core antigen
  • HBV polymerase antigen having an amino acid sequence that is at least 90% identical to SEQ ID NO: 7, wherein the HBV polymerase antigen does not have reverse transcriptase activity and RNase H activity, and
  • a second non-naturally occurring nucleic acid molecule comprising a second polynucleotide sequence encoding the HBV polymerase antigen
  • a therapeutic combination or kit comprises: i) a first non-naturally occurring nucleic acid molecule comprising a first polynucleotide sequence encoding a truncated HBV core antigen consisting of an amino acid sequence that is at least 95% identical to SEQ ID NO: 2; ii) a second non-naturally occurring nucleic acid molecule comprising a second polynucleotide sequence encoding an HBV polymerase antigen having an amino acid sequence that is at least 90% identical to SEQ ID NO: 7, wherein the HBV polymerase antigen does not have reverse transcriptase activity and RNase H activity; and iii) a capsid assembly modulator.
  • the polynucleotides in a vaccine combination or kit can be linked or separate, such that the HBV antigens expressed from such polynucleotides are fused together or produced as separate proteins, whether expressed from the same or different polynucleotides.
  • the first and second polynucleotides are present in separate vectors, e.g., DNA plasmids or viral vectors, used in combination either in the same or separate compositions, such that the expressed proteins are also separate proteins, but used in combination.
  • the HBV antigens encoded by the first and second polynucleotides can be expressed from the same vector, such that an HBV core-pol fusion antigen is produced.
  • the core and pol antigens can be joined or fused together by a short linker.
  • the HBV antigens encoded by the first and second polynucleotides can be expressed independently from a single vector using a using a ribosomal slippage site (also known as cis-hydrolase site) between the core and pol antigen coding sequences.
  • a ribosomal slippage site also known as cis-hydrolase site
  • This strategy results in a bicistronic expression vector in which individual core and pol antigens are produced from a single mRNA transcript.
  • the core and pol antigens produced from such a bicistronic expression vector can have additional N or C-terminal residues, depending upon the ordering of the coding sequences on the mRNA transcript.
  • ribosomal slippage sites examples include, but are not limited to, the FA2 slippage site from foot-and-mouth disease virus (FMDV).
  • FMDV foot-and-mouth disease virus
  • HBV antigens encoded by the first and second polynucleotides can be expressed independently from two separate vectors, one encoding the HBV core antigen and one encoding the HBV pol antigen.
  • the first and second polynucleotides are present in separate vectors, e.g., DNA plasmids or viral vectors.
  • the separate vectors are present in the same composition.
  • a therapeutic combination or kit comprises a first polynucleotide present in a first vector, a second polynucleotide present in a second vector.
  • the first and second vectors can be the same or different.
  • the vectors are DNA plasmids.
  • the first vector is a first DNA plasmid
  • the second vector is a second DNA plasmid.
  • Each of the first and second DNA plasmids comprises an origin of replication, preferably pUC ORI of SEQ ID NO: 21, and an antibiotic resistance cassette, preferably comprising a codon optimized Kanr gene having a polynucleotide sequence that is at least 90% identical to SEQ ID NO: 23, preferably under control of a bla promoter, for instance the bla promoter shown in SEQ ID NO: 24.
  • Each of the first and second DNA plasmids independently further comprises at least one of a promoter sequence, enhancer sequence, and a polynucleotide sequence encoding a signal peptide sequence operably linked to the first polynucleotide sequence or the second polynucleotide sequence.
  • each of the first and second DNA plasmids comprises an upstream sequence operably linked to the first polynucleotide or the second polynucleotide, wherein the upstream sequence comprises, from 5’ end to 3’ end, a promoter sequence of SEQ ID NO: 18 or 19, an enhancer sequence, and a polynucleotide sequence encoding a signal peptide sequence having the amino acid sequence of SEQ ID NO: 9 or 15.
  • Each of the first and second DNA plasmids can also comprise a polyadenylation signal located downstream of the coding sequence of the HBV antigen, such as the bGH polyadenylation signal of SEQ ID NO: 20.
  • the first vector is a viral vector and the second vector is a viral vector.
  • each of the viral vectors is an adenoviral vector, more preferably an Ad26 or Ad35 vector, comprising an expression cassette including the
  • polynucleotide encoding an HBV pol antigen or an truncated HBV core antigen of the application; an upstream sequence operably linked to the polynucleotide encoding the HBV antigen comprising, from 5’ end to 3’ end, a promoter sequence, preferably a CMV promoter sequence of SEQ ID NO: 19, an enhancer sequence, preferably an ApoAI gene fragment sequence of SEQ ID NO: 12, and a polynucleotide sequence encoding a signal peptide sequence, preferably an immunoglobulin secretion signal having the amino acid sequence of SEQ ID NO: 15; and a downstream sequence operably linked to the polynucleotide encoding the HBV antigen comprising a polyadenylation signal, preferably a SV40 polyadenylation signal of SEQ ID NO: 13.
  • the first and second polynucleotides are present in a single vector, e.g., DNA plasmid or viral vector.
  • the single vector is an adenoviral vector, more preferably an Ad26 vector, comprising an expression cassette including a polynucleotide encoding an HBV pol antigen and a truncated HBV core antigen of the application, preferably encoding an HBV pol antigen and a truncated HBV core antigen of the application as a fusion protein; an upstream sequence operably linked to the polynucleotide encoding the HBV pol and truncated core antigens comprising, from 5’ end to 3’ end, a promoter sequence, preferably a CMV promoter sequence of SEQ ID NO: 19, an enhancer sequence, preferably an ApoAI gene fragment sequence of SEQ ID NO: 12, and a polynucleotide sequence encoding a signal peptide sequence, preferably an immuno
  • a therapeutic combination of the application comprises a first vector, such as a DNA plasmid or viral vector, and a second vector, such as a DNA plasmid or viral vector
  • the amount of each of the first and second vectors is not particularly limited.
  • the first DNA plasmid and the second DNA plasmid can be present in a ratio of 10:1 to 1:10, by weight, such as 10:1, 9:1, 8:1, 7:1, 6:1, 5:1, 4:1, 3:1, 2:1, 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, or 1:10, by weight.
  • the first and second DNA plasmids are present in a ratio of 1:1, by weight.
  • the therapeutic combination of the application can further comprise a third vector encoding a third active agent useful for treating an HBV infection.
  • compositions and therapeutic combinations of the application can comprise additional polynucleotides or vectors encoding additional HBV antigens and/or additional HBV antigens or immunogenic fragments thereof, such as an HBsAg, an HBV L protein or HBV envelope protein, or a polynucleotide sequence encoding thereof.
  • additional HBV antigens and/or additional HBV antigens or immunogenic fragments thereof such as an HBsAg, an HBV L protein or HBV envelope protein, or a polynucleotide sequence encoding thereof.
  • the compositions and therapeutic combinations of the application do not comprise certain antigens.
  • composition or therapeutic combination or kit of the application does not comprise a HBsAg or a polynucleotide sequence encoding the HBsAg.
  • composition or therapeutic combination or kit of the application does not comprise an HBV L protein or a polynucleotide sequence encoding the HBV L protein.
  • composition or therapeutic combination of the application does not comprise an HBV envelope protein or a polynucleotide sequence encoding the HBV envelope protein.
  • compositions and therapeutic combinations of the application can also comprise a pharmaceutically acceptable carrier.
  • a pharmaceutically acceptable carrier is non-toxic and should not interfere with the efficacy of the active ingredient.
  • Pharmaceutically acceptable carriers can include one or more excipients such as binders, disintegrants, swelling agents, suspending agents, emulsifying agents, wetting agents, lubricants, flavorants, sweeteners, preservatives, dyes, solubilizers and coatings.
  • Pharmaceutically acceptable carriers can include vehicles, such as lipid nanoparticles (LNPs).
  • suitable carriers and additives include water, glycols, oils, alcohols, preservatives, coloring agents and the like.
  • suitable carriers and additives include starches, sugars, diluents, granulating agents, lubricants, binders, disintegrating agents and the like.
  • the aqueous solution/suspension can comprise water, glycols, oils, emollients, stabilizers, wetting agents, preservatives, aromatics, flavors, and the like as suitable carriers and additives.
  • compositions and therapeutic combinations of the application can be formulated in any matter suitable for administration to a subject to facilitate administration and improve efficacy, including, but not limited to, oral (enteral) administration and parenteral injections.
  • the parenteral injections include intravenous injection or infusion, subcutaneous injection, intradermal injection, and intramuscular injection.
  • Compositions of the application can also be formulated for other routes of administration including transmucosal, ocular, rectal, long acting implantation, sublingual administration, under the tongue, from oral mucosa bypassing the portal circulation, inhalation, or intranasal.
  • compositions and therapeutic combinations of the application are formulated for parental injection, preferably subcutaneous, intradermal injection, or intramuscular injection, more preferably intramuscular injection.
  • compositions and therapeutic combinations for administration will typically comprise a buffered solution in a pharmaceutically acceptable carrier, e.g., an aqueous carrier such as buffered saline and the like, e.g., phosphate buffered saline (PBS).
  • a pharmaceutically acceptable carrier e.g., an aqueous carrier such as buffered saline and the like, e.g., phosphate buffered saline (PBS).
  • PBS phosphate buffered saline
  • a composition or therapeutic combination of the application comprising plasmid DNA can contain phosphate buffered saline (PBS) as the pharmaceutically acceptable carrier.
  • the plasmid DNA can be present in a concentration of, e.g., 0.5 mg/mL to 5 mg/mL, such as 0.5 mg/mL 1, mg/mL, 2 mg/mL, 3 mg/mL, 4 mg/mL, or 5 mg/mL, preferably at 1 mg/mL.
  • Compositions and therapeutic combinations of the application can be formulated as a vaccine (also referred to as an“immunogenic composition”) according to methods well known in the art. Such compositions can include adjuvants to enhance immune responses.
  • the optimal ratios of each component in the formulation can be determined by techniques well known to those skilled in the art in view of the present disclosure.
  • a composition or therapeutic combination is a DNA vaccine.
  • DNA vaccines typically comprise bacterial plasmids containing a polynucleotide encoding an antigen of interest under control of a strong eukaryotic promoter. Once the plasmids are delivered to the cell cytoplasm of the host, the encoded antigen is produced and processed endogenously. The resulting antigen typically induces both humoral and cell-medicated immune responses.
  • DNA vaccines are advantageous at least because they offer improved safety, are temperature stable, can be easily adapted to express antigenic variants, and are simple to produce. Any of the DNA plasmids of the application can be used to prepare such a DNA vaccine.
  • RNA vaccines typically comprise at least one single-stranded RNA molecule encoding an antigen of interest, e.g., a fusion protein or HBV antigen according to the application. Once the RNA is delivered to the cell cytoplasm of the host, the encoded antigen is produced and processed endogenously, inducing both humoral and cell-mediated immune responses, similar to a DNA vaccine.
  • the RNA sequence can be codon optimized to improve translation efficiency.
  • RNA molecule can be modified by any method known in the art in view of the present disclosure to enhance stability and/or translation, such by adding a polyA tail, e.g., of at least 30 adenosine residues; and/or capping the 5-end with a modified ribonucleotide, e.g., 7-methylguanosine cap, which can be incorporated during RNA synthesis or enzymatically engineered after RNA transcription.
  • An RNA vaccine can also be self-replicating RNA vaccine developed from an alphavirus expression vector.
  • Self-replicating RNA vaccines comprise a replicase RNA molecule derived from a virus belonging to the alphavirus family with a subgenomic promoter that controls replication of the fusion protein or HBV antigen RNA followed by an artificial poly A tail located downstream of the replicase.
  • a further adjuvant can be included in a composition or therapeutic combination of the application, or co-administered with a composition or therapeutic combination of the application.
  • another adjuvant is optional, and can further enhance immune responses when the composition is used for vaccination purposes.
  • Other adjuvants suitable for co-administration or inclusion in compositions in accordance with the application should preferably be ones that are potentially safe, well tolerated and effective in humans.
  • An adjuvant can be a small molecule or antibody including, but not limited to, immune checkpoint inhibitors (e.g., anti-PD1, anti-TIM-3, etc.), toll-like receptor agonists (e.g., TLR7 agonists and/or TLR8 agonists), RIG-1 agonists, IL-15 superagonists (Altor Bioscience), mutant IRF3 and IRF7 genetic adjuvants, STING agonists (Aduro), FLT3L genetic adjuvant, and IL-7-hyFc.
  • immune checkpoint inhibitors e.g., anti-PD1, anti-TIM-3, etc.
  • toll-like receptor agonists e.g., TLR7 agonists and/or TLR8 agonists
  • RIG-1 agonists e.g., RIG-1 agonists
  • IL-15 superagonists e.g., IL-15 superagonists (Altor Bioscience)
  • adjuvants can e.g., be chosen from among the following anti-HBV agents: HBV DNA polymerase inhibitors; Immunomodulators; Toll-like receptor 7 modulators; Toll-like receptor 8 modulators; Toll-like receptor 3 modulators; Interferon alpha receptor ligands;
  • 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-HBV antisense oligonucleotides; short interfering RNAs (siRNA), more particularly anti- HBV siRNA; Endonuclease modulators; Inhibitors of ribonucleotide reductase; Hepatitis B virus E antigen inhibitors; HBV antibodies targeting the surface antigens of the hepatitis B virus; HBV antibodies; CCR 2 chemokine antagonists; Thymosin agonists; Cytokines, such as IL12; Capsid Assembly Modulators, Nucleoprotein inhibitors (HBV core or capsid protein inhibitors); Nucleic Acid Polymers (NAPs); Stimulators of retinoi
  • cccDNA inhibitors include PD-L1 inhibitors, PD-1 inhibitors, TIM-3 inhibitors, TIGIT inhibitors, Lag3 inhibitors, CTLA-4 inhibitors; Agonists of co- stimulatory receptors that are expressed on immune cells (more particularly T cells), such as CD27 and CD28; BTK inhibitors; Other drugs for treating HBV; IDO inhibitors; Arginase inhibitors; and KDM5 inhibitors.
  • immune checkpoint inhibitors such as PD-L1 inhibitors, PD-1 inhibitors, TIM-3 inhibitors, TIGIT inhibitors, Lag3 inhibitors, CTLA-4 inhibitors
  • Agonists of co- stimulatory receptors that are expressed on immune cells include CD27 and CD28; BTK inhibitors; Other drugs for treating HBV; IDO inhibitors; Arginase inhibitors; and KDM5 inhibitors.
  • each of the first and second non-naturally occurring nucleic acid molecules is independently formulated with a lipid nanoparticle (LNP).
  • LNP lipid nanoparticle
  • the application also provides methods of making compositions and therapeutic combinations of the application.
  • a method of producing a composition or therapeutic combination comprises mixing an isolated polynucleotide encoding an HBV antigen, vector, and/or polypeptide of the application with one or more pharmaceutically acceptable carriers.
  • One of ordinary skill in the art will be familiar with conventional techniques used to prepare such compositions.
  • the application also provides methods of inducing an immune response against hepatitis B virus (HBV) in a subject in need thereof, comprising administering to the subject an
  • HBV hepatitis B virus
  • compositions or immunogenic composition of the application are immunogenically effective amounts of a composition or immunogenic composition of the application. Any of the compositions and therapeutic combinations of the application described herein can be used in the methods of the application.
  • infectious agents refers to the invasion of a host by a disease causing agent.
  • a disease causing agent is considered to be“infectious” when it is capable of invading a host, and replicating or propagating within the host.
  • infectious agents include viruses, e.g., HBV and certain species of adenovirus, prions, bacteria, fungi, protozoa and the like.
  • HBV infection specifically refers to invasion of a host organism, such as cells and tissues of the host organism, by HBV.
  • the phrase“inducing an immune response” when used with reference to the methods described herein encompasses causing a desired immune response or effect in a subject in need thereof against an infection, e.g., an HBV infection.“Inducing an immune response” also encompasses providing a therapeutic immunity for treating against a pathogenic agent, e.g., HBV.
  • a pathogenic agent e.g., HBV.
  • the term“therapeutic immunity” or“therapeutic immune response” means that the vaccinated subject is able to control an infection with the pathogenic agent against which the vaccination was done, for instance immunity against HBV infection conferred by vaccination with HBV vaccine.
  • “inducing an immune response” means producing an immunity in a subject in need thereof, e.g., to provide a therapeutic effect against a disease, such as HBV infection.
  • “inducing an immune response” refers to causing or improving cellular immunity, e.g., T cell response, against HBV infection.
  • “inducing an immune response” refers to causing or improving a humoral immune response against HBV infection. In certain embodiments,“inducing an immune response” refers to causing or improving a cellular and a humoral immune response against HBV infection.
  • the term“protective immunity” or“protective immune response” means that the vaccinated subject is able to control an infection with the pathogenic agent against which the vaccination was done. Usually, the subject having developed a“protective immune response” develops only mild to moderate clinical symptoms or no symptoms at all. Usually, a subject having a“protective immune response” or“protective immunity” against a certain agent will not die as a result of the infection with said agent.
  • compositions and therapeutic combinations of the application will have a therapeutic aim to generate an immune response against HBV after HBV infection or development of symptoms characteristic of HBV infection, e.g., for therapeutic vaccination.
  • an immunogenically effective amount or“immunologically effective amount” means an amount of a composition, polynucleotide, vector, or antigen sufficient to induce a desired immune effect or immune response in a subject in need thereof.
  • immunogenically effective amount can be an amount sufficient to induce an immune response in a subject in need thereof.
  • An immunogenically effective amount can be an amount sufficient to produce immunity in a subject in need thereof, e.g., provide a therapeutic effect against a disease such as HBV infection.
  • An immunogenically effective amount can vary depending upon a variety of factors, such as the physical condition of the subject, age, weight, health, etc.; the particular application, e.g., providing protective immunity or therapeutic immunity; and the particular disease, e.g., viral infection, for which immunity is desired.
  • An immunogenically effective amount can readily be determined by one of ordinary skill in the art in view of the present disclosure.
  • an immunogenically 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) prevent the recurrence of an HBV infection or symptom associated therewith; (vii) reduce hospitalization of a subject having an HBV infection; (viii) reduce hospitalization length of a subject having an HBV infection; (ix) increase the survival of a subject with an HBV infection; (x) eliminate an HBV infection in a subject; (xi) inhibit or reduce HBV replication in a subject; and/or (xii
  • An immunogenically effective amount can also be an amount 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 within 12 months.
  • a target index include lower HBsAg below a threshold of 500 copies of HBsAg international units (IU) and/or higher CD8 counts.
  • an immunogenically effective amount when used with reference to a DNA plasmid can range from about 0.1 mg/mL to 10 mg/mL of DNA plasmid total, such as 0.1 mg/mL, 0.25 mg/mL, 0.5 mg/mL.0.75 mg/mL 1 mg/mL, 1.5 mg/mL, 2 mg/mL, 3 mg/mL, 4 mg/mL, 5 mg/mL, 6 mg/mL, 7 mg/mL, 8 mg/mL, 9 mg/mL, or 10 mg/mL.
  • an immunogenically effective amount of DNA plasmid is less than 8 mg/mL, more preferably less than 6 mg/mL, even more preferably 3-4 mg/mL.
  • An immunogenically effective amount can be from one vector or plasmid, or from multiple vectors or plasmids.
  • an immunogenically effective amount when used with reference to a peptide can range from about 10 ⁇ g to 1 mg per administration, such as 10, 20, 50, 100, 200, 300, 400, 500, 600, 700, 800, 9000, or 1000 ⁇ g per administration.
  • An immunogenically effective amount can be administered in a single composition, or in multiple compositions, such as 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 compositions (e.g., tablets, capsules or injectables, or any composition adapted to intradermal delivery, e.g., to intradermal delivery using an intradermal delivery patch), wherein the administration of the multiple capsules or injections collectively provides a subject with an immunogenically effective amount.
  • an immunogenically effective amount can be 3-4 mg/mL, with 1.5-2 mg/mL of each plasmid.
  • prime-boost regimen It is also possible to administer an immunogenically effective amount to a subject, and subsequently administer another dose of an immunogenically effective amount to the same subject, in a so-called prime-boost regimen.
  • This general concept of a prime-boost regimen is well known to the skilled person in the vaccine field. Further booster administrations can optionally be added to the regimen, as needed.
  • a therapeutic combination comprising two DNA plasmids, e.g., a first DNA plasmid encoding an HBV core antigen and second DNA plasmid encoding an HBV pol antigen, can be administered to a subject by mixing both plasmids and delivering the mixture to a single anatomic site.
  • two separate immunizations each delivering a single expression plasmid can be performed.
  • the first DNA plasmid and the second DNA plasmid can be administered in a ratio of 10:1 to 1:10, by weight, such as 10:1, 9:1, 8:1, 7:1, 6:1, 5:1, 4:1, 3:1, 2:1, 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, or 1:10, by weight.
  • the first and second DNA plasmids are administered in a ratio of 1:1, by weight.
  • a subject to be treated according to the methods of the application is an HBV- infected subject, particular a subject having chronic HBV infection.
  • Acute HBV infection is characterized by an efficient activation of the innate immune system complemented with a subsequent broad adaptive response (e.g., HBV-specific T-cells, neutralizing antibodies), which usually results in successful suppression of replication or removal of infected hepatocytes.
  • HBV-specific T-cells, neutralizing antibodies e.g., HBV-specific T-cells, neutralizing antibodies
  • HBV envelope proteins are produced in abundance and can be released in sub-viral particles in 1,000- fold excess to infectious virus.
  • Chronic HBV infection is described in phases characterized by viral load, liver enzyme levels (necroinflammatory activity), HBeAg, or HBsAg load or presence of antibodies to these antigens.
  • cccDNA levels stay relatively constant at approximately 10 to 50 copies per cell, even though viremia can vary considerably. The persistence of the cccDNA species leads to chronicity.
  • the phases of chronic HBV infection include: (i) the immune- tolerant phase characterized by high viral load and normal or minimally elevated liver enzymes; (ii) the immune activation HBeAg-positive phase in which lower or declining levels of viral replication with significantly elevated liver enzymes are observed; (iii) the inactive HBsAg carrier phase, which is a low replicative state with low viral loads and normal liver enzyme levels in the serum that may follow HBeAg seroconversion; and (iv) the HBeAg-negative phase in which viral replication occurs periodically (reactivation) with concomitant fluctuations in liver enzyme levels, mutations in the pre-core and/or basal core promoter are common, such that HBeAg is not produced by the infected cell.
  • chronic HBV infection refers to a subject having the detectable presence of HBV for more than 6 months.
  • a subject having a chronic HBV infection can be in any phase of chronic HBV infection.
  • Chronic HBV infection is understood in accordance with its ordinary meaning in the field.
  • Chronic HBV infection can for example be characterized by the persistence of HBsAg for 6 months or more after acute HBV infection.
  • a chronic HBV infection referred to herein follows the definition published by the Centers for Disease Control and Prevention (CDC), according to which a chronic HBV infection can be characterized by laboratory criteria such as: (i) negative for IgM antibodies to hepatitis B core antigen (IgM anti- HBc) and positive for hepatitis B surface antigen (HBsAg), hepatitis B e antigen (HBeAg), or nucleic acid test for hepatitis B virus DNA, or (ii) positive for HBsAg or nucleic acid test for HBV DNA, or positive for HBeAg two times at least 6 months apart.
  • IgM anti- HBc hepatitis B core antigen
  • HBsAg hepatitis B surface antigen
  • HBeAg hepatitis B e antigen
  • nucleic acid test for hepatitis B virus DNA or
  • positive for HBeAg two times at least 6 months apart.
  • an immunogenically effective amount refers to the amount of a composition or therapeutic combination of the application which is sufficient to treat chronic HBV infection.
  • a subject having chronic HBV infection is undergoing nucleoside analog (NUC) treatment, and is NUC-suppressed.
  • NUC-suppressed refers to a subject having an undetectable viral level of HBV and stable alanine aminotransferase (ALT) levels for at least six months.
  • nucleoside/nucleotide analog treatment include HBV polymerase inhibitors, such as entacavir and tenofovir.
  • a subject having chronic HBV infection does not have advanced hepatic fibrosis or cirrhosis.
  • Such subject would typically have a METAVIR score of less than 3 for fibrosis and a fibroscan result of less than 9 kPa.
  • the METAVIR score is a scoring system that is commonly used to assess the extent of inflammation and fibrosis by histopathological evaluation in a liver biopsy of patients with hepatitis B.
  • the scoring system assigns two standardized numbers: one reflecting the degree of inflammation and one reflecting the degree of fibrosis.
  • an immunogenically effective amount is an amount sufficient to achieve persistent loss of HBsAg within 12 months and significant decrease in clinical disease (e.g., cirrhosis, hepatocellular carcinoma, etc.).
  • Methods according to embodiments of the application further comprises administering to the subject in need thereof another immunogenic agent (such as another HBV antigen or other antigen) or another anti-HBV agent (such as a nucleoside analog or other anti-HBV agent) in combination with a composition of the application.
  • another immunogenic agent such as another HBV antigen or other antigen
  • another anti-HBV agent such as a nucleoside analog or other anti-HBV agent
  • another anti-HBV agent or immunogenic agent can be a small molecule or antibody including, but not limited to, immune checkpoint inhibitors (e.g., anti-PD1, anti-TIM-3, etc.), toll-like receptor agonists (e.g., TLR7 agonists and/oror TLR8 agonists), RIG-1 agonists, IL-15 superagonists (Altor Bioscience), mutant IRF3 and IRF7 genetic adjuvants, STING agonists (Aduro), FLT3L genetic adjuvant, IL12 genetic adjuvant, IL-7-hyFc; CAR-T which bind HBV env (S-CAR cells); capsid assembly modulators; cccDNA inhibitors, HBV polymerase inhibitors (e.g., entecavir and tenofovir).
  • immune checkpoint inhibitors e.g., anti-PD1, anti-TIM-3, etc.
  • toll-like receptor agonists e.g., TLR7 agonist
  • the one or other anti-HBV active agents can be, for example, a small molecule, an antibody or antigen binding fragment thereof, a polypeptide, protein, or nucleic acid.
  • the one or other anti-HBV agents can e.g., be chosen from among HBV DNA polymerase inhibitors; Immunomodulators; Toll-like receptor 7 modulators; Toll-like receptor 8 modulators; Toll-like receptor 3 modulators; Interferon alpha receptor ligands; 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-HBV antisense oligonucleotides; short interfering RNAs (siRNA), more particularly anti-HBV siRNA; Endonuclease modulators; Inhibitors of ribonu
  • Cytokines such as IL12; Capsid Assembly Modulators, Nucleoprotein inhibitors (HBV core or capsid protein inhibitors); Nucleic Acid Polymers (NAPs); Stimulators of retinoic acid-inducible gene 1; Stimulators of NOD2; Recombinant thymosin alpha-1; Hepatitis B virus 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.
  • IL12 Capsid Assembly Modulators, Nucleoprotein inhibitors (HBV core or capsid protein inhibitors);
  • compositions and therapeutic combinations of the application can be administered to a subject by any method known in the art in view of the present disclosure, including, but not limited to, parenteral administration (e.g., intramuscular, subcutaneous, intravenous, or intradermal injection), oral administration, transdermal administration, and nasal administration.
  • parenteral administration e.g., intramuscular, subcutaneous, intravenous, or intradermal injection
  • oral administration e.g., oral administration
  • transdermal administration e.g., transdermal administration
  • nasal administration e.g., by intramuscular injection or intradermal injection
  • compositions and therapeutic combinations are administered parenterally (e.g., by intramuscular injection or intradermal injection) or transdermally.
  • administration can be by injection through the skin, e.g., intramuscular or intradermal injection, preferably intramuscular injection.
  • Intramuscular injection can be combined with electroporation, i.e., application of an electric field to facilitate delivery of the DNA plasmids to cells.
  • electroporation i.e., application of an electric field to facilitate delivery of the DNA plasmids to cells.
  • electroporation refers to the use of a transmembrane electric field pulse to induce microscopic pathways (pores) in a bio-membrane.
  • electrical fields of appropriate magnitude and duration are applied to cells, inducing a transient state of enhanced cell membrane permeability, thus enabling the cellular uptake of molecules unable to cross cell membranes on their own.
  • electroporation is combined with intramuscular injection.
  • electroporation with other forms of parenteral administration, e.g., intradermal injection, subcutaneous injection, etc.
  • the electroporation device can include an electroporation component and an electrode assembly or handle assembly.
  • the electroporation component can include one or more of the following components of electroporation devices: controller, current waveform generator, impedance tester, waveform logger, input element, status reporting element, communication port, memory component, power source, and power switch.
  • Electroporation can be accomplished using an in vivo electroporation device. Examples of electroporation devices and electroporation methods that can facilitate delivery of compositions and therapeutic combinations of the application, particularly those comprising DNA plasmids, include
  • Tri-GridTM delivery system (Ichor Medical Systems, Inc., San Diego, CA 92121) and those described in U.S. Patent No.7,664,545, U.S. Patent No.8,209,006, U.S. Patent No.9,452,285, U.S. Patent No.5,273,525, U.S. Patent No.6,110,161, U.S. Patent No. 6,261,281, U.S. Patent No.6,958,060, and U.S. Patent No.6,939,862, U.S. Patent No.7,328,064, U.S. Patent No.6,041,252, U.S. Patent No.5,873,849, U.S. Patent No.6,278,895, U.S.
  • Patent No.6,319,901 U.S. Patent No.6,912,417, U.S. Patent No.8,187,249, U.S. Patent No.9,364,664, U.S. Patent No.9,802,035, U.S. Patent No.6,117,660, and International Patent Application Publication WO2017172838, all of which are herein incorporated by reference in their entireties.
  • Other examples of in vivo electroporation devices are described in International Patent
  • Transdermal administration can be combined with epidermal skin abrasion to facilitate delivery of the DNA plasmids to cells.
  • a dermatological patch can be used for epidermal skin abrasion. Upon removal of the dermatological patch, the composition or therapeutic combination can be deposited on the abraised skin.
  • Methods of delivery are not limited to the above described embodiments, and any means for intracellular delivery can be used.
  • Other methods of intracellular delivery contemplated by the methods of the application include, but are not limited to, liposome encapsulation, lipid nanoparticles (LNPs), etc.
  • a method of inducing an immune response against HBV further comprises administering an adjuvant.
  • adjuvant and “immune stimulant” are used interchangeably herein, and are defined as one or more substances that cause stimulation of the immune system.
  • an adjuvant is used to enhance an immune response to HBV antigens and antigenic HBV polypeptides of the application.
  • an adjuvant can be present in a therapeutic combination or composition of the application, or administered in a separate composition.
  • An adjuvant can be, e.g., a small molecule or an antibody.
  • adjuvants suitable for use in the application include, but are not limited to, immune checkpoint inhibitors (e.g., anti-PD1, anti- TIM-3, etc.), toll-like receptor agonists (e.g., TLR 7 and/or TLR8 agonists), RIG-1 agonists, IL- 15 superagonists (Altor Bioscience), mutant IRF3 and IRF7 genetic adjuvants, STING agonists (Aduro), FLT3L genetic adjuvant, IL12 genetic adjuvant, and IL-7-hyFc.
  • immune checkpoint inhibitors e.g., anti-PD1, anti- TIM-3, etc.
  • toll-like receptor agonists e.g., TLR 7 and/or TLR8 agonists
  • adjuvants can e.g., be chosen from among the following anti-HBV agents: HBV DNA polymerase inhibitors; Immunomodulators; Toll-like receptor 7 modulators; Toll-like receptor 8 modulators; Toll-like receptor 3 modulators; Interferon alpha receptor ligands; 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-HBV antisense oligonucleotides; short interfering RNAs (siRNA), more particularly anti-HBV siRNA; Endonuclease modulators; Inhibitors of ribonucleotide reductase; Hepatitis B virus E antigen inhibitors; HBV antibodies targeting the surface antigens of the hepatitis B virus; HBV
  • compositions and therapeutic combinations of the application can also be administered in combination with at least one other anti-HBV agent.
  • anti-HBV agents suitable for use with the application include, but are not limited to small molecules, antibodies, and/or CAR- T therapies which bind HBV env (S-CAR cells), capsid assembly modulators, TLR agonists (e.g., TLR7 and/or TLR8 agonists), cccDNA inhibitors, HBV polymerase inhibitors (e.g., entecavir and tenofovir), and/or immune checkpoint inhibitors, etc.
  • the at least one anti-HBV agent can e.g., be chosen from among HBV DNA polymerase inhibitors; Immunomodulators; Toll-like receptor 7 modulators; Toll-like receptor 8 modulators; Toll-like receptor 3 modulators; Interferon alpha receptor ligands; 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-HBV antisense oligonucleotides; short interfering RNAs (siRNA), more particularly anti-HBV siRNA; Endonuclease modulators; Inhibitors of ribonucleotide reductase; Hepatitis B virus E antigen inhibitors; HBV antibodies targeting the surface antigens of the hepatitis B virus; HBV antibodies; CCR
  • Embodiments of the application also contemplate administering an immunogenically effective amount of a composition or therapeutic combination to a subject, and subsequently administering another dose of an immunogenically effective amount of a composition or therapeutic combination to the same subject, in a so-called prime-boost regimen
  • a composition or therapeutic combination of the application is a primer vaccine used for priming an immune response.
  • a composition or therapeutic combination of the application is a booster vaccine used for boosting an immune response.
  • the priming and boosting vaccines of the application can be used in the methods of the application described herein. This general concept of a prime-boost regimen is well known to the skilled person in the vaccine field. Any of the compositions and therapeutic combinations of the application described herein can be used as priming and/or boosting vaccines for priming and/or boosting an immune response against HBV.
  • a composition or therapeutic combination of the application can be administered for priming immunization.
  • the composition or therapeutic combination can be re-administered for boosting immunization. Further booster administrations of the composition or vaccine combination can optionally be added to the regimen, as needed.
  • An adjuvant can be present in a composition of the application used for boosting immunization, present in a separate composition to be administered together with the composition or therapeutic combination of the application for the boosting immunization, or administered on its own as the boosting immunization.
  • the adjuvant is preferably used for boosting immunization.
  • Prime-boost regimen includes
  • boosting immunization is first administered about two to six weeks, preferably four weeks after the priming immunization is initially administered.
  • the priming immunization is initially administered, a further boosting immunization of the composition or therapeutic combination, or other adjuvant, is administered.
  • kits comprising a therapeutic combination of the application.
  • a kit can comprise the first polynucleotide, the second polynucleotide, and the at least one capsid assembly modulator in one or more separate compositions, or a kit can comprise the first polynucleotide, the second polynucleotide, and the capsid assembly modulator in a single composition.
  • a kit can further comprise one or more adjuvants or immune stimulants, and/or other anti-HBV agents.
  • Measurement of cellular immunity can be performed by measurement of cytokine profiles secreted by activated effector cells including those derived from CD4+ and CD8+ T-cells (e.g. quantification of IL-10 or IFN gamma-producing cells by ELISPOT), by determination of the activation status of immune effector cells (e.g. T cell proliferation assays by a classical [3H] thymidine uptake or flow cytometry-based assays), by assaying for antigen-specific T lymphocytes in a sensitized subject (e.g. peptide-specific lysis in a cytotoxicity assay, etc.).
  • activated effector cells including those derived from CD4+ and CD8+ T-cells (e.g. quantification of IL-10 or IFN gamma-producing cells by ELISPOT), by determination of the activation status of immune effector cells (e.g. T cell proliferation assays by a classical [3H] thymidine uptake or flow cyto
  • the ability to stimulate a cellular and/or a humoral response can be determined by antibody binding and/or competition in binding (see for example Harlow, 1989, Antibodies, Cold Spring Harbor Press).
  • titers of antibodies produced in response to administration of a composition providing an immunogen can be measured by enzyme-linked immunosorbent assay (ELISA).
  • ELISA enzyme-linked immunosorbent assay
  • the immune responses can also be measured by neutralizing antibody assay, where a neutralization of a virus is defined as the loss of infectivity through
  • the invention provides also the following non-limiting embodiments.
  • Embodiment 1 is a therapeutic combination for use in treating a hepatitis B virus (HBV) infection in a subject in need thereof, comprising:
  • a truncated HBV core antigen consisting of an amino acid sequence that is at least 95%, such as at least 95%, 96%, 97%, 98%, 99% or 100%, identical to SEQ ID NO: 2,
  • a first non-naturally occurring nucleic acid molecule comprising a first polynucleotide sequence encoding the truncated HBV core antigen
  • an HBV polymerase antigen having an amino acid sequence that is at least 90%, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%, identical to SEQ ID NO: 7, wherein the HBV polymerase antigen does not have reverse transcriptase activity and RNase H activity, and
  • a second non-naturally occurring nucleic acid molecule comprising a second polynucleotide sequence encoding the HBV polymerase antigen
  • a capsid assembly modulator such as that described herein.
  • Embodiment 2 is the therapeutic combination of embodiment 1, comprising at least one of the HBV polymerase antigen and the truncated HBV core antigen.
  • Embodiment 3 is the therapeutic combination of embodiment 2, comprising the HBV polymerase antigen and the truncated HBV core antigen.
  • Embodiment 4 is the therapeutic combination of embodiment 1, comprising at least one of the first non-naturally occurring nucleic acid molecule comprising the first polynucleotide sequence encoding the truncated HBV core antigen, and the second non-naturally occurring nucleic acid molecule comprising the second polynucleotide sequence encoding the HBV polymerase antigen.
  • Embodiment 5 is a therapeutic combination for use in treating a hepatitis B virus (HBV) infection in a subject in need thereof, comprising
  • a first non-naturally occurring nucleic acid molecule comprising a first
  • polynucleotide sequence encoding a truncated HBV core antigen consisting of an amino acid sequence that is at least 95% identical to SEQ ID NO: 2; and ii) a second non-naturally occurring nucleic acid molecule comprising a second polynucleotide sequence encoding an HBV polymerase antigen having an amino acid sequence that is at least 90% identical to SEQ ID NO: 7, wherein the HBV polymerase antigen does not have reverse transcriptase activity and RNase H activity; and
  • a a compound of formula (I), (XI), (XVIII), or (XXI), or an enantiomer, diastereomer or pharmaceutically accepted salt, hydrate, or solvate thereof, including a hydrate, a solvate, a pharmaceutically acceptable salt, a prodrug, or a complex thereof described herein;
  • Embodiment 6 is the therapeutic combination of embodiment 4 or 5, wherein the first non-naturally occurring nucleic acid molecule further comprises a polynucleotide sequence encoding a signal sequence operably linked to the N-terminus of the truncated HBV core antigen.
  • Embodiment 6a is the therapeutic combination of any one of embodiments 4 to 6, wherein the second non-naturally occurring nucleic acid molecule further comprises a polynucleotide sequence encoding a signal sequence operably linked to the N-terminus of the HBV polymerase antigen.
  • Embodiment 6b is the therapeutic combination of embodiment 6 or 6a, wherein the signal sequence independently comprises the amino acid sequence of SEQ ID NO: 9 or SEQ ID NO: 15.
  • Embodiment 6c is the therapeutic combination of embodiment 6 or 6a, wherein the signal sequence is independently encoded by the polynucleotide sequence of SEQ ID NO: 8 or SEQ ID NO: 14.
  • Embodiment 7 is the therapeutic combination of any one of embodiments 1-6c, wherein the HBV polymerase antigen comprises an amino acid sequence that is at least 98%, such as at least 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9%, or 100%, identical to SEQ ID NO: 7.
  • Embodiment 7a is the therapeutic combination of embodiment 7, wherein the HBV polymerase antigen comprises the amino acid sequence of SEQ ID NO: 7.
  • Embodiment 7b is the therapeutic combination of any one of embodiments 1 to 7a, wherein and the truncated HBV core antigen consists of the amino acid sequence that is at least 98%, such as at least 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9%, or 100%, identical to SEQ ID NO: 2.
  • Embodiment 7c is the therapeutic combination of embodiment 7b, wherein the truncated HBV antigen consists of the amino acid sequence of SEQ ID NO: 2 or SEQ ID NO: 4.
  • Embodiment 8 is the therapeutic combination of any one of embodiments 1-7c, wherein each of the first and second non-naturally occurring nucleic acid molecules is a DNA molecule.
  • Embodiment 8a is the therapeutic combination of embodiment 8, wherein the DNA molecule is present on a DNA vector.
  • Embodiment 8b is the therapeutic combination of embodiment 8a, wherein the DNA vector is selected from the group consisting of DNA plasmids, bacterial artificial chromosomes, yeast artificial chromosomes, and closed linear deoxyribonucleic acid.
  • Embodiment 8c is the therapeutic combination of embodiment 8, wherein the DNA molecule is present on a viral vector.
  • Embodiment 8d is the therapeutic combination of embodiment 8c, wherein the viral vector is selected from the group consisting of bacteriophages, animal viruses, and plant viruses.
  • Embodiment 8e is the therapeutic combination of any one of embodiments 1-7c, wherein each of the first and second non-naturally occurring nucleic acid molecules is an RNA molecule.
  • Embodiment 8f is the therapeutic combination of embodiment 8e, wherein the RNA molecule is an RNA replicon, preferably a self-replicating RNA replicon, an mRNA replicon, a modified mRNA replicon, or self-amplifying mRNA.
  • the RNA molecule is an RNA replicon, preferably a self-replicating RNA replicon, an mRNA replicon, a modified mRNA replicon, or self-amplifying mRNA.
  • Embodiment 8g is the therapeutic combination of any one of embodiments 1 to 8f, wherein each of the first and second non-naturally occurring nucleic acid molecules is independently formulated with a lipid composition, preferably a lipid nanoparticle (LNP).
  • a lipid composition preferably a lipid nanoparticle (LNP).
  • Embodiment 9 is the therapeutic combination of any one of embodiments 4-8g, comprising the first non-naturally occurring nucleic acid molecule and the second non-naturally occurring nucleic acid molecule in the same non-naturally occurring nucleic acid molecule.
  • Embodiment 10 is the therapeutic combination of any one of embodiments 4-8g, comprising the first non-naturally occurring nucleic acid molecule and the second non-naturally occurring nucleic acid molecule in two different non-naturally occurring nucleic acid molecules.
  • Embodiment 11 is the therapeutic combination of any one of embodiments 4-10, wherein the first polynucleotide sequence comprises a polynucleotide sequence having at least 90%, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%, sequence identity to SEQ ID NO: 1 or SEQ ID NO: 3.
  • Embodiment 11a is the therapeutic combination of embodiment 11, wherein the first polynucleotide sequence comprises a polynucleotide sequence having at least 98%, such as at least 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9%, or 100%, sequence identity to SEQ ID NO: 1 or SEQ ID NO: 3.
  • Embodiment 12 is the therapeutic combination of embodiment 11a, wherein the first polynucleotide sequence comprises the polynucleotide sequence of SEQ ID NO: 1 or SEQ ID NO: 3.
  • Embodiment 13 is the therapeutic combination of any one of embodiments 4 to 12, wherein the second polynucleotide sequence comprises a polynucleotide sequence having at least 90%, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%, sequence identity to SEQ ID NO: 5 or SEQ ID NO: 6.
  • Embodiment 13a is the therapeutic combination of embodiment 13, wherein the second polynucleotide sequence comprises a polynucleotide sequence having at least 98%, such as at least 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9%, or 100%, sequence identity to SEQ ID NO: 5 or SEQ ID NO: 6.
  • Embodiment 14 is the therapeutic combination of embodiment 13a, wherein the second polynucleotide sequence comprises the polynucleotide sequence of SEQ ID NO: 5 or SEQ ID NO: 6.
  • Embodiment 15 is the therapeutic combination of any one of embodiments 1 to 14, wherein the compound is selected from the group consisting of the exemplified compounds, or an enantiomer, diastereomer or pharmaceutically accepted salt, hydrate, or solvate thereof, including a hydrate, a solvate, a pharmaceutically acceptable salt, a prodrug, or a complex thereof.
  • Embodiment 15a is the therapeutic combination of any one of embodiments 1 to 15, wherein the capsid assembly modulator is a compound having formula (I), (XI), (XVIII), or (XXI), as described herein.
  • Embodiment 15b is the therapeutic combination of any one of embodiments 1 to 15, wherein the capsid assembly modulator is a compound selected from the exemplified compounds having formula (I), (XI), (XVIII), or (XXI), as described herein or in WO 2014106019 and US 20160024004, the content of which is incorporated herein by reference.
  • the capsid assembly modulator is a compound selected from the exemplified compounds having formula (I), (XI), (XVIII), or (XXI), as described herein or in WO 2014106019 and US 20160024004, the content of which is incorporated herein by reference.
  • Embodiment 15c is the therapeutic combination of any one of embodiments 1 to 15, wherein the capsid assembly modulator is a compound having (A-I), (A-II), (A-III), (A-IV), (A- V), (A-VI), or (AVII), as described herein.
  • Embodiment 15d is the therapeutic combination of any one of embodiments 1 to 15, wherein the capsid assembly modulator is a compound selected from the exemplified compounds having (A-I), (A-II), (A-III), (A-IV), (A-V), (A-VI), or (AVII), as described herein, or in WO 2017156255, the content of which is incorporated herein by reference.
  • Embodiment 15e is the therapeutic combination of any one of embodiments 1 to 15, wherein the capsid assembly modulator is a compound having formula (B-I) as described herein.
  • Embodiment 15f is the therapeutic combination of any one of embodiments 1 to 15, wherein the capsid assembly modulator is a compound selected from the exemplified compounds having formula (B-I) as described herein, or in WO 2018039531 and US 20180161307, the content of which is incorporated herein by reference.
  • Embodiment 16 is a kit comprising the therapeutic combination of any one of embodiments 1 to 15f, and instructions for using the therapeutic combination in treating a hepatitis B virus (HBV) infection in a subject in need thereof.
  • HBV hepatitis B virus
  • Embodiment 17 is a method of treating a hepatitis B virus (HBV) infection in a subject in need thereof, comprising administering to the subject the therapeutic combination of any one of embodiments 1 to 15f.
  • HBV hepatitis B virus
  • Embodiment 17a is the method of embodiment 17, wherein the treatment induces an immune response against a hepatitis B virus in a subject in need thereof, preferably the subject has chronic HBV infection.
  • Embodiment 17b is the method of embodiment 17 or 17a, wherein the subject has chronic HBV infection.
  • Embodiment 17c is the method of any one of embodiments 17 to 17b, wherein the subject is in need of a treatment of an HBV-induced disease selected from the group consisting of advanced fibrosis, cirrhosis and hepatocellular carcinoma (HCC).
  • HBV-induced disease selected from the group consisting of advanced fibrosis, cirrhosis and hepatocellular carcinoma (HCC).
  • Embodiment 18 is the method of any one of embodiments 17-17c, wherein the therapeutic combination is administered by injection through the skin, e.g., intramuscular or intradermal injection, preferably intramuscular injection.
  • Embodiment 19 is the method of embodiment 18, wherein the therapeutic combination comprises at least one of the first and second non-naturally occurring nucleic acid molecules.
  • Embodiment 19a is the method of embodiment 19, wherein the therapeutic combination comprises the first and second non-naturally occurring nucleic acid molecules.
  • Embodiment 20 is the method of embodiment 19 or 19a, wherein the non-naturally occurring nucleic acid molecules are administered to the subject by intramuscular injection in combination with electroporation.
  • Embodiment 21 is the method of embodiment 19 or 19a, wherein the non-naturally occurring nucleic acid molecules are administered to the subject by a lipid composition, preferably by a lipid nanoparticle.
  • FIG.1A and 1B A schematic representation of the pDK-pol and pDK-core vectors is shown in Fig.1A and 1B, respectively.
  • An HBV core or pol antigen optimized expression cassette containing a CMV promoter (SEQ ID NO: 18), a splicing enhancer (triple composite sequence) (SEQ ID NO: 10), Cystatin S precursor signal peptide SPCS (NP_0018901.1) (SEQ ID NO: 9), and pol (SEQ ID NO: 5) or core (SEQ ID NO: 2) gene was introduced into a pDK plasmid backbone, using standard molecular biology techniques.
  • the plasmids were tested in vitro for core and pol antigen expression by Western blot analysis using core and pol specific antibodies, and were shown to provide consistent expression profile for cellular and secreted core and pol antigens (data not shown).
  • adenovirus vector has been designed as a fusion protein expressed from a single open reading frame. Additional configurations for the expression of the two proteins, e.g. using two separate expression cassettes, or using a 2A-like sequence to separate the two sequences, can also be envisaged.
  • the expression cassettes are comprised of the CMV promoter (SEQ ID NO: 19), an intron (SEQ ID NO:12) (a fragment derived from the human ApoAI gene - GenBank accession X01038 base pairs 295– 523, harboring the ApoAI second intron), followed by the optimized coding sequence– either core alone or the core and polymerase fusion protein preceded by a human immunoglobulin secretion signal coding sequence (SEQ ID NO: 14), and followed by the SV40 polyadenylation signal (SEQ ID NO: 13).
  • a secretion signal was included because of past experience showing improvement in the manufacturability of some adenoviral vectors harboring secreted transgenes, without influencing the elicited T-cell response (mouse experiments).
  • VVMP Polymerase protein
  • the plasmids tested included a pDK-Pol plasmid and pDK-Core plasmid, as shown in FIGS.1A and 1B, respectively, and as described above in Example 1.
  • the pDK-Pol plasmid encoded a polymerase antigen having the amino acid sequence of SEQ ID NO: 7, and the pDK-Core plasmid encoding a Core antigen having the amino acid sequence of SEQ ID NO: 2.
  • T-cell responses induced by each plasmid individually were tested.
  • the DNA plasmid (pDNA) vaccine was intramuscularly delivered via electroporation to Balb/c mice using a commercially available TriGridTM delivery system-intramuscular (TDS-IM) adapted for application in the mouse model in cranialis tibialis.
  • TDS-IM TriGridTM delivery system-intramuscular
  • HBV Hepatitis B Virus
  • the TDS-IM array of a TDS-IM v1.0 device having an electrode array with a 2.5 mm spacing between the electrodes and an electrode diameter of 0.030 inch was inserted percutaneously into the selected muscle, with a conductive length of 3.2 mm and an effective penetration depth of 3.2 mm, and with the major axis of the diamond configuration of the electrodes oriented in parallel with the muscle fibers.
  • the injection was initiated to distribute DNA (e.g., 0.020 ml) in the muscle.
  • a 250 V/cm electrical field (applied voltage of 59.4 -65.6 V, applied current limits of less than 4 A, 0.16 A/sec) was locally applied for a total duration of about 400 ms at a 10% duty cycle (i.e., voltage is actively applied for a total of about 40 ms of the about 400 ms duration) with 6 total pulses.
  • the TriGridTM array was removed and the animals were recovered. High-dose (20 ⁇ g) administration to BALB/c mice was performed as summarized in Table 1.
  • mice Six mice were administered plasmid DNA encoding the HBV core antigen (pDK-core; Group 1), six mice were administered plasmid DNA encoding the HBV pol antigen (pDK-pol; Group 2), and two mice received empty vector as the negative control. Animals received two DNA immunizations two weeks apart and splenocytes were collected one week after the last immunization.
  • Antigen-specific responses were analyzed and quantified by IFN-g enzyme-linked immunospot (ELISPOT).
  • IFN-g enzyme-linked immunospot In this assay, isolated splenocytes of immunized animals were incubated overnight with peptide pools covering the Core protein, the Pol protein, or the small peptide leader and junction sequence (2 ⁇ g/ml of each peptide). These pools consisted of 15 mer peptides that overlap by 11 residues matching the Genotypes BCD consensus sequence of the Core and Pol vaccine vectors. The large 94 kDan HBV Pol protein was split in the middle into two peptide pools. Antigen-specific T cells were stimulated with the homologous peptide pools and IFN-g-positive T cells were assessed using the ELISPOT assay. IFN-g release by a single antigen-specific T cell was visualized by appropriate antibodies and subsequent chromogenic detection as a colored spot on the microplate referred to as spot-forming cell (SFC).
  • SFC spot-
  • mice immunized with the DNA vaccine plasmid pDK-Core (Group 1) reaching 1,000 SFCs per 10 6 cells (FIG.3).
  • Pol T-cell responses towards the Pol 1 peptide pool were strong ( ⁇ 1,000 SFCs per 10 6 cells).
  • the weak Pol-2-directed anti-Pol cellular responses were likely due to the limited MHC diversity in mice, a phenomenon called T-cell immunodominance defined as unequal recognition of different epitopes from one antigen.

Abstract

Therapeutic combinations of hepatitis B virus (HBV) vaccines and capsid assembly modulators such as sulfonamide derivatives are described. Methods of inducing an immune response against HBV or treating an HBV-induced disease, particularly in individuals having chronic HBV infection, using the disclosed therapeutic combinations are also described.

Description

COMBINATION OF HEPATITIS B VIRUS (HBV) VACCINES AND CAPSID
ASSEMBLY MODULATORS BEING AMIDE DERIVATIVES
CROSS REFERENCE TO RELATED APPLICATION
This application claims priority to U.S. Provisional Application No. 62/862,775 filed on June 18, 2019, the disclosure of which is incorporated herein by reference in its entirety.
REFERENCE TO SEQUENCE LISTING SUBMITTED ELECTRONICALLY
This application contains a sequence listing, which is submitted electronically via EFS- Web as an ASCII formatted sequence listing with a file name“065814_36WOl Sequence Listing” and a creation date of June 4, 2020 and having a size of 46 kb. The sequence listing submitted via EFS-Web is part of the specification and is herein incorporated by reference in its entirety.
BACKGROUND OF THE INVENTION
Hepatitis B virus (HBV) is a small 3.2-kb hepatotropic DNA virus that encodes four open reading frames and seven proteins. Approximately 240 million people have chronic hepatitis B infection (chronic HBV), characterized by persistent virus and subvirus particles in the blood for more than 6 months (Cohen et al. J. Viral Hepat. (2011) 18(6), 377-83). Persistent HBV infection leads to T-cell exhaustion in circulating and intrahepatic HBV-specific CD4+ and CD8+ T-cells through chronic stimulation of HBV-specific T-cell receptors with viral peptides and circulating antigens. As a result, T-cell polyfunctionality is decreased (i.e., decreased levels of IL-2, tumor necrosis factor (TNF)-a, IFN-g, and lack of proliferation).
A safe and effective prophylactic vaccine against HBV infection has been available since the 1980s and is the mainstay of hepatitis B prevention (World Health Organization, Hepatitis B: Fact sheet No. 204 [Internet] 2015 March.). The World Health Organization recommends vaccination of all infants, and, in countries where there is low or intermediate hepatitis B endemicity, vaccination of all children and adolescents (<18 years of age), and of people of certain at risk population categories. Due to vaccination, worldwide infection rates have dropped dramatically. However, prophylactic vaccines do not cure established HBV infection. Chronic HBV is currently treated with IFN-a and nucleoside or nucleotide analogs, but there is no ultimate cure due to the persistence in infected hepatocytes of an intracellular viral replication intermediate called covalently closed circular DNA (cccDNA), which plays a fundamental role as a template for viral RNAs, and thus new virions. It is thought that induced virus-specific T-cell and B-cell responses can effectively eliminate cccDNA-carrying hepatocytes. Current therapies targeting the HBV polymerase suppress viremia, but offer limited effect on cccDNA that resides in the nucleus and related production of circulating antigen. The most rigorous form of a cure may be elimination of HBV cccDNA from the organism, which has neither been observed as a naturally occurring outcome nor as a result of any therapeutic intervention. However, loss of HBV surface antigens (HBsAg) is a clinically credible equivalent of a cure, since disease relapse can occur only in cases of severe immunosuppression, which can then be prevented by prophylactic treatment. Thus, at least from a clinical standpoint, loss of HBsAg is associated with the most stringent form of immune reconstitution against HBV.
For example, immune modulation with pegylated interferon (pegIFN)-a has proven better in comparison to nucleoside or nucleotide therapy in terms of sustained off-treatment response with a finite treatment course. Besides a direct antiviral effect, IFN-a is reported to exert epigenetic suppression of cccDNA in cell culture and humanized mice, which leads to reduction of virion productivity and transcripts (Belloni et al. J. Clin. Invest. (2012) 122(2), 529-537). However, this therapy is still fraught with side-effects and overall responses are rather low, in part because IFN-a has only poor modulatory influences on HBV-specific T-cells. In particular, cure rates are low (< 10%) and toxicity is high. Likewise, direct acting HBV antivirals, namely the HBV polymerase inhibitors entecavir and tenofovir, are effective as monotherapy in inducing viral suppression with a high genetic barrier to emergence of drug resistant mutants and consecutive prevention of liver disease progression. However, cure of chronic hepatitis B, defined by HBsAg loss or seroconversion, is rarely achieved with such HBV polymerase inhibitors. Therefore, these antivirals in theory need to be administered indefinitely to prevent reoccurrence of liver disease, similar to antiretroviral therapy for human immunodeficiency virus (HIV).
Therapeutic vaccination has the potential to eliminate HBV from chronically infected patients (Michel et al. J. Hepatol. (2011) 54(6), 1286-1296). Many strategies have been explored, but to date therapeutic vaccination has not proven successful. BRIEF SUMMARY OF THE INVENTION
Accordingly, there is an unmet medical need in the treatment of hepatitis B virus (HBV), particularly chronic HBV, for a finite well-tolerated treatment with a higher cure rate. The invention satisfies this need by providing therapeutic combinations or compositions and methods for inducing an immune response against hepatitis B viruses (HBV) infection. The immunogenic compositions/combinations and methods of the invention can be used to provide therapeutic immunity to a subject, such as a subject having chronic HBV infection.
In a general aspect, the application relates to therapeutic combinations or compositions comprising one or more HBV antigens, or one or more polynucleotides encoding the HBV antigens, and a capsid assembly modulator, for use in treating an HBV infection in a subject in need thereof.
In one embodiment, the therapeutic combination comprises:
i) at least one of:
a) a truncated HBV core antigen consisting of an amino acid sequence that is at least 95%, such as at least 95%, 96%, 97%, 98%, 99% or 100%, identical to SEQ ID NO: 2, b) a first non-naturally occurring nucleic acid molecule comprising a first polynucleotide sequence encoding the truncated HBV core antigen;
c) an HBV polymerase antigen having an amino acid sequence that is at least 90%, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%, identical to SEQ ID NO: 7, wherein the HBV polymerase antigen does not have reverse transcriptase activity and RNase H activity, and
d) a second non-naturally occurring nucleic acid molecule comprising a second
polynucleotide sequence encoding the HBV polymerase antigen; and
ii) a capsid assembly modulator, such as those described herein.
In one embodiment, the truncated HBV core antigen consists of the amino acid sequence of SEQ ID NO: 2 or SEQ ID NO: 4, and the HBV polymerase antigen comprises the amino acid sequence of SEQ ID NO: 7.
In one embodiment, the therapeutic combination comprises at least one of the HBV polymerase antigen and the truncated HBV core antigen. In certain embodiments, the therapeutic combination comprises the HBV polymerase antigen and the truncated HBV core antigen. In one embodiment, the therapeutic combination comprises at least one of the first non- naturally occurring nucleic acid molecule comprising the first polynucleotide sequence encoding the truncated HBV core antigen, and the second non-naturally occurring nucleic acid molecule comprising the second polynucleotide sequence encoding the HBV polymerase antigen. In certain embodiments, the first non-naturally occurring nucleic acid molecule further comprises a polynucleotide sequence encoding a signal sequence operably linked to the N-terminus of the truncated HBV core antigen, and the second non-naturally occurring nucleic acid molecule further comprises a polynucleotide sequence encoding a signal sequence operably linked to the N-terminus of the HBV polymerase antigen, preferably, the signal sequence independently comprises the amino acid sequence of SEQ ID NO: 9 or SEQ ID NO: 15, more preferably, the signal sequence is encoded by the polynucleotide sequence of SEQ ID NO: 8 or SEQ ID NO: 14, respectively.
In certain embodiments, the first polynucleotide sequence comprises the polynucleotide sequence having at least 90%, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%, sequence identity to SEQ ID NO: 1 or SEQ ID NO: 3.
In certain embodiments, the second polynucleotide sequence comprises a polynucleotide sequence having at least 90%, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%, sequence identity to SEQ ID NO: 5 or SEQ ID NO: 6.
In certain embodiments, the capsid modulators useful for the invention, as well as related information such as its structure, production, biological activities, therapeutic applications, etc., are described in International Patent Application Publications WO 2014106019, WO
2017156255, and WO 2018039531, and U.S. Patent Application Publications US 20160024004 and US 20180161307, the contents of which are hereby incorporated by reference in their entireties.
In an embodiment, a therapeutic combination comprises:
a) a first non-naturally occurring nucleic acid molecule comprising a first
polynucleotide sequence encoding a truncated HBV core antigen consisting of an amino acid sequence that is at least 95%, such as at least 95%, 96%, 97%, 98%, 99% or 100%, identical to SEQ ID NO: 2;
b) a second non-naturally occurring nucleic acid molecule comprising a second
polynucleotide sequence encoding an HBV polymerase antigen having an amino acid sequence that is at least 90%, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%, identical to SEQ ID NO: 7, wherein the HBV polymerase antigen does not have reverse transcriptase activity and RNase H activity; and
c) a capsid assembly modulator selected from the group consisting of:
i. a compound of formula (I), (XI), (XVIII), or (XXI), or an enantiomer,
diastereomer or pharmaceutically accepted salt, hydrate, or solvate thereof, including a hydrate, a solvate, a pharmaceutically acceptable salt, a prodrug, or a complex thereof described herein;
ii. a compound of formula (A-I), (A-II), (A-III), (A-IV), (A-V), (A-VI), or
(AVII), or an enantiomer, diastereomer or pharmaceutically accepted salt, hydrate, or solvate thereof, including a hydrate, a solvate, a pharmaceutically acceptable salt, a prodrug, or a complex thereof described herein; and iii. a compound of formula of formula (B-I), or a pharmaceutical salt described herein.
Preferably, the therapeutic combination comprises a) a first non-naturally occurring nucleic acid molecule comprising a first polynucleotide sequence encoding an truncated HBV core antigen consisting of the amino acid sequence of SEQ ID NO: 2 or SEQ ID NO: 4; b) a second non-naturally occurring nucleic acid molecule comprising a second polynucleotide sequence encoding an HBV polymerase antigen having the amino acid sequence of SEQ ID NO: 7, and (c) a compound selected from the group consisting of the exemplified compounds, or an enantiomer, diastereomer or pharmaceutically accepted salt, hydrate, or solvate thereof, including a hydrate, a solvate, a pharmaceutically acceptable salt, a prodrug, or a complex thereof.
Preferably, the therapeutic combination comprises a first non-naturally occurring nucleic acid molecule comprising a polynucleotide sequence having at least 90%, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%, sequence identity to SEQ ID NO: 1 or SEQ ID NO: 3, and a second non-naturally occurring nucleic acid molecule comprising the polynucleotide sequence having at least 90%, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%, sequence identity to SEQ ID NO: 5 or SEQ ID NO: 6. More preferably, the therapeutic combination comprises a) a first non-naturally occurring nucleic acid molecule comprising a first polynucleotide sequence of SEQ ID NO: 1 or SEQ ID NO: 3; b) a second non-naturally occurring nucleic acid molecule comprising a second polynucleotide sequence of SEQ ID NO: 5 or 6; and c) a compound selected from the group consisting of:
Figure imgf000008_0001
Figure imgf000009_0001
, or a pharmaceutically acceptable salt thereof.
In an embodiment, each of the first and the second non-naturally occurring nucleic acid molecules is a DNA molecule, preferably the DNA molecule is present on a plasmid or a viral vector.
In another embodiment, each of the first and the second non-naturally occurring nucleic acid molecules is an RNA molecule, preferably an mRNA or a self-replicating RNA molecule. In some embodiments, each of the first and the second non-naturally occurring nucleic acid molecules is independently formulated with a lipid nanoparticle (LNP).
In another general aspect, the application relates to a kit comprising a therapeutic combination of the application.
The application also relates to a therapeutic combination or kit of the application for use in inducing an immune response against hepatitis B virus (HBV); and use of a therapeutic combination, composition or kit of the application in the manufacture of a medicament for inducing an immune response against hepatitis B virus (HBV). The use can further comprise a combination with another immunogenic or therapeutic agent, preferably another HBV antigen or another HBV therapy. Preferably, the subject has chronic HBV infection.
The application further relates to a therapeutic combination or kit of the application for use in treating an HBV-induced disease in a subject in need thereof; and use of therapeutic combination or kit of the application in the manufacture of a medicament for treating an HBV- induced disease in a subject in need thereof. The use can further comprise a combination with another therapeutic agent, preferably another anti-HBV antigen. Preferably, the subject has chronic HBV infection, and the HBV-induced disease is selected from the group consisting of advanced fibrosis, cirrhosis, and hepatocellular carcinoma (HCC).
The application also relates to a method of inducing an immune response against an HBV or a method of treating an HBV infection or an HBV-induced disease, comprising administering to a subject in need thereof a therapeutic combination according to embodiments of the application.
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
The foregoing summary, as well as the following detailed description of preferred embodiments of the present application, will be better understood when read in conjunction with the appended drawings. It should be understood, however, that the application is not limited to the precise embodiments shown in the drawings. FIG.1A and FIG.1B show schematic representations of DNA plasmids according to embodiments of the application; FIG.1A shows a DNA plasmid encoding an HBV core antigen according to an embodiment of the application; FIG.1B shows a DNA plasmid encoding an HBV polymerase (pol) antigen according to an embodiment of the application; the HBV core and pol antigens are expressed under control of a CMV promoter with an N-terminal cystatin S signal peptide that is cleaved from the expressed antigen upon secretion from the cell; transcriptional regulatory elements of the plasmid include an enhancer sequence located between the CMV promoter and the polynucleotide sequence encoding the HBV antigen and a bGH polyadenylation sequence located downstream of the polynucleotide sequence encoding the HBV antigen; a second expression cassette is included in the plasmid in reverse orientation including a kanamycin resistance gene under control of an Ampr (bla) promoter; an origin of replication (pUC) is also included in reverse orientation.
FIG.2A and FIG.2B. show the schematic representations of the expression cassettes in adenoviral vectors according to embodiments of the application; FIG.2A shows the expression cassette for a truncated HBV core antigen, which contains a CMV promoter, an intron (a fragment derived from the human ApoAI gene - GenBank accession X01038 base pairs 295– 523, harboring the ApoAI second intron), a human immunoglobulin secretion signal, followed by a coding sequence for a truncated HBV core antigen and a SV40 polyadenylation signal; FIG.2B shows the expression cassette for a fusion protein of a truncated HBV core antigen operably linked to an HBV polymerase antigen, which is otherwise identical to the expression cassette for the truncated HBV core antigen except the HBV antigen.
FIG.3 shows ELISPOT responses of Balb/c mice immunized with different DNA plasmids expressing HBV core antigen or HBV pol antigen, as described in Example 3; peptide pools used to stimulate splenocytes isolated from the various vaccinated animal groups are indicated in gray scale; the number of responsive T-cells are indicated on the y-axis expressed as spot forming cells (SFC) per 106 splenocytes; DETAILED DESCRIPTION OF THE INVENTION
Various publications, articles and patents are cited or described in the background and throughout the specification; each of these references is herein incorporated by reference in its entirety. Discussion of documents, acts, materials, devices, articles or the like which has been included in the present specification is for the purpose of providing context for the invention. Such discussion is not an admission that any or all of these matters form part of the prior art with respect to any inventions disclosed or claimed.
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood to one of ordinary skill in the art to which this invention pertains. Otherwise, certain terms used herein have the meanings as set forth in the specification. All patents, published patent applications and publications cited herein are incorporated by reference as if set forth fully herein.
It must be noted that as used herein and in the appended claims, the singular forms“a,” “an,” and“the” include plural reference unless the context clearly dictates otherwise.
Unless otherwise indicated, the term“at least” preceding a series of elements is to be understood to refer to every element in the series. Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be
encompassed by the invention.
Throughout this specification and the claims which follow, unless the context requires otherwise, the word“comprise”, and variations such as“comprises” and“comprising”, will be understood to imply the inclusion of a stated integer or step or group of integers or steps but not the exclusion of any other integer or step or group of integers or steps. When used herein the term“comprising” can be substituted with the term“containing” or“including” or sometimes when used herein with the term“having”.
When used herein“consisting of” excludes any element, step, or ingredient not specified in the claim element. When used herein,“consisting essentially of” does not exclude materials or steps that do not materially affect the basic and novel characteristics of the claim. Any of the aforementioned terms of“comprising”,“containing”,“including”, and“having”, whenever used herein in the context of an aspect or embodiment of the application can be replaced with the term “consisting of” or“consisting essentially of” to vary scopes of the disclosure.
As used herein, the conjunctive term“and/or” between multiple recited elements is understood as encompassing both individual and combined options. For instance, where two elements are conjoined by“and/or,” a first option refers to the applicability of the first element without the second. A second option refers to the applicability of the second element without the first. A third option refers to the applicability of the first and second elements together. Any one of these options is understood to fall within the meaning, and therefore satisfy the requirement of the term“and/or” as used herein. Concurrent applicability of more than one of the options is also understood to fall within the meaning, and therefore satisfy the requirement of the term“and/or.” Unless otherwise stated, any numerical value, such as a concentration or a concentration range described herein, are to be understood as being modified in all instances by the term “about.” Thus, a numerical value typically includes ± 10% of the recited value. For example, a concentration of 1 mg/mL includes 0.9 mg/mL to 1.1 mg/mL. Likewise, a concentration range of 1 mg/mL to 10 mg/mL includes 0.9 mg/mL to 11 mg/mL. As used herein, the use of a numerical range expressly includes all possible subranges, all individual numerical values within that range, including integers within such ranges and fractions of the values unless the context clearly indicates otherwise.
The phrases“percent (%) sequence identity” or“% identity” or“% identical to” when used with reference to an amino acid sequence describe the number of matches (“hits”) of identical amino acids of two or more aligned amino acid sequences as compared to the number of amino acid residues making up the overall length of the amino acid sequences. In other terms, using an alignment, for two or more sequences the percentage of amino acid residues that are the same (e.g.90%, 91%, 92%, 93%, 94%, 95%, 97%, 98%, 99%, or 100% identity over the full- length of the amino acid sequences) may be determined, when the sequences are compared and aligned for maximum correspondence as measured using a sequence comparison algorithm as known in the art, or when manually aligned and visually inspected. The sequences which are compared to determine sequence identity may thus differ by substitution(s), addition(s) or deletion(s) of amino acids. Suitable programs for aligning protein sequences are known to the skilled person. The percentage sequence identity of protein sequences can, for example, be determined with programs such as CLUSTALW, Clustal Omega, FASTA or BLAST, e.g. using the NCBI BLAST algorithm (Altschul SF, et al (1997), Nucleic Acids Res.25:3389-3402).
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” can be administration of the two or more therapies or components at least within the same day. When two components are“administered together with” or“administered in combination with,” they can be administered in separate compositions sequentially within a short time period, such as 24, 20, 16, 12, 8 or 4 hours, or within 1 hour, or they can be administered in a single composition at the same time.“Subsequent administration” can be administration of the two or more therapies or components in the same day or on separate days. 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. first DNA plasmid encoding an HBV antigen) 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 (e.g., second DNA plasmid encoding an HBV antigen), and/or a third therapy or component (e.g.,capsid assembly modulator). In some embodiments, a first therapy or component (e.g. first DNA plasmid encoding an HBV antigen), a second therapy or component (e.g., second DNA plasmid encoding an HBV antigen), and a third therapy or component (e.g., capsid assembly modulator) are administered in the same composition. In other embodiments, a first therapy or component (e.g. first DNA plasmid encoding an HBV antigen), a second therapy or component (e.g., second DNA plasmid encoding an HBV antigen), and a third therapy or component (e.g., capsid assembly modulator) are administered in separate compositions, such as two or three separate compositions.
As used herein, a“non-naturally occurring” nucleic acid or polypeptide, refers to a nucleic acid or polypeptide that does not occur in nature. A“non-naturally occurring” nucleic acid or polypeptide can be synthesized, treated, fabricated, and/or otherwise manipulated in a laboratory and/or manufacturing setting. In some cases, a non-naturally occurring nucleic acid or polypeptide can comprise a naturally-occurring nucleic acid or polypeptide that is treated, processed, or manipulated to exhibit properties that were not present in the naturally-occurring nucleic acid or polypeptide, prior to treatment. As used herein, a“non-naturally occurring” nucleic acid or polypeptide can be a nucleic acid or polypeptide isolated or separated from the natural source in which it was discovered, and it lacks covalent bonds to sequences with which it was associated in the natural source. A“non-naturally occurring” nucleic acid or polypeptide can be made recombinantly or via other methods, such as chemical synthesis. As used herein,“subject” means any animal, preferably a mammal, most preferably a human, to whom will be or has been treated by a method according to an embodiment of the application. The term“mammal” as used herein, encompasses any mammal. Examples of mammals include, but are not limited to, cows, horses, sheep, pigs, cats, dogs, mice, rats, rabbits, guinea pigs, non-human primates (NHPs) such as monkeys or apes, humans, etc., more preferably a human.
As used herein, the term“operably linked” refers to a linkage or a juxtaposition wherein the components so described are in a relationship permitting them to function in their intended manner. For example, a regulatory sequence operably linked to a nucleic acid sequence of interest is capable of directing the transcription of the nucleic acid sequence of interest, or a signal sequence operably linked to an amino acid sequence of interest is capable of secreting or translocating the amino acid sequence of interest over a membrane.
In an attempt to help the reader of the application, the description has been separated in various paragraphs or sections, or is directed to various embodiments of the application. These separations should not be considered as disconnecting the substance of a paragraph or section or embodiments from the substance of another paragraph or section or embodiments. To the contrary, one skilled in the art will understand that the description has broad application and encompasses all the combinations of the various sections, paragraphs and sentences that can be contemplated. The discussion of any embodiment is meant only to be exemplary and is not intended to suggest that the scope of the disclosure, including the claims, is limited to these examples. For example, while embodiments of HBV vectors of the application (e.g., plasmid DNA or viral vectors) described herein may contain particular components, including, but not limited to, certain promoter sequences, enhancer or regulatory sequences, signal peptides, coding sequence of an HBV antigen, polyadenylation signal sequences, etc. arranged in a particular order, those having ordinary skill in the art will appreciate that the concepts disclosed herein may equally apply to other components arranged in other orders that can be used in HBV vectors of the application. The application contemplates use of any of the applicable components in any combination having any sequence that can be used in HBV vectors of the application, whether or not a particular combination is expressly described. The invention generally relates to a therapeutic combination comprising one or more HBV antigens and at least one capsid assembly modulator. Hepatitis B Virus (HBV)
As used herein“hepatitis B virus” or“HBV” refers to a virus of the hepadnaviridae family. HBV is a small (e.g., 3.2 kb) hepatotropic DNA virus that encodes four open reading frames and seven proteins. The seven proteins encoded by HBV include small (S), medium (M), and large (L) surface antigen (HBsAg) or envelope (Env) proteins, pre-Core protein, core protein, viral polymerase (Pol), and HBx protein. HBV expresses three surface antigens, or envelope proteins, L, M, and S, with S being the smallest and L being the largest. The extra domains in the M and L proteins are named Pre-S2 and Pre-S1, respectively. Core protein is the subunit of the viral nucleocapsid. Pol is needed for synthesis of viral DNA (reverse transcriptase, RNaseH, and primer), which takes place in nucleocapsids localized to the cytoplasm of infected hepatocytes. PreCore is the core protein with an N-terminal signal peptide and is proteolytically processed at its N and C termini before secretion from infected cells, as the so-called hepatitis B e-antigen (HBeAg). HBx protein is required for efficient transcription of covalently closed circular DNA (cccDNA). HBx is not a viral structural protein. All viral proteins of HBV have their own mRNA except for core and polymerase, which share an mRNA. With the exception of the protein pre-Core, none of the HBV viral proteins are subject to post-translational proteolytic processing.
The HBV virion contains a viral envelope, nucleocapsid, and single copy of the partially double-stranded DNA genome. The nucleocapsid comprises 120 dimers of core protein and is covered by a capsid membrane embedded with the S, M, and L viral envelope or surface antigen proteins. After entry into the cell, the virus is uncoated and the capsid-containing relaxed circular DNA (rcDNA) with covalently bound viral polymerase migrates to the nucleus. During that process, phosphorylation of the core protein induces structural changes, exposing a nuclear localization signal enabling interaction of the capsid with so-called importins. These importins mediate binding of the core protein to nuclear pore complexes upon which the capsid
disassembles and polymerase/rcDNA complex is released into the nucleus. Within the nucleus the rcDNA becomes deproteinized (removal of polymerase) and is converted by host DNA repair machinery to a covalently closed circular DNA (cccDNA) genome from which overlapping transcripts encode for HBeAg, HBsAg, Core protein, viral polymerase and HBx protein. Core protein, viral polymerase, and pre-genomic RNA (pgRNA) associate in the cytoplasm and self- assemble into immature pgRNA-containing capsid particles, which further convert into mature rcDNA-capsids and function as a common intermediate that is either enveloped and secreted as infectious virus particles or transported back to the nucleus to replenish and maintain a stable cccDNA pool.
To date, HBV is divided into four serotypes (adr, adw, ayr, ayw) based on antigenic epitopes present on the envelope proteins, and into eight genotypes (A, B, C, D, E, F, G, and H) based on the sequence of the viral genome. The HBV genotypes are distributed over different geographic regions. For example, the most prevalent genotypes in Asia are genotypes B and C. Genotype D is dominant in Africa, the Middle East, and India, whereas genotype A is widespread in Northern Europe, sub-Saharan Africa, and West Africa.
HBV Antigens
As used herein, the terms“HBV antigen,”“antigenic polypeptide of HBV,”“HBV antigenic polypeptide,”“HBV antigenic protein,”“HBV immunogenic polypeptide,” and“HBV immunogen” all refer to a polypeptide capable of inducing an immune response, e.g., a humoral and/or cellular mediated response, against an HBV in a subject. The HBV antigen can be a polypeptide of HBV, a fragment or epitope thereof, or a combination of multiple HBV polypeptides, portions or derivatives thereof. An HBV antigen is capable of raising in a host a protective immune response, e.g., inducing an immune response against a viral disease or infection, and/or producing an immunity (i.e., vaccinates) in a subject against a viral disease or infection, that protects the subject against the viral disease or infection. For example, an HBV antigen can comprise a polypeptide or immunogenic fragment(s) thereof from any HBV protein, such as HBeAg, pre-core protein, HBsAg (S, M, or L proteins), core protein, viral polymerase, or HBx protein derived from any HBV genotype, e.g., genotype A, B, C, D, E, F, G, and/or H, or combination thereof.
(1) HBV Core Antigen
As used herein, each of the terms“HBV core antigen,”“HBc” and“core antigen” refers to an HBV antigen capable of inducing an immune response, e.g., a humoral and/or cellular mediated response, against an HBV core protein in a subject. Each of the terms“core,”“core polypeptide,” and“core protein” refers to the HBV viral core protein. Full-length core antigen is typically 183 amino acids in length and includes an assembly domain (amino acids 1 to 149) and a nucleic acid binding domain (amino acids 150 to 183). The 34-residue nucleic acid binding domain is required for pre-genomic RNA encapsidation. This domain also functions as a nuclear import signal. It comprises 17 arginine residues and is highly basic, consistent with its function. HBV core protein is dimeric in solution, with the dimers self-assembling into icosahedral capsids. Each dimer of core protein has four a-helix bundles flanked by an a-helix domain on either side. Truncated HBV core proteins lacking the nucleic acid binding domain are also capable of forming capsids.
In an embodiment of the application, an HBV antigen is a truncated HBV core antigen. As used herein, a“truncated HBV core antigen,” refers to an HBV antigen that does not contain the entire length of an HBV core protein, but is capable of inducing an immune response against the HBV core protein in a subject. For example, an HBV core antigen can be modified to delete one or more amino acids of the highly positively charged (arginine rich) C-terminal nucleic acid binding domain of the core antigen, which typically contains seventeen arginine (R) residues. A truncated HBV core antigen of the application is preferably a C-terminally truncated HBV core protein which does not comprise the HBV core nuclear import signal and/or a truncated HBV core protein from which the C-terminal HBV core nuclear import signal has been deleted. In an embodiment, a truncated HBV core antigen comprises a deletion in the C-terminal nucleic acid binding domain, such as a deletion of 1 to 34 amino acid residues of the C-terminal nucleic acid binding domain, e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, or 34 amino acid residues, preferably a deletion of all 34 amino acid residues. In a preferred embodiment, a truncated HBV core antigen comprises a deletion in the C-terminal nucleic acid binding domain, preferably a deletion of all 34 amino acid residues.
An HBV core antigen of the application can be a consensus sequence derived from multiple HBV genotypes (e.g., genotypes A, B, C, D, E, F, G, and H). As used herein, “consensus sequence” means an artificial sequence of amino acids based on an alignment of amino acid sequences of homologous proteins, e.g., as determined by an alignment (e.g., using Clustal Omega) of amino acid sequences of homologous proteins. It can be the calculated order of most frequent amino acid residues, found at each position in a sequence alignment, based upon sequences of HBV antigens (e.g., core, pol, etc.) from at least 100 natural HBV isolates. A consensus sequence can be non-naturally occurring and different from the native viral sequences. Consensus sequences can be designed by aligning multiple HBV antigen sequences from different sources using a multiple sequence alignment tool, and at variable alignment positions, selecting the most frequent amino acid. Preferably, a consensus sequence of an HBV antigen is derived from HBV genotypes B, C, and D. The term“consensus antigen” is used to refer to an antigen having a consensus sequence.
An exemplary truncated HBV core antigen according to the application lacks the nucleic acid binding function, and is capable of inducing an immune response in a mammal against at least two HBV genotypes. Preferably a truncated HBV core antigen is capable of inducing a T cell response in a mammal against at least HBV genotypes B, C and D. More preferably, a truncated HBV core antigen is capable of inducing a CD8 T cell response in a human subject against at least HBV genotypes A, B, C and D.
Preferably, an HBV core antigen of the application is a consensus antigen, preferably a consensus antigen derived from HBV genotypes B, C, and D, more preferably a truncated consensus antigen derived from HBV genotypes B, C, and D. An exemplary truncated HBV core consensus antigen according to the application consists of an amino acid sequence that is at least 90% identical to SEQ ID NO: 2 or SEQ ID NO: 4, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9%, or 100% identical to SEQ ID NO: 2 or SEQ ID NO: 4. SEQ ID NO: 2 and SEQ ID NO: 4 are core consensus antigens derived from HBV genotypes B, C, and D. SEQ ID NO: 2 and SEQ ID NO: 4 each contain a 34-amino acid C-terminal deletion of the highly positively charged (arginine rich) nucleic acid binding domain of the native core antigen.
In one embodiment of the application, an HBV core antigen is a truncated HBV antigen consisting of the amino acid sequence of SEQ ID NO: 2. In another embodiment, an HBV core antigen is a truncated HBV antigen consisting of the amino acid sequence of SEQ ID NO: 4. In another embodiment, an HBV core antigen further contains a signal sequence operably linked to the N-terminus of a mature HBV core antigen sequence, such as the amino acid sequence of SEQ ID NO: 2 or SEQ ID NO: 4. Preferably, the signal sequence has the amino acid sequence of SEQ ID NO: 9 or SEQ ID NO: 15.
(2) HBV Polymerase Antigen
As used herein, the term“HBV polymerase antigen,”“HBV Pol antigen” or“HBV pol antigen” refers to an HBV antigen capable of inducing an immune response, e.g., a humoral and/or cellular mediated response, against an HBV polymerase in a subject. Each of the terms “polymerase,”“polymerase polypeptide,”“Pol” and“pol” refers to the HBV viral DNA polymerase. The HBV viral DNA polymerase has four domains, including, from the N terminus to the C terminus, a terminal protein (TP) domain, which acts as a primer for minus-strand DNA synthesis; a spacer that is nonessential for the polymerase functions; a reverse transcriptase (RT) domain for transcription; and a RNase H domain.
In an embodiment of the application, an HBV antigen comprises an HBV Pol antigen, or any immunogenic fragment or combination thereof. An HBV Pol antigen can contain further modifications to improve immunogenicity of the antigen, such as by introducing mutations into the active sites of the polymerase and/or RNase domains to decrease or substantially eliminate certain enzymatic activities.
Preferably, an HBV Pol antigen of the application does not have reverse transcriptase activity and RNase H activity, and is capable of inducing an immune response in a mammal against at least two HBV genotypes. Preferably, an HBV Pol antigen is capable of inducing a T cell response in a mammal against at least HBV genotypes B, C and D. More preferably, an HBV Pol antigen is capable of inducing a CD8 T cell response in a human subject against at least HBV genotypes A, B, C and D.
Thus, in some embodiments, an HBV Pol antigen is an inactivated Pol antigen. In an embodiment, an inactivated HBV Pol antigen comprises one or more amino acid mutations in the active site of the polymerase domain. In another embodiment, an inactivated HBV Pol antigen comprises one or more amino acid mutations in the active site of the RNaseH domain. In a preferred embodiment, an inactivated HBV pol antigen comprises one or more amino acid mutations in the active site of both the polymerase domain and the RNaseH domain. For example, the“YXDD” motif in the polymerase domain of an HBV pol antigen that can be required for nucleotide/metal ion binding can be mutated, e.g., by replacing one or more of the aspartate residues (D) with asparagine residues (N), eliminating or reducing metal coordination function, thereby decreasing or substantially eliminating reverse transcriptase function.
Alternatively, or in addition to mutation of the“YXDD” motif, the“DEDD” motif in the RNaseH domain of an HBV pol antigen required for Mg2+ coordination can be mutated, e.g., by replacing one or more aspartate residues (D) with asparagine residues (N) and/or replacing the glutamate residue (E) with glutamine (Q), thereby decreasing or substantially eliminating RNaseH function. In a particular embodiment, an HBV pol antigen is modified by (1) mutating the aspartate residues (D) to asparagine residues (N) in the“YXDD” motif of the polymerase domain; and (2) mutating the first aspartate residue (D) to an asparagine residue (N) and the first glutamate residue (E) to a glutamine residue (N) in the“DEDD” motif of the RNaseH domain, thereby decreasing or substantially eliminating both the reverse transcriptase and RNaseH functions of the pol antigen.
In a preferred embodiment of the application, an HBV pol antigen is a consensus antigen, preferably a consensus antigen derived from HBV genotypes B, C, and D, more preferably an inactivated consensus antigen derived from HBV genotypes B, C, and D. An exemplary HBV pol consensus antigen according to the application comprises an amino acid sequence that is at least 90% identical to SEQ ID NO: 7, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100% identical to SEQ ID NO: 7, preferably at least 98% identical to SEQ ID NO: 7, such as at least 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100% identical to SEQ ID NO: 7. SEQ ID NO: 7 is a pol consensus antigen derived from HBV genotypes B, C, and D comprising four mutations located in the active sites of the polymerase and RNaseH domains. In particular, the four mutations include mutation of the aspartic acid residues (D) to asparagine residues (N) in the“YXDD” motif of the polymerase domain; and mutation of the first aspartate residue (D) to an asparagine residue (N) and mutation of the glutamate residue (E) to a glutamine residue (Q) in the“DEDD” motif of the RNaseH domain.
In a particular embodiment of the application, an HBV pol antigen comprises the amino acid sequence of SEQ ID NO: 7. In other embodiments of the application, an HBV pol antigen consists of the amino acid sequence of SEQ ID NO: 7. In a further embodiment, an HBV pol antigen further contains a signal sequence operably linked to the N-terminus of a mature HBV pol antigen sequence, such as the amino acid sequence of SEQ ID NO: 7. Preferably, the signal sequence has the amino acid sequence of SEQ ID NO: 9 or SEQ ID NO: 15.
(3) Fusion of HBV Core Antigen and HBV Polymerase Antigen
As used herein the term“fusion protein” or“fusion” refers to a single polypeptide chain having at least two polypeptide domains that are not normally present in a single, natural polypeptide. In an embodiment of the application, an HBV antigen comprises a fusion protein comprising a truncated HBV core antigen operably linked to an HBV Pol antigen, or an HBV Pol antigen operably linked to a truncated HBV core antigen, preferably via a linker.
For example, in a fusion protein containing a first polypeptide and a second heterologous polypeptide, a linker serves primarily as a spacer between the first and second polypeptides. In an embodiment, a linker is made up of amino acids linked together by peptide bonds, preferably from 1 to 20 amino acids linked by peptide bonds, wherein the amino acids are selected from the 20 naturally occurring amino acids. In an embodiment, the 1 to 20 amino acids are selected from glycine, alanine, proline, asparagine, glutamine, and lysine. Preferably, a linker is made up of a majority of amino acids that are sterically unhindered, such as glycine and alanine. Exemplary linkers are polyglycines, particularly (Gly)5, (Gly)8; poly(Gly-Ala), and polyalanines. One exemplary suitable linker as shown in the Examples below is (AlaGly)n, wherein n is an integer of 2 to 5.
Preferably, a fusion protein of the application is capable of inducing an immune response in a mammal against HBV core and HBV Pol of at least two HBV genotypes. Preferably, a fusion protein is capable of inducing a T cell response in a mammal against at least HBV genotypes B, C and D. More preferably, the fusion protein is capable of inducing a CD8 T cell response in a human subject against at least HBV genotypes A, B, C and D.
In an embodiment of the application, a fusion protein comprises a truncated HBV core antigen having an amino acid sequence at least 90%, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9%, or 100% identical to SEQ ID NO: 2 or SEQ ID NO: 4, a linker, and an HBV Pol antigen having an amino acid sequence at least 90%, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9%, or 100%, identical to SEQ ID NO: 7.
In a preferred embodiment of the application, a fusion protein comprises a truncated HBV core antigen consisting of the amino acid sequence of SEQ ID NO: 2 or SEQ ID NO: 4, a linker comprising (AlaGly)n, wherein n is an integer of 2 to 5, and an HBV Pol antigen having the amino acid sequence of SEQ ID NO: 7. More preferably, a fusion protein according to an embodiment of the application comprises the amino acid sequence of SEQ ID NO: 16. In one embodiment of the application, a fusion protein further comprises a signal sequence operably linked to the N-terminus of the fusion protein. Preferably, the signal sequence has the amino acid sequence of SEQ ID NO: 9 or SEQ ID NO: 15. In one embodiment, a fusion protein comprises the amino acid sequence of SEQ ID NO: 17.
Additional disclosure on HBV vaccines that can be used for the present invention are described in U.S. Patent Application No: 16/223,251, filed December 18, 2018, the contents of the application, more preferably the examples of the application, are hereby incorporated by reference in their entireties.
Polynucleotides and Vectors
In another general aspect, the application provides a non-naturally occurring nucleic acid molecule encoding an HBV antigen useful for an invention according to embodiments of the application, and vectors comprising the non-naturally occurring nucleic acid. A first or second non-naturally occurring nucleic acid molecule can comprise any polynucleotide sequence encoding an HBV antigen useful for the application, which can be made using methods known in the art in view of the present disclosure. Preferably, a first or second polynucleotide encodes at least one of a truncated HBV core antigen and an HBV polymerase antigen of the application. A polynucleotide can be in the form of RNA or in the form of DNA obtained by recombinant techniques (e.g., cloning) or produced synthetically (e.g., chemical synthesis). The DNA can be single-stranded or double-stranded, or can contain portions of both double-stranded and single- stranded sequence. The DNA can, for example, comprise genomic DNA, cDNA, or
combinations thereof. The polynucleotide can also be a DNA/RNA hybrid. The polynucleotides and vectors of the application can be used for recombinant protein production, expression of the protein in host cell, or the production of viral particles. Preferably, a polynucleotide is DNA.
In an embodiment of the application, a first non-naturally occurring nucleic acid molecule comprises a first polynucleotide sequence encoding a truncated HBV core antigen consisting of an amino acid sequence that is at least 90% identical to SEQ ID NO: 2 or SEQ ID NO: 4, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100% identical to SEQ ID NO: 2, preferably 98%, 99% or 100% identical to SEQ ID NO: 2 or SEQ ID NO: 4. In a particular embodiment of the application, a first non-naturally occurring nucleic acid molecule comprises a first polynucleotide sequence encoding a truncated HBV core antigen consisting the amino acid sequence of SEQ ID NO: 2 or SEQ ID NO: 4.
Examples of polynucleotide sequences of the application encoding a truncated HBV core antigen consisting of the amino acid sequence of SEQ ID NO: 2 or SEQ ID NO: 4 include, but are not limited to, a polynucleotide sequence at least 90% identical to SEQ ID NO: 1 or SEQ ID NO: 3, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100% identical to SEQ ID NO: 1 or SEQ ID NO: 3, preferably 98%, 99% or 100% identical to SEQ ID NO: 1 or SEQ ID NO: 3. Exemplary non-naturally occurring nucleic acid molecules encoding a truncated HBV core antigen have the polynucleotide sequence of SEQ ID NOs: 1 or 3.
In another embodiment, a first non-naturally occurring nucleic acid molecule further comprises a coding sequence for a signal sequence that is operably linked to the N-terminus of the HBV core antigen sequence. Preferably, the signal sequence has the amino acid sequence of SEQ ID NO: 9 or SEQ ID NO: 15. More preferably, the coding sequence for a signal sequence comprises the polynucleotide sequence of SEQ ID NO: 8 or SEQ ID NO: 14.
In an embodiment of the application, a second non-naturally occurring nucleic acid molecule comprises a second polynucleotide sequence encoding an HBV polymerase antigen comprising an amino acid sequence that is at least 90% identical to SEQ ID NO: 7, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100% identical to SEQ ID NO: 7, preferably 100% identical to SEQ ID NO: 7. In a particular embodiment of the application, a second non-naturally occurring nucleic acid molecule comprises a second polynucleotide sequence encoding an HBV polymerase antigen consisting of the amino acid sequence of SEQ ID NO: 7.
Examples of polynucleotide sequences of the application encoding an HBV Pol antigen comprising the amino acid sequence of at least 90% identical to SEQ ID NO: 7 include, but are not limited to, a polynucleotide sequence at least 90% identical to SEQ ID NO: 5 or SEQ ID NO: 6, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100% identical to SEQ ID NO: 5 or SEQ ID NO: 6, preferably 98%, 99% or 100% identical to SEQ ID NO: 5 or SEQ ID NO: 6. Exemplary non-naturally occurring nucleic acid molecules encoding an HBV pol antigen have the polynucleotide sequence of SEQ ID NOs: 5 or 6.
In another embodiment, a second non-naturally occurring nucleic acid molecule further comprises a coding sequence for a signal sequence that is operably linked to the N-terminus of the HBV pol antigen sequence, such as the amino acid sequence of SEQ ID NO: 7. Preferably, the signal sequence has the amino acid sequence of SEQ ID NO: 9 or SEQ ID NO: 15. More preferably, the coding sequence for a signal sequence comprises the polynucleotide sequence of SEQ ID NO: 8 or SEQ ID NO: 14.
In another embodiment of the application, a non-naturally occurring nucleic acid molecule encodes an HBV antigen fusion protein comprising a truncated HBV core antigen operably linked to an HBV Pol antigen, or an HBV Pol antigen operably linked to a truncated HBV core antigen. In a particular embodiment, a non-naturally occurring nucleic acid molecule of the application encodes a truncated HBV core antigen consisting of an amino acid sequence that is at least 90% identical to SEQ ID NO: 2 or SEQ ID NO: 4, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100% identical to SEQ ID NO: 2 or SEQ ID NO: 4, preferably 100% identical to SEQ ID NO: 2 or SEQ ID NO: 4, more preferably 100% identical to SEQ ID NO: 2 or SEQ ID NO:4; a linker; and an HBV polymerase antigen comprising an amino acid sequence that is at least 90% identical to SEQ ID NO: 7, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100% identical to SEQ ID NO: 7, preferably 98%, 99% or 100% identical to SEQ ID NO: 7. In a particular embodiment of the application, a non-naturally occurring nucleic acid molecule encodes a fusion protein comprising a truncated HBV core antigen consisting of the amino acid sequence of SEQ ID NO: 2 or SEQ ID NO: 4, a linker comprising (AlaGly)n, wherein n is an integer of 2 to 5; and an HBV Pol antigen comprising the amino acid sequence of SEQ ID NO: 7. In a particular embodiment of the application, a non-naturally occurring nucleic acid molecule encodes an HBV antigen fusion protein comprising the amino acid sequence of SEQ ID NO: 16.
Examples of polynucleotide sequences of the application encoding an HBV antigen fusion protein include, but are not limited to, a polynucleotide sequence at least 90% identical to SEQ ID NO: 1 or SEQ ID NO: 3, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100% identical to SEQ ID NO: 1 or SEQ ID NO: 3, preferably 98%, 99% or 100% identical to SEQ ID NO: 1 or SEQ ID NO: 3, operably linked to a linker coding sequence at least 90% identical to SEQ ID NO: 11, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100% identical to SEQ ID NO: 11, preferably 98%, 99% or 100% identical to SEQ ID NO: 11, which is further operably linked a polynucleotide sequence at least 90% identical to SEQ ID NO: 5 or SEQ ID NO: 6, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100% identical to SEQ ID NO: 5 or SEQ ID NO: 6, preferably 98%, 99% or 100% identical to SEQ ID NO: 5 or SEQ ID NO: 6. In particular embodiments of the application, a non-naturally occurring nucleic acid molecule encoding an HBV antigen fusion protein comprises SEQ ID NO: 1 or SEQ ID NO: 3, operably linked to SEQ ID NO: 11, which is further operably linked to SEQ ID NO: 5 or SEQ ID NO: 6.
In another embodiment, a non-naturally occurring nucleic acid molecule encoding an HBV fusion further comprises a coding sequence for a signal sequence that is operably linked to the N-terminus of the HBV fusion sequence, such as the amino acid sequence of SEQ ID NO: 16. Preferably, the signal sequence has the amino acid sequence of SEQ ID NO: 9 or SEQ ID NO: 15. More preferably, the coding sequence for a signal sequence comprises the
polynucleotide sequence of SEQ ID NO: 8 or SEQ ID NO: 14. In one embodiment, the encoded fusion protein with the signal sequence comprises the amino acid sequence of SEQ ID NO: 17.
The application also relates to a vector comprising the first and/or second non-naturally occurring nucleic acid molecules. As used herein, a“vector” is a nucleic acid molecule used to carry genetic material into another cell, where it can be replicated and/or expressed. Any vector known to those skilled in the art in view of the present disclosure can be used. Examples of vectors include, but are not limited to, plasmids, viral vectors (bacteriophage, animal viruses, and plant viruses), cosmids, and artificial chromosomes (e.g., YACs). Preferably, a vector is a DNA plasmid. A vector can be a DNA vector or an RNA vector. One of ordinary skill in the art can construct a vector of the application through standard recombinant techniques in view of the present disclosure. A vector of the application can be an expression vector. As used herein, the term “expression vector” refers to any type of genetic construct comprising a nucleic acid coding for an RNA capable of being transcribed. Expression vectors include, but are not limited to, vectors for recombinant protein expression, such as a DNA plasmid or a viral vector, and vectors for delivery of nucleic acid into a subject for expression in a tissue of the subject, such as a DNA plasmid or a viral vector. It will be appreciated by those skilled in the art that the design of the expression vector can depend on such factors as the choice of the host cell to be transformed, the level of expression of protein desired, etc.
Vectors of the application can contain a variety of regulatory sequences. As used herein, the term“regulatory sequence” refers to any sequence that allows, contributes or modulates the functional regulation of the nucleic acid molecule, including replication, duplication, transcription, splicing, translation, stability and/or transport of the nucleic acid or one of its derivative (i.e. mRNA) into the host cell or organism. In the context of the disclosure, this term encompasses promoters, enhancers and other expression control elements (e.g., polyadenylation signals and elements that affect mRNA stability).
In some embodiments of the application, a vector is a non-viral vector. Examples of non- viral vectors include, but are not limited to, DNA plasmids, bacterial artificial chromosomes, yeast artificial chromosomes, bacteriophages, etc. Examples of non-viral vectors include, but are not limited to, RNA replicon, mRNA replicon, modified mRNA replicon or self-amplifying mRNA, closed linear deoxyribonucleic acid, e.g. a linear covalently closed DNA such as linear covalently closed double stranded DNA molecule. Preferably, a non-viral vector is a DNA plasmid. A“DNA plasmid”, which is used interchangeably with“DNA plasmid vector,” “plasmid DNA” or“plasmid DNA vector,” refers to a double-stranded and generally circular DNA sequence that is capable of autonomous replication in a suitable host cell. DNA plasmids used for expression of an encoded polynucleotide typically comprise an origin of replication, a multiple cloning site, and a selectable marker, which for example, can be an antibiotic resistance gene. Examples of DNA plasmids suitable that can be used include, but are not limited to, commercially available expression vectors for use in well-known expression systems (including both prokaryotic and eukaryotic systems), such as pSE420 (Invitrogen, San Diego, Calif.), which can be used for production and/or expression of protein in Escherichia coli; pYES2 (Invitrogen, Thermo Fisher Scientific), which can be used for production and/or expression in Saccharomyces cerevisiae strains of yeast; MAXBAC® complete baculovirus expression system (Thermo Fisher Scientific), which can be used for production and/or expression in insect cells; pcDNATM or pcDNA3TM (Life Technologies, Thermo Fisher Scientific), which can be used for high level constitutive protein expression in mammalian cells; and pVAX or pVAX-1 (Life Technologies, Thermo Fisher Scientific), which can be used for high-level transient expression of a protein of interest in most mammalian cells. The backbone of any commercially available DNA plasmid can be modified to optimize protein expression in the host cell, such as to reverse the orientation of certain elements (e.g., origin of replication and/or antibiotic resistance cassette), replace a promoter endogenous to the plasmid (e.g., the promoter in the antibiotic resistance cassette), and/or replace the polynucleotide sequence encoding transcribed proteins (e.g., the coding sequence of the antibiotic resistance gene), by using routine techniques and readily available starting materials. (See e.g., Sambrook et al., Molecular Cloning a
Laboratory Manual, Second Ed. Cold Spring Harbor Press (1989)).
Preferably, a DNA plasmid is an expression vector suitable for protein expression in mammalian host cells. Expression vectors suitable for protein expression in mammalian host cells include, but are not limited to, pcDNATM, pcDNA3TM, pVAX, pVAX-1, ADVAX, NTC8454, etc. Preferably, an expression vector is based on pVAX-1, which can be further modified to optimize protein expression in mammalian cells. pVAX-1 is commonly used plasmid in DNA vaccines, and contains a strong human intermediate early cytomegalovirus (CMV-IE) promoter followed by the bovine growth hormone (bGH)-derived polyadenylation sequence (pA). pVAX-1 further contains a pUC origin of replication and kanamycin resistance gene driven by a small prokaryotic promoter that allows for bacterial plasmid propagation.
A vector of the application can also be a viral vector. In general, viral vectors are genetically engineered viruses carrying modified viral DNA or RNA that has been rendered non- infectious, but still contains viral promoters and transgenes, thus allowing for translation of the transgene through a viral promoter. Because viral vectors are frequently lacking infectious sequences, they require helper viruses or packaging lines for large-scale transfection. Examples of viral vectors that can be used include, but are not limited to, adenoviral vectors, adeno- associated virus vectors, pox virus vectors, enteric virus vectors, Venezuelan Equine Encephalitis virus vectors, Semliki Forest Virus vectors, Tobacco Mosaic Virus vectors, lentiviral vectors, etc. Examples of viral vectors that can be used include, but are not limited to, arenavirus viral vectors, replication-deficient arenavirus viral vectors or replication-competent arenavirus viral vectors, bi-segmented or tri-segmented arenavirus, infectious arenavirus viral vectors, nucleic acids which comprise an arenavirus genomic segment wherein one open reading frame of the genomic segment is deleted or functionally inactivated (and replaced by a nucleic acid encoding an HBV antigen as described herein), arenavirus such as lymphocytic choriomeningitidis virus (LCMV), e.g., clone 13 strain or MP strain, and arenavirus such as Junin virus e.g., Candid #1 strain. The vector can also be a non-viral vector.
Preferably, a viral vector is an adenovirus vector, e.g., a recombinant adenovirus vector. A recombinant adenovirus vector can for instance be derived from a human adenovirus (HAdV, or AdHu), or a simian adenovirus such as chimpanzee or gorilla adenovirus (ChAd, AdCh, or SAdV) or rhesus adenovirus (rhAd). Preferably, an adenovirus vector is a recombinant human adenovirus vector, for instance a recombinant human adenovirus serotype 26, or any one of recombinant human adenovirus serotype 5, 4, 35, 7, 48, etc. In other embodiments, an adenovirus vector is a rhAd vector, e.g. rhAd51, rhAd52 or rhAd53. A recombinant viral vector useful for the application can be prepared using methods known in the art in view of the present disclosure. For example, in view of the degeneracy of the genetic code, several nucleic acid sequences can be designed that encode the same polypeptide. A polynucleotide encoding an HBV antigen of the application can optionally be codon-optimized to ensure proper expression in the host cell (e.g., bacterial or mammalian cells). Codon-optimization is a technology widely applied in the art, and methods for obtaining codon-optimized polynucleotides will be well known to those skilled in the art in view of the present disclosure.
A vector of the application, e.g., a DNA plasmid or a viral vector (particularly an adenoviral vector), can comprise any regulatory elements to establish conventional function(s) of the vector, including but not limited to replication and expression of the HBV antigen(s) encoded by the polynucleotide sequence of the vector. Regulatory elements include, but are not limited to, a promoter, an enhancer, a polyadenylation signal, translation stop codon, a ribosome binding element, a transcription terminator, selection markers, origin of replication, etc. A vector can comprise one or more expression cassettes. An“expression cassette” is part of a vector that directs the cellular machinery to make RNA and protein. An expression cassette typically comprises three components: a promoter sequence, an open reading frame, and a 3’-untranslated region (UTR) optionally comprising a polyadenylation signal. An open reading frame (ORF) is a reading frame that contains a coding sequence of a protein of interest (e.g., HBV antigen) from a start codon to a stop codon. Regulatory elements of the expression cassette can be operably linked to a polynucleotide sequence encoding an HBV antigen of interest. As used herein, the term“operably linked” is to be taken in its broadest reasonable context, and refers to a linkage of polynucleotide elements in a functional relationship. A polynucleotide is“operably linked” when it is placed into a functional relationship with another polynucleotide. For instance, a promoter is operably linked to a coding sequence if it affects the transcription of the coding sequence. Any components suitable for use in an expression cassette described herein can be used in any combination and in any order to prepare vectors of the application.
A vector can comprise a promoter sequence, preferably within an expression cassette, to control expression of an HBV antigen of interest. The term“promoter” is used in its conventional sense, and refers to a nucleotide sequence that initiates the transcription of an operably linked nucleotide sequence. A promoter is located on the same strand near the nucleotide sequence it transcribes. Promoters can be a constitutive, inducible, or repressible. Promoters can be naturally occurring or synthetic. A promoter can be derived from sources including viral, bacterial, fungal, plants, insects, and animals. A promoter can be a homologous promoter (i.e., derived from the same genetic source as the vector) or a heterologous promoter (i.e., derived from a different vector or genetic source). For example, if the vector to be employed is a DNA plasmid, the promoter can be endogenous to the plasmid (homologous) or derived from other sources (heterologous). Preferably, the promoter is located upstream of the polynucleotide encoding an HBV antigen within an expression cassette.
Examples of promoters that can be used include, but are not limited to, a promoter from simian virus 40 (SV40), a mouse mammary tumor virus (MMTV) promoter, a human immunodeficiency virus (HIV) promoter such as the bovine immunodeficiency virus (BIV) long terminal repeat (LTR) promoter, a Moloney virus promoter, an avian leukosis virus (ALV) promoter, a cytomegalovirus (CMV) promoter such as the CMV immediate early promoter (CMV-IE), Epstein Barr virus (EBV) promoter, or a Rous sarcoma virus (RSV) promoter. A promoter can also be a promoter from a human gene such as human actin, human myosin, human hemoglobin, human muscle creatine, or human metalothionein. A promoter can also be a tissue specific promoter, such as a muscle or skin specific promoter, natural or synthetic. Preferably, a promoter is a strong eukaryotic promoter, preferably a cytomegalovirus immediate early (CMV-IE) promoter. A nucleotide sequence of an exemplary CMV-IE promoter is shown in SEQ ID NO: 18 or SEQ ID NO: 19.
A vector can comprise additional polynucleotide sequences that stabilize the expressed transcript, enhance nuclear export of the RNA transcript, and/or improve transcriptional- translational coupling. Examples of such sequences include polyadenylation signals and enhancer sequences. A polyadenylation signal is typically located downstream of the coding sequence for a protein of interest (e.g., an HBV antigen) within an expression cassette of the vector. Enhancer sequences are regulatory DNA sequences that, when bound by transcription factors, enhance the transcription of an associated gene. An enhancer sequence is preferably located upstream of the polynucleotide sequence encoding an HBV antigen, but downstream of a promoter sequence within an expression cassette of the vector.
Any polyadenylation signal known to those skilled in the art in view of the present disclosure can be used. For example, the polyadenylation signal can be a SV40 polyadenylation signal, LTR polyadenylation signal, bovine growth hormone (bGH) polyadenylation signal, human growth hormone (hGH) polyadenylation signal, or human b-globin polyadenylation signal. Preferably, a polyadenylation signal is a bovine growth hormone (bGH) polyadenylation signal or a SV40 polyadenylation signal. A nucleotide sequence of an exemplary bGH polyadenylation signal is shown in SEQ ID NO: 20. A nucleotide sequence of an exemplary SV40 polyadenylation signal is shown in SEQ ID NO: 13.
Any enhancer sequence known to those skilled in the art in view of the present disclosure can be used. For example, an enhancer sequence can be human actin, human myosin, human hemoglobin, human muscle creatine, or a viral enhancer, such as one from CMV, HA, RSV, or EBV. Examples of particular enhancers include, but are not limited to, Woodchuck HBV Post- transcriptional regulatory element (WPRE), intron/exon sequence derived from human apolipoprotein A1 precursor (ApoAI), untranslated R-U5 domain of the human T-cell leukemia virus type 1 (HTLV-1) long terminal repeat (LTR), a splicing enhancer, a synthetic rabbit b- globin intron, or any combination thereof. Preferably, an enhancer sequence is a composite sequence of three consecutive elements of the untranslated R-U5 domain of HTLV-1 LTR, rabbit b-globin intron, and a splicing enhancer, which is referred to herein as“a triple enhancer sequence.” A nucleotide sequence of an exemplary triple enhancer sequence is shown in SEQ ID NO: 10. Another exemplary enhancer sequence is an ApoAI gene fragment shown in SEQ ID NO: 12.
A vector can comprise a polynucleotide sequence encoding a signal peptide sequence. Preferably, the polynucleotide sequence encoding the signal peptide sequence is located upstream of the polynucleotide sequence encoding an HBV antigen. Signal peptides typically direct localization of a protein, facilitate secretion of the protein from the cell in which it is produced, and/or improve antigen expression and cross-presentation to antigen-presenting cells. A signal peptide can be present at the N-terminus of an HBV antigen when expressed from the vector, but is cleaved off by signal peptidase, e.g., upon secretion from the cell. An expressed protein in which a signal peptide has been cleaved is often referred to as the“mature protein.” Any signal peptide known in the art in view of the present disclosure can be used. For example, a signal peptide can be a cystatin S signal peptide; an immunoglobulin (Ig) secretion signal, such as the Ig heavy chain gamma signal peptide SPIgG or the Ig heavy chain epsilon signal peptide SPIgE.
Preferably, a signal peptide sequence is a cystatin S signal peptide. Exemplary nucleic acid and amino acid sequences of a cystatin S signal peptide are shown in SEQ ID NOs: 8 and 9, respectively. Exemplary nucleic acid and amino acid sequences of an immunoglobulin secretion signal are shown in SEQ ID NOs: 14 and 15, respectively.
A vector, such as a DNA plasmid, can also include a bacterial origin of replication and an antibiotic resistance expression cassette for selection and maintenance of the plasmid in bacterial cells, e.g., E. coli. Bacterial origins of replication and antibiotic resistance cassettes can be located in a vector in the same orientation as the expression cassette encoding an HBV antigen, or in the opposite (reverse) orientation. An origin of replication (ORI) is a sequence at which replication is initiated, enabling a plasmid to reproduce and survive within cells. Examples of ORIs suitable for use in the application include, but are not limited to ColE1, pMB1, pUC, pSC101, R6K, and 15A, preferably pUC. An exemplary nucleotide sequence of a pUC ORI is shown in SEQ ID NO: 21.
Expression cassettes for selection and maintenance in bacterial cells typically include a promoter sequence operably linked to an antibiotic resistance gene. Preferably, the promoter sequence operably linked to an antibiotic resistance gene differs from the promoter sequence operably linked to a polynucleotide sequence encoding a protein of interest, e.g., HBV antigen. The antibiotic resistance gene can be codon optimized, and the sequence composition of the antibiotic resistance gene is normally adjusted to bacterial, e.g., E. coli, codon usage. Any antibiotic resistance gene known to those skilled in the art in view of the present disclosure can be used, including, but not limited to, kanamycin resistance gene (Kanr), ampicillin resistance gene (Ampr), and tetracycline resistance gene (Tetr), as well as genes conferring resistance to chloramphenicol, bleomycin, spectinomycin, carbenicillin, etc.
Preferably, an antibiotic resistance gene in the antibiotic expression cassette of a vector is a kanamycin resistance gene (Kanr). The sequence of Kanr gene is shown in SEQ ID NO: 22. Preferably, the Kanr gene is codon optimized. An exemplary nucleic acid sequence of a codon optimized Kanr gene is shown in SEQ ID NO: 23. The Kanr can be operably linked to its native promoter, or the Kanr gene can be linked to a heterologous promoter. In a particular embodiment, the Kanr gene is operably linked to the ampicillin resistance gene (Ampr) promoter, known as the bla promoter. An exemplary nucleotide sequence of a bla promoter is shown in SEQ ID NO: 24.
In a particular embodiment of the application, a vector is a DNA plasmid comprising an expression cassette including a polynucleotide encoding at least one of an HBV antigen selected from the group consisting of an HBV pol antigen comprising an amino acid sequence at least 90%, such as 90%, 91%, 92%, 93%, 94%, 95%, 96, 97%, preferably at least 98%, such as at least 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100%, identical to SEQ ID NO: 7, and a truncated HBV core antigen consisting of the amino acid sequence at least 95%, such as 95%, 96, 97%, preferably at least 98%, such as at least 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100%, identical of SEQ ID NO: 2 or SEQ ID NO: 4; an upstream sequence operably linked to the polynucleotide encoding the HBV antigen comprising, from 5’ end to 3’ end, a promoter sequence, preferably a CMV promoter sequence of SEQ ID NO: 18, an enhancer sequence, preferably a triple enhancer sequence of SEQ ID NO: 10, and a polynucleotide sequence encoding a signal peptide sequence, preferably a cystatin S signal peptide having the amino acid sequence of SEQ ID NO: 9; and a downstream sequence operably linked to the polynucleotide encoding the HBV antigen comprising a polyadenylation signal, preferably a bGH
polyadenylation signal of SEQ ID NO: 20. Such vector further comprises an antibiotic resistance expression cassette including a polynucleotide encoding an antibiotic resistance gene, preferably a Kanr gene, more preferably a codon optimized Kanr gene of at least 90% identical to SEQ ID NO: 23, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100% identical to SEQ ID NO: 23, preferably 100% identical to SEQ ID NO: 23, operably linked to an Ampr (bla) promoter of SEQ ID NO: 24, upstream of and operably linked to the polynucleotide encoding the antibiotic resistance gene; and an origin of replication, preferably a pUC ori of SEQ ID NO: 21. Preferably, the antibiotic resistance cassette and the origin of replication are present in the plasmid in the reverse orientation relative to the HBV antigen expression cassette.
In another particular embodiment of the application, a vector is a viral vector, preferably an adenoviral vector, more preferably an Ad26 or Ad35 vector, comprising an expression cassette including a polynucleotide encoding at least one of an HBV antigen selected from the group consisting of an HBV pol antigen comprising an amino acid sequence at least 90%, such as 90%, 91%, 92%, 93%, 94%, 95%, 96, 97%, preferably at least 98%, such as at least 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100%, identical to SEQ ID NO: 7, and a truncated HBV core antigen consisting of the amino acid sequence at least 95%, such as 95%, 96, 97%, preferably at least 98%, such as at least 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100%, identical of SEQ ID NO: 2 or SEQ ID NO: 4; an upstream sequence operably linked to the polynucleotide encoding the HBV antigen comprising, from 5’ end to 3’ end, a promoter sequence, preferably a CMV promoter sequence of SEQ ID NO: 19, an enhancer sequence, preferably an ApoAI gene fragment sequence of SEQ ID NO: 12, and a polynucleotide sequence encoding a signal peptide sequence, preferably an immunoglobulin secretion signal having the amino acid sequence of SEQ ID NO: 15; and a downstream sequence operably linked to the polynucleotide encoding the HBV antigen comprising a polyadenylation signal, preferably a SV40 polyadenylation signal of SEQ ID NO: 13.
In an embodiment of the application, a vector, such as a plasmid DNA vector or a viral vector (preferably an adenoviral vector, more preferably an Ad26 or Ad35 vector), encodes an HBV Pol antigen having the amino acid sequence of SEQ ID NO: 7. Preferably, the vector comprises a coding sequence for the HBV Pol antigen that is at least 90% identical to the polynucleotide sequence of SEQ ID NO: 5 or 6, such as 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100% identical to SEQ ID NO: 5 or 6, preferably 100% identical to SEQ ID NO: 5 or 6.
In an embodiment of the application, a vector, such as a plasmid DNA vector or a viral vector (preferably an adenoviral vector, more preferably an Ad26 or Ad35 vector), encodes a truncated HBV core antigen consisting of the amino acid sequence of SEQ ID NO: 2 or SEQ ID NO: 4. Preferably, the vector comprises a coding sequence for the truncated HBV core antigen that is at least 90% identical to the polynucleotide sequence of SEQ ID NO: 1 or SEQ ID NO: 3, such as 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100% identical to SEQ ID NO: 1 or SEQ ID NO: 3, preferably 100% identical to SEQ ID NO: 1 or SEQ ID NO: 3.
In yet another embodiment of the application, a vector, such as a plasmid DNA vector or a viral vector (preferably an adenoviral vector, more preferably an Ad26 or Ad35 vector), encodes a fusion protein comprising an HBV Pol antigen having the amino acid sequence of SEQ ID NO: 7 and a truncated HBV core antigen consisting of the amino acid sequence of SEQ ID NO: 1 or SEQ ID NO: 3. Preferably, the vector comprises a coding sequence for the fusion, which contains a coding sequence for the truncated HBV core antigen at least 90% identical to SEQ ID NO: 1 or SEQ ID NO: 3, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100% identical to SEQ ID NO: 1 or SEQ ID NO: 3, preferably 98%, 99% or 100% identical to SEQ ID NO: 1 or SEQ ID NO: 3, more preferably SEQ ID NO: 1 or SEQ ID NO: 3, operably linked to a coding sequence for the HBV Pol antigen at least 90% identical to SEQ ID NO: 5 or SEQ ID NO: 6, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100% identical to SEQ ID NO: 5 or SEQ ID NO: 6, preferably 98%, 99% or 100% identical to SEQ ID NO: 5 or SEQ ID NO: 6, more preferably SEQ ID NO: 5 or SEQ ID NO: 6. Preferably, the coding sequence for the truncated HBV core antigen is operably linked to the coding sequence for the HBV Pol antigen via a coding sequence for a linker at least 90% identical to SEQ ID NO: 11, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100% identical to SEQ ID NO: 11, preferably 98%, 99% or 100% identical to SEQ ID NO: 11. In particular embodiments of the application, a vector comprises a coding sequence for the fusion having SEQ ID NO: 1 or SEQ ID NO: 3 operably linked to SEQ ID NO: 11, which is further operably linked to SEQ ID NO: 5 or SEQ ID NO: 6.
The polynucleotides and expression vectors encoding the HBV antigens of the application can be made by any method known in the art in view of the present disclosure. For example, a polynucleotide encoding an HBV antigen can be introduced or“cloned” into an expression vector using standard molecular biology techniques, e.g., polymerase chain reaction (PCR), etc., which are well known to those skilled in the art.
Cells, Polypeptides and Antibodies
The application also provides cells, preferably isolated cells, comprising any of the polynucleotides and vectors described herein. The cells can, for instance, be used for recombinant protein production, or for the production of viral particles.
Embodiments of the application thus also relate to a method of making an HBV antigen of the application. The method comprises transfecting a host cell with an expression vector comprising a polynucleotide encoding an HBV antigen of the application operably linked to a promoter, growing the transfected cell under conditions suitable for expression of the HBV antigen, and optionally purifying or isolating the HBV antigen expressed in the cell. The HBV antigen can be isolated or collected from the cell by any method known in the art including affinity chromatography, size exclusion chromatography, etc. Techniques used for recombinant protein expression will be well known to one of ordinary skill in the art in view of the present disclosure. The expressed HBV antigens can also be studied without purifying or isolating the expressed protein, e.g., by analyzing the supernatant of cells transfected with an expression vector encoding the HBV antigen and grown under conditions suitable for expression of the HBV antigen.
Thus, also provided are non-naturally occurring or recombinant polypeptides comprising an amino acid sequence that is at least 90% identical to the amino acid sequence of SEQ ID NO: 2, SEQ ID NO: 4, or SEQ ID NO: 7. As described above and below, isolated nucleic acid molecules encoding these sequences, vectors comprising these sequences operably linked to a promoter, and compositions comprising the polypeptide, polynucleotide, or vector are also contemplated by the application. In an embodiment of the application, a recombinant polypeptide comprises an amino acid sequence that is at least 90% identical to the amino acid sequence of SEQ ID NO: 2, such as 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100% identical to SEQ ID NO: 2. Preferably, a non-naturally occurring or recombinant polypeptide consists of SEQ ID NO: 2.
In another embodiment of the application, a non-naturally occurring or recombinant polypeptide comprises an amino acid sequence that is at least 90% identical to the amino acid sequence of SEQ ID NO: 4, such as 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100% identical to SEQ ID NO: 4. Preferably, a non-naturally occurring or
recombinant polypeptide comprises SEQ ID NO: 4.
In another embodiment of the application, a non-naturally occurring or recombinant polypeptide comprises an amino acid sequence that is at least 90% identical to the amino acid sequence of SEQ ID NO: 7, such as 90%, 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 96.5%, 97%, 97.5%, 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or 100% identical to SEQ ID NO: 7. Preferably, a non-naturally occurring or
recombinant polypeptide consists of SEQ ID NO: 7.
Also provided are antibodies or antigen binding fragments thereof that specifically bind to a non-naturally occurring polypeptide of the application. In an embodiment of the application, an antibody specific to a non-naturally HBV antigen of the application does not bind specifically to another HBV antigen. For example, an antibody of the application that binds specifically to an HBV Pol antigen having the amino acid sequence of SEQ ID NO: 7 will not bind specifically to an HBV Pol antigen not having the amino acid sequence of SEQ ID NO: 7.
As used herein, the term“antibody” includes polyclonal, monoclonal, chimeric, humanized, Fv, Fab and F(ab¢)2; bifunctional hybrid (e.g., Lanzavecchia et al., Eur. J. Immunol. 17:105, 1987), single-chain (Huston et al., Proc. Natl. Acad. Sci. USA 85:5879, 1988; Bird et al., Science 242:423, 1988); and antibodies with altered constant regions (e.g., U.S. Pat. No.
5,624,821).
As used herein, an antibody that“specifically binds to” an antigen refers to an antibody that binds to the antigen with a KD of 1×10-7 M or less. Preferably, an antibody that
“specifically binds to” an antigen binds to the antigen with a KD of 1×10-8 M or less, more preferably 5×10-9 M or less, 1×10-9 M or less, 5×10-10 M or less, or 1×10-10 M or less. The term “KD” refers to the dissociation constant, which is obtained from the ratio of Kd to Ka (i.e., Kd/Ka) and is expressed as a molar concentration (M). KD values for antibodies can be determined using methods in the art in view of the present disclosure. For example, the KD of an antibody can be determined by using surface plasmon resonance, such as by using a biosensor system, e.g., a Biacore® system, or by using bio-layer interferometry technology, such as an Octet RED96 system.
The smaller the value of the KD of an antibody, the higher affinity that the antibody binds to a target antigen.
Capsid Assembly Modulators
A number of capsid assembly modulators can be utilized in the invention. As used herein, the term“capsid assembly modulators” refers to a compound that disrupts or accelerates or inhibits or hinders or delays or reduces or modifies normal capsid assembly (e.g., during maturation) or normal capsid disassembly (e.g., during infectivity) or perturbs capsid stability, thereby inducing aberrant capsid morphology and function. Examples of these compounds, described in more detail below, are selected from (a) specific compounds of formulae (I), (XI), (XVIII), and (XXI), (b) specific compounds of formula (A-I) to formula (A-VII), and (c) specific compounds of formula (B-I). These compounds are described in detail, including the preparation and biological activities thereof, in International Patent Application Publications WO
2014106019, WO 2017156255, and WO 2018039531, and U.S. Patent Application Publications US 20160024004 and US 20180161307, the contents of which are hereby incorporated by reference in their entireties.
(a) Compounds of formulae (I), (XI), (XVIII), and (XXI)
Examples of compounds of formulae (I), (XI), (XVIII), and (XXI) (or enantiomeric and diastereomeric forms, hydrates, solvates, pharmaceutically acceptable salts, prodrugs and complexes thereof), synthesis, biologic activities, uses or other related information thereof are described in International Patent Application Publication WO 2014106019 and U.S. Patent Application Publication US 20160024004, the contents of which are hereby incorporated by reference in their entireties. In certain embodiments, the compounds for use in the invention are those having the formula (I) or enantiomeric and diastereomeric forms, hydrates, solvates, pharmaceutically acceptable salts, prodrugs and complexes thereof:
Figure imgf000039_0001
wherein:
A is selected from a group consisting of SO2 and CO;
R1 is selected from a group consisting of optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted benzyl; R1 may also alternatively or additionally optionally include optionally substituted C1-6 haloalkyl, optionally substituted 3-7 membered cycloheteroalkyl, optionally substituted C2-8 alkenyl, optionally substituted C2-8 alkynyl, optionally substituted C1-6 alkoxy, optionally substituted amine, optionally substituted amidine, optionally substituted carboxyamine, optionally substituted carboxy-C1-6-alkoxide, -SO2-C1- 6alkyl, optionally substituted heterocyclic, or optionally substituted heteroaryl;
R2 is selected from a group consisting of hydrogen and optionally substituted C1-6 linear alkyl; R2 may also alternatively or additionally optionally include optionally substituted cycloalkyl or optionally substituted heterocyclic; or
R1 and R2 are taken together with the atoms to which they are bound to form an optionally substituted heterocycle (including bicyclic or adamantyl structures) with 3 to 10 atoms; and
R3 is selected from a group consisting of optionally substituted aryl, optionally substituted benzyl, optionally substituted alkylaryl, optionally substituted heteroaryl, and optionally substituted alkylheteroaryl ; R3 may also optionally comprise an optionally substituted C1-6 linear alkyl;
R4 is selected from a group consisting of hydrogen and optionally substituted C1-6 linear alkyl; R5 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, optionally substituted C1-6 haloalkyl, and OR9; R5 may also alternatively or additionally optionally include cyano or N(R9)2;
R6 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, optionally substituted C1-6 haloalkyl, and OR9; in some embodiments, R6 may also alternatively or additionally optionally include cyano or N(R9)2; R6 may also alternatively or additionally optionally include cyano or N(R9)2; or
R4 and R6 are taken together with the atoms to which they are bound to form an optionally substituted carbocyclic or heterocyclic ring with 5 to 6 atoms, optionally containing a carbonyl, optionally containing two carbonyls; and
R7 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, optionally substituted C1-6 haloalkyl, and OR9; R7 may also alternatively or additionally optionally include cyano or N(R9)2;
R8 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, optionally substituted C1-6 haloalkyL-and OR9; R8 may also alternatively or additionally optionally include cyano or N( R9)2; or
R2 and R8 are taken together with the atoms to which they are bound to form an optionally substituted carbocyclic or heterocyclic ring with 5 to 6 atoms; and
R9 is independently at each occurrence selected from a group consisting of hydrogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, and optionally substituted C3-7 cycloalkyl; R9 may also alternatively or additionally optionally include independently at each occurrence optionally substituted aryl, optionally substituted benzyl, optionally substituted heterocyclyl, or optionally substituted heteroaryl;
provided that when A is SO2; R4 and R6 taken together with the atoms to which they are bound do not to form an optionally substituted ring; and R2 and R8 taken together with the atoms to which they are bound do not to form an optionally substituted ring, then none of the following (a) through (d) apply:
(a) R3 is an optionally substituted phenyl and R1 or R2 , either individually or when taken together, contain a hydroxyl group, or (b) R3 is an optionally substituted alkyl or phenyl, and N(R1)(R2) is an optionally substituted piperazine or
Figure imgf000041_0001
(c) R3 is optionally substituted alkyl, aryl, or alkaryl and N(R1)(R2) is
Figure imgf000042_0001
(d) either R or R is an unsubstituted or monosubstituted aryl, or an unsubstituted or monosubstituted aralkyl, or unsubstituted or monosubstituted heteroaryl and R1 and R2 are taken together with the atoms to which they are bound to form an optionally substituted heterocyclic ring structure with 6 to 12 atoms; or
provided that the compound is not 3- {[(dicyclopropylmethyl)amino]sulfonyl}-N-(4- isopropoxyphenyl)benzamide; or 3-({[2-(lH-benzimidazol-2-yl)propyl]amino}sulfonyl)-N-(4- isopropoxyphenyl)benzamide; or 3-[(cyclohexylamino)sulfonyl]-N-(4- isopropylphenyl)benzamide; or 3-(anilinosulfonyl)-N-(4-isopropylphenyl)benzamide; or 5- {[(3- {[(4-methoxyphenyl)amino]carbonyl}phenyl)sulfonyl]amino}pentanoic acid; or 3-[(tert- butylamino)sulfonyl]-N-(4-methoxyphenyl)benzamide; or (3S)- l-[(3- {[(5-isopropoxypyridin-2- yl)amino]carbonyl}phenyl)sulfonyl]piperidine-3-carboxamide; or (3R)-l-[(3- {[(5- isopropoxypyridin- 2-yl)amino]carbonyl}phenyl)sulfonyl]piperidine-3-carboxamide; or 3- (piperidin- l-ylsulfonyl)-N- [(1 S)- 1 ,2,3,4-tetrahydronaphthalen- 1 -yljbenzamide; or N-(5- bromo-3-methoxypyridin-2-yl)-3- (piperidin-1-ylsulfonyl)benzamide; or N-(3-methoxy-5- phenylpyridin-2-yl)-3-(pyrrolidin- 1 - ylsulfonyl)benzamide; or N-(3-methoxy-5- phenoxypyridin-2-yl)-3-(pyrrolidin- 1 - ylsulfonyl)benzamide; or N-[3-methoxy-5- (phenylthio)pyridin-2-yl]-3-(pyrrolidin- 1 - ylsulfonyl)benzamide; or N-(5-ethyl-3- methoxypyridin-2-yl)-3-(piperidin- 1 -ylsulfonyl)benzamide; or N-(3-methoxy-5-vinylpyridin-2- yl)-3-(piperidin- 1 -ylsulfonyl)benzamide; or
Figure imgf000043_0001
In certain embodiments, the compounds for use in the invention are those having the formula (XI) or enantiomeric and diastereomeric forms, hydrates, solvates, pharmaceutically acceptable salts, prodrugs and complexes thereof:
Figure imgf000043_0002
wherein:
A is selected from a group consisting of SO2 and CO; R1 is selected from a group consisting of optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted benzyl;
R3 is selected from a group consisting of optionally substituted aryl, optionally substituted benzyl, optionally substituted alkylaryl, optionally substituted heteroaryl, and optionally substituted alkylheteroaryl; in some embodiments, R3 may also comprise an optionally substituted C1-6 linear alkyl;
R4 is selected from a group consisting of hydrogen and optionally substituted C1-6 linear alkyl;
R5 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, optionally substituted C1-6 haloalkyl, and OR9; R5 may also alternatively or additionally optionally include cyano or N(R9)2;
R6 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, optionally substituted C1-6 haloalkyl, and OR9; R6 may also alternatively or additionally optionally include cyano or N(
Figure imgf000044_0001
or
R4 and R6 are taken together with the atoms to which they are bound to form an optionally substituted carbocyclic or heterocyclic ring with 5 to 6 atoms, optionally containing a carbonyl, optionally containing two carbonyls; and
R7 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, optionally substituted C1-6 haloalkyl, and OR9; R7 may also alternatively or additionally optionally include cyano or N(R9)2; R8 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, optionally substituted C1-6 haloalkyl, and OR9;, R8 may also alternatively or additionally optionally include cyano or N(R9)2;
R9 is independently at each occurrence selected from a group consisting of hydrogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, and optionally substituted cycloalkyl, and optionally substituted C1-6 haloalkyl; R9 may also alternatively or additionally optionally include independently at each occurrence optionally substituted aryl, optionally substituted benzyl, optionally substituted heterocyclyl, or optionally substituted heteroaryl;and R11 is selected from a group consisting of hydrogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, and optionally substituted cycloalkyl. In certain embodiments, the compounds for use in the invention are those having the formula (XVIII) or enantiomeric and diastereomeric forms, hydrates, solvates, pharmaceutically acceptable salts, prodrugs and complexes thereof:
Figure imgf000045_0001
wherein:
A is selected from a group consisting of SO2 and CO;
R1 is selected from a group consisting of optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted benzyl;
R5 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, optionally substituted C1-6 haloalkyl, and OR9; R5 may also alternatively or additionally optionally include cyano or N(R9)2;
R6 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, optionally substituted C1-6 haloalkyl, and OR9; R6 may also alternatively or additionally optionally include cyano or N(R9)2;
R7 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, optionally substituted C1-6 haloalkyl, and OR9; R7 may also alternatively or additionally optionally include cyano or N(R9)2;
R8 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, optionally substituted C1-6 haloalkyl, and OR9; R8 may also alternatively or additionally optionally include cyano or N(R9)2; R9 is independently at each occurrence selected from a group consisting of hydrogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, and optionally substituted cycloalkyl; R9 may also alternatively or additionally optionally include independently at each occurrence optionally substituted aryl, optionally substituted benzyl, optionally substituted heterocyclyl, or optionally substituted heteroaryl;
R11 is selected from a group consisting of hydrogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, and optionally substituted cycloalkyl;
R12 is selected from a group consisting of hydrogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, and optionally substituted cycloalkyl; and R13 is selected from a group consisting of optionally substituted aryl, optionally substituted benzyl, optionally substituted alkylaryl, optionally substituted heteroaryl, and optionally substituted alkylheteroaryl. In certain embodiments, the compounds for use in the invention are those having the formula (XXI) or enantiomeric and diastereomeric forms, hydrates, solvates, pharmaceutically
Figure imgf000046_0001
acceptable salts, prodrugs and complexes thereof:
wherein:
R1 is selected from a group consisting of optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted benzyl; R1 may also alternatively or additionally optionally include optionally substituted C1-6 haloalkyl, optionally substituted 3-7 membered cycloheteroalkyl, optionally substituted C2-8 alkenyl, optionally substituted C2-8 alkynyl, optionally substituted C1-6 alkoxy, optionally substituted amine, optionally substituted amidine, optionally substituted carboxyamine, optionally substituted carboxy-C1-6-alkoxide, -S02-C1- 6alkyl, optionally substituted heterocyclic, or optionally substituted heteroaryl;
R2 is selected from a group consisting of hydrogen and optionally substituted C1-6 linear alkyl; R2 may also alternatively or additionally optionally include optionally substituted cycloalkyl or optionally substituted heterocyclic; or
R1 and R2 are taken together with the atoms to which they are bound to form an optionally substituted heterocycle (including cyclic and heterocyclic structures) with 3 to 10 atoms;
R5 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, optionally substituted C1-6 haloalkyl, and OR9; R5 may also alternatively or additionally optionally include cyano or N(R9)2;
R6 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, optionally substituted C1-6 haloalkyl, and OR9; R6 may also alternatively or additionally optionally include cyano or N(R9)2;
R7 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, optionally substituted C1-6 haloalkyl, OR9; R7 may also alternatively or additionally optionally include cyano or N(R9)2;
R8 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, optionally substituted C1-6 haloalkyl, and OR9; R8 may also alternatively or additionally optionally include cyano or N(R9)2;
R9 is independently at each occurrence selected from a group consisting of hydrogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, and optionally substituted cycloalkyl, and optionally substituted C1-6 haloalkyl; R9 may also alternatively or additionally optionally include, independently at each occurrence, optionally substituted aryl, optionally substituted benzyl, optionally substituted heterocyclyl, or optionally substituted heteroaryl;
R12 is selected from a group consisting of hydrogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, and optionally substituted cycloalkyl; and R13 is selected from a group consisting of optionally substituted aryl, optionally substituted benzyl, optionally substituted alkylaryl, optionally substituted heteroaryl, and optionally substituted alkylheteroaryl. Exemplary compounds having formulae (I), (XI), (XVIII), or (XXI) have the following structures, where the substituents are as described previously:
Figure imgf000048_0001
Figure imgf000049_0001
Preferably, exemplary embodiments include compounds having the formula (XXIX) or a pharmaceutically acceptable salt form thereof:
Figure imgf000050_0001
wherein non- limiting examples of R1 and R2 are defined herein below in Table 1.
Table 1: Exemplary embodiments of compounds of the formula (XXIX):
Figure imgf000050_0002
Figure imgf000051_0001
Figure imgf000052_0003
Exemplary embodiments include compounds having the formula (XXX) or a pharmaceutically acceptable salt form thereof:
Figure imgf000052_0001
wherein non- limiting examples of R1 and R2 are defined herein below in Table 2. Table 2: Exemplary embodiments of compounds of the formula (XXX):
Figure imgf000052_0002
Figure imgf000053_0001
Figure imgf000054_0003
Exemplary embodiments include compounds having the formula (XXXI) or a pharmaceutically acceptable salt form thereof:
Figure imgf000054_0001
wherein non- limiting examples of R1 and R2 are defined herein below in Table 3. Table 3. Exemplary embodiments of compounds of the formula (XXXI):
Figure imgf000054_0002
Figure imgf000055_0001
Figure imgf000056_0002
Exemplary embodiments include compounds having the formula (XXXII) or a pharmaceutically acceptable salt form thereof:
Figure imgf000056_0001
wherein non- limiting examples of R1 and R2 are defined herein below in Table 4. Table 4. Exemplary embodiments of compounds of the formula (XXXII):
Figure imgf000056_0003
Figure imgf000057_0001
Figure imgf000058_0003
Exemplary embodiments include compounds having the formula (XXXIII) or a pharmaceutically acceptable salt form thereof:
Figure imgf000058_0001
wherein non- limiting examples of R1 and R2 are defined herein below in Table 5. Table 5. Exemplary embodiments of compounds of the formula (XXXIII):
Figure imgf000058_0002
Figure imgf000059_0001
Figure imgf000060_0002
Exemplary embodiments include compounds having the formula (XXXIV) or a pharmaceutically acceptable salt form thereof:
Figure imgf000060_0001
wherein non- limiting examples of R1 and R2 are defined herein below in Table 6. Table 6. Exemplary embodiments of compounds of the formula (XXXIV):
Figure imgf000060_0003
Figure imgf000061_0001
Figure imgf000062_0002
Exemplary embodiments include compounds having the formula (XXXV) or a pharmaceutically acceptable salt form thereof:
Figure imgf000062_0001
wherein non- limiting examples of R1 and R2 are defined herein below in Table 7. Table 7. Exemplary embodiments of compounds of the formula (XXXV):
Figure imgf000062_0003
Figure imgf000063_0001
Figure imgf000064_0002
Exemplary embodiments include compounds having the formula (XXXVI) or a pharmaceutically acceptable salt form thereof:
Figure imgf000064_0001
wherein non- limiting examples of R1 and R2 are defined herein below in Table 8. Table 8. Exemplary embodiments of compounds of the formula (XXXVI):
Figure imgf000064_0003
Figure imgf000065_0001
Figure imgf000066_0002
Exemplary embodiments include compounds having the formula (XXXVII) or a pharmaceutically acceptable salt form thereof:
Figure imgf000066_0001
wherein non- limiting examples of R2 , R3, and R4 are defined herein below in Table 9. Table 9. Exemplary embodiments of compounds of the formula (XXXVII):
Figure imgf000066_0003
Figure imgf000067_0001
Figure imgf000068_0002
Exemplary embodiments include compounds having the formula (XXXVIII) or a pharmaceutically acceptable salt form thereof:
Figure imgf000068_0001
wherein non- limiting examples of R2 , R3, and R4 are defined herein below in Table 10. Table 10. Exemplary embodiments of compounds of the formula (XXXVIII):
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0005
Figure imgf000071_0004
Figure imgf000071_0002
Exemplary embodiments include compounds having the formula (XXXIX) or a pharmaceutically acceptable salt form thereof:
Figure imgf000071_0001
wherein non- limiting examples of R2 , R3, and R4 are defined herein below in Table 11. Table 11. Exemplary embodiments of compounds of the formula (XXXIX):
Figure imgf000071_0003
Figure imgf000072_0001
Figure imgf000073_0002
Exemplary embodiments include compounds having the formula (XXXX) or a pharmaceutically acceptable salt form thereof:
Figure imgf000073_0001
wherein non- limiting examples of R5, R6, R7, and R8are defined herein below in Table 12.
Table 12: Exemplary embodiments of compounds of the formula (XXXX):
Figure imgf000074_0003
Exemplary embodiments include compounds having the formula (XXXXI-A or XXXXI- B) or a pharmaceutically acceptable salt form thereof:
Figure imgf000074_0001
wherein non- limiting examples of R5, R6, R7, and R8 are defined herein below in Table 13.
Table 13: Exemplary embodiments of compounds of the formula (XXXXI-A or XXXXI- B):
Figure imgf000074_0004
Figure imgf000074_0002
Exemplary embodiments include compounds having the formula (XXXXII) or a pharmaceutically acceptable salt form thereof:
Figure imgf000075_0001
wherein non- limiting examples of R1 , R2 , R3 and z are defined herein below in Table 14. Table 14: Exemplary embodiments of compounds of the formula (XXXXII):
Figure imgf000075_0003
Exemplary embodiments include compounds having the formula (XXXXIII) or a pharmaceutically acceptable salt form thereof:
Figure imgf000075_0002
wherein non- limiting examples of R1 , R2 , R3 and f are defined herein below in Table 15. Table 15: Exemplary embodiments of compounds of the formula (XXXXIII):
Figure imgf000076_0003
Exemplary embodiments include compounds having the formula (XXXXIV) or a pharmaceutically acceptable salt form thereof:
Figure imgf000076_0001
wherein non- limiting examples of R1 , R2 , R3 and z are defined herein below in Table 16. Table 16: Exemplary embodiments of compounds of the formula (XXXXIV):
Figure imgf000076_0002
Exemplary embodiments include compounds having the formula (XXXXV) or a pharmaceutically acceptable salt form thereof:
Figure imgf000077_0001
wherein non- limiting examples of R1 , R2 , R3 and f are defined herein below in Table 17. Table 17: Exemplary embodiments of compounds of the formula (XXXXV):
Figure imgf000077_0003
Exemplary embodiments include compounds having the formula (XXXXVI) or a pharmaceutically acceptable salt form thereof:
Figure imgf000077_0002
wherein non- limiting examples of R1 , R3, R4 and y are defined herein below in Table 18. Table 18: Exemplary embodiments of compounds of the formula (XXXXVI):
Figure imgf000078_0003
Exemplary embodiments include compounds having the formula (XXXXVII) or a pharmaceutically acceptable salt form thereof:
Figure imgf000078_0001
wherein non- limiting examples of R1 , R3, and R4 are defined herein below in Table 19. Table 19: Exemplary embodiments of compounds of the formula (XXXXVII):
Figure imgf000078_0004
Exemplary embodiments include compounds having the formula (XXXXVIII) or a pharmaceutically acceptable salt form thereof:
Figure imgf000078_0002
wherein non- limiting examples of R1 , and R13 are defined herein below in Table 20. Table 20: Exemplary embodiments of compounds of the formula (XXXXVIII):
Figure imgf000079_0001
Specific compounds having the formulae (I), (XI), (XVIII), and (XXI) which are appropriate for use in embodiments according to the invention include, but are not limited to:
Figure imgf000080_0001
The effective amount of the compound having formula (I), (XI), (XVIII), or (XXI), or a pharmaceutically acceptable salt, an enantiomer, or a diastereomer thereof may be determined by routine experimentation, but can be in range of about 0.001 to 1000 mg, preferably from about 0.01 to 100 mg, more preferably from about 0.1 to 10 mg.
Suitable processes for synthesizing these compounds can be synthesized by any method or synthetic scheme known in the art or to be discovered. For example, the compounds can be prepared according to Scheme 1, shown below. Examples of specific synthetic compounds are described in WO 2014106019 and US 20160024004.
Figure imgf000081_0001
(b) Compounds of formula (A-I) to formula (A-VII)
Examples of compounds of formula (A-I) to formula (A-VII) (or enantiomeric and diastereomeric forms, hydrates, solvates, pharmaceutically acceptable salts, prodrugs and complexes thereof), synthesis, biologic activities, uses or other related information thereof are described in International Patent Application Publication WO 2017156255, the contents of which are hereby incorporated by reference in their entireties.
In one embodiment, the compounds for use in the invention are those having formula (A- I):
Figure imgf000081_0002
or an enantiomer, diastereomer or pharmaceutically accepted salt, hydrate, or solvate thereof, including a hydrate, a solvate, a pharmaceutically acceptable salt, a prodrug, or a complex thereof, wherein
A is phenyl, a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms, a five-membered heteroaromatic ring containing one, two, or three heteroatoms which are, independently, N, O, or S; a C4-14 bicyclic ring, alkylheteroaryl, or alkylaryl; B is a six or seven-membered ring or a six or seven-membered bridged or spiro-fused ring containing zero, one, or two heteroatoms, which are, independently, N, O, or S, a five- membered ring containing zero, one, or two heteroatoms, which are, independently, N, O, or S; a four-membered ring containing zero, one, or two heteroatoms, which are, independently, N, O, or S, or a C5-14 bicyclic ring,
When R1 and R1’ are attached to a carbon, they are, independently, hydrogen, halogen (including F, Cl, Br, and I), SF5, CF3, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C1-6 alkoxy, C1-6 haloalkoxy, C2-6 alkenyl, cyano, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C1-6 alkyl, arylalkoxycarbonyl, carboxy, C1-6 haloalkyl, heterocyclylalkyl, or C1-6 hydroxyalkyl;
When R1 and R1’ are attached to a nitrogen, they are, independently, hydrogen, C2-6 alkoxy, C3-6 alkoxyalkyl, C2-6 alkenyl, alkoxycarbonyl, carbonylalkyl, carbonyl aryl, C1-6 alkyl, heterocyclylalkyl, C2-6 hydroxyalkyl, or S(O)2R’;
Each R’ is independently H, C1-6 alkyl, C1-6 haloalkyl, C1-6 alkoxy, C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, aryl, heteroaryl, alkylaryl, or arylalkyl, or if two R’ reside on the same nitrogen atom, they can come together to form a C3-6 ring optionally containing a N, O, or S heteroatom;
The R’ groups can optionally be substituted with one or more substituents, which substituents are, independently, halo, C1-6 haloalkyl, C1-6 hydroxyalkyl, hydroxyl, carboxyl, acyl, aryl, acyloxy, amino, amido, carboxyl derivatives, alkylamino, dialkylamino, arylamino, alkoxy, alkoxyalkyl, aryloxy, nitro, cyano, sulfonic acid, thiol, imine, sulfonyl, sulfanyl, sulfinyl, sulfamonyl, ester, carboxylic acid, amide, phosphonyl, phosphinyl, phosphoryl, phosphine, thioester, thioether, acid halide, anhydride, oxime, hydrozine, carbamate, phosphonic acid, or phosphonate, either unprotected, or protected as necessary, as known to those skilled in the art, for example, as taught in Greene, et al., Protective Groups in Organic Synthesis, John Wiley and Sons, Second Edition, 1991, hereby incorporated by reference;
u and v are independently 0, 1, 2, 3, 4 or 5;
X is
Figure imgf000082_0001
R3 is H, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, or C2-6 alkynyl; R2 is C1-6 alkyl, C1-6 haloalkyl, C2-8 alkoxyalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, such as phenyl, heteroaryl, including six-membered heteroaromatic rings containing one, two, or three nitrogen atoms and five-membered heteroaromatic rings containing one, two, or three heteroatoms, which, independently, are N, O, or S, alkylaryl, arylalkyl, a six-membered ring or a six-membered bridged or spiro-fused ring containing zero, one, or two heteroatoms which are, independently, N, O, or S, a seven-membered bridged or spiro-fused ring containing zero, one, or two heteroatoms which are, independently, N, O, or S, a five-membered ring containing zero, one, or two heteroatoms which are, independently, N, O, or S; a four-membered ring containing zero, one, or two heteroatoms which are, independently, N, O, or S; cycloalkyl, alkylheteroaryl, or alkylaryl;
R2 is optionally substituted with one or more substituents, which each, independently, are halogen (including F, Cl, Br, and I), CF3, SF5, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C1-6 alkoxy, C1-6 haloalkoxy, cyano, azido, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C1-6 alkyl, arylalkoxycarbonyl, carboxy, C1-6 haloalkyl, heterocyclylalkyl, or C1-6 hydroxyalkyl; or is optionally substituted with aryl, substituted aryl, heteroaryl, or substituted heteroaryl, where substituents on the substituted aryl and substituted heteroaryl are selected from the group consisting of halogen, SF5, CF3, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C(O)R’, C1-6 alkoxy, cyano, azido, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, and C1-6 alkyl,
R2 and R3 can come together with the nitrogen to which they are attached to form a 6-10 membered bicyclic or bridged ring, a 3 to 8 saturated ring, or a 5 membered unsaturated ring; such bicyclic, bridged, saturated and unsaturated rings optionally containing one or more additional heteroatoms, where each is, independently, O, S or N, and optionally being substituted with one or more substituents, wherein each, independently, is halogen (including F, Cl, Br, I), CF3, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C1-6 alkoxy, cyano, azido, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C1-6 alkyl, arylalkoxycarbonyl, carboxy, C1-6 haloalkyl, heterocyclylalkyl, or C1-6 hydroxyalkyl.
In a second embodiment, the compounds for use in the invention are those having formula (A-II):
Figure imgf000084_0002
or an enantiomer, diastereomer or pharmaceutically accepted salt, hydrate, or solvate thereof, including a hydrate, a solvate, a pharmaceutically acceptable salt, a prodrug, or a complex thereof,
wherein
R1 and R1’ are as defined with respect to formula (A-I);
u and v are independently 0, 1, 2, 3, 4 or 5;
C is phenyl, a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms, a five-membered heteroaromatic ring containing one, two, or three heteroatoms which are, independently, N, O, or S; a C4-14 bicyclic ring, alkylaryl, or alkylheteroaryl;
D is phenyl, a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms, a five-membered heteroaromatic ring containing one, two, or three heteroatoms, which are, independently, N, O, or S, or a C5-14 bicyclic ring,
Figure imgf000084_0001
R4 is H or C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl;
where, in one embodiment, if C is phenyl, D is not phenyl or a 5-membered ring heteroaryl, and in another embodiment, if C is phenyl and D is phenyl or a 5-membered ring heteroaryl, then R5 is not alkylaryl, alkenyl, or a six-membered bridged ring;
R5 is alkylaryl, arylalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, such as phenyl, heteroaryl, including six-membered heteroaromatic rings containing one, two, or three nitrogen atoms and five-membered heteroaromatic rings containing one, two, or three heteroatoms, which, independently, are N, O, or S; and a six-membered bridged or spiro-fused ring containing zero, one, or two heteroatoms which are, independently, N, O, or S;
R5 is optionally substituted with one or more substituents, each of which is,
independently, halogen (including F, Cl, Br, and I), CF3, SF5, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C1-6 alkoxy, C1-6 haloalkoxy, cyano, azido, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C1-6 alkyl, cycloalkyl, arylalkoxycarbonyl, carboxy, C1-6 haloalkyl, heterocyclylalkyl, or C1-6 hydroxyalkyl; or is substituted with aryl, substituted aryl, heteroaryl, or substituted heteroaryl, where substituents on the substituted aryl and substituted heteroaryl are selected from the group consisting of halogen, SF5, CF3, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C(O)R’, C1-6 alkoxy, cyano, azido, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, and C1-6 alkyl;
or when Y is
Figure imgf000085_0001
, R4 and R5 together with the nitrogen to which they are attached form a 3 to 4 membered ring optionally substituted with one or more substituents, each of which is, independently, halogen (including F, Cl, Br, I), CF3, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C1-6 alkoxy, cyano, azido, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C1-6 alkyl, arylalkoxycarbonyl, carboxy, C1-6 haloalkyl, heterocyclylalkyl, or C1-6 hydroxyalkyl.
Figure imgf000085_0002
In one embodiment of the compounds of Formula II, D is , where R6 is H, Cl, F or Br, and R7 is H, methyl, F or Cl.
In one aspect of this embodiment, when Y is
Figure imgf000085_0003
, R5 is not
Figure imgf000085_0004
Figure imgf000085_0005
In another aspect of this embodiment, when R4 is ethyl, then R5 is not
Figure imgf000085_0006
. In one embodiment of the compounds of formula (A-II), C is a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms, a five-membered
heteroaromatic ring containing one, two, or three heteroatoms which are, independently, N, O, or S; a C4-14 bicyclic ring, alkylaryl, or alkylheteroaryl.
In one embodiment of the compounds of formula (A-II), D is a C4-14 bicyclic ring.
In another embodiment of the compounds of formula (A-II), R5 is arylalkyl, C2-6 alkynyl, aryl, such as phenyl, heteroaryl, including six-membered heteroaromatic rings containing one, two, or three nitrogen atoms and five-membered heteroaromatic rings containing one, two, or three heteroatoms, which, independently, are N, O, or S; and a six-membered spiro-fused ring containing zero, one, or two heteroatoms which are, independently, N, O, or S. In a third embodiment, the compounds for use in the invention are those having formula (A-III):
Figure imgf000086_0001
or an enantiomer, diastereomer or pharmaceutically accepted salt, hydrate, or solvate thereof, including a hydrate, a solvate, a pharmaceutically acceptable salt, a prodrug, or a complex thereof,
wherein
R1 and R1’ are as defined with respect to formula (A-I);
u and v are independently 0, 1, 2, 3, 4 or 5;
E is a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms, a five-membered heteroaromatic ring containing one, two, or three heteroatoms, where each is, independently, N, O, or S; a C4-14 bicyclic ring, alkylheteroaryl, or alkylaryl;
F is a five-membered heteroaromatic ring containing one, two, or three heteroatoms which are, independently, N, O, or S, or a C4-14 bicyclic ring;
Figure imgf000086_0002
R8 is H, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, or C2-6 alkynyl;
R9 is C1-6 alkyl, C1-6 haloalkyl, C2-8 alkoxyalkyl, a six-membered ring or a six- membered bridged or spiro-fused ring containing zero, one, or two heteroatoms, which are independently N, O, or S, a seven-membered bridged or spiro-fused ring containing zero, one, or two heteroatoms, which are, independently, N, O, or S, a five- membered ring containing zero, one, or two heteroatoms, which are, independently, N, O, or S; a four-membered ring containing zero, one, or two heteroatoms, which are, independently, N, O, or S, or a three membered ring;
R9 is optionally substituted with one or more substituents, each of which is independently halogen (including F, Cl, Br, and I), CF3, SF5, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C1-6 alkoxy, C1-6 haloalkoxy, cyano, azido, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C1-6 alkyl, cycloalkyl, arylalkoxycarbonyl, carboxy, C1-6 haloalkyl, heterocyclylalkyl, or C1-6 hydroxyalkyl; or is substituted with aryl, substituted aryl, heteroaryl, or substituted heteroaryl, where substituents on the substituted aryl and substituted heteroaryl are selected from the group consisting of halogen, SF5, CF3, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C(O)R’, C1-6 alkoxy, cyano, azido, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, and C1-6 alkyl;
R8 and R9 can come together with the nitrogen to which they are attached to form a 6-10 membered bicyclic or bridged ring or a 3 to 8 saturated ring; such bicyclic, bridged and saturated ring moiety optionally containing one or more additional heteroatoms which, independently, are O, S or N and optionally being substituted with one or more substituents, each, independently, is halogen (including F, Cl, Br, and I), CF3, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C1-6 alkoxy, cyano, azido, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C1-6 alkyl, arylalkoxycarbonyl, carboxy, C1-6 haloalkyl, heterocyclylalkyl, or C1-6 hydroxyalkyl. In a fourth embodiment, the compounds for use in the invention are those having formula (A-IV):
Figure imgf000088_0001
or an enantiomer, diastereomer or pharmaceutically accepted salt, hydrate, or solvate thereof, including a hydrate, a solvate, a pharmaceutically acceptable salt, a prodrug, or a complex thereof,
wherein
G is phenyl, a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms, a five-membered heteroaromatic ring containing one, two, or three heteroatoms, which are, independently, N, O, or S; a C4-14 bicyclic ring, alkylheteroaryl, or alkylaryl;
H is phenyl, a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms, a six-membered non-aromatic ring optionally containing one, two, or three heteroatoms, which are, independently, N, O, or S; or a C4-14 bicyclic ring;
When R1 and R1’ are attached to a carbon they are, independently, hydrogen, halogen (including F, Cl, Br, and I), CF3, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C1-6 alkoxy, cyano, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C1-6 alkyl, arylalkoxycarbonyl, carboxy, C1-6 haloalkyl, heterocyclylalkyl, or C1-6 hydroxyalkyl;
When R1 and R1’ are attached to a nitrogen they are, independently, hydrogen, C1-6 alkoxy, C3-6 alkoxyalkyl, alkoxycarbonyl, carbonylalkyl, carbonyl aryl, C1-6 alkyl, C2-6 alkynyl, C2-6 alkenyl, heterocyclylalkyl, C1-6 hydroxyalkyl, or S(O)2R’;
Each R’ is independently H, C1-6 alkyl, C1-6 haloalkyl, C1-6 alkoxy, C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, aryl, heteroaryl, alkylaryl, or arylalkyl, or if two R’ reside on the same nitrogen atom they can come together to form a C3-6 alkyl ring optionally containing a N, O, or S; wherein the R’ groups can be substituted with one or more substituents as defined above, for example, C1-6 hydroxyalkyl, aminoalkyl, and alkoxyalkyl;
u and v are independently 0, 1, 2, 3, 4 or 5;
W is
Figure imgf000088_0002
R10 is H, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, or C2-6 alkynyl; R11 is C1-6 alkyl, C1-6 haloalkyl, C2-8 alkoxyalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, heteroaryl, alkylaryl, arylalkyl, phenyl, a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms, a six-membered ring or a six-membered bridged or spiro-fused ring containing zero, one, or two heteroatoms, which are, independently, N, O, or S, a seven- membered bridged or spiro-fused ring containing zero, one, or two heteroatoms, which are, independently, N, O, or S, a five-membered heteroaromatic ring containing one, two, or three heteroatoms, which are, independently, N, O, or S, a five-membered ring containing zero, one, or two heteroatoms, which are, independently, N, O, or S; a four-membered ring containing zero, one, or two heteroatoms, which are, independently, N, O, or S; a three membered ring, alkylheteroaryl, or alkylaryl;
wherein R11 is optionally substituted with one or more substituents selected from the group consisting of halogen (including F, Cl, Br, and I), SF5, CF3, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C1-6 alkoxy, C1-6 haloalkoxy, C2-6 alkenyl, cyano, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C1-6 alkyl, arylalkoxycarbonyl, carboxy, C1-6 haloalkyl, heterocyclylalkyl, C1-6 hydroxyalkyl, aryl, substituted aryl, heteroaryl, and substituted heteroaryl, where substituents on the substituted aryl and substituted heteroaryl are selected from the group consisting of halogen, SF5, CF3, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C(O)R’, C1-6 alkoxy, cyano, azido, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl,
alkoxycarbonylalkyl, and C1-6 alkyl;
or R10 and R11 can come together with the nitrogen to which they are attached to form a 6-10 membered bicyclic or bridged ring or a 3 to 8 saturated ring; such bicyclic, bridged or saturated ring moiety optionally containing one or more additional heteroatoms, which are each, independently, O, S or N, and optionally substituted with one or more substituents, each of which is, independently, halogen (including F, Cl, Br, and I), CF3, SF5, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C1-6 alkoxy, cyano, azido, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C1- 6 alkyl, arylalkoxycarbonyl, carboxy, C1-6 haloalkyl, heterocyclylalkyl, or C1-6 hydroxyalkyl; In a fifth embodiment, the compounds for use in the invention are those having formula (A-V):
Figure imgf000090_0001
or an enantiomer, diastereomer or pharmaceutically accepted salt, hydrate, or solvate thereof, including a hydrate, a solvate, a pharmaceutically acceptable salt, a prodrug, or a complex thereof,
wherein
R1 and R1’ are as defined with respect to formula (A-I);
u and v are independently 0, 1, 2, 3, 4 or 5;
I is phenyl, a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms, a five-membered heteroaromatic ring containing one, two, or three heteroatoms, which are, independently, N, O, or S, a C4-14 bicyclic ring; alkylheteroaryl, or alkylaryl;
J is a five-membered heteroaromatic ring containing one, two, or three heteroatoms, which are, independently, N, O, or S, a six or seven-membered ring or a six or seven-membered bridged or spiro-fused ring containing zero, one, or two heteroatoms which are, independently, N, O, or S, a five-membered ring containing zero, one, or two heteroatoms which are, independently, N, O, or S; or a four- membered ring containing zero, one, or two heteroatoms which are, independently, N, O, or S,
W is
Figure imgf000090_0002
R12 is H, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, or C2-6 alkynyl;
R13 is C2-6 alkenyl, C2-6 alkynyl, aryl, including phenyl, heteroaryl, including six- membered heteroaromatic rings containing one, two, or three nitrogen atoms and five-membered heteroaromatic rings containing one, two, or three heteroatoms, which are, independently, N, O, or S; alkylaryl, arylalkyl, a C4-14 bicyclic ring; a six- membered bridged or spiro-fused ring containing zero, one, or two heteroatoms which are, independently, N, O, or S,
R13 is optionally substituted with one or more substituents each independently selected from the group consisting of hydrogen, halogen (F, Cl, Br, I), CF3, SF5, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C(O)R’, C1-6 alkoxy, C1-6 haloalkoxy, cyano, azido, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C1- 6 alkyl, cycloalkyl, arylalkoxycarbonyl, carboxyl, haloalkyl, heterocyclylalkyl, or C1-6 hydroxyalkyl; or is optionally substituted with aryl, substituted aryl, heteroaryl, and substituted heteroaryl, where substituents on the substituted aryl and substituted heteroaryl are selected from the group consisting of halogen, SF5, CF3, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C(O)R’, C1-6 alkoxy, cyano, azido, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, and C1-6 alkyl;
or R12 and R13 together with the nitrogen to which they are attached form a 3 to 4 membered ring optionally substituted with one or more substituents each independently selected from the group consisting of hydrogen, halogen (F, Cl, Br, I), CF3, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C1-6 alkoxy, cyano, azido, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C1-6 alkyl, arylalkoxycarbonyl, carboxy, C1-6 haloalkyl, heterocyclylalkyl, and C1-6 hydroxyalkyl; In a fifth embodiment, the compounds for use in the invention are those having formula (A-VI):
or an enantiomer, diastereomer or pharmaceutically
Figure imgf000091_0001
accepted salt, hydrate, or solvate thereof, including a hydrate, a solvate, a pharmaceutically acceptable salt, a prodrug, or a complex thereof,
wherein
R1 and R1’ are as defined with respect to formula (A-I);
u and v are independently 0, 1, 2, 3, 4 or 5;
K is a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms, a five-membered heteroaromatic ring containing one, two, or three heteroatoms which are, independently, N, O, or S; a C4-14 bicyclic ring, alkylheteroaryl, or alkylaryl;
L is a five-membered heteroaromatic ring containing one, two, or three heteroatoms which are, independently, N, O, or S, a six or seven -membered ring or a six or seven-membered bridged or spiro-fused ring containing zero, one, or two heteroatoms which are, independently, N, O, or S, a five-membered ring containing zero, one, or two heteroatoms which are, independently, N, O, or S; a four-membered ring containing zero, one, or two heteroatoms which are, independently, N, O, or S, or a C4-14 bicyclic ring;
W is
Figure imgf000092_0001
R14 is H, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, or C2-6 alkynyl;
R15 is C1-6 alkyl, C1-6 haloalkyl, C2-8 alkoxyalkyl, a six-membered ring or a six- membered bridged or spiro-fused ring containing zero, one, or two heteroatoms which are, independently, N, O, or S, a seven-membered bridged or spiro-fused ring containing zero, one, or two heteroatoms which are, independently, N, O, or S, a five- membered ring containing zero, one, or two heteroatoms which are, independently, N, O, or S; a four-membered ring containing zero, one, or two heteroatoms which are, independently, N, O, or S;
R15 is optionally substituted with one or more substituents which are, independently, halogen (F, Cl, Br, I), SF5, CF3, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C1-6 alkoxy, C1-6 haloalkoxy, cyano, azido, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl,
alkoxycarbonylalkyl, C1-6 alkyl, cycloalkyl, arylalkoxycarbonyl, carboxy, C1-6 haloalkyl, heterocyclylalkyl, or C1-6 hydroxyalkyl; or is optionally substituted with aryl, substituted aryl, heteroaryl, or substituted heteroaryl, where substituents on the substituted aryl and substituted heteroaryl are selected from the group consisting of halogen, SF5, CF3, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C(O)R’, C1-6 alkoxy, cyano, azido, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, and C1-6 alkyl;
or R14 and R15 can come together with the nitrogen to which they are attached to form a 6-10 membered bicyclic or bridged ring or a 3 to 8 saturated ring; such bicyclic, bridged and saturated ring moiety optionally containing one or more additional heteroatoms which are, independently, O, S or N, and optionally being substituted with one or more substituents each independently selected from the group consisting of halogen (including F, Cl, Br, and I), SF5, CF3, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C1-6 alkoxy, cyano, azido, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C1-6 alkyl, arylalkoxycarbonyl, carboxy, C1-6 haloalkyl, heterocyclylalkyl, and C1-6 hydroxyalkyl. In a seventh embodiment, the compounds for use in the invention are those having formula (A-VII):
Figure imgf000093_0001
or an enantiomer, diastereomer or pharmaceutically accepted salt, hydrate, or solvate thereof, including a hydrate, a solvate, a pharmaceutically acceptable salt, a prodrug, or a complex thereof,
wherein
R1 and R1’ are as defined with respect to formula (A-I);
u and v are independently 0, 1, 2, 3, 4 or 5;
M is phenyl, a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms, a five-membered heteroaromatic ring containing one, two, or three heteroatoms which are, independently, N, O, or S, a C4-14 bicyclic ring, alkylheteroaryl, or alkylaryl;
N is phenyl, a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms, a five-membered heteroaromatic ring containing one, two, or three heteroatoms independently N, O, or S, a six or seven-membered ring or a six or seven- membered bridged or spiro-fused ring containing zero, one, or two heteroatoms which are, independently, N, O, or S, a five-membered ring containing zero, one, or two heteroatoms which are, independently, N, O, or S; a four-membered ring containing zero, one, or two heteroatoms which are, independently, N, O, or S; or a C4-14 bicyclic ring,
V is
Figure imgf000093_0002
R16 is C1-6 alkyl, C1-6 haloalkyl, C2-8 alkoxyalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, such as phenyl, heteroaryl, such as a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms or a five-membered heteroaromatic ring containing one, two, or three heteroatoms which are, independently, N, O, or S; a six- membered ring or a six-membered bridged or spiro-fused ring containing zero, one, or two heteroatoms which are, independently, N, O, or S, a five-membered ring containing zero, one, or two heteroatoms which are, independently, N, O, or S; a four- membered ring containing zero, one, or two heteroatoms which are, independently, N, O, or S; alkylaryl, arylalkyl, alkylheteroaryl, or alkylaryl,
wherein R16 is optionally substituted with one or more substituents selected from the group consisting of halogen (including F, Cl, Br, and I), SF5, CF3, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C1-6 alkoxy, C1-6 haloalkoxy, C2-6 alkenyl, cyano, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C1-6 alkyl, arylalkoxycarbonyl, carboxy, C1-6 haloalkyl, heterocyclylalkyl, C1-6 hydroxyalkyl, aryl, substituted aryl, heteroaryl, and substituted heteroaryl, where substituents on the substituted aryl and substituted heteroaryl are selected from the group consisting of halogen, SF5, CF3, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C(O)R’, C1-6 alkoxy, cyano, azido, C2-6 alkynyl, C3-6 alkoxyalkyl,
alkoxycarbonyl, alkoxycarbonylalkyl, and C1-6 alkyl. Specific compounds having formulae (A-I) to (A-VII) which are appropriate for use in embodiments according to the invention include, but are not limited to:
Figure imgf000094_0001
Figure imgf000095_0001
Figure imgf000096_0001
Figure imgf000097_0002
Particularly preferred compounds include:
Figure imgf000097_0001
, or a pharmaceutically acceptable salt or prodrug thereof.
A particularly preferred compound has the formula:
Figure imgf000098_0001
or a pharmaceutically acceptable salt thereof. The effective amount of the compound having formulae (A-I) to (A-VII) or a
pharmaceutically acceptable salt, an enantiomer, or a diastereomer thereof may be determined by routine experimentation, but can be in range of 7 to 600 mg, preferably from 70 to 600 mg per unit dosage form.
Suitable processes for synthesizing these compounds can be synthesized by any method or synthetic scheme known in the art or to be discovered. For example, the compounds can be prepared according to Scheme 1, shown below. Examples of specific synthetic compounds are described in WO 2017156255.
Scheme 1
Figure imgf000098_0002
(c) Compounds of formula (B-I)
Examples of compounds of formula (B-I) (or pharmaceutically acceptable salts thereof), synthesis, biologic activities, uses or other related information thereof are described in International Patent Application Publication WO 2018039531 and U.S. Patent Application Publication US 20180161307, the contents of which are hereby incorporated by reference in their entireties.
In one embodiment, the compounds for use in the invention are those having formula (B- I):
, or pharmaceutically acceptable salts thereof, wherein:
Figure imgf000099_0001
R1 is C1-6 alkyl optionally substituted with 1 to 3 R1A , C3-8cycloalkyl optionally substituted with 1 to 4 R1B , or 3 to 8 membered monocyclic or bicyclic heterocyclyl having 1 to 3 heteroatoms selected from N, O, and S, optionally substituted with 1 to 3 R1C;
each R1A is independently halogen, -OH, -CN, -C1-2 haloalkyl, -C(O)NRXRY , C6-10 aryl optionally substituted with 1 to 3 R1D , or a 5 to 8 membered heteroaryl having 1 to 3 heteroatoms selected from N, O, and S, optionally substituted with 1 to 3 R1D , provided no more than 1 R1A is C6-10 aryl optionally substituted with 1 to 3 R1Dor 5 to 8 membered heteroaryl having 1 to 3 heteroatoms selected from N, O, and S optionally substituted with 1 to 3 R1D;
each R1B is independently -CN, halogen, C1-6alkyl optionally substituted with 1 to 3 -OH or -NRaRb, C2-4 alkynyl, C1-4 alkoxy, C1-2 haloalkyl, C3-6 cycloalkyl, -C(O)NRXRY , or a 5 to 8 membered heteroaryl having 1 to 3 heteroatoms selected from N, O, and S optionally substituted with 1 to 3 R1D , provided no more than 1 R1B is C3-6 cycloalkyl or 5 to 8 membered heteroaryl having 1 to 3 heteroatoms selected from N, O, and S optionally substituted with 1 to 3 R1D; each R1C is independently C1-6 alkyl, oxo, C1-4haloalkyl, -C(O)H, -C(O)C1-4 alkyl, - C(O)OC1-4alkyl, or a 5 to 12 membered heteroaryl having 1 to 3 heteroatoms selected from N, O, and S optionally substituted with 1 to 3 R1D , provided no more than 1 R1C is a 5 to 12 membered heteroaryl having 1 to 3 heteroatoms selected from N, O, and S optionally substituted with 1 to 3 R1D; each RX is independently -H, C3-6 cycloalkyl, C1-6 alkyl optionally substituted with 1 to 3 RZ , 3 to 8 membered monocyclic or bicyclic heterocyclyl having 1 to 3 heteroatoms selected from N, O, and S, optionally substituted with 1 to 3 RZ;
each RY is independently -H or C1 alkyl optionally substituted with 1 to 3 RZ;
or RX and RY are taken together to form a 3 to 8 membered monocyclic or bicyclic heterocyclyl having 1 to 3 heteroatoms selected from N, O, and S, optionally substituted with 1 to 3 RZ;
wherein each RZ is independently halogen, methyl, ethyl, oxo, -OH, -S(O)2C1-3alkyl, or 3 to 8 membered monocyclic or bicyclic heterocyclyl having 1 to 3 heteroatoms selected from N, O, and S;
each Ra is -H, C1-3alkyl, or a 3 to 8 membered monocyclic or bicyclic heterocyclyl having 1 to 3 heteroatoms selected from N, O, and S, optionally substituted with 1 to 3 RZ;
each Rb is -H or C1-3alkyl; or
Ra and Rb taken together form a 3 to 8 membered monocyclic or bicyclic heterocycle optionally substituted with 1 to 3 RZ;
the moiety
Figure imgf000100_0001
is a pyrrolidine or a 5-7 membered bicyclic heterocycle having one nitrogen, optionally substituted with 1 to 6 R2 groups; wherein each R2 is independently halogen, C1-3alkyl, -OH, or -OC1-3alkyl;
R3 is -H, halogen, or C1-4 alkyl;
R4 is C6-10 aryl optionally substituted with 1 to 5 R4A , or 5 to 12 membered heteroaryl having 1 to 3 heteroatoms selected from N, O, and S, optionally substituted with 1 to 4 R4B; and each R1D , R4A , and R4B are independently -CN, halogen, C1-4 alkyl, -OC1-4alkyl, -OC1- 4 haloalkyl, or C1-4 haloalkyl. In certain embodiments, R4 is phenyl optionally substituted with 1 to 5 R4A or pyridinyl optionally substituted with 1 to 4 R4B .
In certain embodiments, R4 is phenyl optionally substituted with 1 to 3 R4A .
In certain embodiments, each R4A is independently CI, F, -CF3, -CHF2,-CH3, -OCF3, - OCF2H, or - CN. In certain embodiments, R3 is -Cl or -CH3, preferably -CH3.
In certain embodiments, the moiety
Figure imgf000101_0001
Figure imgf000101_0002
In certain embodiments, the moiety
Figure imgf000101_0003
Figure imgf000101_0004
Exemplary compounds having formula (B-I) have the following structures, where the substituents are as described previously or in WO 2018039531 and US 20180161307:
Figure imgf000102_0001
Specific compounds having formula (B-I) which are appropriate for use in embodiments according to the invention include, but are not limited to:
Figure imgf000103_0001
Figure imgf000104_0001
Figure imgf000105_0001
Figure imgf000106_0001
Figure imgf000107_0001
Figure imgf000108_0001
Figure imgf000109_0001
Figure imgf000110_0001
Figure imgf000111_0001
Figure imgf000112_0001
Figure imgf000113_0001
Figure imgf000114_0001
Figure imgf000115_0002
, or pharmaceutically acceptable salts. Preferably, the specific compounds of formula (B-I) include:
Figure imgf000115_0001
The effective amount of the compound having formula (B-I) or a pharmaceutically acceptable salt thereof, may be determined by routine experimentation, but can be in range of from about 0.00001 mg/kg body weight per day to about 10 mg/kg body weight per day, such as from about 0.0001 mg/kg body weight per day to about 10 mg/kg body weight per day, or such as from about 0.001 mg/kg body weight per day to about 1 mg/kg body weight per day, or such as from about 0.01 mg/kg body weight per day to about 1 mg/kg body weight per day, or such as from about 0.05 mg/kg body weight per day to about 0.5 mg/kg body weight per day, or such as from about 0.3 mg to about 30 mg per day, or such as from about 30 mg to about 300 mg per day.
Suitable processes for synthesizing these compounds can be synthesized by any method or synthetic scheme known in the art or to be discovered. For example, the compounds can be prepared according to Schemes 1 and 2, shown below. Examples of specific synthetic compounds are described in WO 2018039531 and US 20180161307.
Scheme 1
Figure imgf000116_0001
Al can be converted to A2 by treatment with an appropriate alkyl halide such as (3- bromopropoxy)(tert-butyl)diphenylsilane in the presence of a suitable catalyst such as palladium dichloride bis(acetonitrile. A2 can be converted to A3 through a deprotection with a suitable reagent such as tetrabutylammonium fluoride and further halogenated to A4 with a suitable reagents such as carbon tetrabromide and triphenylphosphine. Cyclization to A5 can be effected with an appropriate radical initiator such as 2,2'-azobis(2-methylpropionitrile) in the presence other reagents such as tributyl tin hydride. Ester hydrolysis with a suitable reagent such as lithium hydroxide followed by amide formation via treatment with an appropriate coupling reagent such as HATU and the appropriate aniline or by conversion to the acid chloride with a reagent such a thionyl chloride or oxalyl chloride followed by treatment with the appropriate aniline gives A6. The aniline may be varied based on the R4 groups disclosued herein.
Formation of A7 can be effected by treatment with a suitable reagent such as oxalyl chloride or ethyl 2-chloro-2-oxoacetate and may or may not require the addition of catalyst such as aluminum chloride. Hydrolysis to A8 via a suitable reagent such as lithium hydroxide is followed by preparation of A9 by the coupling of the appropriate amine such as l,l,l- trifluoropropan-2-amine in the presence of a coupling reagent such as HATU. The amine may be varied for particular R1 groups disclosed herein. Scheme 2
Figure imgf000117_0001
Bl can be converted to B2 by treatment with methyl 2-chloro-2-oxoacetate. Conversion to B3 can be carried out with an appropriate hydroxide reagent such as lithium hydroxide or in some cases with hydrogen gas and a suitable catalyst if the ester is a benzyl ester. B3 can be converted to B4 by treatment with a suitable reagent such as N,N'-diisopropylcarbodiimide and alkyne reagent B3B at elevated temperatures. Alternatively, B3 can be treated with oxalyl chloride, worked up and treated with B3B and a suitable base such as 2,6-di-tertbutylpyridine to give B4. Hydrolysis to B5 can be effected with a suitable reagent such as lithium hydroxide. Conversion to B6 occurs by addition of an appropriate amine (e.g. R1- NH2 ) and an amide coupling reagent such as HATU. B6 can be converted to desired B7 by treatment with a suitable reagent such as lithium hydroxide at elevated temperatures followed a second amide coupling with an appropriate aniline or heteroaryl amine (e.g R4-NH2) and a coupling reagent such as HATU. Alternatively, B5 can be treated with an aniline or heteroaryl amine in the presence of a reagent such as Lithium bis(trimethylsilyl)amide to give B6B followed by treatment with an appropriate amine and a coupling reagent such as HATU to give desired B7. Compositions, Therapeutic Combinations, and Vaccines
The application also relates to compositions, therapeutic combinations, more particularly kits, and vaccines comprising one or more HBV antigens, polynucleotides, and/or vectors encoding one or more HBV antigens according to the application. Any of the HBV antigens, polynucleotides (including RNA and DNA), and/or vectors of the application described herein can be used in the compositions, therapeutic combinations or kits, and vaccines of the application.
In an embodiment of the application, a composition comprises an isolated or non-naturally occurring nucleic acid molecule (DNA or RNA) comprising polynucleotide sequence encoding a truncated HBV core antigen consisting of an amino acid sequence that is at least 90% identical to SEQ ID NO: 2 or SEQ ID NO: 4, or an HBV polymerase antigen comprising an amino acid sequence that is at least 90% identical to SEQ ID NO: 7, a vector comprising the isolated or non- naturally occurring nucleic acid molecule, and/or an isolated or non-naturally occurring polypeptide encoded by the isolated or non-naturally occurring nucleic acid molecule.
In an embodiment of the application, a composition comprises an isolated or non-naturally occurring nucleic acid molecule (DNA or RNA) comprising a polynucleotide sequence encoding an HBV Pol antigen comprising an amino acid sequence that is at least 90% identical to SEQ ID NO: 7, preferably 100% identical to SEQ ID NO: 7. In an embodiment of the application, a composition comprises an isolated or non-naturally occurring nucleic acid molecule (DNA or RNA) encoding a truncated HBV core antigen consisting of an amino acid sequence that is at least 90% identical to SEQ ID NO: 2 or SEQ ID NO: 4, preferably 100% identical to SEQ ID NO: 2 or SEQ ID NO: 4.
In an embodiment of the application, a composition comprises an isolated or non-naturally occurring nucleic acid molecule (DNA or RNA) comprising a polynucleotide sequence encoding a truncated HBV core antigen consisting of an amino acid sequence that is at least 90% identical to SEQ ID NO: 2 or SEQ ID NO: 4, preferably 100% identical to SEQ ID NO: 2 or SEQ ID NO: 4; and an isolated or non-naturally occurring nucleic acid molecule (DNA or RNA) comprising a polynucleotide sequence encoding an HBV Pol antigen comprising an amino acid sequence that is at least 90% identical to SEQ ID NO: 7, preferably 100% identical to SEQ ID NO: 7. The coding sequences for the truncated HBV core antigen and the HBV Pol antigen can be present in the same isolated or non-naturally occurring nucleic acid molecule (DNA or RNA), or in two different isolated or non-naturally occurring nucleic acid molecules (DNA or RNA).
In an embodiment of the application, a composition comprises a vector, preferably a DNA plasmid or a viral vector (such as an adenoviral vector) comprising a polynucleotide encoding a truncated HBV core antigen consisting of an amino acid sequence that is at least 90% identical to SEQ ID NO: 2 or SEQ ID NO: 4, preferably 100% identical to SEQ ID NO: 2 or SEQ ID NO: 4.
In an embodiment of the application, a composition comprises a vector, preferably a DNA plasmid or a viral vector (such as an adenoviral vector), comprising a polynucleotide encoding an HBV Pol antigen comprising an amino acid sequence that is at least 90% identical to SEQ ID NO: 7, preferably 100% identical to SEQ ID NO: 7.
In an embodiment of the application, a composition comprises a vector, preferably a DNA plasmid or a viral vector (such as an adenoviral vector), comprising a polynucleotide encoding a truncated HBV core antigen consisting of an amino acid sequence that is at least 90% identical to SEQ ID NO: 2 or SEQ ID NO: 4, preferably 100% identical to SEQ ID NO: 2 or SEQ ID NO: 4; and a vector, preferably a DNA plasmid or a viral vector (such as an adenoviral vector), comprising a polynucleotide encoding an HBV Pol antigen comprising an amino acid sequence that is at least 90% identical to SEQ ID NO: 7, preferably 100% identical to SEQ ID NO: 7. The vector comprising the coding sequence for the truncated HBV core antigen and the vector comprising the coding sequence for the HBV Pol antigen can be the same vector, or two different vectors.
In an embodiment of the application, a composition comprises a vector, preferably a DNA plasmid or a viral vector (such as an adenoviral vector), comprising a polynucleotide encoding a fusion protein comprising a truncated HBV core antigen consisting of an amino acid sequence that is at least 90% identical to SEQ ID NO: 2 or SEQ ID NO: 4, preferably 100% identical to SEQ ID NO: 2 or SEQ ID NO: 4, operably linked to an HBV Pol antigen comprising an amino acid sequence that is at least 90% identical to SEQ ID NO: 7, preferably 100% identical to SEQ ID NO: 7, or vice versa. Preferably, the fusion protein further comprises a linker that operably links the truncated HBV core antigen to the HBV Pol antigen, or vice versa. Preferably, the linker has the amino acid sequence of (AlaGly)n, wherein n is an integer of 2 to 5.
In an embodiment of the application, a composition comprises an isolated or non-naturally occurring truncated HBV core antigen consisting of an amino acid sequence that is at least 90% identical to SEQ ID NO: 2 or SEQ ID NO: 4, preferably 100% identical to SEQ ID NO: 2 or SEQ ID NO: 4.
In an embodiment of the application, a composition comprises an isolated or non-naturally occurring HBV Pol antigen comprising an amino acid sequence that is at least 90% identical to SEQ ID NO: 7, preferably 100% identical to SEQ ID NO: 7.
In an embodiment of the application, a composition comprises an isolated or non-naturally occurring truncated HBV core antigen consisting of an amino acid sequence that is at least 90% identical to SEQ ID NO: 2 or SEQ ID NO: 4, preferably 100% identical to SEQ ID NO: 2 or SEQ ID NO: 4; and an isolated or non-naturally occurring HBV Pol antigen comprising an amino acid sequence that is at least 90% identical to SEQ ID NO: 7, preferably 100% identical to SEQ ID NO: 7.
In an embodiment of the application, a composition comprises an isolated or non-naturally occurring fusion protein comprising a truncated HBV core antigen consisting of an amino acid sequence that is at least 90% identical to SEQ ID NO: 2 or SEQ ID NO: 14, preferably 100% identical to SEQ ID NO: 2 or SEQ ID NO: 4, operably linked to an HBV Pol antigen comprising an amino acid sequence that is at least 90% identical to SEQ ID NO: 7, preferably 100% identical to SEQ ID NO: 7, or vice versa. Preferably, the fusion protein further comprises a linker that operably links the truncated HBV core antigen to the HBV Pol antigen, or vice versa. Preferably, the linker has the amino acid sequence of (AlaGly)n, wherein n is an integer of 2 to 5.
The application also relates to a therapeutic combination or a kit comprising
polynucleotides expressing a truncated HBV core antigen and an HBV pol antigen according to embodiments of the application. Any polynucleotides and/or vectors encoding HBV core and pol antigens of the application described herein can be used in the therapeutic combinations or kits of the application.
According to embodiments of the application, a therapeutic combination or kit for use in treating an HBV infection in a subject in need thereof, comprises:
i) at least one of:
a) a truncated HBV core antigen consisting of an amino acid sequence that is at least 95% identical to SEQ ID NO: 2, and
b) a first non-naturally occurring nucleic acid molecule comprising a first polynucleotide sequence encoding the truncated HBV core antigen
c) an HBV polymerase antigen having an amino acid sequence that is at least 90% identical to SEQ ID NO: 7, wherein the HBV polymerase antigen does not have reverse transcriptase activity and RNase H activity, and
d) a second non-naturally occurring nucleic acid molecule comprising a second polynucleotide sequence encoding the HBV polymerase antigen; and
ii) a capsid assembly modulator.
In a particular embodiment of the application, a therapeutic combination or kit comprises: i) a first non-naturally occurring nucleic acid molecule comprising a first polynucleotide sequence encoding a truncated HBV core antigen consisting of an amino acid sequence that is at least 95% identical to SEQ ID NO: 2; ii) a second non-naturally occurring nucleic acid molecule comprising a second polynucleotide sequence encoding an HBV polymerase antigen having an amino acid sequence that is at least 90% identical to SEQ ID NO: 7, wherein the HBV polymerase antigen does not have reverse transcriptase activity and RNase H activity; and iii) a capsid assembly modulator.
According to embodiments of the application, the polynucleotides in a vaccine combination or kit can be linked or separate, such that the HBV antigens expressed from such polynucleotides are fused together or produced as separate proteins, whether expressed from the same or different polynucleotides. In an embodiment, the first and second polynucleotides are present in separate vectors, e.g., DNA plasmids or viral vectors, used in combination either in the same or separate compositions, such that the expressed proteins are also separate proteins, but used in combination. In another embodiment, the HBV antigens encoded by the first and second polynucleotides can be expressed from the same vector, such that an HBV core-pol fusion antigen is produced. Optionally, the core and pol antigens can be joined or fused together by a short linker. Alternatively, the HBV antigens encoded by the first and second polynucleotides can be expressed independently from a single vector using a using a ribosomal slippage site (also known as cis-hydrolase site) between the core and pol antigen coding sequences. This strategy results in a bicistronic expression vector in which individual core and pol antigens are produced from a single mRNA transcript. The core and pol antigens produced from such a bicistronic expression vector can have additional N or C-terminal residues, depending upon the ordering of the coding sequences on the mRNA transcript. Examples of ribosomal slippage sites that can be used for this purpose include, but are not limited to, the FA2 slippage site from foot-and-mouth disease virus (FMDV). Another possibility is that the HBV antigens encoded by the first and second polynucleotides can be expressed independently from two separate vectors, one encoding the HBV core antigen and one encoding the HBV pol antigen.
In a preferred embodiment, the first and second polynucleotides are present in separate vectors, e.g., DNA plasmids or viral vectors. Preferably, the separate vectors are present in the same composition.
According to preferred embodiments of the application, a therapeutic combination or kit comprises a first polynucleotide present in a first vector, a second polynucleotide present in a second vector. The first and second vectors can be the same or different. Preferably the vectors are DNA plasmids.
In a particular embodiment of the application, the first vector is a first DNA plasmid, the second vector is a second DNA plasmid. Each of the first and second DNA plasmids comprises an origin of replication, preferably pUC ORI of SEQ ID NO: 21, and an antibiotic resistance cassette, preferably comprising a codon optimized Kanr gene having a polynucleotide sequence that is at least 90% identical to SEQ ID NO: 23, preferably under control of a bla promoter, for instance the bla promoter shown in SEQ ID NO: 24. Each of the first and second DNA plasmids independently further comprises at least one of a promoter sequence, enhancer sequence, and a polynucleotide sequence encoding a signal peptide sequence operably linked to the first polynucleotide sequence or the second polynucleotide sequence. Preferably, each of the first and second DNA plasmids comprises an upstream sequence operably linked to the first polynucleotide or the second polynucleotide, wherein the upstream sequence comprises, from 5’ end to 3’ end, a promoter sequence of SEQ ID NO: 18 or 19, an enhancer sequence, and a polynucleotide sequence encoding a signal peptide sequence having the amino acid sequence of SEQ ID NO: 9 or 15. Each of the first and second DNA plasmids can also comprise a polyadenylation signal located downstream of the coding sequence of the HBV antigen, such as the bGH polyadenylation signal of SEQ ID NO: 20.
In one particular embodiment of the application, the first vector is a viral vector and the second vector is a viral vector. Preferably, each of the viral vectors is an adenoviral vector, more preferably an Ad26 or Ad35 vector, comprising an expression cassette including the
polynucleotide encoding an HBV pol antigen or an truncated HBV core antigen of the application; an upstream sequence operably linked to the polynucleotide encoding the HBV antigen comprising, from 5’ end to 3’ end, a promoter sequence, preferably a CMV promoter sequence of SEQ ID NO: 19, an enhancer sequence, preferably an ApoAI gene fragment sequence of SEQ ID NO: 12, and a polynucleotide sequence encoding a signal peptide sequence, preferably an immunoglobulin secretion signal having the amino acid sequence of SEQ ID NO: 15; and a downstream sequence operably linked to the polynucleotide encoding the HBV antigen comprising a polyadenylation signal, preferably a SV40 polyadenylation signal of SEQ ID NO: 13.
In another preferred embodiment, the first and second polynucleotides are present in a single vector, e.g., DNA plasmid or viral vector. Preferably, the single vector is an adenoviral vector, more preferably an Ad26 vector, comprising an expression cassette including a polynucleotide encoding an HBV pol antigen and a truncated HBV core antigen of the application, preferably encoding an HBV pol antigen and a truncated HBV core antigen of the application as a fusion protein; an upstream sequence operably linked to the polynucleotide encoding the HBV pol and truncated core antigens comprising, from 5’ end to 3’ end, a promoter sequence, preferably a CMV promoter sequence of SEQ ID NO: 19, an enhancer sequence, preferably an ApoAI gene fragment sequence of SEQ ID NO: 12, and a polynucleotide sequence encoding a signal peptide sequence, preferably an immunoglobulin secretion signal having the amino acid sequence of SEQ ID NO: 15; and a downstream sequence operably linked to the polynucleotide encoding the HBV antigen comprising a polyadenylation signal, preferably a SV40 polyadenylation signal of SEQ ID NO: 13.
When a therapeutic combination of the application comprises a first vector, such as a DNA plasmid or viral vector, and a second vector, such as a DNA plasmid or viral vector, the amount of each of the first and second vectors is not particularly limited. For example, the first DNA plasmid and the second DNA plasmid can be present in a ratio of 10:1 to 1:10, by weight, such as 10:1, 9:1, 8:1, 7:1, 6:1, 5:1, 4:1, 3:1, 2:1, 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, or 1:10, by weight. Preferably, the first and second DNA plasmids are present in a ratio of 1:1, by weight. The therapeutic combination of the application can further comprise a third vector encoding a third active agent useful for treating an HBV infection.
Compositions and therapeutic combinations of the application can comprise additional polynucleotides or vectors encoding additional HBV antigens and/or additional HBV antigens or immunogenic fragments thereof, such as an HBsAg, an HBV L protein or HBV envelope protein, or a polynucleotide sequence encoding thereof. However, in particular embodiments, the compositions and therapeutic combinations of the application do not comprise certain antigens.
In a particular embodiment, a composition or therapeutic combination or kit of the application does not comprise a HBsAg or a polynucleotide sequence encoding the HBsAg.
In another particular embodiment, a composition or therapeutic combination or kit of the application does not comprise an HBV L protein or a polynucleotide sequence encoding the HBV L protein.
In yet another particular embodiment of the application, a composition or therapeutic combination of the application does not comprise an HBV envelope protein or a polynucleotide sequence encoding the HBV envelope protein.
Compositions and therapeutic combinations of the application can also comprise a pharmaceutically acceptable carrier. A pharmaceutically acceptable carrier is non-toxic and should not interfere with the efficacy of the active ingredient. Pharmaceutically acceptable carriers can include one or more excipients such as binders, disintegrants, swelling agents, suspending agents, emulsifying agents, wetting agents, lubricants, flavorants, sweeteners, preservatives, dyes, solubilizers and coatings. Pharmaceutically acceptable carriers can include vehicles, such as lipid nanoparticles (LNPs). The precise nature of the carrier or other material can depend on the route of administration, e.g., intramuscular, intradermal, subcutaneous, oral, intravenous, cutaneous, intramucosal (e.g., gut), intranasal or intraperitoneal routes. For liquid injectable preparations, for example, suspensions and solutions, suitable carriers and additives include water, glycols, oils, alcohols, preservatives, coloring agents and the like. For solid oral preparations, for example, powders, capsules, caplets, gelcaps and tablets, suitable carriers and additives include starches, sugars, diluents, granulating agents, lubricants, binders, disintegrating agents and the like. For nasal sprays/inhalant mixtures, the aqueous solution/suspension can comprise water, glycols, oils, emollients, stabilizers, wetting agents, preservatives, aromatics, flavors, and the like as suitable carriers and additives.
Compositions and therapeutic combinations of the application can be formulated in any matter suitable for administration to a subject to facilitate administration and improve efficacy, including, but not limited to, oral (enteral) administration and parenteral injections. The parenteral injections include intravenous injection or infusion, subcutaneous injection, intradermal injection, and intramuscular injection. Compositions of the application can also be formulated for other routes of administration including transmucosal, ocular, rectal, long acting implantation, sublingual administration, under the tongue, from oral mucosa bypassing the portal circulation, inhalation, or intranasal.
In a preferred embodiment of the application, compositions and therapeutic combinations of the application are formulated for parental injection, preferably subcutaneous, intradermal injection, or intramuscular injection, more preferably intramuscular injection.
According to embodiments of the application, compositions and therapeutic combinations for administration will typically comprise a buffered solution in a pharmaceutically acceptable carrier, e.g., an aqueous carrier such as buffered saline and the like, e.g., phosphate buffered saline (PBS). The compositions and therapeutic combinations can also contain pharmaceutically acceptable substances as required to approximate physiological conditions such as pH adjusting and buffering agents. For example, a composition or therapeutic combination of the application comprising plasmid DNA can contain phosphate buffered saline (PBS) as the pharmaceutically acceptable carrier. The plasmid DNA can be present in a concentration of, e.g., 0.5 mg/mL to 5 mg/mL, such as 0.5 mg/mL 1, mg/mL, 2 mg/mL, 3 mg/mL, 4 mg/mL, or 5 mg/mL, preferably at 1 mg/mL. Compositions and therapeutic combinations of the application can be formulated as a vaccine (also referred to as an“immunogenic composition”) according to methods well known in the art. Such compositions can include adjuvants to enhance immune responses. The optimal ratios of each component in the formulation can be determined by techniques well known to those skilled in the art in view of the present disclosure.
In a particular embodiment of the application, a composition or therapeutic combination is a DNA vaccine. DNA vaccines typically comprise bacterial plasmids containing a polynucleotide encoding an antigen of interest under control of a strong eukaryotic promoter. Once the plasmids are delivered to the cell cytoplasm of the host, the encoded antigen is produced and processed endogenously. The resulting antigen typically induces both humoral and cell-medicated immune responses. DNA vaccines are advantageous at least because they offer improved safety, are temperature stable, can be easily adapted to express antigenic variants, and are simple to produce. Any of the DNA plasmids of the application can be used to prepare such a DNA vaccine.
In other particular embodiments of the application, a composition or therapeutic combination is an RNA vaccine. RNA vaccines typically comprise at least one single-stranded RNA molecule encoding an antigen of interest, e.g., a fusion protein or HBV antigen according to the application. Once the RNA is delivered to the cell cytoplasm of the host, the encoded antigen is produced and processed endogenously, inducing both humoral and cell-mediated immune responses, similar to a DNA vaccine. The RNA sequence can be codon optimized to improve translation efficiency. The RNA molecule can be modified by any method known in the art in view of the present disclosure to enhance stability and/or translation, such by adding a polyA tail, e.g., of at least 30 adenosine residues; and/or capping the 5-end with a modified ribonucleotide, e.g., 7-methylguanosine cap, which can be incorporated during RNA synthesis or enzymatically engineered after RNA transcription. An RNA vaccine can also be self-replicating RNA vaccine developed from an alphavirus expression vector. Self-replicating RNA vaccines comprise a replicase RNA molecule derived from a virus belonging to the alphavirus family with a subgenomic promoter that controls replication of the fusion protein or HBV antigen RNA followed by an artificial poly A tail located downstream of the replicase.
In certain embodiments, a further adjuvant can be included in a composition or therapeutic combination of the application, or co-administered with a composition or therapeutic combination of the application. Use of another adjuvant is optional, and can further enhance immune responses when the composition is used for vaccination purposes. Other adjuvants suitable for co-administration or inclusion in compositions in accordance with the application should preferably be ones that are potentially safe, well tolerated and effective in humans. An adjuvant can be a small molecule or antibody including, but not limited to, immune checkpoint inhibitors (e.g., anti-PD1, anti-TIM-3, etc.), toll-like receptor agonists (e.g., TLR7 agonists and/or TLR8 agonists), RIG-1 agonists, IL-15 superagonists (Altor Bioscience), mutant IRF3 and IRF7 genetic adjuvants, STING agonists (Aduro), FLT3L genetic adjuvant, and IL-7-hyFc. For example, adjuvants can e.g., be chosen from among the following anti-HBV agents: HBV DNA polymerase inhibitors; Immunomodulators; Toll-like receptor 7 modulators; Toll-like receptor 8 modulators; Toll-like receptor 3 modulators; Interferon alpha receptor ligands;
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-HBV antisense oligonucleotides; short interfering RNAs (siRNA), more particularly anti- HBV siRNA; Endonuclease modulators; Inhibitors of ribonucleotide reductase; Hepatitis B virus E antigen inhibitors; HBV antibodies targeting the surface antigens of the hepatitis B virus; HBV antibodies; CCR2 chemokine antagonists; Thymosin agonists; Cytokines, such as IL12; Capsid Assembly Modulators, Nucleoprotein inhibitors (HBV core or capsid protein inhibitors); Nucleic Acid Polymers (NAPs); Stimulators of retinoic acid-inducible gene 1; Stimulators of NOD2; Recombinant thymosin alpha-1; Hepatitis B virus replication inhibitors; PI3K inhibitors;
cccDNA inhibitors; immune checkpoint inhibitors, such as PD-L1 inhibitors, PD-1 inhibitors, TIM-3 inhibitors, TIGIT inhibitors, Lag3 inhibitors, CTLA-4 inhibitors; Agonists of co- stimulatory receptors that are expressed on immune cells (more particularly T cells), such as CD27 and CD28; BTK inhibitors; Other drugs for treating HBV; IDO inhibitors; Arginase inhibitors; and KDM5 inhibitors.
In certain embodiments, each of the first and second non-naturally occurring nucleic acid molecules is independently formulated with a lipid nanoparticle (LNP).
The application also provides methods of making compositions and therapeutic combinations of the application. A method of producing a composition or therapeutic combination comprises mixing an isolated polynucleotide encoding an HBV antigen, vector, and/or polypeptide of the application with one or more pharmaceutically acceptable carriers. One of ordinary skill in the art will be familiar with conventional techniques used to prepare such compositions.
Methods of Inducing an Immune Response or Treating an HBV Infection
The application also provides methods of inducing an immune response against hepatitis B virus (HBV) in a subject in need thereof, comprising administering to the subject an
immunogenically effective amount of a composition or immunogenic composition of the application. Any of the compositions and therapeutic combinations of the application described herein can be used in the methods of the application.
As used herein, the term“infection” refers to the invasion of a host by a disease causing agent. A disease causing agent is considered to be“infectious” when it is capable of invading a host, and replicating or propagating within the host. Examples of infectious agents include viruses, e.g., HBV and certain species of adenovirus, prions, bacteria, fungi, protozoa and the like. “HBV infection” specifically refers to invasion of a host organism, such as cells and tissues of the host organism, by HBV.
The phrase“inducing an immune response” when used with reference to the methods described herein encompasses causing a desired immune response or effect in a subject in need thereof against an infection, e.g., an HBV infection.“Inducing an immune response” also encompasses providing a therapeutic immunity for treating against a pathogenic agent, e.g., HBV. As used herein, the term“therapeutic immunity” or“therapeutic immune response” means that the vaccinated subject is able to control an infection with the pathogenic agent against which the vaccination was done, for instance immunity against HBV infection conferred by vaccination with HBV vaccine. In an embodiment,“inducing an immune response” means producing an immunity in a subject in need thereof, e.g., to provide a therapeutic effect against a disease, such as HBV infection. In certain embodiments,“inducing an immune response” refers to causing or improving cellular immunity, e.g., T cell response, against HBV infection. In certain
embodiments,“inducing an immune response” refers to causing or improving a humoral immune response against HBV infection. In certain embodiments,“inducing an immune response” refers to causing or improving a cellular and a humoral immune response against HBV infection.
As used herein, the term“protective immunity” or“protective immune response” means that the vaccinated subject is able to control an infection with the pathogenic agent against which the vaccination was done. Usually, the subject having developed a“protective immune response” develops only mild to moderate clinical symptoms or no symptoms at all. Usually, a subject having a“protective immune response” or“protective immunity” against a certain agent will not die as a result of the infection with said agent.
Typically, the administration of compositions and therapeutic combinations of the application will have a therapeutic aim to generate an immune response against HBV after HBV infection or development of symptoms characteristic of HBV infection, e.g., for therapeutic vaccination.
As used herein,“an immunogenically effective amount” or“immunologically effective amount” means an amount of a composition, polynucleotide, vector, or antigen sufficient to induce a desired immune effect or immune response in a subject in need thereof. An
immunogenically effective amount can be an amount sufficient to induce an immune response in a subject in need thereof. An immunogenically effective amount can be an amount sufficient to produce immunity in a subject in need thereof, e.g., provide a therapeutic effect against a disease such as HBV infection. An immunogenically effective amount can vary depending upon a variety of factors, such as the physical condition of the subject, age, weight, health, etc.; the particular application, e.g., providing protective immunity or therapeutic immunity; and the particular disease, e.g., viral infection, for which immunity is desired. An immunogenically effective amount can readily be determined by one of ordinary skill in the art in view of the present disclosure.
In particular embodiments of the application, an immunogenically 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) prevent the recurrence of an HBV infection or symptom associated therewith; (vii) reduce hospitalization of a subject having an HBV infection; (viii) reduce hospitalization length of a subject having an HBV infection; (ix) increase the survival of a subject with an HBV infection; (x) eliminate an HBV infection in a subject; (xi) inhibit or reduce HBV replication in a subject; and/or (xii) enhance or improve the prophylactic or therapeutic effect(s) of another therapy. An immunogenically effective amount can also be an amount 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 within 12 months. Examples of a target index include lower HBsAg below a threshold of 500 copies of HBsAg international units (IU) and/or higher CD8 counts.
As general guidance, an immunogenically effective amount when used with reference to a DNA plasmid can range from about 0.1 mg/mL to 10 mg/mL of DNA plasmid total, such as 0.1 mg/mL, 0.25 mg/mL, 0.5 mg/mL.0.75 mg/mL 1 mg/mL, 1.5 mg/mL, 2 mg/mL, 3 mg/mL, 4 mg/mL, 5 mg/mL, 6 mg/mL, 7 mg/mL, 8 mg/mL, 9 mg/mL, or 10 mg/mL. Preferably, an immunogenically effective amount of DNA plasmid is less than 8 mg/mL, more preferably less than 6 mg/mL, even more preferably 3-4 mg/mL. An immunogenically effective amount can be from one vector or plasmid, or from multiple vectors or plasmids. As further general guidance, an immunogenically effective amount when used with reference to a peptide can range from about 10 µg to 1 mg per administration, such as 10, 20, 50, 100, 200, 300, 400, 500, 600, 700, 800, 9000, or 1000 µg per administration. An immunogenically effective amount can be administered in a single composition, or in multiple compositions, such as 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 compositions (e.g., tablets, capsules or injectables, or any composition adapted to intradermal delivery, e.g., to intradermal delivery using an intradermal delivery patch), wherein the administration of the multiple capsules or injections collectively provides a subject with an immunogenically effective amount. For example, when two DNA plasmids are used, an immunogenically effective amount can be 3-4 mg/mL, with 1.5-2 mg/mL of each plasmid. It is also possible to administer an immunogenically effective amount to a subject, and subsequently administer another dose of an immunogenically effective amount to the same subject, in a so- called prime-boost regimen. This general concept of a prime-boost regimen is well known to the skilled person in the vaccine field. Further booster administrations can optionally be added to the regimen, as needed.
A therapeutic combination comprising two DNA plasmids, e.g., a first DNA plasmid encoding an HBV core antigen and second DNA plasmid encoding an HBV pol antigen, can be administered to a subject by mixing both plasmids and delivering the mixture to a single anatomic site. Alternatively, two separate immunizations each delivering a single expression plasmid can be performed. In such embodiments, whether both plasmids are administered in a single immunization as a mixture of in two separate immunizations, the first DNA plasmid and the second DNA plasmid can be administered in a ratio of 10:1 to 1:10, by weight, such as 10:1, 9:1, 8:1, 7:1, 6:1, 5:1, 4:1, 3:1, 2:1, 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, or 1:10, by weight.
Preferably, the first and second DNA plasmids are administered in a ratio of 1:1, by weight.
Preferably, a subject to be treated according to the methods of the application is an HBV- infected subject, particular a subject having chronic HBV infection. Acute HBV infection is characterized by an efficient activation of the innate immune system complemented with a subsequent broad adaptive response (e.g., HBV-specific T-cells, neutralizing antibodies), which usually results in successful suppression of replication or removal of infected hepatocytes. In contrast, such responses are impaired or diminished due to high viral and antigen load, e.g., HBV envelope proteins are produced in abundance and can be released in sub-viral particles in 1,000- fold excess to infectious virus.
Chronic HBV infection is described in phases characterized by viral load, liver enzyme levels (necroinflammatory activity), HBeAg, or HBsAg load or presence of antibodies to these antigens. cccDNA levels stay relatively constant at approximately 10 to 50 copies per cell, even though viremia can vary considerably. The persistence of the cccDNA species leads to chronicity. More specifically, the phases of chronic HBV infection include: (i) the immune- tolerant phase characterized by high viral load and normal or minimally elevated liver enzymes; (ii) the immune activation HBeAg-positive phase in which lower or declining levels of viral replication with significantly elevated liver enzymes are observed; (iii) the inactive HBsAg carrier phase, which is a low replicative state with low viral loads and normal liver enzyme levels in the serum that may follow HBeAg seroconversion; and (iv) the HBeAg-negative phase in which viral replication occurs periodically (reactivation) with concomitant fluctuations in liver enzyme levels, mutations in the pre-core and/or basal core promoter are common, such that HBeAg is not produced by the infected cell.
As used herein,“chronic HBV infection” refers to a subject having the detectable presence of HBV for more than 6 months. A subject having a chronic HBV infection can be in any phase of chronic HBV infection. Chronic HBV infection is understood in accordance with its ordinary meaning in the field. Chronic HBV infection can for example be characterized by the persistence of HBsAg for 6 months or more after acute HBV infection. For example, a chronic HBV infection referred to herein follows the definition published by the Centers for Disease Control and Prevention (CDC), according to which a chronic HBV infection can be characterized by laboratory criteria such as: (i) negative for IgM antibodies to hepatitis B core antigen (IgM anti- HBc) and positive for hepatitis B surface antigen (HBsAg), hepatitis B e antigen (HBeAg), or nucleic acid test for hepatitis B virus DNA, or (ii) positive for HBsAg or nucleic acid test for HBV DNA, or positive for HBeAg two times at least 6 months apart.
Preferably, an immunogenically effective amount refers to the amount of a composition or therapeutic combination of the application which is sufficient to treat chronic HBV infection.
In some embodiments, a subject having chronic HBV infection is undergoing nucleoside analog (NUC) treatment, and is NUC-suppressed. As used herein,“NUC-suppressed” refers to a subject having an undetectable viral level of HBV and stable alanine aminotransferase (ALT) levels for at least six months. Examples of nucleoside/nucleotide analog treatment include HBV polymerase inhibitors, such as entacavir and tenofovir. Preferably, a subject having chronic HBV infection does not have advanced hepatic fibrosis or cirrhosis. Such subject would typically have a METAVIR score of less than 3 for fibrosis and a fibroscan result of less than 9 kPa. The METAVIR score is a scoring system that is commonly used to assess the extent of inflammation and fibrosis by histopathological evaluation in a liver biopsy of patients with hepatitis B. The scoring system assigns two standardized numbers: one reflecting the degree of inflammation and one reflecting the degree of fibrosis.
It is believed that elimination or reduction of chronic HBV may allow early disease interception of severe liver disease, including virus-induced cirrhosis and hepatocellular carcinoma. Thus, the methods of the application can also be used as therapy to treat HBV- induced diseases. Examples of HBV-induced diseases include, but are not limited to cirrhosis, cancer (e.g., hepatocellular carcinoma), and fibrosis, particularly advanced fibrosis characterized by a METAVIR score of 3 or higher for fibrosis. In such embodiments, an immunogenically effective amount is an amount sufficient to achieve persistent loss of HBsAg within 12 months and significant decrease in clinical disease (e.g., cirrhosis, hepatocellular carcinoma, etc.).
Methods according to embodiments of the application further comprises administering to the subject in need thereof another immunogenic agent (such as another HBV antigen or other antigen) or another anti-HBV agent (such as a nucleoside analog or other anti-HBV agent) in combination with a composition of the application. For example, another anti-HBV agent or immunogenic agent can be a small molecule or antibody including, but not limited to, immune checkpoint inhibitors (e.g., anti-PD1, anti-TIM-3, etc.), toll-like receptor agonists (e.g., TLR7 agonists and/oror TLR8 agonists), RIG-1 agonists, IL-15 superagonists (Altor Bioscience), mutant IRF3 and IRF7 genetic adjuvants, STING agonists (Aduro), FLT3L genetic adjuvant, IL12 genetic adjuvant, IL-7-hyFc; CAR-T which bind HBV env (S-CAR cells); capsid assembly modulators; cccDNA inhibitors, HBV polymerase inhibitors (e.g., entecavir and tenofovir). The one or other anti-HBV active agents can be, for example, a small molecule, an antibody or antigen binding fragment thereof, a polypeptide, protein, or nucleic acid. The one or other anti-HBV agents can e.g., be chosen from among HBV DNA polymerase inhibitors; Immunomodulators; Toll-like receptor 7 modulators; Toll-like receptor 8 modulators; Toll-like receptor 3 modulators; Interferon alpha receptor ligands; 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-HBV antisense oligonucleotides; short interfering RNAs (siRNA), more particularly anti-HBV siRNA; Endonuclease modulators; Inhibitors of ribonucleotide reductase; Hepatitis B virus E antigen inhibitors; HBV antibodies targeting the surface antigens of the hepatitis B virus; HBV antibodies; CCR2 chemokine antagonists; Thymosin agonists;
Cytokines, such as IL12; Capsid Assembly Modulators, Nucleoprotein inhibitors (HBV core or capsid protein inhibitors); Nucleic Acid Polymers (NAPs); Stimulators of retinoic acid-inducible gene 1; Stimulators of NOD2; Recombinant thymosin alpha-1; Hepatitis B virus 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.
Methods of Delivery
Compositions and therapeutic combinations of the application can be administered to a subject by any method known in the art in view of the present disclosure, including, but not limited to, parenteral administration (e.g., intramuscular, subcutaneous, intravenous, or intradermal injection), oral administration, transdermal administration, and nasal administration. Preferably, compositions and therapeutic combinations are administered parenterally (e.g., by intramuscular injection or intradermal injection) or transdermally.
In some embodiments of the application in which a composition or therapeutic combination comprises one or more DNA plasmids, administration can be by injection through the skin, e.g., intramuscular or intradermal injection, preferably intramuscular injection.
Intramuscular injection can be combined with electroporation, i.e., application of an electric field to facilitate delivery of the DNA plasmids to cells. As used herein, the term“electroporation” refers to the use of a transmembrane electric field pulse to induce microscopic pathways (pores) in a bio-membrane. During in vivo electroporation, electrical fields of appropriate magnitude and duration are applied to cells, inducing a transient state of enhanced cell membrane permeability, thus enabling the cellular uptake of molecules unable to cross cell membranes on their own. Creation of such pores by electroporation facilitates passage of biomolecules, such as plasmids, oligonucleotides, siRNAs, drugs, etc., from one side of a cellular membrane to the other. In vivo electroporation for the delivery of DNA vaccines has been shown to significantly increase plasmid uptake by host cells, while also leading to mild-to-moderate inflammation at the injection site. As a result, transfection efficiency and immune response are significantly improved (e.g., up to 1,000 fold and 100 fold respectively) with intradermal or intramuscular electroporation, in comparison to conventional injection.
In a typical embodiment, electroporation is combined with intramuscular injection.
However, it is also possible to combine electroporation with other forms of parenteral administration, e.g., intradermal injection, subcutaneous injection, etc.
Administration of a composition, therapeutic combination or vaccine of the application via electroporation can be accomplished using electroporation devices that can be configured to deliver to a desired tissue of a mammal a pulse of energy effective to cause reversible pores to form in cell membranes. The electroporation device can include an electroporation component and an electrode assembly or handle assembly. The electroporation component can include one or more of the following components of electroporation devices: controller, current waveform generator, impedance tester, waveform logger, input element, status reporting element, communication port, memory component, power source, and power switch. Electroporation can be accomplished using an in vivo electroporation device. Examples of electroporation devices and electroporation methods that can facilitate delivery of compositions and therapeutic combinations of the application, particularly those comprising DNA plasmids, include
CELLECTRA® (Inovio Pharmaceuticals, Blue Bell, PA), Elgen electroporator (Inovio
Pharmaceuticals, Inc.) Tri-GridTM delivery system (Ichor Medical Systems, Inc., San Diego, CA 92121) and those described in U.S. Patent No.7,664,545, U.S. Patent No.8,209,006, U.S. Patent No.9,452,285, U.S. Patent No.5,273,525, U.S. Patent No.6,110,161, U.S. Patent No. 6,261,281, U.S. Patent No.6,958,060, and U.S. Patent No.6,939,862, U.S. Patent No.7,328,064, U.S. Patent No.6,041,252, U.S. Patent No.5,873,849, U.S. Patent No.6,278,895, U.S. Patent No.6,319,901, U.S. Patent No.6,912,417, U.S. Patent No.8,187,249, U.S. Patent No.9,364,664, U.S. Patent No.9,802,035, U.S. Patent No.6,117,660, and International Patent Application Publication WO2017172838, all of which are herein incorporated by reference in their entireties. Other examples of in vivo electroporation devices are described in International Patent
Application entitled“Method and Apparatus for the Delivery of Hepatitis B Virus (HBV) Vaccines,” filed on the same day as this application with the Attorney Docket Number 688097- 405WO, the contents of which are hereby incorporated by reference in their entireties. Also contemplated by the application for delivery of the compositions and therapeutic combinations of the application are use of a pulsed electric field, for instance as described in, e.g., U.S. Patent No. 6,697,669, which is herein incorporated by reference in its entirety.
In other embodiments of the application in which a composition or therapeutic
combination comprises one or more DNA plasmids, the method of administration is transdermal. Transdermal administration can be combined with epidermal skin abrasion to facilitate delivery of the DNA plasmids to cells. For example, a dermatological patch can be used for epidermal skin abrasion. Upon removal of the dermatological patch, the composition or therapeutic combination can be deposited on the abraised skin.
Methods of delivery are not limited to the above described embodiments, and any means for intracellular delivery can be used. Other methods of intracellular delivery contemplated by the methods of the application include, but are not limited to, liposome encapsulation, lipid nanoparticles (LNPs), etc.
Adjuvants
In some embodiments of the application, a method of inducing an immune response against HBV further comprises administering an adjuvant. The terms“adjuvant” and "immune stimulant" are used interchangeably herein, and are defined as one or more substances that cause stimulation of the immune system. In this context, an adjuvant is used to enhance an immune response to HBV antigens and antigenic HBV polypeptides of the application.
According to embodiments of the application, an adjuvant can be present in a therapeutic combination or composition of the application, or administered in a separate composition. An adjuvant can be, e.g., a small molecule or an antibody. Examples of adjuvants suitable for use in the application include, but are not limited to, immune checkpoint inhibitors (e.g., anti-PD1, anti- TIM-3, etc.), toll-like receptor agonists (e.g., TLR7 and/or TLR8 agonists), RIG-1 agonists, IL- 15 superagonists (Altor Bioscience), mutant IRF3 and IRF7 genetic adjuvants, STING agonists (Aduro), FLT3L genetic adjuvant, IL12 genetic adjuvant, and IL-7-hyFc. Examples of adjuvants can e.g., be chosen from among the following anti-HBV agents: HBV DNA polymerase inhibitors; Immunomodulators; Toll-like receptor 7 modulators; Toll-like receptor 8 modulators; Toll-like receptor 3 modulators; Interferon alpha receptor ligands; 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-HBV antisense oligonucleotides; short interfering RNAs (siRNA), more particularly anti-HBV siRNA; Endonuclease modulators; Inhibitors of ribonucleotide reductase; Hepatitis B virus E antigen inhibitors; HBV antibodies targeting the surface antigens of the hepatitis B virus; HBV antibodies; CCR2 chemokine antagonists; Thymosin agonists; Cytokines, such as IL12; Capsid Assembly Modulators, Nucleoprotein inhibitors (HBV core or capsid protein inhibitors); Nucleic Acid Polymers (NAPs); Stimulators of retinoic acid-inducible gene 1; Stimulators of NOD2; Recombinant thymosin alpha-1; Hepatitis B virus 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.
Compositions and therapeutic combinations of the application can also be administered in combination with at least one other anti-HBV agent. Examples of anti-HBV agents suitable for use with the application include, but are not limited to small molecules, antibodies, and/or CAR- T therapies which bind HBV env (S-CAR cells), capsid assembly modulators, TLR agonists (e.g., TLR7 and/or TLR8 agonists), cccDNA inhibitors, HBV polymerase inhibitors (e.g., entecavir and tenofovir), and/or immune checkpoint inhibitors, etc.
The at least one anti-HBV agent can e.g., be chosen from among HBV DNA polymerase inhibitors; Immunomodulators; Toll-like receptor 7 modulators; Toll-like receptor 8 modulators; Toll-like receptor 3 modulators; Interferon alpha receptor ligands; 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-HBV antisense oligonucleotides; short interfering RNAs (siRNA), more particularly anti-HBV siRNA; Endonuclease modulators; Inhibitors of ribonucleotide reductase; Hepatitis B virus E antigen inhibitors; HBV antibodies targeting the surface antigens of the hepatitis B virus; HBV antibodies; CCR2 chemokine antagonists; Thymosin agonists; Cytokines, such as IL12; Capsid Assembly Modulators, Nucleoprotein inhibitors (HBV core or capsid protein inhibitors); Nucleic Acid Polymers (NAPs); Stimulators of retinoic acid-inducible gene 1; Stimulators of NOD2; Recombinant thymosin alpha-1; Hepatitis B virus 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. Such anti-HBV agents can be administered with the compositions and therapeutic combinations of the application simultaneously or sequentially.
Methods of Prime/Boost Immunization
Embodiments of the application also contemplate administering an immunogenically effective amount of a composition or therapeutic combination to a subject, and subsequently administering another dose of an immunogenically effective amount of a composition or therapeutic combination to the same subject, in a so-called prime-boost regimen Thus, in an embodiment, a composition or therapeutic combination of the application is a primer vaccine used for priming an immune response. In another embodiment, a composition or therapeutic combination of the application is a booster vaccine used for boosting an immune response. The priming and boosting vaccines of the application can be used in the methods of the application described herein. This general concept of a prime-boost regimen is well known to the skilled person in the vaccine field. Any of the compositions and therapeutic combinations of the application described herein can be used as priming and/or boosting vaccines for priming and/or boosting an immune response against HBV.
In some embodiments of the application, a composition or therapeutic combination of the application can be administered for priming immunization. The composition or therapeutic combination can be re-administered for boosting immunization. Further booster administrations of the composition or vaccine combination can optionally be added to the regimen, as needed. An adjuvant can be present in a composition of the application used for boosting immunization, present in a separate composition to be administered together with the composition or therapeutic combination of the application for the boosting immunization, or administered on its own as the boosting immunization. In those embodiments in which an adjuvant is included in the regimen, the adjuvant is preferably used for boosting immunization.
An illustrative and non-limiting example of a prime-boost regimen includes
administering a single dose of an immunogenically effective amount of a composition or therapeutic combination of the application to a subject to prime the immune response; and subsequently administering another dose of an immunogenically effective amount of a composition or therapeutic combination of the application to boost the immune response, wherein the boosting immunization is first administered about two to six weeks, preferably four weeks after the priming immunization is initially administered. Optionally, about 10 to 14 weeks, preferably 12 weeks, after the priming immunization is initially administered, a further boosting immunization of the composition or therapeutic combination, or other adjuvant, is administered.
Kits
Also provided herein is a kit comprising a therapeutic combination of the application. A kit can comprise the first polynucleotide, the second polynucleotide, and the at least one capsid assembly modulator in one or more separate compositions, or a kit can comprise the first polynucleotide, the second polynucleotide, and the capsid assembly modulator in a single composition. A kit can further comprise one or more adjuvants or immune stimulants, and/or other anti-HBV agents.
The ability to induce or stimulate an anti-HBV immune response upon administration in an animal or human organism can be evaluated either in vitro or in vivo using a variety of assays which are standard in the art. For a general description of techniques available to evaluate the onset and activation of an immune response, see for example Coligan et al. (1992 and 1994, Current Protocols in Immunology; ed. J Wiley & Sons Inc, National Institute of Health).
Measurement of cellular immunity can be performed by measurement of cytokine profiles secreted by activated effector cells including those derived from CD4+ and CD8+ T-cells (e.g. quantification of IL-10 or IFN gamma-producing cells by ELISPOT), by determination of the activation status of immune effector cells (e.g. T cell proliferation assays by a classical [3H] thymidine uptake or flow cytometry-based assays), by assaying for antigen-specific T lymphocytes in a sensitized subject (e.g. peptide-specific lysis in a cytotoxicity assay, etc.).
The ability to stimulate a cellular and/or a humoral response can be determined by antibody binding and/or competition in binding (see for example Harlow, 1989, Antibodies, Cold Spring Harbor Press). For example, titers of antibodies produced in response to administration of a composition providing an immunogen can be measured by enzyme-linked immunosorbent assay (ELISA). The immune responses can also be measured by neutralizing antibody assay, where a neutralization of a virus is defined as the loss of infectivity through
reaction/inhibition/neutralization of the virus with specific antibody. The immune response can further be measured by Antibody-Dependent Cellular Phagocytosis (ADCP) Assay. EMBODIMENTS
The invention provides also the following non-limiting embodiments.
Embodiment 1 is a therapeutic combination for use in treating a hepatitis B virus (HBV) infection in a subject in need thereof, comprising:
i) at least one of:
a) a truncated HBV core antigen consisting of an amino acid sequence that is at least 95%, such as at least 95%, 96%, 97%, 98%, 99% or 100%, identical to SEQ ID NO: 2,
b) a first non-naturally occurring nucleic acid molecule comprising a first polynucleotide sequence encoding the truncated HBV core antigen
c) an HBV polymerase antigen having an amino acid sequence that is at least 90%, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%, identical to SEQ ID NO: 7, wherein the HBV polymerase antigen does not have reverse transcriptase activity and RNase H activity, and
d) a second non-naturally occurring nucleic acid molecule comprising a second polynucleotide sequence encoding the HBV polymerase antigen; and
ii) a capsid assembly modulator, such as that described herein.
Embodiment 2 is the therapeutic combination of embodiment 1, comprising at least one of the HBV polymerase antigen and the truncated HBV core antigen.
Embodiment 3 is the therapeutic combination of embodiment 2, comprising the HBV polymerase antigen and the truncated HBV core antigen.
Embodiment 4 is the therapeutic combination of embodiment 1, comprising at least one of the first non-naturally occurring nucleic acid molecule comprising the first polynucleotide sequence encoding the truncated HBV core antigen, and the second non-naturally occurring nucleic acid molecule comprising the second polynucleotide sequence encoding the HBV polymerase antigen.
Embodiment 5 is a therapeutic combination for use in treating a hepatitis B virus (HBV) infection in a subject in need thereof, comprising
i) a first non-naturally occurring nucleic acid molecule comprising a first
polynucleotide sequence encoding a truncated HBV core antigen consisting of an amino acid sequence that is at least 95% identical to SEQ ID NO: 2; and ii) a second non-naturally occurring nucleic acid molecule comprising a second polynucleotide sequence encoding an HBV polymerase antigen having an amino acid sequence that is at least 90% identical to SEQ ID NO: 7, wherein the HBV polymerase antigen does not have reverse transcriptase activity and RNase H activity; and
iii) a compound selected from the group consisting of:
a. a compound of formula (I), (XI), (XVIII), or (XXI), or an enantiomer, diastereomer or pharmaceutically accepted salt, hydrate, or solvate thereof, including a hydrate, a solvate, a pharmaceutically acceptable salt, a prodrug, or a complex thereof described herein;
b. a compound of formula (A-I), (A-II), (A-III), (A-IV), (A-V), (A-VI), or (AVII), or an enantiomer, diastereomer or pharmaceutically accepted salt, hydrate, or solvate thereof, including a hydrate, a solvate, a pharmaceutically acceptable salt, a prodrug, or a complex thereof described herein; and
c. a compound of formula (B-I), or a pharmaceutically salt described herein. Embodiment 6 is the therapeutic combination of embodiment 4 or 5, wherein the first non-naturally occurring nucleic acid molecule further comprises a polynucleotide sequence encoding a signal sequence operably linked to the N-terminus of the truncated HBV core antigen.
Embodiment 6a is the therapeutic combination of any one of embodiments 4 to 6, wherein the second non-naturally occurring nucleic acid molecule further comprises a polynucleotide sequence encoding a signal sequence operably linked to the N-terminus of the HBV polymerase antigen.
Embodiment 6b is the therapeutic combination of embodiment 6 or 6a, wherein the signal sequence independently comprises the amino acid sequence of SEQ ID NO: 9 or SEQ ID NO: 15.
Embodiment 6c is the therapeutic combination of embodiment 6 or 6a, wherein the signal sequence is independently encoded by the polynucleotide sequence of SEQ ID NO: 8 or SEQ ID NO: 14.
Embodiment 7 is the therapeutic combination of any one of embodiments 1-6c, wherein the HBV polymerase antigen comprises an amino acid sequence that is at least 98%, such as at least 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9%, or 100%, identical to SEQ ID NO: 7.
Embodiment 7a is the therapeutic combination of embodiment 7, wherein the HBV polymerase antigen comprises the amino acid sequence of SEQ ID NO: 7.
Embodiment 7b is the therapeutic combination of any one of embodiments 1 to 7a, wherein and the truncated HBV core antigen consists of the amino acid sequence that is at least 98%, such as at least 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9%, or 100%, identical to SEQ ID NO: 2.
Embodiment 7c is the therapeutic combination of embodiment 7b, wherein the truncated HBV antigen consists of the amino acid sequence of SEQ ID NO: 2 or SEQ ID NO: 4. Embodiment 8 is the therapeutic combination of any one of embodiments 1-7c, wherein each of the first and second non-naturally occurring nucleic acid molecules is a DNA molecule.
Embodiment 8a is the therapeutic combination of embodiment 8, wherein the DNA molecule is present on a DNA vector.
Embodiment 8b is the therapeutic combination of embodiment 8a, wherein the DNA vector is selected from the group consisting of DNA plasmids, bacterial artificial chromosomes, yeast artificial chromosomes, and closed linear deoxyribonucleic acid.
Embodiment 8c is the therapeutic combination of embodiment 8, wherein the DNA molecule is present on a viral vector.
Embodiment 8d is the therapeutic combination of embodiment 8c, wherein the viral vector is selected from the group consisting of bacteriophages, animal viruses, and plant viruses.
Embodiment 8e is the therapeutic combination of any one of embodiments 1-7c, wherein each of the first and second non-naturally occurring nucleic acid molecules is an RNA molecule.
Embodiment 8f is the therapeutic combination of embodiment 8e, wherein the RNA molecule is an RNA replicon, preferably a self-replicating RNA replicon, an mRNA replicon, a modified mRNA replicon, or self-amplifying mRNA.
Embodiment 8g is the therapeutic combination of any one of embodiments 1 to 8f, wherein each of the first and second non-naturally occurring nucleic acid molecules is independently formulated with a lipid composition, preferably a lipid nanoparticle (LNP).
Embodiment 9 is the therapeutic combination of any one of embodiments 4-8g, comprising the first non-naturally occurring nucleic acid molecule and the second non-naturally occurring nucleic acid molecule in the same non-naturally occurring nucleic acid molecule.
Embodiment 10 is the therapeutic combination of any one of embodiments 4-8g, comprising the first non-naturally occurring nucleic acid molecule and the second non-naturally occurring nucleic acid molecule in two different non-naturally occurring nucleic acid molecules.
Embodiment 11 is the therapeutic combination of any one of embodiments 4-10, wherein the first polynucleotide sequence comprises a polynucleotide sequence having at least 90%, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%, sequence identity to SEQ ID NO: 1 or SEQ ID NO: 3.
Embodiment 11a is the therapeutic combination of embodiment 11, wherein the first polynucleotide sequence comprises a polynucleotide sequence having at least 98%, such as at least 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9%, or 100%, sequence identity to SEQ ID NO: 1 or SEQ ID NO: 3.
Embodiment 12 is the therapeutic combination of embodiment 11a, wherein the first polynucleotide sequence comprises the polynucleotide sequence of SEQ ID NO: 1 or SEQ ID NO: 3.
Embodiment 13 is the therapeutic combination of any one of embodiments 4 to 12, wherein the second polynucleotide sequence comprises a polynucleotide sequence having at least 90%, such as at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%, sequence identity to SEQ ID NO: 5 or SEQ ID NO: 6.
Embodiment 13a is the therapeutic combination of embodiment 13, wherein the second polynucleotide sequence comprises a polynucleotide sequence having at least 98%, such as at least 98%, 98.5%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9%, or 100%, sequence identity to SEQ ID NO: 5 or SEQ ID NO: 6.
Embodiment 14 is the therapeutic combination of embodiment 13a, wherein the second polynucleotide sequence comprises the polynucleotide sequence of SEQ ID NO: 5 or SEQ ID NO: 6.
Embodiment 15 is the therapeutic combination of any one of embodiments 1 to 14, wherein the compound is selected from the group consisting of the exemplified compounds, or an enantiomer, diastereomer or pharmaceutically accepted salt, hydrate, or solvate thereof, including a hydrate, a solvate, a pharmaceutically acceptable salt, a prodrug, or a complex thereof.
Embodiment 15a is the therapeutic combination of any one of embodiments 1 to 15, wherein the capsid assembly modulator is a compound having formula (I), (XI), (XVIII), or (XXI), as described herein.
Embodiment 15b is the therapeutic combination of any one of embodiments 1 to 15, wherein the capsid assembly modulator is a compound selected from the exemplified compounds having formula (I), (XI), (XVIII), or (XXI), as described herein or in WO 2014106019 and US 20160024004, the content of which is incorporated herein by reference.
Embodiment 15c is the therapeutic combination of any one of embodiments 1 to 15, wherein the capsid assembly modulator is a compound having (A-I), (A-II), (A-III), (A-IV), (A- V), (A-VI), or (AVII), as described herein. Embodiment 15d is the therapeutic combination of any one of embodiments 1 to 15, wherein the capsid assembly modulator is a compound selected from the exemplified compounds having (A-I), (A-II), (A-III), (A-IV), (A-V), (A-VI), or (AVII), as described herein, or in WO 2017156255, the content of which is incorporated herein by reference.
Embodiment 15e is the therapeutic combination of any one of embodiments 1 to 15, wherein the capsid assembly modulator is a compound having formula (B-I) as described herein.
Embodiment 15f is the therapeutic combination of any one of embodiments 1 to 15, wherein the capsid assembly modulator is a compound selected from the exemplified compounds having formula (B-I) as described herein, or in WO 2018039531 and US 20180161307, the content of which is incorporated herein by reference.
Embodiment 16 is a kit comprising the therapeutic combination of any one of embodiments 1 to 15f, and instructions for using the therapeutic combination in treating a hepatitis B virus (HBV) infection in a subject in need thereof.
Embodiment 17 is a method of treating a hepatitis B virus (HBV) infection in a subject in need thereof, comprising administering to the subject the therapeutic combination of any one of embodiments 1 to 15f.
Embodiment 17a is the method of embodiment 17, wherein the treatment induces an immune response against a hepatitis B virus in a subject in need thereof, preferably the subject has chronic HBV infection.
Embodiment 17b is the method of embodiment 17 or 17a, wherein the subject has chronic HBV infection.
Embodiment 17c is the method of any one of embodiments 17 to 17b, wherein the subject is in need of a treatment of an HBV-induced disease selected from the group consisting of advanced fibrosis, cirrhosis and hepatocellular carcinoma (HCC).
Embodiment 18 is the method of any one of embodiments 17-17c, wherein the therapeutic combination is administered by injection through the skin, e.g., intramuscular or intradermal injection, preferably intramuscular injection.
Embodiment 19 is the method of embodiment 18, wherein the therapeutic combination comprises at least one of the first and second non-naturally occurring nucleic acid molecules.
Embodiment 19a is the method of embodiment 19, wherein the therapeutic combination comprises the first and second non-naturally occurring nucleic acid molecules. Embodiment 20 is the method of embodiment 19 or 19a, wherein the non-naturally occurring nucleic acid molecules are administered to the subject by intramuscular injection in combination with electroporation.
Embodiment 21 is the method of embodiment 19 or 19a, wherein the non-naturally occurring nucleic acid molecules are administered to the subject by a lipid composition, preferably by a lipid nanoparticle. EXAMPLES
It will be appreciated by those skilled in the art that changes could be made to the embodiments described herein without departing from the broad inventive concept thereof. It is understood, therefore, that this invention is not limited to the particular embodiments disclosed, but it is intended to cover modifications within the spirit and scope of the present invention as defined by the present description.
Example 1. HBV core plasmid & HBV pol plasmid
A schematic representation of the pDK-pol and pDK-core vectors is shown in Fig.1A and 1B, respectively. An HBV core or pol antigen optimized expression cassette containing a CMV promoter (SEQ ID NO: 18), a splicing enhancer (triple composite sequence) (SEQ ID NO: 10), Cystatin S precursor signal peptide SPCS (NP_0018901.1) (SEQ ID NO: 9), and pol (SEQ ID NO: 5) or core (SEQ ID NO: 2) gene was introduced into a pDK plasmid backbone, using standard molecular biology techniques.
The plasmids were tested in vitro for core and pol antigen expression by Western blot analysis using core and pol specific antibodies, and were shown to provide consistent expression profile for cellular and secreted core and pol antigens (data not shown).
Example 2. Generation of Adenoviral Vectors Expressing a Fusion of Truncated HBV Core Antigen with HBV Pol Antigen
The creation of an adenovirus vector has been designed as a fusion protein expressed from a single open reading frame. Additional configurations for the expression of the two proteins, e.g. using two separate expression cassettes, or using a 2A-like sequence to separate the two sequences, can also be envisaged.
Design of expression cassettes for adenoviral vectors The expression cassettes (diagrammed in FIG.2A and FIG.2B) are comprised of the CMV promoter (SEQ ID NO: 19), an intron (SEQ ID NO:12) (a fragment derived from the human ApoAI gene - GenBank accession X01038 base pairs 295– 523, harboring the ApoAI second intron), followed by the optimized coding sequence– either core alone or the core and polymerase fusion protein preceded by a human immunoglobulin secretion signal coding sequence (SEQ ID NO: 14), and followed by the SV40 polyadenylation signal (SEQ ID NO: 13).
A secretion signal was included because of past experience showing improvement in the manufacturability of some adenoviral vectors harboring secreted transgenes, without influencing the elicited T-cell response (mouse experiments).
The last two residues of the Core protein (VV) and the first two residues of the
Polymerase protein (MP) if fused results in a junction sequence (VVMP) that is present on the human dopamine receptor protein (D3 isoform), along with flanking homologies.
The interjection of an AGAG linker between the core and the polymerase sequences eliminates this homology and returned no further hits in a Blast of the human proteome.
Example 3. In Vivo Immunogenicity Study of DNA Vaccine in Mice
An immunotherapeutic DNA vaccine containing DNA plasmids encoding an HBV core antigen or HBV polymerase antigen was tested in mice. The purpose of the study was designed to detect T-cell responses induced by the vaccine after intramuscular delivery via electroporation into BALB/c mice. Initial immunogenicity studies focused on determining the cellular immune responses that would be elicited by the introduced HBV antigens.
In particular, the plasmids tested included a pDK-Pol plasmid and pDK-Core plasmid, as shown in FIGS.1A and 1B, respectively, and as described above in Example 1. The pDK-Pol plasmid encoded a polymerase antigen having the amino acid sequence of SEQ ID NO: 7, and the pDK-Core plasmid encoding a Core antigen having the amino acid sequence of SEQ ID NO: 2. First, T-cell responses induced by each plasmid individually were tested. The DNA plasmid (pDNA) vaccine was intramuscularly delivered via electroporation to Balb/c mice using a commercially available TriGridTM delivery system-intramuscular (TDS-IM) adapted for application in the mouse model in cranialis tibialis. See International Patent Application Publication WO2017172838, and U.S. Patent Application No.62/607,430, entitled“Method and Apparatus for the Delivery of Hepatitis B Virus (HBV) Vaccines,” filed on December 19, 2017 for additional description on methods and devices for intramuscular delivery of DNA to mice by electroporation, the disclosures of which are hereby incorporated by reference in their entireties. In particular, the TDS-IM array of a TDS-IM v1.0 device having an electrode array with a 2.5 mm spacing between the electrodes and an electrode diameter of 0.030 inch was inserted percutaneously into the selected muscle, with a conductive length of 3.2 mm and an effective penetration depth of 3.2 mm, and with the major axis of the diamond configuration of the electrodes oriented in parallel with the muscle fibers. Following electrode insertion, the injection was initiated to distribute DNA (e.g., 0.020 ml) in the muscle. Following completion of the IM injection, a 250 V/cm electrical field (applied voltage of 59.4 -65.6 V, applied current limits of less than 4 A, 0.16 A/sec) was locally applied for a total duration of about 400 ms at a 10% duty cycle (i.e., voltage is actively applied for a total of about 40 ms of the about 400 ms duration) with 6 total pulses. Once the electroporation procedure was completed, the TriGridTM array was removed and the animals were recovered. High-dose (20 µg) administration to BALB/c mice was performed as summarized in Table 1. Six mice were administered plasmid DNA encoding the HBV core antigen (pDK-core; Group 1), six mice were administered plasmid DNA encoding the HBV pol antigen (pDK-pol; Group 2), and two mice received empty vector as the negative control. Animals received two DNA immunizations two weeks apart and splenocytes were collected one week after the last immunization.
Table 1: Mouse immunization experimental design of the pilot study.
Figure imgf000147_0001
Antigen-specific responses were analyzed and quantified by IFN-g enzyme-linked immunospot (ELISPOT). In this assay, isolated splenocytes of immunized animals were incubated overnight with peptide pools covering the Core protein, the Pol protein, or the small peptide leader and junction sequence (2µg/ml of each peptide). These pools consisted of 15 mer peptides that overlap by 11 residues matching the Genotypes BCD consensus sequence of the Core and Pol vaccine vectors. The large 94 kDan HBV Pol protein was split in the middle into two peptide pools. Antigen-specific T cells were stimulated with the homologous peptide pools and IFN-g-positive T cells were assessed using the ELISPOT assay. IFN-g release by a single antigen-specific T cell was visualized by appropriate antibodies and subsequent chromogenic detection as a colored spot on the microplate referred to as spot-forming cell (SFC).
Substantial T-cell responses against HBV Core were achieved in mice immunized with the DNA vaccine plasmid pDK-Core (Group 1) reaching 1,000 SFCs per 106 cells (FIG.3). Pol T-cell responses towards the Pol 1 peptide pool were strong (~1,000 SFCs per 106 cells). The weak Pol-2-directed anti-Pol cellular responses were likely due to the limited MHC diversity in mice, a phenomenon called T-cell immunodominance defined as unequal recognition of different epitopes from one antigen. A confirmatory study was performed confirming the results obtained in this study (data not shown).
The above results demonstrate that vaccination with a DNA plasmid vaccine encoding HBV antigens induces cellular immune responses against the administered HBV antigens in mice. Similar results were also obtained with non-human primates (data not shown).
It is understood that the examples and embodiments described herein are for illustrative purposes only, and that changes could be made to the embodiments described above without departing from the broad inventive concept thereof. It is understood, therefore, that this invention is not limited to the particular embodiments disclosed, but it is intended to cover modifications within the spirit and scope of the invention as defined by the appended claims.

Claims

CLAIMS It is claimed:
1. A therapeutic combination for use in treating a hepatitis B virus (HBV) infection in a subject in need thereof, comprising:
i) at least one of:
a) a truncated HBV core antigen consisting of an amino acid sequence that is at least 95% identical to SEQ ID NO: 2, and
b) a first non-naturally occurring nucleic acid molecule comprising a first polynucleotide sequence encoding the truncated HBV core antigen. c) an HBV polymerase antigen having an amino acid sequence that is at least 90% identical to SEQ ID NO: 7, wherein the HBV polymerase antigen does not have reverse transcriptase activity and RNase H activity, and
d) a second non-naturally occurring nucleic acid molecule comprising a second polynucleotide sequence encoding the HBV polymerase antigen; and ii) a capsid assembly modulator selected from the group consisting of:
i. a compound of formula (I), (XI), (XVIII), or (XXI), or an
enantiomer, diastereomer or pharmaceutically accepted salt, hydrate, or solvate thereof, including a hydrate, a solvate, a pharmaceutically acceptable salt, a prodrug, or a complex thereof described herein;
ii. a compound of formula (A-I), (A-II), (A-III), (A-IV), (A-V), (A- VI), or (AVII), or an enantiomer, diastereomer or pharmaceutically accepted salt, hydrate, or solvate thereof, including a hydrate, a solvate, a pharmaceutically acceptable salt, a prodrug, or a complex thereof described herein; and
iii. a compound of formula of formula (B-I), or a pharmaceutical salt described herein.
2. The therapeutic combination of claim 1, comprising at least one of the HBV polymerase antigen and the truncated HBV core antigen.
3. The therapeutic combination of claim 2, comprising the HBV polymerase antigen and the truncated HBV core antigen.
4. The therapeutic combination of claim 1, comprising at least one of the first non-naturally occurring nucleic acid molecule comprising the first polynucleotide sequence encoding the truncated HBV core antigen and the second non-naturally occurring nucleic acid molecule comprising the second polynucleotide sequence encoding the HBV polymerase antigen.
5. A therapeutic combination for use in treating a hepatitis B virus (HBV) infection in a subject in need thereof, comprising
i) a first non-naturally occurring nucleic acid molecule comprising a first
polynucleotide sequence encoding a truncated HBV core antigen consisting of an amino acid sequence that is at least 95% identical to SEQ ID NO: 2; and ii) a second non-naturally occurring nucleic acid molecule comprising a second polynucleotide sequence encoding an HBV polymerase antigen having an amino acid sequence that is at least 90% identical to SEQ ID NO: 7, wherein the HBV polymerase antigen does not have reverse transcriptase activity and RNase H activity; and
iii) a compound selected from the group consisting of:
a. a compound of formula (I), (XI), (XVIII), or (XXI), or an enantiomer, diastereomer or pharmaceutically accepted salt, hydrate, or solvate thereof, including a hydrate, a solvate, a pharmaceutically acceptable salt, a prodrug, or a complex thereof;
b. a compound of formula (A-I), (A-II), (A-III), (A-IV), (A-V), (A-VI), or (AVII), or an enantiomer, diastereomer or pharmaceutically accepted salt, hydrate, or solvate thereof, including a hydrate, a solvate, a
pharmaceutically acceptable salt, a prodrug, or a complex thereof; and c. a compound of formula (B-I), or a pharmaceutically salt thereof.
6. The therapeutic combination of claim 4 or 5, wherein the first non-naturally occurring nucleic acid molecule further comprises a polynucleotide sequence encoding a signal sequence operably linked to the N-terminus of the truncated HBV core antigen, and the second non-naturally occurring nucleic acid molecule further comprises a polynucleotide sequence encoding a signal sequence operably linked to the N-terminus of the HBV polymerase antigen, preferably, the signal sequence independently comprises the amino acid sequence of SEQ ID NO: 9 or SEQ ID NO: 15, preferably the signal sequence is independently encoded by the polynucleotide sequence of SEQ ID NO: 8 or SEQ ID NO: 14.
7. The therapeutic combination of any one of claims 1-6, wherein
a) the truncated HBV core antigen consists of the amino acid sequence of SEQ ID NO: 2 or SEQ ID NO: 4; and
b) the HBV polymerase antigen comprises the amino acid sequence of SEQ ID NO: 7.
8. The therapeutic combination of any one of claims 1-7, wherein each of the first, and second non-naturally occurring nucleic acid molecules is a DNA molecule, preferably the DNA molecule is present on a plasmid or a viral vector.
9. The therapeutic combination of any one of claims 4 to 8, comprising the first non- naturally occurring nucleic acid molecule and the second non-naturally occurring nucleic acid molecule in the same non-naturally nucleic acid molecule.
10. The therapeutic combination of any one of claims 4 to 8, comprising the first non- naturally occurring nucleic acid molecule and the second non-naturally occurring nucleic acid molecule in two different non-naturally occurring nucleic acid molecules.
11. The therapeutic combination of any one of claims 4 to 10, wherein the first
polynucleotide sequence comprises a polynucleotide sequence having at least 90% sequence identity to SEQ ID NO: 1 or SEQ ID NO: 3.
12. The therapeutic combination of claim 11, wherein the first polynucleotide sequence
comprises the polynucleotide sequence of SEQ ID NO: 1 or SEQ ID NO: 3.
13. The therapeutic combination of any one of claims 4 to 12, wherein the second
polynucleotide sequence comprises a polynucleotide sequence having at least 90% sequence identity to SEQ ID NO: 5 or SEQ ID NO: 6.
14. The therapeutic combination of claim 13, wherein the second polynucleotide sequence comprises the polynucleotide sequence of SEQ ID NO: 5 or SEQ ID NO: 6.
15. The therapeutic combination of any one of claims 1-14, wherein the capsid assembly modulator is a compound of formula (I), (XI), (XVIII), or (XXI), or an enantiomer, diastereomer or pharmaceutically accepted salt, hydrate, or solvate thereof, including a hydrate, a solvate, a pharmaceutically acceptable salt, a prodrug, or a complex thereof, wherein
i) the compound of formula (I) is
Figure imgf000152_0001
wherein
A is selected from a group consisting of SO2 and CO;
R1 is selected from a group consisting of optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted aryl, and optionally substituted benzyl; R1 may also alternatively or additionally optionally include optionally substituted C1-6 haloalkyl, optionally substituted 3-7 membered cycloheteroalkyl, optionally substituted C2-8 alkenyl, optionally substituted C2-8 alkynyl, optionally substituted C1-6 alkoxy, optionally substituted amine, optionally substituted amidine, optionally substituted carboxyamine, optionally substituted carboxy- C1-6 -alkoxide, -SO2- C1-6 alkyl, optionally substituted heterocyclic, or optionally substituted heteroaryl;
R2 is selected from a group consisting of hydrogen and optionally substituted C1-6 linear alkyl; R2 may also alternatively or additionally optionally include optionally substituted C3-7 cycloalkyl or optionally substituted heterocyclic; or
R1 and R2 are taken together with the atoms to which they are bound to form an optionally substituted heterocycle (including bicyclic or adamantyl structures) with 3 to 10 atoms; and
R3 is selected from a group consisting of optionally substituted aryl, optionally substituted benzyl, optionally substituted alkylaryl, optionally substituted heteroaryl, and optionally substituted alkylheteroaryl; in some embodiments, R3 may also comprise an optionally substituted C1-6 linear alkyl;
R4 is selected from a group consisting of hydrogen and optionally substituted C1-6 linear alkyl; R5 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, optionally substituted C1-6 haloalkyl, and OR9; R5 may also alternatively or additionally optionally include cyano or N(R9)2;
R6 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, optionally substituted C1-6 haloalkyl, and OR9; R6 may also alternatively or additionally optionally include cyano or N(R9)2; or
R4 and R6 are taken together with the atoms to which they are bound to form an optionally substituted carbocyclic or heterocyclic ring with 5 to 6 atoms, optionally containing a carbonyl, optionally containing two carbonyls; and
R7 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, and OR9; R7 may also alternatively or additionally optionally include cyano or N(R9)2;
R8 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, and OR9; R8 may also alternatively or additionally optionally include cyano or N(R9)2; or
R2 and R8 are taken together with the atoms to which they are bound to form an optionally substituted carbocyclic or heterocyclic ring with 5 to 6 atoms; and
R9 is independently at each occurrence selected from a group consisting of hydrogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, and optionally substituted cycloalkyl; R9 may also alternatively or additionally optionally include independently at each occurrence optionally substituted aryl, optionally substituted benzyl, optionally substituted heterocyclyl, or optionally substituted heteroaryl;
provided that when A is SO2; R4 and R6 taken together with the atoms to which they are bound do not to form an optionally substituted ring; and R2 and R8 taken together with the atoms to which they are bound do not to form an optionally substituted ring, then none of the following (a) through (d) apply:
(a) R3 is an optionally substituted phenyl and R1 or R2 , either individually or when taken together, contain a hydroxyl group, or (b) R3 is an optionally substituted alkyl or phenyl, and N(R1)(R2) is an optionally
substituted piperazine or
Figure imgf000154_0001
Figure imgf000154_0002
(c) R3 is optionally substituted alkyl, aryl, or alkaryl and N(R1)(R2) is
Figure imgf000155_0001
(d) either R3 or R4 is an unsubstituted or monosubstituted aryl, or an unsubstituted or monosubstituted aralkyl, or unsubstituted or monosubstituted heteroaryl and R1 and R2 are taken together with the atoms to which they are bound to form an optionally substituted heterocyclic ring structure with 6 to 12 atoms; or
provided that the compound is not 3- {[(dicyclopropylmethyl)amino]sulfonyl}-N-(4- isopropoxyphenyl)benzamide; or 3-({[2-(lH-benzimidazol-2-yl)propyl]amino}sulfonyl)- N-(4- isopropoxyphenyl)benzamide; or 3-[(cyclohexylamino)sulfonyl]-N-(4- isopropylphenyl)benzamide; or 3-(anilinosulfonyl)-N-(4-isopropylphenyl)benzamide; or 5- {[(3- {[(4- methoxyphenyl)amino]carbonyl}phenyl)sulfonyl]amino}pentanoic acid; or 3-[(tert- butylamino)sulfonyl]-N-(4-methoxyphenyl)benzamide; or (3S)- l-[(3- {[(5- isopropoxypyridin-2- yl)amino]carbonyl}phenyl)sulfonyl]piperidine-3-carboxamide; or (3R)-l-[(3- {[(5-isopropoxypyridin- 2-yl)amino]carbonyl}phenyl)sulfonyl]piperidine-3- carboxamide; or 3-(piperidin- l-ylsulfonyl)-N- [(1 S)- 1 ,2,3,4-tetrahydronaphthalen- 1 - yljbenzamide; or N-(5-bromo-3-methoxypyridin-2-yl)-3- (piperidin- 1 - ylsulfonyl)benzamide; or N-(3-methoxy-5-phenylpyridin-2-yl)-3-(pyrrolidin- 1 - ylsulfonyl)benzamide; or N-(3-methoxy-5-phenoxypyridin-2-yl)-3-(pyrrolidin- 1 - ylsulfonyl)benzamide; or N-[3-methoxy-5-(phenylthio)pyridin-2-yl]-3-(pyrrolidin- 1 - ylsulfonyl)benzamide; or N-(5-ethyl-3-methoxypyridin-2-yl)-3-(piperidin- 1 - ylsulfonyl)benzamide; or N-(3-methoxy-5-vinylpyridin-2-yl)-3-(piperidin-1- ylsulfonyl)benzamide; or
Figure imgf000156_0001
;
ii) the compound of formula (XI) is:
Figure imgf000156_0002
wherein
A is selected from a group consisting of SO2 and CO;
R1 is selected from a group consisting of optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted benzyl; R1 may also alternatively or additionally optionally include optionally substituted C1-6 haloalkyl, optionally substituted 3-7 membered cycloheteroalkyl, optionally substituted C2-8 alkenyl, optionally substituted C2-8 alkynyl, optionally substituted C1-6 alkoxy, optionally substituted amine, optionally substituted amidine, optionally substituted carboxyamine, optionally substituted carboxy- C1-6 -alkoxide, -SO2- C1-6 alkyl, optionally substituted heterocyclic, or optionally substituted heteroaryl;
R3 is selected from a group consisting of optionally substituted aryl, optionally substituted benzyl, optionally substituted alkylaryl, optionally substituted heteroaryl, and optionally substituted alkylheteroaryl; in some embodiments, R3 may also comprise an optionally substituted C1-6 linear alkyl;
R4 is selected from a group consisting of hydrogen and optionally substituted C1-6 linear alkyl;
R5 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, optionally substituted C1-6 haloalkyl, and OR9; R5 may also alternatively or additionally optionally include cyano or N(R9)2;
R6 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C
Figure imgf000157_0001
6 branched alkyl, optionally substituted C1
Figure imgf000157_0002
haloalkyl, and OR9; R6 may also alternatively or additionally optionally include cyano or N(R9)2; or
R4 and R6 are taken together with the atoms to which they are bound to form an optionally substituted carbocyclic or heterocyclic ring with 5 to 6 atoms, optionally containing a carbonyl, optionally containing two carbonyls; and
R7 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, optionally substituted C1-6 haloalkyl, and OR9; R7 may also alternatively or additionally optionally include cyano or N(R9)2;
R8 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, optionally substituted C1-6 haloalkyl, and OR9; R8 may also alternatively or additionally optionally include cyano or N(R9)2;
R9 is independently at each occurrence selected from a group consisting of hydrogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, and optionally substituted cycloalkyl; R9 may also alternatively or additionally optionally include independently at each occurrence optionally substituted aryl, optionally substituted benzyl, optionally substituted heterocyclyl, or optionally substituted heteroaryl; and
R11 is selected from a group consisting of hydrogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, and optionally substituted cycloalkyl; iii) the compound of formula (XVIII) is
Figure imgf000158_0001
or an enantiomer, diastereomer or pharmaceutically accepted salt, hydrate, or solvate thereof, including a hydrate, a solvate, a
pharmaceutically acceptable salt, a prodrug, or a complex thereof,
wherein
A is selected from a group consisting of SO2 and CO;
R1 is selected from a group consisting of optionally substituted C1-6 linear alkyl, optionally substituted C
Figure imgf000158_0002
branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted aryl, and optionally substituted benzyl; R1 may also alternatively or additionally optionally include optionally substituted 3-7 membered cycloheteroalkyl, optionally substituted C2-8 alkenyl, optionally substituted C2-8 alkynyl, optionally substituted C1-6 alkoxy, optionally substituted amine, optionally substituted amidine, optionally substituted carboxyamine, optionally substituted carboxy- C1-6 -alkoxide, - SO2- C1-6 alkyl, optionally substituted heterocyclic, or optionally substituted heteroaryl; R5 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, optionally substituted C1-6 haloalkyl, and OR9; R5 may also alternatively or additionally optionally include cyano or N(R9)2;
R6 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, and OR9; R6 may also
alternatively or additionally optionally cyano or N(R9)2; R7 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, and OR9; R7 may also alternatively or additionally optionally include cyano or N(R9)2; R8 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, and OR9; R8 may also alternatively or additionally optionally include cyano or N(R9)2;
R9 is independently at each occurrence selected from a group consisting of hydrogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, and optionally substituted C1-6; R9 may also alternatively or additionally optionally include independently at each occurrence optionally substituted aryl, optionally substituted benzyl, optionally substituted heterocyclyl, or optionally substituted heteroaryl;
R11 is selected from a group consisting of hydrogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, and optionally substituted C3-7 cycloalkyl;
R12 is selected from a group consisting of hydrogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, and optionally substituted C3-7 cycloalkyl; and
R13 is selected from a group consisting of optionally substituted aryl, optionally substituted benzyl, optionally substituted alkylaryl, optionally substituted heteroaryl, and optionally substituted alkylheteroaryl; and iv) the compound of formula (XXI) is
Figure imgf000159_0001
,
wherein
R1 is selected from a group consisting of optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted aryl, and optionally substituted benzyl; R1 may also alternatively or additionally optionally include optionally substituted 3-7 membered cycloheteroalkyl, optionally substituted C2-8 alkenyl, optionally substituted C2-8 alkynyl, optionally substituted C1-6 alkoxy, optionally substituted amine, optionally substituted amidine, optionally substituted carboxyamine, optionally substituted carboxy- C1-6 -alkoxide, - SO2- C1-6 alkyl, optionally substituted heterocyclic, or optionally substituted heteroaryl; R2 is selected from a group consisting of hydrogen and optionally substituted C1-6 linear alkyl; R2 may also alternatively or additionally optionally include optionally substituted C3-7 cycloalkyl or optionally substituted heterocyclic; or
R1 and R2 are taken together with the atoms to which they are bound to form an optionally substituted heterocycle (including bicyclic or adamantyl structures) with 3 to 10 atoms;
R5 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, and OR9; R5 may also
alternatively or additionally optionally include cyano or N(R9)2;
R6 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, and OR9; R6 may also
alternatively or additionally optionally include cyano or N(R9)2;
R7 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, and OR9; R7 may also
alternatively or additionally optionally include cyano or N(R9)2;
R8 is selected from a group consisting of hydrogen, halogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, and OR9; R8 may also
alternatively or additionally optionally include cyano or N(R9)2;
R9 is independently at each occurrence selected from a group consisting of hydrogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, and optionally substituted cycloalkyl, and optionally substituted C1-6 haloalkyl; R9 may also alternatively or additionally optionally include independently at each occurrence optionally substituted aryl, optionally substituted benzyl, optionally substituted heterocyclyl, or optionally substituted heteroaryl; R12 is selected from a group consisting of hydrogen, optionally substituted C1-6 linear alkyl, optionally substituted C1-6 branched alkyl, and optionally substituted cycloalkyl; and
R13 is selected from a group consisting of optionally substituted aryl, optionally substituted benzyl, optionally substituted alkylaryl, optionally substituted heteroaryl, and optionally substituted alkylheteroaryl;
16. The therapeutic combination of any one of claims 1-14, wherein the compound is
selected from the group consisting of:
Figure imgf000161_0001
Figure imgf000161_0002
, or a pharmaceutically acceptable salt thereof.
17. The therapeutic combination of any one of claims 1-14, wherein the capsid assembly modulator is a compound of formula (A-I), (A-II), (A-III), (A-IV), (A-V), (A-VI), or (AVII), or an enantiomer, diastereomer or pharmaceutically accepted salt, hydrate, or solvate thereof, including a hydrate, a solvate, a pharmaceutically acceptable salt, a prodrug, or a complex thereof, wherein i) the compound of formula (A-I) is
Figure imgf000162_0001
wherein
A is phenyl, a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms, a five-membered heteroaromatic ring containing one, two, or three heteroatoms which are, independently, N, O, or S; a C4-14 bicyclic ring, alkylheteroaryl, or alkylaryl; B is a six or seven-membered ring or a six or seven-membered bridged or spiro-fused ring containing zero, one, or two heteroatoms, which are, independently, N, O, or S, a five-membered ring containing zero, one, or two heteroatoms, which are, independently, N, O, or S; a four-membered ring containing zero, one, or two heteroatoms, which are, independently, N, O, or S, or a C5-14 bicyclic ring,
When R1 and R1’ are attached to a carbon, they are, independently, hydrogen, halogen (including F, Cl, Br, and I), SF5, CF3, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C1- 6 alkoxy, C1-6 haloalkoxy, C2-6 alkenyl, cyano, C2-6 alkynyl, C3-6 alkoxyalkyl,
alkoxycarbonyl, alkoxycarbonylalkyl, C1-6 alkyl, arylalkoxycarbonyl, carboxy, C1-6 haloalkyl, heterocyclylalkyl, or C1-6 hydroxyalkyl;
When R1 and R1’ are attached to a nitrogen, they are, independently, hydrogen, C2-6 alkoxy, C3-6 alkoxyalkyl, C2-6 alkenyl, alkoxycarbonyl, carbonylalkyl, carbonyl aryl, C1-6 alkyl, heterocyclylalkyl, C2-6 hydroxyalkyl, or S(O)2R’;
Each R’ is independently H, C1-6 alkyl, C1-6 haloalkyl, C1-6 alkoxy, C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, aryl, heteroaryl, alkylaryl, or arylalkyl, or if two R’ reside on the same nitrogen atom, they can come together to form a C3-6 ring optionally containing a N, O, or S heteroatom;
The R’ groups can optionally be substituted with one or more substituents, which substituents are, independently, halo, C1-6 haloalkyl, C1-6 hydroxyalkyl, hydroxyl, carboxyl, acyl, aryl, acyloxy, amino, amido, carboxyl derivatives, alkylamino, dialkylamino, arylamino, alkoxy, alkoxyalkyl, aryloxy, nitro, cyano, sulfonic acid, thiol, imine, sulfonyl, sulfanyl, sulfinyl, sulfamonyl, ester, carboxylic acid, amide, phosphonyl, phosphinyl, phosphoryl, phosphine, thioester, thioether, acid halide, anhydride, oxime, hydrozine, carbamate, phosphonic acid, or phosphonate, either unprotected, or protected as necessary, as known to those skilled in the art, for example, as taught in Greene, et al., Protective Groups in Organic Synthesis, John Wiley and Sons, Second Edition, 1991, hereby incorporated by reference;
u and v are independently 0, 1, 2, 3, 4 or 5;
X is
Figure imgf000163_0001
R3 is H, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, or C2-6 alkynyl;
R2 is C1-6 alkyl, C1-6 haloalkyl, C2-8 alkoxyalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, such as phenyl, heteroaryl, including six-membered heteroaromatic rings containing one, two, or three nitrogen atoms and five-membered heteroaromatic rings containing one, two, or three heteroatoms, which, independently, are N, O, or S, alkylaryl, arylalkyl, a six- membered ring or a six-membered bridged or spiro-fused ring containing zero, one, or two heteroatoms which are, independently, N, O, or S, a seven-membered bridged or spiro-fused ring containing zero, one, or two heteroatoms which are, independently, N, O, or S, a five-membered ring containing zero, one, or two heteroatoms which are, independently, N, O, or S; a four-membered ring containing zero, one, or two
heteroatoms which are, independently, N, O, or S; cycloalkyl, alkylheteroaryl, or alkylaryl;
R2 is optionally substituted with one or more substituents, which each, independently, are halogen (including F, Cl, Br, and I), CF3, SF5, hydroxy, N(R’)S(O)2R’, S(O)2R’,
S(O)2N(R’)2, C1-6 alkoxy, C1-6 haloalkoxy, cyano, azido, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C1-6 alkyl, arylalkoxycarbonyl, carboxy, C1-6 haloalkyl, heterocyclylalkyl, or C1-6 hydroxyalkyl; or is optionally substituted with aryl, substituted aryl, heteroaryl, or substituted heteroaryl, where substituents on the substituted aryl and substituted heteroaryl are selected from the group consisting of halogen, SF5, CF3, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C(O)R’, C1-6 alkoxy, cyano, azido, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, and C1-6 alkyl, or R2 and R3 can come together with the nitrogen to which they are attached to form a 6- 10 membered bicyclic or bridged ring, a 3 to 8 saturated ring, or a 5 membered unsaturated ring; such bicyclic, bridged, saturated and unsaturated rings optionally containing one or more additional heteroatoms, where each is, independently, O, S or N, and optionally being substituted with one or more substituents, wherein each, independently, is halogen (including F, Cl, Br, I), CF3, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C1-6 alkoxy, cyano, azido, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C1-6 alkyl, arylalkoxycarbonyl, carboxy, C1-6 haloalkyl, heterocyclylalkyl, or C1-6 hydroxyalkyl; ii) the compound of formula (A-II) is
Figure imgf000164_0001
wherein
R1 and R1’ are as defined with respect to formula (A-I);
u and v are independently 0, 1, 2, 3, 4 or 5;
C is phenyl, a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms, a five-membered heteroaromatic ring containing one, two, or three heteroatoms which are, independently, N, O, or S; a C4-14 bicyclic ring, alkylaryl, or alkylheteroaryl; D is phenyl, a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms, a five-membered heteroaromatic ring containing one, two, or three heteroatoms, which are, independently, N, O, or S, or a C5-14 bicyclic ring,
Figure imgf000164_0002
;
R4 is H or C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl;
R5 is alkylaryl, arylalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, such as phenyl, heteroaryl, including six-membered heteroaromatic rings containing one, two, or three nitrogen atoms and five-membered heteroaromatic rings containing one, two, or three
heteroatoms, which, independently, are N, O, or S; and a six-membered bridged or spiro- fused ring containing zero, one, or two heteroatoms which are, independently, N, O, or S; R5 is optionally substituted with one or more substituents, each of which is,
independently, halogen (including F, Cl, Br, and I), CF3, SF5, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C1-6 alkoxy, C1-6 haloalkoxy, cyano, azido, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C1-6 alkyl, cycloalkyl,
arylalkoxycarbonyl, carboxy, C1-6 haloalkyl, heterocyclylalkyl, or C1-6 hydroxyalkyl; or is substituted with aryl, substituted aryl, heteroaryl, or substituted heteroaryl, where substituents on the substituted aryl and substituted heteroaryl are selected from the group consisting of halogen, SF5, CF3, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C(O)R’, C1-6 alkoxy, cyano, azido, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl,
alkoxycarbonylalkyl, and C1-6 alkyl; iii) the compound of formula (A-III) is
Figure imgf000165_0001
wherein
R1 and R1’ are as defined with respect to formula (A-I);
u and v are independently 0, 1, 2, 3, 4 or 5;
E is a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms, a five-membered heteroaromatic ring containing one, two, or three heteroatoms, where each is, independently, N, O, or S; a C4-14 bicyclic ring, alkylheteroaryl, or alkylaryl; F is a five-membered heteroaromatic ring containing one, two, or three heteroatoms which are, independently, N, O, or S, or a C4-14 bicyclic ring;
Figure imgf000165_0002
R8 is H, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, or C2-6 alkynyl;
R9 is C1-6 alkyl, C1-6 haloalkyl, C2-8 alkoxyalkyl, a six-membered ring or a six- membered bridged or spiro-fused ring containing zero, one, or two heteroatoms, which are independently N, O, or S, a seven-membered bridged or spiro-fused ring containing zero, one, or two heteroatoms, which are, independently, N, O, or S, a five- membered ring containing zero, one, or two heteroatoms, which are, independently, N, O, or S; a four- membered ring containing zero, one, or two heteroatoms, which are, independently, N, O, or S, or a three membered ring;
R9 is optionally substituted with one or more substituents, each of which is independently halogen (including F, Cl, Br, and I), CF3, SF5, hydroxy, N(R’)S(O)2R’, S(O)2R’,
S(O)2N(R’)2, C1-6 alkoxy, C1-6 haloalkoxy, cyano, azido, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C1-6 alkyl, cycloalkyl, arylalkoxycarbonyl, carboxy, C1-6 haloalkyl, heterocyclylalkyl, or C1-6 hydroxyalkyl; or is substituted with aryl, substituted aryl, heteroaryl, or substituted heteroaryl, where substituents on the substituted aryl and substituted heteroaryl are selected from the group consisting of halogen, SF5, CF3, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C(O)R’, C1-6 alkoxy, cyano, azido, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, and C1-6 alkyl;
or R8 and R9 can come together with the nitrogen to which they are attached to form a 6- 10 membered bicyclic or bridged ring or a 3 to 8 saturated ring; such bicyclic, bridged and saturated ring moiety optionally containing one or more additional heteroatoms which, independently, are O, S or N and optionally being substituted with one or more substituents, each, independently, is halogen (including F, Cl, Br, and I), CF3, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C1-6 alkoxy, cyano, azido, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C1-6 alkyl, arylalkoxycarbonyl, carboxy, C1-6 haloalkyl, heterocyclylalkyl, or C1-6 hydroxyalkyl; iv) the compound of formula (A-IV) is
Figure imgf000167_0001
wherein
G is phenyl, a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms, a five-membered heteroaromatic ring containing one, two, or three heteroatoms, which are, independently, N, O, or S; a C4-14 bicyclic ring, alkylheteroaryl, or alkylaryl; H is phenyl, a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms, a six-membered non-aromatic ring optionally containing one, two, or three heteroatoms, which are, independently, N, O, or S; or a C4-14 bicyclic ring;
When R1 and R1’ are attached to a carbon they are, independently, hydrogen, halogen (including F, Cl, Br, and I), CF3, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C1-6 alkoxy, cyano, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C1-6 alkyl, arylalkoxycarbonyl, carboxy, C1-6 haloalkyl, heterocyclylalkyl, or C1-6
hydroxyalkyl;
When R1 and R1’ are attached to a nitrogen they are, independently, hydrogen, C1-6 alkoxy, C3-6 alkoxyalkyl, alkoxycarbonyl, carbonylalkyl, carbonyl aryl, C1-6 alkyl, C2-6 alkynyl, C2-6 alkenyl, heterocyclylalkyl, C1-6 hydroxyalkyl, or S(O)2R’;
Each R’ is independently H, C1-6 alkyl, C1-6 haloalkyl, C1-6 alkoxy, C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, aryl, heteroaryl, alkylaryl, or arylalkyl, or if two R’ reside on the same nitrogen atom they can come together to form a C3-6 alkyl ring optionally containing a N, O, or S; wherein the R’ groups can be substituted with one or more substituents as defined above, for example, C1-6 hydroxyalkyl, aminoalkyl, and alkoxyalkyl;
u and v are independently 0, 1, 2, 3, 4 or 5;
W is
Figure imgf000167_0002
;
R10 is H, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, or C2-6 alkynyl; R11 is C1-6 alkyl, C1-6 haloalkyl, C2-8 alkoxyalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, heteroaryl, alkylaryl, arylalkyl, phenyl, a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms, a six-membered ring or a six-membered bridged or spiro-fused ring containing zero, one, or two heteroatoms, which are, independently, N, O, or S, a seven-membered bridged or spiro-fused ring containing zero, one, or two heteroatoms, which are, independently, N, O, or S, a five-membered heteroaromatic ring containing one, two, or three heteroatoms, which are, independently, N, O, or S, a five- membered ring containing zero, one, or two heteroatoms, which are, independently, N, O, or S; a four-membered ring containing zero, one, or two heteroatoms, which are, independently, N, O, or S; a three membered ring, alkylheteroaryl, or alkylaryl;
wherein R11 is optionally substituted with one or more substituents selected from the group consisting of halogen (including F, Cl, Br, and I), SF5, CF3, hydroxy,
N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C1-6 alkoxy, C1-6 haloalkoxy, C2-6 alkenyl, cyano, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C1-6 alkyl, arylalkoxycarbonyl, carboxy, C1-6 haloalkyl, heterocyclylalkyl, C1-6 hydroxyalkyl, aryl, substituted aryl, heteroaryl, and substituted heteroaryl, where substituents on the substituted aryl and substituted heteroaryl are selected from the group consisting of halogen, SF5, CF3, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C(O)R’, C
Figure imgf000168_0001
lkoxy, cyano, azido, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, and C1-6 alkyl;
or R10 and R11 can come together with the nitrogen to which they are attached to form a 6-10 membered bicyclic or bridged ring or a 3 to 8 saturated ring; such bicyclic, bridged or saturated ring moiety optionally containing one or more additional heteroatoms, which are each, independently, O, S or N, and optionally substituted with one or more substituents, each of which is, independently, halogen (including F, Cl, Br, and I), CF3, SF5, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C1-6 alkoxy, cyano, azido, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C1- 6 alkyl,
arylalkoxycarbonyl, carboxy, C1-6 haloalkyl, heterocyclylalkyl, or C1-6 hydroxyalkyl; v) the compound of formula (A-V) is
Figure imgf000169_0001
wherein
R1 and R1’ are as defined with respect to formula (A-I);
u and v are independently 0, 1, 2, 3, 4 or 5;
I is phenyl, a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms, a five-membered heteroaromatic ring containing one, two, or three heteroatoms, which are, independently, N, O, or S, a C4-14 bicyclic ring; alkylheteroaryl, or alkylaryl; J is a five-membered heteroaromatic ring containing one, two, or three heteroatoms, which are, independently, N, O, or S, a six or seven-membered ring or a six or seven- membered bridged or spiro-fused ring containing zero, one, or two heteroatoms which are, independently, N, O, or S, a five-membered ring containing zero, one, or two heteroatoms which are, independently, N, O, or S; or a four- membered ring containing zero, one, or two heteroatoms which are, independently, N, O, or S,
W is
Figure imgf000169_0002
R12 is H, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, or C2-6 alkynyl;
R13 is C2-6 alkenyl, C2-6 alkynyl, aryl, including phenyl, heteroaryl, including six- membered heteroaromatic rings containing one, two, or three nitrogen atoms and five- membered heteroaromatic rings containing one, two, or three heteroatoms, which are, independently, N, O, or S; alkylaryl, arylalkyl, a C4-14 bicyclic ring; a six- membered bridged or spiro-fused ring containing zero, one, or two heteroatoms which are, independently, N, O, or S,
R13 is optionally substituted with one or more substituents each independently selected from the group consisting of hydrogen, halogen (F, Cl, Br, I), CF3, SF5, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C(O)R’, C1-6 alkoxy, C1-6 haloalkoxy, cyano, azido, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C1- 6 alkyl, cycloalkyl, arylalkoxycarbonyl, carboxyl, haloalkyl, heterocyclylalkyl, or C1-6 hydroxyalkyl; or is optionally substituted with aryl, substituted aryl, heteroaryl, and substituted heteroaryl, where substituents on the substituted aryl and substituted heteroaryl are selected from the group consisting of halogen, SF5, CF3, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C(O)R’, C1-6 alkoxy, cyano, azido, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, and C1-6 alkyl;
or R12 and R13 together with the nitrogen to which they are attached form a 3 to 4 membered ring optionally substituted with one or more substituents each independently selected from the group consisting of hydrogen, halogen (F, Cl, Br, I), CF3, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C1-6 alkoxy, cyano, azido, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C1-6 alkyl, arylalkoxycarbonyl, carboxy, C1-6 haloalkyl, heterocyclylalkyl, and C1-6 hydroxyalkyl; vi) the compound of formula (A-VI) is
Figure imgf000170_0001
wherein
R1 and R1’ are as defined with respect to formula (A-I);
u and v are independently 0, 1, 2, 3, 4 or 5;
K is a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms, a five-membered heteroaromatic ring containing one, two, or three heteroatoms which are, independently, N, O, or S; a C4-14 bicyclic ring, alkylheteroaryl, or alkylaryl;
L is a five-membered heteroaromatic ring containing one, two, or three heteroatoms which are, independently, N, O, or S, a six or seven -membered ring or a six or seven- membered bridged or spiro-fused ring containing zero, one, or two heteroatoms which are, independently, N, O, or S, a five-membered ring containing zero, one, or two heteroatoms which are, independently, N, O, or S; a four-membered ring containing zero, one, or two heteroatoms which are, independently, N, O, or S, or a C4-14 bicyclic ring;
W is
Figure imgf000171_0001
R14 is H, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, or C2-6 alkynyl;
R15 is C1-6 alkyl, C1-6 haloalkyl, C2-8 alkoxyalkyl, a six-membered ring or a six- membered bridged or spiro-fused ring containing zero, one, or two heteroatoms which are, independently, N, O, or S, a seven-membered bridged or spiro-fused ring containing zero, one, or two heteroatoms which are, independently, N, O, or S, a five- membered ring containing zero, one, or two heteroatoms which are, independently, N, O, or S; a four-membered ring containing zero, one, or two heteroatoms which are, independently, N, O, or S;
R15 is optionally substituted with one or more substituents which are, independently, halogen (F, Cl, Br, I), SF5, CF3, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C1-6 alkoxy, C1-6 haloalkoxy, cyano, azido, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C1-6 alkyl, cycloalkyl, arylalkoxycarbonyl, carboxy, C1-6 haloalkyl, heterocyclylalkyl, or C1-6 hydroxyalkyl; or is optionally substituted with aryl, substituted aryl, heteroaryl, or substituted heteroaryl, where substituents on the substituted aryl and substituted heteroaryl are selected from the group consisting of halogen, SF5, CF3, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C(O)R’, C1-6 alkoxy, cyano, azido, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, and C1-6 alkyl;
or R14 and R15 can come together with the nitrogen to which they are attached to form a 6-10 membered bicyclic or bridged ring or a 3 to 8 saturated ring; such bicyclic, bridged and saturated ring moiety optionally containing one or more additional heteroatoms which are, independently, O, S or N, and optionally being substituted with one or more substituents each independently selected from the group consisting of halogen (including F, Cl, Br, and I), SF5, CF3, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C1-6 alkoxy, cyano, azido, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C1-6 alkyl, arylalkoxycarbonyl, carboxy, C1-6 haloalkyl, heterocyclylalkyl, and C1-6 hydroxyalkyl; and vii) the compound of formula (A-VII) is
Figure imgf000172_0001
wherein
R1 and R1’ are as defined with respect to formula (A-I);
u and v are independently 0, 1, 2, 3, 4 or 5;
M is phenyl, a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms, a five-membered heteroaromatic ring containing one, two, or three heteroatoms which are, independently, N, O, or S, a C4-14 bicyclic ring, alkylheteroaryl, or alkylaryl; N is phenyl, a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms, a five-membered heteroaromatic ring containing one, two, or three heteroatoms independently N, O, or S, a six or seven-membered ring or a six or seven- membered bridged or spiro-fused ring containing zero, one, or two heteroatoms which are, independently, N, O, or S, a five-membered ring containing zero, one, or two
heteroatoms which are, independently, N, O, or S; a four-membered ring containing zero, one, or two heteroatoms which are, independently, N, O, or S; or a C4-14 bicyclic ring,
V is
Figure imgf000172_0002
R16 is C1-6 alkyl, C1-6 haloalkyl, C2-8 alkoxyalkyl, C2-6 alkenyl, C2-6 alkynyl, aryl, such as phenyl, heteroaryl, such as a six-membered heteroaromatic ring containing one, two, or three nitrogen atoms or a five-membered heteroaromatic ring containing one, two, or three heteroatoms which are, independently, N, O, or S; a six- membered ring or a six- membered bridged or spiro-fused ring containing zero, one, or two heteroatoms which are, independently, N, O, or S, a five-membered ring containing zero, one, or two heteroatoms which are, independently, N, O, or S; a four- membered ring containing zero, one, or two heteroatoms which are, independently, N, O, or S; alkylaryl, arylalkyl, alkylheteroaryl, or alkylaryl,
wherein R16 is optionally substituted with one or more substituents selected from the group consisting of halogen (including F, Cl, Br, and I), SF5, CF3, hydroxy,
N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C1-6 alkoxy, C1-6 haloalkoxy, C2-6 alkenyl, cyano, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, C1-6 alkyl, arylalkoxycarbonyl, carboxy, C1-6 haloalkyl, heterocyclylalkyl, C1-6 hydroxyalkyl, aryl, substituted aryl, heteroaryl, and substituted heteroaryl, where substituents on the substituted aryl and substituted heteroaryl are selected from the group consisting of halogen, SF5, CF3, hydroxy, N(R’)S(O)2R’, S(O)2R’, S(O)2N(R’)2, C(O)R’, C1-6 alkoxy, cyano, azido, C2-6 alkynyl, C3-6 alkoxyalkyl, alkoxycarbonyl,
alkoxycarbonylalkyl, and C1-6 alkyl.
18. The therapeutic combination of any one of claims 1-14, wherein the compound is
selected from the group consisting of:
,
Figure imgf000173_0001
Figure imgf000174_0001
Figure imgf000175_0001
Figure imgf000176_0001
or a pharmaceutically acceptable salt thereof.
19. The therapeutic combination of any one of claims 1-14, wherein the capsid assembly modulator is a compound of formula (B-I) or a pharmaceutically accepted salt thereof,
Figure imgf000177_0001
, wherein:
R1 is C1-6 alkyl optionally substituted with 1 to 3 R1A , C3-8cycloalkyl optionally substituted with 1 to 4 R1B , or 3 to 8 membered monocyclic or bicyclic heterocyclyl having 1 to 3 heteroatoms selected from N, O, and S, optionally substituted with 1 to 3 R1C;
each R1A is independently halogen, -OH, -CN, -C1-2 haloalkyl, -C(O)NRXRY , C6-10 aryl optionally substituted with 1 to 3 R1D , or a 5 to 8 membered heteroaryl having 1 to 3 heteroatoms selected from N, O, and S, optionally substituted with 1 to 3 R1D , provided no more than 1 R1A is C6-10 aryl optionally substituted with 1 to 3 R1Dor 5 to 8 membered heteroaryl having 1 to 3 heteroatoms selected from N, O, and S optionally substituted with 1 to 3 R1D;
each R1B is independently -CN, halogen, C1-6alkyl optionally substituted with 1 to 3 -OH or -NRaRb, C2-4 alkynyl, C1-4 alkoxy, C1-2 haloalkyl, C3-6 cycloalkyl, -C(O)NRXRY , or a 5 to 8 membered heteroaryl having 1 to 3 heteroatoms selected from N, O, and S optionally substituted with 1 to 3 R1D , provided no more than 1 R1B is C3-6 cycloalkyl or 5 to 8 membered heteroaryl having 1 to 3 heteroatoms selected from N, O, and S optionally substituted with 1 to 3 R1D;
each R1C is independently C1-6 alkyl, oxo, C1-4haloalkyl, -C(O)H, -C(O)C1-4 alkyl, - C(O)OC1-4alkyl, or a 5 to 12 membered heteroaryl having 1 to 3 heteroatoms selected from N, O, and S optionally substituted with 1 to 3 R1D , provided no more than 1 R1C is a 5 to 12 membered heteroaryl having 1 to 3 heteroatoms selected from N, O, and S optionally substituted with 1 to 3 R1D;
each RX is independently -H, C3-6 cycloalkyl, C1-6 alkyl optionally substituted with 1 to 3 RZ , 3 to 8 membered monocyclic or bicyclic heterocyclyl having 1 to 3 heteroatoms selected from N, O, and S, optionally substituted with 1 to 3 RZ; each RY is independently -H or C1-6 alkyl optionally substituted with 1 to 3 RZ;
or RX and RY are taken together to form a 3 to 8 membered monocyclic or bicyclic heterocyclyl having 1 to 3 heteroatoms selected from N, O, and S, optionally substituted with 1 to 3 RZ;
wherein each RZ is independently halogen, methyl, ethyl, oxo, -OH, -S(O)2C1-3alkyl, or 3 to 8 membered monocyclic or bicyclic heterocyclyl having 1 to 3 heteroatoms selected from N, O, and S;
each Ra is -H, C1-3alkyl, or a 3 to 8 membered monocyclic or bicyclic heterocyclyl having 1 to 3 heteroatoms selected from N, O, and S, optionally substituted with 1 to 3 RZ; each Rb is -H or C1-3alkyl; or
Ra and Rb taken together form a 3 to 8 membered monocyclic or bicyclic heterocycle optionally substituted with 1 to 3 RZ;
the moiety is a pyrrolidine or a 5-7 membered bicyclic heterocycle having
Figure imgf000178_0001
one nitrogen, optionally substituted with 1 to 6 R2 groups; wherein each R2 is independently halogen, C1-3alkyl, -OH, or -OC1-3alkyl;
R3 is -H, halogen, or C1-4 alkyl;
R4 is C6-10 aryl optionally substituted with 1 to 5 R4A , or 5 to 12 membered heteroaryl having 1 to 3 heteroatoms selected from N, O, and S, optionally substituted with 1 to 4 R4B; and
each R1D , R4A , and R4B are independently -CN, halogen, C1-4 alkyl, -OC1-4alkyl, -OC1- 4 haloalkyl, or C1-4 haloalkyl.
20. The therapeutic combination of any one of claims 1-14, wherein the capsid assembly modulator is selected from the group consisting of:
Figure imgf000179_0001
Figure imgf000180_0001
Figure imgf000181_0001
Figure imgf000182_0001
Figure imgf000183_0001
Figure imgf000184_0001
Figure imgf000185_0001
Figure imgf000186_0001
Figure imgf000187_0001
Figure imgf000188_0001
Figure imgf000189_0001
Figure imgf000190_0001
Figure imgf000191_0001
, or a pharmaceutically acceptable salt thereof.
21. A kit comprising the therapeutic combination of any one of claims 1-20, and instructions for using the therapeutic combination in treating a hepatitis B virus (HBV) infection in a subject in need thereof.
22. The therapeutic combination of any one of claims 1 to 20 for use in treating a hepatitis B virus (HBV) infection in a subject in need thereof.
PCT/IB2020/055704 2019-06-18 2020-06-18 Combination of hepatitis b virus (hbv) vaccines and capsid assembly modulators being amide derivatives WO2020255013A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201962862775P 2019-06-18 2019-06-18
US62/862,775 2019-06-18

Publications (1)

Publication Number Publication Date
WO2020255013A1 true WO2020255013A1 (en) 2020-12-24

Family

ID=71728799

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2020/055704 WO2020255013A1 (en) 2019-06-18 2020-06-18 Combination of hepatitis b virus (hbv) vaccines and capsid assembly modulators being amide derivatives

Country Status (1)

Country Link
WO (1) WO2020255013A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022143610A1 (en) * 2020-12-29 2022-07-07 广东东阳光药业有限公司 Novel amide pyrrole compounds and use thereof in medicaments
WO2022156758A1 (en) * 2021-01-22 2022-07-28 广东东阳光药业有限公司 Novel amide pyrrole compound and use thereof in drugs
CN116120219A (en) * 2022-12-27 2023-05-16 吉斯凯(苏州)制药有限公司 Polysubstituted pyrrole compound, preparation method thereof and application thereof in preparation of HBV capsid protein inhibitor
CN116504302A (en) * 2023-06-21 2023-07-28 南京大学 Novel hepatitis B virus capsid assembly regulator de novo design and virtual screening method based on generation model and computational chemistry

Citations (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA92121A (en) 1904-11-07 1905-03-14 William Waid Paddock Pressure filter
US5273525A (en) 1992-08-13 1993-12-28 Btx Inc. Injection and electroporation apparatus for drug and gene delivery
US5624821A (en) 1987-03-18 1997-04-29 Scotgen Biopharmaceuticals Incorporated Antibodies with altered effector functions
US5873849A (en) 1997-04-24 1999-02-23 Ichor Medical Systems, Inc. Electrodes and electrode arrays for generating electroporation inducing electrical fields
US6041252A (en) 1995-06-07 2000-03-21 Ichor Medical Systems Inc. Drug delivery system and method
US6110161A (en) 1997-04-03 2000-08-29 Electrofect As Method for introducing pharmaceutical drugs and nucleic acids into skeletal muscle
US6117660A (en) 1997-06-10 2000-09-12 Cytopulse Sciences, Inc. Method and apparatus for treating materials with electrical fields having varying orientations
US6261281B1 (en) 1997-04-03 2001-07-17 Electrofect As Method for genetic immunization and introduction of molecules into skeletal muscle and immune cells
US6319901B1 (en) 1998-10-15 2001-11-20 Ichor Medical Systems, Inc. Methods for prolonging cell membrane permeability
US6697669B2 (en) 1998-07-13 2004-02-24 Genetronics, Inc. Skin and muscle-targeted gene therapy by pulsed electrical field
US6912417B1 (en) 2002-04-05 2005-06-28 Ichor Medical Systmes, Inc. Method and apparatus for delivery of therapeutic agents
US6939862B2 (en) 1997-06-30 2005-09-06 Aventis Pharma S.A. Method for transferring nucleic acid into striated muscles
US7328064B2 (en) 2002-07-04 2008-02-05 Inovio As Electroporation device and injection apparatus
US7664545B2 (en) 2002-03-07 2010-02-16 Vgx Pharmaceuticals, Inc. Electrode assembly for constant-current electroporation and use
US8209006B2 (en) 2002-03-07 2012-06-26 Vgx Pharmaceuticals, Inc. Constant current electroporation device and methods of use
WO2014106019A2 (en) 2012-12-27 2014-07-03 Philadelphia Health & Education Corporation, D/B/A Drexel Novel antiviral agents against hbv infection
WO2014184350A1 (en) * 2013-05-17 2014-11-20 Janssen R&D Ireland Sulphamoylpyrrolamide derivatives and the use thereof as medicaments for the treatment of hepatitis b
US20150274652A1 (en) * 2014-03-27 2015-10-01 Novira Therapeutics, Inc. Piperidine derivatives and methods of treating hepatitis b infections
WO2015172128A1 (en) * 2014-05-09 2015-11-12 Indiana University Research And Technology Corporation Methods and compositions for treating hepatitis b virus infections
WO2016020538A1 (en) * 2014-08-08 2016-02-11 Transgene Sa Hbv vaccine and antibody combination therapy to treat hbv infections
US9364664B2 (en) 2004-03-08 2016-06-14 Ichor Medical Systems, Inc. Apparatus for electrically mediated delivery of therapeutic agents
WO2016149581A1 (en) * 2015-03-19 2016-09-22 Novira Therapeutics, Inc. Azocane and azonane derivatives and methods of treating hepatitis b infections
US9452285B2 (en) 2006-10-17 2016-09-27 Vgx Pharmaceuticals, Inc. Electroporation devices and methods of using same for electroporation of cells in mammals
WO2017156255A1 (en) 2016-03-09 2017-09-14 Emory University Elimination of hepatitis b virus with antiviral agents
WO2017172838A1 (en) 2016-03-28 2017-10-05 Ichor Medical Systems, Inc. Method and apparatus for delivery of therapeutic agents
WO2018039531A1 (en) 2016-08-26 2018-03-01 Gilead Sciences, Inc. Substituted pyrrolizine compounds and uses thereof

Patent Citations (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA92121A (en) 1904-11-07 1905-03-14 William Waid Paddock Pressure filter
US5624821A (en) 1987-03-18 1997-04-29 Scotgen Biopharmaceuticals Incorporated Antibodies with altered effector functions
US5273525A (en) 1992-08-13 1993-12-28 Btx Inc. Injection and electroporation apparatus for drug and gene delivery
US6041252A (en) 1995-06-07 2000-03-21 Ichor Medical Systems Inc. Drug delivery system and method
US6958060B2 (en) 1997-04-03 2005-10-25 Genetronics, Inc. Method for muscle delivery of drugs, nucleic acids and other compounds
US6110161A (en) 1997-04-03 2000-08-29 Electrofect As Method for introducing pharmaceutical drugs and nucleic acids into skeletal muscle
US6261281B1 (en) 1997-04-03 2001-07-17 Electrofect As Method for genetic immunization and introduction of molecules into skeletal muscle and immune cells
US5873849A (en) 1997-04-24 1999-02-23 Ichor Medical Systems, Inc. Electrodes and electrode arrays for generating electroporation inducing electrical fields
US6278895B1 (en) 1997-04-24 2001-08-21 Ichor Medical Systems, Inc. Electrodes and electrode arrays for generating electroporation inducing electrical fields
US6117660A (en) 1997-06-10 2000-09-12 Cytopulse Sciences, Inc. Method and apparatus for treating materials with electrical fields having varying orientations
US6939862B2 (en) 1997-06-30 2005-09-06 Aventis Pharma S.A. Method for transferring nucleic acid into striated muscles
US6697669B2 (en) 1998-07-13 2004-02-24 Genetronics, Inc. Skin and muscle-targeted gene therapy by pulsed electrical field
US6319901B1 (en) 1998-10-15 2001-11-20 Ichor Medical Systems, Inc. Methods for prolonging cell membrane permeability
US8209006B2 (en) 2002-03-07 2012-06-26 Vgx Pharmaceuticals, Inc. Constant current electroporation device and methods of use
US7664545B2 (en) 2002-03-07 2010-02-16 Vgx Pharmaceuticals, Inc. Electrode assembly for constant-current electroporation and use
US8187249B2 (en) 2002-04-05 2012-05-29 Ichor Medical Systems, Inc. Method and apparatus for delivery of therapeutic agents
US6912417B1 (en) 2002-04-05 2005-06-28 Ichor Medical Systmes, Inc. Method and apparatus for delivery of therapeutic agents
US7328064B2 (en) 2002-07-04 2008-02-05 Inovio As Electroporation device and injection apparatus
US9364664B2 (en) 2004-03-08 2016-06-14 Ichor Medical Systems, Inc. Apparatus for electrically mediated delivery of therapeutic agents
US9802035B2 (en) 2004-03-08 2017-10-31 Ichor Medical Systems, Inc. Apparatus for electrically mediated delivery of therapeutic agents
US9452285B2 (en) 2006-10-17 2016-09-27 Vgx Pharmaceuticals, Inc. Electroporation devices and methods of using same for electroporation of cells in mammals
WO2014106019A2 (en) 2012-12-27 2014-07-03 Philadelphia Health & Education Corporation, D/B/A Drexel Novel antiviral agents against hbv infection
US20160024004A1 (en) 2012-12-27 2016-01-28 Drexel University Novel antiviral agents against hbv infection
WO2014184350A1 (en) * 2013-05-17 2014-11-20 Janssen R&D Ireland Sulphamoylpyrrolamide derivatives and the use thereof as medicaments for the treatment of hepatitis b
US20150274652A1 (en) * 2014-03-27 2015-10-01 Novira Therapeutics, Inc. Piperidine derivatives and methods of treating hepatitis b infections
WO2015172128A1 (en) * 2014-05-09 2015-11-12 Indiana University Research And Technology Corporation Methods and compositions for treating hepatitis b virus infections
WO2016020538A1 (en) * 2014-08-08 2016-02-11 Transgene Sa Hbv vaccine and antibody combination therapy to treat hbv infections
WO2016149581A1 (en) * 2015-03-19 2016-09-22 Novira Therapeutics, Inc. Azocane and azonane derivatives and methods of treating hepatitis b infections
WO2017156255A1 (en) 2016-03-09 2017-09-14 Emory University Elimination of hepatitis b virus with antiviral agents
WO2017172838A1 (en) 2016-03-28 2017-10-05 Ichor Medical Systems, Inc. Method and apparatus for delivery of therapeutic agents
WO2018039531A1 (en) 2016-08-26 2018-03-01 Gilead Sciences, Inc. Substituted pyrrolizine compounds and uses thereof
US20180161307A1 (en) 2016-08-26 2018-06-14 Gilead Sciences, Inc. Substituted pyrrolizine compounds and uses thereof

Non-Patent Citations (11)

* Cited by examiner, † Cited by third party
Title
ALTSCHUL SF ET AL., NUCLEIC ACIDS RES., vol. 25, 1997, pages 3389 - 3402
BELLONI ET AL., J. CLIN. INVEST., vol. 122, no. 2, 2012, pages 529 - 537
BIRD ET AL., SCIENCE, vol. 242, 1988, pages 423
COHEN ET AL., J. VIRAL HEPAT., vol. 18, no. 6, 2011, pages 377 - 83
COLIGAN ET AL.: "Current Protocols in Immunology", 1992, J WILEY & SONS INC, NATIONAL INSTITUTE OF HEALTH
HUSTON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 5879
LANZAVECCHIA ET AL., EUR. J. IMMUNOL., vol. 17, 1987, pages 105
MICHEL ET AL., J. HEPATOL., vol. 54, no. 6, 2011, pages 1286 - 1296
SAMBROOK ET AL.: "Molecular Cloning a Laboratory Manual", 1989, COLD SPRING HARBOR PRESS
TAUGHT IN GREENE ET AL.: "Protective Groups in Organic Synthesis", 1991, HEREBY INCORPORATED BY REFERENCE
WORLD HEALTH ORGANIZATION, HEPATITIS B: FACT SHEET NO. 204 [INTERNET, March 2015 (2015-03-01)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022143610A1 (en) * 2020-12-29 2022-07-07 广东东阳光药业有限公司 Novel amide pyrrole compounds and use thereof in medicaments
WO2022156758A1 (en) * 2021-01-22 2022-07-28 广东东阳光药业有限公司 Novel amide pyrrole compound and use thereof in drugs
CN116120219A (en) * 2022-12-27 2023-05-16 吉斯凯(苏州)制药有限公司 Polysubstituted pyrrole compound, preparation method thereof and application thereof in preparation of HBV capsid protein inhibitor
CN116504302A (en) * 2023-06-21 2023-07-28 南京大学 Novel hepatitis B virus capsid assembly regulator de novo design and virtual screening method based on generation model and computational chemistry
CN116504302B (en) * 2023-06-21 2023-11-17 南京大学 Novel hepatitis B virus capsid assembly regulator de novo design and virtual screening method based on generation model and computational chemistry

Similar Documents

Publication Publication Date Title
US11725194B2 (en) Hepatitis B virus (HBV) vaccines and uses thereof
EP3727445B1 (en) Hepatitis b virus (hbv) vaccines and uses thereof
WO2020255013A1 (en) Combination of hepatitis b virus (hbv) vaccines and capsid assembly modulators being amide derivatives
US20220305116A1 (en) Combination of hepatitis b virus (hbv) vaccines and capsid assembly modulators being sulfonamide derivatives
US20220233526A1 (en) Combination of hepatitis b virus (hbv) vaccines and dihydropyrimidine derivatives as capsid assembly modulators
US20220305118A1 (en) Carbohydrate nanocarrier delivery of hepatitis b virus (hbv) vaccines
US20220249647A1 (en) Combination of hepatitis b virus (hbv) vaccines and dihydropyrimidine derivatives as capsid assembly modulators
US20220226467A1 (en) Arenavirus vectors for hepatitis b virus (hbv) vaccines and uses thereof
US20220305114A1 (en) Combination of hepatitis b virus (hbv) vaccines and small molecule pdl1 or pd1 inhibitor
WO2020255010A1 (en) Combination of recombinant interleukin 12 construct and hepatitis b virus (hbv) vaccines
WO2020254876A1 (en) Virus-like particle delivery of hepatitis b virus (hbv) vaccines
US20220324916A1 (en) Hepatitis B Virus (HBV) Vaccines and Uses Thereof
WO2020255042A1 (en) Combination of hepatitis b virus (hbv) vaccines and a pyrimidine derivative
US20220305108A1 (en) Lipid nanoparticle or liposome delivery of hepatitis b virus (hbv) vaccines
WO2020255035A1 (en) Combination of hepatitis b virus (hbv) vaccines and pyrimidine derivatives
WO2020255019A1 (en) Combination of hepatitis b virus (hbv) vaccines and a quinazoline derivative
OA19833A (en) Hepatitis B virus (HBV) vaccines and uses thereof.
EA045279B1 (en) VACCINES AGAINST HEPATITIS B VIRUS (HBV) AND THEIR USE
TW201930594A (en) Hepatitis B virus (HBV) vaccines and uses thereof

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: 20743290

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: 20743290

Country of ref document: EP

Kind code of ref document: A1