USE OF COPS3 INHIBITORS FOR TREATING HEPATITIS B VIRUS INFECTION

Information

  • Patent Application
  • 20230122751
  • Publication Number
    20230122751
  • Date Filed
    June 21, 2022
    2 years ago
  • Date Published
    April 20, 2023
    a year ago
Abstract
The present invention relates to a COPS3 inhibitor for use in treatment of an HBV infection, in particular a chronic HBV infection. The invention in particular relates to the use of COPS3 inhibitors for destabilizing cccDNA, such as HBV cccDNA. The invention also relates to nucleic acid molecules which are complementary to COPS3 and capable of reducing the level of a COPS3 mRNA. Also comprised in the present invention is a pharmaceutical composition and its use in the treatment of a HBV infection.
Description
FIELD OF INVENTION

The present invention relates to COPS3 inhibitors for use in treating and/or preventing a hepatitis B virus (HBV) infection, in particular a chronic HBV infection. The invention in particular relates to the use of COPS3 inhibitors for destabilizing cccDNA, such as HBV cccDNA. The invention also relates to nucleic acid molecules, such as oligonucleotides including siRNA, shRNA and antisense oligonucleotides, that are complementary to COPS3, and capable of reducing the expression of COPS3. Also comprised in the present invention is a pharmaceutical composition and its use in the treatment and/or prevention of a HBV infection.


BACKGROUND

Hepatitis B is an infectious disease caused by the hepatitis B virus (HBV), a small hepatotropic virus that replicates through reverse transcription. Chronic HBV infection is a key factor for severe liver diseases such as liver cirrhosis and hepatocellular carcinoma. Current treatments for chronic HBV infection are based on administration of pegylated type 1 interferons or nucleos(t)ide analogues, such as lamivudine, adefovir, entecavir, tenofovir disoproxil, and tenofovir alafenamide, which target the viral polymerase, a multifunctional reverse transcriptase. Treatment success is usually measured as loss of hepatitis B surface antigen (HBsAg). However, a complete HBsAg clearance is rarely achieved since Hepatitis B virus DNA persists in the body after infection. HBV persistence is mediated by an episomal form of the HBV genome which is stably maintained in the nucleus. This episomal form is called “covalently closed circular DNA” (cccDNA). The cccDNA serves as a template for all HBV transcripts, including pregenomic RNA (pgRNA), a viral replicative intermediate. The presence of a few copies of cccDNA might be sufficient to reinitiate a full-blown HBV infection. Current treatments for HBV do not target cccDNA. A cure of chronic HBV infection, however, would require the elimination of cccDNA (reviewed by Nassal, Gut. 2015 Dec;64(12):1972-84. doi: 10.1136/gutjnl-2015-309809).


The COP9 (Constitutive photomorphogenesis 2) signalosome is a protein complex having isopeptidase activity. It catalyses the hydrolysis of NEDD8 protein from the cullin subunit of Cullin-RING ubiquitin ligases and it is responsible for deneddylation of Cullin-RING ubiquitin ligases. Further, it is capable to bind the denedyllated cullin-RING complex, thereby retaining the complex in deactivated form. Accordingly, the COP9 signalosome functions as deactivator of Cullin-RING ubiquitin ligases. In mammals, the signalosome is involved in various processes such as signal transduction, protein stability, protein phosphorylation, cell cycle regulation and apoptosis. The COP signalosome is found in all eukaryotic organisms. In humans, the COP9 signalosome comprises eight subunits and has a size of about 350 kDa. All subunits seem to be essential for full function of the signalosome. (Lingaraju et al. (2014) Nature. 512 (7513): 161-5. doi:10.1038/nature13566. PMID 25043011).


COPS3 (COP9 Signalosome Subunit a) is the third subunit of the signalosome and maintains the integrity of the complex. It has been shown to bind to the striated muscle-specific βD integrin tail, and its subcellular localization is altered in differentiated skeletal muscle cells. Other names for COPS3 are JAB1-Containing Signalosome Subunit 3, Signalosome Subunit 3, CSN3 and SGN3.


Various publications describe the down-regulation of COPS3 in target cells using RNAi based Technologies, some of these publications are cited below.


Ba et al. describe the use of shRNAs for down-regulation of COPS3 in C2C12 cells. Downregulation of COPS3 was accompanied by destabilization of several COP9 subunits and increased nuclear NF-κB localization and reduced growth rate (Ba et al., BMC Pharmacol Toxicol. 2017 Jun 17;18(1):47. doi: 10.1186/s40360-017-0154-5).


Kim et al. examined the effects of siRNA mediated knockdown of each subunit of the COP9 signalosome in oocytes. COPS3 knockdown leads to meiosis I arrest, disruption of maturation promoting factor (MPF) activity, and decreased degradation of anaphase-promoting complex/cyclosome (APC/C) substrates (Kim et al., PLoS One. 2011; 6(10): e25870. doi: 10.1371/journal.pone.0025870).


Yoneda-Kato et al. showed that Myeloid leukemia factor 1 regulates p53 by suppressing COP1 via COP9 signalosome subunit 3. Specifically, reduction in the level of COPS3 protein with siRNA abrogated MLF1-induced G1 arrest and impaired the activation of p53 by genotoxic stress (Yoneda-Kato et al., The EMBO Journal (2005) 24, 1739-1749. doi:10.1038/sj.emboj.7600656).


COPS3 further plays a role in cancer. E.g., Pang et al. showed that knockdown of COPS3 with shRNA inhibits lung cancer tumor growth in nude mice. (Pang et al., J Cancer. 2017 Apr 9;8(7):1129-1136. doi: 10.7150/jca.16201). Similarly, Yan et al. demonstrated that siRNA mediated COPS3 gene silencing reduced proliferation and migration of HOS cells and may be relevant for metastasis (Yan et al., Cancer Gene Therapy (2011) 18, 450-456). Yu et al. showed that knockdown of COPS3 expression using shRNA in hepatocellular carcinoma cell lines (SMMC-7721 and Hep3B) showed in vitro growth inhibition as well as in vivo tumor weight reduction in xenograft mice (Yu et al., Cancer Chemother Pharmacol (2012) 69:1173-1180, DOI 10.1007/s00280 1810-x).


To our knowledge COPS3 has never been identified as a cccDNA dependency factor in the context of cccDNA stability and maintenance, nor have molecules inhibiting COPS3 ever been suggested as cccDNA destabilizers for the treatment of HBV infection.


OBJECTIVE OF THE INVENTION

The present invention shows that there is an association between the inhibition of COPS3 (COP9 Signalosome Subunit 3 or Constitutive photomorphogenesis 9 Signalosome Subunit 3) and reduction of cccDNA in an HBV infected cell, which is relevant in the treatment of HBV infected individuals. An objective of the present invention is to identify COPS3 inhibitors which reduce cccDNA in an HBV infected cell. Such COPS3 inhibitors can be used in the treatment of HBV infection.


The present invention further identifies novel nucleic acid molecules, which are capable of inhibiting the expression of COPS3 in vitro and in vivo.


SUMMARY OF INVENTION

The present invention relates to oligonucleotides targeting a nucleic acid capable of modulating the expression of COPS3 and to treat or prevent diseases related to the functioning of the COPS3.


Accordingly, in a first aspect the invention provides a COPS3 inhibitor for use in the treatment and/or prevention of Hepatitis B virus (HBV) infection. In particular, a COPS3 inhibitor capable of reducing HBV cccDNA and/or HBV pre-genomic RNA (pgRNA) is useful. Such an inhibitor is advantageously a nucleic acid molecule of 12 to 60 nucleotides in length, which is capable of reducing COPS3 mRNA.


In a further aspect, the invention relates to a nucleic acid molecule of 12-60 nucleotides, such as of 12-30 nucleotides, comprising a contiguous nucleotides sequence of at least 12 nucleotides, in particular of 16 to 20 nucleotides, which is at least 90% complementary to a mammalian COPS3, e.g. a human COPS3, a mouse COPS3 or a cynomolgus monkey COPS3. Such a nucleic acid molecule is capable of inhibiting the expression of COPS3 in a cell expressing COPS3. The inhibition of COPS3 allows for a reduction of the amount of cccDNA present in the cell. The nucleic acid molecule can be selected from a single stranded antisense oligonucleotide, a double stranded siRNA molecule or a shRNA nucleic acid molecule (in particular a chemically produced shRNA molecules).


A further aspect of the present invention relates to single stranded antisense oligonucleotides or siRNA's that inhibit expression and/or activity of COPS3. In particular, modified antisense oligonucleotides or modified siRNA comprising one or more 2′ sugar modified nucleoside(s) and one or more phosphorothioate linkage(s), which reduce COPS3 mRNA are of advantageous.


In a further aspect, the invention provides pharmaceutical compositions comprising the COPS3 inhibitor of the present invention, such as the antisense oligonucleotide or siRNA of the invention and a pharmaceutically acceptable excipient.


In a further aspect, the invention provides methods for in vivo or in vitro modulation of COPS3 expression in a target cell which is expressing COPS3, by administering a COPS3 inhibitor of the present invention, such as an antisense oligonucleotide or composition of the invention in an effective amount to said cell. In some embodiments, the COPS3 expression is reduced by at least 50%, or at least 60% in the target cell compared to the level without any treatment or treated with a control. In some embodiments, the target cell is infected with HBV and the cccDNA in an HBV infected cell is reduced by at least 50%, or at least 60% in the HBV infected target cell compared to the level without any treatment or treated with a control. In some embodiments, the target cell is infected with HBV and the cccDNA in an HBV infected cell is reduced by at least 25%, such as by at least 40%, in the HBV infected target cell compared to the level without any treatment or treated with a control. In some embodiments, the target cell is infected with HBV and the pgRNA in an HBV infected cell is reduced by at least 50%, or at least 60%, or at least 70%, or at least 80%, in the HBV infected target cell compared to the level without any treatment or treated with a control.


In a further aspect, the invention provides methods for treating or preventing a disease, disorder or dysfunction associated with in vivo activity of COPS3 comprising administering a therapeutically or prophylactically effective amount of the COPS3 inhibitor of the present invention, such as the antisense oligonucleotide or siRNA of the invention to a subject suffering from or susceptible to the disease, disorder or dysfunction.


Further aspects of the invention are conjugates of nucleic acid molecules of the invention and pharmaceutical compositions comprising the molecules of the invention. In particular conjugates targeting the liver are of interest, such as GaINAc clusters.





BRIEF DESCRIPTION OF FIGURES


FIG. 1A-1 to FIG. 1L: Illustrate exemplary antisense oligonucleotide conjugates, wherein the oligonucleotide is represented by the term “Oligonucleotide” and the asialoglycoprotein receptor targeting conjugate moieties are trivalent N-acetylgalactosamine moieties. Compounds in FIG. 1A-1 to FIG. 1D-2 comprise a di-lysine brancher molecule, a PEG3 spacer and three terminal GaINAc carbohydrate moieties.

    • FIG. 1A-1 and FIG. 1A-2 show two different diastereoisomers of the same compound. In the compounds in FIG. 1A-1 and FIG. 1A-2, the oligonucleotide is attached directly to the asialoglycoprotein receptor targeting conjugate moiety without a linker.
    • FIG. 1B-1 and FIG. 1B-2 show two different diastereoisomers of the same compound. In the compounds in FIG. 1B-1 and FIG. 1B-2, the oligonucleotide is attached directly to the asialoglycoprotein receptor targeting conjugate moiety without a linker.
    • FIG. 1C-1 and FIG. 1C-2 show two different diastereoisomers of the same compound. In the compounds in FIG. 1C-1 and FIG. 1C-2, the oligonucleotide is attached to the asialoglycoprotein receptor targeting conjugate moiety via a C6 linker.
    • FIG. 1D-1 and FIG. 1D-2 show two different diastereoisomers of the same compound. In the compounds in FIG. 1D-1 and FIG. 1D-2, the oligonucleotide is attached to the asialoglycoprotein receptor targeting conjugate moiety via a C6 linker.
    • The compounds in FIG. 1E, FIG. 1F, FIG. 1G, FIG. 1H, FIG. 1I, FIG. 1J, and FIG. 1K comprise a commercially available trebler brancher molecule and spacers of varying length and structure and three terminal GaINAc carbohydrate moieties.
    • The compound in FIG. 1L is composed of monomeric GaINAc phosphoramidites added to the oligonucleotide while still on the solid support as part of the synthesis, wherein X=S or O, and independently Y=S or O, and n=1-3 (see WO 2017/178656). FIG. 1B-1, FIG. 1B-2, FIG. 1D-1, and FIG. 1D-2 are also termed GaINAc2 or GN2 herein, without and with C6 linker respectively.
    • The two different diastereoisomers shown in each of FIG. 1A-1 to FIG. 1D-2 are the result of the conjugation reaction. A pool of a specific antisense oligonucleotide conjugate can therefore contain only one of the two different diastereoisomers, or a pool of a specific antisense oligonucleotide conjugate can contain a mixture of the two different diastereoisomers.





Definitions

HBV Infection


The term “hepatitis B virus infection” or “HBV infection” is commonly known in the art and refers to an infectious disease that is caused by the hepatitis B virus (HBV) and affects the liver. A HBV infection can be an acute or a chronic infection. Chronic hepatitis B virus (CHB) infection is a global disease burden affecting 248 million individuals worldwide. Approximately 686,000 deaths annually are attributed to HBV-related end-stage liver diseases and hepatocellular carcinoma (HCC) (GBD 2013; Schweitzer et al., Lancet. 2015 Oct 17;386(10003):1546-55). WHO projected that without expanded intervention, the number of people living with CHB infection will remain at the current high levels for the next 40-50 years, with a cumulative 20 million deaths occurring between 2015 and 2030 (WHO 2016). CHB infection is not a homogenous disease with singular clinical presentation. Infected individuals have progressed through several phases of CHB-associated liver disease in their life; these phases of disease are also the basis for treatment with standard of care (SOC). Current guidelines recommend treating only selected CHB-infected individuals based on three criteria - serum ALT level, HBV DNA level, and severity of liver disease (EASL, 2017). This recommendation was due to the fact that SOC i.e. nucleos(t)ide analogs (NAs) and pegylated interferon-alpha (PEG-IFN), are not curative and must be administered for long periods of time thereby increasing their safety risks. NAs effectively suppress HBV DNA replication; however, they have very limited/no effect on other viral markers. Two hallmarks of HBV infection, hepatitis B surface antigen (HBsAg) and covalently closed circular DNA (cccDNA), are the main targets of novel drugs aiming for HBV cure. In the plasma of CHB individuals, HBsAg subviral (empty) particles outnumber HBV virions by a factor of 103 to 105 (Ganem & Prince, N Engl J Med. 2004 Mar 11;350(11):1118-29); its excess is believed to contribute to immunopathogenesis of the disease, including inability of individuals to develop neutralizing anti-HBs antibody, the serological marker observed following resolution of acute HBV infection.


In some embodiments, the term “HBV infection” refers to “chronic HBV infection”.


Further, the term encompasses infection with any HBV genotype.


In some embodiments, the patient to be treated is infected with HBV genotype A.


In some embodiments, the patient to be treated is infected with HBV genotype B.


In some embodiments, the patient to be treated is infected with HBV genotype C.


In some embodiments, the patient to be treated is infected with HBV genotype D.


In some embodiments, the patient to be treated is infected with HBV genotype E.


In some embodiments, the patient to be treated is infected with HBV genotype F.


In some embodiments, the patient to be treated is infected with HBV genotype G.


In some embodiments, the patient to be treated is infected with HBV genotype H.


In some embodiments, the patient to be treated is infected with HBV genotype I.


In some embodiments, the patient to be treated is infected with HBV genotype J.


cccDNA (covalently closed circular DNA) cccDNA is the viral genetic template of HBV that resides in the nucleus of infected hepatocytes, where it gives rise to all HBV RNA transcripts needed for productive infection and is responsible for viral persistence during natural course of chronic HBV infection (Locarnini & Zoulim, Antivir Ther. 2010; 15 Suppl 3:3-14. doi: 10.3851/IMP1619). Acting as a viral reservoir, cccDNA is the source of viral rebound after cessation of treatment, necessitating long term, often lifetime treatment. PEG-IFN can only be administered to a small subset of CHB due to its various side effects.


Consequently, novel therapies that can deliver a complete cure, defined by degradation or elimination of HBV cccDNA, to the majority of CHB patients are highly needed.


Compound Herein, the term “compound” means any molecule capable of inhibition COPS3 expression or activity. Particular compounds of the invention are nucleic acid molecules, such as RNAi molecules or antisense oligonucleotides according to the invention or any conjugate comprising such a nucleic acid molecule. For example, herein the compound may be a nucleic acid molecule targeting COPS3, in particular an antisense oligonucleotide or a siRNA.


Oligonucleotide The term “oligonucleotide” as used herein is defined as it is generally understood by the skilled person as a molecule comprising two or more covalently linked nucleosides. Such covalently bound nucleosides may also be referred to as nucleic acid molecules or oligomers.


The oligonucleotides referred to in the description and claims are generally therapeutic oligonucleotides below 70 nucleotides in length. The oligonucleotide may be or comprise a single stranded antisense oligonucleotide, or may be another nucleic acid molecule, such as a CRISPR


RNA, a siRNA, shRNA, an aptamer, or a ribozyme. Therapeutic oligonucleotide molecules are commonly made in the laboratory by solid-phase chemical synthesis followed by purification and isolation. shRNA's are however often delivered to cells using lentiviral vectors from which they are then transcribed to produce the single stranded RNA that will form a stem loop (hairpin) RNA structure that is capable of interacting with the RNA interference machinery (including the RNA-induced silencing complex (RISC)). In an embodiment of the present invention the shRNA is chemically produced shRNA molecules (not relying on cell based expression from plasmids or viruses). When referring to a sequence of the oligonucleotide, reference is made to the sequence or order of nucleobase moieties, or modifications thereof, of the covalently linked nucleotides or nucleosides. Generally, the oligonucleotide of the invention is man-made, and is chemically synthesized, and is typically purified or isolated. Although in some embodiments the oligonucleotide of the invention is a shRNA transcribed from a vector upon entry into the target cell. The oligonucleotide of the invention may comprise one or more modified nucleosides or nucleotides.


In some embodiments, the oligonucleotide of the invention comprises or consists of 10 to 70 nucleotides in length, such as from 12 to 60, such as from 13 to 50, such as from 14 to 40, such as from 15 to 30, such as from 16 to 25, such as from 16 to 22, such as from 16 to 20 contiguous nucleotides in length. Accordingly, the oligonucleotide of the present invention, in some embodiments, may have a length of 12 to 25 nucleotides. Alternatively, the oligonucleotide of the present invention, in some embodiments, may have a length of 15 to 22 nucleotides.


In some embodiments, the oligonucleotide or contiguous nucleotide sequence thereof comprises or consists of 24 or less nucleotides, such as 22, such as 20 or less nucleotides, such as 18 or less nucleotides, such as 14, 15, 16 or 17 nucleotides. It is to be understood that any range given herein includes the range endpoints. Accordingly, if a nucleic acid molecule is said to include from 12 to 25 nucleotides, both 12 and 25 nucleotides are included.


In some embodiments, the contiguous nucleotide sequence comprises or consists of 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 or 22 contiguous nucleotides in length


The olignucleotide(s) are for modulating the expression of a target nucleic acid in a mammal. In some embodiments, the nucleic acid molecules, such as for siRNAs, shRNAs and antisense oligonucleotides, are typically for inhibiting the expression of a target nucleic acid(s).


In one embodiment, of the invention oligonucleotide is selected from a RNAi agent, such as a siRNA or shRNA. In another embodiment, the oligonucleotide is a single stranded antisense oligonucleotide, such as a high affinity modified antisense oligonucleotide interacting with RNase H.


In some embodiments, the oligonucleotide of the invention may comprise one or more modified nucleosides or nucleotides, such as 2′ sugar modified nucleosides.


In some embodiments, the oligonucleotide comprises phosphorothioate internucleoside linkages.


In some embodiments, the oligonucleotide may be conjugated to non-nucleosidic moieties (conjugate moieties).


A library of oligonucleotides is to be understood as a collection of variant oligonucleotides. The purpose of the library of oligonucleotides can vary. In some embodiments, the library of oligonucleotides is composed of oligonucleotides with overlapping nucleobase sequence targeting one or more mammalian COPS3 target nucleic acids with the purpose of identifying the most potent sequence within the library of oligonucleotides. In some embodiments, the library of oligonucleotides is a library of oligonucleotide design variants (child nucleic acid molecules) of a parent or ancestral oligonucleotide, wherein the oligonucleotide design variants retaining the core nucleobase sequence of the parent nucleic acid molecule.


Antisense oligonucleotides The term “antisense oligonucleotide” or “ASO” as used herein is defined as oligonucleotides capable of modulating expression of a target gene by hybridizing to a target nucleic acid, in particular to a contiguous sequence on a target nucleic acid. The antisense oligonucleotides are not essentially double stranded and are therefore not siRNAs or shRNAs. Preferably, the antisense oligonucleotides of the present invention are single stranded. It is understood that single stranded oligonucleotides of the present invention can form hairpins or intermolecular duplex structures (duplex between two molecules of the same oligonucleotide), as long as the degree of intra or inter self complementarity is less than 50% across of the full length of the oligonucleotide.


Advantageously, the single stranded antisense oligonucleotide of the invention does not contain RNA nucleosides, since this will decrease nuclease resistance.


Advantageously, the oligonucleotide of the invention comprises one or more modified nucleosides or nucleotides, such as 2′ sugar modified nucleosides. Furthermore, it is advantageous that the nucleosides which are not modified are DNA nucleosides.


RNAi molecules Herein, the term “RNA interference (RNAi) molecule” refers to short double-stranded oligonucleotide containing RNA nucleosides and which mediates targeted cleavage of an RNA transcript via the RNA-induced silencing complex (RISC), where they interact with the catalytic RISC component argonaute. The RNAi molecule modulates, e g., inhibits, the expression of the target nucleic acid in a cell, e.g. a cell within a subject. such as a mammalian subject. RNAi molecules includes single stranded RNAi molecules (Lima at al 2012 Cell 150: 883) and double stranded siRNAs, as well as short hairpin RNAs (shRNAs). In some embodiments of the invention, the oligonucleotide of the invention or contiguous nucleotide sequence thereof is a RNAi agent, such as a siRNA.


siRNA The term “small interfering ribonucleic acid” or “siRNA” refers to a small interfering ribonucleic acid RNAi molecule. It is a class of double-stranded RNA molecules, also known in the art as short interfering RNA or silencing RNA. siRNAs typically comprise a sense strand (also referred to as a passenger strand) and an antisense strand (also referred to as the guide strand), wherein each strand are of 17-30 nucleotides in length, typically 19-25 nucleosides in length, wherein the antisense strand is complementary, such as at least 95% complementary, such as fully complementary, to the target nucleic acid (suitably a mature mRNA sequence), and the sense strand is complementary to the antisense strand so that the sense strand and antisense strand form a duplex or duplex region. siRNA strands may form a blunt ended duplex, or advantageously the sense and antisense strand 3′ ends may form a 3′ overhang of e.g. 1, 2 or 3 nucleosides to resemble the product produced by Dicer, which forms the RISC substrate in vivo. Effective extended forms of Dicer substrates have been described in U.S. Pat. Nos. 8,349,809 and 8,513,207, hereby incorporated by reference. In some embodiments, both the sense strand and antisense strand have a 2nt 3′ overhang. The duplex region may therefore be, for example 17-25 nucleotides in length, such as 21-23 nucleotides in length.


Once inside a cell the antisense strand is incorporated into the RISC complex which mediate target degradation or target inhibition of the target nucleic acid. siRNAs typically comprise modified nucleosides in addition to RNA nucleosides. In one embodiment, the siRNA molecule may be chemically modified using modified internucleotide linkages and 2′ sugar modified nucleosides, such as 2′-4′ bicyclic ribose modified nucleosides, including LNA and cET or 2′ substituted modifications like of 2′-O-alkyl-RNA, 2′-O-methyl-RNA, 2′-alkoxy-RNA, 2′-O-methoxyethyl-RNA (MOE), 2′-amino-DNA, 2′-fluoro-DNA, arabino nucleic acid (ANA), 2′-fluoro-ANA. In particular 2′ fluoro, 2′-O-methyl or 2′-O-methoxyethyl may be incorporated into siRNAs.


In some embodiments, all of the nucleotides of an siRNA sense (passenger) strand may be modified with 2′ sugar modified nucleosides such as LNA (see WO2004/083430, WO2007/085485 for example). In some embodiments, the passenger stand of the siRNA may be discontinuous (see WO2007/107162 for example). The incorporation of thermally destabilizing nucleotides occurring at a seed region of the antisense strand of siRNAs have been reported as useful in reducing off-target activity of siRNAs (see WO2018/098328 for example). Suitably the siRNA comprises a 5′ phosphate group or a 5′-phosphate mimic at the 5′ end of the antisense strand. In some embodiments, the 5′ end of the antisense strand is a RNA nucleoside.


In one embodiment, the siRNA molecule further comprises at least one phosphorothioate or methylphosphonate internucleoside linkage. The phosphorothioaie or methylphosphonate internucleoside linkage may be at the 3′- terminus one or both strand (e.g., the antisense strand; or the sense strand); or the phosphorothioate or methylphosphonate internucleoside linkage may be at the 5′-terminus of one or both strands (e.g., the antisense strand; or the sense strand); or the phosphorothioate or methylphosphonate internucleoside linkage may be at the both the 5′- and 3′-terminus of one or both strands (e.g., the antisense strand; or the sense strand). In some embodiments, the remaining internucleoside linkages are phosphodiester linkages. In some embodiments, siRNA molecules comprise one or more phosphorothioate internucleoside linkages. In siRNA molecules phosphorothioate internucleoside linkages may reduce or the nuclease cleavage in RICS, it is therefore advantageous that not all internucleoside linkages in the antisense strand are modified.


The siRNA molecule may further comprise a ligand. In some embodiments, the ligand is conjugated to the 3′ end of the sense strand.


For biological distribution, siRNAs may be conjugated to a targeting ligand, and/or be formulated into lipid nanoparticles.


Other aspects of the invention relate to pharmaceutical compositions comprising these dsRNA, such as siRNA molecules suitable for therapeutic use, and methods of inhibiting the expression of the target gene by administering the dsRNA molecules such as siRNAs of the invention, e.g., for the treatment of various disease conditions as disclosed herein.


shRNA The term “short hairpin RNA” or “shRNA” refers to molecules that are generally between 40 and 70 nucleotides in length, such as between 45 and 65 nucleotides in length, such as 50 and 60 nucleotides in length and form a stem loop (hairpin) RNA structure which interacts with the endonuclease known as Dicer which is believed to processes dsRNA into 19-23 base pair short interfering RNAs with characteristic two base 3′ overhangs which are then incorporated into an RNA-induced silencing complex (RISC). Upon binding to the appropriate target mRNA, one or more endonucleases within the RISC cleave the target to induce silencing. shRNA oligonucleotides may be chemically modified using modified internucleotide linkages and 2′ sugar modified nucleosides, such as 2′-4′ bicyclic ribose modified nucleosides, including LNA and cET or 2′ substituted modifications like of 2′-O-alkyl-RNA, 2′-O-methyl-RNA, 2′-alkoxy-RNA, 2′-O-methoxyethyl-RNA (MOE), 2′-amino-DNA, 2′-fluoro-DNA, arabino nucleic acid (ANA), 2′-fluoro-ANA.


In some embodiments, shRNA molecule comprises one or more phosphorothioate internucleoside linkages. In RNAi molecules phosphorothioate internucleoside linkages may reduce or the nuclease cleavage in RICS it is therefore advantageous that not al internucleoside linkages in the stem loop of the shRNA molecule are modified. Phosphorothioate internucleoside linkages can advantageously be placed in the 3′ and/or 5′ end of the stem loop of the shRNA molecule, in particular in the part of the molecule that is not complementary to the target nucleic acid. The region of the shRNA molecule that is complementary to the target nucleic acid may however also be modified in the first 2 to 3 internucleoside linkages in the part that is predicted to become the 3′ and/or 5′ terminal following cleavage by Dicer.


Contiguous Nucleotide Sequence The term “contiguous nucleotide sequence” refers to the region of the nucleic acid molecule which is complementary to the target nucleic acid. The term is used interchangeably herein with the term “contiguous nucleobase sequence” and the term “oligonucleotide motif sequence”. In some embodiments, all the nucleotides of the oligonucleotide constitute the contiguous nucleotide sequence. In some embodiments, the contiguous nucleotide sequence is included in the guide strand of an siRNA molecule. In some embodiments, the contiguous nucleotide sequence is the part of an shRNA molecule which is 100% complementary to the target nucleic acid. In some embodiments, the oligonucleotide comprises the contiguous nucleotide sequence, such as a F-G-F′ gapmer region, and may optionally comprise further nucleotide(s), for example a nucleotide linker region which may be used to attach a functional group (e.g. a conjugate group for targeting) to the contiguous nucleotide sequence. The nucleotide linker region may or may not be complementary to the target nucleic acid. In some embodiments, the nucleobase sequence of the antisense oligonucleotide is the contiguous nucleotide sequence. In some embodiments, the contiguous nucleotide sequence is 100% complementary to the target nucleic acid.


Nucleotides and nucleosides Nucleotides and nucleosides are the building blocks of oligonucleotides and polynucleotides, and for the purposes of the present invention include both naturally occurring and non-naturally occurring nucleotides and nucleosides. In nature, nucleotides, such as DNA and RNA nucleotides comprise a ribose sugar moiety, a nucleobase moiety and one or more phosphate groups (which is absent in nucleosides). Nucleosides and nucleotides may also interchangeably be referred to as “units” or “monomers”.


Modified nucleoside The term “modified nucleoside” or “nucleoside modification” as used herein refers to nucleosides modified as compared to the equivalent DNA or RNA nucleoside by the introduction of one or more modifications of the sugar moiety or the (nucleo)base moiety. Advantageously, one or more of the modified nucleoside comprises a modified sugar moiety. The term modified nucleoside may also be used herein interchangeably with the term “nucleoside analogue” or modified “units” or modified “monomers”. Nucleosides with an unmodified DNA or RNA sugar moiety are termed DNA or RNA nucleosides herein. Nucleosides with modifications in the base region of the DNA or RNA nucleoside are still generally termed DNA or RNA if they allow Watson Crick base pairing.


Modified internucleoside linkage The term “modified internucleoside linkage” is defined as generally understood by the skilled person as linkages other than phosphodiester (PO) linkages, that covalently couples two nucleosides together. The oligonucleotides of the invention may therefore comprise one or more modified internucleoside linkages, such as a one or more phosphorothioate internucleoside linkages, or one or more phoshporodithioate internucleoside linkages.


With the oligonucleotide of the invention it is advantageous to use phosphorothioate internucleoside linkages.


Phosphorothioate internucleoside linkages are particularly useful due to nuclease resistance, beneficial pharmacokinetics and ease of manufacture. In some embodiments, at least 50% of the internucleoside linkages in the oligonucleotide, or contiguous nucleotide sequence thereof, are phosphorothioate, such as at least 60%, such as at least 70%, such as at least 75%, such as at least 80% or such as at least 90% of the internucleoside linkages in the oligonucleotide, or contiguous nucleotide sequence thereof, are phosphorothioate. In some embodiments, all of the internucleoside linkages of the oligonucleotide, or contiguous nucleotide sequence thereof, are phosphorothioate.


In some advantageous embodiments, all the internucleoside linkages of the contiguous nucleotide sequence of the oligonucleotide are phosphorothioate, or all the internucleoside linkages of the oligonucleotide are phosphorothioate linkages.


It is recognized that, as disclosed in EP 2 742 135, antisense oligonucleotides may comprise other internucleoside linkages (other than phosphodiester and phosphorothioate), for example alkyl phosphonate/methyl phosphonate internucleoside linkages, which according to EP 2 742 135 may for example be tolerated in an otherwise DNA phosphorothioate gap region.


Nucleobase The term nucleobase includes the purine (e.g. adenine and guanine) and pyrimidine (e.g. uracil, thymine and cytosine) moiety present in nucleosides and nucleotides which form hydrogen bonds in nucleic acid hybridization. In the context of the present invention the term nucleobase also encompasses modified nucleobases which may differ from naturally occurring nucleobases, but are functional during nucleic acid hybridization. In this context “nucleobase” refers to both naturally occurring nucleobases such as adenine, guanine, cytosine, thymidine, uracil, xanthine and hypoxanthine, as well as non-naturally occurring variants. Such variants are for example described in Hirao et al (2012) Accounts of Chemical Research vol 45 page 2055 and Bergstrom (2009) Current Protocols in Nucleic Acid Chemistry Suppl. 37 1.4.1.


In a some embodiments the nucleobase moiety is modified by changing the purine or pyrimidine into a modified purine or pyrimidine, such as substituted purine or substituted pyrimidine, such as a nucleobased selected from isocytosine, pseudoisocytosine, 5-methyl cytosine, 5-thiozolo-cytosine, 5-propynyl-cytosine, 5-propynyl-uracil, 5-bromouracil 5-thiazolo-uracil, 2-thio-uracil, 2′ thio-thymine, inosine, diaminopurine, 6-aminopurine, 2-aminopurine, 2,6-diaminopurine and 2-chloro-6-am inopuri ne.


The nucleobase moieties may be indicated by the letter code for each corresponding nucleobase, e.g. A, T, G, C or U, wherein each letter may optionally include modified nucleobases of equivalent function. For example, in the exemplified oligonucleotides, the nucleobase moieties are selected from A, T, G, C, and 5-methyl cytosine. Optionally, for LNA gapmers, 5-methyl cytosine LNA nucleosides may be used.


Modified oligonucleotide The term “modified oligonucleotide” describes an oligonucleotide comprising one or more sugar-modified nucleosides and/or modified internucleoside linkages. The term chimeric” oligonucleotide is a term that has been used in the literature to describe oligonucleotides comprising modified nucleosides and DNA nucleosides. The antisense oligonucleotide of the invention is advantageously a chimeric oligonucleotide.


Complementarity The term “complementarity” or “complementary” describes the capacity for Watson-Crick base-pairing of nucleosides/nucleotides. Watson-Crick base pairs are guanine (G)-cytosine (C) and adenine (A) - thymine (T)/uracil (U). It will be understood that oligonucleotides may comprise nucleosides with modified nucleobases, for example 5-methyl cytosine is often used in place of cytosine, and as such the term complementarity encompasses Watson Crick base-paring between non-modified and modified nucleobases (see for example Hirao et al (2012) Accounts of Chemical Research vol 45 page 2055 and Bergstrom (2009) Current Protocols in Nucleic Acid Chemistry Suppl. 37 1.4.1).


The term “% complementary” as used herein, refers to the proportion of nucleotides (in percent) of a contiguous nucleotide sequence in a nucleic acid molecule (e.g. oligonucleotide) which across the contiguous nucleotide sequence, are complementary to a reference sequence (e.g. a target sequence or sequence motif). The percentage of complementarity is thus calculated by counting the number of aligned nucleobases that are complementary (from Watson Crick base pair) between the two sequences (when aligned with the target sequence 5′-3′ and the oligonucleotide sequence from 3′-5′), dividing that number by the total number of nucleotides in the oligonucleotide and multiplying by 100. In such a comparison a nucleobase/nucleotide which does not align (form a base pair) is termed a mismatch. Insertions and deletions are not allowed in the calculation of % complementarity of a contiguous nucleotide sequence. It will be understood that in determining complementarity, chemical modifications of the nucleobases are disregarded as long as the functional capacity of the nucleobase to form Watson Crick base pairing is retained (e.g. 5′-methyl cytosine is considered identical to a cytosine for the purpose of calculating % identity).


The term “fully complementary”, refers to 100% complementarity.


Identity The term “Identity” as used herein, refers to the proportion of nucleotides (expressed in percent) of a contiguous nucleotide sequence in a nucleic acid molecule (e.g. oligonucleotide) which across the contiguous nucleotide sequence, are identical to a reference sequence (e.g. a sequence motif). The percentage of identity is thus calculated by counting the number of aligned nucleobases that are identical (a Match) between two sequences (in the contiguous nucleotide sequence of the compound of the invention and in the reference sequence), dividing that number by the total number of nucleotides in the oligonucleotide and multiplying by 100. Therefore, Percentage of Identity=(Matches x 100)/Length of aligned region (e.g. the contiguous nucleotide sequence). Insertions and deletions are not allowed in the calculation the percentage of identity of a contiguous nucleotide sequence. It will be understood that in determining identity, chemical modifications of the nucleobases are disregarded as long as the functional capacity of the nucleobase to form Watson Crick base pairing is retained (e.g. 5-methyl cytosine is considered identical to a cytosine for the purpose of calculating % identity).


Hybridization The term “hybridizing” or “hybridizes” as used herein is to be understood as two nucleic acid strands (e.g. an oligonucleotide and a target nucleic acid) forming hydrogen bonds between base pairs on opposite strands thereby forming a duplex. The affinity of the binding between two nucleic acid strands is the strength of the hybridization. It is often described in terms of the melting temperature (Tm) defined as the temperature at which half of the oligonucleotides are duplexed with the target nucleic acid. At physiological conditions Tm is not strictly proportional to the affinity (Mergny and Lacroix, 2003, Oligonucleotides 13:515-537). The standard state Gibbs free energy ΔG° is a more accurate representation of binding affinity and is related to the dissociation constant (Kd) of the reaction by ΔG°=-RTIn(Kd), where R is the gas constant and T is the absolute temperature. Therefore, a very low ΔG° of the reaction between an oligonucleotide and the target nucleic acid reflects a strong hybridization between the oligonucleotide and target nucleic acid. ΔG° is the energy associated with a reaction where aqueous concentrations are 1M, the pH is 7, and the temperature is 37° C. The hybridization of oligonucleotides to a target nucleic acid is a spontaneous reaction and for spontaneous reactions ΔG° is less than zero. ΔG° can be measured experimentally, for example, by use of the isothermal titration calorimetry (ITC) method as described in Hansen et al., 1965, Chem, Comm, 36-38 and Holdgate et al., 2005, Drug Discov Today. The skilled person will know that commercial equipment is available for ΔG° measurements. ΔG° can also be estimated numerically by using the nearest neighbor model as described by SantaLucia, 1998, Proc Natl Acad Sci USA. 95: 1460-1465 using appropriately derived thermodynamic parameters described by Sugimoto et al., 1995, Biochemistry 34:11211-11216 and McTigue et al., 2004, Biochemistry 43:5388-5405. In order to have the possibility of modulating its intended nucleic acid target by hybridization, oligonucleotides of the present invention hybridize to a target nucleic acid with estimated ΔG° values below −10 kcal for oligonucleotides that are 10 to30 nucleotides in length. In some embodiments, the degree or strength of hybridization is measured by the standard state Gibbs free energy ΔG°. The oligonucleotides may hybridize to a target nucleic acid with estimated ΔG° values below −10 kcal, such as below −15 kcal, such as below −20 kcal and such as below −25 kcal for oligonucleotides that are 8 to30 nucleotides in length. In some embodiments, the oligonucleotides hybridize to a target nucleic acid with an estimated ΔG° value of −10 to −60 kcal, such as in the range of −12 to −40, such as from −15 to −30 kcal or −16 to −27 kcal such as −18 to −25 kcal.


Target Nucleic Acid


According to the present invention, the target nucleic acid is a nucleic acid which encodes mammalian COPS3 and may for example be a gene, a RNA, a mRNA, and pre-mRNA, a mature mRNA or a cDNA sequence. The target may therefore be referred to as COPS3 target nucleic acid. Suitably, the target nucleic acid encodes a COPS3 protein, in particular mammalian COPS3, such as the human COPS3 gene encoding pre-mRNA or mRNA sequences provided herein as SEQ ID NO: 1, 4, 5, 6, 7, 8 and/or 9.


The therapeutic nucleic acid molecules of the invention may for example target exon regions of a mammalian COPS3 (in particular siRNA and shRNA, but also antisense oligonucleotides), or may for example target any intron region in the COPS3 pre-mRNA (in particular antisense oligonucleotides). The human COPS3 gene encodes 18 transcripts, six of which are protein coding and therefore potential nucleic acid targets.


Table 1 lists predicted exon and intron regions of SEQ ID NO: 1, i.e. of the human COPS3 pre-mRNA sequence.









TABLE 1







Exon and intron regions in the human COPS3 pre-mRNA.









Exonic regions in the
Intronic regions in the



human COPS3 premRNA
human COPS3 premRNA


(SEQ ID NO: 1)
(SEQ ID NO: 1)












ID
start
end
ID
start
end















E1
28
142
I1
143
5109


E2
5110
5239
I2
5240
10265


E3
10266
10378
I3
10379
10466


E4
10467
10516
I4
10517
13296


E5
13297
13389
I5
13390
16292


E6
16293
16472
I6
16473
19167


E7
19168
19308
I7
19309
20799


E8
20800
20973
I8
20974
26328


E9
26329
26415
I9
26416
32234


E10
32235
32348
I10
32349
33713


E11
33714
33794
I11
33795
34122


E12
34123
34658









Suitably, the target nucleic acid encodes a COPS3 protein, in particular mammalian COPS3, such as human COPS3 (See for example Table 2 and Table 3) which provides an overview on the genomic sequences of human, cyno monkey and mouse COPS3 (Table 2) and on pre-mRNA sequences for human, monkey and mouse COPS3 and for the mature mRNAs for human COPS3 (Table 3).


In some embodiments, the target nucleic acid is selected from the group consisting of SEQ ID NO: 1, 2, 3, 4, 6, 7, 8, and/or 9, or naturally occurring variants thereof (e.g. sequences encoding a mammalian COPS3).









TABLE 2







Genome and assembly information for COPS3 across species.















Genomic coordinates

ensembl













Species
Chr.
Stand
Start
End
Assembly
gene_id





Human
17
Rv
17246616
17281273
GRCh38.p12
ENSG00000141030


Cyno
160qq
Rv
17560038
17601679
Macaca_fascicularis_5.0
ENSMFAG00000001157


monkey








Mouse
11
Rv
59817795
59839838
GRCm38.p4
ENSMUSG00000019373





Fwd = forward strand. Rv = reverse strand. The genome coordinates provide the pre-mRNA sequence (genomic sequence).






If employing the nucleic acid molecule of the invention in research or diagnostics the target nucleic acid may be a cDNA or a synthetic nucleic acid derived from DNA or RNA.


For in vivo or in vitro application, the therapeutic nucleic acid molecule of the invention is typically capable of inhibiting the expression of the COPS3 target nucleic acid in a cell which is expressing the COPS3 target nucleic acid. In some embodiments, said cell comprises HBV cccDNA. The contiguous sequence of nucleobases of the nucleic acid molecule of the invention is typically complementary to a conserved region of the COPS3 target nucleic acid, as measured across the length of the nucleic acid molecule, optionally with the exception of one or two mismatches, and optionally excluding nucleotide based linker regions which may link the oligonucleotide to an optional functional group such as a conjugate, or other non-complementary terminal nucleotides. The target nucleic acid is a messenger RNA, such as a pre-mRNA which encodes mammalian COPS3 protein, such as human COPS3, e.g. the human COPS3 pre-mRNA sequence, such as that disclosed as SEQ ID NO: 1, the monkey COPS3 pre-mRNA sequence, such as that disclosed as SEQ ID NO: 2, or the mouse COPS3 pre-mRNA sequence, such as that disclosed as SEQ ID NO: 3, or a mature COPS3 mRNA, such as that a human mature mRNA disclosed as SEQ ID NO: 4, 5, 6, 7, 8 or 9. SEQ ID NOs: 1-9 are DNA sequences—it will be understood that target RNA sequences have uracil (U) bases in place of the thymidine bases (T).


Further information on exemplary target nucleic acids is provided in Tables 2 and 3.









TABLE 3







Overview on target nucleic acids.










Target Nucleic Acid, Species, Reference
Sequence ID







COPS3 Homo sapiens pre-mRNA
SEQ ID NO: 1



COPS3 Macaca fascicularis pre-mRNA
SEQ ID NO: 2



COPS3 Mus musculus pre-mRNA
SEQ ID NO: 3



COPS3 Homo sapiens mature mRNA,
SEQ ID NO: 4



variant 1 (ENST00000268717.10)



COPS3 Homo sapiens mature mRNA,
SEQ ID NO: 5



variant 2 (ENST00000539941.6)



COPS3 Homo sapiens mature mRNA,
SEQ ID NO: 6



variant 3 (ENST00000439936.6)



COPS3 Homo sapiens mature mRNA,
SEQ ID NO: 7



variant 4 (ENST00000417352.5)



COPS3 Homo sapiens mature mRNA,
SEQ ID NO: 8



variant 5 (ENST00000579716.2)



COPS3 Homo sapiens mature mRNA,
SEQ ID NO: 9



variant 6 (ENST00000583160.5)










Note SEQ ID NO: 2 comprises regions of multiple NNNNs, where the sequencing has been unable to accurately refine the sequence, and a degenerate sequence is therefore included. For the avoidance of doubt the compounds of the invention are complementary to the actual target sequence and are not therefore degenerate compounds.


In some embodiments, the target nucleic acid is SEQ ID NO: 1.


In some embodiments, the target nucleic acid is SEQ ID NO: 2.


In some embodiments, the target nucleic acid is SEQ ID NO: 3.


In some embodiments, the target nucleic acid is SEQ ID NO: 4.


In some embodiments, the target nucleic acid is SEQ ID NO: 5.


In some embodiments, the target nucleic acid is SEQ ID NO: 6.


In some embodiments, the target nucleic acid is SEQ ID NO: 7.


In some embodiments, the target nucleic acid is SEQ ID NO: 8.


In some embodiments, the target nucleic acid is SEQ ID NO: 9.


In some embodiments, the target nucleic acid is SEQ ID NO: 1, 4, 5, 6 and/or 8.


In some embodiments, the target nucleic acid is SEQ ID NO: 1, 4 and/or 5.


Target Sequence The term “target sequence” as used herein refers to a sequence of nucleotides present in the target nucleic acid which comprises the nucleobase sequence which is complementary to the oligonucleotide or nucleic acid molecule of the invention. In some embodiments, the target sequence consists of a region on the target nucleic acid with a nucleobase sequence that is complementary to the contiguous nucleotide sequence of the oligonucleotide of the invention. This region of the target nucleic acid may interchangeably be referred to as the target nucleotide sequence, target sequence or target region. In some embodiments, the target sequence is longer than the complementary sequence of a nucleic acid molecule of the invention, and may, for example represent a preferred region of the target nucleic acid which may be targeted by several nucleic acid molecules of the invention.


In some embodiments, the target sequence is a sequence selected from the group consisting of a human COPS3 mRNA exon, such as a human COPS3 mRNA exon selected from the group consisting of e1, e2, e3, e4, e5, e6, e7, e8, e9, e10, e11 and e12 (see for example Table 1 above).


Accordingly, the invention provides for an oligonucleotide, wherein said oligonucleotide comprises a contiguous sequence which is at least 90% complementary, such as fully complementary to an exon region of SEQ ID NO: 1, selected from the group consisting of e1— e12 (see Table 1).


In some embodiments, the target sequence is a sequence selected from the group consisting of a human COPS3 mRNA intron, such as a human COPS3 mRNA intron selected from the group consisting of i1, i2, i3, i4, i5, i6, i7, i9, i10 and i11 (see for example Table 1 above).


Accordingly, the invention provides for an oligonucleotide, wherein said oligonucleotide comprises a contiguous sequence which is at least 90% complementary, such as fully complementary to an intron region of SEQ ID NO: 1, selected from the group consisting of i1— i11 (see Table 1).


In some embodiments, the target sequence is selected from the group consisting of SEQ ID NOs: 10, 11, 12 and 13. In some embodiments, the contiguous nucleotide sequence as referred to herein is at least 90% complementary, such as at least 95% complementary to a target sequence selected from the group consisting of SEQ ID NOs: 10, 11, 12 and 13. In some embodiments, the contiguous nucleotide sequence is fully complementary to a target sequence selected from the group consisting of SEQ ID NOs: 10, 11, 12 and 13.


The nucleic acid molecule of the invention comprises a contiguous nucleotide sequence which is complementary to or hybridizes to a region on the target nucleic acid, such as a target sequence described herein.


The target nucleic acid sequence to which the therapeutic nucleic acid molecule is complementary or hybridizes to generally comprises a stretch of contiguous nucleobases of at least 10 nucleotides. The contiguous nucleotide sequence is between 12 to 70 nucleotides, such as 12 to 50, such as 13 to 30, such as 14 to 25, such as 15 to 20, such as 16 to 18 contiguous nucleotides.


In some embodiments, the nucleic acid molecule of the present invention targets a region shown in Table 4 or 5.









TABLE 4







Exemplary target regions









Target
start SEQ
end SEQ


region
ID NO: 1
ID NO: 1












 1A
10
53


 2A
64
125


 3A
157
185


 4A
197
217


 5A
229
245


 6A
269
286


 7A
288
308


 8A
337
405


 9A
414
451


 10A
455
474


 11A
476
520


 12A
543
580


 13A
623
654


 14A
656
704


 15A
736
755


 16A
743
777


 17A
744
759


 18A
745
759


 19A
746
760


 20A
746
765


 21A
764
778


 22A
765
791


 23A
793
815


 24A
795
815


 25A
803
820


 26A
809
830


 27A
829
845


 28A
833
849


 29A
857
873


 30A
861
886


 31A
861
885


 32A
861
875


 33A
861
883


 34A
863
879


 35A
863
885


 36A
880
898


 37A
890
905


 38A
894
915


 39A
894
914


 40A
901
916


 41A
902
916


 42A
903
917


 43A
947
964


 44A
948
972


 45A
950
972


 46A
952
972


 47A
960
977


 48A
966
987


 49A
966
988


 50A
976
1005


 51A
1002
1017


 52A
1004
1050


 53A
1004
1031


 54A
1005
1065


 55A
1005
1033


 56A
1011
1027


 57A
1013
1031


 58A
1013
1046


 59A
1017
1050


 60A
1022
1050


 61A
1033
1054


 62A
1034
1054


 63A
1076
1101


 64A
1092
1133


 65A
1100
1119


 66A
1100
1133


 67A
1104
1126


 68A
1109
1132


 69A
1116
1133


 70A
1126
1153


 71A
1163
1177


 72A
1173
1187


 73A
1175
1192


 74A
1185
1201


 75A
1235
1271


 76A
1236
1257


 77A
1239
1259


 78A
1261
1283


 79A
1262
1276


 80A
1272
1287


 81A
1289
1303


 82A
1305
1328


 83A
1344
1364


 84A
1403
1426


 85A
1438
1467


 86A
1483
1509


 87A
1537
1558


 88A
1562
1592


 89A
1594
1610


 90A
1631
1650


 91A
1652
1705


 92A
1707
1733


 93A
1746
1767


 94A
1769
1795


 95A
1803
1825


 96A
1848
1883


 97A
1885
1904


 98A
1925
1958


 99A
1960
1983


100A
1987
2014


101A
2016
2032


102A
2034
2053


103A
2060
2078


104A
2080
2109


105A
2124
2145


106A
2133
2157


107A
2138
2153


108A
2165
2184


109A
2166
2184


110A
2166
2180


111A
2172
2187


112A
2189
2220


113A
2200
2226


114A
2208
2232


115A
2228
2242


116A
2230
2251


117A
2255
2273


118A
2266
2298


119A
2266
2282


120A
2300
2317


121A
2314
2330


122A
2314
2329


123A
2318
2335


124A
2332
2346


125A
2355
2373


126A
2361
2375


127A
2363
2395


128A
2380
2395


129A
2382
2396


130A
2383
2400


131A
2383
2397


132A
2389
2404


133A
2401
2415


134A
2444
2463


135A
2477
2510


136A
2520
2551


137A
2553
2608


138A
2610
2640


139A
2648
2671


140A
2667
2709


141A
2711
2757


142A
2759
2785


143A
2787
2804


144A
2819
2839


145A
2841
2901


146A
2905
2955


147A
2957
2996


148A
3014
3031


149A
3058
3082


150A
3070
3086


151A
3101
3117


152A
3110
3125


153A
3127
3143


154A
3129
3143


155A
3131
3157


156A
3132
3157


157A
3145
3168


158A
3170
3185


159A
3174
3190


160A
3193
3211


161A
3223
3241


162A
3229
3247


163A
3236
3266


164A
3236
3258


165A
3254
3305


166A
3258
3276


167A
3261
3278


168A
3275
3294


169A
3283
3302


170A
3288
3304


171A
3293
3322


172A
3310
3331


173A
3311
3331


174A
3319
3334


175A
3319
3333


176A
3322
3361


177A
3329
3360


178A
3337
3353


179A
3368
3390


180A
3378
3410


181A
3378
3397


182A
3407
3438


183A
3415
3431


184A
3446
3468


185A
3456
3488


186A
3456
3489


187A
3456
3475


188A
3477
3541


189A
3543
3559


190A
3575
3616


191A
3604
3622


192A
3623
3642


193A
3675
3697


194A
3699
3731


195A
3722
3739


196A
3723
3739


197A
3733
3759


198A
3741
3759


199A
3781
3814


200A
3826
3862


201A
3864
3927


202A
3944
3962


203A
3972
3994


204A
4009
4053


205A
4055
4077


206A
4101
4122


207A
4127
4155


208A
4162
4219


209A
4226
4270


210A
4272
4287


211A
4314
4339


212A
4341
4364


213A
4372
4391


214A
4393
4451


215A
4462
4479


216A
4468
4486


217A
4468
4482


218A
4473
4499


219A
4474
4489


220A
4474
4513


221A
4474
4499


222A
4474
4494


223A
4480
4499


224A
4511
4536


225A
4523
4537


226A
4524
4538


227A
4530
4544


228A
4534
4550


229A
4560
4575


230A
4563
4577


231A
4565
4579


232A
4578
4596


233A
4584
4610


234A
4592
4610


235A
4635
4650


236A
4643
4665


237A
4644
4659


238A
4644
4663


239A
4653
4673


240A
4684
4701


241A
4686
4701


242A
4688
4702


243A
4689
4703


244A
4716
4779


245A
4789
4803


246A
4805
4829


247A
4860
4887


248A
4889
4912


249A
5016
5031


250A
5040
5056


251A
5064
5078


252A
5080
5097


253A
5099
5245


254A
5247
5264


255A
5297
5312


256A
5300
5315


257A
5309
5325


258A
5325
5345


259A
5326
5342


260A
5344
5378


261A
5350
5367


262A
5355
5377


263A
5355
5375


264A
5360
5377


265A
5362
5377


266A
5363
5377


267A
5388
5412


268A
5392
5412


269A
5397
5412


270A
5401
5415


271A
5417
5437


272A
5426
5448


273A
5439
5469


274A
5473
5503


275A
5475
5494


276A
5476
5500


277A
5480
5496


278A
5482
5500


279A
5482
5515


280A
5486
5519


281A
5491
5519


282A
5508
5529


283A
5508
5524


284A
5531
5548


285A
5550
5577


286A
5557
5575


287A
5560
5575


288A
5562
5591


289A
5565
5602


290A
5583
5597


291A
5590
5608


292A
5593
5611


293A
5596
5614


294A
5610
5627


295A
5612
5627


296A
5615
5630


297A
5624
5640


298A
5624
5646


299A
5624
5642


300A
5625
5642


301A
5626
5642


302A
5626
5643


303A
5672
5695


304A
5735
5757


305A
5799
5815


306A
5803
5818


307A
5838
5853


308A
5854
5869


309A
5888
5902


310A
5890
5905


311A
5985
6008


312A
6031
6045


313A
6036
6052


314A
6037
6052


315A
6040
6058


316A
6094
6114


317A
6098
6114


318A
6102
6117


319A
6102
6137


320A
6102
6118


321A
6121
6137


322A
6125
6151


323A
6160
6179


324A
6162
6177


325A
6162
6178


326A
6504
6519


327A
6506
6520


328A
6585
6607


329A
6595
6615


330A
6620
6646


331A
6621
6636


332A
6621
6646


333A
6621
6641


334A
6627
6646


335A
6634
6649


336A
6653
6667


337A
6656
6676


338A
6664
6687


339A
6685
6700


340A
6685
6699


341A
6694
6723


342A
6706
6724


343A
6706
6748


344A
6711
6748


345A
6715
6732


346A
6716
6748


347A
6716
6732


348A
6747
6762


349A
6750
6767


350A
6758
6779


351A
6764
6779


352A
6767
6784


353A
6769
6784


354A
6788
6803


355A
6791
6805


356A
6819
6834


357A
6821
6837


358A
6822
6837


359A
6825
6844


360A
6825
6840


361A
6832
6847


362A
6834
6848


363A
6835
6852


364A
6835
6849


365A
6835
6853


366A
6836
6853


367A
7290
7304


368A
7331
7345


369A
7369
7383


370A
7379
7396


371A
7453
7469


372A
7471
7486


373A
7518
7536


374A
7568
7584


375A
7587
7603


376A
7613
7632


377A
7620
7640


378A
7649
7667


379A
7655
7670


380A
7672
7688


381A
7745
7760


382A
7764
7779


383A
7767
7802


384A
7767
7800


385A
7786
7801


386A
8398
8415


387A
8556
8570


388A
8564
8583


389A
8571
8587


390A
8576
8590


391A
8602
8617


392A
8605
8620


393A
8608
8629


394A
8608
8623


395A
8842
8858


396A
8874
8894


397A
8879
8896


398A
8881
8896


399A
8882
8896


400A
8887
8901


401A
8895
8914


402A
8898
8913


403A
8916
8946


404A
8916
8936


405A
8934
8957


406A
8936
8957


407A
8940
8957


408A
8946
8961


409A
8946
8960


410A
8964
8978


411A
8980
9005


412A
9007
9021


413A
9009
9025


414A
9015
9033


415A
9037
9056


416A
9058
9108


417A
9084
9099


418A
9089
9108


419A
9110
9128


420A
9134
9150


421A
9138
9154


422A
9180
9201


423A
9180
9195


424A
9180
9198


425A
9192
9208


426A
9237
9265


427A
9253
9343


428A
9414
9428


429A
9416
9433


430A
9446
9466


431A
9468
9489


432A
9469
9486


433A
9476
9497


434A
9477
9499


435A
9477
9497


436A
9482
9499


437A
9484
9499


438A
9485
9499


439A
9487
9505


440A
9501
9516


441A
9501
9531


442A
9506
9528


443A
9507
9528


444A
9507
9531


445A
9524
9540


446A
9544
9566


447A
9568
9595


448A
9583
9607


449A
9602
9616


450A
9604
9627


451A
9604
9622


452A
9648
9663


453A
9677
9698


454A
9700
9727


455A
9701
9724


456A
9708
9725


457A
9717
9731


458A
9737
9752


459A
9740
9768


460A
9781
9796


461A
9798
9815


462A
9818
9839


463A
9836
9850


464A
9849
9868


465A
9856
9883


466A
9863
9880


467A
9868
9888


468A
9871
9900


469A
9876
9892


470A
9897
9917


471A
9905
9971


472A
9940
9957


473A
9998
10022


474A
10011
10025


475A
10035
10060


476A
10043
10059


477A
10062
10078


478A
10069
10088


479A
10076
10091


480A
10076
10094


481A
10079
10094


482A
10082
10102


483A
10104
10118


484A
10106
10175


485A
10178
10197


486A
10208
10247


487A
10249
10301


488A
10303
10325


489A
10339
10443


490A
10445
10552


491A
10554
10574


492A
10599
10654


493A
10685
10701


494A
10722
10745


495A
10749
10770


496A
10776
10819


497A
10821
10847


498A
10849
10865


499A
10908
10926


500A
10930
10945


501A
10942
10958


502A
10955
10975


503A
10991
11056


504A
11069
11104


505A
11118
11136


506A
11118
11133


507A
11138
11159


508A
11147
11162


509A
11148
11162


510A
11172
11193


511A
11187
11208


512A
11208
11289


513A
11208
11223


514A
11213
11231


515A
11213
11235


516A
11277
11306


517A
11280
11305


518A
11294
11310


519A
11306
11321


520A
11311
11326


521A
11311
11329


522A
11320
11338


523A
11340
11361


524A
11340
11366


525A
11350
11365


526A
11369
11399


527A
11370
11384


528A
11377
11395


529A
11387
11421


530A
11423
11437


531A
11439
11455


532A
11459
11503


533A
11524
11541


534A
11543
11561


535A
11574
11594


536A
11609
11626


537A
11656
11681


538A
11683
11750


539A
11696
11710


540A
11699
11720


541A
11710
11728


542A
11722
11745


543A
11724
11746


544A
11725
11740


545A
11764
11790


546A
11774
11792


547A
11774
11796


548A
11778
11805


549A
11780
11804


550A
11781
11798


551A
11793
11807


552A
11812
11842


553A
11832
11849


554A
11837
11861


555A
11840
11854


556A
11849
11887


557A
11850
11868


558A
11850
11865


559A
11857
11873


560A
11867
11884


561A
11875
11892


562A
11893
11911


563A
11899
11913


564A
11902
11923


565A
11915
11934


566A
11940
11955


567A
11943
11959


568A
11944
11961


569A
11947
11968


570A
11949
11968


571A
11956
11975


572A
11963
11979


573A
11967
11981


574A
11984
12002


575A
11984
11998


576A
11988
12003


577A
11990
12036


578A
11990
12017


579A
11991
12019


580A
11997
12013


581A
11999
12017


582A
11999
12032


583A
12003
12036


584A
12008
12036


585A
12024
12038


586A
12070
12092


587A
12094
12128


588A
12102
12116


589A
12128
12143


590A
12131
12145


591A
12169
12195


592A
12197
12218


593A
12206
12221


594A
12258
12325


595A
12328
12405


596A
12405
12422


597A
12410
12426


598A
12440
12456


599A
12449
12467


600A
12464
12489


601A
12477
12499


602A
12477
12498


603A
12478
12499


604A
12478
12498


605A
12485
12500


606A
12486
12500


607A
12487
12501


608A
12487
12506


609A
12499
12513


610A
12501
12523


611A
12502
12517


612A
12511
12529


613A
12520
12535


614A
12524
12550


615A
12528
12550


616A
12538
12570


617A
12538
12553


618A
12546
12565


619A
12551
12569


620A
12551
12573


621A
12558
12575


622A
12567
12581


623A
12590
12608


624A
12590
12604


625A
12594
12609


626A
12596
12623


627A
12603
12619


628A
12605
12623


629A
12654
12671


630A
12659
12677


631A
12662
12677


632A
12685
12714


633A
12702
12762


634A
12717
12731


635A
12786
12800


636A
12802
12846


637A
12848
12874


638A
12876
12916


639A
12918
12936


640A
12938
12982


641A
12984
13005


642A
13015
13126


643A
13128
13144


644A
13146
13223


645A
13225
13396


646A
13422
13439


647A
13441
13471


648A
13486
13509


649A
13512
13529


650A
13531
13560


651A
13569
13589


652A
13610
13624


653A
13618
13642


654A
13626
13640


655A
13629
13649


656A
13630
13652


657A
13664
13687


658A
13664
13681


659A
13666
13681


660A
13667
13681


661A
13683
13698


662A
13683
13742


663A
13691
13721


664A
13706
13722


665A
13730
13744


666A
13780
13804


667A
13784
13800


668A
13786
13804


669A
13792
13818


670A
13805
13826


671A
13806
13826


672A
13814
13828


673A
13816
13831


674A
13816
13833


675A
13836
13854


676A
13842
13857


677A
13853
13883


678A
13865
13880


679A
13865
13883


680A
13868
13883


681A
13871
13904


682A
13892
13907


683A
13900
13914


684A
13913
13931


685A
13917
13931


686A
13925
13946


687A
13934
13958


688A
13934
13948


689A
13934
13956


690A
13936
13952


691A
13936
13958


692A
13947
13966


693A
13950
13966


694A
13952
13966


695A
13960
13978


696A
13983
13998


697A
13983
14016


698A
13983
14010


699A
13992
14016


700A
13996
14016


701A
14001
14016


702A
14005
14019


703A
14005
14029


704A
14018
14069


705A
14032
14054


706A
14036
14062


707A
14051
14065


708A
14086
14100


709A
14088
14106


710A
14121
14151


711A
14133
14149


712A
14142
14161


713A
14165
14198


714A
14168
14183


715A
14168
14186


716A
14168
14196


717A
14171
14186


718A
14174
14193


719A
14186
14209


720A
14189
14203


721A
14283
14299


722A
14293
14313


723A
14301
14319


724A
14301
14316


725A
14316
14337


726A
14321
14338


727A
14348
14373


728A
14375
14391


729A
14387
14412


730A
14421
14435


731A
14451
14468


732A
14462
14481


733A
14469
14485


734A
14473
14487


735A
14481
14495


736A
14483
14509


737A
14484
14509


738A
14497
14518


739A
14498
14518


740A
14506
14522


741A
14506
14525


742A
14521
14535


743A
14527
14557


744A
14545
14566


745A
14545
14569


746A
14571
14589


747A
14571
14593


748A
14613
14629


749A
14616
14643


750A
14617
14645


751A
14623
14639


752A
14625
14643


753A
14625
14656


754A
14630
14657


755A
14633
14658


756A
14639
14657


757A
14661
14680


758A
14661
14684


759A
14672
14688


760A
14702
14719


761A
14707
14722


762A
14707
14725


763A
14707
14741


764A
14707
14735


765A
14710
14725


766A
14713
14732


767A
14749
14778


768A
14804
14830


769A
14832
14865


770A
14867
14884


771A
14893
14907


772A
14909
14934


773A
14936
14967


774A
14995
15014


775A
15016
15032


776A
15040
15060


777A
15062
15079


778A
15105
15137


779A
15139
15175


780A
15185
15225


781A
15227
15263


782A
15269
15318


783A
15325
15342


784A
15345
15362


785A
15353
15371


786A
15389
15408


787A
15428
15448


788A
15450
15481


789A
15489
15505


790A
15507
15522


791A
15524
15576


792A
15578
15592


793A
15586
15606


794A
15594
15622


795A
15641
15655


796A
15643
15662


797A
15649
15667


798A
15650
15668


799A
15671
15685


800A
15675
15701


801A
15679
15702


802A
15682
15702


803A
15689
15732


804A
15690
15713


805A
15715
15732


806A
15728
15745


807A
15730
15745


808A
15733
15747


809A
15733
15754


810A
15733
15750


811A
15733
15749


812A
15734
15749


813A
15741
15758


814A
15742
15758


815A
15762
15788


816A
15783
15802


817A
15784
15802


818A
15790
15805


819A
15793
15810


820A
15795
15810


821A
15814
15829


822A
15814
15833


823A
15821
15852


824A
15856
15871


825A
15856
15872


826A
15859
15873


827A
15860
15874


828A
15888
15921


829A
15926
15951


830A
15956
15998


831A
16000
16037


832A
16039
16109


833A
16111
16162


834A
16164
16218


835A
16232
16288


836A
16288
16339


837A
16341
16387


838A
16389
16417


839A
16419
16478


840A
16479
16510


841A
16513
16528


842A
16530
16554


843A
16565
16582


844A
16584
16600


845A
16624
16654


846A
16656
16679


847A
16684
16710


848A
16727
16789


849A
16801
16848


850A
16850
16887


851A
16891
16905


852A
16907
16963


853A
16989
17022


854A
17029
17043


855A
17065
17086


856A
17088
17113


857A
17122
17152


858A
17162
17192


859A
17194
17210


860A
17212
17230


861A
17232
17257


862A
17262
17278


863A
17334
17349


864A
17366
17393


865A
17469
17512


866A
17471
17491


867A
17477
17493


868A
17478
17495


869A
17479
17504


870A
17480
17495


871A
17498
17513


872A
17515
17532


873A
17520
17540


874A
17534
17552


875A
17541
17557


876A
17555
17594


877A
17569
17585


878A
17572
17587


879A
17594
17612


880A
17608
17645


881A
17608
17624


882A
17647
17668


883A
17647
17665


884A
17649
17667


885A
17670
17696


886A
17675
17690


887A
17684
17698


888A
17699
17715


889A
17706
17720


890A
17711
17725


891A
17717
17738


892A
17732
17751


893A
17737
17761


894A
17739
17762


895A
17740
17759


896A
17742
17757


897A
17742
17758


898A
17750
17764


899A
17751
17768


900A
17751
17765


901A
17751
17769


902A
17752
17769


903A
17786
17813


904A
17815
17837


905A
17816
17831


906A
17817
17831


907A
17839
17859


908A
17853
17872


909A
17860
17875


910A
17860
17882


911A
17884
17911


912A
17899
17916


913A
17928
17943


914A
17931
17945


915A
17931
17948


916A
17931
17947


917A
17932
17947


918A
17939
17953


919A
17952
17966


920A
17955
17970


921A
17971
17986


922A
17974
17988


923A
17992
18006


924A
18000
18018


925A
18000
18026


926A
18008
18029


927A
18009
18031


928A
18009
18029


929A
18014
18031


930A
18016
18031


931A
18017
18031


932A
18022
18036


933A
18046
18066


934A
18051
18066


935A
18055
18069


936A
18071
18091


937A
18097
18112


938A
18107
18125


939A
18114
18128


940A
18119
18140


941A
18119
18137


942A
18130
18169


943A
18137
18155


944A
18137
18168


945A
18142
18169


946A
18145
18170


947A
18151
18169


948A
18158
18174


949A
18163
18184


950A
18163
18188


951A
18163
18179


952A
18176
18230


953A
18176
18204


954A
18186
18203


955A
18203
18219


956A
18232
18255


957A
18291
18309


958A
18293
18308


959A
18316
18338


960A
18317
18332


961A
18317
18336


962A
18379
18394


963A
18403
18422


964A
18534
18550


965A
18534
18587


966A
18534
18556


967A
18566
18588


968A
18566
18587


969A
18567
18588


970A
18567
18587


971A
18574
18589


972A
18575
18589


973A
18591
18606


974A
18591
18618


975A
18606
18627


976A
18632
18654


977A
18634
18653


978A
18638
18654


979A
18639
18656


980A
18641
18669


981A
18641
18671


982A
18642
18663


983A
18642
18671


984A
18644
18663


985A
18644
18668


986A
18659
18673


987A
18659
18683


988A
18677
18699


989A
18682
18698


990A
18694
18708


991A
18696
18714


992A
18696
18727


993A
18718
18740


994A
18754
18777


995A
18786
18808


996A
18793
18809


997A
18810
18826


998A
18833
18866


999A
18883
18904


1000A
18910
18934


1001A
18936
18951


1002A
18968
19003


1003A
18983
18997


1004A
19006
19022


1005A
19013
19027


1006A
19042
19062


1007A
19083
19113


1008A
19115
19145


1009A
19160
19208


1010A
19210
19277


1011A
19279
19319


1012A
19320
19358


1013A
19360
19390


1014A
19479
19520


1015A
19523
19551


1016A
19553
19570


1017A
19563
19577


1018A
19581
19596


1019A
19594
19613


1020A
19615
19645


1021A
19618
19632


1022A
19647
19670


1023A
19658
19679


1024A
19667
19685


1025A
19673
19709


1026A
19724
19740


1027A
19724
19742


1028A
19725
19742


1029A
19726
19742


1030A
19728
19744


1031A
19728
19746


1032A
19729
19746


1033A
19730
19746


1034A
19751
19766


1035A
19776
19798


1036A
19776
19793


1037A
19782
19796


1038A
19792
19815


1039A
19795
19810


1040A
19803
19833


1041A
19817
19843


1042A
19821
19843


1043A
19831
19846


1044A
19848
19874


1045A
19862
19876


1046A
19869
19886


1047A
19876
19919


1048A
19923
19945


1049A
19959
20019


1050A
19975
19995


1051A
19985
20004


1052A
20000
20014


1053A
20007
20030


1054A
20014
20028


1055A
20039
20067


1056A
20070
20129


1057A
20136
20169


1058A
20184
20217


1059A
20436
20451


1060A
20454
20471


1061A
20459
20473


1062A
20459
20481


1063A
20461
20477


1064A
20483
20513


1065A
20493
20514


1066A
20494
20516


1067A
20494
20514


1068A
20499
20516


1069A
20501
20516


1070A
20502
20516


1071A
20507
20521


1072A
20517
20536


1073A
20519
20536


1074A
20524
20545


1075A
20535
20563


1076A
20535
20556


1077A
20557
20578


1078A
20565
20581


1079A
20566
20583


1080A
20569
20590


1081A
20571
20590


1082A
20578
20597


1083A
20578
20606


1084A
20618
20634


1085A
20622
20649


1086A
20637
20653


1087A
20652
20669


1088A
20698
20718


1089A
20706
20721


1090A
20718
20733


1091A
20723
20737


1092A
20746
20764


1093A
20774
20891


1094A
20917
20983


1095A
20985
21010


1096A
21012
21040


1097A
21054
21114


1098A
21116
21140


1099A
21142
21212


1100A
21214
21286


1101A
21312
21332


1102A
21339
21354


1103A
21380
21412


1104A
21414
21428


1105A
21444
21474


1106A
21462
21476


1107A
21464
21479


1108A
21465
21479


1109A
21472
21489


1110A
21472
21490


1111A
21478
21520


1112A
21492
21506


1113A
21504
21519


1114A
21508
21526


1115A
21514
21535


1116A
21523
21542


1117A
21531
21546


1118A
21558
21574


1119A
21593
21608


1120A
21598
21624


1121A
21626
21640


1122A
21673
21694


1123A
21696
21779


1124A
21742
21762


1125A
21779
21799


1126A
21815
21830


1127A
21837
21855


1128A
21873
21907


1129A
21909
21940


1130A
21954
21997


1131A
22011
22032


1132A
22037
22083


1133A
22104
22118


1134A
22130
22173


1135A
22175
22211


1136A
22250
22267


1137A
22269
22326


1138A
22336
22350


1139A
22362
22376


1140A
22383
22420


1141A
22420
22442


1142A
22444
22458


1143A
22460
22481


1144A
22483
22501


1145A
22503
22552


1146A
22572
22615


1147A
22635
22662


1148A
22677
22700


1149A
22702
22797


1150A
22803
22822


1151A
22813
22830


1152A
22830
22846


1153A
22833
22847


1154A
22833
22849


1155A
22834
22849


1156A
22854
22873


1157A
22857
22872


1158A
22890
22906


1159A
22890
22911


1160A
22897
22913


1161A
22898
22913


1162A
22899
22914


1163A
22902
22918


1164A
22908
22931


1165A
22913
22929


1166A
22917
22931


1167A
22942
22964


1168A
22958
22974


1169A
22975
22998


1170A
23000
23023


1171A
23033
23059


1172A
23083
23116


1173A
23132
23175


1174A
23177
23197


1175A
23199
23245


1176A
23259
23293


1177A
23291
23316


1178A
23318
23369


1179A
23378
23394


1180A
23401
23445


1181A
23457
23477


1182A
23461
23476


1183A
23465
23489


1184A
23465
23479


1185A
23465
23487


1186A
23467
23483


1187A
23467
23489


1188A
23478
23497


1189A
23481
23497


1190A
23483
23497


1191A
23485
23500


1192A
23496
23511


1193A
23500
23520


1194A
23522
23545


1195A
23533
23551


1196A
23547
23589


1197A
23553
23584


1198A
23557
23589


1199A
23562
23582


1200A
23573
23595


1201A
23577
23603


1202A
23591
23605


1203A
23591
23606


1204A
23610
23628


1205A
23630
23657


1206A
23630
23660


1207A
23648
23668


1208A
23670
23706


1209A
23677
23694


1210A
23694
23710


1211A
23698
23735


1212A
23708
23723


1213A
23708
23726


1214A
23711
23726


1215A
23714
23733


1216A
23723
23737


1217A
23740
23759


1218A
23759
23815


1219A
23836
23850


1220A
23889
23908


1221A
23896
23911


1222A
23896
23910


1223A
23917
23932


1224A
23930
23948


1225A
23936
23955


1226A
23946
23960


1227A
23975
23995


1228A
23983
24016


1229A
23993
24008


1230A
24012
24036


1231A
24013
24030


1232A
24024
24038


1233A
24024
24039


1234A
24024
24078


1235A
24043
24064


1236A
24043
24061


1237A
24058
24074


1238A
24060
24078


1239A
24091
24106


1240A
24091
24108


1241A
24091
24111


1242A
24092
24111


1243A
24099
24115


1244A
24100
24115


1245A
24102
24121


1246A
24103
24123


1247A
24114
24143


1248A
24127
24141


1249A
24143
24158


1250A
24143
24161


1251A
24149
24165


1252A
24167
24192


1253A
24176
24192


1254A
24202
24234


1255A
24203
24219


1256A
24244
24263


1257A
24328
24356


1258A
24358
24440


1259A
24477
24492


1260A
24496
24513


1261A
24518
24532


1262A
24524
24538


1263A
24524
24576


1264A
24564
24581


1265A
24614
24655


1266A
24643
24665


1267A
24644
24658


1268A
24656
24698


1269A
24700
24727


1270A
24739
24757


1271A
24762
24778


1272A
24796
24816


1273A
24828
24854


1274A
24856
24878


1275A
24880
24900


1276A
24902
24918


1277A
24941
24980


1278A
25001
25025


1279A
25072
25086


1280A
25074
25093


1281A
25098
25112


1282A
25114
25152


1283A
25117
25139


1284A
25117
25138


1285A
25118
25139


1286A
25118
25138


1287A
25123
25145


1288A
25123
25140


1289A
25129
25143


1290A
25156
25176


1291A
25165
25179


1292A
25171
25202


1293A
25180
25200


1294A
25183
25205


1295A
25185
25204


1296A
25189
25205


1297A
25190
25207


1298A
25193
25214


1299A
25195
25214


1300A
25195
25219


1301A
25207
25223


1302A
25221
25250


1303A
25226
25250


1304A
25244
25260


1305A
25248
25268


1306A
25254
25268


1307A
25256
25274


1308A
25256
25289


1309A
25277
25293


1310A
25291
25311


1311A
25313
25331


1312A
25335
25349


1313A
25339
25361


1314A
25349
25365


1315A
25356
25370


1316A
25358
25375


1317A
25368
25386


1318A
25368
25382


1319A
25393
25410


1320A
25394
25410


1321A
25396
25412


1322A
25396
25414


1323A
25397
25414


1324A
25398
25414


1325A
25400
25416


1326A
25400
25418


1327A
25401
25418


1328A
25402
25418


1329A
25404
25420


1330A
25404
25422


1331A
25405
25422


1332A
25406
25422


1333A
25408
25424


1334A
25408
25426


1335A
25409
25426


1336A
25410
25426


1337A
25412
25428


1338A
25412
25430


1339A
25413
25430


1340A
25414
25430


1341A
25416
25432


1342A
25416
25434


1343A
25417
25434


1344A
25418
25434


1345A
25418
25435


1346A
25432
25455


1347A
25433
25447


1348A
25445
25471


1349A
25492
25523


1350A
25525
25544


1351A
25543
25564


1352A
25548
25564


1353A
25552
25566


1354A
25552
25572


1355A
25560
25580


1356A
25592
25610


1357A
25616
25635


1358A
25623
25638


1359A
25661
25682


1360A
25670
25695


1361A
25684
25708


1362A
25693
25708


1363A
25760
25788


1364A
25792
25808


1365A
25803
25822


1366A
25810
25830


1367A
25835
25852


1368A
25840
25862


1369A
25841
25862


1370A
25852
25873


1371A
25861
25891


1372A
25891
25905


1373A
25893
25921


1374A
25894
25915


1375A
25896
25915


1376A
25896
25920


1377A
25928
25942


1378A
25950
25967


1379A
25956
25979


1380A
25957
25974


1381A
25967
25987


1382A
25992
26009


1383A
26028
26049


1384A
26030
26048


1385A
26039
26063


1386A
26040
26059


1387A
26051
26075


1388A
26053
26075


1389A
26055
26075


1390A
26063
26080


1391A
26069
26083


1392A
26071
26099


1393A
26071
26101


1394A
26072
26093


1395A
26072
26101


1396A
26074
26093


1397A
26074
26098


1398A
26128
26161


1399A
26133
26161


1400A
26150
26166


1401A
26169
26185


1402A
26187
26201


1403A
26192
26206


1404A
26199
26216


1405A
26204
26219


1406A
26204
26222


1407A
26204
26232


1408A
26207
26222


1409A
26210
26229


1410A
26219
26233


1411A
26236
26254


1412A
26239
26253


1413A
26246
26269


1414A
26282
26342


1415A
26344
26366


1416A
26377
26396


1417A
26404
26422


1418A
26436
26456


1419A
26436
26457


1420A
26440
26455


1421A
26445
26461


1422A
26447
26461


1423A
26451
26465


1424A
26461
26485


1425A
26463
26477


1426A
26473
26489


1427A
26488
26504


1428A
26512
26529


1429A
26517
26538


1430A
26517
26541


1431A
26534
26556


1432A
26544
26594


1433A
26550
26571


1434A
26595
26609


1435A
26598
26612


1436A
26611
26631


1437A
26612
26628


1438A
26614
26632


1439A
26614
26645


1440A
26619
26646


1441A
26622
26647


1442A
26628
26646


1443A
26635
26651


1444A
26635
26653


1445A
26641
26678


1446A
26646
26663


1447A
26656
26675


1448A
26657
26677


1449A
26681
26696


1450A
26684
26698


1451A
26700
26719


1452A
26763
26786


1453A
26796
26818


1454A
26859
26880


1455A
26905
26924


1456A
26951
26968


1457A
26956
26970


1458A
26956
26981


1459A
26981
26998


1460A
27000
27033


1461A
27010
27028


1462A
27021
27056


1463A
27058
27075


1464A
27058
27076


1465A
27058
27092


1466A
27060
27082


1467A
27063
27091


1468A
27063
27093


1469A
27064
27085


1470A
27064
27093


1471A
27066
27085


1472A
27066
27090


1473A
27080
27097


1474A
27104
27134


1475A
27137
27159


1476A
27147
27169


1477A
27173
27188


1478A
27198
27212


1479A
27202
27219


1480A
27245
27270


1481A
27270
27286


1482A
27297
27327


1483A
27315
27333


1484A
27335
27350


1485A
27368
27389


1486A
27372
27389


1487A
27378
27399


1488A
27378
27400


1489A
27378
27409


1490A
27413
27434


1491A
27437
27453


1492A
27461
27475


1493A
27477
27493


1494A
27510
27558


1495A
27561
27591


1496A
27593
27608


1497A
27665
27679


1498A
27683
27718


1499A
27730
27756


1500A
27768
27783


1501A
27785
27799


1502A
27807
27826


1503A
27828
27863


1504A
27883
27923


1505A
27925
27966


1506A
27968
27999


1507A
28001
28017


1508A
28022
28040


1509A
28046
28061


1510A
28063
28088


1511A
28070
28092


1512A
28070
28091


1513A
28071
28092


1514A
28071
28091


1515A
28076
28093


1516A
28090
28137


1517A
28142
28164


1518A
28168
28195


1519A
28183
28202


1520A
28229
28254


1521A
28256
28287


1522A
28276
28294


1523A
28279
28294


1524A
28282
28297


1525A
28282
28309


1526A
28287
28306


1527A
28297
28315


1528A
28317
28355


1529A
28357
28398


1530A
28411
28425


1531A
28427
28462


1532A
28502
28529


1533A
28546
28576


1534A
28624
28671


1535A
28673
28714


1536A
28716
28738


1537A
28773
28792


1538A
28804
28828


1539A
28808
28828


1540A
28836
28863


1541A
28839
28863


1542A
28843
28863


1543A
28871
28898


1544A
28874
28898


1545A
28878
28898


1546A
28913
28933


1547A
28980
29000


1548A
29010
29024


1549A
29012
29028


1550A
29026
29059


1551A
29068
29104


1552A
29103
29128


1553A
29160
29179


1554A
29181
29196


1555A
29195
29211


1556A
29195
29217


1557A
29206
29225


1558A
29209
29225


1559A
29211
29225


1560A
29213
29243


1561A
29216
29243


1562A
29223
29240


1563A
29231
29250


1564A
29241
29255


1565A
29241
29263


1566A
29249
29263


1567A
29251
29273


1568A
29252
29267


1569A
29261
29279


1570A
29275
29297


1571A
29285
29317


1572A
29290
29310


1573A
29301
29323


1574A
29305
29331


1575A
29305
29341


1576A
29320
29334


1577A
29349
29380


1578A
29356
29374


1579A
29358
29385


1580A
29358
29388


1581A
29368
29393


1582A
29382
29403


1583A
29382
29407


1584A
29382
29398


1585A
29395
29423


1586A
29405
29422


1587A
29418
29438


1588A
29426
29445


1589A
29426
29459


1590A
29430
29452


1591A
29435
29458


1592A
29442
29459


1593A
29452
29466


1594A
29471
29503


1595A
29523
29537


1596A
29565
29593


1597A
29596
29623


1598A
29637
29657


1599A
29659
29685


1600A
29687
29706


1601A
29696
29711


1602A
29707
29723


1603A
29709
29729


1604A
29711
29729


1605A
29711
29733


1606A
29711
29726


1607A
29714
29732


1608A
29737
29760


1609A
29741
29760


1610A
29742
29760


1611A
29742
29756


1612A
29747
29773


1613A
29748
29763


1614A
29748
29773


1615A
29748
29768


1616A
29754
29773


1617A
29761
29776


1618A
29761
29783


1619A
29771
29787


1620A
29776
29809


1621A
29778
29800


1622A
29781
29801


1623A
29784
29809


1624A
29796
29810


1625A
29797
29811


1626A
29834
29849


1627A
29837
29851


1628A
29837
29853


1629A
29838
29853


1630A
29844
29875


1631A
29882
29917


1632A
29915
29939


1633A
29927
29949


1634A
29942
29957


1635A
29965
29980


1636A
29989
30009


1637A
30004
30025


1638A
30013
30033


1639A
30015
30033


1640A
30015
30030


1641A
30026
30043


1642A
30039
30062


1643A
30080
30102


1644A
30091
30117


1645A
30108
30122


1646A
30110
30130


1647A
30133
30148


1648A
30136
30154


1649A
30136
30178


1650A
30141
30178


1651A
30145
30162


1652A
30146
30178


1653A
30146
30162


1654A
30180
30197


1655A
30194
30209


1656A
30199
30214


1657A
30217
30239


1658A
30218
30233


1659A
30218
30237


1660A
30227
30245


1661A
30271
30287


1662A
30289
30307


1663A
30319
30351


1664A
30354
30369


1665A
30393
30409


1666A
30411
30429


1667A
30431
30452


1668A
30469
30494


1669A
30497
30534


1670A
30536
30563


1671A
30584
30607


1672A
30622
30649


1673A
30651
30675


1674A
30677
30696


1675A
30698
30718


1676A
30742
30759


1677A
30776
30792


1678A
30794
30826


1679A
30803
30818


1680A
30828
30842


1681A
30830
30845


1682A
30845
30864


1683A
30849
30863


1684A
30875
30901


1685A
30896
30910


1686A
30903
30928


1687A
30928
30946


1688A
30934
30949


1689A
30934
30956


1690A
30959
30987


1691A
30967
30985


1692A
30986
31006


1693A
30995
31019


1694A
30998
31017


1695A
31000
31015


1696A
31000
31016


1697A
31006
31020


1698A
31007
31021


1699A
31048
31068


1700A
31079
31120


1701A
31122
31160


1702A
31162
31191


1703A
31204
31227


1704A
31241
31256


1705A
31292
31313


1706A
31317
31341


1707A
31355
31394


1708A
31384
31399


1709A
31387
31402


1710A
31388
31402


1711A
31405
31426


1712A
31428
31453


1713A
31431
31446


1714A
31431
31453


1715A
31441
31457


1716A
31463
31477


1717A
31466
31480


1718A
31467
31481


1719A
31501
31526


1720A
31502
31517


1721A
31555
31575


1722A
31563
31579


1723A
31564
31589


1724A
31567
31592


1725A
31586
31609


1726A
31589
31609


1727A
31597
31631


1728A
31610
31624


1729A
31629
31645


1730A
31649
31666


1731A
31668
31686


1732A
31676
31692


1733A
31688
31715


1734A
31690
31711


1735A
31691
31717


1736A
31696
31711


1737A
31703
31719


1738A
31704
31719


1739A
31726
31741


1740A
31729
31756


1741A
31758
31773


1742A
31775
31789


1743A
31775
31792


1744A
31803
31819


1745A
31816
31857


1746A
31829
31855


1747A
31837
31855


1748A
31839
31857


1749A
31865
31881


1750A
31869
31884


1751A
31869
31885


1752A
31888
31904


1753A
31892
31915


1754A
31919
31933


1755A
31922
31942


1756A
31931
31953


1757A
31935
31957


1758A
31941
31958


1759A
31956
31977


1760A
31982
32003


1761A
32005
32023


1762A
32017
32047


1763A
32056
32098


1764A
32106
32145


1765A
32158
32192


1766A
32194
32210


1767A
32212
32226


1768A
32233
32275


1769A
32277
32335


1770A
32337
32389


1771A
32366
32380


1772A
32391
32405


1773A
32413
32460


1774A
32462
32487


1775A
32489
32511


1776A
32508
32526


1777A
32517
32532


1778A
32534
32548


1779A
32540
32555


1780A
32572
32609


1781A
32579
32593


1782A
32609
32624


1783A
32626
32676


1784A
32668
32690


1785A
32684
32704


1786A
32692
32707


1787A
32699
32713


1788A
32722
32741


1789A
32723
32741


1790A
32723
32737


1791A
32729
32744


1792A
32729
32749


1793A
32751
32767


1794A
32762
32782


1795A
32795
32809


1796A
32799
32816


1797A
32812
32829


1798A
32814
32829


1799A
32817
32831


1800A
32817
32834


1801A
32817
32833


1802A
32817
32853


1803A
32818
32833


1804A
32862
32881


1805A
32898
32920


1806A
32899
32914


1807A
32899
32918


1808A
32900
32928


1809A
32908
32926


1810A
32908
32935


1811A
32930
32946


1812A
32931
32946


1813A
32934
32949


1814A
32976
33006


1815A
33008
33029


1816A
33031
33046


1817A
33051
33069


1818A
33082
33097


1819A
33099
33118


1820A
33119
33137


1821A
33156
33172


1822A
33174
33211


1823A
33260
33279


1824A
33283
33305


1825A
33307
33322


1826A
33345
33376


1827A
33402
33437


1828A
33465
33522


1829A
33542
33569


1830A
33571
33605


1831A
33608
33626


1832A
33648
33677


1833A
33682
33727


1834A
33729
33809


1835A
33817
33845


1836A
33847
33868


1837A
33885
33900


1838A
33902
33949


1839A
33952
34007


1840A
34019
34042


1841A
34044
34066


1842A
34079
34112


1843A
34114
34227


1844A
34229
34314


1845A
34316
34335


1846A
34337
34391









In some embodiments, the target sequence is selected from the group consisting of target regions 1A to 1846A as shown in Table 4 above.









TABLE 5







Exemplary target regions









Target
start SEQ
end SEQ


region
ID NO: 1
ID NO: 1












 1C
84
98


 2C
100
125


 3C
419
446


 4C
635
649


 5C
860
873


 6C
861
875


 7C
862
875


 8C
2384
2397


 9C
2386
2400


 10C
3264
3279


 11C
3264
3278


 12C
3328
3341


 13C
3826
3840


 14C
3832
3846


 15C
3839
3852


 16C
4425
4438


 17C
4686
4699


 18C
4690
4703


 19C
5121
5134


 20C
5136
5149


 21C
5151
5170


 22C
5172
5191


 23C
5309
5322


 24C
5312
5325


 25C
5624
5637


 26C
5627
5640


 27C
5627
5641


 28C
5628
5641


 29C
5890
5904


 30C
6504
6517


 31C
6821
6837


 32C
6822
6835


 33C
6825
6839


 34C
6836
6849


 35C
6838
6852


 36C
6838
6853


 37C
7613
7628


 38C
7614
7628


 39C
9042
9056


 40C
9326
9340


 41C
9327
9340


 42C
10259
10277


 43C
10282
10301


 44C
10309
10325


 45C
10351
10373


 46C
10461
10526


 47C
11014
11032


 48C
11827
11842


 49C
11832
11847


 50C
11835
11849


 51C
11845
11862


 52C
11846
11862


 53C
12405
12418


 54C
13191
13212


 55C
13225
13256


 56C
13258
13301


 57C
13303
13352


 58C
13378
13392


 59C
13618
13638


 60C
13619
13632


 61C
13620
13637


 62C
13621
13638


 63C
13623
13639


 64C
13624
13639


 65C
13626
13640


 66C
13627
13640


 67C
13925
13945


 68C
13925
13946


 69C
13926
13939


 70C
13927
13944


 71C
13928
13945


 72C
13929
13946


 73C
13931
13947


 74C
13932
13947


 75C
13934
13948


 76C
13935
13948


 77C
14455
14468


 78C
14666
14680


 79C
15109
15122


 80C
15894
15913


 81C
16320
16339


 82C
16341
16387


 83C
16431
16453


 84C
17752
17765


 85C
17754
17768


 86C
17754
17769


 87C
17971
17984


 88C
17971
17986


 89C
17972
17985


 90C
17974
17988


 91C
17975
17988


 92C
19166
19190


 93C
19210
19230


 94C
19232
19268


 95C
19303
19317


 96C
19562
19575


 97C
19563
19576


 98C
19658
19671


 99C
19724
19737


100C
19727
19740


101C
19727
19741


102C
19728
19741


103C
19731
19744


104C
19731
19745


105C
19732
19745


106C
19872
19886


107C
20436
20451


108C
20437
20451


109C
20456
20472


110C
20457
20472


111C
20459
20473


112C
20460
20473


113C
20821
20834


114C
20836
20852


115C
20938
20954


116C
21054
21069


117C
21159
21180


118C
21180
21196


119C
21339
21352


120C
21360
21373


121C
21444
21458


122C
23004
23019


123C
23459
23475


124C
23461
23474


125C
23461
23476


126C
23462
23475


127C
23463
23478


128C
23465
23479


129C
23466
23479


130C
23892
23908


131C
23894
23907


132C
23895
23908


133C
23896
23909


134C
25244
25257


135C
25245
25261


136C
25246
25261


137C
25395
25408


138C
25395
25409


139C
25396
25409


140C
25399
25412


141C
25399
25413


142C
25400
25413


143C
25403
25416


144C
25403
25417


145C
25404
25417


146C
25407
25420


147C
25407
25421


148C
25408
25421


149C
25411
25424


150C
25411
25425


151C
25412
25425


152C
25415
25428


153C
25415
25429


154C
25416
25429


155C
25419
25432


156C
25419
25433


157C
25420
25433


158C
25971
25987


159C
26315
26330


160C
26344
26363


161C
26438
26454


162C
26440
26453


163C
26440
26455


164C
26441
26454


165C
26442
26459


166C
26447
26460


167C
26951
26968


168C
26953
26966


169C
26954
26969


170C
26956
26970


171C
26957
26970


172C
29014
29028


173C
30271
30284


174C
31931
31948


175C
31932
31948


176C
31932
31945


177C
31934
31947


178C
31935
31952


179C
31936
31952


180C
31936
31949


181C
31938
31951


182C
31939
31956


183C
31940
31956


184C
31940
31953


185C
31942
31955


186C
31944
31957


187C
32211
32224


188C
32212
32225


189C
32250
32275


190C
32280
32305


191C
32310
32335


192C
32588
32601


193C
32593
32606


194C
32831
32845


195C
32930
32946


196C
32931
32944


197C
32934
32948


198C
33182
33195


199C
33729
33742


200C
33753
33769


201C
34126
34145


202C
34318
34335


203C
34337
34355









In some embodiments, the target sequence is selected from the group consisting of target regions 10 to 178C as shown in Table 5 above.


Target Cell The term a “target cell” as used herein refers to a cell which is expressing the target nucleic acid. For the therapeutic use of the present invention it is advantageous if the target cell is infected with HBV. In some embodiments, the target cell may be in vivo or in vitro. In some embodiments, the target cell is a mammalian cell such as a rodent cell, such as a mouse cell or a rat cell, or a woodchuck cell or a primate cell such as a monkey cell (e.g. a cynomolgus monkey cell) or a human cell.


In preferred embodiments, the target cell expresses COPS3 mRNA, such as the COPS3 pre-mRNA or COPS3 mature mRNA. The poly A tail of COPS3 mRNA is typically disregarded for antisense oligonucleotide targeting.


Further, the target cell may be a hepatocyte. In one embodiment, the target cell is HBV infected primary human hepatocytes, either derived from HBV infected individuals or from a HBV infected mouse with a humanized liver (PhoenixBio, PXB-mouse).


In accordance with the present invention, the target cell may be infected with HBV. Further, the target cell may comprise HBV cccDNA. Thus, the target cell preferably comprises COPS3 mRNA, such as the COPS3 pre-mRNA or COPS3 mature mRNA, and HBV cccDNA.


Naturally occurring variant The term “naturally occurring variant” refers to variants of COPS3 gene or transcripts which originate from the same genetic loci as the target nucleic acid, but may differ for example, by virtue of degeneracy of the genetic code causing a multiplicity of codons encoding the same amino acid, or due to alternative splicing of pre-mRNA, or the presence of polymorphisms, such as single nucleotide polymorphisms (SNPs), and allelic variants. Based on the presence of the sufficient complementary sequence to the oligonucleotide, the oligonucleotide of the invention may therefore target the target nucleic acid and naturally occurring variants thereof.


In some embodiments, the naturally occurring variants have at least 95% such as at least 98% or at least 99% homology to a mammalian COPS3 target nucleic acid, such as a target nucleic acid of SEQ ID NO: 1 and/or SEQ ID NO: 2. In some embodiments, the naturally occurring variants have at least 99% homology to the human COPS3 target nucleic acid of SEQ ID NO: 1. In some embodiments, the naturally occurring variants are known polymorphisms.


Inhibition of expression The term “inhibition of expression” as used herein is to be understood as an overall term for an COPS3 (COP9 Signalosome Subunit 3) inhibitor's ability to inhibit, i.e. to reduce, the amount or the activity of COPS3 in a target cell. Inhibition of expression or activity may be determined by measuring the level of COPS3 pre-mRNA or COPS3 mRNA, or by measuring the level of COPS3 protein or activity in a cell. Inhibition of expression may be determined in vitro or in vivo. Advantageously, the inhibition is assessed in relation to the amount of COPS3 before administration of the COPS3 inhibitor. Alternatively, inhibition is determined by reference to a control. It is generally understood that the control is an individual or target cell treated with a saline composition or an individual or target cell treated with a non-targeting oligonucleotide (mock).


The term “inhibition” or “inhibit” may also be referred to as down-regulate, reduce, suppress, lessen, lower, decrease the expression or activity of COPS3.


The inhibition of expression of COPS3 may occur e.g. by degradation of pre-mRNA or mRNA e.g. using RNase H recruiting oligonucleotides, such as gapmers, or nucleic acid molecules that function via the RNA interference pathway, such as siRNA or shRNA. Alternatively, the inhibitor of the present invention may bind to COPS3 polypeptide and inhibit the activity of COPS3 or prevent its binding to other molecules.


In some embodiments, the inhibition of expression of the COPS3 target nucleic acid or the activity of COPS3 protein results in a decreased amount of HBV cccDNA in the target cell. Preferably, the amount of HBV cccDNA is decreased as compared to a control. In some embodiments, the decrease in amount of HBV cccDNA is at least 20%, at least 30%, as compared to a control. In some embodiments, the amount of cccDNA in an HBV infected cell is reduced by at least 50%, such as 60% when compared to a control.


In some embodiments, the inhibition of expression of the COPS3 target nucleic acid or the activity of COPS3 protein results in a decreased amount of HBV pgRNA in the target cell. Preferably, the amount of HBV pgRNA is decreased as compared to a control. In some embodiments, the decrease in amount of HBV pgRNA is at least 20%, at least 30%, as compared to a control. In some embodiments, the amount of pgRNA in an HBV infected cell is reduced by at least 50%, such as 60%, when compared to a control.


Sugar modifications The oligonucleotide of the invention may comprise one or more nucleosides which have a modified sugar moiety, i.e. a modification of the sugar moiety when compared to the ribose sugar moiety found in DNA and RNA.


Numerous nucleosides with modification of the ribose sugar moiety have been made, primarily with the aim of improving certain properties of oligonucleotides, such as affinity and/or nuclease resistance.


Such modifications include those where the ribose ring structure is modified, e.g. by replacement with a hexose ring (HNA), or a bicyclic ring, which typically have a biradical bridge between the C2 and C4 carbons on the ribose ring (LNA), or an unlinked ribose ring which typically lacks a bond between the C2 and C3 carbons (e.g. UNA). Other sugar modified nucleosides include, for example, bicyclohexose nucleic acids (WO2011/017521) or tricyclic nucleic acids (WO2013/154798). Modified nucleosides also include nucleosides where the sugar moiety is replaced with a non-sugar moiety, for example in the case of peptide nucleic acids (PNA), or morpholino nucleic acids.


Sugar modifications also include modifications made via altering the substituent groups on the ribose ring to groups other than hydrogen, or the 2′—OH group naturally found in DNA and RNA nucleosides. Substituents may, for example be introduced at the 2′, 3′, 4′ or 5′ positions.


High affinity modified nucleosides A high affinity modified nucleoside is a modified nucleotide which, when incorporated into the oligonucleotide enhances the affinity of the oligonucleotide for its complementary target, for example as measured by the melting temperature (Tm). A high affinity modified nucleoside of the present invention preferably result in an increase in melting temperature in the range of +0.5 to +12° C., more preferably in the range of +1.5 to +10° C. and most preferably in the range of +3 to +8° C. per modified nucleoside. Numerous high affinity modified nucleosides are known in the art and include for example, many 2′ substituted nucleosides as well as locked nucleic acids (LNA) (see e.g. Freier & Altmann; Nucl. Acid Res., 1997, 25, 4429-4443 and Uhlmann; Curr. Opinion in Drug Development, 2000, 3(2), 293-213).


2′ sugar modified nucleosides A 2′ sugar modified nucleoside is a nucleoside which has a substituent other than H or —OH at the 2′ position (2′ substituted nucleoside) or comprises a 2′ linked biradical capable of forming a bridge between the 2′ carbon and a second carbon in the ribose ring, such as LNA (2′-4′ biradical bridged) nucleosides.


Indeed, much focus has been spent on developing 2′ sugar substituted nucleosides, and numerous 2′ substituted nucleosides have been found to have beneficial properties when incorporated into oligonucleotides. For example, the 2′ modified sugar may provide enhanced binding affinity and/or increased nuclease resistance to the oligonucleotide. Examples of 2′ substituted modified nucleosides are 2′-O-alkyl-RNA, 2′-O-methyl-RNA, 2′-alkoxy-RNA, 2′-O-methoxyethyl-RNA (MOE), 2′-amino-DNA, 2′-Fluoro-RNA, and 2′-F-ANA nucleoside. For further examples, please see e.g. Freier & Altmann; Nucl. Acid Res., 1997, 25, 4429-4443 and Uhlmann; Curr. Opinion in Drug Development, 2000, 3(2), 293-213, and Deleavey and Damha, Chemistry and Biology 2012, 19, 937. Below are illustrations of some 2′ substituted modified nucleosides.




embedded image


In relation to the present invention 2′ substituted sugar modified nucleosides does not include 2′ bridged nucleosides like LNA.


Locked Nucleic Acid Nucleosides (LNA nucleoside) A “LNA nucleoside” is a 2′-sugar modified nucleoside which comprises a biradical linking the C2′ and C4′ of the ribose sugar ring of said nucleoside (also referred to as a “2′- 4′ bridge”), which restricts or locks the conformation of the ribose ring. These nucleosides are also termed bridged nucleic acid or bicyclic nucleic acid (BNA) in the literature. The locking of the conformation of the ribose is associated with an enhanced affinity of hybridization (duplex stabilization) when the LNA is incorporated into an oligonucleotide for a complementary RNA or DNA molecule. This can be routinely determined by measuring the melting temperature of the oligonucleotide/complement duplex.


Non limiting, exemplary LNA nucleosides are disclosed in WO 99/014226, WO 00/66604, WO 98/039352, WO 2004/046160, WO 00/047599, WO 2007/134181, WO 2010/077578, WO 2010/036698, WO 2007/090071, WO 2009/006478, WO 2011/156202, WO 2008/154401, WO 2009/067647, WO 2008/150729, Morita et al., Bioorganic & Med. Chem. Lett. 12, 73-76, Seth et al. J. Org. Chem. 2010, Vol 75(5) pp. 1569-81, Mitsuoka et al., Nucleic Acids Research 2009, 37(4), 1225-1238, and Wan and Seth, J. Medical Chemistry 2016, 59, 9645-9667.


Particular examples of LNA nucleosides of the invention are presented in Scheme 1 (wherein B is as defined above).




embedded image


embedded image


Particular LNA nucleosides are beta-D-oxy-LNA, 6′-methyl-beta-D-oxy LNA such as (S)-6′-methyl-beta-D-oxy-LNA (ScET) and ENA.


RNase H Activity and Recruitment


The RNase H activity of an antisense oligonucleotide refers to its ability to recruit RNase H when in a duplex with a complementary RNA molecule. WO01/23613 provides in vitro methods for determining RNase H activity, which may be used to determine the ability to recruit RNase H. Typically an oligonucleotide is deemed capable of recruiting RNase H if it, when provided with a complementary target nucleic acid sequence, has an initial rate, as measured in pmol/l/min, of at least 5%, such as at least 10% or more than 20% of the of the initial rate determined when using a oligonucleotide having the same base sequence as the modified oligonucleotide being tested, but containing only DNA monomers with phosphorothioate linkages between all monomers in the oligonucleotide, and using the methodology provided by Example 91-95 of WO 01/23613 (hereby incorporated by reference). For use in determining RHase H activity, recombinant human RNase H1 is available from Creative Biomart® (Recombinant Human RNase H1 fused with His tag expressed in E. coli).


Gapmer


The antisense oligonucleotide of the invention, or contiguous nucleotide sequence thereof, may be a gapmer, also termed gapmer oligonucleotide or gapmer designs. The antisense gapmers are commonly used to inhibit a target nucleic acid via RNase H mediated degradation. A gapmer oligonucleotide comprises at least three distinct structural regions a 5′-flank, a gap and a 3′-flank, F-G-F′ in the ‘5->3’ orientation. The “gap” region (G) comprises a stretch of contiguous DNA nucleotides which enable the oligonucleotide to recruit RNase H. The gap region is flanked by a 5′ flanking region (F) comprising one or more sugar modified nucleosides, advantageously high affinity sugar modified nucleosides, and by a 3′ flanking region (F′) comprising one or more sugar modified nucleosides, advantageously high affinity sugar modified nucleosides. The one or more sugar modified nucleosides in region F and F′ enhance the affinity of the oligonucleotide for the target nucleic acid (i.e. are affinity enhancing sugar modified nucleosides). In some embodiments, the one or more sugar modified nucleosides in region F and F′ are 2′ sugar modified nucleosides, such as high affinity 2′ sugar modifications, such as independently selected from LNA and 2′-MOE.


In a gapmer design, the 5′ and 3′ most nucleosides of the gap region are DNA nucleosides, and are positioned adjacent to a sugar modified nucleoside of the 5′ (F) or 3′ (F′) region respectively. The flanks may further be defined by having at least one sugar modified nucleoside at the end most distant from the gap region, i.e. at the 5′ end of the 5′ flank and at the 3′ end of the 3′ flank.


Regions F-G-F′ form a contiguous nucleotide sequence. Antisense oligonucleotides of the invention, or the contiguous nucleotide sequence thereof, may comprise a gapmer region of formula F-G-F′.


The overall length of the gapmer design F-G-F′ may be, for example 12 to 32 nucleosides, such as 13 to 24, such as 14 to 22 nucleosides, such as from 15 to 20, such as 16 to 18 nucleosides. By way of example, the gapmer oligonucleotide of the present invention can be represented by the following formulae:


F1-8-G5-18-F′1-8, such as


F1-8-G7-18-F′2-8


with the proviso that the overall length of the gapmer regions F-G-F′ is at least 12, such as at least 14 nucleotides in length.


In an aspect of the invention, the antisense oligonucleotide or contiguous nucleotide sequence thereof consists of or comprises a gapmer of formula 5′-F-G-F′-3′, where region F and F′ independently comprise or consist of 1-8 nucleosides, of which 1-4 are 2′ sugar modified and defines the 5′ and 3′ end of the F and F′ region, and G is a region between 6 and 18 nucleosides which are capable of recruiting RNase H. In some embodiments, the G region consists of DNA nucleosides.


In some embodiments, region F and F′ independently consists of or comprises a contiguous sequence of sugar modified nucleosides. In some embodiments, the sugar modified nucleosides of region F may be independently selected from 2′-O-alkyl-RNA units, 2′-O-methyl-RNA, 2′-amino-DNA units, 2′-fluoro-DNA units, 2′-alkoxy-RNA, MOE units, LNA units, arabino nucleic acid (ANA) units and 2′-fluoro-ANA units.


In some embodiments, region F and F′ independently comprises both LNA and a 2′-substituted sugar modified nucleotide (mixed wing design). In some embodiments, the 2′-substituted sugar modified nucleotide is independently selected from the group consisting of 2′-O-alkyl-RNA units, 2′-O-methyl-RNA, 2′-amino-DNA units, 2′-fluoro-DNA units, 2′-alkoxy-RNA, MOE units, arabino nucleic acid (ANA) units and 2′-fluoro-ANA units.


In some embodiments, all the modified nucleosides of region F and F′ are LNA nucleosides, such as independently selected from beta-D-oxy LNA, ENA or ScET nucleosides, wherein region F or F′, or F and F′ may optionally comprise DNA nucleosides. In some embodiments, all the modified nucleosides of region F and F′ are beta-D-oxy LNA nucleosides, wherein region F or F′, or F and F′ may optionally comprise DNA nucleosides. In such embodiments, the flanking region F or F′, or both F and F′ comprise at least three nucleosides, wherein the 5′ and 3′ most nucleosides of the F and/or F′ region are LNA nucleosides.


LNA Gapmer An LNA gapmer is a gapmer wherein either one or both of region F and F′ comprises or consists of LNA nucleosides. A beta-D-oxy gapmer is a gapmer wherein either one or both of region F and F′ comprises or consists of beta-D-oxy LNA nucleosides.


In some embodiments, the LNA gapmer is of formula: [LNA]i1-5-[region G]6-18-[LNA]1-5, wherein region G is as defined in the Gapmer region G definition.


MOE Gapmers


A MOE gapmers is a gapmer wherein regions F and F′ consist of MOE nucleosides. In some embodiments, the MOE gapmer is of design [MOE]1-8-[Region G]5-16-[MOE]1-8, such as [MOE]2-7-[Region G]6-14-[MOE]2-7, such as [MOE]3-6-[Region G]8-12-[MOE]3-6, such as [MOE]5-[Region G]10- [MOE]s wherein region G is as defined in the Gapmer definition. MOE gapmers with a 5-10-5 design (MOE-DNA-MOE) have been widely used in the art.


Region D′ or D″ in an oligonucleotide The oligonucleotide of the invention may in some embodiments comprise or consist of the contiguous nucleotide sequence of the oligonucleotide which is complementary to the target nucleic acid, such as a gapmer region F-G-F′, and further 5′ and/or 3′ nucleosides. The further 5′ and/or 3′ nucleosides may or may not be fully complementary to the target nucleic acid. Such further 5′ and/or 3′ nucleosides may be referred to as region D′ and D″ herein.


The addition of region D′ or D″ may be used for the purpose of joining the contiguous nucleotide sequence, such as the gapmer, to a conjugate moiety or another functional group. When used for joining the contiguous nucleotide sequence with a conjugate moiety is can serve as a biocleavable linker. Alternatively, it may be used to provide exonuclease protection or for ease of synthesis or manufacture.


Region D′ and D″ can be attached to the 5′ end of region F or the 3′ end of region F′, respectively to generate designs of the following formulas D′-F-G-F′, F-G-F′-D″ or D′-F-G-F′-D″. In this instance the F-G-F′ is the gapmer portion of the oligonucleotide and region D′ or D″ constitute a separate part of the oligonucleotide.


Region D′ or D″ may independently comprise or consist of 1, 2, 3, 4 or 5 additional nucleotides, which may be complementary or non-complementary to the target nucleic acid. The nucleotide adjacent to the F or F′ region is not a sugar-modified nucleotide, such as a DNA or RNA or base modified versions of these. The D′ or D″ region may serve as a nuclease susceptible biocleavable linker (see definition of linkers). In some embodiments, the additional 5′ and/or 3′ end nucleotides are linked with phosphodiester linkages, and are DNA or RNA. Nucleotide based biocleavable linkers suitable for use as region D′ or D″ are disclosed in WO2014/076195, which include by way of example a phosphodiester linked DNA dinucleotide. The use of biocleavable linkers in poly-oligonucleotide constructs is disclosed in WO2015/113922, where they are used to link multiple antisense constructs (e.g. gapmer regions) within a single oligonucleotide.


In one embodiment, the oligonucleotide of the invention comprises a region D′ and/or D″ in addition to the contiguous nucleotide sequence which constitutes the gapmer.


In some embodiments, the oligonucleotide of the present invention can be represented by the following formulae:


F-G-F′; in particular F1-8-G5-18-F′2-8


D′-F-G-F′, in particular D′1-3-F1-8-G5-18-F2-8


F-G-F′-D″, in particular F1-8-G5-18-F′2-8-D″1-3


D′-F-G-F′-D″, in particular D′1-3- F1-8-G5-18-F2-8-D″1-3


In some embodiments, the internucleoside linkage positioned between region D′ and region F is a phosphodiester linkage. In some embodiments, the internucleoside linkage positioned between region F′ and region D″ is a phosphodiester linkage.


Conjugate


The term conjugate as used herein refers to an oligonucleotide which is covalently linked to a non-nucleotide moiety (conjugate moiety or region C or third region). The conjugate moiety may be covalently linked to the antisense oligonucleotide, optionally via a linker group, such as region D′ or D″


Oligonucleotide conjugates and their synthesis have been reported in comprehensive reviews by Manoharan in Antisense Drug Technology, Principles, Strategies, and Applications, S.T. Crooke, ed., Ch. 16, Marcel Dekker, Inc., 2001 and Manoharan, Antisense and Nucleic Acid Drug Development, 2002, 12, 103, each of which is incorporated herein by reference in its entirety.


In some embodiments, the non-nucleotide moiety (conjugate moiety) is selected from the group consisting of carbohydrates (e.g. galactose or N-acetylgalactosamine (GaINAc)), cell surface receptor ligands, drug substances, hormones, lipophilic substances, polymers, proteins (e.g. antibodies), peptides, toxins (e.g. bacterial toxins), vitamins, viral proteins (e.g. capsids) or combinations thereof.


Exemplary conjugate moieties are those capable of binding to the asialoglycoprotein receptor (ASGPR). In particular, tri-valent N-acetylgalactosamine conjugate moieties are suitable for binding to the ASGPR, see for example WO 2014/076196, WO 2014/207232 and WO 2014/179620 (hereby incorporated by reference). Such conjugates serve to enhance uptake of the oligonucleotide to the liver.


Linkers


A linkage or linker is a connection between two atoms that links one chemical group or segment of interest to another chemical group or segment of interest via one or more covalent bonds. Conjugate moieties can be attached to the oligonucleotide directly or through a linking moiety (e.g. linker or tether). Linkers serve to covalently connect a third region, e.g. a conjugate moiety (region C), to a first region, e.g. an oligonucleotide or contiguous nucleotide sequence complementary to the target nucleic acid (region A).


In some embodiments of the invention the conjugate or oligonucleotide conjugate of the invention may optionally, comprise a linker region (second region or region B and/or region Y) which is positioned between the oligonucleotide or contiguous nucleotide sequence complementary to the target nucleic acid (region A or first region) and the conjugate moiety (region C or third region).


Region B refers to biocleavable linkers comprising or consisting of a physiologically labile bond that is cleavable under conditions normally encountered or analogous to those encountered within a mammalian body. Conditions under which physiologically labile linkers undergo chemical transformation (e.g., cleavage) include chemical conditions such as pH, temperature, oxidative or reductive conditions or agents, and salt concentration found in or analogous to those encountered in mammalian cells. Mammalian intracellular conditions also include the presence of enzymatic activity normally present in a mammalian cell such as from proteolytic enzymes or hydrolytic enzymes or nucleases. In one embodiment, the biocleavable linker is susceptible to S1 nuclease cleavage. In a preferred embodiment the nuclease susceptible linker comprises between 1 and 5 nucleosides, such as 1, 2, 3, 4 or 5 nucleosides, more preferably between 2 and 4 nucleosides and most preferably 2 or 3 linked nucleosides comprising at least two consecutive phosphodiester linkages, such as at least 3 or 4 or 5 consecutive phosphodiester linkages. Preferably the nucleosides are DNA or RNA. Phosphodiester containing biocleavable linkers are described in more detail in WO 2014/076195 (hereby incorporated by reference).


Region Y refers to linkers that are not necessarily biocleavable but primarily serve to covalently connect a conjugate moiety (region C or third region), to an oligonucleotide (region A or first region). The region Y linkers may comprise a chain structure or an oligomer of repeating units such as ethylene glycol, amino acid units or amino alkyl groups The oligonucleotide conjugates of the present invention can be constructed of the following regional elements A-C, A-B-C, A-B-Y-C, A-Y-B-C or A-Y-C. In some embodiments, the linker (region Y) is an amino alkyl, such as a C2-C36 amino alkyl group, including, for example C6 to C12 amino alkyl groups. some embodiments the linker (region Y) is a C6 amino alkyl group.


Treatment The term “treatment” as used herein refers to both treatment of an existing disease (e.g. a disease or disorder as herein referred to), or prevention of a disease, i.e. prophylaxis. It will therefore be recognized that treatment as referred to herein may, in some embodiments, be prophylactic. Prophylactic can be understood as preventing an HBV infection from turning into a chronic HBV infection or the prevention of severe liver diseases such as liver cirrhosis and hepatocellular carcinoma caused by a chronic HBV infection.


Patient For the purposes of the present invention the “subject” (or “patient”) may be a vertebrate. In context of the present invention, the term “subject” includes both humans and other animals, particularly mammals, and other organisms. Thus, the herein provided means and methods are applicable to both human therapy and veterinary applications. Preferably, the subject is a mammal. More preferably the subject is human.


As described elsewhere herein, the patient to be treated may suffers from HBV infection, such as chronic HBV infection. In some embodiments, the patient suffering from HBV infection may suffer from hepatocellular carcinoma (HCC). In some embodiments, the patient suffering from HBV infection does not suffer from hepatocellular carcinoma.


DETAILED DESCRIPTION OF THE INVENTION

HBV cccDNA in infected hepatocytes is responsible for persistent chronic infection and reactivation, being the template for all viral subgenomic transcripts and pre-genomic RNA (pgRNA) to ensure both newly synthesized viral progeny and cccDNA pool replenishment via intracellular nucleocapsid recycling. In the context of the present invention it was for the first time shown that COPS3 is associated with cccDNA stability. This knowledge allows for the opportunity to destabilize cccDNA in HBV infected subjects which in turn opens the opportunity for a complete cure of chronically infected HBV patients.


One aspect of the present invention is a COPS3 inhibitor for use in the treatment and/or prevention of Hepatitis B virus (HBV) infection, in particular a chronic HBV infection.


The COPS3 inhibitor can for example be a small molecule that specifically binds to COPS3 protein, wherein said inhibitor prevents or reduces binding of COPS3 protein to cccDNA.


An embodiment of the invention is a COPS3 inhibitor which is capable of reducing cccDNA and/or pgRNA in an infected cell, such as an HBV infected cell.


In a further embodiment, the COPS3 inhibitor is capable of reducing HBsAg and/or HBeAg in vivo in an HBV infected individual.


COPS3 inhibitors for use in treatment of HBV Without being bound by theory, it is believed that COPS3 is involved in the stabilization of the cccDNA in the cell nucleus, either via direct or indirect binding to the cccDNA, and by preventing the binding/association of COPS3 with cccDNA, the cccDNA is destabilized and becomes prone to degradation. One embodiment of the invention is therefore a COPS3 inhibitor which interacts with the COPS3 protein, and prevents or reduces its binding/association to cccDNA.


In some embodiments of the present invention, the inhibitor is an antibody, antibody fragment or a small molecule compound. In some embodiments, the inhibitor may be an antibody, antibody fragment or a small molecule that specifically binds to the COPS3 protein, such as the COPS3 protein encoded by SEQ ID NO: 1, 4, 5, 6, or 7.


Nucleic acid molecules of the Invention Therapeutic nucleic acid molecules are potentially excellent COPS3 inhibitors since they can target the COPS3 transcript and promote its degradation either via the RNA interference pathway or via RNase H cleavage. Alternatively, oligonucleotides such as aptamers can also act as inhibitors of COPS3 protein interactions.


One aspect of the present invention is a COPS3 targeting nucleic acid molecule for use in treatment and/or prevention of Hepatitis B virus (HBV) infection. Such a nucleic acid molecule can be selected from the group consisting of single stranded antisense oligonucleotide, siRNA molecule, and shRNA molecule.


The present section describes novel nucleic acid molecule suitable for use in treatment and/or prevention of Hepatitis B virus (HBV) infection.


The nucleic acid molecule of the present invention is capable of inhibiting expression of COPS3 in vitro and in vivo. The inhibition is achieved by hybridizing an oligonucleotide to a target nucleic acid encoding COPS3 or which is involved in the regulation of COPS3. The target nucleic acid may be a mammalian COPS3 sequence. In some embodiments, the target nucleic acid may be a human COPS3 pre-mRNA sequence such as the sequence of SEQ ID NO: 1 or a human COPS3 mRNA sequence selected from SEQ ID NO: 4 to 9. In some embodiments, the target nucleic acid may be a cynomolgus monkey COPS3 sequence such as the sequence of SEQ ID NO: 2.


In some embodiments, the nucleic acid molecule of the invention is capable of modulating the expression of the target by inhibiting or down-regulating it. Preferably, such modulation produces an inhibition of expression of at least 20% compared to the normal expression level of the target, more preferably at least 30%, at least 40%, at least 50%, at least 60% inhibition compared to the normal expression level of the target. In some embodiments, the nucleic acid molecule of the invention may be capable of inhibiting expression levels of COPS3 mRNA by at least 60% or 70% in vitro by transfecting 25 nM nucleic acid molecule into PXB-PHH cells, this range of target reduction is advantageous in terms of selecting nucleic acid molecules with good correlation to the cccDNA reduction. Suitably, the examples provide assays which may be used to measure COPS3 RNA or protein inhibition (e.g. example 1 and the “Materials and Methods” section). The target inhibition is triggered by the hybridization between a contiguous nucleotide sequence of the oligonucleotide, such as the guide strand of a siRNA or gapmer region of an antisense oligonucleotide, and the target nucleic acid. In some embodiments, the nucleic acid molecule of the invention comprises mismatches between the oligonucleotide and the target nucleic acid. Despite mismatches hybridization to the target nucleic acid may still be sufficient to show a desired inhibition of COPS3 expression. Reduced binding affinity resulting from mismatches may advantageously be compensated by increased number of nucleotides in the oligonucleotide complementary to the target nucleic acid and/or an increased number of modified nucleosides capable of increasing the binding affinity to the target, such as 2′ sugar modified nucleosides, including LNA, present within the oligonucleotide sequence.


An aspect of the present invention relates to a nucleic acid molecules of 12 to 60 nucleotides in length, which comprises a contiguous nucleotide sequence of at least 12 nucleotides in length, such as at least 12 to 30 nucleotides in length, which is at least 95% complementary, such as fully complementary, to a mammalian COPS3 target nucleic acid, in particular a human COPS3 nucleic acid. These nucleic acid molecules are capable of inhibiting the expression of COPS3.


An aspect of the invention relates to a nucleic acid molecule of 12 to 30 nucleotides in length, comprising a contiguous nucleotide sequence of at least 12 nucleotides, such as 12 to 30 nucleotides in length which is at least 90% complementary, such as fully complementary, to a mammalian COPS3 target sequence.


A further aspect of the present invention relates to a nucleic acid molecule according to the invention comprising a contiguous nucleotide sequence of 12 to 20 nucleotides in length with at least 90% complementary, such as fully complementary, to the target nucleic acid of SEQ ID NO: 1.


In some embodiments, the nucleic acid molecule comprises a contiguous sequence of 12 to 30 nucleotides in length, which is at least 90% complementary, such as at least 91%, such as at least 92%, such as at least 93%, such as at least 94%, such as at least 95%, such as at least 96%, such as at least 97%, such as at least 98%, or 100% complementary with a region of the target nucleic acid or a target sequence.


It is advantageous if the nucleic acid molecule, or contiguous nucleotide sequence thereof is fully complementary (100% complementary) to a region of the target nucleic acid, or in some embodiments may comprise one or two mismatches between the oligonucleotide and the target nucleic acid.


In some embodiments, the oligonucleotide sequence is 100% complementary to a target nucleic acid region of SEQ ID NO: 1 and/or SEQ ID NO: 4, 5, 6, 7, 8 and/or 9.


In some embodiments, the nucleic acid molecule or the contiguous nucleotide sequence of the invention is at least 90% or 95% complementary, such as fully (or 100%) complementary, to the target nucleic acid of SEQ ID NO: 1 and 2.


In some embodiments, the oligonucleotide or the contiguous nucleotide sequence of the invention is at least 90% or 95% complementary, such as fully (or 100%) complementary, to the target nucleic acid of SEQ ID NO: 2 and SEQ ID NO: 4, 5, 6, 7, 8 or 9.


In some embodiments, the oligonucleotide or the contiguous nucleotide sequence of the invention is at least 90% or 95% complementary, such as fully (or 100%) complementary, to the target nucleic acid of SEQ ID NO: 1 and SEQ ID NO: 2 and SEQ ID NO: 3.


In some embodiments, the contiguous sequence of the nucleic acid molecule of the present invention is least 90% complementary, such as fully complementary to a region of SEQ ID NO: 1, selected from the group consisting of target regions 1A to 1846A as shown in Table 4.


In some embodiments, the contiguous sequence of the nucleic acid molecule of the present invention is least 90% complementary, such as fully complementary to a region of SEQ ID NO: 1, selected from the group consisting of target regions 10 to 203C as shown in Table 5.


In some embodiments, the nucleic acid molecule of the invention comprises or consists of 12 to 60 nucleotides in length, such as from 13 to 50, such as from 14 to 35, such as 15 to 30, such as from 16 to 22 contiguous nucleotides in length. In a preferred embodiment, the nucleic acid molecule comprises or consists of 15, 16, 17, 18, 19, 20, 21 or 22 nucleotides in length.


In some embodiments, the contiguous nucleotide sequence of the nucleic acid molecule which is complementary to the target nucleic acids comprises or consists of 12 to 30, such as from 13 to 25, such as from 15 to 23, such as from 16 to 22, contiguous nucleotides in length.


In some embodiments, the oligonucleotide is selected from the group consisting of an antisense oligonucleotide, siRNA and shRNA.


In some embodiments, the contiguous nucleotide sequence of the siRNA or shRNA which is complementary to the target nucleic acids comprises or consists of 18 to 28, such as from 19 to 26, such as from 20 to 24, such as from 21 to 23, contiguous nucleotides in length.


In some embodiments, the contiguous nucleotide sequence of the antisense oligonucleotide which is complementary to the target nucleic acids comprises or consists of 12 to 22, such as from 14 to 20, such as from 16 to 20, such as from 15 to 18, such as from 16 to 18, such as from 16, 17, 18, 19 or 20 contiguous nucleotides in length.


It is understood that the contiguous oligonucleotide sequence (motif sequence) can be modified to, for example, increase nuclease resistance and/or binding affinity to the target nucleic acid.


The pattern in which the modified nucleosides (such as high affinity modified nucleosides) are incorporated into the oligonucleotide sequence is generally termed oligonucleotide design.


The nucleic acid molecule of the invention may be designed with modified nucleosides and RNA nucleosides (in particular for siRNA and shRNA molecules) or DNA nucleosides (in particular for single stranded antisense oligonucleotides). Advantageously, high affinity modified nucleosides are used.


In advantageous embodiments, the nucleic acid molecule or contiguous nucleotide sequence comprises one or more sugar modified nucleosides, such as 2′ sugar modified nucleosides, such as comprise one or more 2′ sugar modified nucleoside independently selected from the group consisting of 2′-O-alkyl-RNA, 2′-O-methyl-RNA, 2′-alkoxy-RNA, 2′-O-methoxyethyl-RNA, 2′-amino-DNA, 2′-fluoro-DNA, arabino nucleic acid (ANA), 2′-fluoro-ANA and LNA nucleosides. It is advantageous if one or more of the modified nucleoside(s) is a locked nucleic acid (LNA).


In some embodiments, the contiguous nucleotide sequence comprises LNA nucleosides.


In some embodiments, the contiguous nucleotide sequence comprises LNA nucleosides and DNA nucleosides.


In some embodiments, the contiguous nucleotide sequence comprises 2′-O-methoxyethyl (2′MOE) nucleosides.


In some embodiments, the contiguous nucleotide sequence comprises 2′-O-methoxyethyl (2′MOE) nucleosides and DNA nucleosides.


Advantageously, the 3′ most nucleoside of the antisense oligonucleotide, or contiguous nucleotide sequence thereof is a 2′ sugar modified nucleoside.


In a further embodiment the nucleic acid molecule comprises at least one modified internucleoside linkage. Suitable internucleoside modifications are described in the “Definitions” section under “Modified internucleoside linkage”.


Advantageously, the oligonucleotide comprises at least one modified internucleoside linkage, such as phosphorothioate or phosphorodithioate.


In some embodiments, at least one internucleoside linkage in the contiguous nucleotide sequence is a phosphodiester internucleoside linkages.


It is advantageous if at least 2 to 3 internucleoside linkages at the 5′ or 3′ end of the oligonucleotide are phosphorothioate internucleoside linkages.


For single stranded antisense oligonucleotides it is advantageous if at least 75%, such as all, the internucleoside linkages within the contiguous nucleotide sequence are phosphorothioate internucleoside linkages. In some embodiments, all the internucleotide linkages in the contiguous sequence of the single stranded antisense oligonucleotide are phosphorothioate linkages.


In an advantageous embodiment of the invention the antisense oligonucleotide of the invention is capable of recruiting RNase H, such as RNase H1. An advantageous structural design is a gapmer design as described in the “Definitions” section under for example “Gapmer”, “LNA Gapmer” and “MOE gapmer”. In the present invention it is advantageous if the antisense oligonucleotide of the invention is a gapmer with an F-G-F′ design.


In all instances the F-G-F′ design may further include region D′ and/or D″ as described in the “Definitions” section under “Region D′ or D” in an oligonucleotide“.


The invention provides antisense oligonucleotides according to the invention, such as antisense oligonucleotides 12-24, such as 12-18 in length, nucleosides in length wherein the antisense oligonucleotide comprises a contiguous nucleotide sequence comprising at least 14, such as at least 15, such as 16 contiguous nucleotides present in SEQ ID NO 23.


The invention provides antisense oligonucleotides according to the invention, such as antisense oligonucleotides 12-24, such as 12-18 in length, nucleosides in length wherein the antisense oligonucleotide comprises a contiguous nucleotide sequence comprising at least 14, such as at least 15, such as 16 contiguous nucleotides present in SEQ ID NO 24.


The invention provides antisense oligonucleotides according to the invention, such as antisense oligonucleotides 12-24, such as 12-18 in length, nucleosides in length wherein the antisense oligonucleotide comprises a contiguous nucleotide sequence comprising at least 14, such as at least 15, such as 16 contiguous nucleotides present in SEQ ID NO 25.


The invention provides LNA gapmers according to the invention comprising or consisting of a contiguous nucleotide sequence shown in SEQ ID NO 23, 24 or 25. In some embodiments, the LNA gapmer is a LNA gapmer with CMP ID NO: 23_1, 24_1 or 25_1 in Table 7.


In a further aspect, of the invention the nucleic acid molecules, such as the antisense oligonucleotide, siRNA or shRNA, of the invention can be targeted directly to the liver by covalently attaching them to a conjugate moiety capable of binding to the asialoglycoprotein receptor (ASGPr), such as divalent or trivalent GaINAc cluster.


Conjugates Since HBV infection primarily affects the hepatocytes in the liver it is advantageous to conjugate the COPS3 inhibitor to a conjugate moiety that will increase the delivery of the inhibitor to the liver compared to the unconjugated inhibitor. In one embodiment, liver targeting moieties are selected from moieties comprising cholesterol or other lipids or conjugate moieties capable of binding to the asialoglycoprotein receptor (ASGPR).


In some embodiments, the invention provides a conjugate comprising a nucleic acid molecule of the invention covalently attached to a conjugate moiety.


The asialoglycoprotein receptor (ASGPR) conjugate moiety comprises one or more carbohydrate moieties capable of binding to the asialoglycoprotein receptor (ASPGR targeting moieties) with affinity equal to or greater than that of galactose. The affinities of numerous galactose derivatives for the asialoglycoprotein receptor have been studied (see for example: Jobst, S. T. and Drickamer, K. JB.C. 1996, 271, 6686) or are readily determined using methods typical in the art.


In one embodiment, the conjugate moiety comprises at least one asialoglycoprotein receptor targeting moiety selected from group consisting of galactose, galactosamine, N-formyl-galactosamine, N-acetylgalactosamine, N-propionyl-galactosamine, N-n-butanoyl-galactosamine and N-isobutanoylgalactosamine. Advantageously, the asialoglycoprotein receptor targeting moiety is N-acetylgalactosamine (GaINAc).


To generate the ASGPR conjugate moiety the ASPGR targeting moieties (preferably GaINAc) can be attached to a conjugate scaffold. Generally, the ASPGR targeting moieties can be at the same end of the scaffold. In one embodiment, the conjugate moiety consists of two to four terminal GaINAc moieties linked to a spacer which links each GaINAc moiety to a brancher molecule that can be conjugated to the antisense oligonucleotide.


In a further embodiment, the conjugate moiety is mono-valent, di-valent, tri-valent or tetra-valent with respect to asialoglycoprotein receptor targeting moieties. Advantageously, the asialoglycoprotein receptor targeting moiety comprises N-acetylgalactosamine (GaINAc) moieties.


GaINAc conjugate moieties can include, for example, those described in WO 2014/179620 and WO 2016/055601 and PCT/EP2017/059080 (hereby incorporated by reference), as well as small peptides with GaINAc moieties attached such as Tyr-Glu-Glu-(aminohexyl GaINAc)3 (YEE(ahGalNAc)3; a glycotripeptide that binds to asialoglycoprotein receptor on hepatocytes, see, e.g., Duff, et al., Methods Enzymol, 2000, 313, 297); lysine-based galactose clusters (e.g., L3G4; Biessen, et al., Cardovasc. Med., 1999, 214); and cholane-based galactose clusters (e.g., carbohydrate recognition motif for asialoglycoprotein receptor).


The ASGPR conjugate moiety, in particular a trivalent GaINAc conjugate moiety, may be attached to the 3′- or 5′-end of the oligonucleotide using methods known in the art. In one embodiment, the ASGPR conjugate moiety is linked to the 5′-end of the oligonucleotide.


In one embodiment, the conjugate moiety is a tri-valent N-acetylgalactosamine (GaINAc), such as those shown in FIG. 1A-1 to FIG. 1D-2. In one embodiment, the conjugate moiety is the tri-valent N-acetylgalactosamine (GaINAc) of FIG. 1A-1 or FIG. 1A-2, or a mixture of both. In one embodiment, the conjugate moiety is the tri-valent N-acetylgalactosamine (GaINAc) of FIG. 1B-1 or FIG. 1B-2, or a mixture of both. In one embodiment, the conjugate moiety is the tri-valent N-acetylgalactosamine (GaINAc) of FIG. 1C-1 or FIG. 1C-2, or a mixture of both. In one embodiment, the conjugate moiety is the tri-valent N-acetylgalactosamine (GaINAc) of FIG. 1D-1 or FIG. 1D-2, or a mixture of both.


Method of Manufacture


In a further aspect, the invention provides methods for manufacturing the oligonucleotides of the invention comprising reacting nucleotide units and thereby forming covalently linked contiguous nucleotide units comprised in the oligonucleotide. Preferably, the method uses phophoramidite chemistry (see for example Caruthers et al, 1987, Methods in Enzymology vol. 154, pages 287-313). In a further embodiment, the method further comprises reacting the contiguous nucleotide sequence with a conjugating moiety (ligand) to covalently attach the conjugate moiety to the oligonucleotide. In a further aspect, a method is provided for manufacturing the composition of the invention, comprising mixing the oligonucleotide or conjugated oligonucleotide of the invention with a pharmaceutically acceptable diluent, solvent, carrier, salt and/or adjuvant.


Pharmaceutical Salt


The compounds according to the present invention may exist in the form of their pharmaceutically acceptable salts. The term “pharmaceutically acceptable salt” refers to conventional acid-addition salts or base-addition salts that retain the biological effectiveness and properties of the compounds of the present invention.


In a further aspect, the invention provides a pharmaceutically acceptable salt of the nucleic acid molecules or a conjugate thereof, such as a pharmaceutically acceptable sodium salt, ammonium salt or potassium salt.


Pharmaceutical Composition


In a further aspect, the invention provides pharmaceutical compositions comprising any of the compounds of the invention, in particular the aforementioned nucleic acid molecules and/or nucleic acid molecule conjugates or salts thereof and a pharmaceutically acceptable diluent, carrier, salt and/or adjuvant. A pharmaceutically acceptable diluent includes phosphate-buffered saline (PBS) and pharmaceutically acceptable salts include, but are not limited to, sodium and potassium salts. In some embodiments, the pharmaceutically acceptable diluent is sterile phosphate buffered saline. In some embodiments, the nucleic acid molecule is used in the pharmaceutically acceptable diluent at a concentration of 50 to 300 μM solution.


Suitable formulations for use in the present invention are found in Remington's Pharmaceutical Sciences, Mack Publishing Company, Philadelphia, Pa., 17th ed., 1985. For a brief review of methods for drug delivery, see, e.g., Langer (Science 249:1527-1533, 1990). WO 2007/031091 provides further suitable and preferred examples of pharmaceutically acceptable diluents, carriers and adjuvants (hereby incorporated by reference). Suitable dosages, formulations, administration routes, compositions, dosage forms, combinations with other therapeutic agents, prodrug formulations are also provided in WO2007/031091.


In some embodiments, the nucleic acid molecule or the nucleic acid molecule conjugates of the invention, or pharmaceutically acceptable salt thereof is in a solid form, such as a powder, such as a lyophilized powder.


Compounds, nucleic acid molecules or nucleic acid molecule conjugates of the invention may be mixed with pharmaceutically acceptable active or inert substances for the preparation of pharmaceutical compositions or formulations. Compositions and methods for the formulation of pharmaceutical compositions are dependent upon a number of criteria, including, but not limited to, route of administration, extent of disease, or dose to be administered.


These compositions may be sterilized by conventional sterilization techniques, or may be sterile filtered. The resulting aqueous solutions may be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile aqueous carrier prior to administration. The pH of the preparations typically will be between 3 and 11, more preferably between 5 and 9 or between 6 and 8, and most preferably between 7 and 8, such as 7 to 7.5. The resulting compositions in solid form may be packaged in multiple single dose units, each containing a fixed amount of the above-mentioned agent or agents, such as in a sealed package of tablets or capsules. The composition in solid form can also be packaged in a container for a flexible quantity, such as in a squeezable tube designed for a topically applicable cream or ointment.


In some embodiments, the nucleic acid molecule or nucleic acid molecule conjugate of the invention is a prodrug. In particular, with respect to nucleic acid molecule conjugates the conjugate moiety is cleaved off the nucleic acid molecule once the prodrug is delivered to the site of action, e.g. the target cell.


Administration


The compounds, nucleic acid molecules or nucleic acid molecule conjugates or pharmaceutical compositions of the present invention may be administered topical (such as, to the skin, inhalation, ophthalmic or otic) or enteral (such as, orally or through the gastrointestinal tract) or parenteral (such as, intravenous, subcutaneous, intra-muscular, intracerebral, intracerebroventricular or intrathecal).


In a preferred embodiment the oligonucleotide or pharmaceutical compositions of the present invention are administered by a parenteral route including intravenous, intraarterial, subcutaneous, intraperitoneal or intramuscular injection or infusion. In one embodiment, the active nucleic acid molecule or nucleic acid molecule conjugate is administered intravenously. In another embodiment, the active nucleic acid molecule or nucleic acid molecule conjugate is administered subcutaneously.


In some embodiments, the nucleic acid molecule, nucleic acid molecule conjugate or pharmaceutical composition of the invention is administered at a dose of 0.1-15 mg/kg, such as from 0.2-10 mg/kg, such as from 0.25-5 mg/kg. The administration can be once a week, every second week, every third week or even once a month.


The invention also provides for the use of the COPS3 inhibitor, such as the nucleic acid molecule or nucleic acid molecule conjugate of the invention as described for the manufacture of a medicament wherein the medicament is in a dosage form for subcutaneous administration.


Combination Therapies


In some embodiments, the inhibitor of the present invention such as the nucleic acid molecule, nucleic acid molecule conjugate or pharmaceutical composition of the invention is for use in a combination treatment with another therapeutic agent. The therapeutic agent can for example be the standard of care for the diseases or disorders described above.


By way of example, the COPS3 inhibitor, such as the nucleic acid molecule or the nucleic acid molecule conjugate of the present invention may be used in combination with other actives, such as oligonucleotide-based antivirals—such as sequence specific oligonucleotide-based antivirals -acting either through antisense (including other LNA oligomers), siRNAs (such as ARC520), aptamers, morpholinos or any other antiviral, nucleotide sequence-dependent mode of action.


By way of further example, the COPS3 inhibitor, such as the nucleic acid molecule or the nucleic acid molecule conjugate of the present invention may be used in combination with other actives, such as immune stimulatory antiviral compounds, such as interferon (e.g. pegylated interferon alpha), TLR7 agonists (e.g. GS-9620), or therapeutic vaccines.


By way of further example, the COPS3 inhibitor, such as the nucleic acid molecule or the nucleic acid molecule conjugate of the present invention may be used in combination with other actives, such as small molecules, with antiviral activity. These other actives could be, for example, nucleoside/nucleotide inhibitors (eg entecavir or tenofovir disoproxil fumarate), encapsidation inhibitors, entry inhibitors (eg Myrcludex B).


In certain embodiments, the additional therapeutic agent may be an HBV agent, a Hepatitis C virus (HCV) agent, a chemotherapeutic agent, an antibiotic, an analgesic, a nonsteroidal anti-inflammatory (NSAID) agent, an antifungal agent, an antiparasitic agent, an anti-nausea agent, an anti-diarrheal agent, or an immunosuppressant agent.


In particular related embodiments, the additional HBV agent may be interferon alpha-2b, interferon alpha-2a, and interferon alphacon-1 (pegylated and unpegylated), ribavirin; an HBV RNA replication inhibitor; a second antisense oligomer; an HBV therapeutic vaccine; an HBV prophylactic vaccine; lamivudine (3TC); entecavir (ETV); tenofovir diisoproxil fumarate (TDF); telbivudine (LdT); adefovir; or an HBV antibody therapy (monoclonal or polyclonal).


In other particular related embodiments, the additional HCV agent may be interferon alpha-2b, interferon alpha-2a, and interferon alphacon-1 (pegylated and unpegylated); ribavirin; pegasys; an HCV RNA replication inhibitor (e.g., ViroPharma's VP50406 series); an HCV antisense agent; an HCV therapeutic vaccine; an HCV protease inhibitor; an HCV helicase inhibitor; or an HCV monoclonal or polyclonal antibody therapy.


Applications The nucleic acid molecules of the invention may be utilized as research reagents for, for example, diagnostics, therapeutics and prophylaxis.


In research, such nucleic acid molecules may be used to specifically modulate the synthesis of COPS3 protein in cells (e.g. in vitro cell cultures) and experimental animals thereby facilitating functional analysis of the target or an appraisal of its usefulness as a target for therapeutic intervention. Typically, the target modulation is achieved by degrading or inhibiting the mRNA producing the protein, thereby prevent protein formation or by degrading or inhibiting a modulator of the gene or mRNA producing the protein.


If employing the nucleic acid molecules of the invention in research or diagnostics the target nucleic acid may be a cDNA or a synthetic nucleic acid derived from DNA or RNA.


Also encompassed by the present invention is an in vivo or in vitro method for modulating COPS3 expression in a target cell which is expressing COPS3, said method comprising administering a nucleic acid molecule, conjugate compound or pharmaceutical composition of the invention in an effective amount to said cell.


In some embodiments, the target cell, is a mammalian cell in particular a human cell. The target cell may be an in vitro cell culture or an in vivo cell forming part of a tissue in a mammal. In preferred embodiments, the target cell is present in in the liver. The target cell may be a hepatocyte.


One aspect of the present invention is related the COPS3 inhibitor, such as the nucleic acid molecules, conjugate compounds or pharmaceutical compositions of the invention for use as a medicament.


In an aspect of the invention, the COPS3 inhibitor, such as the nucleic acid molecules, conjugate compound or pharmaceutical composition of the invention is capable of reducing the cccDNA level in the infected cells and therefore inhibiting HBV infection. In particular, the nucleic acid molecule is capable of affecting one or more of the following parameters i) reducing cccDNA and/or ii) reducing pgRNA and/or iii) reducing HBV DNA and/or iv) reducing HBV viral antigens in an infected cell.


For example, nucleic acid molecule that inhibits HBV infection may reduce i) the cccDNA levels in an infected cell by at least 40% such as 50% or 60% reduction compared to controls; or ii) the level of pgRNA by at least 40% such as 50% or 60% reduction compared to controls. The controls may be untreated cells or animals, or cells or animals treated with an appropriate negative control.


Inhibition of HBV infection may be measured in vitro using HBV infected primary human hepatocytes or in vivo using humanized hepatocytes PXB mouse model (available at PhoenixBio, see also Kakuni et al 2014 Int. J. Mol. Sci. 15:58-74). Inhibition of secretion of HBsAg and/or HBeAg may be measured by ELISA, e.g. by using the CLIA ELISA Kit (Autobio Diagnostic) according to the manufacturers' instructions. Reduction of intracellular cccDNA or HBV mRNA and pgRNA may be measured by qPCR, e.g. as described in the Materials and Methods section. Further methods for evaluating whether a test compound inhibits HBV infection are measuring secretion of HBV DNA by qPCR e.g. as described in WO 2015/173208 or using Northern Blot; in-situ hybridization, or immuno-fluorescence.


Due to the reduction of COPS3 levels the nucleic acid molecules, conjugate compounds or pharmaceutical compositions of the present invention can be used to inhibit development of or in the treatment of HBV infection. In particular, through the destabilization and reduction of the cccDNA, the nucleic acid molecules, conjugate compounds or pharmaceutical compositions of the present invention more efficiently inhibits development of or treats a chronic HBV infection as compared to a compound that only reduces secretion of HBsAg.


Accordingly, one aspect of the present invention is related to use of the COPS3 inhibitor, such as the nucleic acid molecule, conjugate compounds or pharmaceutical compositions of the invention to reduce cccDNA and/or pgRNA in an HBV infected individual.


A further aspect of the invention relates to the use of the COPS3 inhibitor, such as the nucleic acid molecules, conjugate compounds or pharmaceutical compositions of the invention to inhibit development of or treat a chronic HBV infection.


A further aspect of the invention relates to the use of the COPS3 inhibitor, such as the nucleic acid molecules, conjugate compounds or pharmaceutical compositions of the invention to reduce the infectiousness of a HBV infected person. In a particular aspect of the invention, the COPS3 inhibitor, such as the nucleic acid molecules, conjugate compounds or pharmaceutical compositions of the invention inhibits development of a chronic HBV infection.


The subject to be treated with the COPS3 inhibitor, such as the nucleic acid molecules, conjugate compounds or pharmaceutical compositions of the invention (or which prophylactically receives nucleic acid molecules, conjugate compounds or pharmaceutical compositions of the present invention) is preferably a human, more preferably a human patient who is HBsAg positive and/or HBeAg positive, even more preferably a human patient that is HBsAg positive and HBeAg positive.


Accordingly, the present invention relates to a method of treating a HBV infection, wherein the method comprises administering an effective amount of the COPS3 inhibitor, such as the nucleic acid molecules, conjugate compounds or pharmaceutical compositions of the invention. The present invention further relates to a method of preventing liver cirrhosis and hepatocellular carcinoma caused by a chronic HBV infection. In one embodiment, the COPS3 inhibitors of the present invention is not intended for the treatment of hepatocellular carcinoma, only its prevention.


The invention also provides for the use of a COPS3 inhibitor, such as a nucleic acid molecule, a conjugate compound or a pharmaceutical composition of the invention for the manufacture of a medicament, in particular a medicament for use in the treatment of HBV infection or chronic HBV infection or reduction of the infectiousness of a HBV infected person. In preferred embodiments, the medicament is manufactured in a dosage form for subcutaneous administration.


The invention also provides for the use of a COPS3 inhibitor, such as a nucleic acid molecule, a conjugate compound, the pharmaceutical composition of the invention for the manufacture of a medicament wherein the medicament is in a dosage form for intravenous administration.


The COPS3 inhibitor, such as the nucleic acid molecule, conjugate or the pharmaceutical composition of the invention may be used in a combination therapy. For example, the COPS3 inhibitor, such as the nucleic acid molecule, conjugate or the pharmaceutical composition of the invention may be combined with other anti-HBV agents such as interferon alpha-2b, interferon alpha-2a, and interferon alphacon-1 (pegylated and unpegylated), ribavirin, lamivudine (3TC), entecavir, tenofovir, telbivudine (LdT), adefovir, or other emerging anti-HBV agents such as a HBV RNA replication inhibitor, a HBsAg secretion inhibitor, a HBV capsid inhibitor, an antisense oligomer (e.g. as described in WO2012/145697, WO 2014/179629 and WO2017/216390), a siRNA (e.g. described in WO 2005/014806, WO 2012/024170, WO 2012/2055362, WO 2013/003520, WO 2013/159109, WO 2017/027350 and WO2017/015175), a HBV therapeutic vaccine, a HBV prophylactic vaccine, a HBV antibody therapy (monoclonal or polyclonal), or TLR 2, 3, 7, 8 or 9 agonists for the treatment and/or prophylaxis of HBV.


Embodiments of the Invention

The following embodiments of the present invention may be used in combination with any other embodiments described herein. The definitions and explanations provided herein above, in particular in the sections “SUMMARY OF INVENTION”, “DEFINITIONS” and DETAILED DESCRIPTION OF THE INVENTION″ apply mutatis mutandis to the following.


1. A COPS3 inhibitor for use in the in the treatment and/or prevention of Hepatitis B virus (HBV) infection.


2. The COPS3 inhibitor for the use of embodiment 1, wherein the COPS3 inhibitor is administered in an effective amount.


3. The COPS3 inhibitor for the use of embodiment 1 or 2, wherein the HBV infection is a chronic infection.


4. The COPS3 inhibitor for the use of embodiments 1 to 3, wherein the COPS3 inhibitor is capable of reducing cccDNA and/or pgRNA in an infected cell.


5. The COPS3 inhibitor for the use of any one of embodiments 1 to 4, wherein the COPS3 inhibitor prevents or reduces the association of COPS3 to cccDNA.


6. COPS3 inhibitor for the use of embodiment 5, wherein said inhibitor is a small molecule that specifically binds to COPS3 protein, wherein said inhibitor prevents or reduces association of COPS3 protein to cccDNA.


7. COPS3 inhibitor for the use of embodiment 6, wherein the COPS3 protein is encoded by SEQ ID NO: 4, 5, 6, 7, 8 or 9.


8. The COPS3 inhibitor for the use of any one of embodiments 1 to 7, wherein said inhibitor is a nucleic acid molecule of 12-60 nucleotides in length comprising or consisting of a contiguous nucleotide sequence of at least 12 nucleotides in length which is at least 90% complementary to a mammalian COPS3 target nucleic acid.


9. The COPS3 inhibitor for the use of embodiment 8, which is capable of reducing the level of the mammalian COPS3 target nucleic acid.


10. The COPS3 inhibitor for the use of embodiment 8 or 9, wherein the mammalian COPS3 target nucleic acid is RNA.


11. The COPS3 inhibitor for the use of embodiment 10, wherein the RNA is pre-mRNA.


12. The COPS3 inhibitor for the use of any one of embodiments 8 to 11, wherein the nucleic acid molecule is selected from the group consisting of antisense oligonucleotide, siRNA and shRNA.


13. The COPS3 inhibitor for the use of embodiment 12, wherein the nucleic acid molecule is a single stranded antisense oligonucleotide or a double stranded siRNA.


14. The COPS3 inhibitor for the use of any one of embodiments 8 to 13, wherein the mammalian COPS3 target nucleic acid is selected from the group consisting of SEQ ID NO: 1, 4, 5, 6,7,8 and 9.


15. The COPS3 inhibitor for the use of any one of embodiments 8 to 13, wherein the contiguous nucleotide sequence of the nucleic acid molecule is at least 98% complementary to the target nucleic acid of SEQ ID NO: 1 and SEQ ID NO: 2.


16. The COPS3 inhibitor for the use of any one of embodiments 8 to 13, wherein the contiguous nucleotide sequence of the nucleic acid molecule is at least 98% complementary to the target nucleic acid of SEQ ID NO: 1 and SEQ ID NO: 2 and SEQ ID NO: 3.


17. The COPS3 inhibitor for the use of any one of embodiments 1 to 16, wherein the cccDNA in an HBV infected cell is reduced by at least 50%, such as 60%, when compared to a control.


18. The COPS3 inhibitor for the use of any one of embodiments 1 to 16, wherein the pgRNA in an HBV infected cell is reduced by at least 50%, such as 60%, when compared to a control.


19. The COPS3 inhibitor for the use of any one of embodiments 8 to 18, wherein the mammalian COPS3 target nucleic acid is reduced by at least 50%, such as 60%, when compared to a control.


20. A nucleic acid molecule of 12 to 60 nucleotides in length which comprises or consists of a contiguous nucleotide sequence of 12 to 30 nucleotides in length wherein the contiguous nucleotide sequence is at least 90% complementary, such as 95%, such as 98%, such as fully complementary, to a mammalian COPS3 target nucleic acid.


21. The nucleic acid molecule of embodiment 20, wherein the nucleic acid molecule is chemically produced.


22. The nucleic acid molecule of embodiment 20 or 21, wherein the mammalian COPS3 target nucleic acid is selected from the group consisting of SEQ ID NO: 1, 4, 5, 6, 7, 8 and 9.


23. The nucleic acid molecule of embodiment 20 or 21, wherein the contiguous nucleotide sequence is at least 98% complementary to the target nucleic acid of SEQ ID NO: 1 and SEQ ID NO: 2.


24. The nucleic acid molecule of embodiment 20 or 21, wherein the contiguous nucleotide sequence is fully complementary to the target nucleic acid of SEQ ID NO: 1 and SEQ ID NO: 2 and SEQ ID NO: 3.


25. The nucleic acid molecule of any one of embodiments 20 to 23, wherein the nucleic acid molecule is 12 to 30 nucleotides in length.


26. The nucleic acid molecule of any one of embodiments 20 to 25, wherein the nucleic acid molecule is a RNAi molecule, such as a double stranded siRNA or shRNA.


27. The nucleic acid molecule of any one of embodiments 20 to 25, wherein the nucleic acid molecule is a single stranded antisense oligonucleotide.


28. The nucleic acid molecule of any one of embodiments 20 to 27, wherein the contiguous nucleotide sequence is fully complementary to a target nucleic acid sequence selected from Table 4 or Table 5.


29. The nucleic acid molecule of any one of embodiments 20 to 28, which is capable of hybridizing to a target nucleic acid of SEQ ID NO: 1 and SEQ ID NO: 2 with a ΔG° below −15 kcal.


30. The nucleic acid molecule of any one of embodiments 20 to 29, wherein the contiguous nucleotide sequence comprises or consists of at least 14 contiguous nucleotides, particularly 15, 16, 17, 18, 19, 20, 21 or 22 contiguous nucleotides.


31. The nucleic acid molecule of any one of embodiments 20 to 29, wherein the contiguous nucleotide sequence comprises or consists of from 14 to 22 nucleotides.


32. The nucleic acid molecule of embodiment 31, wherein the contiguous nucleotide sequence comprises or consists of 16 to 20 nucleotides.


33. The nucleic acid molecule of any one of embodiments 20 to 32, wherein the nucleic acid molecule comprises or consists of 14 to 25 nucleotides in length.


34. The nucleic acid molecule of embodiment 33, wherein the nucleic acid molecule comprises or consists of at least one oligonucleotide strand of 16 to 22 nucleotides in length.


35. The nucleic acid molecule of any one of embodiment 20 to 34, wherein the contiguous nucleotide sequence is fully complementary to a target sequence selected from the group consisting of SEQ ID NOs: 10, 11, 12 and 13.


36. The nucleic acid molecule of any one of embodiments 20 to 35, wherein the contiguous nucleotide sequence has zero to three mismatches compared to the mammalian COPS3 target nucleic acid it is complementary to.


37. The nucleic acid molecule of embodiment 36, wherein the contiguous nucleotide sequence has one mismatch compared to the mammalian COPS3 target nucleic acid.


38. The nucleic acid molecule of embodiment 36, wherein the contiguous nucleotide sequence has two mismatches compared to the mammalian COPS3 target nucleic acid.


39. The nucleic acid molecule of embodiment 36, wherein the contiguous nucleotide sequence is fully complementary to the mammalian COPS3 target nucleic acid.


40. The nucleic acid molecule of any one of embodiments 20 to 39, comprising one or more modified nucleosides.


41. The nucleic acid molecule of embodiment 40, wherein the one or more modified nucleosides are high-affinity modified nucleosides.


42. The nucleic acid molecule of embodiment 40 or 41, wherein the one or more modified nucleosides are 2′ sugar modified nucleosides.


43. The nucleic acid molecule of embodiment 42, wherein the one or more 2′ sugar modified nucleosides are independently selected from the group consisting of 2′-O-alkyl-RNA, 2′-O-methyl-RNA, 2′-alkoxy-RNA, 2′-O-methoxyethyl-RNA, 2′-amino-DNA, 2′-fluoro-DNA, 2′-fluoro-ANA and LNA nucleosides.


44. The nucleic acid molecule of any one of embodiments 40 to 43, wherein the one or more modified nucleosides are LNA nucleosides.


45. The nucleic acid molecule of embodiment 44, wherein the modified LNA nucleosides are selected from the group consisting of oxy-LNA, amino-LNA, thio-LNA, cET, and ENA.


46. The nucleic acid molecule of embodiment 44 or 45, wherein the modified LNA nucleosides are oxy-LNA with the following 2′-4′ bridge —O—CH2-.


47. The nucleic acid molecule of embodiment 46, wherein the oxy-LNA is beta-D-oxy-LNA.


48. The nucleic acid molecule of embodiment 44 or 45, wherein the modified LNA nucleosides are cET with the following 2′-4′ bridge —O—CH(CH3)—.


49. The nucleic acid molecule of embodiment 48, wherein the cET is (S)cET, i.e. 6′(S)methyl-beta-D-oxy-LNA.


50. The nucleic acid molecule of embodiment 44 or 45, wherein the LNA is ENA, with the following 2′-4′ bridge —O—CH2—CH2-.


51. The nucleic acid molecule of any one of embodiments 20 to 50, wherein the nucleic acid molecule comprises at least one modified internucleoside linkage.


52. The nucleic acid molecule of embodiment 51, wherein the at least one modified internucleoside linkage is a phosphorothioate internucleoside linkage.


53. The nucleic acid molecule of any one of embodiments 20 to 52, wherein the nucleic acid molecule is an antisense oligonucleotide capable of recruiting RNase H.


54. The nucleic acid molecule of embodiment 53, wherein the antisense oligonucleotide or the contiguous nucleotide sequence is a gapmer.


55. The nucleic acid molecule of embodiment 54, wherein the antisense oligonucleotide or contiguous nucleotide sequence thereof consists of or comprises a gapmer of formula 5′-F-G-F′-3′, where region F and F′ independently comprise or consist of 1-4 2′ sugar modified nucleosides and G is a region between 6 and 18 nucleosides which are capable of recruiting RNase H.


56. The nucleic acid molecule of embodiment 55, wherein the 1-4 2′ sugar modified nucleosides are independently selected from the group consisting of 2′-O-alkyl-RNA, 2′-O-methyl-RNA, 2′-alkoxy-RNA, 2′-O-methoxyethyl-RNA, 2′-amino-DNA, 2′-fluoro-DNA, arabino nucleic acid (ANA), 2′-fluoro-ANA and LNA nucleosides.


57. The nucleic acid molecule of embodiment 55 or 56, wherein one or more of the 1-4 2′ sugar modified nucleosides in region F and F′ are LNA nucleosides.


58. The nucleic acid molecule of embodiment 57, wherein all the 2′ sugar modified nucleosides in region F and F′ are LNA nucleosides.


59. The nucleic acid molecule of any one of embodiments 56 to 58, wherein the LNA nucleosides are selected from the group consisting of beta-D-oxy-LNA, alpha-L-oxy-LNA, beta-D-amino-LNA, alpha-L-amino-LNA, beta-D-thio-LNA, alpha-L-thio-LNA, (S)cET, (R)cET beta-D-ENA and alpha-L-ENA.


60. The nucleic acid molecule of any one of embodiments 56 to 59, wherein region F and F′ consist of identical LNA nucleosides.


61. The nucleic acid molecule of any one of embodiments 56 to 60, wherein all the 2′ sugar modified nucleosides in region F and F′ are oxy-LNA nucleosides.


62. The nucleic acid molecule of any one of embodiments 55 to 61, wherein the nucleosides in region G are DNA nucleosides.


63. The nucleic acid molecule of embodiment 62, wherein region G consists of at least 75% DNA nucleosides.


64. The nucleic acid molecule of embodiment 63, where all the nucleosides in region G are DNA nucleosides.


65. A conjugate compound comprising a nucleic acid molecule according to any one of embodiments 20 to 64, and at least one conjugate moiety covalently attached to said nucleic acid molecule.


66. The conjugate compound of embodiment 65, wherein the nucleic acid molecule is a double stranded siRNA and the conjugate moiety is covalently attached to the sense strand of the siRNA.


67. The conjugate compound of embodiment 65 or 66, wherein the conjugate moiety is selected from carbohydrates, cell surface receptor ligands, drug substances, hormones, lipophilic substances, polymers, proteins, peptides, toxins, vitamins, viral proteins or combinations thereof.


68. The conjugate compound of any one of embodiments 65 to 67, wherein the conjugate moiety is capable of binding to the asialoglycoprotein receptor.


69. The conjugate compound of embodiment 68, wherein the conjugate moiety comprises at least one asialoglycoprotein receptor targeting moiety selected from group consisting of galactose, galactosamine, N-formyl-galactosamine, N-acetylgalactosamine, N-propionyl-galactosamine, N-n-butanoyl-galactosamine and N-isobutanoylgalactosamine.


70. The conjugate compound of embodiment 69, wherein the asialoglycoprotein receptor targeting moiety is N-acetylgalactosamine (GaINAc).


71. The conjugate compound of embodiment 69 or 70, wherein the conjugate moiety is mono-valent, di-valent, tri-valent or tetra-valent with respect to asialoglycoprotein receptor targeting moieties.


72. The conjugate compound of embodiment 71, wherein the conjugate moiety consists of two to four terminal GaINAc moieties and a spacer linking each GaINAc moiety to a brancher molecule that can be conjugated to the antisense compound.


73. The conjugate compound of embodiment 72, wherein the spacer is a PEG spacer.


74. The conjugate compound of any one of embodiments 68 to 73, wherein the conjugate moiety is a tri-valent N-acetylgalactosamine (GaINAc) moiety.


75. The conjugate compound of any one of embodiments 68 to 74, wherein the conjugate moiety is selected from one of the trivalent GaINAc moieties in FIG. 1A-1 to FIG. 1K.


76. The conjugate compound of embodiment 75, wherein the conjugate moiety is the trivalent


GaINAc moiety in FIG. 1D-1 or FIG. 1D-2, ora mixture of both.


77. The conjugate compound of any one of embodiments 65 to 76, comprising a linker which is positioned between the nucleic acid molecule and the conjugate moiety.


78. The conjugate compound of embodiment 77, wherein the linker is a physiologically labile linker.


79. The conjugate compound of embodiment 78, wherein the physiologically labile linker is nuclease susceptible linker.


80. The conjugate compound of embodiment 78 or 79, wherein the physiologically labile linker is composed of 2 to 5 consecutive phosphodiester linkages.


81. The conjugate compound of any one of embodiments 68 to 80, which display improved cellular distribution between liver vs. kidney or improved cellular uptake into the liver of the conjugate compound as compared to an unconjugated nucleic acid.


82. A pharmaceutical composition comprising a nucleic acid molecule of any one of embodiments 20 to 64, a conjugate compound of embodiment 65 to 81, or acceptable salts thereof, and a pharmaceutically acceptable diluent, carrier, salt and/or adjuvant.


83. A method for identifying a compound that prevents, ameliorates and/or inhibits a hepatitis B virus (HBV) infection, comprising:

    • a. contacting a test compound with
      • i. a COPS3 polypeptide; or
      • ii. a cell expressing COPS3;
    • b. measuring the expression and/or activity of COPS3 in the presence or absence of said test compound; and
    • c. identifying a compound that reduces the expression and/or activity COPS3 and reduces cccDNA.


84. An in vivo or in vitro method for modulating COPS3 expression in a target cell which is expressing COPS3, said method comprising administering the nucleic acid molecule of any one of embodiments 20 t 64, a conjugate compound of any one of embodiments 65 to 81 or the pharmaceutical composition of embodiment 82 in an effective amount to said cell.


85. The method of embodiment 84, wherein the COPS3 expression is reduced by at least 50%, or at least 60% in the target cell compared to the level without any treatment or treated with a control.


86. The method of embodiment 84, wherein the target cell is infected with HBV and the cccDNA in an HBV infected cell is reduced by at least 50%, or at least 60% in the HBV infected target cell compared to the level without any treatment or treated with a control.


87. A method for treating or preventing a disease, such as HBV infection, comprising administering a therapeutically or prophylactically effective amount of the nucleic acid molecule any one of embodiments 20 to 64, a conjugate compound of any of embodiments 65 to 81, or the pharmaceutical composition of embodiment 82 to a subject suffering from or susceptible to the disease.


88. The nucleic acid molecule of any one of embodiments 20 to 64, or the conjugate compound of any one of embodiments 65 to 81 or the pharmaceutical composition of embodiment 82, for use as a medicament for treatment or prevention of a disease, such as HBV infection, in a subject.


89. Use of the nucleic acid molecule of any one of embodiments 20 to 64, or the conjugate compound of any one of embodiments 65 to 81 for the preparation of a medicament for treatment or prevention of a disease, such as HBV infection, in a subject.


90. The method, the nucleic acid molecule, the antisense oligonucleotide, the conjugate compound or the use of any one of embodiments 87 to 89, wherein the subject is a mammal.


91. The method, the nucleic acid molecule, the conjugate compound, or the use of embodiment 90, wherein the mammal is human.


92. The conjugate compound of embodiment 75, wherein the conjugate moiety is the trivalent GaINAc moiety of FIG. 1B-1 or FIG. 1B-2, or a mixture of both.


The invention will now be illustrated by the following examples which have no limiting character.


Examples

Materials and Methods


siRNA Sequences and Compounds









TABLE 6A







Human COPS3 sequences targeted by the individual


components of the siRNA pool










SEQ ID

Position on SEQ ID



NO:
COPS3 target sequence
NO: 1
Exon





10
GCACAAGUGUAUUCAACCA
20827-20845
8





11
CAAUGCAUACCACGAGUUA
20808-20826
8





12
CAAACCAGCUGACCUCAAU
13352-13370
5





13
GAAUUGGCAUCCUUAAGCA
13307-13325
5









The pool of siRNA (ON-TARGETplus SMART pool siRNA Cat. No. W-011494-00-0005, Dharmacon) contains four individual siRNA molecules targeting the sequences listed in the above table.









TABLE 6B







Control compounds















SEQ





Sequence
ID


Name
Supplier
Order number
5′ to 3′ sense strand
NO





Non-targeting
Dharmacon
#D-001810-01-
UGGUUUACAUGUCGACUAA
14


negative control

05




siRNA#1









Hbx positive
GA life
Custom made
GCACUUCGCUUCACCUCUG
15


control
science









Oligonucleotide Synthesis


Oligonucleotide synthesis is generally known in the art. Below is a protocol which may be applied. The oligonucleotides of the present invention may have been produced by slightly varying methods in terms of apparatus, support and concentrations used.


Oligonucleotides are synthesized on uridine universal supports using the phosphoramidite approach on an Oligomaker 48 at 1 μmol scale. At the end of the synthesis, the oligonucleotides are cleaved from the solid support using aqueous ammonia for 5-16 hours at 60° C. The oligonucleotides are purified by reverse phase HPLC (RP-HPLC) or by solid phase extractions and characterized by UPLC, and the molecular mass is further confirmed by ESI-MS.


Elongation of the oligonucleotide: The coupling of β-cyanoethyl- phosphoramidites (DNA-A(Bz), DNA- G(ibu), DNA- C(Bz), DNA-T, LNA-5-methyl-C(Bz), LNA-A(Bz), LNA- G(dmf), or LNA-T) is performed by using a solution of 0.1 M of the 5′-O-DMT-protected amidite in acetonitrile and DCI (4,5—dicyanoimidazole) in acetonitrile (0.25 M) as activator. For the final cycle a phosphoramidite with desired modifications can be used, e.g. a C6 linker for attaching a conjugate group or a conjugate group as such. Thiolation for introduction of phosphorthioate linkages is carried out by using xanthane hydride (0.01 M in acetonitrile/pyridine 9:1). Phosphordiester linkages can be introduced using 0.02 M iodine in THF/Pyridine/water 7:2:1. The rest of the reagents are the ones typically used for oligonucleotide synthesis.


For post solid phase synthesis conjugation a commercially available C6 aminolinker phorphoramidite can be used in the last cycle of the solid phase synthesis and after deprotection and cleavage from the solid support the aminolinked deprotected oligonucleotide is isolated. The conjugates are introduced via activation of the functional group using standard synthesis methods.


Purification by RP-HPLC:


The crude compounds are purified by preparative RP-HPLC on a Phenomenex Jupiter C18 10 μm 150×10 mm column. 0.1 M ammonium acetate pH 8 and acetonitrile is used as buffers at a flow rate of 5 mL/min. The collected fractions are lyophilized to give the purified compound typically as a white solid.


Abbreviations:

  • DCI: 4,5-Dicyanoimidazole
  • DCM: Dichloromethane
  • DMF: Dimethylformamide
  • DMT: 4,4′-Dimethoxytrityl
  • THF: Tetrahydrofurane
  • Bz: Benzoyl
  • Ibu: Isobutyryl
  • RP-HPLC: Reverse phase high performance liquid chromatography


Tm Assay:


Oligonucleotide and RNA target (phosphate linked, PO) duplexes are diluted to 3 mM in 500 ml RNase-free water and mixed with 500 ml 2x Tm-buffer (200 mM NaCl, 0.2 mM EDTA, 20 mM Naphosphate, pH 7.0). The solution is heated to 95° C. for 3 min and then allowed to anneal in room temperature for 30 min. The duplex melting temperatures (Tm) is measured on a Lambda 40 UV/VIS Spectrophotometer equipped with a Peltier temperature programmer PTP6 using PE Templab software (Perkin Elmer). The temperature is ramped up from 20° C. to 95° C. and then down to 25° C., recording absorption at 260 nm. First derivative and the local maximums of both the melting and annealing are used to assess the duplex Tm.


Clonal growth medium (dHCGM). dHCGM is a DMEM medium containing 100 U/mI Penicillin, 100 μg/ml Streptomycin, 20 mM Hepes, 44 mM NaHCO3, 15 μg/ml L-proline, 0.25 μg/ml insulin, 50 nM Dexamethazone, 5 ng/ml EGF, 0.1 mM Asc-2P, 2% DMSO and 10% FBS (Ishida et al., 2015). Cells were cultured at 37° C. incubator in a humidified atmosphere with 5% CO2. Culture medium was replaced 24 h post-plating and every 2 days until harvest.


ASOs Sequences and Compounds









TABLE 7







list of oligonucleotide motif sequences of the


invention (indicated by SEQ ID NO), as well as


specific oligonucleotide compounds of the inven-


tion (indicated by CMP ID NO) designed based


on the motif sequence.









SEQ ID NO
CMP ID NO
Oligonucleotide Compound





23
23_1
TGTtgtactttgcCAA





24
24_1
AAGcatggctgggtTA





25
25_1
GAggtccagacaacTG





The heading “Oligonucleotide compound” in the table represents specific designs of a motif sequence. Capital letters are beta-D-oxy LNA nucleosides, lowercase letters are DNA nucleosides, all LNA C are 5-methyl cytosine, all internucleoside linkages are phosphorothioate internucleoside linkages (CMP ID NO = Compound ID NO)






HBV Infected PHH Cells


Fresh primary human hepatocytes (PHH) were provided by PhoenixBio, Higashi-Hiroshima City, Japan (PXB-cells also described in Ishida et al 2015 Am J Pathol. 185(5):1275-85) in 70,000 cells/well in 96-well plate format.


Upon arrival, PHH were infected either with an MOI of 2 GE/mL using HepG2 2.2.15-derived HBV (batch Z12) or with an MOI of 7E08 GE/mL using chronic patient-derived purified inoculum (genotype C) by incubating the PHH cells with HBV in 4% (v/v) PEG in PHH medium for 16 hours. The cells were then washed three times with PBS and cultured a humidified atmosphere with 5% CO2 in fresh PHH medium consisting of DMEM (GIBCO, Cat#21885) supplemented with 10% (v/v) heat-inactivated fetal bovine serum (GIBCO, Cat#10082), 2% (v/v) DMSO, 1% (v/v) Penicillin/Streptomycin (GIBCO, Cat#15140-148), 20 mM HEPES (GIBCO, Cat#15630-080), 44 mM NaHCO3 (Wako, Cat#195-14515), 15 μg/ml L-proline (MP-Biomedicals, Cat#0219472825), 0.25 μg/ml Insulin (Sigma, Cat#11882), 50 nM Dexamethasone (Sigma, Cat# D8893), 5 ng/ml EGF (Sigma, Cat# E9644), and 0.1 mM L-Ascorbic acid 2-phosphate (Wako, Cat#013-12061). Cells were cultured at 37° C. incubator in a humidified atmosphere with 5% CO2. Culture medium was replaced 24 hours post-plating and three times a week until harvest.


siRNA Transfection


Four days post-infection the cells were transfected with the COPS3 siRNA pool in triplicates. No drug controls (NDC), negative control siRNA and HBx siRNA were included as controls (see Table 6A).


Per well a transfection mixture was prepared with 2 μl of either negative control siRNA (stock concentration 1 uM), COPS3 siRNA pool (stock concentration 1 uM), HBx control siRNA (stock concentration 0.12 uM) or H2O (NDC) with 18.2 μl OptiMEM (Thermo Fisher Scientific Reduced Serum media) and 0.6 μl Lipofectamine® RNAiMAX Transfection Reagent (Thermofisher Scientific catalog No. 13778). The transfection mixture was mixed and incubated at room temperature 5 minutes prior to transfection. Prior to transfection the medium was removed from the PHH cells and replaced by 100 μl/well William's E Medium+GlutaMAX (Gibco, #32551) supplemented with HepaRG supplement without P/S (Biopredic International, #ADD711C). 20 ul of transfection mix was added to each well yielding a final concentration of 16 nM for the negative control siRNA or COPS3 siRNA pool, or 1.92 nM for the HBx control siRNA and the plates gently rocked before placing into the incubator. The medium was replaced with PHH medium after 6 hours. The siRNA treatment was repeated on day 6 post-infection as described above. On day 8 post-infection the supernatants were harvested and stored at −20° C. HBsAg and HBeAg can be determined from the supernatants if desired.


LNA Treatment


Two LNA master mix plates from a 500 μM stock were prepared. For LNA treatment at a final concentration of 25 μM, 200 uL of a 500 μM stock LNA is prepared in the first master mix plate. A second master mix plate including COPS3 LNAs at 100 μM was prepared for LNA treatment at a final concentration of 5 μM, mixing 40 μL of each COPS3 LNA at 500 μM and 160 μL of PBS.


Four days post-infection the cells were treated with COPS3 LNAs at final concentration of 25 μM (see Table 7) in either duplicate or triplicates or with PBS as no drug control (NDC). Prior to the LNA treatment, the old medium was removed from the cells and replaced by 114 μl/well of fresh PHH medium. Per well, 6 μL of each COPS3 LNA either at 500 uM or PBS as NDC were added to the 114 μL PHH medium. The same treatment was repeted 3 times at day 4, 11 and 18 post-infection. Cell culture medium was changed with fresh one every three days at day 7, 14 and 21 post infection.


For the quantification of cccDNA, the infected cells were treated with entecavir (ETV) at 10 nM final concentration from day 7 to day 21 post infection. Fresh ETV treatment was repeated 5 times at day 7, 11, 14, 18 and 21 post infection. This ETV treatment was used to inhibit the synthesis of new viral DNA intermediates and to detect specifically HBV cccDNA sequences.


Measurement of HBV Antigen Expression


HBV antigen expression and secretion can be measured in the collected supernatants if desired. The HBV propagation parameters, HBsAg and HBeAg levels, are measured using CLIA ELISA Kits (Autobio Diagnostic #CL0310-2, #CL0312-2), according to the manufacturer's protocol. Briefly, 25 μL of supernatant per well is transferred to the respective antibody coated microtiter plate and 25 μL of enzyme conjugate reagent is added. The plate is incubated for 60 min on a shaker at room temperature before the wells are washed five times with washing buffer using an automatic washer. 25 μL of substrate A and B were added to each well. The plates are incubated on a shaker for 10 min at room temperature before luminescence is measured using an EnVision® luminescence reader (Perkin Elmer).


Cell Viability Measurements


The cell viability was measured on the supernatant free cells by the Cell Counting Kit— 8 (CCK8 from Sigma Aldrich, #96992). For the measurement the CCK8 reagent was diluted 1:10 in normal culture medium and 100 μl/well added to the cells. After 1h incubation in the incubator 80 μl of the supernatants were transferred to a clear flat bottom 96 well plate, and the absorbance at 450 nm was read using a microplate reader (Tecan). Absorbance values were normalized to the NDC which was set at 100% to calculate the relative cell viabilities.


Cell viability measurements are used to confirm that any reduction in the viral parameters is not the cause of cell death, the closer the value is to 100% the lower the toxicity. LNA treatment giving cellular viability values equal or below 20% to the NDC were excluded from further analysis.


Real-time PCR for measuring COPS3 mRNA expression and the viral parameters pgRNA, cccDNA and HBV DNA quantification


Following cell viability determination the cells were washed with PBS once. For siRNA treatment cells were lysed with 50 μl/well lysis solution from the TaqMan® Gene Expression Cells-to-CT™ Kit (Thermo Fisher Scientific, #AM1729) and stored at −80° C. For cells treated with LNAs, total RNA was extracted using a MagNA Pure robot and the MagNA Pure 96 Cellular RNA Large Volume Kit (Roche, #05467535001) according to the manufacturer's protocol. For quantification of COPS3 RNA and viral pgRNA levels and the normalization control, GUS B, the TaqMan® RNA-to-Ct™ 1-Step Kit (Life Technologies, #4392656) has been used. For each reaction 2 or 4 μl of cell lysate, 0.5 μl 20x COPS3 Taqman primer/probe, 0.5 μl 20x GUS B Taqman primer/probe, 5 μl 2x TaqMan® RT-PCR Mix, 0.25 μl 40x TaqMan® RT Enzyme Mix, and 1.75 μl DEPC-treated water is used. Primers used for GUS B RNA and target mRNA quantification are listed in Table 8. Technical replicates are run for each sample and minus RT controls included to evaluate potential amplification due to DNA present.


The target mRNA expression levels, as well as the viral pgRNA, were quantified in technical duplicates by RT-qPCR using a QuantStudio 12K Flex (Applied Biosystems) with the following protocol, 48° C. for 15 min, 95° C. for 10 min, then 40 cycles with 95° C. for 15 seconds, and 60° C. for 60 seconds.


COPS3 mRNA and pgRNA expression levels were analyzed using the comparative cycle threshold 2-ΔΔCt method normalized to the reference gene GUS B and non-transfected cells. The expression levels in siRNA-treated cells are presented as % of the average no-drug control samples (i.e. the lower the value the larger the inhibition/reduction). In LNA-treated cells, the expression levels are presented as inhibitory effect compared to non-treated cells (NDC) set as 100% and is expressed as a percentage of the mean+SD from two independent biological replicates are measured.For cccDNA quantification, total DNA was extracted from HBV infected Primary Human Hepatocytes treated with siRNA or with LNAs. Prior to the cccDNA qPCR analysis, a fraction of the siRNA treated cell lysate was digested with T5 enzyme (10U/500 ng DNA; New England Biolabs, #M0363L) to remove viral DNA intermediates and to quantify the cccDNA molecule only. T5 digestion was done at 37° C. for 30 min. T5 digestion was not applied on LNA treated cell lysates to avoid qPCR interference in the assay To remove HBV DNA intermediates and quantify cccDNA level in LNA treated cells, cells were treated with entecavir (10 nM) for 3 weeks as described in LNA treatment section


For the quantification of cccDNA in siRNA-treated cells, each reaction mix per well contained 2 μl T5-digested cell lysate, 0.5 μl 20x cccDNA_DANDRI Taqman primer/probe (Life Technologies, custom #AI1RW7N, FAM-dye listed in the Table below), 5 μl TaqMan® Fast Advanced Master Mix (Applied Biosystems, #4444557) and 2.5 p1 DEPC-treated water were used. Technical triplicates were run for each sample.














Primers for siRNA-treated cells









Primer

SEQ


name
Sequence
ID





CCCDNA_
CCGTGTGCACTTCGCTTCA
16


DANDRI_F







CCCDNA_
GCACAGCTTGGAGGCTTGA
17


DANDRI_R







CCCDNA_
5′-[6FAM]CATGGAGACCACCGTGAACGCCC
18


DANDRI_M
[BHQ1] -3′










Primers for LNA-treated cells









Primer




name
Sequence





CCCDNA_
5′- CGTCTGTGCCTTCTCATCTGC-3′
19


Fwd







CCCDNA_
5′- GCACAGCTTGGAGGCTTGAA -3′
20


Rev







Mito Fwd
CCGTCTGAACTATCCTGCCC
21





Mito Rev
GCCGTAGTCGGTGTACTCGT
22









For the quantification of cccDNA in LNA-treated cells by qPCR, a master mix of 16 uL/well, with 10ul 2× Fast SYBR™ Green Master Mix (Applied Biosystems, #4385614), 2ul cccDNA Primer Mix (1 uM of each forward and reverse), and 4ul nuclease-free water per well is prepared. A master mix with 10ul 2× Fast SYBR™ Green Master Mix (Applied Biosystems, #4385614), 2ul mitochondrial genome primer mix (1 uM of each forward and reverse), and 4ul nuclease-free water per well is also prepared for normalization of the cccDNA.


For quantification of intracellular HBV DNA and the normalization control, human hemoglobin beta (HBB), each reaction mix contained 2 μl undigested cell lysate, 0.5 μl 20x HBV Taqman primer/probe (Life Technologies, #Pa03453406_s1, FAM-dye), 0.5 μl 20x HBB Taqman primer/probe (Life Technologies, #Hs00758889_s1, VIC-dye), 5 μl TaqMan® Fast Advanced Master Mix (Applied Biosystems, #4444557) and 2 μl DEPC-treated water were used. Technical triplicates were run for each sample.


The qPCR was run on the QuantStudio™ K12 Flex with standard settings for the fast heating block (95° C. for 20 seconds, then 40 cycles with 95° C. for 1 second and 60C for 20 seconds).


Any outliers were removed from the data set by excluding values with more than 0.9 difference to the median Ct of all the three biological replicates for each treatment condition. Fold changes of cccDNA (siRNA and LNA treated cells) and total HBV DNA (only siRNA treated cells) were determined from the Ct values via the 2-ddCT method and normalized to the HBB or mitochondrial DNA as housekeeping genes. For siRNA-treated cells, expression levels are presented as % of the average no drug control samples (i.e. the lower the value the larger the inhibition/reduction). For LNA treated cells, the inhibitory effect on cccDNA was expressed as a percentage of the mean+/−SD from three independent biological replicates compared to non-treated cells (NDC) set as 100%.









TABLE 8





GUS B and COPS3 mRNA qPCR primers


(Thermo Fisher Scientific)

















COPS3 (FAM): Hs00182547_m1



Housekeeping gene primers GUS B (VIC): Hs00939627_m1



pgRNA (FAM): AILIKX5










Example 1: Measurement of the Reduction of COPS3 mRNA, HBV Intracellular DNA and cccDNA in HBV Infected PHH Cells Resulting from siRNA Treatment

In the following experiment, the effect of COPS3 knock-down on the HBV parameters, HBV DNA and cccDNA, was tested.


HBV infected PHH cells were treated with the pool of siRNAs from Dharmacon (LU-011494-00-0005, see Table 6A) as described in the Materials and Methods section “siRNA transfection”. Following the 4 days-treatment, COPS3 mRNA, cccDNA and intracellular HBV DNA were measured by qPCR as described in the Materials and Methods section “Real-time PCR for measuring COPS3 mRNA expression and the viral parameters pgRNA, cccDNA, and HBV DNA”.


The results are shown in Table 9 as % of the average no drug control samples (i.e. the lower the value the larger the inhibition/reduction)









TABLE 9







Effect on HBV parameters following knockdown of COPS3


with pool of siRNA. Values are given as the


average of biological and technical triplicates.












HBV




COPS3
intracellular




mRNA
DNA
cccDNA













Treatment
Mean
SD
Mean
SD
Mean
SD
















COPS3 siRNA
20
2
42
13
31
8


HBx positive
ND
ND
52
9
74
14


control








siRNA negative
ND
ND
103
21
67
2


control











ND = not determined






From this, it can be seen that the COPS3 siRNA pool is capable of reducing COPS3 mRNA, cccDNA as well as HBV DNA quite efficiently. The positive control reduce intracellular HBV DNA as expected but had no effect on cccDNA when compared to the negative control.


Example 2: Measurement of the Reduction of COPS3 mRNA, HBV Intracellular pgRNA and cccDNA in HBV Infected PHH Cells Resulting from LNA Treatment

In the following experiment, the effect of COPS3 knock-down on the HBV parameters, HBV DNA and cccDNA, was tested.


HBV infected PHH cells were treated with COPS3 naked LNAs (see Table 7) as described in the Materials and Methods section “LNA treatment”.


Following 21 days-treatment, COPS3 mRNA, cccDNA, and intracellular HBV pgRNA were measured by qPCR as described in the Materials and Methods section “Real-time PCR for measuring COPS3 mRNA expression and the viral parameters pgRNA, cccDNA, and HBV DNA”.


The results are shown in Table 10 as inhibitory effect compared to non-treated cells (NDC) set as 100% and are expressed as a percentage of the mean+SD from two independent biological replicates are measured.









TABLE 10







Effect on HBV parameters following knockdown of COPS3 with


naked LNAs. Values are given as the average of either two or


three biological replicates. Data show the effect with LNA at a


final concentration of 25 mM.










CMP
COPS3 mRNA
pgRNA
cccDNA













ID
Mean %
SD
Mean %
SD
Mean %
SD
















23_1
4.66%
0.08%
44.56%
0.54%
50.79%
2.79%


24_1
34.44%
0.85%
69.52%
5.25%
61.28%
2.42%


25_1
48.00%
0.22%
36.92%
1.98%
63.28%
5.36%


NDC
100.00%
0.00%
100.00%
0.00%
96.42%
4.10%









From this, it can be seen that SCAMP3 LNAs are capable of sensibly reducing SCAMP3 mRNA expression resulting in a quite efficient reduction in expression level for both pgRNA and cccDNA.

Claims
  • 1. A method of treating or preventing a Hepatitis B virus (HBV) infection in a subject in need thereof, the method comprising administering to the subject a therapeutically or prophylactically effective amount of a COPS3 (COPS Signalosome Subunit 3) inhibitor.
  • 2. The method according to claim 1, wherein the HBV infection is a chronic infection, and/or wherein the COPS3 inhibitor is capable of reducing the amount of cccDNA (covalently closed circular DNA) in an HBV infected cell.
  • 3. (canceled)
  • 4. The method according to claim 1, wherein said inhibitor is an nucleic acid molecule of 12 to 60 nucleotides in length comprising a contiguous nucleotide sequence of at least 12 nucleotides in length which is at least 95% complementary to a mammalian COPS3 target nucleic acid and is capable of reducing the expression of COPS3 mRNA.
  • 5. The method according to claim 1, wherein said inhibitor is selected from the group consisting of a single stranded antisense oligonucleotide, an siRNA and a shRNA.
  • 6. The method according to claim 4, wherein the mammalian COPS3 target sequence is selected from the group consisting of SEQ ID NOs: 1, 4, 5, 6, 7, 8, and 9.
  • 7. The method according to claim 4, wherein the contiguous nucleotide sequence is at least 98% complementary to the target sequence of SEQ ID NO: 1 and SEQ ID NO: 2.
  • 8. The method according to claim 3, wherein the amount of cccDNA in the HBV infected cell is reduced by at least 60%.
  • 9. The method according to claim 4, wherein the amount of COPS3 mRNA is reduced by at least 60%.
  • 10. A nucleic acid molecule of 12 to 30 nucleotides in length comprising a contiguous nucleotide sequence of at least 12 nucleotides which is 90% complementary to a mammalian COPS3 target sequence, wherein the nucleic acid molecule is capable of inhibiting the expression of COPS3 mRNA.
  • 11. The nucleic acid molecule according to claim 10, wherein the contiguous nucleotide sequence is fully complementary to a sequence selected from the group consisting of SEQ ID NOs: 1, 4, 5, 6, 7, 8 and 9, and/or wherein the nucleic acid molecule comprises a contiguous nucleotide sequence of 12 to 25.
  • 12. (canceled)
  • 13. The nucleic acid molecule of claim 10, wherein the nucleic acid molecule is a RNAi molecule.
  • 14. The nucleic acid molecule of claim 10, wherein the nucleic acid molecule is a single stranded antisense oligonucleotide.
  • 15. The nucleic acid molecule according to claim 10, wherein the nucleic acid molecule comprises one or more 2′ sugar modified nucleosides.
  • 16. The nucleic acid molecule according to claim 15, wherein the one or more 2′ sugar modified nucleosides are independently selected from the group consisting of 2′-O-alkyl-RNA, 2′-O-methyl-RNA, 2′-alkoxy-RNA, 2′-O-methoxyethyl-RNA, 2′-amino-DNA, 2′-fluoro-DNA, arabino nucleic acid (ANA), 2′-fluoro-ANA and LNA nucleosides.
  • 17. (canceled)
  • 18. The nucleic acid molecule according to claim 10, wherein the contiguous nucleotide sequence comprises at least one phosphorothioate internucleoside linkage.
  • 19. The nucleic acid molecule according to claim 18, wherein all the internucleoside linkages within the contiguous nucleotide sequence are phosphorothioate internucleoside linkages.
  • 20. The nucleic acid molecule according to claim 10, wherein the nucleic acid molecule is capable of recruiting RNase H.
  • 21. The nucleic acid molecule according to claim 10, wherein the nucleic acid molecule, or contiguous nucleotide sequence thereof, comprises a gapmer of formula 5′-F-G-F′-3′, wherein regions F and F′ independently comprise 1-4 2′ sugar modified nucleosides and G is a region between 6 and 18 nucleosides which are capable of recruiting RNase H.
  • 22. A conjugate compound comprising a nucleic acid molecule according to claim 10 and at least one conjugate moiety covalently attached to said nucleic acid molecule.
  • 23. The conjugate compound of claim 22, wherein the conjugate moiety is or comprises a GalNAc moiety.
  • 24. The conjugate compound of claim 22, wherein the conjugate compound comprises a physiologically labile linker composed of 2 to 5 linked nucleosides comprising at least two consecutive phosphodiester linkages, wherein the physiologically labile linker is covalently bound at the 5′ or 3′ terminal of the nucleic acid molecule.
  • 25. A pharmaceutically acceptable salt of a nucleic acid molecule according to claim 10.
  • 26. A pharmaceutical composition comprising a nucleic acid molecule according to claim 10 and a pharmaceutically acceptable excipient.
  • 27. An in vivo or in vitro method for inhibiting COPS3 expression in a target cell which is expressing COPS3, said method comprising administering a nucleic acid molecule according to claim 10 in an effective amount to said cell.
  • 28. A method for treating or preventing a disease in a subject suffering from or susceptible to the disease, the method comprising administering to the subject a therapeutically or prophylactically effective amount of a nucleic acid molecule according to claim 10.
  • 29. (canceled)
  • 30. (canceled)
  • 31. (canceled)
  • 32. (canceled)
Priority Claims (1)
Number Date Country Kind
19217769.9 Dec 2019 EP regional
CROSS-REFERENCE TO RELATED APPLICATIONS

This application is a continuation of International PCT Application No. PCT/EP2020/086676 filed on Dec. 17, 2020, which claims priority to European Patent Application No. 19217769.9 filed on Dec. 19, 2019, the contents of each application are incorporated herein by reference in their entireties.

Continuations (1)
Number Date Country
Parent PCT/EP2020/086676 Dec 2020 US
Child 17845847 US