COMPLEMENT COMPONENT C1R INHIBITORS FOR TREATING A NEUROLOGICAL DISEASE, AND RELATED COMPOSITIONS, SYSTEMS AND METHODS OF USING SAME

Abstract
The present invention relates to complement C1r subcomponent (C1R) inhibitors for use in treatment of neurological diseases. The invention in particular relates to the use of C1R inhibitors for down-regulation of C1R expression. The invention also relates to nucleic acid molecules, which are complementary to C1R and capable of reducing the level of an C1R mRNA. Also comprised in the present invention is a pharmaceutical composition and its use in the treatment of neurological diseases.
Description
CROSS-REFERENCE TO RELATED APPLICATIONS

This application relates to U.S. Provisional Application filed May 11, 2020, entitled “Complement Component C4 Inhibitors For Treating A Neurological Disease, And Related Compositions, Systems And Methods Of Using Same” and US Provisional Application filed May 11, 2020, entitled “Complement Component C1S Inhibitors For Treating A Neurological Disease, And Related Compositions, Systems And Methods Of Using Same,” the contents of which are both incorporated herein by reference in their entireties. This application claims priority to U.S. Provisional Application No. 63/023,113, filed May 11, 2020, entitled “Complement Component C1R Inhibitors For Treating A Neurological Disease, And Related Compositions, Systems And Methods Of Using Same,” the contents of which are incorporated herein by reference in its entirety.


SEQUENCE LISTING

This application contains a Sequence Listing, which has been submitted electronically in XML format and is hereby incorporated by reference in its entirety. Said XML copy, created on May 3, 2023, is named P36090-US-1_SL.xml and is 891,056 bytes in size.


FIELD OF INVENTION

The present invention relates to complement C1r subcomponent (C1R) inhibitors for use in treatment of neurological diseases. The invention in particular relates to the use of C1R inhibitors for down-regulation of C1R expression. The invention also relates to nucleic acid molecules, which are complementary to C1R and capable of reducing the level of an C1R mRNA. Also comprised in the present invention is a pharmaceutical composition and its use in the treatment of neurological diseases.


BACKGROUND

The complement system is a part of the innate immune system that enhances the clearance of microbes or damaged cells by phagocytes and promotes inflammation. The complement system also participates in synaptic pruning in the brain, with the classical pathway of the complement system mediating synapse removal. This process involves initiation of the classical pathway by the complement component 1 (C1) complex (consisting of C1C), C1S and C1R), leading to cleavage of complement component 2 (C2) and complement component 4 (C4), which in turn lead to cleavage of complement component 3 (C3) followed by engulfment of synapses by microglia cells. Beyond roles in normal brain circuitry refinement during early development, it is well established that aberrant activity of the classical complement pathway can mediate synapse loss and neurodegeneration in various neurological diseases. Observations of elevated complement levels in patient samples and beneficial effects of reducing or eliminating complement components in mouse models have identified a damaging role for complement in conditions including, Alzheimer's disease, frontotemporal dementia, multiple sclerosis, amyotrophic lateral sclerosis, Huntington's disease, Parkinson's disease, virus-induced cognitive impairment, glaucoma, macular degeneration, myasthenia gravis, Guillain-Barré syndrome, neuromyelitis optica, central nervous system lupus erythematosus and schizophrenia.


There remains a need in the art for therapeutic and prognostic agents to address such conditions. The present invention meets these and other needs.


OBJECTIVE OF THE INVENTION

The present invention provides nucleic acid inhibitors of complement component 1R (C1R) which may be used both in vivo and in vitro for down-regulation of C1R expression and for the prophylactic and therapeutic intervention in neurological diseases. The present invention further identifies novel nucleic acid molecules, such as antisense oligonucleotides, which are capable of inhibiting the expression of C1R in vitro and in vivo.


SUMMARY OF INVENTION

The present invention relates to oligonucleotides targeting a nucleic acid and capable of modulating the expression of C1R, useful, for example, to treat or prevent diseases related to the functioning of the C1R.


Accordingly, in a first aspect, the invention provides a C1R inhibitor for use in the treatment and/or prevention of neurological disease, such as tauopathies or schizophrenia, in particular, a C1R inhibitor is capable of reducing the amount of C1R, such as C1R mRNA and/or C1R protein. Such an inhibitor is advantageously a nucleic acid molecule of 12 to 60 nucleotides in length, which is capable of reducing C1R mRNA levels.


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 nucleotide sequence of at least 10 nucleotides, in particular of 16 to 20 nucleotides, which is at least 90% complementary, such as 90-95%, 95-98%, or fully complementary to a mammalian C1R, e.g. a human C1R, a mouse C1ra, C1rb or a cynomolgus monkey C1R. Such a nucleic acid molecule is capable of inhibiting the expression of C1R in a cell expressing C1R. The inhibition of C1R allows for a reduction of the amount of C1R protein 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 siRNAs that inhibit the expression and/or activity of C1R. In particular, modified antisense oligonucleotides or modified siRNAs comprising one or more 2′ sugar modified nucleoside(s) and one or more phosphorothioate linkage(s), which reduce C1R mRNA are advantageous.


In a further aspect, the invention provides pharmaceutical compositions comprising the C1R 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 C1R expression in a target cell, which is expressing C1R, by administering an C1R 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 C1R expression is reduced by at least 50%, e.g., 50-60%; or at least 60%, e.g., 60-70%; or at least 70%, e.g., 70-80%; or at least 80%, e.g., 80-90%; or at least 90%, e.g., 90-95%, in the 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 C1R comprising administering a therapeutically or prophylactically effective amount of the C1R 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.


Definitions
Compound

Herein, the term “compound”, with respect to a compound of the invention, means any molecule capable of inhibition C1R 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 C1R, in particular an antisense oligonucleotide or a siRNA. In some embodiments, the compound is herein also referred to as an “inhibitor” or a “C1R inhibitor”.


Oligonucleotide

The term “oligonucleotide” as used herein is defined as it is generally understood by the skilled person, such as, as a molecule comprising two or more covalently linked nucleosides. An oligonucleotide is also referred to herein as a “nucleic acid” or “nucleic acid molecule”. 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, an siRNA, an 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 often delivered to cells using lentiviral vectors from which they are then transcribed to produce single stranded RNA that will form a stem loop (hairpin) RNA structure capable of interacting with RNA interference machinery (including the RNA-induced silencing complex (RISC)). In an embodiment of the present invention, the shRNA is a chemically produced shRNA molecule (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 an 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 term oligonucleotide of the invention also includes pharmaceutically acceptable salts, esters, solvates and prodrugs thereof.


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 21 nucleotides.


In some embodiments, the oligonucleotide, or a contiguous nucleotide sequence thereof, comprises or consists of 24 or less nucleotides, such as 22, such as 20 or less nucleotides, such as 14, 15, 16, 17, 18, 19, 20 or 21 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 15 to 20 nucleotides, both 15 and 20 nucleotide lengths 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 oligonucleotide(s) can modulate the expression of a target nucleic acid in a mammal or in a mammalian cell. In some embodiments, the nucleic acid molecules, such as for siRNAs, shRNAs and antisense oligonucleotides inhibit expression of a target nucleic acid(s).


In one embodiment of the invention, the oligonucleotide is selected from an RNAi agent, such as an 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.


A library of oligonucleotides is to be understood as a collection of different 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 C1R target nucleic acids, designed for the purpose of identifying potent sequences, e.g., 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 retain a core nucleobase sequence of the parent nucleic acid molecule, e.g., a conserved sequence of the parent.


Antisense Oligonucleotides

The term “antisense oligonucleotide” or “ASO” as used herein is defined as oligonucleotides capable of hybridizing to a target nucleic acid, in particular to a contiguous sequence on a target nucleic acid, e.g., to modulate expression of the corresponding target gene. Generally, nucleic acid molecules of the invention are antisense nucleic acids. The antisense oligonucleotides are not essentially double stranded and need not be 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), e.g., where the degree of intra or inter self-complementarity is less than 50% across of the full length of the oligonucleotide.


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


Advantageously, in some embodiments, the oligonucleotide of the invention comprises one or more modified nucleosides or nucleotides, such as 2′ sugar modified nucleosides. Furthermore, it is advantageous that, some, most, or all of the nucleosides, which are not modified, are DNA nucleosides, e.g., 50%, 75%, 95%, or 100% of 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 that generally interferes with the expression of an mRNA. The term refers to 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 one or both strands are of 17 to 30 nucleotides in length, typically 19 to 25 nucleosides in length, wherein the antisense strand is complementary, such as at least 90%, e.g., 90-95% complementary, or 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/or antisense strand 3′ end may form a 3′ overhang of, e.g. 1, 2, or 3 nucleosides (e.g., 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 to 25 nucleotides in length, such as 21 to 23 nucleotide in length.


Once inside a cell the antisense strand can be 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, some, most, or all (e.g., 75-90%, 80-95%, 90-99%, or 100%) of the nucleotides of an siRNA sense (passenger) strand may be modified with 2′ sugar modified nucleosides such as LNA (see WO2004/083430 and WO2007/085485, for example). In some embodiments, the passenger stand of the siRNA may be discontinuous (see WO2007/107162 for example). In some embodiments, thermally destabilizing nucleotides at a seed region of the antisense strand of siRNAs are useful in reducing off-target activity of the siRNAs (see WO2018/098328 for example). In some embodiments, 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 of one or both strands (e.g., the antisense strand and/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 and/or the sense strand); or the phosphorothioate or methylphosphonate internucleoside linkage may be at both the 5′- and 3′-termini of one or both strands (e.g., the antisense strand and/or the sense strand). In some embodiments, the remaining internucleoside linkages are phosphodiester linkages. In some embodiments, the siRNA molecule comprises one or more phosphorothioate internucleoside linkages. In siRNA molecules, phosphorothioate internucleoside linkages may reduce or inhibit nuclease cleavage in RICS. Accordingly, in some embodiments, not all internucleoside linkages in the antisense strand are modified, e.g., in some embodiments, 10-90%, 20-80%, 30-70%, or 40-60% of 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. In a particular example, the nucleic acid molecule is conjugated to a moiety that targets a brain cell or other cell of the CNS. Thus, the nucleic acid molecule may conjugated to a moiety that facilitates delivery across the blood brain barrier. For example, the nucleic acid molecule may be conjugated to an antibody or antibody fragment targeting the transferrin receptor.


Other aspects of the invention relate to pharmaceutical compositions, in particular, pharmaceutical compositions comprising dsRNA, such as siRNA molecules suitable for therapeutic use, and methods of inhibiting the expression of a 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 can interact with the endonuclease known as Dicer (believed to processes dsRNA into 19-23 base pair short interfering RNAs with characteristic two base 3′ overhangs which then can be 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, an shRNA molecule comprises one or more phosphorothioate internucleoside linkages. In RNAi molecules, phosphorothioate internucleoside linkages may reduce or inhibit nuclease cleavage in RICS. Accordingly, not all internucleoside linkages in the stem loop of the shRNA molecule are modified, e.g., in some embodiments, 10-90%, 20-80%, 30-70%, or 40-60% of internucleoside linkages in the antisense strand 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, e.g., 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 95%, 98%, 99%, or 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, in some embodiments, 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 unit” or “modified monomer”. 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, such as, as being a linkage 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 phosphorodithioate internucleoside linkages.


With the oligonucleotide of the invention, it can be advantageous to use phosphorothioate internucleoside linkages, e.g., for 10-90%, 20-80%, 30-70%, or 40-60% of 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%, e.g., 60-80%; such as at least 70%, e.g., 70-85%; such as at least 75%, e.g., 75-90%; such as at least 80%, e.g. 80-95%; or such as at least 90%, e.g., 90-99%, 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.


In some embodiments, the antisense oligonucleotides may comprise other internucleoside linkages (other than phosphodiester and phosphorothioate), for example alkyl phosphonate/methyl phosphonate internucleoside linkages, which may be tolerated in an otherwise DNA phosphorothioate gap region (e.g., as in EP 2 742 135).


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 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 nucleobase 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-aminopurine.


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 and/or modified nucleobases. 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×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 referring to 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°=−RTln(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 a nucleic acid target by hybridization, oligonucleotides of the present invention hybridize to a target nucleic acid with estimated ΔG° values below −10 kcal/mol for oligonucleotides that are 10 to 30 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/mol, such as below −15 kcal/mol, such as below −20 kcal/mol and such as below −25 kcal/mol for oligonucleotides that are 8 to 30 nucleotides in length. In some embodiments, the oligonucleotides hybridize to a target nucleic acid with an estimated ΔG° value in the range of −10 to −60 kcal/mol, such as −12 to −40, such as from −15 to −30 kcal/mol or −16 to −27 kcal/mol such as −18 to −25 kcal/mol.


Target Nucleic Acid

According to the present invention, the target nucleic acid is a nucleic acid, which encodes a mammalian C1R 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 C1R target nucleic acid.


The therapeutic oligonucleotides of the invention may for example target exon regions of a mammalian C1R (in particular siRNA and shRNA, but also antisense oligonucleotides), or may for example target any intron region in the C1R pre-mRNA (in particular antisense oligonucleotides).


Table 1a lists predicted exon and intron regions of SEQ ID NO: 3, i.e. of the human C1R pre-mRNA sequence.









TABLE 1a







Exons and introns in the human C1R pre-mRNA.








Exemplary exonic regions in
Exemplary intronic regions in


the human C1R premRNA
the human C1R premRNA


(SEQ ID NO 3)
(SEQ ID NO 3)












ID
start
end
ID
start
end















E1
163
221
11
222
927


E2
928
1156
12
1157
2359


E3
2360
2552
13
2553
2874


E4
2875
3021
14
3022
3118


E5
3119
3315
5
3316
3621


E6
3622
3769
16
3770
3876


E7
3877
3998
17
3999
6150


E8
6151
6229
18
6230
6591


E9
6592
6747
19
6748
10501


E10
10502
10576
110
10577
11306


E11
11307
12409









In some embodiments, the target nucleic acid encodes a C1R protein, in particular a mammalian C1R protein, such as a human C1R protein. See for example Table 2 and Table 3, which provides an overview on the genomic sequences of human, cyno monkey and mouse C1R (Table 2) and on pre-mRNA sequences for human, monkey and mouse C1R and for the mature mRNAs for human C1R (Table 3).


In some embodiments, the target nucleic acid is selected from the group consisting of SEQ ID NO: 1, 2, 3, 4, 5, and 6, or naturally occurring variants thereof (e.g. sequences encoding a mammalian C1R).









TABLE 2







Genome and assembly information for C1R across species.









NCBI reference



sequence*











Genomic coordinates

accession number













Species
Chr
Strand
Start
End
Assembly
for mRNA
















Mouse
6
Fwd
124512405
124523443
GRCm38.p6
NM_023143.3


Mouse
6
Fwd
124570294
124581171
GRCm38.p6
NM_001113356.1


Human
12
Rev
7080214
7092540
GRCh38.p12
NM_001733.7 and








NM_001354346.2


Cyno
11
Rev
7511281
7524092
Macaca_fascicularis_5



Cyno
11
Rev
7412536
7495192
Macaca_fascicularis_5






Fwd = forward strand. Rev = 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 C1R target nucleic acid in a cell, which is expressing the C1R target nucleic acid. The contiguous sequence of nucleobases of the nucleic acid molecule of the invention is typically complementary to a conserved region of the C1R target nucleic acid, as measured across the length of the nucleic acid molecule, optionally with the exception of one or two mismatches. In some embodiments, the target nucleic acid is a messenger RNA, such as a pre-mRNA which encodes mammalian C1R protein, such as mouse C1ra, e.g. the mouse C1ra pre-mRNA sequence, such as that disclosed as SEQ ID NO: 1, the human C1R pre-mRNA sequence, such as that disclosed as SEQ ID NO: 3, or the cyno monkey C1R pre-mRNA sequence, such as that disclosed as SEQ ID NO: 4, or a mature C1R mRNA, such as that of a human mature mRNA disclosed as SEQ ID NO: 6. In some embodiments, the target nucleic acid is a messenger RNA, such as a pre-mRNA which encodes mammalian C1R protein, such as mouse C1rb, e.g. the mouse C1rb pre-mRNA sequence, such as that disclosed as SEQ ID NO: 2, the human C1R pre-mRNA sequence, such as that disclosed as SEQ ID NO: 3, or the cyno monkey C1R pre-mRNA sequence, such as that disclosed as SEQ ID NO: 5, or a mature C1R mRNA, such as that of a human mature mRNA disclosed as SEQ ID NO: 6. SEQ ID NOs: 1, 2, 3, 4, 5 and 6 are DNA sequences—it will be understood that target RNA sequences have uracil (U) bases in place of the thymidine bases (T).


It is known that different, i.e. shorter, annotated mRNA isoforms of the above sequences exist. The isoforms are well-known in the art and can be derived from the known sequence databases.


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







C1ra Mus musculus pre-mRNA
SEQ ID NO: 1



C1rb Mus musculus pre-mRNA
SEQ ID NO: 2



C1R Homo sapiens pre-mRNA
SEQ ID NO: 3



C1R Macaca fascicularis pre-mRNA
SEQ ID NO: 4



C1R Macaca fascicularis pre-mRNA
SEQ ID NO: 5



C1R Homo sapiens mature mRNA
SEQ ID NO: 6







Note:



SEQ ID NO: 3, 5 and 6 comprise 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, compounds of the present invention do not bind to the regions comprising multiple NNNNs.






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.


Target

The term “target” as used herein refers to the complement C1r subcomponent (C1R), which can in the context of this disclosure be C1R. C1R is also referred to as complement C1r. Further, the term “target” can refer to the C1R target nucleic acid, as well as the C1R protein.


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 comprises or 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. It is well known in the art that C1R genes display high level of variability between individuals. The term “target sequence” encompasses all publicly annotated variants of C1R.


In some embodiments, the target sequence is a sequence selected from the group consisting of a human C1R mRNA exon, such as a human C1R mRNA exon selected from the group consisting of Ea1-Ea11 (see for example Table 1a above).


Accordingly, the invention provides for an oligonucleotide, wherein said oligonucleotide comprises a contiguous sequence, which is at least 90% complementary, such as 90-95% or fully complementary, to an exon region of SEQ ID NO: 3, selected from the group consisting of Ea1-Ea11 (see Table 1a).


In some embodiments, the target sequence is a sequence selected from the group consisting of a human C1R mRNA exon, such as a human C1R mRNA intron selected from the group consisting of la1-la10 (see for example Table 1a above).


Accordingly, the invention provides an oligonucleotide, wherein said oligonucleotide comprises a contiguous sequence, which is at least 90% complementary, such as 90-95% or fully complementary, to an intron region of SEQ ID NO: 3, selected from the group consisting of la1-la10 (see Table 1a).


In some embodiments, the target sequence is SEQ ID NO: 6. In some embodiments, the contiguous nucleotide sequence as referred to herein is at least 90% (e.g., 90-95%) complementary, such as at least 95% (e.g., 95-98) complementary to the target sequence of SEQ ID NO: 6. In some embodiments, the contiguous nucleotide sequence is fully complementary to a target sequence of SEQ ID NO: 6.


The oligonucleotide 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 oligonucleotide 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 21 contiguous nucleotides.


In some embodiments, the oligonucleotide of the present invention targets a region shown in Table 4a.









TABLE 4a







Exemplary target regions on SEQ ID NO: 3












start
end



Target region
SEQ ID NO: 3
SEQ ID NO: 3















 1A
775
796



 2A
810
824



 3A
845
859



 4A
862
906



 5A
918
945



 6A
997
1017



 7A
1019
1046



 8A
1048
1063



 9A
1065
1080



 10A
1089
1110



 11A
1112
1139



 12A
1155
1175



 13A
1175
1193



 14A
1198
1269



 15A
1310
1324



 16A
1332
1362



 17A
1362
1391



 18A
1421
1440



 19A
1457
1483



 20A
1507
1543



 21A
1567
1582



 22A
1585
1606



 23A
1608
1626



 24A
1634
1662



 25A
1664
1692



 26A
1694
1716



 27A
1820
1835



 28A
1837
1857



 29A
1885
1907



 30A
1933
1947



 31A
2021
2056



 32A
2072
2086



 33A
2088
2123



 34A
2125
2140



 35A
2142
2177



 36A
2194
2214



 37A
2223
2242



 38A
2282
2309



 39A
2343
2372



 40A
2380
2427



 41A
2429
2445



 42A
2450
2472



 43A
2483
2594



 44A
2604
2629



 45A
2641
2670



 46A
2672
2706



 47A
2760
2788



 48A
2790
2812



 49A
2856
2895



 50A
2897
2912



 51A
2919
2945



 52A
2947
2970



 53A
2958
2999



 54A
3010
3043



 55A
3111
3152



 56A
3154
3179



 57A
3204
3224



 58A
3226
3241



 59A
3271
3351



 60A
3353
3367



 61A
3390
3419



 62A
3422
3445



 63A
3489
3539



 64A
3530
3561



 65A
3563
3582



 66A
3620
3674



 67A
3682
3748



 68A
3756
3782



 69A
3785
3809



 70A
3837
3854



 71A
3888
3916



 72A
3919
3946



 73A
3952
3978



 74A
3980
4037



 75A
4056
4079



 76A
4105
4140



 77A
4142
4172



 78A
4174
4198



 79A
4210
4237



 80A
4222
4237



 81A
4354
4374



 82A
4391
4416



 83A
4500
4517



 84A
4542
4559



 85A
4614
4632



 86A
4634
4666



 87A
4672
4695



 88A
4736
4750



 89A
4779
4799



 90A
4808
4884



 91A
4886
4917



 92A
4919
4952



 93A
4971
4991



 94A
5021
5041



 95A
5043
5068



 96A
5083
5106



 97A
5108
5139



 98A
5194
5212



 99A
5238
5270



100A
5351
5365



101A
5382
5405



102A
5529
5547



103A
5555
5611



104A
5613
5656



105A
5682
5724



106A
5738
5754



107A
5773
5808



108A
5814
5839



109A
5841
5863



110A
5883
5908



111A
5926
5940



112A
5955
5969



113A
5972
6012



114A
5991
6012



115A
6008
6029



116A
6025
6046



117A
6061
6084



118A
6106
6128



119A
6136
6163



120A
6169
6200



121A
6202
6227



122A
6229
6253



123A
6277
6302



124A
6304
6331



125A
6346
6361



126A
6370
6401



127A
6402
6426



128A
6438
6455



129A
6510
6529



130A
6539
6554



131A
6556
6616



132A
6638
6655



133A
6676
6694



134A
6696
6721



135A
6723
6746



136A
6775
6875



137A
6901
6919



138A
6925
6952



139A
6954
6995



140A
7024
7040



141A
7029
7058



142A
7031
7045



143A
7060
7081



144A
7083
7100



145A
7093
7107



146A
7108
7122



147A
7130
7166



148A
7136
7166



149A
7136
7157



150A
7138
7157



151A
7138
7155



152A
7146
7160



153A
7147
7162



154A
7164
7178



155A
7166
7183



156A
7168
7187



157A
7168
7186



158A
7170
7196



159A
7170
7188



160A
7171
7198



161A
7173
7188



162A
7173
7198



163A
7174
7188



164A
7186
7201



165A
7189
7205



166A
7194
7212



167A
7217
7231



168A
7219
7234



169A
7231
7253



170A
7231
7246



171A
7231
7245



172A
7242
7258



173A
7243
7257



174A
7246
7267



175A
7246
7262



176A
7248
7267



177A
7255
7269



178A
7269
7283



179A
7271
7319



180A
7271
7285



181A
7283
7319



182A
7292
7306



183A
7294
7312



184A
7295
7318



185A
7296
7312



186A
7297
7311



187A
7302
7318



188A
7317
7332



189A
7318
7332



190A
7332
7364



191A
7338
7352



192A
7338
7364



193A
7341
7364



194A
7348
7364



195A
7352
7366



196A
7354
7373



197A
7354
7372



198A
7361
7391



199A
7361
7382



200A
7363
7382



201A
7363
7380



202A
7371
7385



203A
7372
7387



204A
7401
7416



205A
7401
7426



206A
7402
7416



207A
7414
7429



208A
7417
7433



209A
7438
7457



210A
7438
7455



211A
7443
7463



212A
7445
7459



213A
7445
7463



214A
7446
7463



215A
7456
7478



216A
7456
7471



217A
7456
7470



218A
7467
7483



219A
7468
7482



220A
7471
7487



221A
7507
7543



222A
7516
7530



223A
7518
7536



224A
7519
7542



225A
7520
7536



226A
7521
7535



227A
7526
7542



228A
7541
7556



229A
7542
7556



230A
7565
7588



231A
7572
7588



232A
7576
7590



233A
7578
7597



234A
7578
7596



235A
7585
7606



236A
7587
7606



237A
7587
7604



238A
7594
7608



239A
7596
7611



240A
7596
7612



241A
7604
7624



242A
7606
7620



243A
7606
7624



244A
7607
7624



245A
7617
7632



246A
7617
7631



247A
7634
7653



248A
7641
7655



249A
7719
7733



250A
7734
7753



251A
7735
7753



252A
7741
7756



253A
7751
7765



254A
7773
7799



255A
7773
7791



256A
7776
7791



257A
7777
7791



258A
7823
7849



259A
7837
7855



260A
7839
7855



261A
7840
7854



262A
7843
7864



263A
7843
7859



264A
7843
7870



265A
7845
7864



266A
7849
7870



267A
7849
7867



268A
7855
7870



269A
7864
7879



270A
7909
7932



271A
7910
7926



272A
7911
7925



273A
7916
7932



274A
7929
7950



275A
7929
7947



276A
7935
7950



277A
7943
7957



278A
8962
8976



279A
9000
9014



280A
9002
9016



281A
9021
9037



282A
9034
9052



283A
9036
9054



284A
9039
9054



285A
9040
9054



286A
9060
9074



287A
9060
9086



288A
9063
9086



289A
9070
9086



290A
9074
9088



291A
9076
9094



292A
9096
9128



293A
9121
9138



294A
9213
9231



295A
9216
9231



296A
9217
9231



297A
9236
9253



298A
9253
9272



299A
9253
9270



300A
9258
9278



301A
9260
9274



302A
9260
9278



303A
9261
9278



304A
9276
9314



305A
9283
9304



306A
9283
9299



307A
9283
9310



308A
9285
9304



309A
9289
9310



310A
9289
9307



311A
9295
9310



312A
9298
9313



313A
9298
9312



314A
9312
9328



315A
9340
9355



316A
9351
9371



317A
9352
9370



318A
9367
9385



319A
9381
9398



320A
9416
9470



321A
9480
9509



322A
9522
9538



323A
9540
9586



324A
9586
9636



325A
9639
9712



326A
9734
9787



327A
9820
9840



328A
9842
9856



329A
9858
9873



330A
9880
9903



331A
9905
9936



332A
9938
9958



333A
9985
10022



334A
10132
10150



335A
10133
10150



336A
10216
10231



337A
10272
10297



338A
10299
10343



339A
10345
10390



340A
10427
10450



341A
10452
10468



342A
10470
10499



343A
10501
10542



344A
10554
10596



345A
10643
10660



346A
10708
10734



347A
10739
10765



348A
10793
10861



349A
10823
10860



350A
10888
10917



351A
10944
10970



352A
10972
10989



353A
11004
11020



354A
11078
11100



355A
11140
11165



356A
11189
11204



357A
11210
11227



358A
11272
11293



359A
11295
11320



360A
11322
11414



361A
11427
11454



362A
11456
11487



363A
11507
11526



364A
11537
11553



365A
11560
11581



366A
11583
11611



367A
11613
11637



368A
11639
11703



369A
11714
11733



370A
11747
11807



371A
11824
11880



372A
11882
11901



373A
11903
11918



374A
11920
11935










Target Cell

The term “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 a brain cell. In some embodiments, the brain cell is selected from the group consisting of a neuron and a microglia cell. 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 some embodiments, the target cell expresses C1R mRNA, such as the C1R pre-mRNA or C1R mature mRNA. The poly A tail of the C1R mRNA is typically disregarded for antisense oligonucleotide targeting.


Naturally Occurring Variant

The term “naturally occurring variant” refers to variants of the C1R 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 sufficiently complementary sequence of 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% (e.g., 95-98%), such as at least 98% (e.g., 99-99%), or at least 99% (e.g., 99-100%) homology to a mammalian C1R target nucleic acid, such as a target nucleic acid of SEQ ID NO: 3 and/or SEQ ID NO: 4. In some embodiments, the naturally occurring variants have at least 99% (e.g., 99-100%) homology to the human C1R target nucleic acid of SEQ ID NO: 3. In some embodiments, the naturally occurring variants have at least 95% (e.g., 95-98%), such as at least 98% (e.g., 98-99%), or at least 99% (e.g., 99-100%) homology to a mammalian C1R target nucleic acid, such as a target nucleic acid of SEQ ID NO: 3 and/or SEQ ID NO: 5. 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 a C1R inhibitor's ability to inhibit an amount or the activity of C1R in a target cell. Inhibition of expression or activity may be determined by measuring the level of C1R pre-mRNA or C1R mRNA, or by measuring the level of C1R protein or activity in a cell. Inhibition of expression may be determined in vitro or in vivo. 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.


The term “inhibitor,” “inhibition” or “inhibit” may also be referred to as down-regulate, reduce, suppress, lessen, lower, or decrease the amount, expression, and/or activity of C1R.


The inhibition of expression of C1R 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 C1R mRNA or polypeptide and inhibit the activity of C1R or prevent its binding to other molecules.


In some embodiments, the inhibition of expression of the C1R target nucleic acid results in a decreased amount of C1R protein in the target cell. Preferably, the amount of C1R protein is decreased as compared to a control. In some embodiments, the decrease in amount of C1R protein is at least 20%, at least 30%, as compared to a control. In some embodiments, the amount of C1R protein in the target cell is reduced by at least 50%, e.g., 50-60%, or at least 60%, e.g., 60-70%, or at least 70%, e.g., 70-80%, at least 80%, e.g., 80-90%, or at least 90%, e.g., 90-95%, 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 one or more 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 results in an increase in melting temperature in the range of +0.5 to +12C, 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, a 2′ substituted sugar modified nucleoside does not include 2′ bridged nucleosides like LNA.


Locked Nucleic Acid Nucleosides (LNA Nucleoside)

A “LNA nucleoside” is a 2′-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 acids or bicyclic nucleic acids (BNAs) 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, and 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).




text missing or illegible when filed


Particular LNA nucleosides for use in molecules of the invention are beta-D-oxy-LNA, 6′-methyl-beta-D-oxy LNA such as (S)-6′-methyl-beta-D-oxy-LNA (ScET) and ENA. A particularly advantageous LNA is beta-D-oxy-LNA.


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, for example, provides in vitro methods for determining RNase H activity, which may be used to determine 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%-15% or more than 20%, e.g., 20-25%, or 20-30%, 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 RNase 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. 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) and/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 15 to 21 nucleosides.


By way of example, the gapmer oligonucleotide of the present invention can be represented by the following formulae:





F1-8-G5-16-F′1-8, such as





F1-8-G7-16-F′2-8


with the proviso that the overall length of the gapmer regions F-G-F′ is at least 12 (e.g., 12-15 nucleotides), such as at least 14 nucleotides (e.g., 14-20 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 define the 5′ and 3′ ends of the F and F′ region, respectively, and G is a region between 6 and 16 nucleosides which are capable of recruiting RNaseH.


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 define the 5′ and 3′ end of the F and F′ region, respectively, 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]1-5-[region G]six-18-[LNA]1-5, wherein region G is as defined in the Gapmer region G definition.


MOE Gapmers

An “MOE gapmers” is a gapmer wherein regions F and F′ consist of MOE (methoxyethy) 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]5 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 may further comprise 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 exonucleoase 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. In some embodiments, 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, for example, 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, for example, 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 one or more of the following formulae:





F-G-F′; in particular F1-8-G5-18-F2-8





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.


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. Prophylaxis also includes delaying or reducing the likelihood of disease occurrence, delaying or reducing frequency of relapse of the disease, and/or reducing severity or duration of the disease if the subject eventually succumbs to the disease. It will therefore be recognized that treatment as referred to herein may, in some embodiments, be prophylactic. In some embodiments, treatment is performed on a patient who has been diagnosed with a complement mediated neurological disease, such as a neurological disease selected from the group consisting of Alzheimer's disease, frontotemporal dementia, multiple sclerosis, amyotrophic lateral sclerosis, Huntington's disease, Parkinson's disease, virus-induced cognitive impairment, glaucoma, macular degeneration, myasthenia gravis, Guillain-Barre syndrome, neuromyelitis optica, central nervous system lupus erythematosus, and schizophrenia. In some embodiments, the compounds of the invention are for use in the treatment of a tauopathy, such as Alzheimer's disease. In some embodiments, the compounds of the invention are for use in the treatment of schizophrenia.


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 suffer from or be susceptible to a neurological disease or neurodegenerative disorder. A patient “susceptible to” a disease or disorder is one who is pre-disposed thereto and/or otherwise at risk of developing or having a recurrence of the disease or disorder. A susceptible patient can be understood a patent likely to develop the disease or disorder, to the extent that the patient would benefit from prophylactic treatment or intervention.


By “neurological disease” is meant a disease or disorder of the nervous system including, but not limited to, neurological conditions associated with cancer, and neurodegenerative disease.


By “neurodegenerative disease” is meant diseases including, but not limited to Alzheimer's disease, frontotemporal dementia, multiple sclerosis, amyotrophic lateral sclerosis, Huntington's disease, Parkinson's disease, virus-induced cognitive impairment, glaucoma, macular degeneration, myasthenia gravis, Guillain-Barre syndrome, neuromyelitis optica, central nervous system lupus erythematosus, and schizophrenia. In some embodiments, the patient to be treated suffers from a tauopathy, such as Alzheimer's disease. In some embodiments, the patient to be treated suffers from schizophrenia.


Alzheimer's disease (AD), also referred to as Alzheimer disease or “Alzheimer's,” is a chronic neurodegenerative disorder typically characterized by progressive cognitive deterioration, as well as increasing memory loss, problems with language, judgment, and/or problem solving, and that can lead to inability to perform daily tasks, and eventually dementia.







DETAILED DESCRIPTION OF THE INVENTION

Synapse removal and neuronal damage can be mediated by the classical pathway of the complement system, which is initiated by activation of the C1, leading to cleavage of C2 and C4, which in turn lead to cleavage of C3 which can trigger phagocytosis as well as inflammation and further downstream complement activation, Complement C1r subcomponent (C1R) is a protein involved in the complement system.


C1R, C1Q and C1S form the C1 complex, which is the first component of the serum complement system. C1R is serine protease that activates CIS to its active form by proteolytic cleavage. After proteolytic cleavage, C1S activates C2 and C4, which leads to the cleavage of C3.


In the context of the present invention, the present inventors have shown that nucleic acid molecules, such as antisense oligonucleotides, inhibit the expression of C1R. Reduced expression of C1R can lead to reduced cleavage of C1S, C2, C4 and C3, and thereby to reduced engulfment of synapses by microglia cells and other harmful effects of complement activation.


One aspect of the present invention is a C1R inhibitor for use in the treatment and/or prevention of a neurological disease, in particular a neurological disease selected from a tauopathy and schizophrenia. In some embodiments, the tauopathy is Alzheimer's disease. The C1R inhibitor can for example be a small molecule that specifically binds to the C1R protein, wherein said inhibitor prevents or reduces cleavage of the C1S protein.


An embodiment of the invention is a C1R inhibitor, which is capable of preventing or reducing expression of C1R protein thereby leading to reduced cleavage of C1S. In some embodiments, the C1R inhibitor leads to inhibition of engulfment of synapses by microglia cells.


C1R Inhibitors for Use in Treatment of Neurological Diseases

Without being bound by theory, it is believed that C1R is involved in the in the cleavage of C1S, which may lead to the cleavage of C3 (via cleavage of C2 and C4). Therefore, it is believed that C1R is involved in the engulfment of synapses by microglia cells.


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 C1R protein. In some embodiments, the C1R protein is encoded by a sequence selected from SEQ ID NO: 3, 4, 5, and 6. In some embodiments, the C1R protein is encoded by a sequence selected from SEQ ID NO: 6.


Nucleic Acid Molecules of the Invention

Therapeutic nucleic acid molecules are potentially excellent C1R inhibitors since they can target C1R transcripts and promote their degradation, e.g., either via the RNA interference pathway or via RNase H cleavage. Alternatively, oligonucleotides such as aptamers can also act as inhibitors of C1R proteins.


One aspect of the present invention is a C1R targeting nucleic acid molecule for use in treatment and/or prevention of Neurological diseases. Such a nucleic acid molecule can be selected from the group consisting of a single stranded antisense oligonucleotide, an siRNA, and a shRNA.


The present section describes novel nucleic acid molecules suitable for use in treatment and/or prevention of a neurological disease. In some embodiments, the neurological disease is selected from the group consisting of Alzheimer's disease, frontotemporal dementia, multiple sclerosis, amyotrophic lateral sclerosis, Huntington's disease, Parkinson's disease, virus-induced cognitive impairment, glaucoma, macular degeneration, myasthenia gravis, Guillain-Barre syndrome, neuromyelitis optica, central nervous system lupus erythernatosus, and schizophrenia. In some embodiments, the neurological disease is a tauopathy, such as Alzheimer's disease. In some embodiments, the neurological disease is schizophrenia.


The nucleic acid molecules of the present invention are capable of inhibiting C1R mRNA and/or expression of C1R protein in vitro and in vivo. The inhibition may be achieved by hybridizing an oligonucleotide to a target nucleic acid encoding a C1R protein. In some embodiments, the target nucleic acid may be a mammalian C1R sequence. In some embodiments, the target nucleic acid may be a human C1R pre-mRNA sequence such as the sequence of SEQ ID NO: 3 or a human mature C1R mRNA sequence such as the sequence of SEQ ID NO:6. In some embodiments, the target nucleic acid may be a mammalian C1R sequence. In some embodiments, the target nucleic acid may be a cynomolgus monkey C1R sequence such as the sequence of SEQ ID NO: 4 or 5.


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% (e.g., 20-30%) compared to the normal expression level of the target, more preferably at least 30% (e.g., 30-40%), at least 40% (e.g., 40-50%), or at least 50% (e.g., 50-60%), 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 C1R mRNA by at least (e.g., 50-60%) or 60% (e.g., 50-60%) in vitro by using 20-50 nM nucleic acid molecule for transfection. In some embodiments, the nucleic acid molecule of the invention may be capable of inhibiting expression levels of C1R mRNA by at least 50% (e.g., 50-60%) or 60% (e.g., 50-60%) in vitro by using 50-350 nM nucleic acid molecule for gymnosis. Suitably, the examples provide assays, which may be used to measure C1R mRNA inhibition (e.g. Example 1 and the “Materials and Methods” section). C1R 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 C1R 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 molecule 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 C1R target nucleic acid, in particular a human C1R mRNA. These nucleic acid molecules are capable of inhibiting the expression of C1R mRNA and/or C1R protein.


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, or such as 15 to 21 nucleotides in length, which is at least 90% complementary, such as fully complementary, to a mammalian C1R 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 14 to 22, such as 15 to 21 nucleotides in length with at least 90% complementary, such as fully complementary, to the target sequence of SEQ ID NO: 3.


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


In some embodiments, the oligonucleotide sequence is 100% complementary to a region of the target sequence of SEQ ID NO: 3, and/or SEQ ID NO: 6.


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: 3.


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: 4 and 5 and/or SEQ ID NO: 6.


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 2, and/or SEQ ID NO: 3, and/or SEQ ID NO: 4 and 5.


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: 3, selected from the group consisting of target regions 1A to 374A as shown in Table 4a.


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 15 to 21 contiguous nucleotides in length. In a preferred embodiment, the nucleic acid molecule comprises or consists of 15, 16, 17, 18, 19, 20 or 21 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 21 contiguous nucleotides in length.


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


In some embodiments, the contiguous nucleotide sequence of the siRNA or shRNA, which is complementary to the target sequence, 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 21, such as from 15 to 21, such as from 15, 16, 17, 18, 19, 20, or 21 contiguous nucleotides in length.


In some embodiments, the oligonucleotide or contiguous nucleotide sequence comprises or consists of a sequence selected from the group consisting of sequences listed in Table 8 (Materials and Methods section).


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).


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 linkage.


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 70-80%, at least 90%, such as 90-95%, or 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 some embodiments, 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”.


In some embodiments, the inhibitor of the present invention is a nucleic acid capable of inducing the process of RNA interference (as described, e.g., in WO 2014/089121).


Method of Manufacture

In a further aspect, the invention provides methods for manufacturing the oligonucleotide of the invention. In some embodiments, the method comprises reacting nucleotide units and thereby forming covalently linked contiguous nucleotide units comprised in the oligonucleotide in a sequence according to a nucleic acid molecule of the present invention. Preferably, the method uses phophoramidite chemistry (see for example Caruthers et al, 1987, Methods in Enzymology vol. 154, pages 287-313).


The manufactured oligonucleotides may comprise one or more modifications as described herein. For example, the manufactured oligonucleotides may comprise one or more sugar-modified nucleosides, one or more modified internucleoside linkages and/or one or more modified nucleobases. Accordingly, the method for manufacturing the oligonucleotide of the invention may further comprise the introduction of such modifications into the oligonucleotide.


In some embodiments, one or more modified internucleoside linkages, such as phosphorothioate internucleoside linkages, may be introduced into the oligonucleotide. In some embodiments, one or more sugar-modified nucleosides, such as 2′ sugar modified nucleosides, may be introduced. In some embodiments, one or more high affinity modified nucleosides and/or one or more LNA nucleosides may be introduced into the oligonucleotide. In some embodiments, region D′ and/or D″ as described elsewhere herein are added to the oligonucleotide.


In a further aspect, a method is provided for manufacturing the pharmaceutical composition of the invention, comprising mixing the oligonucleotides of the invention with a pharmaceutically acceptable diluent, solvent, carrier, salt and/or adjuvant.


As described elsewhere herein in more detail, the oligonucleotide of the invention may exist in the form of its pharmaceutically acceptable salts, esters, solvates or in the form of prodrugs. Accordingly, methods are provided for manufacturing the oligonucleotide of the invention in such forms.


Pharmaceutically Salts

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, such as a pharmaceutically acceptable sodium salt, ammonium salt or potassium salt. By way of example, the following salts may be mentioned: Alkaline metal salts such as sodium salts, potassium salts or lithium salts; alkaline earth metal salts such as calcium salts or magnesium salts; metal salts such as aluminum salts, iron salts, zinc salts, copper salts; amine salts including inorganic salts such as ammonium salts and organic salts such as t-octylamine salts, dibenzylamine salts, morpholine salts, glucosamine salts, phenylglycine alkyl ester salts, ethylenediamine salts, N-methylglucamine salts, guanidine salts, diethylamine salts, triethylamine salts, dicyclohexylamine salts, N,N′-dibenzylethylenediamine salts, chloroprocaine salts, procaine salts, diethanolamine salts, N-benzyl-phenethylamine salts, piperazine salts, tetramethylammonium salts or tris(hydroxymethyl)aminomethane salts; inorganic acid salts including hydrohalogenic acid salts such as hydrofluorides, hydrochlorides, hydrobromides or hydroiodides, sulfates or phosphates; organic acid salts including lower alkane sulfonic acid salts such as methanesulfonates, trifluoromethanesulfonates or ethanesulfonates, arylsulfonic acid salts such as benzenesulfonates or p-toluenesulfonates, acetates, malates, fumarates, succinates, citrates, tartrates, oxalates or maleates; and amino acid salts such as glycine salts, lysine salts, arginine salts, ornithine salts, glutamic acid salts or aspartic acid salts. These salts may be prepared by known methods.


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


Solvates

The compounds according to the present invention may exist in the form of solvates. The term ‘solvate’ is used herein to describe a molecular complex comprising the oligonucleotide of the invention and one or more pharmaceutically acceptable solvent molecules, for example, ethanol or water. If the solvent is water, the solvate is a ‘hydrate’. Pharmaceutically acceptable solvates within the meaning of the present invention include hydrates and other solvates.


Prodrugs

Further, the compounds according to the present invention may be administered in the form of a prodrug. A prodrug is defined as a compound that undergoes transformations in vivo to yield the parent active drug. Because cell membranes are lipophilic in nature, cellular uptake of oligonucleotides is often reduced compared to neutral or lipophilic equivalents. One solution is to use a prodrug approach (see e.g. Crooke, R. M. (1998) in Crooke, S. T. Antisense research and Application. Springer-Verlag, Berlin, Germany, vol. 131, pp. 103-140). Examples of such prodrug include, but are not limited to, amides, esters, carbamates, carbonates, ureides and phosphates. These prodrugs may be prepared by known methods.


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 or salts thereof and a pharmaceutically acceptable diluent, carrier, salt and/or adjuvant. A pharmaceutically acceptable diluent includes, but is not limited to, phosphate-buffered saline (PBS). 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, e.g., 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, pro-drug formulations, and the like, are also provided, e.g., in WO2007/031091. In some embodiments, the nucleic acid molecule of the invention or pharmaceutically acceptable salt thereof is in a solid form, such as a powder, such as a lyophilized powder. Compounds or nucleic acid molecules 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.


Administration

The oligonucleotides or pharmaceutical compositions of the present invention may be administered via parenteral (such as, intravenous, subcutaneous, intra-muscular, intranasal, intracerebral, intracerebroventricular intraocular, or intrathecal administration).


In some embodiments, the administration is via intrathecal administration, e.g., by lumbar puncture.


Advantageously, e.g. for treatment of neurological disorders, the oligonucleotide or pharmaceutical compositions of the present invention are administered intrathecally or intracranially, e.g. via intracerebral or intraventricular administration.


The invention also provides for the use of the oligonucleotide or conjugate thereof, such as pharmaceutical salts or compositions of the invention, for the manufacture of a medicament wherein the medicament is in a dosage form for subcutaneous administration.


The invention also provides for the use of the oligonucleotide of the invention, or conjugate thereof, such as pharmaceutical salts or compositions of the invention, for the manufacture of a medicament wherein the medicament is in a dosage form for intrathecal administration.


In some embodiments, a therapeutically or prophylactically effective amount of the oligonucleotide or pharmaceutical composition of the present invention is administered.


Delivery Platforms

Delivery of the oligonucleotides to the target tissue may be enhanced by carrier-mediated delivery including, but not limited to, cationic liposomes, cyclodextrins, porphyrin derivatives, branched chain dendrimers, polyethylenimine polymers, nanoparticles, cell-penetrating peptides, and microspheres (see e.g. Dass, C R. J Pharm Pharmacol 2002; 54(1):3-27).


In some embodiments, the inhibitors of the present invention, such as the oligonucleotides of the present invention, are targeted to the brain. For example, delivery to the brain might be achieved by conjugating said inhibitor to a moiety that facilitates delivery across the blood brain barrier, such as an antibody or antibody fragment targeting the transferrin receptor.


Combination Therapies

In some embodiments, the inhibitor of the present invention such as the nucleic acid molecule, nucleic acid molecule conjugate, pharmaceutically acceptable salt, 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 inhibitor of the present invention may be used in combination with other actives, such as oligonucleotide-based therapeutic agents—such as sequence specific oligonucleotide-based therapeutic agents—acting through nucleotide sequence-dependent mode of action.


By way of further example, the inhibitor of the present invention may be used in combination with one or more acetylcholinesterase inhibitors and/or one or more NMDA receptor antagonists. A cholinesterase inhibitor may be, for example, donepezil, tacrine, galantamine or rivastigmine. A NMDA receptor antagonist may be, for example, memantine.


By way of further example, the inhibitor of the present invention may be used in combination with one or more typical antipsychotics and/or one or more atypical antipsychotics. A typical antipsychotic may be, for example, chlorpromazine, fluphenazine, haloperidol, perphenazine, thioridazine, thiothixene, or trifluoperazine. An atypical antipsychotic may be, for example, aripiprazole, aripiprazole lauroxil, asenapine, brexpiprazole, cariprazine, clozapine, Iloperidone, lumateperone tosylate, lurasidone, olanzapine, paliperidone, aliperidone palmitate, or ziprasidone.


In some embodiments, the inhibitor of the present invention is used in combination with one or more of the following: an antisense compound that targets C9ORT72 (e.g., as described in WO 2014/062736); an antisense oligonucleotide, aptamer, miRNA, ribozyme, or siRNA that blocks expression of one or more of C3 convertase, C5, C6, C7, C8, and C9 (e.g., as described in WO 2008/044928); an antibody that blocks the activity of one or more of C3 convertase, C5, C6, C7, C8, and C9 (e.g., as described in WO 2008/044928); an antisense or double stranded RNA that decreases activity of the complement cascade (e.g., as described in WO 2005/060667); and an antibody that binds C1s protein, e.g., to inhibit proteolytic activity of C1s (e.g., as described in WO 2014/066744).


In some embodiments, the inhibitor of the present invention is used in combination with an antibody that binds to complement C4 or the C4b portion of C4 (e.g., as described in WO 2017/196969).


In some embodiments, the inhibitor of the present invention is used in combination with one or more nucleic acid molecules disclosed in U.S. Provisional Application filed May 11, 2020, entitled “Complement Component C4 Inhibitors For Treating A Neurological Disease, And Related Compositions, Systems And Methods Of Using Same” and US Provisional Application filed May 11, 2020, entitled “Complement Component C1S Inhibitors For Treating A Neurological Disease, And Related Compositions, Systems And Methods Of Using Same,”


Applications

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


In research, such nucleic acid molecules may be used to specifically modulate the synthesis of a C1R protein in cells (e.g. in vitro cell cultures) and animal models 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 corresponding to the protein, thereby preventing 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.


Methods of Detection or Diagnosis

Further encompassed by the present invention is a method for diagnosing a neurological disease in a patient suspected of a having a neurological disease, said method comprising the steps of

    • a) determining the amount of one or more C1R nucleic acids, such as C1R mRNA or cDNA derived from C1R mRNA, in a sample from the subject, wherein the determination comprises contacting the sample with one or more oligonucleotides of the present invention,
    • b) comparing the amount determined in step a) to a reference amount, and
    • c) diagnosing whether the subject suffers from the neurological disease, or not, based on the results of step c).


In some embodiments, the method of diagnosing a neurological disease is an in vitro method.


The term “neurological disease” has been defined elsewhere herein. The definition applies accordingly. In some embodiments, the neurological disease to be diagnosed is a tauopathy, such as Alzheimer's disease. In some embodiments, the neurological disease to be diagnosed is schizophrenia.


The term “sample” refers to a sample of a body fluid, to a sample of separated cells or to a sample from a tissue or an organ. Samples of body fluids can be obtained by well-known techniques and include samples of blood, plasma, serum, urine, lymphatic fluid, sputum, ascites, saliva, and lacrimal fluid. In some embodiments, the sample is a cerebrospinal fluid sample.


Tissue or organ samples may be obtained from any tissue or organ by, e.g., biopsy. In some embodiments, the sample is a neural tissue sample, such as a brain tissue sample or spinal cord sample.


In some embodiments, the sample comprises neuron, astrocytes, oligodendrocytes, and/or microglia cells.


The subject may be a mammal. In some embodiments, the subject is a human. In some embodiments, the subject is a human. In some embodiments, the subject is a cynomolgus monkey.


In step a) of the aforementioned method, the amount of C1R nucleic acid present in the sample shall be determined. The C1R nucleic acid to be determined shall be a nucleic acid encoding a C1R protein. In some embodiments, the C1R nucleic acid is mammalian C1R nucleic acid. In some embodiments, the C1R nucleic acid is a human C1R nucleic acid.


The C1R nucleic acid may for example be a gene, a RNA, a mRNA, and pre-mRNA, a mature mRNA or a cDNA sequence. In an embodiment, the nucleic acid is a C1R mRNA, such as. In another embodiment, the C1R nucleic acid is cDNA derived from a C1R mRNA.


In step b) of aforementioned method, the amount of the C1R nucleic acid shall be compared to a reference, i.e. to a reference amount. The terms “reference amount” or “reference” are well understood by the skilled person. Suitable reference amounts can, in principle, be calculated for a cohort of subjects based on the average or mean values for a given biomarker by applying standard methods of statistics. A suitable reference shall allow for the diagnosis of the neurological disease. Accordingly, the reference shall allow for differentiating between a patient suffering from a neurological disease and a subject who is not suffering from a neurological disease. In some embodiments, the reference is a predetermined value.


In some embodiments, an amount larger than the reference amount is indicative for a patient suffering from a neurological disease, whereas an amount of lower than the reference amount is indicative for a patient not suffering from neurological disease.


The determination of the amount of the one or more nucleic acids in step a) shall comprise contacting the sample with one or more oligonucleotides of the present invention. For example, the sample is contacted with said one or more oligonucleotides under conditions, which allow for the hybridization of said one or more oligonucleotides to the one or more C1R nucleic acids present in the sample (such as the C1R mRNA), thereby forming duplexes of said oligonucleotides and said C1R nucleic acids. In some embodiments, the amount of the one or more C1R nucleic acids is determined by determining the amount of the formed duplexes, e.g. via a detectable label.


Accordingly, the one or more oligonucleotides to be used in the above methods may comprise a detectable label.


Further encompassed by the present invention is a method for detecting one or more C1R nucleic acids in a sample, for example, in a sample as defined above. The method may comprise contacting the sample with one or more oligonucleotides of the present invention as described above. In some embodiments, the sample is from a patient having or suspected of a having a neurological disease.


Also encompassed by the present invention is an in vivo or in vitro method for modulating C1R expression in a target cell which is expressing C1R, 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 the brain. The target cell may be a brain cell. In some embodiments, the brain cell is selected from the group consisting of a neuron and a microglia cell.


One aspect of the present invention is related to the nucleic acid molecules or pharmaceutical compositions of the invention for use as a medicament.


In an aspect of the invention, the C1R inhibitor, such as a nucleic acid molecule or pharmaceutical composition of the invention is capable of reducing the amount of C1R in a cell expressing C1R.


For example, a nucleic acid molecule that inhibits C1R expression may reduce the C1R protein in an affected cell by at least 50% (e.g., 50-60%), or at least 60% (e.g., 60-70%), or at least 70% (e.g., 70-80%), at least 80% (e.g., 80-90%), or at least 90% (e.g., 90-95%) reduction compared to controls. The controls may be untreated cells or animals, or cells or animals treated with an appropriate control.


Inhibition of C1R expression may be measured by RT-qPCR, e.g. as described in the Materials and Methods section.


Due to the decrease of C1R levels, the nucleic acid molecules or pharmaceutical compositions of the present invention can be used to inhibit development of or in the treatment of neurological diseases.


Accordingly, one aspect of the present invention is related to use of an C1R inhibitor, such as the nucleic acid molecule or pharmaceutical compositions of the invention to decrease C1R protein in an individual having or susceptible to a neurological disease.


The subject to be treated with the C1R inhibitor, such as the nucleic acid molecules or pharmaceutical compositions of the invention (or who prophylactically receives nucleic acid molecules or pharmaceutical compositions of the present invention) is preferably a human, more preferably a human patient who has a neurological diseases, even more preferably a human patient having a tauopathy, even more preferably a human patient having Alzheimer's disease. In some embodiments, the human patient has schizophrenia.


Accordingly, the present invention relates to a method of treating a neurological disease, wherein the method comprises administering an effective amount of a C1R inhibitor, such as a nucleic acid molecule or pharmaceutical compositions of the invention. The present invention further relates to a method of preventing neurological diseases. In one embodiment, the C1R inhibitors of the present invention is not intended for the treatment of neurological diseases, only its prevention.


In some embodiments, the subject to be treated does not have a cardiovascular disorder or disease (e.g., as described in WO 2014/089121). In some embodiments, the subject to be treated does not require treatment for pain (e.g., as described in WO 2005/060667).


The invention also provides for the use of a C1R inhibitor, such as nucleic acid molecule or a pharmaceutical composition of the invention for the manufacture of a medicament, in particular a medicament for use in the treatment of a neurological disease. In preferred embodiments, the medicament is manufactured in a dosage form for intrathecal or intracranial administration.


The invention also provides for the use of the nucleic acid molecule or the pharmaceutical composition of the invention for the manufacture of a medicament wherein the medicament is in a dosage form for intravenous administration.


Kits

The invention also provides a kit containing the C1R inhibitor of the present invention, such as the nucleic acid molecule or pharmaceutical composition of the present invention, and instructions for administering the C1R inhibitor. The instructions may indicate that the C1R inhibitor may be used for the treatment of a neurological disease or neurodegenerative disorder as referred to herein, such as Alzheimer's disease or Schizophrenia.


The term “kit” as used herein refers to a packaged product comprising components with which to administer the C1R inhibitor of the present invention. The kit may comprise a box or container that holds the components of the kit. The kit can also include instructions for administering the C1R inhibitor of the present invention of the invention.


EXAMPLES
Materials and Methods
Example 1: Testing In Vitro Efficacy of Antisense Oligonucleotides Targeting C1R in Primary Mouse Hepatocytes

Cells were maintained in a humidified incubator as recommended by the supplier. The vendor and recommended culture conditions are reported in Table 5.









TABLE 5







Cell culture details.
















Incub.
Incub.





Seeding
time
time





density
before
with


Cell


(cells/
oligo
oligo


Line
Vendor
Culture Condition
well)
(hrs)
(hrs)





mouse
Minerva
WME (Sigma #W1878)
25000
24
72


hepatocytes
Imaging
w/FBS:







complemented with







1x







Pen/Strep/Glutamine







(freshly added), 10%







(v/v) FBS (Sigma







F7524), non-heat







inactivated.









For assays, cells were seeded in a 96-multi well plate in culture media and incubated as reported in Table 5 before addition of oligonucleotides dissolved in PBS. The seeding density of the cells is reported in Table 5.


Oligonucleotides were added at the concentrations reported in Table 7. The cells were harvested 72 hours after the addition of oligonucleotides (see Table 5). RNA was extracted using the RNeasy 96 kit (Qiagen) according to the manufacturer's instructions and eluted in 200 μL of water. The RNA was subsequently heated to 90° C. for one minute.


For gene expressions analysis, One Step RT-qPCR was performed using gScript™ XLT One-Step RT-qPCR ToughMix®, Low ROX™ (Quantabio) in a duplex set up. The primer assays used for qPCR are collated in Table 6 for both target and endogenous control.









TABLE 6







qPCR primer-probe details.












Endogen
Endog.
Endogen.


Target


contr.
contr.
contr.
Target
Target
fluoro-


assay
vendor
fluorophore
assay
vendor
phore





RPLP0:
IDT
HEX-ZEN
C1ra:
IDT
6-


Rplp0_


Mm.PT.58.13565

FAM/


MmPT5843


159

ZEN/


894205




IBFQ









The relative mouse C1ra and mouse C1rb mRNA expression level in Table 7 is shown as percent of control (PBS-treated cells). More information on the tested compounds can be found in Table 8.









TABLE 7







mRNA expression level (% of PBS-treated cells).

















C1RS mRNA


C1RS mRNA


C1RS mRNA




qPCR SP


qPCR SP


qPCR SP




probe1 mouse


probe1 mouse


probe1 mouse




hepatocytes


hepatocytes


hepatocytes


CMP ID

C1ra:
CMP ID

C1ra:
CMP ID
Conc
C1ra:


NO
Conc
AP015387
NO
Conc
AP015387
NO2
2
AP0153872


















100_1
0.3
62.2
121_1
0.06
59.1
79_1
0.3
64.9


101_1
0.06
71.9
121_1
0.3
15.5
80_1
0.06
107


101_1
0.3
17.6
122_1
0.06
31.7
80_1
0.3
73


102_1
0.06
84.3
122_1
0.3
9.6
81_1
0.06
67.7


102_1
0.3
68.1
123_1
0.06
33.7
81_1
0.3
38.2


103_1
0.06
56.4
123_1
0.3
8.3
82_1
0.06
83


103_1
0.3
23.9
124_1
0.06
12.5
82_1
0.3
46


104_1
0.06
110
124_1
0.3
4.4
83_1
0.06
70.6


104_1
0.3
59.7
125_1
0.06
24.3
83_1
0.3
55


105_1
0.06
58.9
126_1
0.06
12.2
84_1
0.06
88.5


105_1
0.3
43.6
126_1
0.3
1.9
84_1
0.3
36.6


106_1
0.06
72
65_1
0.06
129
85_1
0.06
80.6


106_1
0.3
37
65_1
0.3
110
85_1
0.3
36


107_1
0.06
84.4
66_1
0.06
75.6
86_1
0.06
90.6


107_1
0.3
38.4
66_1
0.3
101
86_1
0.3
78.2


108_1
0.06
104
67_1
0.06
127
87_1
0.06
136


108_1
0.3
68.2
67_1
0.3
116
87_1
0.3
61.1


109_1
0.06
79.3
68_1
0.06
89.6
88_1
0.06
47.2


109_1
0.3
52.8
68_1
0.3
86.6
88_1
0.3
22.7


110_1
0.06
118
69_1
0.06
79.2
89_1
0.06
85.4


110_1
0.3
46
69_1
0.3
56.4
89_1
0.3
71.8


111_1
0.06
114
70_1
0.06
80.2
90_1
0.06
94.6


111_1
0.3
72.8
70_1
0.3
55.1
90_1
0.3
91.3


112_1
0.06
83.9
71_1
0.06
81.7
91_1
0.06
105


112_1
0.3
76.9
71_1
0.3
62.6
91_1
0.3
90.7


113_1
0.06
90.3
72_1
0.06
64.2
92_1
0.06
78.1


113_1
0.3
73.3
72_1
0.3
66.8
92_1
0.3
75.2


114_1
0.06
110
73_1
0.06
106
93_1
0.06
99.8


114_1
0.3
87.4
73_1
0.3
79.3
93_1
0.3
102


115_1
0.06
70
74_1
0.06
119
94_1
0.06
83


115_1
0.3
35.3
74_1
0.3
67
94_1
0.3
68


116_1
0.06
87.8
75_1
0.06
86
95_1
0.06
79.3


116_1
0.3
95.3
75_1
0.3
61.1
95_1
0.3
59.9


117_1
0.06
75.3
76_1
0.06
121
96_1
0.06
67.1


117_1
0.3
50.3
76_1
0.3
99.6
96_1
0.3
52.7


118_1
0.06
110
77_1
0.06
76.8
97_1
0.06
81.6


118_1
0.3
56.2
77_1
0.3
54.4
97_1
0.3
44.3


119_1
0.06
64.6
78_1
0.06
63.9
98_1
0.06
93.9


119_1
0.3
12.9
78_1
0.3
34.4
98_1
0.3
52.1


120_1
0.06
43.1
79_1
0.06
90.7
99_1
0.06
63.3








99_1
0.3
30









From Table 7 it can be taken that the C1R pool is capable of reducing C1R mRNA efficiently at different concentrations.


The invention provides the following oligonucleotide compounds (Table 8):









TABLE 8







Oligonucleotide compounds















SEQ


SEQ

CMP





ID NO
Motif
Design
ID NO:
Compound
ID NO
ΔG °
start_C1ra
start_C1rb


















23
ATCAGTCTTACTCACAT
2-11-4
 85
ATcagtcttactcACAT
65_1
−19.9
  395
  164





24
TACCCATCAGTCTTAC
4-10-2
 86
TACCcatcagtcttAC
66_1
−21.7
  401
  170





25
CGCCTACCTTAACATA
2-11-3
 87
CGcctaccttaacATA
67_1
−19.9
 1465
 1280





26
ACGCCTACCTTAACAT
3-11-2
 88
ACGcctaccttaacAT
68_1
−19.7
 1466
 1281





27
GACGCCTACCTTAACA
2-12-2
 89
GAcgcctaccttaaCA
69_1
−20.4
 1467
 1282





28
TGACGCCTACCTTAA
2-9-4
 90
TGacgcctaccTTAA
70_1
−19.7
 1469
 1284





29
TCCTCATGAATCTCAC
3-11-2
 91
TCCtcatgaatctcAC
71_1
−19.2
 1636
 1451





30
CTAACTCCAATACATA
4-8-4
 92
CTAActccaataCATA
72_1
−19.9
 1693
 1508





31
TGTCCTAACTCCAATACAT
2-15-2
 93
TGtcctaactccaatacAT
73_1
−20.6
 1694
 1509





32
TCCTAACTCCAATACAT
2-11-4
 94
TOctaactccaatACAT
74_1
−20.1
 1694
 1509





33
GTCCTAACTCCAATACA
3-12-2
 95
GTCctaactccaataCA
75_1
−21.2
 1695
 1510





34
TGTCCTAACTCCAATA
2-11-3
 96
TGtcctaactccaATA
76_1
−18.9
 1697
 1512





35
GTGTCCTAACTCCAAT
2-10-4
 97
GTgtcctaactcCAAT
77_1
−21.5
 1698
 1513





36
CACAGATTCCTTACTTT
3-11-3
 98
CACagattccttacTTT
78_1
−19.9
 2101
 1917





37
TCACAGATTCOTTACTT
2-12-3
 99
TCacagattccttaCTT
79_1
−19.6
 2102
 1918





38
ATCTAATGTTACTATC
4-8-4
100
ATCTaatgttacTATC
80_1
−18.8
 3158
 2815





39
ATTAAGAAATTACTACA
4-9-4
101
ATTAagaaattacTACA
81_1
−16.4
 3532
 3189





40
ACAGCCAATCATATCTCA
2-14-2
102
ACagccaatcatatctCA
82_1
−20.3
 3687
 3344





41
CAGCCAATCATATCTCA
2-13-2
103
CAgccaatcatatctCA
83_1
−20.2
 3687
 3344





42
ATACAGCCAATCATATC
4-10-3
104
ATACagccaatcatATC
84_1
−20.2
 3690
 3347





43
AATACAGCCAATCATAT
4-9-4
105
AATAcagccaatcATAT
85_1
−20.1
 3691
 3348





44
TACTCTAGACCTCTAT
2-10-4
106
TActctagacctCTAT
86_1
−20.5
 4860
 4516





45
CTACTCTAGACCTCTA
2-11-3
107
CTactctagacctCTA
87_1
−20.7
 4861
 4517





46
TTTACACAAGTCCCCT
2-12-2
108
TTtacacaagtcccCT
88_1
−19.7
 4899
 4555





47
TTCTCTTCTTTTTACACAA
3-13-3
109
TTCtcttctttttacaCAA
89_1
−20.6
 4906
 4562





48
GTTCTCTTCTTTTTACA
2-13-2
110
GTtctcttctttttaCA
90_1
−18.5
 4909
 4565





49
AGTTCTCTTCTTTTTACA
3-13-2
111
AGTtctcttctttttaCA
91_1
−21.1
 4909
 4565





50
GTTATAAATAACTTATTA
4-10-4
112
GTTAtaaataacttATTA
92_1
−15.9
 6231
 5969





51
GTTATAAATAACTTATT
4-9-4
113
GTTAtaaataactTATT
93_1
−15.5
 6232
 5970





52
TAAAATCATCCTCATCTAT
3-13-3
114
TAAaatcatcctcatcTAT
94_1
−19.6
 8405
 8499





53
TAAAATCATCCTCATCTA
2-12-4
115
TAaaatcatcctcaTCTA
95_1
−19.8
 8406
 8500





54
TAAAATCATCCTCATCT
3-10-4
116
TAAaatcatcctcATCT
96_1
−18.9
 8407
 8501





55
TTAAAATCATCCTCATC
4-9-4
117
TTAAaatcatcctCATC
97_1
−19.3
 8408
 8502





56
GTTTAAAATCATCCTCATC
3-13-3
118
GTTtaaaatcatcctcATC
98_1
−19.9
 8408
 8502





57
TGTTTAAAATCATCCTCA
4-12-2
119
TGTTtaaaatcatcctCA
99_1
−19.9
 8410
 8504





58
AAGATTAAACCCTTCCTT
2-13-3
120
AAgattaaacccttcCTT
100_1
−19.9
 8594
 8726





59
TAAGATTAAACCCTTCC
3-11-3
121
TAAgattaaaccctTCC
101_1
−20.2
 8596
 8728





60
CTAAGATTAAACCCTTCC
2-14-2
122
CTaagattaaacccttCC
102_1
−20.5
 8596
 8728





61
ACTAAGATTAAACCCTT
4-10-3
123
ACTAagattaaaccCTT
103_1
−19.6
 8598
 8730





62
CTGTATAACTCTTTCCA
3-12-2
124
CTGtataactctttcCA
104_1
−20.2
 8866
 8996





63
CCTGTATAACTCTTTCCA
2-14-2
125
CCtgtataactctttcCA
105_1
−22.5
 8866
 8996





64
CCTGTATAACTOTTTCC
2-13-2
126
CCtgtataactctttCC
106_1
−21.6
 8867
 8997





65
CTTAGCTATATATATTT
4-9-4
127
CTTAgctatatatATTT
107_1
−18.1
 8977
 9090





66
TCATCCACTCTTTACCT
3-12-2
128
TCAtccactctttacCT
108_1
−22.8
 9289
 9407





67
CCTTACTTGATCTCTC
3-11-2
129
CCTtacttgatctcTC
109_1
−20.5
 9356
 9471





68
TOTTGACTTTACTCTCT
3-12-2
130
TCTtgactttactctCT
110_1
−20
 9508
 9623





69
TCTTACTAATTCCTTCT
2-11-4
131
TCttactaattccTTCT
111_1
−20.1
 9534
 9649





70
GTTCTTACTAATTCCTTC
2-13-3
132
GTtcttactaattccTTC
112_1
−19.8
 9535
 9650





71
GTTCTTACTAATTCCTT
3-11-3
133
GTTcttactaattcCTT
113_1
−19.7
 9536
 9651





72
CTGTTCTTACTAATTCC
2-12-3
134
CTgttcttactaatTCC
114_1
−20.1
 9538
 9653





73
CCTGTTCTTACTAATTC
3-12-2
135
CCTgttcttactaatTC
115_1
−20.1
 9539
 9654





74
CCCTGTTCTTACTAATT
2-13-2
136
CCctgttcttactaaTT
116_1
−19.6
 9540
 9655





75
CCGCTGACATAACCC
2-11-2
137
CCgctgacataacCC
117_1
−21.8
10228
10385





76
CATATCCCTCACCACAC
2-13-2
138
CAtatccctcaccacAC
118_1
−21.7
10495
10652





77
TCCAGTCAACATAATTCA
2-12-4
139
TCcagtcaacataaTTCA
119_1
−20.5
10530
10687





78
CCAGTCAACATAATTCA
2-11-4
140
CCagtcaacataaTTCA
120_1
−20.2
10530
10687





79
CCAGTCAACATAATTC
3-9-4
141
CCAgtcaacataATTC
121_1
−20
10531
10688





80
TTTAATACACTATACCCT
4-12-2
142
TTTAatacactataccCT
122_1
−19.8
10603
10760





81
TTTTTAATACACTATACCCT
3-15-2
143
TTTttaatacactataccCT
123_1
−20.3
10603
10760





82
TTAATACACTATACCCT
3-11-3
144
TTAatacactatacCCT
124_1
−20.3
10603
10760





83
TAATACACTATACCCT
4-9-3
145
TAATacactatacCCT
125_1
−20.6
10603
10760





84
TTTTAATACACTATACC
4-9-4
146
TTTTaatacactaTACC
126_1
−19
10605
10762









In the table, capital letters are beta-D-oxy LNA nucleosides, lowercase letters are DNA nucleosides, all LNA C are 5-methyl cytosine, and all internucleoside linkages are phosphorothioate internucleoside linkages.

Claims
  • 1. (canceled)
  • 2. (canceled)
  • 3. (canceled)
  • 4. (canceled)
  • 5. (canceled)
  • 6. (canceled)
  • 7. (canceled)
  • 8. (canceled)
  • 9. A nucleic acid molecule of 12 to 30 nucleotides in length comprising a contiguous nucleotides sequence of at least 12 nucleotides which is at least 95% complementary to a mammalian C1R target sequence, wherein the nucleic acid molecule is capable of inhibiting the expression of a C1R mRNA.
  • 10. The nucleic acid molecule according to claim 9, wherein the contiguous nucleotide sequence is fully complementary to a sequence selected from the group consisting of SEQ ID NOs: 3 and 6.
  • 11. The nucleic acid molecule according to claim 9, wherein the nucleic acid molecule comprises a contiguous nucleotide sequence of 12 to 25 nucleotides in length.
  • 12. The nucleic acid molecule of claim 9, wherein the nucleic acid molecule is a RNAi molecule.
  • 13. The nucleic acid molecule of claim 9, wherein the nucleic acid molecule is a single stranded antisense oligonucleotide.
  • 14. (canceled)
  • 15. The nucleic acid molecule according to claim 9, 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 9, where the contiguous nucleotide sequence comprises at least one phosphorothioate internucleoside linkage.
  • 19. The nucleic acid molecule according to claim 18, wherein at least 90% of the internucleoside linkages within the contiguous nucleotide sequence are phosphorothioate internucleoside linkages.
  • 20. The nucleic acid molecule according to claim 9, 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.
  • 21. A pharmaceutically acceptable salt of a nucleic acid molecule according to claim 9.
  • 22. A pharmaceutical composition comprising a nucleic acid molecule according to claim 9, or a pharmaceutically acceptable salt of the nucleic acid molecule and a pharmaceutically acceptable excipient.
  • 23. An in vivo or in vitro method for inhibiting C1R expression in a target cell which is expressing C1R, said method comprising administering a nucleic acid molecule according to claim 9, a pharmaceutically acceptable salt of the nucleic acid molecule, or a pharmaceutical composition comprising the nucleic acid molecule in an effective amount to said cell.
  • 24. A method for treating a disease comprising administering a therapeutically or prophylactically effective amount of a nucleic acid molecule according to claim 9, a pharmaceutically acceptable salt of the nucleic acid molecule, or a pharmaceutical composition comprising the nucleic acid molecule, to a subject suffering from or susceptible to a neurological disease.
  • 25. A method according to claim 24, wherein a neurological disease is selected from the group consisting of a tauopathy and schizophrenia.
  • 26. (canceled)
  • 27. (canceled)
  • 28. (canceled)
  • 29. (canceled)
  • 30. (canceled)
  • 31. A method for diagnosing a neurological disease in a patient suspected of a having a neurological disease, said method comprising the steps of a) determining the amount of one or more C1R nucleic acids in a sample from the subject, wherein the determination comprises contacting the sample with one or more nucleic acid molecules as defined in claim 9,b) comparing the amount determined in step a) to a reference amount, andc) diagnosing whether the subject suffers from the neurological disease, or not, based on the results of step b).
  • 32. The method of claim 31, wherein the sample is contacted in step a) with said one or more nucleic acid molecules under conditions which allow for the hybridization of said one or more nucleic acid molecules to said one or more C1R nucleic acids present in the sample, thereby forming duplexes of said nucleic acid molecules and said C1R nucleic acids.
  • 33. A method for manufacturing a nucleic acid molecule as defined in claim 9, comprising reacting nucleotide units and thereby forming covalently linked contiguous nucleotide units comprised in the nucleic acid molecule.
  • 34. The method of claim 33, wherein the method comprises the introduction of one or more sugar-modified nucleosides, of one or more modified internucleoside linkages, and/or of one or more modified nucleobases into the nucleic acid molecule.
Provisional Applications (1)
Number Date Country
63023113 May 2020 US
Continuations (1)
Number Date Country
Parent PCT/US2021/031292 May 2021 US
Child 18054113 US