WO2021195723A1 - "methods for treatment of coronavirus infections" - Google Patents

"methods for treatment of coronavirus infections" Download PDF

Info

Publication number
WO2021195723A1
WO2021195723A1 PCT/AU2021/050317 AU2021050317W WO2021195723A1 WO 2021195723 A1 WO2021195723 A1 WO 2021195723A1 AU 2021050317 W AU2021050317 W AU 2021050317W WO 2021195723 A1 WO2021195723 A1 WO 2021195723A1
Authority
WO
WIPO (PCT)
Prior art keywords
lsd1
polypeptide
cell
trans
agent
Prior art date
Application number
PCT/AU2021/050317
Other languages
French (fr)
Inventor
Sudha RAO
Original Assignee
The Council Of The Queensland Institute Of Medical Research
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU2020901054A external-priority patent/AU2020901054A0/en
Application filed by The Council Of The Queensland Institute Of Medical Research filed Critical The Council Of The Queensland Institute Of Medical Research
Publication of WO2021195723A1 publication Critical patent/WO2021195723A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/15Oximes (>C=N—O—); Hydrazines (>N—N<); Hydrazones (>N—N=) ; Imines (C—N=C)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • A61K31/167Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide having the nitrogen of a carboxamide group directly attached to the aromatic ring, e.g. lidocaine, paracetamol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4409Non condensed pyridines; Hydrogenated derivatives thereof only substituted in position 4, e.g. isoniazid, iproniazid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/21Interferons [IFN]
    • A61K38/215IFN-beta
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/44Oxidoreductases (1)
    • A61K38/443Oxidoreductases (1) acting on CH-OH groups as donors, e.g. glucose oxidase, lactate dehydrogenase (1.1)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1137Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against enzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.

Definitions

  • This invention relates generally to methods for treating coronavirus infections. More specifically, the invention relates to the use of LSD antagonists to treat coronavirus infections, including betacoronavirus infections.
  • Coronaviruses are enveloped RNA viruses that infect mammals and birds.
  • the severe acute respiratory syndrome (SARS) and the Middle East respiratory syndrome (MERS) are both members of the genus Betacoronavirus, and responsible for hundreds of deaths in Asia and the Middle East, respectively.
  • SARS-CoV-2 SARS-coronavirus 2
  • the coronaviruses are a virus family grouped into four genera, being the alphacoronavirus, betacoronavirus (b-CoVs), gammacoronavirus, and deltacoronavirus.
  • the alphacoronaviruses and betacoronaviruses infect a wide range of species, including humans.
  • the b-CoVs that are of particular clinical importance in humans include OC43 and HKU1 of the A lineage, Severe Acute Respiratory Syndrome coronavirus (8ARS ⁇ CoV) and SARS-CoV-2 (which causes the disease COVID-19) of the B lineage, and Middle Eastern Respiratory Syndrome-related coronavirus (MERS-CoV) of the C lineage.
  • the present invention is predicated, at least in part, on the discovery that LSD1 plays a part in regulating key demethylation/methylation sites of the ACE2 polypeptide, which is an important part of the molecular machinery used by SARS- CoV-2 to gain entry into the host cell. Additionally, LSD1 also plays a role in demethylation/methylation of key lysine residues present on the C-terminal cytoplasmic tail of the ACE2 polypeptide, which is known to regulate the translocation of the ACE2 polypeptide from the cell membrane to the cell nucleus. Accordingly, these findings lead the present inventors to contemplate the use of LSD1 inhibitors for the treatment and/or prevention of virus infections, namely coronavirus infections.
  • a coronavirus infection in a subject having a coronavirus infection comprising administering to the subject an agent that inhibits or reduces an activity of a lysine-specific demethylase 1 (LSD1) polypeptide, to thereby treat the coronavirus infection in the subject.
  • LSD1 lysine-specific demethylase 1
  • the invention provides methods of preventing a coronavirus from entering a cell, the method comprising, exposing the cell to an agent that inhibits or reduces an activity of a LSD1 polypeptide , to thereby reduce or prevent the coronavirus from entering the cell.
  • the agent that inhibits or reduces an activity of a LSD1 polypeptide is exposed to the cell for a time and under conditions sufficient to antagonise a TMPRSS2 polypeptide.
  • the agent that inhibits or reduces an activity of a LSD1 polypeptide is exposed to the cell for a time and under conditions sufficient to antagonise a virus cell entry receptor polypeptide.
  • the virus cell entry receptor polypeptide is selected from the group comprising an ACE2 polypeptide, a DPP4 polypeptide, and a CD13 polypeptide.
  • the invention provides a method of reducing or preventing entry of a coronavirus into a cell, the method comprising exposing the cell to an agent that inhibits an activity of a lysine-specific demethylase 1 (LSD1) polypeptide, to thereby antagonise, or otherwise reduce the level or amount of TMPRSS2 polypeptide present on the surface of the cell.
  • LSD1 lysine-specific demethylase 1
  • the invention provides a method of reducing or preventing entry of a coronavirus into a cell, the method comprising exposing the cell to an agent that inhibits an activity of a lysine-specific demethylase 1 (LSD1) polypeptide, to thereby antagonise, or otherwise reduce the level or amount of a virus cell entry receptor polypeptide present on the surface of the cell.
  • LSD1 lysine-specific demethylase 1
  • the agent is a selective LSD1 inhibitor.
  • the agent that inhibits or reduces an activity of a LSD1 polypeptide is selected from the group comprising: a small molecule; a polypeptide; and an antigen-binding molecule.
  • the agent is a monoamine oxidase (MAO) inhibitor.
  • MAO monoamine oxidase
  • a large number of MAO inhibitor are known to be suitable for the purposes of the present invention.
  • the MAO inhibitor comprises, consists, or consists essentially of, phenelzine.
  • the agent is a selective LSD1 inhibitor.
  • the selective LSD1 inhibitor can be selected from the group comprising or consisting of GSK-LSD1 , SP-2509, and SP-2577.
  • the agent is GSK-LSD1 , which has the following molecular structure:
  • the agent is SP-2509, which has the following molecular structure:
  • the agent is SP-2577, which has the following molecular structure:
  • the agent is bizine, which has the following molecular structure:
  • the agent is an isolated or purified proteinaceous molecule comprising, consisting or consisting essentially of sequence corresponding to residues 108 to 118 of LSD1.
  • the isolated or purified proteinaceous molecule is an isolated or purified proteinaceous molecule represented by Formula (I):
  • Zi and Z2 are independently absent or are independently selected from at least one of a proteinaceous moiety comprising from about 1 to about 50 amino acid residues (and all integers in between), and a protecting moiety; and
  • Xi is selected from small amino acid residues, including S, T, A, G, and modified forms thereof.
  • Xi is selected from S and A.
  • Zi is a proteinaceous molecule represented by Formula (II):
  • X2 is absent or is a protecting moiety
  • X3 is absent or is selected from any amino acid residue
  • X4 is selected from any amino acid residue.
  • X3 is selected from basis amino acid residues including R, K, and modified forms thereof.
  • X4 is selected from aromatic amino acid residues, including F, Y, W, and modified forms thereof.
  • Z2 is absent.
  • the isolated or purified proteinaceous molecule of Formula (I) comprises, consists or consists essentially of an amino acid sequence represented by SEQ ID NO: 1 , 2, or 3 (RRTSRRKRAKV [SEQ ID NO: 1]; RRTARRKRAKV [SEQ ID NO: 2]; or RWRRTARRKRAKV [SEQ ID NO: 3]).
  • the proteinaceous molecule of Formula (I) further comprises at least one membrane permeating moiety.
  • the membrane permeating moiety is a lipid moiety.
  • the membrane permeating moiety is a myristoyl group.
  • the coronavirus is selected from the group comprising SARS-CoV, SARS-CoV-2, and MERS-CoV.
  • the coronavirus is SARS-CoV-2.
  • the subject is a human.
  • the agent that inhibits or reduces an activity of a LSD1 polypeptide is an indirect inhibitor of the LSD1 polypeptide.
  • the agent that inhibits or reduces an activity of a LSD polypeptide may prevent the interaction between an LSD polypeptide and a RKOQ polypeptide.
  • the agent prevents the phosphorylation of a phosphorylation site of the LSD polypeptide.
  • the present invention provides a method of treating a coronavirus infection in a subject having a coronavirus infection, the method comprising administering to the subject an agent that reduces the level of a lysine-specific demethylase (LSD) polypeptide, to thereby treat the coronavirus infection in the subject.
  • the agent may be an interfering nucleic acid.
  • the LSD polypeptide is selected from an LSD1 polypeptide or an LSD2 polypeptide. In some preferred embodiments, the LSD polypeptide is an LSD1 polypeptide.
  • the present invention provides a method of treating a coronavirus infection in a subject having a coronavirus infection, the method comprising administering to the subject an agent that reduces the level or activity of a CoREST polypeptide, to thereby treat the coronavirus infection in the subject.
  • an agent that reduces the level or activity of a CoREST polypeptide to thereby treat the coronavirus infection in the subject.
  • the present invention provides a LSD1 inhibitor for use in the treatment of a coronavirus infection.
  • the present invention provides a PKC9 inhibitor for use in the treatment of a coronavirus infection.
  • the coronavirus is a betacoronavirus.
  • the betacoronavirus is SARS- CoV-2.
  • the LSD1 inhibitor is selected from GSK-LSD1 , SP-2509, SP-2577, bizine, and phenelzine.
  • the present invention provides the use of a LSD1 inhibitor in the manufacture of a medicament for the prevention and/or treatment of a coronavirus infection (e.g., a betacoronavirus infection).
  • a coronavirus infection e.g., a betacoronavirus infection
  • compositions for treating coronavirus comprising (i) an agent that inhibits or reduces an activity of a lysine-specific demethylase-1 (LSD1) polypeptide; and (ii) an antiviral agent.
  • LSD1 lysine-specific demethylase-1
  • the antiviral agent is selected from the group comprising hydroxychloroquine, chloroquine, lopinavir, ritonavir, favipiravir, and remdesivir.
  • the antiviral agent comprises an IFN-b polypeptide.
  • the present invention provides methods of inhibiting the phosphorylating activity of a protein kinase C (PKC), comprising contacting a cell infected with a coronavirus with an isolated or purified proteinaceous molecule comprising, consisting or consisting essentially of a sequence corresponding to residues 108 to 118 of LSD1.
  • PLC protein kinase C
  • the present invention provides methods of preventing S protein priming by a coronavirus, the method comprising administering a TMPRSS2 antagonist to a cell infected by the coronavirus, wherein the TMPRSS2 antagonist is an inhibitor of at least one interaction between an LSD1 polypeptide and another polypeptide (e.g., a PKC0 polypeptide).
  • a TMPRSS2 antagonist is an inhibitor of at least one interaction between an LSD1 polypeptide and another polypeptide (e.g., a PKC0 polypeptide).
  • the present invention provides a method of preventing or reducing coronavirus entry into a cell, the method comprising exposing the cell to an LSD1 inhibitor, wherein the LSD1 inhibitor reduces the expression of ACE2 by the cell, to thereby inhibit coronavirus entry into the cell.
  • Figure 1 provides a graphical representations showing the role of LSD1 on virus entry.
  • A Nanostring analysis of TMPRSS2 expression in MDA- MB-231 (TNBC breast cancer cell line) or MCF7 (epithelial breast cancer cell line) treated with siRNA targeting LSD1 or control siRNA.
  • B Nanostring analysis of TMPRSS2 expression in MDA-MB-231 (TNBC breast cancer cell line) MCF7 (epithelial breast cancer cell line), CT26 (Colorectal cancer cell line) or Huh7 (liver cancer cell line).
  • GSK-LSD1 catalytic LSD1 inhibitor
  • Phenelzine Dual catalytic/nuclear axis LSD1p inhibitor.
  • FIG. 2 shows that LSD1 and ACE2 associate as a complex on cell surface in SARS-CoV-2 susceptible cells.
  • A Representative images of CaCo2 cells imaged with the ASI digital pathology system. Cells are either permeabilized (intracellular) or not permeabilized (surface) and stained for expression of ACE2, LSD1 and TRMPSS2. Scale bar represents 10 pm.
  • B Dot graphs display the nuclear fluorescence intensity in Caco-2 cells for ACE2, LSD1 and TRMPSS2 from (A). > 50 cells counted per group. Data represent mean ⁇ SE. Mann-Whitney-test. **p ⁇ 0.01 , ***p ⁇ 0.001 , ****p ⁇ 0.0001 denote significant differences n.s. denotes non-significant.
  • +1 perfect colocalization.
  • C Representative FACS plot showing the cell surface and intracellular expression of ACE2 and LSD1 in Caco-2 cells. The numbers in each quadrant indicate the percentage of the total cell population, which also shown in dot plot (D). Data in dot plot represent two independent biological replicates.
  • E Representative image of MRC5 cells imaged with the ASI digital pathology system, that are either permeabilized (intracellular) or not permeabilized (surface) and stained for expression of ACE2, LSD1 and TRMPSS2. Scale bar represents 10 pm.
  • F Dot graphs displays the nuclear fluorescence intensity in MRC5 cells for ACE2, LSD1 and TRMPSS2 from (E). > 50 cells counted per group. Data represent mean ⁇ SE. Mann-Whitney-test. **p ⁇ 0.01 , ***p ⁇ 0.001 , ****p ⁇ 0.0001 denote significant differences n.s. denotes non-significant.
  • Figure 3 provides graphical representation showing LSD1 and ACE2 have increased association on the cell surface in SARS-CoV-2 infected cells.
  • A, B Representative image of Caco-2-SARS-CoV-2 infected cells imaged with the ASI digital pathology system (MRC5/Caco-2 uninfected not show) are shown, cells were either (A) permeabilized (intracellular) with 0.5% Triton X-100 for 15 minutes or (B) not permeabilized (surface) and stained for with primary antibodies against ACE2, LSD1 and SARS-CoV-2 nucleocapsid protein. Scale bar represents 12 mm.
  • Dot graphs displays the nuclear fluorescence intensity in Caco-2 cells for ACE2, LSD1 and SARS-CoV-2 from (A). >50 cells counted per group. Data represent mean ⁇ SE. Mann-Whitney-test. **p ⁇ 0.01 , ***p ⁇ 0.001 , ****p ⁇ 0.0001 denote significant differences n.s. denotes non-significant.
  • C Representative image of Caco-2 or Caco-2-SARS-CoV-2 infected cells imaged with the ASI digital pathology system are shown, cells were either permeabilized (intracellular) with 0.5% Triton X-100 for 15 minutes or not permeabilized (surface) and stained for with primary antibodies against ACE2, LSD1 and SARS-CoV-2 SPIKE protein. Scale bar represents 12 mm. Dot graphs displays the nuclear fluorescence intensity in Caco-2 cells for ACE2, LSD1 and SARS-CoV-2 from (C). > 50 cells counted per group. Data represent mean ⁇ SE. Mann-Whitney-test.
  • (E) FACS analysis of the expression of SARS-CoV-2 nucleocapsid protein, cell surface ACE2 and intracellular LSD1 in Caco-2 cells after 48 hours post infection. The unit of / axis indicates the percentage of the total cell population. Data represent mean ⁇ SD, n 2.
  • FIG. 4 provides graphical representations showing LSD1 directly interacting with the ACE2 cytoplasmic tail that harbours high affinity LSD1 demethylation domain.
  • A, B Representative image of Caco-2 cells imaged with the ASI digital pathology system are shown. Caco-2 cells were treated with vehicle control or 200 mM of phenelzine and imaged with the ASI digital pathology system are shown, cells were either (A) permeabilized (intracellular) with 0.5% Triton X-100 for 15 minutes or (B) non-permeabilized (surface) and stained for with primary antibodies against ACE2, LSD1 and SARS-CoV-2 nucleocapsid protein. Scale bar represents 12 mm.
  • Figure 5 provides a graphical representation of LSD1 inhibitors abrogating ACE2 expression and inhibiting SARS-CoV-2 expression.
  • n.s. denotes non-significant.
  • C Representative image of CaCo2-SARS-CoV-2 infected cells treated with phenelzine (P400 mM), GSK (G400 mM) or EPI-111 (50 mM) imaged with the ASI digital pathology system are shown, cells were permeabilized with 0.5% Triton X-100 for 15 minutes and stained for with primary antibodies against ACE2, LSD1 and SARS-CoV-2. Scale bar represents 15 mm.
  • Representative images are show for the following pairs: LSD1 & ACE2.
  • PLA signal intensity of the Duolink® assay is shown for average dot intensity (single Duolink® dot) or overall cell intensity for each cell (G)
  • Duolink® proximity ligation assay measurements of protein interactions were performed on Caco-2 cells infected with SARS-CoV-2 and treated with Control, Nardil® 400 mM, or GSK-LSD1 400 mM, permeabilized with 0.5% Triton X-100 and subject to Duolink® assay which produces a single bright dot per interaction within the cell.
  • Representative images are show for the following pairs: LSD1 & ACE2.
  • PLA signal intensity of the Duolink® assay is shown for average dot intensity (single Duolink® dot) or overall cell intensity for each cell.
  • FIG. 6 provides a graphical representation of a global transcript analysis.
  • A Caco-2 cells were treated with phenelzine, GSK-LSD1 or L1 and global RNA transcriptome analysis shows that key anti-viral and transcription processes are impacted.
  • the heat map above focuses on a DEGs list related to ISG, IFN-I, cytokine/chemokine activity, and viral entry, nuclear import/RNA synthesis, translation and replication.
  • the heatmap graph depicts the log2 (fold change) of DEGs of inhibition treated compared with control cells. Those selected DEGs have a log2(fold change) of more than 1 and FDR value of less than 0.01.
  • the heatmap values depict the log2-fold change (logFC) of DEGs from treated cells compared with control cells (GSK-LSD1 vs.
  • E Dot plot visualization of the top enriched Reactome pathways in treated cells compared to control cells. The dot colour represents the false discovery rate (FDR) value for each enriched Reactome pathway and size represents the gene ratio.
  • F Dot plot visualization of enriched Reactome pathways for GSK-LSD1 vs. control (left) and phenelzine vs. control (right). The dot colour represents the false discovery rate (FDR) value for each enriched Reactome pathway and size represents the gene ratio.
  • Figure 7 shows a graphical representation of the interplay of intracellular ACE2 in infected cells.
  • A Representative image of Caco-2 or MRC5 SARS-CoV-2 infected cells are depicted. Scale bar represents 15 mm.
  • B Cells were permeabilized and imaged with the ASI digital pathology system are shown, cells were stained for with primary antibodies against SARS-CoV-2 (nucleocapsid), ACE2 and LSD1. Dot graphs displays the nuclear fluorescence intensity in Caco-2 cells for ACE2, LSD1. 20 or more cells counted per group. Data represent mean ⁇ SEM. Mann-Whitney-test.
  • Dot graphs displays the nuclear fluorescence intensity in Caco-2 cells for ACE2, LSD1. 20 or more cells counted per group. Data represent mean ⁇ SEM. Mann-Whitney-test. * p ⁇ 0.0181 , **p ⁇ 0.01 , ***p ⁇ 0.001 , ****p ⁇ 0.0001 denote significant differences n.s. denotes non-significant.
  • the nuclear/cytoplasmic fluorescence ratio (Fn/c) using the equation: Fn/c (Fn- Fb)/(Fc - Fb), where Fn is nuclear fluorescence, Fc is cytoplasmic fluorescence, and the dotted line indicates background fluorescence.
  • the Mann- Whitney nonparametric test (GraphPad Prism, GraphPad Software, San Diego, CA) was used to determine significant differences between datasets.
  • Figure 8 shows graphical representations of ACE2 peptide inhibitor effect on nucleocapsid and Spike protein of SARS-Cov-2.
  • Figure 9 shows a graphical representation showing HBEC ALI cell culture with SARS-CoV-2 infection.
  • HBECs were pre-treated with and without Phenelzine (400 mM) for 24hr, followed by SARS-CoV-2 infection (10 4 PFU) for 2 hr.
  • Virus was removed by HBSS wash.
  • Infected HEBCs were cultured with and without Phenelzine (400 mM) for 24 hr and 48 hr post-infection.
  • qRT-PCR analysis was performed to detect the replicates of SARS-CoV-2 in infected cells at indicated time points of post-infection. The limit of detection is 100 copies/ml.
  • FIG. 10 provides graphical representations of LSD1-ACE2 interactions and the effect of LSD1 on the spike protein.
  • A Dot plot quantification of the fluorescence intensity (cell surface) of SARS-CoV-2 spike protein in SARS-CoV-2- infected Caco-2 cells with phenelzine or GSK treatment. >50 cells were analyzed for each group and were quantified using the digital pathology assay (ASI system).
  • the graph illustrates the number of gene transcripts from three replicates normalized to the geometric mean of HPRT1, GAPDH, and ACTB. Statistical significance was calculated using one-way ANOVA. NS, not significant.
  • D Schematic of SARS-CoV-2 infection assays. Caco-2 cells were seeded 24 h before the experiment. Then, cells were treated with each drug component for 48 h followed by SARS-CoV-2 infection
  • Representative images are shown for ACE2 and pan-methylation lysine antibody.
  • Duolink ® proximity ligation assay measurements of protein interactions were performed on unpermeabilized Caco-2 cells infected with SARS-CoV-2 and treated with control, or GSK inhibitors. The Duolink assay produces a single bright dot per interaction within the cell.
  • Representative images are shown for ACE2 and SARS-CoV-2 Spike Duolink ® .
  • a cell means one cell or more than one cell.
  • administering concurrently or “co-administering” and the like refer to the administration of a single composition containing two or more actives, or the administration of each active as separate compositions and/or delivered by separate routes either contemporaneously or simultaneously or sequentially within a short enough period of time that the effective result is equivalent to that obtained when all such actives are administered as a single composition.
  • simultaneous is meant that the active agents are administered at substantially the same time, and desirably together in the same formulation.
  • temporary it is meant that the active agents are administered closely in time, e.g., one agent is administered within from about one minute to within about one day before or after another.
  • any contemporaneous time is useful. However, it will often be the case that when not administered simultaneously, the agents will be administered within about one minute to within about eight hours and suitably within less than about one to about four hours. When administered contemporaneously, the agents are suitably administered at the same site on the subject.
  • the term “same site” includes the exact location, but can be within about 0.5 to about 15 cm, preferably from within about 0.5 to about 5 cm.
  • the term “separately” as used herein means that the agents are administered at an interval, for example at an interval of about a day to several weeks or months.
  • the active agents may be administered in either order.
  • agent includes a compound that induces a desired pharmacological and/or physiological effect.
  • agent also encompasses pharmaceutically acceptable and pharmacologically active ingredients of those compounds specifically mentioned herein including but not limited to salts, esters, amides, prodrugs, active metabolites, analogues and the like.
  • agent perse as well as pharmaceutically acceptable, pharmacologically active salts, esters, amides, prodrugs, metabolites, analogues, etc.
  • agent is not to be construed narrowly but extends to small molecules, proteinaceous molecules such as peptides, polypeptides and proteins as well as compositions comprising them and genetic molecules such as RNA, DNA and mimetics and chemical analogues thereof as well as cellular agents.
  • agent includes a cell that is capable of producing and secreting a polypeptide referred to herein as well as a polynucleotide comprising a nucleotide sequence that encodes that polypeptide.
  • agent extends to nucleic acid constructs including vectors such as viral or non-viral vectors, expression vectors and plasmids for expression in and secretion in a range of cells.
  • the “amount” or “level” of a biomarker is a detectable level in a sample. These can be measured by methods known to one skilled in the art and also disclosed herein. The expression level or amount of biomarker assessed can be used to determine the response to treatment.
  • antagonist refers to a substance that prevents, blocks, inhibits, neutralizes, or reduces a biological activity or effect of another molecule, such as a receptor.
  • binding refers to measurable and reproducible interactions such as binding between a target and a binding molecule, which is determinative of the presence of the target in the presence of a heterogeneous population of molecules including biological molecules.
  • binding molecule that binds to or specifically binds to a target is a molecule that binds this target with greater affinity, avidity, more readily, and/or with greater duration than it binds to other targets.
  • the extent of binding of a binding molecule to an unrelated target is less than about 10% of the binding of the molecule to the target as measured, e.g., by a radioimmunoassay (RIA).
  • a binding molecule that specifically binds to a target has a dissociation constant (Kd) of ⁇ 1 mM, ⁇ 100 nM, ⁇ 10 nM, ⁇ 1 nM, or ⁇ 0.1 nM.
  • Kd dissociation constant
  • a binding molecule specifically binds to a region on a protein that is conserved among the protein from different species.
  • specific binding can include, but does not require exclusive binding.
  • binding agent refers to an agent that binds to a target antigen and does not significantly bind to unrelated compounds.
  • binding agents that can be effectively employed in the disclosed methods include, but are not limited to, lectins, proteins, and antibodies, such as monoclonal antibodies, chimeric antibodies, or polyclonal antibodies, or antigen binding fragments thereof, as well as aptamers, Fc domain fusion proteins, and aptamers having or fused to hydrophobic protein domain, e.g., Fc domain, etc.
  • the binding agent is an exogenous antibody.
  • An exogenous antibody is an antibody not naturally produced in a mammal, e.g. in a human, by the mammalian immune system.
  • the term “complex” refers to an assemblage or aggregate of molecules ⁇ e.g., peptides, polypeptides, etc.) in direct or indirect contact with one another.
  • “contact”, or more particularly, “direct contact” means two or more molecules are close enough so that attractive noncovalent interactions, such as Van der Waal forces, hydrogen bonding, ionic and hydrophobic interactions, and the like, dominate the interaction of the molecules.
  • a complex of molecules ⁇ e.g., a peptide and polypeptide) is formed under conditions such that the complex is thermodynamically favoured ⁇ e.g., compared to a non-aggregated, or non-complexed, state of its component molecules).
  • polypeptide complex or “protein complex,” as used herein, refers to a trimer, tetramer, pentamer, hexamer, heptamer, octamer, nonamer, decamer, undecamer, dodecamer, or higher order oligomer.
  • amino acid sequence that displays substantial sequence similarity or identity to a reference amino acid sequence.
  • amino acid sequence will display at least about 70, 71 , 72, 73, 74, 75, 76, 77, 78, 79, 80, 81 , 82, 83, 84, 85, 86, 97, 88, 89, 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99% or even up to 100% sequence similarity or identity to at least a portion of the reference amino acid sequence.
  • an “effective amount” is at least the minimum amount required to effect a measurable improvement or prevention of a particular disorder.
  • An effective amount herein may vary according to factors such as the disease state, age, sex, and weight of the patient, and the ability of the antibody to elicit a desired response in the individual.
  • An effective amount is also one in which any toxic or detrimental effects of the treatment are outweighed by the therapeutically beneficial effects.
  • beneficial or desired results include results such as eliminating or reducing the risk, lessening the severity, or delaying the onset of the disease, including biochemical, histological and/or behavioural symptoms of the disease, its complications and intermediate pathological phenotypes presenting during development of the disease.
  • beneficial or desired results include clinical results such as decreasing one or more symptoms resulting from the disease, increasing the quality of life of those suffering from the disease, decreasing the dose of other medications required to treat the disease, enhancing effect of another medication such as via targeting, delaying the progression of the disease, and/or prolonging survival.
  • an effective amount of the drug may have the effect in reducing pathogen (bacterium, virus, etc.) titres in the circulation or tissue; reducing the number of pathogen infected cells; inhibiting (/.
  • an effective amount can be administered in one or more administrations.
  • an effective amount of drug, compound, or pharmaceutical composition is an amount sufficient to accomplish prophylactic or therapeutic treatment either directly or indirectly.
  • an effective amount of a drug, compound, or pharmaceutical composition may or may not be achieved in conjunction with another drug, compound, or pharmaceutical composition.
  • an “effective amount” may be considered in the context of administering one or more therapeutic agents, and a single agent may be considered to be given in an effective amount if, in conjunction with one or more other agents, a desirable result may be or is achieved.
  • an “effective response” of a patient or a patient’s “responsiveness” to treatment with a medicament and similar wording refers to the clinical or therapeutic benefit imparted to a patient at risk for, or suffering from, a disease or disorder, such as a pathogenic infection.
  • a disease or disorder such as a pathogenic infection.
  • such benefit includes any one or more of: extending survival (including overall survival and progression free survival); resulting in an objective response (including a complete response or a partial response); or improving signs or symptoms of the pathogenic infection.
  • a patient who “does not have an effective response” to treatment refers to a patient who does not have any one of extending survival (including overall survival and progression free survival); resulting in an objective response (including a complete response or a partial response); or improving signs or symptoms of the pathogenic infection.
  • RNA transcript e.g ., mRNA, antisense RNA, siRNA, shRNA, miRNA, etc.
  • expression of a coding sequence results from transcription and translation of the coding sequence.
  • expression of a non-coding sequence results from the transcription of the non-coding sequence.
  • level of expression or “expression level” in general are used interchangeably and generally refer to the amount of a biomarker in a sample. “Expression” generally refers to the process by which information (e.g ., gene- encoded and/or epigenetic) is converted into the structures present and operating in the cell. Therefore, as used herein, “expression” may refer to transcription into a polynucleotide, translation into a polypeptide, or even polynucleotide and/or polypeptide modifications ⁇ e.g., posttranslational modification of a polypeptide).
  • Fragments of the transcribed polynucleotide, the translated polypeptide, or polynucleotide and/or polypeptide modifications ⁇ e.g., post-translational modification of a polypeptide) shall also be regarded as expressed whether they originate from a transcript generated by alternative splicing or a degraded transcript, or from a post translational processing of the polypeptide, e.g., by proteolysis.
  • “Expressed genes” include those that are transcribed into a polynucleotide as mRNA and then translated into a polypeptide, and also those that are transcribed into RNA but not translated into a polypeptide ⁇ e.g., transfer and ribosomal RNAs).
  • “Elevated expression”, “elevated expression levels”, or “elevated levels” refers to an increased expression or increased levels of a biomarker in an individual or part of an individual ⁇ e.g., a cell, tissue or organ) relative to a control, such as an individual or individuals who are not suffering from the disease or disorder ⁇ e.g., T cell dysfunctional disorder) or parts thereof ⁇ e.g., a cell, tissue or organ) or an internal control ⁇ e.g., housekeeping biomarker).
  • Reduced expression refers to a decreased expression or decreased levels of a biomarker in an individual or part of an individual ⁇ e.g., a cell, tissue or organ) relative to a control, such as an individual or individuals who are not suffering from the disease or disorder ⁇ e.g., T-cell dysfunctional disorder) or parts thereof ⁇ e.g., a cell, tissue or organ) or an internal control ⁇ e.g., housekeeping biomarker).
  • reduced expression is little or no expression.
  • infection refers to invasion of body tissues by disease- causing microorganisms, their multiplication and the reaction of body tissues to these microorganisms and the toxins they produce. “Infection” includes but are not limited to infections by viruses, prions, bacteria, viroids, parasites, protozoans and fungi. In the context of the present invention, however, “infection” generally refers to virus infection of the family Coronnavitidae ( e.g ., coronaviruses);
  • “instructional material” includes a publication, a recording, a diagram, or any other medium of expression which can be used to communicate the usefulness of the compositions and methods of the invention.
  • the instructional material of the kit of the invention may, for example, be affixed to a container which contains the therapeutic or diagnostic agents of the invention or be shipped together with a container which contains the therapeutic or diagnostic agents of the invention.
  • LSD1 refers to any form of LSD1 and variants thereof that retain at least part of the activity of LSD1. Unless indicated differently, such as by specific reference to human LSD1 , LSD1 includes all mammalian species of native sequence LSD1, e.g., human, canine, feline, equine, and bovine. One exemplary human LSD1 is found as UniProt Accession Number 060341.
  • LSD2 refers to any form of LSD2 and variants thereof that retain at least part of the activity of LSD2. Unless indicated differently, such as by specific reference to human LSD2, LSD2 includes all mammalian species of native sequence LSD2, e.g., human, canine, feline, equine, and bovine. One exemplary human wild-type LSD2 peptide sequence is deposited as UniProt Accession Number Q8NB78.
  • LSD inhibitor refers to a molecule that decreases, blocks, inhibits, abrogates or interferes with the epigenetic function of an LSD (e.g., LSD1 or LSD2).
  • the LSD inhibitor is a direct inhibitor of LSD.
  • the LSD inhibitor is a molecule that inhibits the binding of LSD to one or more of its binding partners.
  • the LSD inhibitor inhibits the binding of the LSD binding partner PKC0 to LSD.
  • the LSD inhibitor is an indirect inhibitor of LSD.
  • the LSD inhibitors contemplated include molecules that bind specifically to PKC0, and thus decrease, block, inhibit, abrogate or interfere with the phosphorylation (and therefore subsequent activation) of LSD.
  • patient refers to any subject, particularly a vertebrate subject, and even more particularly a mammalian subject, for whom therapy or prophylaxis is desired.
  • Suitable vertebrate animals that fall within the scope of the invention include, but are not restricted to, any member of the subphylum Chordata including primates (e.g ., humans, monkeys and apes, and includes species of monkeys such from the genus Macaca ⁇ e.g., cynomologus monkeys such as Macaca fascicularis, and/or rhesus monkeys (Macaca mulatta)) and baboon (Papio ursinus), as well as marmosets (species from the genus Callithrix), squirrel monkeys (species from the genus Saimiri) and tamarins (species from the genus Saguinus), as well as species of apes such as chimpanzees (Pan troglodytes)), rodents ⁇ e.g., mice rats, guinea pigs), lagomorphs ⁇ e.g., rabbits, hares), bovines ⁇ e.
  • composition or “pharmaceutical formulation” refers to a preparation which is in such form as to permit the biological activity of the active ingredient(s) to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the composition or formulation would be administered. Such formulations are sterile. “Pharmaceutically acceptable” excipients (vehicles, additives) are those which can reasonably be administered to a subject mammal to provide an effective dose of the active ingredient employed.
  • the terms “prevent”, “prevented”, or “preventing”, refer to a prophylactic treatment which increases the resistance of a subject to developing the disease or condition or, in other words, decreases the likelihood that the subject will develop the disease or condition as well as a treatment after the disease or condition has begun in order to reduce or eliminate it altogether or prevent it from becoming worse. These terms also include within their scope preventing the disease or condition from occurring in a subject which may be predisposed to the disease or condition but has not yet been diagnosed as having it.
  • selective refers to compounds that inhibit or display antagonism towards a LSD without displaying substantial inhibition or antagonism towards another LSD or another enzyme such as a monoamine oxidase (MAO) (e.g ., MAO A or MAO B). Accordingly, a compound that is selective for LSD1 exhibits a LSD1 selectivity of greater than about 2-fold, 5-fold, 10-fold, 20-fold, 50-fold or greater than about 100-fold with respect to inhibition or antagonism of another LSD (i.e., a LSD other than LSD1 such as LSD2) or of another enzyme ⁇ e.g., a MAO).
  • a LSD other than LSD1 such as LSD2
  • another enzyme ⁇ e.g., a MAO
  • selective compounds display at least 50-fold greater inhibition or antagonism towards a specified LSD than towards another LSD or another enzyme ⁇ e.g., a MAO). In still other embodiments, selective compounds inhibit or display at least 100-fold greater inhibition or antagonism towards a specified LSD than towards another LSD or another enzyme ⁇ e.g., a MAO). In still other embodiments, selective compounds display at least 500-fold greater inhibition or antagonism towards a specified LSD than towards another LSD or another enzyme ⁇ e.g., a MAO). In still other embodiments, selective compounds display at least 1000- fold greater inhibition or antagonism towards a specified LSD than towards another LSD or another enzyme ⁇ e.g., a MAO).
  • sequence identity refers to the extent that sequences are identical on a nucleotide-by-nucleotide basis or an amino acid-by- amino acid basis over a window of comparison.
  • a “percentage of sequence identity” is calculated by comparing two optimally aligned sequences over the window of comparison, determining the number of positions at which the identical nucleic acid base ⁇ e.g., A, T, C, G, I) or the identical amino acid residue ⁇ e.g., Ala, Pro, Ser, Thr, Gly, Val, Leu, lie, Phe, Tyr, Trp, Lys, Arg, His, Asp, Glu, Asn, Gin, Cys, and Met) occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window of comparison ⁇ i.e., the window size), and multiplying the result by 100 to yield the percentage of sequence identity.
  • sequence identity will be understood to mean the “match percentage” calculated by an appropriate method.
  • sequence identity analysis may be carried out using the DNASIS computer program (Version 2.5 for windows; available from Hitachi Software engineering Co., Ltd., South San Francisco, California, USA) using standard defaults as used in the reference manual accompanying the software.
  • small molecule refers to a compound that has a molecular weight of less than 3 kilodalton (kDa), and typically less than 1.5 kDa, and more preferably less than about 1 kDa.
  • Small molecules may be nucleic acids, peptides, polypeptides, peptidomimetics, carbohydrates, lipids or other organic (carbon-containing) or inorganic molecules.
  • kDa kilodalton
  • small molecules may be nucleic acids, peptides, polypeptides, peptidomimetics, carbohydrates, lipids or other organic (carbon-containing) or inorganic molecules.
  • extensive libraries of chemical and/or biological mixtures often fungal, bacterial, or algal extracts, may be screened with any of the assays of the invention to identify compounds that modulate a bioactivity.
  • a “small organic molecule” is an organic compound (or organic compound complexed with an inorganic compound (e.g ., metal)) that has a molecular weight of less than 3 kDa, less than 1.5 kDa, or even less than about 1 kDa.
  • “Stringency” of hybridization reactions is readily determinable by one of ordinary skill in the art, and generally is an empirical calculation dependent upon probe length, washing temperature, and salt concentration. In general, longer probes require higher temperatures for proper annealing, while shorter probes need lower temperatures. Hybridization generally depends on the ability of denatured DNA to reanneal when complementary strands are present in an environment below their melting temperature. The higher the degree of desired homology between the probe and hybridisable sequence, the higher the relative temperature which can be used. As a result, it follows that higher relative temperatures would tend to make the reaction conditions more stringent, while lower temperatures less so. For additional details and explanation of stringency of hybridization reactions, see Ausubel etal., Current Protocols in Molecular Biology, Wiley Interscience Publishers, (1995).
  • “Stringent conditions” or “high stringency conditions”, as defined herein, can be identified by those that: (1) employ low ionic strength and high temperature for washing, for example 15 mM sodium chloride/1.5 mM sodium citrate/0.1 % sodium dodecyl sulphate at 50 Q C; (2) employ during hybridization a denaturing agent, such as formamide, for example, 50% (v/v) formamide with 0.1% bovine serum albumin/0.1% Ficoll/0.1% polyvinylpyrrolidone/50 mM sodium phosphate buffer at pH 6.5 with 750 mM sodium chloride, 75 mM sodium citrate at 42 Q C; or (3) overnight hybridization in a solution that employs 50% formamide, 5 x SSC (0.75 M NaCI, 75 mM sodium citrate), 50 mM sodium phosphate (pH 6.8), 0.1% sodium pyrophosphate, 5 x Denhardt's solution, sonicated salmon sperm DNA (50 pg/mL),
  • the term “synergistic” means that the therapeutic effect of a LSD inhibitor (e.g ., a LSD1 inhibitor or a LSD2 inhibitor) when administered in combination with an antiviral agent (or vice-versa) is greater than the predicted additive therapeutic effects of the LSD inhibitor and the antiviral agent when administered alone.
  • a LSD inhibitor e.g ., a LSD1 inhibitor or a LSD2 inhibitor
  • compositions generally a pharmaceutical formulation
  • an antiviral agent refers to the amount of each component in a composition (generally a pharmaceutical formulation), which is effective for enhancing immune effector function including any one or more of increased recognition of an antigen or an antigen peptide derived from an antigen in the context of MHC class II molecules by T cell receptors, increased release of cytokines and/or the activation of CD8 + lymphocytes (CTLs) and/or B cells, increased recognition of an antigen or an antigen peptide derived from an antigen in the context of MHC class I molecules by T cell receptors, increased elimination of cells presented in the context of MHC class I molecules, i.e., cells characterized by presentation of an antigen with class I MHC, for example, via apoptosis or perforin- mediated cell lysis, increased production of cytokines such as IL-2, IFN-y and TNF-a, and increased specific cytolytic killing of antigen expressing target cells
  • CTLs CD8 + lymphocytes
  • the dose response curve used to determine synergy in the art is described for example by Sande etal., (see, p. 1080-1105 in A. Goodman, The Pharmacological Basis of Therapeutics, MacMillan Publishing Co., Inc., New York (1980)).
  • the optimum synergistic amounts can be determined, using a 95% confidence limit, by varying factors such as dose level, schedule and response, and using a computer-generated model that generates isobolograms from the dose response curves for various combinations of the LSD inhibitor and the antiviral agent.
  • the highest enhancement of immune effector function on the dose response curve correlates with the optimum dosage levels.
  • treatment refers to clinical intervention designed to alter the natural course of the individual or cell being treated during the course of clinical pathology. Desirable effects of treatment include decreasing the rate of disease progression, ameliorating or palliating the disease state, and remission or improved prognosis.
  • an individual is successfully “treated” if one or more symptoms associated with a T cell dysfunctional disorder are mitigated or eliminated, including, but are not limited to, reducing the proliferation of (or destroying) cancerous cells, reducing pathogen infection, decreasing symptoms resulting from the disease, increasing the quality of life of those suffering from the disease, decreasing the dose of other medications required to treat the disease, and/or prolonging survival of individuals.
  • underscoring or italicizing the name of a gene shall indicate the gene, in contrast to its protein product, which is indicated by the name of the gene in the absence of any underscoring or italicizing.
  • LSD1 shall mean the LSD1 gene
  • LSD1 shall indicate the protein product or products generated from transcription and translation and/or alternative splicing of the LSD1 gene.
  • coronaviruses display spike (S) protein trimers on their cell surface, and these glycosylated proteins accommodate binding to a host cell surface receptor. Upon receptor binding, the viral membrane fuses with that of the host cell, allowing the viral RNA entry into the host cell.
  • S spike
  • the coronavirus S protein mediates the first essential step in coronavirus infection, i.e., viral entry into host cells.
  • the S protein contains an N-terminal signal peptide which primes the nascent polypeptide for import into the endoplasmic reticulum (ER).
  • ER endoplasmic reticulum
  • the S protein is extensively modified with N-linked glycans, which are thought to provide protection against the neutralising antibodies of the host.
  • the S proteins which are present as trimers on the virus surface, combine two biological functions. First, its surface unit, Si, binds to a specific receptor located at the surface of host cells (e.g., the ACE protein receptor) and thereby determines cellular tropism and, as a consequence, viral pathogenesis. Second, the transmembrane unit, S2, mediates fusion between the viral envelope and a target cell membrane. Priming (the proteolytic separation of the Si and S2 subunits) provides the coronavirus S protein with the structural flexibility required for the subsequent membrane fusion reaction.
  • Si its surface unit
  • S2 mediates fusion between the viral envelope and a target cell membrane. Priming (the proteolytic separation of the Si and S2 subunits) provides the coronavirus S protein with the structural flexibility required for the subsequent membrane fusion reaction.
  • TMPRSS2 polypeptide has been demonstrated to cleave and activate S protein (Glowacha etal., 2011). Activation by the TMPRSS2 polypeptide is therefore believed to occur at the plasma membrane, likely after S protein binding to a virus cell entry receptor polypeptide (e.g., an ACE2 polypeptide). Notably, TMPRSS2 is reported to bind the virus cell entry polypeptide (e.g., ACE2, as described in Shulla etal., (2011)), and it is likely that the conformational changes in the S protein that are induced upon virus cell entry receptor polypeptide-binding expose the TMPRSS2 polypeptide cleavage site in the S protein.
  • virus cell entry polypeptide e.g., ACE2
  • the present inventors have determined that post-translational modifications play a significant role in the regulation of functional activity of the TMPRSS2 polypeptide. For example, a plurality of methylation sites have been identified in both (i) nuclear localisation signal domain of the TMPRSS2 polypeptide; and (ii) the catalytic domain of the TMPRSS2 polypeptide. Accordingly, administering an LSD1 inhibitor reduces the demethylation activity asserted on the TMPRSS2, causing a decrease in its functional activity.
  • an LSD inhibitor ⁇ e.g., a LSD1 inhibitor or a LSD2 inhibitor
  • a subject is administered to a subject to prevent or reduce the ability of the TMPRSS2 polypeptide from transporting the coronavirus into the host cell.
  • the TMPRSS2 polypeptide is a human TMPRSS2 polypeptide, with the amino acid sequence identified as UniProt Accession No. 015393, and set forth below: MALNSGSPPAIGPYYENHGYQPENPYPAQPTVVPTVYEVHPAQYYPSPVP
  • Lysine residues K340 and K362 of the TMPRSS2 amino acid sequence set forth above are identified as being key LSD1 -mediated methylation/demethylation residues. Accordingly, in some embodiments the present invention provides methods of antagonising TMPRSS2 polypeptide, the method comprising administering an inhibitor of LSD1 -mediated demethylation of one or both residues K340 and K362.
  • the present inventors have also determined that post-translational modifications play a significant role in the regulation of functional activity of the viral cell entry receptor polypeptides (e.g ., an ACE2 polypeptide). For example, a plurality of methylation sites are identified in both (i) nuclear localisation signal domain of the viral cell entry receptor polypeptide; and (ii) the catalytic domain of the viral cell entry receptor polypeptide. Accordingly, administering an LSD1 inhibitor reduces the demethylation activity asserted on the viral cell entry receptor polypeptide, causing a decrease in its functional activity. Accordingly, in some embodiments, an LSD inhibitor ⁇ e.g., a LSD1 inhibitor) is administered to a subject to prevent or reduce the ability of the virus cell entry receptor polypeptide to transporting a coronavirus into the host cell.
  • an LSD inhibitor ⁇ e.g., a LSD1 inhibitor
  • the virus cell entry polypeptide is a human ACE2 polypeptide, with the amino acid sequence identified as UniProt Accession No. Q9BYF1 , and set forth below:
  • Lysine residues K26 and K353 of the ACE2 polypeptide amino acid sequence set forth above are identified as key LSD1 -mediated methylation/ demethylation residues. Accordingly, in some embodiments the present invention provides methods of antagonising an ACE2 polypeptide, the method comprising administering an inhibitor of LSD1 -mediated demethylation. In some embodiments, the LSD1 -mediated demethylation includes demethylation of residues K26 and/or K353 of the ACE2 polypeptide.
  • the present invention provides methods of antagonising an ACE2 polypeptide, the method comprising administering an inhibitor of LSD1 -mediated demethylation of one or both residues K340 and K362.
  • lysine residue K31 of the ACE2 polypeptide amino acid sequence set forth above is identified as key LSD1 -mediated methylation/demethylation residue for virus entry into the host cell. Accordingly, in some embodiments, the present invention provides methods of preventing entry of a coronavirus into a host cell, the method comprising administering an inhibitor of LSD1 -mediated demethylation of an ACE polypeptide (e.g., a human ACE polypeptide as set forth in UniProt Accession No. Q9BYF1).
  • an ACE polypeptide e.g., a human ACE polypeptide as set forth in UniProt Accession No. Q9BYF1
  • the LSD1 -mediated demethylation of the ACE polypeptide includes demethylation of residue K31 of the ACE2 polypeptide [0104]
  • the viral cell entry receptor polypeptide is selected from the group comprising an ACE2 polypeptide, a dipeptidyl peptidase 4 (DPP4) polypeptide, and a CD13 polypeptide.
  • the present invention extends to a method of inhibiting the entry of a coronavirus into a cell of the host, the method comprising administering to the subject an LSD1 inhibitor (e.g ., a selective LSD1 inhibitor).
  • an LSD1 inhibitor e.g ., a selective LSD1 inhibitor.
  • the coronavirus cell entry machinery is prevented from transporting the coronavirus into the cell.
  • post-translational modifications by way of phosphorylation plays a significant role in the regulation of functional activity of the TMPRSS2 polypeptide.
  • at least one PKC0 serine phosphorylation site has been identified in the catalytic domain of the TMPRSS2 polypeptide. Accordingly, administering a PKC0 inhibitor reduces the phosphorylation activity asserted on the TMPRSS2, causing a decrease in its functional activity. Accordingly, in some embodiments, a PKC0 inhibitor is administered to a subject to prevent or reduce the ability of the TMPRSS2 polypeptide from transporting the coronavirus into the host cell.
  • Serine residue S441 of the TMPRSS2 amino acid sequence set forth above is a key PKC0-mediated phosphorylated residue. Accordingly, in some embodiments the present invention provides methods of antagonising TMPRSS2 polypeptide, the method comprising administering an inhibitor of the PKC0-mediated phosphorylation of one or both residues S441.
  • phosphorylation post-translational modifications play a significant role in the regulation of functional activity of the viral cell entry receptor polypeptides ⁇ e.g., an ACE2 polypeptide).
  • a plurality of PKC0-mediated phosphorylation sites are present in both (i) the nuclear localisation signal domain of the viral cell entry receptor polypeptide; and (ii) the catalytic domain of the viral cell entry receptor polypeptide.
  • administering a PKC0 inhibitor reduces the phosphorylation activity asserted on the viral cell entry receptor polypeptide, causing a decrease in its functional activity.
  • a PKC0 inhibitor is administered to a subject to prevent or reduce the ability of the virus cell entry receptor polypeptide to transporting a coronavirus into the host cell.
  • Residues S47, S109, S254 and T763 of the ACE2 polypeptide amino acid sequence set forth above are identified as key PKC0-mediated phosphorylation sites. Accordingly, in some embodiments the present invention provides methods of antagonising an ACE2 polypeptide, the method comprising administering an inhibitor of PKC0-mediated phosphorylation of one or more residues S47, S109, S254 and T763 of the ACE2 polypeptide.
  • post-translational modifications by way of phosphorylation plays a significant role in the regulation of functional activity of the viral cell entry receptor polypeptides (e.g ., an ACE2 polypeptide).
  • a plurality of methylation sites are identified in both (i) nuclear localisation signal domain of the viral cell entry receptor polypeptide; and (ii) the catalytic domain of the viral cell entry receptor polypeptide. Accordingly, administering an LSD1 inhibitor reduces the demethylation activity asserted on the viral cell entry receptor polypeptide, causing a decrease in its functional activity.
  • an LSD inhibitor ⁇ e.g., a LSD1 inhibitor or a LSD2 inhibitor
  • a subject is administered to a subject to prevent or reduce the ability of the virus cell entry receptor polypeptide to transporting a coronavirus into the host cell.
  • the coronavirus is a SARS-CoV-2.
  • the present invention is based in part on the determination that coronavirus infections take advantage of host machinery to enter the host cell, and that these essential host machinery are regulated at the post-translational level and the transcriptional level by methylation, by LSD1.
  • LSDs including LSD1 , play a critical role in two levels: (1) the post-transcriptional methylation of the chromatin domains across the regulatory regions of the cell entry machinery and that prevention of the demethylation by LSD1 prevents the ability of the cell to express the polypeptide expression products of the TMPRSS2 gene and/or the virus cell entry receptor gene; and (2) the post-translational methylation of the active (e.g., catalytic) domains of the TMPRSS2 polypeptide and/or the viral host cell entry polypeptide (e.g., an ACE2 polypeptide).
  • active e.g., catalytic
  • LSD1 inhibitor e.g., a selective LSD1 inhibitor
  • the LSD1 inhibitor is used in combination with an additional antiviral agent.
  • the methods and compositions of the present invention are thus particularly useful in the treatment or prophylaxis of a coronavirus infection ⁇ e.g., a coronavirus infection), as described hereafter.
  • the LSD inhibitor includes and encompasses any active agent that reduces the accumulation, function or stability of a LSD; or decrease expression of a LSD gene, and such inhibitors include without limitation, small molecules and macromolecules such as nucleic acids, peptides, polypeptides, peptidomimetics, carbohydrates, polysaccharides, lipopolysaccharides, lipids or other organic (carbon containing) or inorganic molecules.
  • the LSD inhibitor is an antagonistic nucleic acid molecule that functions to inhibit the transcription or translation of LSD (e.g., LSD1 or LSD2) transcripts.
  • LSD LSD1 or LSD2
  • Representative transcripts of this type include nucleotide sequences corresponding to any one the following sequences: (1) human LSD1 nucleotide sequences as set forth for example in GenBank Accession Nos.
  • NP_694587.3 nucleotide sequences that encode an amino acid sequence that shares at least 70, 71 , 72, 73, 74, 75, 76, 77, 78, 79, 80, 81 , 82, 83,
  • nucleotide sequences that encode an amino acid sequence that shares at least 70, 71 , 72, 73, 74, 75, 76, 77,
  • Illustrative antagonist nucleic acid molecules include antisense molecules, aptamers, ribozymes and triplex forming molecules, RNAi and external guide sequences.
  • the nucleic acid molecules can act as effectors, inhibitors, modulators, and stimulators of a specific activity possessed by a target molecule, or the functional nucleic acid molecules can possess a de novo activity independent of any other molecules.
  • Antagonist nucleic acid molecules can interact with any macromolecule, such as DNA, RNA, polypeptides, or carbohydrate chains.
  • antagonist nucleic acid molecules can interact with LSD (e.g., LSD1) mRNA or the genomic DNA of LSD (e.g., LSD1) or they can interact with a LSD polypeptide (e.g., LSD1).
  • LSD LSD1
  • LSD polypeptide e.g., LSD1
  • antagonist nucleic acid molecules are designed to interact with other nucleic acids based on sequence homology between the target molecule and the antagonist nucleic acid molecule.
  • the specific recognition between the antagonist nucleic acid molecule and the target molecule is not based on sequence homology between the antagonist nucleic acid molecule and the target molecule, but rather is based on the formation of tertiary structure that allows specific recognition to take place.
  • anti-sense RNA or DNA molecules are used to directly block the translation of LSD (e.g., LSD1) by binding to targeted mRNA and preventing protein translation.
  • Antisense molecules are designed to interact with a target nucleic acid molecule through either canonical or non-canonical base pairing. The interaction of the antisense molecule and the target molecule may be designed to promote the destruction of the target molecule through, for example, RNAseH mediated RNA-DNA hybrid degradation. Alternatively, the antisense molecule may be designed to interrupt a processing function that normally would take place on the target molecule, such as transcription or replication. Antisense molecules can be designed based on the sequence of the target molecule.
  • Non-limiting methods include in vitro selection experiments and DNA modification studies using DMS and DEPC.
  • the antisense molecules bind the target molecule with a dissociation constant (Kd) less than or equal to 10 6 , 10 8 , 10 10 , or 10 12 .
  • Kd dissociation constant
  • antisense oligodeoxyribonucleotides derived from the translation initiation site e.g., between - 10 and +10 regions are employed.
  • Aptamers are molecules that interact with a target molecule, suitably in a specific way.
  • Aptamers are generally small nucleic acids ranging from 15-50 bases in length that fold into defined secondary and tertiary structures, such as stem-loops or G-quartets.
  • Aptamers can bind small molecules, such as ATP and theophiline, as well as large molecules, such as reverse transcriptase and thrombin.
  • Aptamers can bind very tightly with KdS from the target molecule of less than 10 12 M.
  • the aptamers bind the target molecule with a Kd less than 10 6 , 10 8 , 10 10 , or 10 12
  • Aptamers can bind the target molecule with a very high degree of specificity.
  • aptamers have been isolated that have greater than a 10,000-fold difference in binding affinities between the target molecule and another molecule that differ at only a single position on the molecule. It is desirable that an aptamer have a Kd with the target molecule at least 10-, 100-, 1000-, 10,000-, or 100,000-fold lower than the Kd with a background-binding molecule.
  • a suitable method for generating an aptamer to a target of interest is the “Systematic Evolution of Ligands by Exponential Enrichment” (SELEXTM).
  • SELEXTM Systematic Evolution of Ligands by Exponential Enrichment
  • the SELEXTM method is described in U.S. Pat. Nos. 5,475,096 and 5,270,163 (see also WO 91/19813). Briefly, a mixture of nucleic acids is contacted with the target molecule under conditions favourable for binding. The unbound nucleic acids are partitioned from the bound nucleic acids, and the nucleic acid -target complexes are dissociated.
  • the dissociated nucleic acids are amplified to yield a ligand-enriched mixture of nucleic acids, which is subjected to repeated cycles of binding, partitioning, dissociating and amplifying as desired to yield highly specific high affinity nucleic acid ligands to the target molecule.
  • anti-LSD1 ribozymes are used for catalysing the specific cleavage of LSD1 RNA.
  • the mechanism of ribozyme action involves sequence specific hybridization of the ribozyme molecule to complementary target RNA, followed by an endonucleolytic cleavage.
  • ribozymes that catalyse nuclease or nucleic acid polymerase type reactions, which are based on ribozymes found in natural systems, such as hammerhead ribozymes, hairpin ribozymes, and tetrahymena ribozymes.
  • ribozymes that are not found in natural systems, but which have been engineered to catalyse specific reactions de novo.
  • Representative ribozymes cleave RNA or DNA substrates.
  • ribozymes that cleave RNA substrates are employed.
  • Specific ribozyme cleavage sites within potential RNA targets are initially identified by scanning the target molecule for ribozyme cleavage sites, which include the following sequences, GUA, GUU and GUC.
  • RNA sequences of between 15 and 20 ribonucleotides corresponding to the region of the target gene containing the cleavage site may be evaluated for predicted structural features such as secondary structure that may render the oligonucleotide sequence unsuitable.
  • the suitability of candidate targets may also be evaluated by testing their accessibility to hybridization with complementary oligonucleotides, using ribonuclease protection assays.
  • Triplex forming functional nucleic acid molecules are molecules that can interact with either double-stranded or single-stranded nucleic acid.
  • triplex molecules When triplex molecules interact with a target region, a structure called a triplex is formed, in which there are three strands of DNA forming a complex dependent on both Watson-Crick and Hoogsteen base pairing. Triplex molecules are preferred because they can bind target regions with high affinity and specificity. It is generally desirable that the triplex forming molecules bind the target molecule with a Kd less than 10 6 , 10 8 , 10 10 , or 10- 12 .
  • EGSs External guide sequences
  • RNAse P RNAse P
  • RNAse P RNAse P
  • tRNA transfer RNA
  • RNAse P Bacterial RNAse P can be recruited to cleave virtually any RNA sequence by using an EGS that causes the target RNA: EGS complex to mimic the natural tRNA substrate. Similarly, eukaryotic EGS/RNAse P-directed cleavage of RNA can be utilized to cleave desired targets within eukaryotic cells.
  • RNA molecules that mediate RNA interference (RNAi) of a LSD1 gene or LSD1 transcript can be used to reduce or abrogate gene expression.
  • RNAi refers to interference with or destruction of the product of a target gene by introducing a single-stranded or usually a double- stranded RNA (dsRNA) that is homologous to the transcript of a target gene.
  • dsRNAi methods including double-stranded RNA interference (dsRNAi) or small interfering RNA (siRNA), have been extensively documented in a number of organisms, including mammalian cells and the nematode C. elegans (Fire etal., 1998. Nature 391 , 806-811 ).
  • RNAi can be triggered by 21 - to 23-nucleotide (nt) duplexes of small interfering RNA (siRNA) (Chiu etal., 2002 Mol. Cell. 10: 549- 561 ; Elbashir et al., 2001. Nature 411 : 494-498), or by micro-RNAs (miRNA), functional small-hairpin RNA (shRNA), or other dsRNAs which are expressed in vivo using DNA templates with RNA polymerase III promoters (Zeng et al., 2002. Mol.
  • siRNA small interfering RNA
  • dsRNA per se and especially dsRNA- producing constructs corresponding to at least a portion of a LSD1 gene are used to reduce or abrogate its expression.
  • RNAi-mediated inhibition of gene expression may be accomplished using any of the techniques reported in the art, for instance by transfecting a nucleic acid construct encoding a stem-loop or hairpin RNA structure into the genome of the target cell, or by expressing a transfected nucleic acid construct having homology for a LSD1 gene from between convergent promoters, or as a head to head or tail to tail duplication from behind a single promoter.
  • Any similar construct may be used so long as it produces a single RNA having the ability to fold back on itself and produce a dsRNA, or so long as it produces two separate RNA transcripts, which then anneal to form a dsRNA having homology to a target gene.
  • RNAi-encoding nucleic acids can vary in the level of homology they contain toward the target gene transcript, i.e., with dsRNAs of 100 to 200 base pairs having at least about 85% homology with the target gene, and longer dsRNAs, i.e., 300 to 100 base pairs, having at least about 75% homology to the target gene.
  • RNA-encoding constructs that express a single RNA transcript designed to anneal to a separately expressed RNA, or single constructs expressing separate transcripts from convergent promoters are suitably at least about 100 nucleotides in length.
  • RNA-encoding constructs that express a single RNA designed to form a dsRNA via internal folding are usually at least about 200 nucleotides in length.
  • the promoter used to express the dsRNA-forming construct may be any type of promoter if the resulting dsRNA is specific for a gene product in the cell lineage targeted for destruction.
  • the promoter may be lineage specific in that it is only expressed in cells of a particular development lineage. This might be advantageous where some overlap in homology is observed with a gene that is expressed in a non-targeted cell lineage.
  • the promoter may also be inducible by externally controlled factors, or by intracellular environmental factors.
  • RNA molecules of about 21 to about 23 nucleotides which direct cleavage of specific mRNA to which they correspond, as for example described by in U.S. Pat. Pub. No. 2002/0086356, can be utilized for mediating RNAi.
  • Such 21- to 23-nt RNA molecules can comprise a 3' hydroxyl group, can be single-stranded or double stranded (as two 21 - to 23-nt RNAs) wherein the dsRNA molecules can be blunt ended or comprise overhanging ends ( e.g ., 5', 3').
  • the antagonist nucleic acid molecule is a siRNA.
  • siRNAs can be prepared by any suitable method. For example, reference may be made to International PCT Pat. Pub. No. WO 02/44321 , which discloses siRNAs capable of sequence-specific degradation of target mRNAs when base- paired with 3' overhanging ends, which is incorporated by reference herein. Sequence specific gene silencing can be achieved in mammalian cells using synthetic, short double-stranded RNAs that mimic the siRNAs produced by the enzyme dicer.
  • siRNA can be chemically or in v/ ro-synthesized or can be the result of short double-stranded hairpin-like RNAs (shRNAs) that are processed into siRNAs inside the cell.
  • Synthetic siRNAs are generally designed using algorithms and a conventional DNA/RNA synthesizer. Suppliers include Ambion (Austin, Tex.), ChemGenes (Ashland, Mass.), Dharmacon (Lafayette, Colo.), Glen Research (Sterl ing, Va.), MWB Biotech (Esbersberg, Germany), Proligo (Boulder, Colo.), and Qiagen (Vento, The Netherlands).
  • siRNA can also be synthesized in vitro using kits such as Ambion's SILENCERTM siRNA Construction Kit.
  • siRNAs short hairpin RNAs
  • Kits for the production of vectors comprising shRNA are available, such as, for example, Imgenex's GENESUPPRESSORTM Construction Kits and Invitrogen's BLOCK-ITTM inducible RNAi plasmid and lentivirus vectors.
  • methods for formulation and delivery of siRNAs to a subject are also well known in the art. See, e.g.,
  • RNAi molecules ⁇ e.g., LSD ⁇ e.g., LSD1 or LSD2) siRNA and shRNA
  • LSD Long RNA
  • shRNA siRNA
  • siRNA and shRNA are described in the art ⁇ e.g., Yang, etat, 2010. Proc. Natl. Acad. Sci. USA 107: 21499-21504 and He et al., 2012. Transcription 3: 1-16) or available commercially from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA, USA) and OriGene Technologies, Inc. (Rockville, MD, USA) .
  • the present invention further contemplates peptide or polypeptide- based inhibitor compounds.
  • BHC80 also known as PHD finger protein 21 A
  • the present invention further contemplates the use of BHC80 or biologically active fragments thereof for inhibiting LSD1 enzymatic activity.
  • Amino acid sequences of BHC80 polypeptides, and nucleotide sequences encoding BHC80 polypeptides, are publicly available. In this regard, reference may be made for example to (1) GenBank Accession No.
  • NP057705 for a Homo sapiens BHC80 amino acid sequence
  • GenBank NM016621 for a nucleotide sequence encoding the amino acid sequence set forth in GenBank Accession No. NP057705
  • GenBank Accession No. NP620094 for a Mus musculus BHC80 amino acid sequence
  • GenBank NM138755 for a nucleotide sequence encoding the amino acid sequence set forth in GenBank Accession No. NP620094
  • GenBank Accession No. NP00118576.1 for a Gallus gallus BHC80 amino acid sequence
  • GenBankNMOO1 199647 for a nucleotide sequence encoding the amino acid sequence set forth in GenBank Accession No. NP00118576.1
  • GenBank Accession No. DAA21793 for a Bos taurus BHC80 amino acid sequence.
  • Illustrative BHC80 polypeptides are selected from the group consisting of: (1) a polypeptide comprising an amino acid sequence that shares at least 70, 71 , 72, 73, 74, 75, 76, 77, 78, 79, 80, 81 , 82, 83, 84, 85, 86, 87, 88, 89, 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99% sequence similarity with the amino acid sequence listed in any one of the GenBank BHC80 polypeptide entries noted above; (2) a polypeptide comprising an amino acid sequence that shares at least 70, 71 , 72, 73, 74, 75, 76, 77, 78, 79, 80, 81 , 82, 83, 84, 85, 86, 87, 88, 89, 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99% sequence identity with
  • a BHC80 polypeptide can be introduced into a cell by delivering a polypeptide perse, or by introducing into the cell a BHC80 nucleic acid comprising a nucleotide sequence encoding a BHC80 polypeptide.
  • a BHC80 nucleic acid comprises a nucleotide sequence selected from: (1) a BHC80 nucleotide sequence listed in any one of the GenBank BHC80 polynucleotide entries noted above; (2) a nucleotide sequence that shares at least 70, 71 , 72, 73, 74, 75,
  • the BHC80 nucleic acid can be in the form of a recombinant expression vector.
  • the BHC80 nucleotide sequence can be operably linked to a transcriptional control element(s), e.g., a promoter, in the expression vector.
  • Suitable vectors include, e.g., recombinant retroviruses, lentiviruses, and adenoviruses; retroviral expression vectors, lentiviral expression vectors, nucleic acid expression vectors, and plasmid expression vectors.
  • the expression vector is integrated into the genome of a cell. In other cases, the expression vector persists in an episomal state in a cell.
  • Suitable expression vectors include, but are not limited to, viral vectors ⁇ e.g., viral vectors based on vaccinia virus; poliovirus; adenovirus (see, e.g.,
  • WO 93/03769 WO 93/19191 ; WO 94/28938; WO 95/11984 and WO 95/00655); adeno-associated virus (see, e.g., AN etal., Hum Gene Ther 9 :8186, 1998,
  • SV40 herpes simplex virus
  • human immunodeficiency virus see, e.g., Miyoshi etal., PNAS 94: 1031923, 1997; Takahashi etal., J Virol 73: 78127816, 1999
  • a retroviral vector ⁇ e.g., Murine Leukemia Virus, spleen necrosis virus, and vectors derived from retroviruses such as Rous Sarcoma Virus, Harvey Sarcoma Virus, avian leukosis virus, a lentivirus, human immunodeficiency virus, myeloproliferative sarcoma virus, and mammary tumour virus
  • retroviral vector ⁇ e.g., Murine Leukemia Virus, spleen necrosis virus, and vectors derived from retroviruses such as Rous Sarcoma Virus, Harvey Sarcoma Virus, avian leukosis virus, a lentivirus, human immunodeficiency virus, mye
  • Small molecule agents that reduce enzymatic activity of LSD1 that are suitable for use in the present invention include monoamine oxidase (MAO) inhibitors that also inhibit LSD1 enzymatic activity; polyamine compounds that inhibit LSD1 enzymatic activity; phenylcyclopropylamine derivatives that inhibit LSD1 enzymatic activity; and the like.
  • MAO monoamine oxidase
  • MAO inhibitors include MAO-A-selective inhibitors, MAO-B-selective inhibitors, and MAO non-selective inhibitors.
  • Illustrative examples of MAO inhibitors include reported inhibitors of the MAO-A isoform, which preferentially deaminates 5-hydroxytryptamine (serotonin) (5-HT) and norepinephrine (NE), and/or the MAO-B isoform, which preferentially deaminates phenylethylamine (PEA) and benzylamine (both MAO-A and MAO-B metabolize Dopamine (DA)).
  • MAO inhibitors may be irreversible or reversible ⁇ e.g., reversible inhibitors of MAO-A (RIMA)), and may have varying potencies against MAO-A and/or MAO-B ⁇ e.g., non-selective dual inhibitors or isoform-selective inhibitors).
  • RIMA reversible inhibitors of MAO-A
  • the MAO inhibitors are selected from : clorgyline; L-deprenyl; isocarboxazid (MARPLANTM); ayahuasca; nialamide; iproniazide; iproclozide; moclobemide (AURORIXTM; 4-chloro-N-(2-morpholin-4- ylethyl)benzamide); phenelzine (NARDILTM; ( ⁇ )-2-phenylethylhydrazine); tranylcypromine (PARNATETM; ( ⁇ )-trans-2-phenylcyclopropan-l-amine) (the congeneric of phenelzine); bizine (described in Prusevich et al., ACS Chem.
  • selegiline hydrochloride L-deprenyl, ELDEPRYLTM, ZELAPARTM
  • dimethylselegilene dimethylselegilene
  • safinamide rasagiline
  • AZILECTTM bifemelane
  • desoxypeganine harmine (also known as telepathine or banasterine); linezolid (ZYVOXTM, ZYVOXIDTM); pargyiine (EUDATINTM, SUPIRDYLTM); dienolide kavapyrone desmethoxyyangonin; 5-(4- Arylmethoxyphenyl)-2-(2-cyanoethyl)tetrazoles; and the like.
  • Small molecule LSD1 inhibitors may also be selected from polyamine compounds as described for example in United States Publication No.
  • Illustrative polyamine inhibitors of LSD1 include compounds according to formula (I):
  • n is an integer from 1 to 12; m and p are independently an integer from 1 to 5; q is 0 or 1 ; each Ri is independently selected from the group consisting of C-i-Cs alkyl, C4-C15 cycloalkyl, C3-C15 branched alkyl, C6-C20 aryl, C6-C20 heteroaryl, C7-C24 aralkyl, C7-C24 heteroaralkyl, and
  • R3 is selected from the group consisting of C-i-Cs alkyl, C4-C15 cycloalkyl, C3-C15 branched alkyl, C6-C20 aryl, C6-C20 heteroaryl, C7-C24 aralkyl and C7-C24 heteroaralkyl;
  • each R2 is independently selected from hydrogen or a C-i-Cs alkyl.
  • a suitable polyamine compound is a compound of Formula (I), wherein one or both Ri is a C6-C20 aryl, such as a single ring aryl, including without limitation, a phenyl.
  • the compound is of the formula (I) and each Ri is phenyl.
  • q is 1 , m and p are 3, and n is 4. In another embodiment, q is 1 , m and p are 3, and n is 7.
  • a suitable polyamine compound is a compound of Formula (I), where at least one or both Ri is a C8-C12 or a C-i-Cs alkyl, such as a linear alkyl.
  • One or both Ri may be a C-i-Cs linear alkyl, such as methyl or ethyl.
  • each Ri is methyl.
  • One or both Ri may comprise or be a C4-C15 cycloalkyl group, such as a cycloalkyl group containing a linear alkyl group, where the cycloalkyl group is connected to the molecule either via its alkyl or cycloalkyl moiety.
  • one or both Ri may be cyclopropylmethyl or cyclohexylmethyl.
  • one Ri is cyclopropylmethyl or cyclohexyl methyl and the other Ri is a linear alkyl group, such as a linear C-i-Cs unsubstituted alkyl group, including without limitation an ethyl group.
  • Ri is a C3-C15 branched alkyl group such as isopropyl.
  • the substituted alkyl may be substituted with any substituent, including a primary, secondary, tertiary or quaternary amine.
  • Ri is a C-i-Cs alkyl group substituted with an amine such that Ri may be e.g., alkyl-NH2 or an alkyl-amine-alkyl moiety such as -(CFl2)yNFI(CFl2)zCFl3 where y and z are independently an integer from 1 to 8. In one embodiment, Ri is -(CFl2)3NFl2.
  • the compound is of the formula (I) where one or both Ri is a C7-C24 substituted or unsubstituted aralkyl, which in one embodiment is an aralkyl connected to the molecule via its alkyl moiety ⁇ e.g., benzyl).
  • both Ri are aralkyl moieties wherein the alkyl portion of the moiety is substituted with two aryl groups and the moiety is connected to the molecule via its alkyl group.
  • one or both Ri is a C7-C24 aralkyl wherein the alkyl portion is substituted with two phenyl groups, such as when Ri is
  • both Ri of Formula (I) is
  • each Ri of Formula (I) is
  • n 1 , 2 or 5 and m and p are each 1.
  • At least one Ri is hydrogen.
  • the other Ri may be any moiety listed above for Ri, including an aryl group such as benzyl.
  • Any of the compounds of Formula (I) listed above include compounds where at least one or both of R2 is hydrogen or a C-i-Cs substituted or unsubstituted alkyl.
  • each R2 is an unsubstituted alkyl such as methyl.
  • each R2 is hydrogen.
  • Any of the compounds of Formula (I) listed above may be compounds where q is 1 and m and p are the same.
  • the polyaminoguanidines of Formula (I) may be symmetric with reference to the polyaminoguanidine core ⁇ e.g., excluding Ri).
  • the compounds of Formula (I) may be asymmetric, e.g., when q is 0.
  • m and p are 1 .
  • q is 0.
  • n is an integer from 1 to 5.
  • the compound is a polyaminobiguanide or N- alkylated polyaminobiguanide.
  • An N-alkylated polyaminobiguanide intends a polyaminobiguanide where at least one imine nitrogen of at least one biguanide is alkylated.
  • the compound is a polyaminobiguanide of the Formula (I), or a salt, solvate, or hydrate thereof, where q is 1 , and at least one or each R, is of the structure:
  • each R3 is independently selected from the group consisting of C1-C8 alkyl, C6-C20 aryl, C6-C20 heteroaryl, C7-C24 aralkyl, and C7-C24 heteroaralkyl; and each R2 is independently hydrogen or a C-i-Cs alkyl.
  • each R3 is a C-i-Cs alkyl.
  • the alkyl may be substituted with any substituent, including a primary, secondary, tertiary or quaternary amine.
  • R3 is a C-i-Cs alkyl group substituted with an amine such that R3 may be e.g., alkyl-NFte or an alkyl-amine-alkyl moiety such as -(CH2) y NH(CH2)zCH3 where y and z are independently an integer from 1 to 8.
  • R3 is -(CH2)3NH2.
  • R3 may also be a C4-C15 cycloalkyl or a C3-C15 branched alkyl.
  • at least one or each R3 is a C6-C20 aryl.
  • q is 1 , m and p are 3, and n is 4.
  • q is 1 , m and p are 3, and n is 7.
  • the compound is a polyaminobiguanide of Formula (I) where at least one R3 is a C7-C24 aralkyl, which in one embodiment is an aralkyl connected to the molecule via its alkyl moiety.
  • each R3 is an aralkyl moiety where the alkyl portion of the moiety is substituted with one or two aryl groups and the moiety is connected to the molecule via its alkyl moiety.
  • at least one or each R3 is an aralkyl where the alkyl portion is substituted with two phenyl or benzyl groups, such as when R3 is 2,2- diphenylethyl or 2,2-dibenzylethyl.
  • each R3 is 2,2- diphenylethyl and n is 1 , 2 or 5.
  • each R3 is 2,2-diphenylethyl and n is 1 , 2 or 5 and m and p are each 1.
  • any of the polyaminobiguanide compounds of Formula (I) listed above include compounds where at least one or both of R2 is hydrogen or a C-i-Cs alkyl.
  • each R2 is an unsubstituted alkyl, such as methyl.
  • each R2 is a hydrogen.
  • any of the polyaminobiguanide compounds of formula (I) listed above include compounds where q is 1 and m and p are the same. Accordingly, the polyaminobiguanides of Formula (I) may be symmetric with reference to the polyaminobiguanide core. Alternatively, the compounds of Formula (I) may be asymmetric. In one embodiment, m and p are 1. In some embodiments, q is 0. In some embodiments, n is an integer from 1 to 5. In some embodiments, q, m and p are each 1 and n is 1 , 2 or 5.
  • each Ri, R2, R3, m, n, p and q disclosed in reference to Formula (I) intends and includes all combinations thereof the same as if each and every combination of R-i, R2, R3, m, n, p and q were specifically and individually listed.
  • Representative compounds of the Formula (I) include:
  • polyamine compound is represented by the structure according to Formula (II):
  • n 1 , 2 or 3;
  • each L is independently a linker of from about 2 to 14 carbons in length, for example of about 2, 3, 4, 5, 6, 8, 10, 12 or 14 carbon atoms in length, where the linker backbone atoms may be saturated or unsaturated, usually not more than one, two, three, or four unsaturated atoms will be present in a tether backbone, where each of the backbone atoms may be substituted or unsubstituted (for example with a C1-C8 alkyl), where the linker backbone may include a cyclic group (for example, a cyclohex-1 ,3-diyl group where 3 atoms of the cycle are included in the backbone);
  • each R12 is independently selected from hydrogen and a C-i-Cs alkyl
  • each Rn is independently selected from hydrogen, C2-C8alkenyl, Ci- C8 alkyl or C3-C8 branched alkyl ( e.g ., methyl, ethyl, tert-butyl, isopropyl, pentyl, cyclobutyl, cyclopropylmethyl, 3-methylbutyl, 2-ethylbutyl, 5-NH2-pent-1-yl, propyl-1 - ylmethyl(phenyl)phosphinate, dimethylbicyclo[3.1.1 ]heptyl)ethyl, 2- (decahydronaphthyl)ethyl and the like), C6-C20 aryl or heteroaryl, C1-C23 aralkyl or heteroaralkyl (2-phenylbenzyl, 4-phenylbenzyl, 2-benzylbenzyl, 3-benzylbenzyl, 3,3- diphenylpropyl, 3-(benzo
  • each L is independently selected from: -CHRi3-(CH )m-, -CHRi3-(CH )n-CHRi3-, -(CH )mCHRi3-, -CH2-A-CH2- and -(CH 2 )p- where :
  • m is an integer from 1 to 5;
  • A is (CH2)m, ethane-1, 1-diyl or cyclohex-1 ,3-diyl;
  • p is an integer from 2 to 14, such as 1 , 2, 3, 4 or 5;
  • n is an integer from 1 to 12;
  • Ri3 is a C-i-Cs alkyl.
  • the alkyl portion of the aralkyl or heteroaralkyl moiety is connected to the molecule via its alkyl moiety.
  • Rn may be an aralkyl moiety such as 2-phenylbenzyl, 4-phenylbenzyl, 3,3,-diphenylpropyl, 2-(2- phenylethyl)benzyl, 2-methyl-3-phenylbenzyl, 2-napthylethyl, 4-(pyrenyl)butyl, 2-(3- methylnapthyl)ethyl, 2-(1 ,2-dihydroacenaphth-4-yl)ethyl and the like.
  • aralkyl moiety such as 2-phenylbenzyl, 4-phenylbenzyl, 3,3,-diphenylpropyl, 2-(2- phenylethyl)benzyl, 2-methyl-3-phenylbenzyl, 2-napthylethyl, 4-(pyrenyl)butyl, 2-(3- methylnapthyl)ethyl, 2-(1 ,2-dihydroacenaphth-4-
  • Rn may be a heteroaralkyl moiety such as 3-(benzoimidazolyl)propanoyl, 1 -(benzoimidazolyl)methanoyl, 2-(benzoimidazolyl)ethanoyl, 2-(benzoimidazolyl)ethyl and the like.
  • the compound of Formula (II) comprises at least one moiety selected from the group consisting of t-butyl, isopropyl, 2-ethylbutyl, 1-methylpropyl, 1-methylbutyl, 3-butenyl, isopent-2-enyl, 2-methylpropan-3-olyl, ethylthiyl, phenylthiyl, propynoyl, 1 -methyl-1 H-pyrrole-2-yl; trifluoromethyl, cyclopropanecarbaldehyde, halo-substituted phenyl, nitro-substituted phenyl, alkyl- substituted phenyl, 2,4,6-trimethylbenzyl, halo-5-substituted phenyl (such as para- (F3S)-phenyl, azido and 2-methylbutyl.
  • t-butyl isopropyl, 2-ethylbutyl, 1-methylpropyl,
  • each Rn is independently selected from hydrogen, n-butyl, ethyl, cyclohexylmethyl, cyclopentylmethyl, cyclopropylmethyl, cycloheptylmethyl, cyclohexyleth-2-yl, and benzyl.
  • the polyamine compound is of the structure of Formula (II), where n is 3, such that the compound has a structure according to Formula (III):
  • Li, L2 and L3 are independently selected from -CHRi3-(CH )m-, -CHRi3-(CH 2 )n-CHRi3-, -(CH 2 )m-CHRi 3 -, -CH 2 -A-CH 2 - and -(CH 2 )p-;
  • the polyamine compound is of the structure of Formula (III) where: Li is -CHRi3-(CH 2 )m-; l_ 2 is -CHRi3-(CH 2 ) n -CHRi3-; and L3 is - (CH 2 )m-CHRi3-; where Rn, R12, R13, m and n are as defined above.
  • the polyamine compound is of the structure of Formula (III) where: Li, l_2, and L3 are independently -CH 2 -A-CH 2 -; and R12 is hydrogen; where R11 and A are as defined above.
  • at least one of an A and an Rn comprises an alkenyl moiety.
  • the polyamine compound is of the structure of Formula (III) where: Li, L2 and l_3are independently -(CFl2)p- where p is as defined above; and R121S hydrogen.
  • Li and L3 are independently -(CFl2)p- where p is as defined above; and R121S hydrogen.
  • p is an integer from 3 to 7
  • L3 p is an integer from 3 to 14.
  • the polyamine compound is of the structure of Formula (III) where: Li, and L3 are independently -(CH2) P -; L2 is -CFI2-A-CFI2-; and R12 is hydrogen; where R12, p and A are as defined above.
  • p is an integer from 2 to 6
  • L3 A is (CFl2) x where x is an integer from 1 to 5, or cyclohex-1 ,3-diyl.
  • the polyamine compound is of the structure of Formula (II), where n is 2, such that the compound has a structure according to Formula (IV):
  • Li and L2 are independently selected from -CHRi3-(CH2)m- CHRi3-(CH 2 )n-CHRi3-, -(CH 2 )n, CHR13-, -CH2-A-CH2- and -(CH2 ) P -;
  • the polyamine compound is of the structure of Formula (IV) where: Li is -(CH2) P -; and L2 is -(CH2)m-CHRi3-; where R13, m and p are as defined above.
  • Li p is an integer from 3 to 10
  • L2 n is an integer from 2 to 9.
  • the polyamine compound is of the structure of Formula (IV) where: Li and L2 are -(CH2) P -; where p is as defined above. In particular embodiments, p is an integer from 3 to 7. [0184] In certain embodiments, the polyamine compound is of the structure of Formula (II), where n is 1 , such that the compound has a structure according to Formula (V);
  • Li is -(CFl2)p- where p is as defined above.
  • p is an integer from 2 to 6.
  • one Rn is an amino- substituted cycloalkyl (e.g ., a cycloalkyl group substituted with a primary, secondary, tertiary or quaternary amine) or a C2-C8 alkanoyl (which alkanoyl may be substituted with one or more substituents such as a methyl or an alkylazide group); and the other R11 is a C-i-Cs alkyl or a C7-C24 aralkyl.
  • a cycloalkyl group substituted with a primary, secondary, tertiary or quaternary amine e.g a cycloalkyl group substituted with a primary, secondary, tertiary or quaternary amine
  • a C2-C8 alkanoyl which alkanoyl may be substituted with one or more substituents such as a methyl or an alkylazide group
  • the other R11 is a C-i-Cs
  • Representative compounds of the Formula (II) include, e.g. ⁇
  • Phenylcyclopropylamine derivatives that are inhibitors of include compounds represented by Formula (VI):
  • each of R1-R5 is independently selected from H, halo, alkyl, alkoxy, cycloalkoxy, haloalkyl, haloalkoxy, -L-aryl, -L-heterocyclyl, -L-carbocyclyl, acylamino, acyloxy, alkylthio, cycloalkylthio, alkynyl, amino, alkylamino, aryl, arylalkyl, arylalkenyl, arylalkynyl, arylalkoxy, aryloxy, arylthio, heteroarylthio, cyano, cyanato, haloaryl, hydroxyl, heteroaryloxy, heteroarylalkoxy, isocyanato, isothiocyanate, nitro, sulfinyl, sulfonyl, sulfonamide, thiocarbonyl, thiocyanato,
  • R6 is H or alkyl
  • R7 is H, alkyl, or cycloalkyl
  • R8 is a -L-heterocyclyl wherein the ring or ring system of the -L-heterocyclyl has from 0 to 3 substituents selected from halo, alkyl, alkoxy, cycloalkoxy, haloalkyl, haloalkoxy, -L-aryl, -L-heterocyclyl, -L-carbocyclyl, acylamino, acyloxy, alkylthio, cycloalkylthio, alkynyl, amino, alkylamino, aryl, arylalkyl, arylalkenyl, arylalkynyl, arylalkoxy, aryloxy, arylthio, heteroarylthio, cyano, cyanato, haloaryl, hydroxyl, heteroaryloxy, heteroarylalkoxy, isocyanato, isothiocyanate, nitro, sulfin,
  • R8 is -L-aryl wherein the ring or ring system of the -L-aryl has from 1 to 3 substituents selected from halo, alkyl, alkoxy, cycloalkoxy, haloalkyl, haloalkoxy, -L-aryl, -L-heterocyclyl, -L-carbocyclyl, acylamino, acyloxy, alkylthio, cycloalkylthio, alkynyl, amino, alkylamino, aryl, arylalkyl, arylalkenyl, arylalkynyl, arylalkoxy, aryloxy, arylthio, heteroarylthio, cyano, cyanato, haloaryl, hydroxyl, heteroaryloxy, heteroarylalkoxy, isocyanato, isothiocyanate, nitro, sulfinyl, sulfony
  • each L is independently selected from -(CH 2 )n- (CH 2 )n-, -(CH2)nNH(CH 2 )n-, -(CH 2 ) n O(CH 2 ) n -, and -(CH 2 )nS(CH 2 ) n -, and where each n is independently chosen from 0, 1 , 2, and 3;
  • L is a covalent bond.
  • R6 and R7 are hydro.
  • one of R1 -R5 is selected from -L-aryl, -L-heterocyclyl, and -L-carbocyclyl.
  • a compound of the invention is of Formula (VI) where: [0200] each R1 -R5 is optionally substituted and independently chosen from -H, halo, alkyl, alkoxy, cycloalkoxy, haloalkyl, haloalkoxy, -L-aryl, -L-heteroaryl, -L- heterocyclyl, -L-carbocyclyl, acylamino, acyloxy, alkylthio, cycloalkylthio, alkynyl, amino, aryl, arylalkyl, arylalkenyl, arylalkynyl, arylalkoxy, aryloxy, arylthio, heteroarylthio, cyano, cyanato, haloaryl, hydroxyl, heteroaryloxy, heteroarylalkoxy, isocyanato, isothiocyanato, nitro, sulf
  • R6 is chosen from -H and alkyl
  • R7 is chosen from -H, alkyl, and cycloalkyl
  • Rx when present is chosen from -H, alkyl, alkynyl, alkenyl, -L- carbocyclyl, -L-aryl, and -L-heterocyclyl, all of which are optionally substituted (except -H);
  • Ry when present is chosen from -H, alkyl, alkynyl, alkenyl, -L- carbocyclyl, -L-aryl, and -L-heterocyclyl, all of which are optionally substituted (except -H), where Rx and Ry may be cyclically linked;
  • Rz when present is chosen from -H, alkoxy, -L-carbocyclyl, -L- heterocyclyl, -L-aryl, wherein the aryl, heterocyclyl, or carbocyclyl are optionally substituted; each L is a linker that links the main scaffold of Formula I to a carbocyclyl, heterocyclyl, or aryl group, wherein the hydrocarbon portion of the linker -L- is saturated, partially saturated, or unsaturated, and is independently chosen from a saturated parent group having a formula of -(CH2)n-(CH2)2-,
  • optionally substituted refers to zero or 1 to 4 optional substituents independently chosen from acylamino, acyloxy, alkenyl, alkoxy, cycloalkoxy, alkyl, alkylthio, cycloalkylthio, alkynyl, amino, aryl, arylalkyl, arylalkenyl, arylalkynyl, arylalkoxy, aryloxy, arylthio, heteroarylthio, carbocyclyl, cyano, cyanato, halo, haloalkyl, haloaryl, hydroxyl, heteroaryl, heteroaryloxy, heterocyclyl, heteroarylalkoxy, isocyanato, isothiocyanato, nitro, sulfinyl, sulfonyl, sulfonamide, thiocarbonyl, thiocyanato, trihalomethanesulphonamido, O-
  • R8 is -CORz, such that the compound is of the following structure:
  • R1-R7 are described above; and Rz is -L-heterocyclyl which is optionally substituted with from 1-4 optional substituents independently chosen from acylamino, acyloxy, alkenyl, alkoxy, cycloalkoxy, alkyl, alkylthio, cycloalkylthio, alkynyl, amino, aryl, arylalkyl, arylalkenyl, arylalkynyl, arylalkoxy, aryloxy, arylthio, heteroarylthio, carbocyclyl, cyano, cyanato, halo, haloalkyl, haloaryl, hydroxyl, heteroaryl, heteroaryloxy, heterocyclyl, heteroarylalkoxy, isocyanato, isothiocyanato, nitro, sulfinyl, sulfonyl, sulfonamide, thiocarbonyl
  • each L is independently chosen from -(CH 2 )n-(CH 2 )n- and -(CH 2 )n-0-(CH 2 )n where each n is independently chosen from 0, 1 , 2, and 3.
  • each L is chosen from a bond, -CH 2 -, -CH2CH2-, -OCH2-, -OCH2CH2-, -CH2OCH2-, -CH2CH2CH2-, -OCH2CH2CH2-, and -CH2OCH2CH2-.
  • each L is chosen from a bond, -CH2-, -CH2CH2-, OCH2-, and -CH2CH2CH2-.
  • L is chosen from a bond and -CH2-.
  • Exemplary compounds of Formula (VI) include:
  • Exemplary compounds of Formula VI include: N-cyclopropyl-2- ⁇ [(trans)-2-phenylcyclopropyl]amino ⁇ acetamide; 2- ⁇ [(trans)-2- phenylcyclopropyl]amino acetamide; N-cyclopropyl-2- ⁇ [(trans)-2- phenylcyclopropyl]amino ⁇ propanamide; 2- ⁇ [(trans)-2-phenylcyclopropyl]amino ⁇ -N- prop-2-ynylacetamide; N-isopropyl-2- ⁇ [(trans)-2-phenylcyclopropyl]amino ⁇ acetamide; N-(tert-butyl)-2- ⁇ [(trans)-2-phenylcyclopropyl]amino ⁇ acetamide; N-(2-morpholin-4-yl- 2-oxoethyl)-N-[(trans)-2-phenylcyclopropyl]amine; 2- ⁇ [(trans)
  • Alternative small molecule LSD inhibitor compounds may be selected from selective LSD1 and LSD1/MAOB dual inhibitors disclosed for example in International PCT Patent Pub. No. WO 2010/043,721 , WO 2010/084,160,
  • Representative compounds of this type include phenylcyclopropylamine derivatives or homologs, illustrative examples of which include phenylcyclopropylamine with one or two substitutions on the amine group; phenylcyclopropylamine with zero, one or two substitutions on the amine group and one, two, three, four, or five substitution on the phenyl group; phenylcyclopropylamine with one, two, three, four, or five substitution on the phenyl group; phenylcyclopropylamine with zero, one or two substitutions on the amine group wherein the phenyl group of PCPA is substituted with (exchanged for) another ring system chosen from aryl or heterocyclyl to give an aryl- or heteroaryl- cycl
  • Non-limiting embodiments of phenylcyclopropylamine derivatives or analogues include “cyclopropylamine amide” derivatives and “cyclopropylamine” derivatives.
  • Specific examples of “cyclopropylamine acetamide” derivatives include, but are not limited to: N-cyclopropyl-2- ⁇ [(trans)-2-phenylcyclopropyl]amino ⁇ acetamide; 2- ⁇ [(trans)-2-phenylcyclopropyl]amino ⁇ acetamide; N-cyclopropyl-2- ⁇ [(trans)-2-phenylcyclopropyl]amino ⁇ propanamide; 2- ⁇ [(trans)-2-phenylcyclopropyl] amino ⁇ -N-prop-2-ynylacetamide; N-isopropyl-2- ⁇ [(trans)-2-phenylcyclopropyl]amino ⁇ acetamide; N-(tert-butyl)-2- ⁇ [(trans)-2-
  • cyclopropylamine derivatives, include, but are not limited to: N-4-fluorobenzyl-N- ⁇ (trans)-2-[4-(benzyloxy)phenyl]cyclopropyl ⁇ amine, N-4-methoxybenzyl-N- ⁇ (trans)-2-[4-(benzyloxy)phenyl]cyclopropyl ⁇ amine, N-benzyl- N- ⁇ (trans)-2-[4-(benzyloxy)phenyl]cyclopropyl ⁇ amine, N-[(trans)-2- phenylcyclopropyl]amino-methyl)pyridin-3-ol, N-[(trans)-2-phenylcyclopropyl]-N-(3- methylpyridin-2-ylmethyl)amine, N-[(trans)-2-phenylcyclopropyl]-N-(4-chloropyridin-3- ylmethyl)amine, N-[(trans)-2-phenylcyclopropyl]
  • LSD1 inhibitors are, e.g., phenelzine or pargyline (propargylamine) or a derivative or analogue thereof.
  • Derivatives and analogues of phenelzine and pargyline (propargylamine) include, but are not limited to, compounds where the phenyl group of the parent compound is replaced with a heteroaryl or optionally substituted cyclic group or the phenyl group of the parent compound is optionally substituted with a cyclic group.
  • the phenelzine or pargyline derivative or analogue thereof has selective LSD1 or dual LSD1/MAOB inhibitory activity as described herein.
  • the phenelzine derivative or analogue has one, two, three, four or five substituents on the phenyl group.
  • the phenelzine derivative or analogue has the phenyl group substituted with (exchanged for) an aryl or heterocyclyl group wherein said aryl or heterocyclyl group has zero, one, two, three, four or five substituents.
  • the pargyline derivative or analogue has one, two, three, four or five substituents on the phenyl group.
  • the pargyline derivative or analogue has the phenyl group substituted with (exchanged for) an aryl or heterocyclyl group wherein said aryl or heterocyclyl group has zero, one, two, three, four or five substituents.
  • the present invention also contemplates tranylcypromine derivatives as described for example by Binda etal., (2010. J . Am. Chem. Soc. 132: 6827- 6833, which is hereby incorporated by reference herein in its entirety) as inhibitors of LSD (e.g ., LSD1 and/or LSD2) enzymatic function.
  • LSD e.g ., LSD1 and/or LSD2
  • LSD1 inhibitor compounds may be selected from tranylcypromine analogues described by Benelkebir etal. (2011. Bioorg. Med. Chem. doi: 10.1016/j.bmc.2011.02.017, which is hereby incorporated by reference herein in its entirety),
  • Representative analogues of this type, including o-, m-, and p- bromo analogues include: (1 R, 2S)-2-(4-bromophenyl)cyclopropanamine hydrochloride (Compound 4c), (1 R, 2S)-2-(3-bromophenyl)cyclopropanamine hydrochloride (Compound 4d), : (1 R, 2S)-2-(2-bromophenyl)cyclopropanamine hydrochloride (Compound 4e), : (1 R, 2S)-2-(biphenyl-4-yl)cyclopropanamine hydrochloride (Compound 4f).
  • Non-limiting compounds disclosed by Culhane etal. include propargyl-Lys-4, N-methylpropargyl-Lys-4 H3- 21 , c/s-3-chloroallyl-Lys-4 H3-21 , trans- 3-chloroallyl-Lys-4 H3-21 , exo-cyclopropyl- Lys-4 H3-21 , endo- cyclopropyl-Lys-4 H3-21 , enc/o-dimethylcyclopropyl-Lys-4, hydrazino-Lys-4 H3-21 and hydrazino-Lys-4 H3-21.
  • cyclopropylamine compounds that are useful for inhibiting LSD1 include those disclosed by Fyfe etal., in U.S. Pat. Pub. No. 2013/0197013, which is incorporated herein by reference in its entirety.
  • Illustrative cyclopropylamine inhibitors of LSD1 which are disclosed as being selective for inhibiting LSD1 , include compounds according to Formula VII:
  • X 1 and X 2 are independently C(R2) or N;
  • X 3 and X 4 when present, are independently C(R2) or N;
  • (G) is a cyclyl group (as shown in Formula VII, the cyclyl group (G) has n substituents (R1));
  • each (R1) is independently chosen from alkyl, alkenyl, alkynyl, cyclyl, -L1 -cyclyl, -L1 -amino, -L1 -hydroxyl, amino, amido, nitro, halo, haloalkyl, haloalkoxy, cyano, sulfinyl, sulfonyl, sulphonamide, hydroxyl, alkoxy, urea, carbamate, acyl, or carboxyl;
  • each (R2) is independently chosen from -H, alkyl, alkenyl, alkynyl, cyclyl, -L1 -cyclyl, -L1 -amino, -L1 -hydroxyl, amino, amido, nitro, halo, haloalkyl, haloalkoxy, cyano, sulfinyl, sulfonyl, sulphonamide, hydroxyl, alkoxy, urea, carbamate, acyl, or carboxyl, wherein each (R2) group has 1 , 2, or 3 independently chosen optional substituents or two (R2) groups can be taken together to form a heterocyclyl or aryl group having 1 , 2, or 3 independently chosen optional substituents, wherein said optional substituents are independently chosen from alkyl, alkanoyl, heteroalkyl, heterocyclyl, haloalkyl, cycloalkyl, carbocyclyl, arylalk
  • R3 is -H or a (Ci-C6)alkyl group
  • each L1 is independently alkylene or heteroalkylene
  • n 0, 1 , 2, 3, 4 or 5
  • compounds of Formula (VII) are represented by Formula (VIII):
  • X 1 is CH or N;
  • G is a cyclyl group;
  • each (R1) is independently chosen from alkyl, alkenyl, alkynyl, cyclyl, -L1 -cyclyl, -L1 -amino, -L1 -hydroxyl, amino, amido, nitro, halo, haloalkyl, haloalkoxy, cyano, sulfinyl, sulfonyl, sulphonamide, hydroxyl, alkoxy, urea, carbamate, acyl, or carboxyl;
  • each (R2) is independently chosen from alkyl, alkenyl, alkynyl, cyclyl, -L1 -cyclyl, -L1 -amino, -L1 -hydroxyl, amino, amido, nitro, halo, haloalkyl, haloalkoxy, cyano, sulfinyl, sulfonyl, sulphonamide, hydroxyl, alkoxy, urea, carbamate, acyl, or carboxyl, wherein each (R2) group has 1 , 2, or 3 optional substituents, wherein said optional substituents are independently chosen from alkyl, alkanoyl, heteroalkyl, heterocyclyl, haloalkyl, cycloalkyl, carbocyclyl, arylalkoxy, heterocyclylalkoxy, aryl, aryloxy, heterocyclyloxy, alkoxy, haloalkoxy, oxo,
  • each L1 is independently alkylene or heteroalkylene
  • (G) is a cyclyl group
  • each (R1) is independently chosen from alkyl, alkenyl, alkynyl, cyclyl, -L1 -cyclyl, -L1 -amino, -L1 -hydroxyl, amino, amido, nitro, halo, haloalkyl, haloalkoxy, cyano, sulfinyl, sulfonyl, sulphonamide, hydroxyl, alkoxy, urea, carbamate, acyl, or carboxyl;
  • each (R2) is independently chosen from alkyl, alkenyl, alkynyl, cyclyl, -L1 -cyclyl, -L1 -amino, -L1 -hydroxyl, amino, amido, nitro, halo, haloalkyl, haloalkoxy, cyano, sulfinyl, sulfonyl, sulphonamide, hydroxyl, alkoxy, urea, carbamate, acyl, or carboxyl, wherein each (R2) group has 0, 1 , 2, or 3 optional substituents, wherein said optional substituents are independently chosen from alkyl, alkanoyl, heteroalkyl, heterocyclyl, haloalkyl, cycloalkyl, carbocyclyl, arylalkoxy, heterocyclylalkoxy, aryl, aryloxy, heterocyclyloxy, alkoxy, haloalkoxy, oxo,
  • each L1 is independently alkylene or heteroalkylene; m is 0, 1 , 2 or 3; and
  • n 0, 1 , 2, 3, 4 or 5
  • (G) is a cyclyl group; each (R1 ) is independently chosen from alkyl, alkenyl, alkynyl, cyclyl, -L1 -cyclyl, -L1 -amino, -L1 -hydroxyl, amino, amido, nitro, halo, haloalkyi, haloalkoxy, cyano, sulphinyl, sulphonyl, sulphonamide, hydroxyl, alkoxy, urea, carbamate, acyl, or carboxyl;
  • each (R2) is independently chosen from alkyl, alkenyl, alkynyl, cyclyl, - L1 -cyclyl, -L1 -amino, -L1 -hydroxyl, amino, amido, nitro, halo, haloalkyi, haloalkoxy, cyano, sulphinyl, sulphonyl, sulphonamide, hydroxyl, alkoxy, urea, carbamate, acyl, or carboxyl, wherein each (R2) group has 1 , 2, or 3 optional substituents, wherein said optional substituents are independently chosen from alkyl, alkanoyl, heteroalkyl, heterocyclyl, haloalkyi, cycloalkyi, carbocyclyl, arylalkoxy, heterocyclylalkoxy, aryl, aryloxy, heterocyclyloxy, alkoxy, haloalkoxy, oxo, acyl
  • R3 is -H or a (Ci-C6)alkyl group; each L1 is alkylene or heteroalkylene; and n is 0, 1 , 2, 3, 4 or 5,
  • X 1 , X 2 , X 3 and X 4 are independently CH or N, provided that at least one of X 1 , X 2 , X 3 and X 4 is N;
  • (G) is a cyclyl group; each (R1 ) is independently chosen from alkyl, alkenyl, alkynyl, cyclyl, -L1 -cyclyl, -L1 -amino, -L1 -hydroxyl, amino, amido, nitro, halo, haloalkyl, haloalkoxy, cyano, sulfinyl, sulphonyl, sulphonamide, hydroxyl, alkoxy, urea, carbamate, acyl, or carboxyl;
  • each (R2) is independently chosen from alkyl, alkenyl, alkynyl, cyclyl, -L1 -cyclyl, -L1 -amino, -L1 -hydroxyl, amino, amido, nitro, halo, haloalkyl, haloalkoxy, cyano, sulphinyl, sulphonyl, sulphonamide, hydroxyl, alkoxy, urea, carbamate, acyl, or carboxyl, wherein each (R2) group has 1 , 2, or 3 optional substituents, wherein said optional substituents are independently chosen from alkyl, alkanoyl, heteroalkyl, heterocyclyl, haloalkyl, cycloalkyl, carbocyclyl, arylalkoxy, heterocyclylalkoxy, aryl, aryloxy, heterocyclyloxy, alkoxy, haloalkoxy, oxo, acyl
  • Representative compounds according to Formula VII are suitably selected from: (trans)-2-(3'-(trifluoromethyl)biphenyl-4-yl)cyclopropanamine; (trans)- 2-(terphenyl-4-yl)cyclopropanamine; 4'-((trans)-2-aminocyclopropyl)biphenyl-4-ol; 4'- ((trans)-2-aminocyclopropyl)biphenyl-3-ol; (trans)-2-(6-(3- (trifluoromethyl)phenyl)pyridin-3-yl)cyclopropanamine; (Trans)-2-(6-(3,5- dichlorophenyl)pyridin-3-yl)cyclopropanamine; (trans)-2-(6-(4-chlorophenyl)pyridin-3- yl)cyclopropanamine; (trans)-2-(6-(3-chlorophenyl)pyridin-3-yl)cyclopropanamine
  • LSD1 inhibitor compounds are selected from phenylcyclopropylamine derivatives, as described for example by Ogasawara etal. (2013, Angew. Chem. Int. Ed. 52: 8620-8624, which is hereby incorporated by reference herein in its entirety).
  • Representative compounds of this type are represented by Formula (XII):
  • An is a 5 to 7 membered aryl or heteroaryl ring
  • Ar2 and Ar3 are each independently selected from a 5 to 7 membered aryl or heteroaryl ring, optionally substituted with 1 to 3 substituents;
  • R3 is selected from hydrogen, -Ci ealkyl or -OH; [0268] m is an integer from 1 to 5; and
  • n is an integer from 1 to 3;
  • An is a six membered aryl or heteroaryl ring, especially phenyl, pyridine, pyrimidine, pyrazine 1 ,3,5-triazine, 1 ,2,4-trazine and 1 ,2,3-triazine, more especially phenyl;
  • Ar2 is a six membered aryl or heteroaryl ring, especially phenyl, pyridine, pyrimidine, pyrazine 1 ,3,5-triazine, 1 ,2,4-trazine and 1 ,2,3-triazine, especially phenyl; especially where the six membered aryl or heteroaryl ring is optionally substituted with one optional substituent, especially in the 3 or 4 position;
  • Ar3 is a six membered aryl or heteroaryl ring, especially phenyl, pyridine, pyrimidine, pyrazine 1 ,3,5-triazine, 1 ,2,4-trazine and 1 ,2,3-triazine, especially phenyl; especially where the six membered aryl or heteroaryl ring is optionally substituted with one optional substituent, especially in the 3 or 4 position.
  • Particular optional substituents for An and Ar2 include -Ci ealkyl, -C2- 6alkenyl, -CH2F, -CFIF2, -CF3, halo, aryl, heteroaryl, -C(0)NFICi-6alkyl, -C(0)NFICI-6 alkylNH2, -C(0)-heterocyclyl, especially methyl, ethyl, propyl, butyl, t-butyl, -CFI2F, - CHF2, -CH3, Cl, F, phenyl, -C(0)NH(CH2)I- NH and -C(0)-heterocyclyl ;
  • R3 is FI, -Ci salkyl or -OFI, especially FI, -CFI3 or -OFI.
  • m is 2 to 5, especially 3 to 5, more especially 4,
  • n is 1 or 2, especially 1.
  • the compounds of Formula (XII) are compounds of Formula (Xlla): (XI la)
  • Non-limiting compounds represented by Formula (XII) include the following:
  • LSD1 inhibitors include, but are not limited to those, e.g., disclosed in Ueda et al. (2009. J. Am. Chem. Soc. 131 (48): 17536-17537) and Mimasu et al., (2010, Biochemistry, 49(30): 6494-503).
  • Other phenylcyclopropylamine derivatives and analogues are found, e.g., in Kaiser etal. (1962, J. Med. Chem. 5: 1243-1265); Zirkle etal. (1962. J. Med. Chem. 1265-1284; U.S. Pat. Nos.
  • the inhibitory agent is GSK- LSD1 , which has the following molecular structure:
  • the agent is SP- 2509, which has the following molecular structure:
  • the agent is SP- 2577, which has the following molecular structure:
  • the invention not only encompasses known LSD (e.g ., LSD1 or LSD2) inhibitors but LSD inhibitors identified by any suitable screening assay. Accordingly, the present invention extends to methods of screening for modulatory agents that are useful for inhibiting a LSD1 and, in turn, for preventing demethylation of an ACE2 polypeptide.
  • LSD e.g ., LSD1 or LSD2
  • the screening methods comprise (1) contacting a preparation with a test agent, wherein the preparation comprises (i) a polypeptide comprising an amino acid sequence corresponding to at least a biologically active fragment of LSD1 or to a variant or derivative thereof; or (ii) a polynucleotide comprising a nucleotide sequence from which a transcript of a LSD1 gene or portion thereof is producible, or (iii) a polynucleotide comprising at least a portion of a genetic sequence (e.g.
  • a transcriptional element that regulates the expression of a LSD1 gene, which is operably linked to a reporter gene; and (2) detecting a change in the level or functional activity of the polypeptide, the polynucleotide or an expression product of the reporter gene, relative to a reference level or functional activity in the absence of the test agent.
  • a detected reduction in the level and/or functional activity of the polypeptide, transcript or transcript portion or an expression product of the reporter gene, relative to a normal or reference level and/or functional activity in the absence of the test agent indicates that the agent is useful for preventing demethylation of an ACE2 polypeptide.
  • this is confirmed by analysing or determining whether the test agent prevents the demethylation of an ACE2 polypeptide, and would therefore be useful as an agent for preventing or treatment a coronavirus infection.
  • Modulators falling within the scope of the present invention include inhibitors of the level or functional activity of a LSD (e.g., LSD1 or LSD2), including antagonistic antigen-binding molecules, and inhibitor peptide fragments, antisense molecules, ribozymes, RNAi molecules and co-suppression molecules as well as polysaccharide and lipopolysaccharide inhibitors of a LSD (e.g., LSD1 or LSD2) .
  • Candidate agents encompass numerous chemical classes, though typically they are organic molecules, preferably small organic compounds having a molecular weight of more than 50 and less than about 2,500 Dalton.
  • Candidate agents comprise functional groups necessary for structural interaction with proteins, particularly hydrogen bonding, and typically include at least an amine, carbonyl, hydroxyl or carboxyl group, desirably at least two of the functional chemical groups.
  • the candidate agent often comprises cyclical carbon or heterocyclic structures or aromatic or polyaromatic structures substituted with one or more of the above functional groups.
  • Candidate agents are also found among biomolecules including, but not limited to: peptides, saccharides, fatty acids, steroids, purines, pyrimidines, derivatives, structural analogues or combinations thereof.
  • Small (non-peptide) molecule modulators of a LSD ⁇ e.g., LSD1 or LSD2) are particularly advantageous.
  • small molecules are desirable because such molecules are more readily absorbed after oral administration, have fewer potential antigenic determinants, or are more likely to cross the cell membrane than larger, protein-based pharmaceuticals.
  • Small organic molecules may also have the ability to gain entry into an appropriate cell and affect the expression of a gene ( e.g ., by interacting with the regulatory region or transcription factors involved in gene expression); or affect the activity of a gene by inhibiting or enhancing the binding of accessory molecules.
  • libraries of natural compounds in the form of bacterial, fungal, plant and animal extracts are available or readily produced.
  • natural or synthetically produced libraries and compounds are readily modified through conventional chemical, physical and biochemical means, and may be used to produce combinatorial libraries.
  • Known pharmacological agents may be subjected to directed or random chemical modifications, such as acylation, alkylation, esterification, amidification, etc. to produce structural analogues.
  • Screening may also be directed to known pharmacologically active compounds and chemical analogues thereof.
  • Screening for modulatory agents according to the invention can be achieved by any suitable method.
  • the method may include contacting a cell expressing a polynucleotide corresponding to a gene that encodes a LSDIwith an agent suspected of having the modulatory activity and screening for the modulation of the level or functional activity of the LSD1 , or the modulation of the level of a transcript encoded by the polynucleotide, or the modulation of the activity or expression of a downstream cellular target of the polypeptide or of the transcript (hereafter referred to as target molecules).
  • target molecules a downstream cellular target of the polypeptide or of the transcript
  • Detecting such modulation can be achieved utilizing techniques including, but not restricted to, ELISA, cell-based ELISA, inhibition ELISA, Western blots, immunoprecipitation, slot or dot blot assays, immunostaining, RIA, scintillation proximity assays, fluorescent immunoassays using antigen-binding molecule conjugates or antigen conjugates of fluorescent substances such as fluorescein or rhodamine, Ouchterlony double diffusion analysis, immunoassays employing an avidin-biotin or a streptavidin-biotin detection system, and nucleic acid detection assays including reverse transcriptase polymerase chain reaction (RT-PCR).
  • a polynucleotide from which LSD1 is regulated or expressed may be naturally occurring in the cell which is the subject of testing or it may have been introduced into the host cell for the purpose of testing.
  • the naturally-occurring or introduced polynucleotide may be constitutively expressed - thereby providing a model useful in screening for agents which down- regulate expression of an encoded product of the sequence wherein the down regulation can be at the nucleic acid or expression product level.
  • a polynucleotide may comprise the entire coding sequence that codes for LSD1 or it may comprise a portion of that coding sequence (e.g., the active site of LSD1) or a portion that regulates expression of the corresponding gene that encodes LSD1 (e.g., a LSD1 promoter).
  • the promoter that is naturally associated with the polynucleotide may be introduced into the cell that is the subject of testing.
  • detecting modulation of the promoter activity can be achieved, for example, by operably linking the promoter to a suitable reporter polynucleotide including, but not restricted to, green fluorescent protein (GFP), luciferase, b-galactosidase and catecholamine acetyl transferase (CAT). Modulation of expression may be determined by measuring the activity associated with the reporter polynucleotide.
  • GFP green fluorescent protein
  • CAT catecholamine acetyl transferase
  • These methods provide a mechanism for performing high throughput screening of putative modulatory agents such as proteinaceous or non- proteinaceous agents comprising synthetic, combinatorial, chemical and natural libraries. These methods will also facilitate the detection of agents which bind either the polynucleotide encoding the target molecule or which modulate the expression of an upstream molecule, which subsequently modulates the expression of the polynucleotide encoding the target molecule. Accordingly, these methods provide a mechanism of detecting agents that either directly or indirectly modulate the expression or activity of a target molecule according to the invention.
  • test agents are screened using commercially available assays, illustrative examples of which include EpiQuik Flistone Demethylase LSD1 Inhibitor Screening Assay Kit (Epigentek Group, Brooklyn, NY) or the LSD1 Inhibitor Screening Assay Kit (Cayman Chemical Company, Ann Arbor, Ml).
  • Compounds may be further tested in the animal models to identify those compounds having the most potent in vivo effects. These molecules may serve as “lead compounds” for the further development of pharmaceuticals by, for example, subjecting the compounds to sequential modifications, molecular modelling, and other routine procedures employed in rational drug design.
  • the LSD1 inhibitor inhibits the binding between an LSD1 polypeptide and a CoRest polypeptide.
  • the inhibitor specifically binds a polypeptide that is known to play an important role in activating LSD1.
  • a polypeptide that is known to play an important role in activating LSD1.
  • PKC0 activates the transcription of LDS1
  • any inhibitor sufficient to reduce or prevent the phosphorylation of LSD by PKC0 is equally as applicable for use with the present invention.
  • an isolated or purified proteinaceous molecule represented by any one of SEQ ID NOs:1 or 2:
  • RKEIDPPFRPKVK [SEQ ID NO: 1 ]
  • the proteinaceous molecule of SEQ ID NO: 1 is also referred to herein as “importinib4759” and the proteinaceous molecule of SEQ ID NO: 2 is also referred to herein as "importinib4759_01”.
  • the present invention also contemplates proteinaceous molecules that are variants of SEQ ID NO: 1 and/or 2.
  • variant proteinaceous molecules include proteins derived from the native protein by deletion (so-called truncation) or addition of one or more amino acids to the N-terminal and/or C-terminal end of the native protein; deletion or addition of one or more amino acids at one or more sites in the native protein; or substitution of one or more amino acids at one or more sites in the native protein.
  • variant proteins encompassed by the present invention are biologically active, that is, they continue to possess the desired biological activity of the native protein. Such variants may result from, for example, genetic polymorphism or from human manipulation.
  • the proteinaceous molecules of SEQ ID NO: 1 and/or 2 may be altered in various ways including amino acid substitutions, deletions, truncations, and insertions. Methods for such manipulations are generally known in the art.
  • amino acid sequence variants of SEQ ID NO: 1 and/or 2 can be prepared by mutagenesis of nucleic acids encoding the amino acid sequence of SEQ ID NO: 1 and/or 2. Methods for mutagenesis and nucleotide sequence alterations are well known in the art.
  • Variant peptides or polypeptides of the invention may contain conservative amino acid substitutions at various locations along their sequence, as compared to a parent (e.g ., naturally- occurring or reference) amino acid sequence, such as SEQ ID NO: 1 and/or 2.
  • a “conservative amino acid substitution” is one in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art as discussed in detail below.
  • the amino acid sequence of the proteinaceous molecules of the invention is defined in terms of amino acids of certain characteristics or sub-classes. Amino acid residues are generally sub-classified into major sub-classes as follows:
  • Acidic The residue has a negative charge due to loss of a proton at physiological pH and the residue is attracted by aqueous solution so as to seek the surface positions in the conformation of a peptide in which it is contained when the peptide is in aqueous medium at physiological pH.
  • Amino acids having an acidic side chain include glutamic acid and aspartic acid.
  • Basic The residue has a positive charge due to association with protons at physiological pH or within one or two pH units thereof (e.g. histidine) and the residue is attracted by aqueous solution so as to seek the surface positions in the conformation of a peptide in which it is contained when the peptide is in aqueous medium at physiological pH.
  • Amino acids having a basic side chain include arginine, lysine and histidine.
  • the residue is charged at physiological pH and, therefore, includes amino acids having acidic or basic side chains, such as glutamic acid, aspartic acid, arginine, lysine and histidine.
  • Hydrophobic The residue is not charged at physiological pH and the residue is repelled by aqueous solution so as to seek the inner positions in the conformation of a peptide in which it is contained when the peptide is in aqueous medium at physiological pH.
  • Amino acids having a hydrophobic side chain include tyrosine, valine, isoleucine, leucine, methionine, phenylalanine and tryptophan.
  • Amino acids having a neutral/polar side chain include asparagine, glutamine, cysteine, histidine, serine and threonine.
  • proline differs from all the other naturally-occurring amino acids in that its side chain is bonded to the nitrogen of the a-amino group, as well as the a-carbon.
  • amino acid similarity matrices e.g ., PAM120 matrix and PAM250 matrix as disclosed for example by Dayhoff etal., (1978), A model of evolutionary change in proteins. Matrices for determining distance relationships In M. O. Dayhoff, (ed.), Atlas of protein sequence and structure, Vol. 5, pp.
  • proline in the same group as glycine, serine, alanine and threonine. Accordingly, for the purposes of the present invention, proline is classified as a “small” amino acid.
  • Amino acid residues can be further sub-classified as cyclic or non- cyclic, and aromatic or non-aromatic, self-explanatory classifications with respect to the side-chain substituent groups of the residues, and as small or large.
  • the residue is considered small if it contains a total of four carbon atoms or less, inclusive of the carboxyl carbon, provided an additional polar substituent is present; three or less if not.
  • Small amino acid residues are, of course, always non-aromatic.
  • amino acid residues may fall in two or more classes.
  • sub classification according to this scheme is presented in Table 1.
  • Conservative amino acid substitution also includes groupings based on side chains.
  • a group of amino acids having aliphatic side chains is glycine, alanine, valine, leucine, and isoleucine; a group of amino acids having aliphatic-hydroxyl side chains is serine and threonine; a group of amino acids having amide-containing side chains is asparagine and glutamine; a group of amino acids having aromatic side chains is phenylalanine, tyrosine, and tryptophan; a group of amino acids having basic side chains is lysine, arginine, and histidine; and a group of amino acids having sulphur-containing side chains is cysteine and methionine.
  • Amino acid substitutions falling within the scope of the invention are, in general, accomplished by selecting substitutions that do not differ significantly in their effect on maintaining (a) the structure of the peptide backbone in the area of the substitution, (b) the charge or hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain. After the substitutions are introduced, the variants are screened for biological activity.
  • amino acids for making conservative substitutions can be grouped into three categories based on the identity of the side chains.
  • the first group includes glutamic acid, aspartic acid, arginine, lysine, histidine, which all have charged side chains;
  • the second group includes glycine, serine, threonine, cysteine, tyrosine, glutamine, asparagine;
  • the third group includes leucine, isoleucine, valine, alanine, proline, phenylalanine, tryptophan, methionine, as described in Zubay, Biochemistry, third edition, Wm.C. Brown Publishers (1993).
  • a predicted non-essential amino acid residue in a peptide of the invention is typically replaced with another amino acid residue from the same side chain family.
  • mutations can be introduced randomly along all or part of the coding sequence of a peptide of the invention, such as by saturation mutagenesis, and the resultant mutants can be screened for an activity of the parent polypeptide, as described for example herein, to identify mutants which retain that activity.
  • the encoded peptide can be expressed recombinantly and its activity determined.
  • a “non-essential” amino acid residue is a residue that can be altered from the wild-type sequence of an embodiment peptide of the invention without abolishing or substantially altering one or more of its activities.
  • the alteration does not substantially alter one of these activities, for example, the activity is at least 20%, 40%, 60%, 70% or 80% of that of the wild-type.
  • an “essential” amino acid residue is a residue that, when altered from the wild-type sequence of an embodiment peptide of the invention, results in abolition of an activity of the parent molecule such that less than 20% of the wild-type activity is present.
  • variants of the proteinaceous molecules of SEQ ID NO 1 and/or 2 of the invention wherein the variants are distinguished from the parent sequence by the addition, deletion, or substitution of one or more amino acid residues.
  • variants will display at least about 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% sequence similarity to a parent or reference proteinaceous molecule sequence as, for example, set forth in SEQ ID NO: 1 or 2, as determined by sequence alignment programs described elsewhere herein using default parameters.
  • variants will have at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% sequence identity to a parent or reference peptide sequence as, for example, set forth in SEQ ID NO: 1 or 2, as determined by sequence alignment programs described herein using default parameters.
  • Variants of importinib4759 and importinib4759_01 which fall within the scope of a variant peptide of the invention, may differ from the parent molecule generally by at least 1 , but by less than 5, 4, 3, 2 or 1 amino acid residue(s).
  • a variant peptide of the invention differs from the corresponding sequence in SEQ ID NO: 1 or 2 by at least 1 , but by less than 5, 4, 3, 2 or 1 amino acid residue(s).
  • the amino acid sequence of the variant peptide of the invention comprises lie (or modified form thereof) at position 5, Asp (or modified form thereof) at position 6, Pro (or modified form thereof) at position 8 and/or Pro (or modified form thereof) at position 9, relative to the numbering of SEQ ID NO: 2.
  • the variant peptide of the invention inhibits PKC- Q nuclear translocation.
  • agents that inhibit LSD1 e.g ., a selective LSD1 inhibitor
  • actives and/or pharmaceutical compositions for treating or preventing a virus infection ⁇ e.g., a coronavirus infection.
  • treatment or prevention includes the prevention of incurring a symptom, holding in check such symptoms, or treating existing symptoms associated with the pathogenic infection, when administered to an individual in need thereof.
  • the S protein of many betacoronaviruses including but not limited to SARS-CoV and SARS-CoV-2, have been demonstrated to employ an ACE2 polypeptide for entry into the host cell.
  • the virus host cell entry receptor polypeptide used by MERS-CoV is a DPP4 polypeptide, and for hCoV-229, a CD13polypeptide.
  • Any LSD1 inhibitor can be used in the compositions and methods of the present invention, provided that the inhibitor is pharmaceutically active.
  • a “pharmaceutically active” LSD1 inhibitor is in a form that results in the treatment and/or prevention of a pathogenic infection, particularly a coronavirus infection, including the prevention of incurring a symptom, holding in check such symptoms or treating existing symptoms associated with the metastatic cancer, when administered to an individual in need thereof.
  • Modes of administration, amounts of LSD1 inhibitor administered, and LSD1 inhibitor formulations, for use in the methods of the present invention, are routine and within the skill of practitioners in the art. Whether a pathogenic infection, particularly a coronavirus infection, has been treated is determined by measuring one or more diagnostic parameters indicative of the course of the disease, compared to a suitable control. In the case of an animal experiment, a “suitable control” is an animal not treated with the LSD1 inhibitor, or treated with the pharmaceutical composition without the LSD1 inhibitor. In the case of a human subject, a “suitable control” may be the individual before treatment, or may be a human ( e.g ., an age- matched or similar control) treated with a placebo.
  • the treatment of a pathogenic infection includes and encompasses without limitation: (1) preventing the uptake of a pathogenic infection ⁇ e.g., a coronavirus infection) into a cell of the host; (2) treating a pathogenic infection ⁇ e.g., a coronavirus infection) in a subject; (3) preventing a pathogenic infection ⁇ e.g., a coronavirus infection) in a subject that has a predisposition to the pathogenic infection but has not yet been diagnosed with the pathogenic infection and, accordingly, the treatment constitutes prophylactic treatment of the pathogenic infection; or (iii) causing regression of a pathogenic infection ⁇ e.g., a coronavirus infection).
  • compositions and methods of the present invention are thus suitable for treating an individual who has been diagnosed with a coronavirus infection, who is suspected of having a coronavirus infection, who is known to be susceptible and who is considered likely to develop a coronavirus infection, or who is considered likely to develop a recurrence of a previously treated coronavirus infection.
  • the coronavirus infection may be a SARS-CoV-2 infection.
  • the coronavirus infection is a SARS-CoV-1 or a SARS-CoV-2 infection.
  • the LSD1 inhibitor-containing compositions will generally contain about 0.000001% to 90%, about 0.0001% to 50%, or about 0.01% to about 25%, by weight of LSD inhibitor, the remainder being suitable pharmaceutical carriers or diluents etc.
  • the dosage of the LSD1 inhibitor can depend on a variety of factors, such as mode of administration, the species of the affected subject, age, sex, weight and general health condition, and can be easily determined by a person of skill in the art using standard protocols. The dosages will also take into consideration the binding affinity of the LSD1 inhibitor to its target molecule, its bioavailability and its in vivo and pharmacokinetic properties.
  • precise amounts of the agents for administration can also depend on the judgment of the practitioner.
  • the physician or veterinarian may evaluate the progression of the disease or condition over time.
  • suitable dosages of the LSD1 inhibitor without undue experimentation.
  • the dosage of the actives administered to a patient should be sufficient to effect a beneficial response in the patient over time such as impairment, abrogation or prevention in the uptake of the virus into a cell of the host, and/or in the treatment and/or prevention of a pathogenic infection (e.g ., a coronavirus infection).
  • the dosages may be administered at suitable intervals to ameliorating the symptoms of the hematologic malignancy.
  • Such intervals can be ascertained using routine procedures known to persons of skill in the art and can vary depending on the type of active agent employed and its formulation. For example, the interval may be daily, every other day, weekly, fortnightly, monthly, bimonthly, quarterly, half-yearly or yearly.
  • Dosage amount and interval may be adjusted individually to provide plasma levels of the active agent, which are sufficient to maintain LSD-inhibitory effects.
  • Usual patient dosages for systemic administration range from 1 -2000 mg/day, commonly from 1-250 mg/day, and typically from 10-150 mg/day. Stated in terms of patient body weight, usual dosages range from 0.02-25 mg/kg/day, commonly from 0.02-3 mg/kg/day, typically from 0.2-1.5 mg/kg/day. Stated in terms of patient body surface areas, usual dosages range from 0.5-1200 mg/m 2 /day, commonly from 0.5-150 mg/m 2 /day, typically from 5-100 mg/m 2 /day.
  • the present invention further contemplates administering the LSD1 inhibitor concurrently with at least one antiviral agent.
  • the LSD1 inhibitor may be used therapeutically after the antiviral agent or may be used before the antiviral agent is administered or together with the antiviral agent.
  • combination therapies which employ a LSD1 inhibitor and concurrent administration of an antiviral agent, non-limiting examples of which include: broad-spectrum antiviral agents and coronavirus-specific antivirus agents.
  • the LSD inhibitors described above or elsewhere herein are particularly effective antiviral agents for monotherapeutic or combined-therapeutic use in treating coronavirus infection.
  • One of the benefits of such combination therapies is that lower doses of the other antiviral agents can be administered while still achieving a similar level of antiviral efficacy.
  • Such lower dosages can be particularly advantageous for drugs known to have genotoxicity and mitochondrial toxicity (for example, some nucleoside analogues).
  • greater efficacy might be achieved using therapeutic doses of two drugs than could be achieved using only a single drug.
  • Examples of other cancer therapies include phototherapy, cryotherapy, toxin therapy or pro-apoptosis therapy.
  • phototherapy is not exhaustive of the types of treatment modalities available for cancer and other hyperplastic lesions.
  • the antiviral drug is suitably selected from antimicrobials, which include without limitation compounds that kill or inhibit the growth of microorganisms (including viruses), and antivirals.
  • Illustrative antivirals include abacavir sulphate, acyclovir sodium, amantadine hydrochloride, amprenavir, chloroquine, cidofovir, delavirdine mesylate, didanosine, efavirenz, favipiravir, famciclovir, fomivirsen sodium, foscarnet sodium, ganciclovir, hydroxychloroquine, hydroquinone.
  • indinavir sulphate lamivudine, lamivudine/zidovudine, lopinavir, nelfinavir mesylate, nevirapine, oseltamivir phosphate, ribavirin, remdesivir, rimantadine hydrochloride, ritonavir, saquinavir, saquinavir mesylate, stavudine, valacyclovir hydrochloride, zalcitabine, zanamivir, and zidovudine.
  • the LSD1 inhibitor may be co administered with an antimicrobial agent including chloroquine, hydroxychloroquine and/or hydroquinone.
  • the antiviral agent comprises a recombinant IFN-b polypeptide (UniProt Accession No. P01574). In some embodiments of this type, the antiviral agent comprises at least a portion of an IFN-b polypeptide, or a variant of an IFN-b polypeptide.
  • the present invention encompasses co administration of an LSD1 inhibitor in concert with an additional agent.
  • the dosages of the actives in the combination may on their own comprise an effective amount and the additional agent(s) may further augment the therapeutic or prophylactic benefit to the patient.
  • the LSD1 inhibitor and the additional agent(s) may together comprise an effective amount for preventing or treating the pathogenic infection.
  • effective amounts may be defined in the context of particular treatment regimens, including, e.g., timing and number of administrations, modes of administrations, formulations, etc.
  • the LSD1 inhibitor and optionally the antiviral agent are administered on a routine schedule.
  • routine schedule refers to a predetermined designated period of time.
  • the routine schedule may encompass periods of time which are identical or which differ in length, as long as the schedule is predetermined.
  • routine schedule may involve administration of the LSD inhibitor on a daily basis, every two days, every three days, every four days, every five days, every six days, a weekly basis, a monthly basis or any set number of days or weeks there-between, every two months, three months, four months, five months, six months, seven months, eight months, nine months, ten months, eleven months, twelve months, etc.
  • the predetermined routine schedule may involve concurrent administration of the LSD1 inhibitor and the antiviral agent on a daily basis for the first week, followed by a monthly basis for several months, and then every three months after that. Any particular combination would be covered by the routine schedule as long as it is determined ahead of time that the appropriate schedule involves administration on a certain day.
  • the present invention provides pharmaceutical compositions for reducing or abrogating the uptake a viruses ⁇ e.g., a coronavirus) to a cell of the host, the pharmaceutical compositions comprising a LSD inhibitor and optionally an antiviral agent useful for treating malignancies.
  • the formulations of the invention are administered in pharmaceutically acceptable solutions, which may routinely contain pharmaceutically acceptable concentrations of salt, buffering agents, preservatives, compatible carriers, adjuvants, and optionally other therapeutic ingredients. Depending on the specific conditions being treated, the formulations may be administered systemically or locally. Techniques for formulation and administration may be found in “Remington’s Pharmaceutical Sciences”, Mack Publishing Co., Easton, Pa., latest edition.
  • Suitable routes may, for example, include oral, rectal, transmucosal, or intestinal administration; parenteral delivery, including intramuscular, subcutaneous, intramedullary injections, as well as intrathecal, direct intraventricular, intravenous, intraperitoneal, intranasal, or intraocular injections.
  • the active agents or drugs of the invention may be formulated in aqueous solutions, suitably in physiologically compatible buffers such as Hanks’ solution, Ringer’s solution, or physiological saline buffer.
  • penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
  • the drugs can be formulated readily using pharmaceutically acceptable carriers well known in the art into dosages suitable for oral administration.
  • Such carriers enable the compounds of the invention to be formulated in dosage forms such as tablets, pills, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated.
  • These carriers may be selected from sugars, starches, cellulose and its derivatives, malt, gelatin, talc, calcium sulphate, vegetable oils, synthetic oils, polyols, alginic acid, phosphate buffered solutions, emulsifiers, isotonic saline, and pyrogen-free water.
  • compositions for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances, which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents, which increase the solubility of the compounds to allow for the preparation of highly, concentrated solutions.
  • compositions for oral use can be obtained by combining the active compounds with solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
  • suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, or polyvinylpyrrolidone (PVP).
  • disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • Such compositions may be prepared by any of the methods of pharmacy but all methods include the step of bringing into association one or more drugs as described above with the carrier, which constitutes one or more necessary ingredients.
  • the pharmaceutical compositions of the present invention may be manufactured in a manner that is itself known, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes.
  • Dragee cores are provided with suitable coatings.
  • suitable coatings may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
  • compositions which can be used orally include push- fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers may be added.
  • Dosage forms of the drugs of the invention may also include injecting or implanting controlled releasing devices designed specifically for this purpose or other forms of implants modified to act additionally in this fashion.
  • Controlled release of an agent of the invention may be achieved by coating the same, for example, with hydrophobic polymers including acrylic resins, waxes, higher aliphatic alcohols, polylactic and polyglycolic acids and certain cellulose derivatives such as hydroxypropyl methyl cellulose.
  • controlled release may be achieved by using other polymer matrices, liposomes or microspheres.
  • the drugs of the invention may be provided as salts with pharmaceutically compatible counterions.
  • Pharmaceutically compatible salts may be formed with many acids, including but not limited to hydrochloric, sulphuric, acetic, lactic, tartaric, malic, succinic, etc. Salts tend to be more soluble in aqueous or other protonic solvents that are the corresponding free base forms.
  • the therapeutically effective dose can be estimated initially from cell culture assays.
  • a dose can be formulated in animal models to achieve a circulating concentration range that includes the IC50 as determined in cell culture (e.g ., the concentration of an active agent, which achieves a half-maximal inhibition in activity of a LSD1 polypeptide). Such information can be used to more accurately determine useful doses in humans.
  • Toxicity and therapeutic efficacy of such drugs can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50:ED50.
  • Compounds that exhibit large therapeutic indices are preferred.
  • the data obtained from these cell culture assays and animal studies can be used in formulating a range of dosage for use in human.
  • the dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity.
  • the dosage may vary within this range depending upon the dosage form employed and the route of administration utilised.
  • the exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient’s condition (see, for example, Fingl etal., 1975, in “The Pharmacological Basis of Therapeutics”, Ch. 1 ).
  • the liposomes will be targeted to and taken up selectively by the tissue.
  • the effective local concentration of the agent may not be related to plasma concentration.
  • MAO inhibitors such as phenelzine, tranylcypromine, and isocarboxazid are currently used clinically to treat depression and cancer, and Phase 1 b clinical trials are currently in progress for metastatic breast cancer which show that LSD1 inhibitors, including phenelzine (NARDIL), are safe and re-invigorate T cell-mediated immunity in humans.
  • LSD1 inhibitors including phenelzine (NARDIL)
  • LSD1 inhibitors can be re-purposed for SARS-CoV-2 therapy by targeting viral entry into host cells and enhancing T cell-mediated immunity. Proving the efficacy of pre-existing drugs would rapidly transform clinical practice and outcomes in the current pandemic.
  • Our efficacy, safety, and toxicity data on LSD1 inhibitors and optimised biomarkers in the oncology setting are readily transferrable to SARS-CoV-2 patients and provide not only a drug but also the biomarkers to monitor the exhaustion phenotype and disease progression.
  • a series of protein domains within both the ACE2 protein and the TMPRSS2 protein were identified as being critical for the entry of the SARS-CoV-2 into the cell. These protein domains are subject to epigenetic post-translational modification (lysine methylation, de-methylation, sumoylation and phosphorylation).
  • the present inventors identified within the TMPRSS2 and ACE2 proteins a series of key serine residues that are phosphorylated by PKCq and key lysine residue methylation sites, these methylated lysine residues also represent sites of LSD1 -mediated de-methylation.
  • Other proteins have been demonstrated to be dynamically regulated by lysine methylation and demethylation or phosphorylation include p53.
  • LSD1 is a key epigenetic regulator
  • the importance of LSD1 on the direct regulation of TMPRSS2 was investigated. Specifically, an examination was performed on the effect of LSD1p inhibition or LSDIp’s upstream regulator PKC0 on the expression dynamics of TMPRSS2.
  • LSD1 siRNA knockouts resulted in significant loss of TMPRSS2 mRNA in both MDA- MB-231 and MCF7 breast cancer cell lines ( Figure 1 A).
  • Samples were either control or phenelzine (FAD and nuclear LSD1p:CoREST inhibitor) or C27 (PKCq catalytic inhibitor) and CHiP enrichment assay carried out by pulling down chromatin fragments with either LSD1p or PKC0 antibodies.
  • PKCq catalytic inhibitor CHiP enrichment assay carried out by pulling down chromatin fragments with either LSD1p or PKC0 antibodies.
  • RNA from each treatment group/sample was hybridized using the human Nanostring custom Panel.
  • the absolute RNA counts were quantified by the nCounter NanoString system (NanoString Technologies, Seattle, WA, USA) and mRNA expression was analysed using the nSolver software and R v.3.3.2 for the Advanced analysis (NanoString Technologies), following the manufacturer’s recommendations) and plotted as fold change versus control.
  • ChIP assays were performed in accordance with the protocol supplied by Upstate Biotechnology. Fixation steps were performed as detailed, and fixed chromatin was sonicated with an Ultrasonic processor (Cole/Parmer) under optimized conditions that gave average DNA fragments of approximately 500 bp, as determined by 2% agarose gel electrophoresis. Prior to antibody addition, samples were precleared with salmon sperm DNA-protein A-agarose and the soluble chromatin fraction was incubated overnight at 4°C with 5 to 10 pg of LSD1p (Merck Millipore) or PKC-0 (Abeam) antibody or without antibody as a control.
  • LSD1p Merck Millipore
  • PKC-0 Abeam
  • Immune complexes were bound to salmon sperm DNA-protein A-agarose and then washed and eluted as described. Protein-DNA cross-links were reversed by incubation at 65°C overnight, and the DNA in each sample was recovered by phenol-chloroform extraction and ethanol precipitation. DNA pellets were washed in 70% ethanol, resuspended in Tris (pH 8.0), and subsequently used for SYBR green real-time PCR amplification (Applied Biosystems). Standard curves were generated for each primer set to correct for differences in primer efficiency. ChIP enrichment ratios were calculated for TMPRSS2 as per standard methods.
  • MDA-MB-231 mesenchymal cells or MCF7 epithelial cells were treated with either control, phenelzine or GSK-LSD1. Cells were then fixed for IFA Microscopy Analysis. Cells were permeabilised by incubating with 1% Triton X-100 for 20 min and were probed with rabbit TRMPSS2 mouse and visualized with a donkey anti-rabbit AF 488. Cover slips were mounted on glass microscope slides with ProLong Glass Antifade reagent (Life Technologies). Protein targets were localised by confocal laser scanning microscopy. Single 0.5 pm sections were obtained using the Metagene-ASI Digital Pathology platform. The final image was obtained by averaging four sequential images of the same section. Digital images were analysed using ASI proprietary analysis software to determine the mean Fluorescent Intensity (mean FI). Graph represents the mean FI values for TMPRS2 using ImageJ to select the nucleus minus background (n > 50 individual cells).
  • LSD1 inhibition significantly inhibits both message and protein expression of TMPRSS2 in a variety of cell lines including in a siLSDI knockdown model. This clearly demonstrates the specificity of LSD1p in regulating TMPRSS2 expression. Flowever, there was a slight decrease on ACE2 transcription in metastatic lung tissue. Despite this it is still likely that LSD1 plays a role in post-translationally modifying ACE2.
  • LSD1 is a key eraser enzyme, that demethylates key histone proteins and key proteins such as transcription factors whereby this demethylation/ methylation post-translational modification has resulted in induction, inhibition or stabilization of the expression of the targeted proteins such as p53. Based on these data, the role of LSD1 as a key regulator of the receptor ACE2 and TMPRSS2 responsible for shuttling SARS-CoV-2 into the cell was investigated.
  • LSD1 significantly co-localized higher with ACE2 in cells infected with SARS-CoV-2, as compared to ACE2/LSD1 expression in uninfected CaCo2 and MRC5 cells (which are not susceptible to SARS-CoV-2 infection) as demonstrated by the PCC(r) co efficient which adjudicates the degree of co-localisation between two protein targets, as well as significantly upregulated expression of both LSD1 and ACE2 was demonstrated in infected cells.
  • MRC5 had no expression of ACE2 and no staining of virus proteins (Figure 3D).
  • LSD1 co-localized with both SARS-CoV-2 spike protein and the nucleocapsid proteins.
  • ACE2 exhibits three high probability lysine residues for post-translation modification by LSD1 and that these lysine residues are part of the C-terminal domain and a novel, putative nuclear localization sequence (NLS).
  • NLS nuclear localization sequence
  • Ddigital images were also analysed using ImageJ software (ImageJ, NIH, Bethesda, MD, USA) to determine the total cell fluorescence or cell surface only fluorescence for non-permeabilised cells.
  • Digital images were analysed using ImageJ software (ImageJ, NIH, Bethesda, MD, USA) to determine the either the Total Nuclear Fluorescent Intensity (TNFI), the Total Cytoplasmic Fluorescent Intensity (TCFI).
  • ImageJ software with automatic thresholding and manual selection of regions of interest (ROIs) specific for cell nuclei was used to calculate the Pearson’s co-efficient correlation (PCC) for each pair of antibodies.
  • the Mann-Whitney nonparametric test (GraphPad Prism, GraphPad Software, San Diego, CA) was used to determine significant differences between datasets.
  • Binding affinity measurements were performed on a Monolith NT.115 (NanoTemper Technologies).
  • the fluorescein-Ahx tagged Ace2 peptide sequence RDRKKKNKARSGEN was manufactured by Genescript. Purified Each reaction consisted of 10 pL of the labelled peptide at 444 nM, mixed with unlabelled LSD1 at the indicated concentrations. All experiments were measured at 25°C with laser off/on/off times of 5/30/5 s. Experiments were conducted at 20% light-emitting diode power and 20-40% MST infra-red laser power. Data from three independently performed experiments were fitted to the single binding model via the NT. Analysis software version 1.5.41 (NanoTemper Technologies) using the signal from Thermophoresis + T-Jump.
  • LSD1 regulates ACE2
  • SARS-CoV-2 expression will be inhibited by LSD1 inhibition via blocking viral entry by down-regulation of ACE2.
  • LSD1 expression at the cell surface is inhibited more significantly by phenelzine but not GSK-LSD1 or L1 (EPI-111) ( Figure 5). This is explained by GSK- LSD1 being only a FAD domain demethylase inhibitor whereas phenelzine is known to be a FAD domain inhibitor that also causes structural alterations in LSD1.
  • L1 (EPI- 111) is a nuclear translocation inhibitor, with its effect attributed to blocking the shuttling of LSD1 into the nucleus.
  • phenelzine is superior at inhibiting expression of ACE2, LSD1 and consequently expression of SARS-CoV-2 is also inhibited.
  • Duolink analysis which detects interacting proteins only demonstrated that LSD1 and ACE2 strongly interact and this interaction is significantly inhibited by phenelzine and to a lesser extent, GSK-LSD1.
  • RT-PCR analysis to measure the impact on SARS-CoV-2 RNA transcription showed that the nuclear targeting inhibitor L1 (EPI-111) was the most superior at inhibiting transcription, with both GSK-LSD1 and phenelzine having an effect (Figure 5I).
  • Phenelzine also induced higher levels of SARS-CoV-2 RNA in the supernatant as a consequence of disrupting virus nucleocapsid formation which encapsulates the virus RNA. Furthermore, while LSD1, ACE2, and TMPRSS2 transcript remained unaltered in SARS-CoV-2-infected Caco-2 cells following phenelzine or GSK-LSD1 treatment, a limited type I interferon response was induced in Caco-2 cells following SARS-CoV-2 infection (Figure 5J).
  • phenelzine treatment showed a 1.72-fold increase of cellular viral RNA replication compared to control, much less viral replication increase was detected in cell supernatant in phenelzine treatment between 24 hpi and 48 hpi (data not shown). This indicated that the virus can replicate within phenelzine-treated cells but much less virus were released into cell culture supernatant compared to control cells. This potentially suggests that replicated viral RNA was accumulated in phenelzine-treated cells due to a block in viral protein translation and viral assembly processes, such that less intact viral pathogens were released to infect other cells.
  • TaqMan quantitative real-time PCR was performed using the Applied Biosystems ViiATM7 Real-Time PCR System (Applied Biosystems, Waltham, MA) with the following human TaqMan probes: ACTB (Hs01060665_g1), GAPDH (Hs02786624_g1), HPRT1 (Hs02800695_m1), ACE2 (Hs01085333_m1), TMPRSS2 (Hs01122322_m1), IFNa (Hs03044218_g1 ), /F/V3 (Hs01077958_s1 ), IFNA (Hs00820125_g1), DDX58 (Hs01061436_m1), IFIH1 (Hs00223420_m1), ISG15 (Hs01921425_s1), OASL (Hs00984387_m1 ), IL-6 (Hs00174131_m1), IL-10 (Hs00961622_m1
  • RNA sequencing was performed to identify global gene expression programs impacted by LSD1 inhibition in SARS-CoV-2-infected Caco-2 cells. LSD1 inhibition albeit at different degrees, impacts on key anti-viral processes, key proteins responsible for viral entry and the transcription and replication of the SARS-CoV-2 virus in the host cell (Figure 6A). Principal component analysis (PCA) demonstrated good separation of samples according to their treatment type and high similarity between biological replicates ( Figure 6B). Principal component (PC)1 separated phenelzine-treated samples from all other samples and accounted for 66% of the variance, whilst PC2 further separated GSK-treated samples from the control samples and accounted for 12% of the variance ( Figure 6B).
  • PCA Principal component analysis
  • the Fn/c ratio (a score > than 1 indicates nuclear bias) of ACE2 also increased significantly upon infection. A similar pattern was demonstrated for LSD1.
  • RNA-seq data were obtained from Caco-2 cell line infected with SARS-CoV-2. Three different treatments were tested (named Phe, GSK, and L1), with each of the treatments targeting the same gene but in different ways. A total of 8 samples from four experimental groups were collected:
  • RNA-seq data were generated, fastq data were downloaded to the QIMR Berghofer server, and then archived to the HSM by Scott Wood. Sequence reads were trimmed for adapter sequences using Cutadapt (version 1.9; Martin (2011 )) and aligned using STAR (version 2.5.2a; Dobin et al. (2013)) to the GRCh37 assembly with the gene, transcript, and exon features of Ensembl (release 89) gene model, and the SARS-CoV-2 RefSeq accession NC_045512. Quality control metrics were computed using RNA-SeQC (version 1.1.8; DeLuca et al. (2012)) and expression was estimated using RSEM (version 1.2.30; Li and Dewey (2011)).
  • RNA-Seq samples The quality control of RNA-seq samples is an important step to guarantee quality and reproducible analytical results.
  • RNA-SeQC was run for this purpose, the results of which can be found on the HPC cluster.
  • Another common quality metric is whether the RNA sample is contaminated with mitochondrial DNA (mtDNA) or whether there is a high amount of ribosomal RNA (rRNA) in the sample.
  • mtDNA mitochondrial DNA
  • rRNA ribosomal RNA
  • the aim of normalisation is to remove differences between samples based on systematic technical effects to warrant that these technical biases have a minimal effect on the results.
  • the library size is important to correct for as differences in the initial RNA quantity sequenced will have an impact on the number of reads sequenced. Differences in RNA sequence composition occurs when RNAs are over-represented in one sample compared to others. In these samples, other RNAs will be under-sampled which will lead to higher false-positive rates when predicting differentially expressed genes.
  • HBECs were obtained from two healthy donor (#501936 and #549138) and cultured in PneumaCultTM-ALI Medium on collagen-coated transwell inserts with a 0.4-micron pore size (Costar, Corning, Tewksbury, MA, USA) and inserted into 24 well culture plates according to manufacturer instructions (StemCell Technologies, Cambridge, MA, USA). HBECs were maintained at air-liquid interface allowing them to differentiate. Medium in the basal chamber was changed every 2-3 days (500 mI).
  • HBECs 24 h prior to infection, HBECs were treated with Phenelzine (400 mM; Sigma) in the basal chamber. All infection experiments were performed in a PC3 facility. At the time of infection, the inhibitor-containing media was removed and replaced with fresh media in the basal chamber. 10 4 plaque forming units (PFU) of SARS-CoV-2 virus inoculum in 100 mI was added to the apical compartment. After 2hr of virus adsorption at 37 °C, 5% C02, the unbound virus inoculum was removed, and cells were cultured with an air-liquid interface with inhibitor-containing media in the basal chamber for 24 hr and 48 hr post infection (24 hpi and 48 hpi).
  • PFU plaque forming units
  • ASI digital pathology cells were fixed with 4% formaldehyde for 30 min at room temperature
  • Viral titers (TCID50 equivalents per ml) in the extracted RNA was determined by qRT-PCR, using Real-time fluorescent RT-PCR kit for detecting 2019- nCoV (BGI, China) following the manufacturer’s instructions. Positive control (mix of pseudo-virus with target virus genes and internal reference) and blank control (DNase/RNase free water) were used as quality control. Limit of detection is 100 copies/ml. The quantity of viral genomes was calculated by normalizing to a viral stock with a known viral titer.
  • BALCs Bronchoalveolar lavage cells
  • PBMCs peripheral blood mononuclear cells
  • a proximity ligation assay was used to assess the co-localization of ACE2 and spike protein at the surface of SARS-CoV-2 infected Caco-2 cells.
  • GSK- LSD1 treatment significantly decreased interaction between ACE2 spike protein Figure 9F. This suggests that methylation of ACE2 via inhibition of LSD1 activity contributes to blocking access to ACE2 by the SARS-CoV-2 spike protein.
  • Caco-2 cells were tested negative for Mycoplasma contamination prior to experiments. Caco-2 cells were seeded at 2 x 10 5 cells per well in six-well plates in DEME (10% FBS) and incubated overnight at 37°C, 5% CO2. 48 h prior to infection, cells were treated with either 400 mM phenelzine or 400 mM GSK-LSD1. At the time of infection, plates were transferred to the BSL3 facility and inhibitor- containing medium was removed and replaced with SARS-CoV-2 virus inoculum (MOI 1) containing DEME (5% FBS).
  • MOI 1 SARS-CoV-2 virus inoculum
  • the virus inoculum was removed and cells were washed three times with PBS to remove unbound virus prior to adding inhibitor-containing medium.
  • 48 h post infection 48 hpi
  • the cell culture supernatant and cells were collected separately in TRIzol reagent and RNA was extracted using the Direct-zol RNA miniprep kit (Zymo Research, Irvine, CA) following the manufacturer’s instructions.
  • RNA miniprep kit Zymo Research, Irvine, CA
  • Viral titers (TCID50 equivalents per ml) in the extracted RNA were determined by qRT-PCR using a real-time fluorescent RT-PCR kit for detecting 2019-nCoV (BGI Genomics, China) following the manufacturer’s instructions.
  • Positive control mixture of pseudo-virus with target virus genes and internal reference
  • blank control DNase/RNase free water
  • the limit of detection was 100 copies/ml.
  • the quantity of viral genomes was calculated by normalizing to a viral stock with a known viral titre.
  • PBMCs are pre-treated with inhibitors and killing assays performed using the xCELLigence® Real Time Cell Analyzer.
  • the Duolink proximity ligation assay was employed using PLA probe anti-mouse PLUS (DU092001), PLA probe anti-rabbit MINUS (DU092005), and Duolink In Situ Detection Reagent Red Kit (DU092008) (Sigma Aldrich). Cells were fixed, permeabilized, and incubated with primary antibodies targeting LSD1 and ACE2. Cells were processed according to the manufacturer’s recommendations. Finally, coverslips were mounted onto slides and examined as above
  • RNA will be extracted from cell supernatants and quantified by qRT- PCR (Thevarajan etai, 2020) and inhibition of viral replication assayed using a modified plaque reduction assay and EC50s.
  • the SARS-CoV-2 highly susceptible cell lines (Floffman etal., 2020) Calu-3, H1299, FlepG2, and Caco- 2 will be virus infected (MRC-5 cells as negative control).
  • SARS-CoV-2-infected cells with/without inhibitor treatments will be assayed by qRT-PCR for ACE2 and TMPRSS2 and flow cytometry and digital pathology using antibodies targeting ACE2 and TMPRSS2.
  • PBMCs will be pre-treated with inhibitors and killing assays performed using the xCELLigence® Real Time Cell Analyzer.
  • Enriched BALCs and PBMCs will be treated in vitro with 3 MOAi and FACS and ASI’s mIF digital analysis used to: (i) phenotype immune cells; (ii) quantify T cell exhaustion, proliferation, and effector markers. High-resolution single-cell RNA, ATAC-seq, and LSD1p CFIIP-seq will be performed with our bioinformatics pipeline we will identify predictive biomarkers of SARS-CoV-2 progression related to immune exhaustion.
  • Enriched BALCs and PBMCs will be profiled with our established liquid biopsy digital pathology platform for our novel EOMES exhaustion biomarker in samples taken within 48 hours of presentation and matched samples taken 7-9 and 20 days later and related to clinical progression.
  • MOA inhibitors are assessed in vivo at two doses with four animals per dose. Parameters include: Haematological, Clinical follow up (body weight, temperature), Xray scanner, blood chemistry, local and systemic reaction. Inflammation parameters, Full pathology after necropsy in case of dose lethality. This will track both the safety profile of our MOAi inhibitors as well as their effect on effect on the NHP immunological model and if MOAi inhibition is able to improve recovery from SARS-CoV-2 challenge.
  • MOA inhibitors were assessed at two doses for each. Four animals per dose are tested (8 animals per group), challenged with SARS-CoV-2. A control group with no drugs will be included. In total the study will include 4 groups with 8 animals for 3 of them and 4 animals for the control group (28 animals). We will test multiple parameters will be followed and studied at the IDMIT, to check safety and efficacy.
  • the facility is equipped with state-of-the-art instruments for in vivo imaging technologies with the following objectives: (1) track antigens and therapeutics in infected hosts; (2) visualize host responses to vaccination and/or treatment, with a specific focus on dynamics of immune effectors: (3) track microbes dissemination in the host; (4) refine and reduce using high numbers of animal models for preclinical research; (5) translate part of the technology to the clinical practice in humans.

Abstract

Disclosed are methods for treating coronavirus infections. More particularly, disclosed is the use of LSD inhibitors to treat coronavirus infections, including betacoronavirus infections.

Description

TITLE OF THE INVENTION
“METHODS FOR TREATMENT OF CORONAVIRUS INFECTIONS”
FIELD OF THE INVENTION
[0001] This invention relates generally to methods for treating coronavirus infections. More specifically, the invention relates to the use of LSD antagonists to treat coronavirus infections, including betacoronavirus infections.
BACKGROUND OF THE INVENTION
[0002] The reference in this specification to any prior publication (or information derived from it), or to any matter which is known, is not, and should not be taken as an acknowledgement or admission or any form of suggestion that the prior publication (or information derived from it) or known matter forms part of the common general knowledge in the field of endeavour to which this specification relates.
[0003] Coronaviruses are enveloped RNA viruses that infect mammals and birds. The severe acute respiratory syndrome (SARS) and the Middle East respiratory syndrome (MERS) are both members of the genus Betacoronavirus, and responsible for hundreds of deaths in Asia and the Middle East, respectively. The late 2019 emergence in China of the novel, SARS-coronavirus 2 (SARS-CoV-2) pathogen, with rapid human to human transmission and international spread, poses an immediate global health emergency. In response, a global effort for effective treatments is underway following the World Health Organisation’s (WHO) declaration of a pandemic, based on the substantial number of cases of the SARS-CoV-2 illness (COVID-19) in over 110 countries and territories in only a few months, and with a sustained risk of further global spread. There is an urgent need for both an effective coronavirus vaccine to prevent the spread of this virus and in parallel, novel therapeutic strategies to reduce the global mortality numbers, which stands currently at just around 5,000 (March 2020). This is compounded by the fact that there is no immunity in the community against this virus. Furthermore, the elderly and the sick are the most at risk with mortality due, mostly, to the weakening of their immune system.
[0004] Identifying therapeutic strategies are considered to be the fastest means of addressing this pandemic. One strategy being adopted in treatment developments is combining know drugs for other pathogenic diseases to determine any effectiveness in treating coronavirus infection. Advanced studies are progressing using combinations including an HIV drug and chloroquine (an antimalaria drug, now rarely used as the malaria pathogen has become resistant to it); and between two existing drugs lopinavir and ritonavir (see, Cao et al, 2020). However, due to unintended side effects, in addition to a lack of substantial evidence to demonstrate their efficacy in treating coronavirus infection, there is still a clear unmet clinical need to develop new treatment options specifically for coronavirus.
[0005] The coronaviruses are a virus family grouped into four genera, being the alphacoronavirus, betacoronavirus (b-CoVs), gammacoronavirus, and deltacoronavirus. The alphacoronaviruses and betacoronaviruses infect a wide range of species, including humans. In this regard, the b-CoVs that are of particular clinical importance in humans include OC43 and HKU1 of the A lineage, Severe Acute Respiratory Syndrome coronavirus (8ARS~CoV) and SARS-CoV-2 (which causes the disease COVID-19) of the B lineage, and Middle Eastern Respiratory Syndrome-related coronavirus (MERS-CoV) of the C lineage.
SUMMARY OF THE INVENTION
[0006] The present invention is predicated, at least in part, on the discovery that LSD1 plays a part in regulating key demethylation/methylation sites of the ACE2 polypeptide, which is an important part of the molecular machinery used by SARS- CoV-2 to gain entry into the host cell. Additionally, LSD1 also plays a role in demethylation/methylation of key lysine residues present on the C-terminal cytoplasmic tail of the ACE2 polypeptide, which is known to regulate the translocation of the ACE2 polypeptide from the cell membrane to the cell nucleus. Accordingly, these findings lead the present inventors to contemplate the use of LSD1 inhibitors for the treatment and/or prevention of virus infections, namely coronavirus infections. [0007] Accordingly, in one aspect of the invention provides methods of treating a coronavirus infection in a subject having a coronavirus infection, the method comprising administering to the subject an agent that inhibits or reduces an activity of a lysine-specific demethylase 1 (LSD1) polypeptide, to thereby treat the coronavirus infection in the subject.
[0008] In another aspect, the invention provides methods of preventing a coronavirus from entering a cell, the method comprising, exposing the cell to an agent that inhibits or reduces an activity of a LSD1 polypeptide , to thereby reduce or prevent the coronavirus from entering the cell.
[0009] Typically, the agent that inhibits or reduces an activity of a LSD1 polypeptide is exposed to the cell for a time and under conditions sufficient to antagonise a TMPRSS2 polypeptide. In some of the same embodiments and some alternative embodiments, the agent that inhibits or reduces an activity of a LSD1 polypeptide is exposed to the cell for a time and under conditions sufficient to antagonise a virus cell entry receptor polypeptide. In some preferred embodiments, wherein the virus cell entry receptor polypeptide is selected from the group comprising an ACE2 polypeptide, a DPP4 polypeptide, and a CD13 polypeptide.
[0010] In yet another aspect, the invention provides a method of reducing or preventing entry of a coronavirus into a cell, the method comprising exposing the cell to an agent that inhibits an activity of a lysine-specific demethylase 1 (LSD1) polypeptide, to thereby antagonise, or otherwise reduce the level or amount of TMPRSS2 polypeptide present on the surface of the cell.
[0011] In still yet another aspect, the invention provides a method of reducing or preventing entry of a coronavirus into a cell, the method comprising exposing the cell to an agent that inhibits an activity of a lysine-specific demethylase 1 (LSD1) polypeptide, to thereby antagonise, or otherwise reduce the level or amount of a virus cell entry receptor polypeptide present on the surface of the cell.
[0012] In some preferred embodiments, the agent is a selective LSD1 inhibitor.
[0013] Preferably, the agent that inhibits or reduces an activity of a LSD1 polypeptide is selected from the group comprising: a small molecule; a polypeptide; and an antigen-binding molecule. In some embodiments, the agent is a monoamine oxidase (MAO) inhibitor. In this regard, a large number of MAO inhibitor are known to be suitable for the purposes of the present invention. For example, in some embodiments, the selected from the group comprising phenelzine, bizine, tranylcypromine, pargyline, selegiline, selegiline hydrochloride, dimethylselegilene, brofaromine, moclobemide, beflozatone, safinamide, isocarboxazid, nialamide, rasagiline, iproniazide, iproclozide, toloxatone, bifemelane, desoxypeganine, harmine, harmaline, and linezolid. By way of an illustrative example, the MAO inhibitor comprises, consists, or consists essentially of, phenelzine.
[0014] In some embodiments, the agent is a selective LSD1 inhibitor. For example, the selective LSD1 inhibitor can be selected from the group comprising or consisting of GSK-LSD1 , SP-2509, and SP-2577.
[0015] In some preferred embodiments, the agent is GSK-LSD1 , which has the following molecular structure:
Figure imgf000005_0001
[0016] In some alternative preferred embodiments, the agent is SP-2509, which has the following molecular structure:
Figure imgf000005_0002
[0017] In some alternative preferred embodiments, the agent is SP-2577, which has the following molecular structure:
Figure imgf000005_0003
[0018] In some alternative preferred embodiments, the agent is bizine, which has the following molecular structure:
Figure imgf000006_0001
[0019] In some alternative embodiments, the agent is an isolated or purified proteinaceous molecule comprising, consisting or consisting essentially of sequence corresponding to residues 108 to 118 of LSD1.
[0020] In some embodiments, the isolated or purified proteinaceous molecule is an isolated or purified proteinaceous molecule represented by Formula (I):
Z1RRTX1RRKRAKVZ2 (I)
[0021] wherein:
[0022] Zi and Z2 are independently absent or are independently selected from at least one of a proteinaceous moiety comprising from about 1 to about 50 amino acid residues (and all integers in between), and a protecting moiety; and
[0023] Xi is selected from small amino acid residues, including S, T, A, G, and modified forms thereof.
[0024] In some embodiments of this type, Xi is selected from S and A.
[0025] In some of the same and other embodiments, Zi is a proteinaceous molecule represented by Formula (II):
X2X3X4 (II)
[0026] wherein: X2 is absent or is a protecting moiety; X3 is absent or is selected from any amino acid residue; and X4 is selected from any amino acid residue.
[0027] In some of the same embodiments and some other embodiments, X3 is selected from basis amino acid residues including R, K, and modified forms thereof.
[0028] In some embodiments, X4 is selected from aromatic amino acid residues, including F, Y, W, and modified forms thereof.
[0029] In some embodiments, Z2 is absent. The method according to any one of claims [0019]-[0029], wherein the isolated or purified proteinaceous molecule of Formula (I) comprises, consists or consists essentially of an amino acid sequence represented by SEQ ID NO: 1 , 2, or 3 (RRTSRRKRAKV [SEQ ID NO: 1]; RRTARRKRAKV [SEQ ID NO: 2]; or RWRRTARRKRAKV [SEQ ID NO: 3]).
[0030] In some embodiments, the proteinaceous molecule of Formula (I) further comprises at least one membrane permeating moiety. In some embodiments of this type, the membrane permeating moiety is a lipid moiety. Furthermore, the membrane permeating moiety is a myristoyl group.
[0031] Typically, the coronavirus is selected from the group comprising SARS-CoV, SARS-CoV-2, and MERS-CoV. In this regard, in some embodiments the coronavirus is SARS-CoV-2. Typically, the subject is a human.
[0032] In some embodiments, the agent that inhibits or reduces an activity of a LSD1 polypeptide is an indirect inhibitor of the LSD1 polypeptide. By way of an illustrative example, the agent that inhibits or reduces an activity of a LSD polypeptide may prevent the interaction between an LSD polypeptide and a RKOQ polypeptide. Thus, the agent prevents the phosphorylation of a phosphorylation site of the LSD polypeptide.
[0033] In yet another aspect, the present invention provides a method of treating a coronavirus infection in a subject having a coronavirus infection, the method comprising administering to the subject an agent that reduces the level of a lysine-specific demethylase (LSD) polypeptide, to thereby treat the coronavirus infection in the subject. In embodiments of this type, the agent may be an interfering nucleic acid.
[0034] In some embodiments, the LSD polypeptide is selected from an LSD1 polypeptide or an LSD2 polypeptide. In some preferred embodiments, the LSD polypeptide is an LSD1 polypeptide.
[0035] In still yet another aspect, the present invention provides a method of treating a coronavirus infection in a subject having a coronavirus infection, the method comprising administering to the subject an agent that reduces the level or activity of a CoREST polypeptide, to thereby treat the coronavirus infection in the subject. In the absence of an interaction with CoREST, it is demonstrated that LSD1 is rapidly degraded in its environment.
[0036] In still yet another aspect, the present invention provides a LSD1 inhibitor for use in the treatment of a coronavirus infection.
[0037] In still yet another aspect, the present invention provides a PKC9 inhibitor for use in the treatment of a coronavirus infection.
[0038] In some preferred embodiments, the coronavirus is a betacoronavirus. In some embodiments of this type, the betacoronavirus is SARS- CoV-2.
[0039] In some preferred embodiments of this type, the LSD1 inhibitor is selected from GSK-LSD1 , SP-2509, SP-2577, bizine, and phenelzine.
[0040] In still yet another aspect, the present invention provides the use of a LSD1 inhibitor in the manufacture of a medicament for the prevention and/or treatment of a coronavirus infection (e.g., a betacoronavirus infection).
[0041] In yet another aspect, the present invention provides compositions for treating coronavirus, comprising (i) an agent that inhibits or reduces an activity of a lysine-specific demethylase-1 (LSD1) polypeptide; and (ii) an antiviral agent.
[0042] In some embodiments of this type, the antiviral agent is selected from the group comprising hydroxychloroquine, chloroquine, lopinavir, ritonavir, favipiravir, and remdesivir.
[0043] In some of the same embodiments and some alternative embodiments, the antiviral agent comprises an IFN-b polypeptide.
[0044] In yet another aspect, the present invention provides methods of inhibiting the phosphorylating activity of a protein kinase C (PKC), comprising contacting a cell infected with a coronavirus with an isolated or purified proteinaceous molecule comprising, consisting or consisting essentially of a sequence corresponding to residues 108 to 118 of LSD1.
[0045] In still yet another aspect, the present invention provides methods of preventing S protein priming by a coronavirus, the method comprising administering a TMPRSS2 antagonist to a cell infected by the coronavirus, wherein the TMPRSS2 antagonist is an inhibitor of at least one interaction between an LSD1 polypeptide and another polypeptide (e.g., a PKC0 polypeptide).
[0046] In yet another aspect, the present invention provides a method of preventing or reducing coronavirus entry into a cell, the method comprising exposing the cell to an LSD1 inhibitor, wherein the LSD1 inhibitor reduces the expression of ACE2 by the cell, to thereby inhibit coronavirus entry into the cell.
BRIEF DESCRIPTION OF THE FIGURES
[0047] Figure 1 provides a graphical representations showing the role of LSD1 on virus entry. (A) Nanostring analysis of TMPRSS2 expression in MDA- MB-231 (TNBC breast cancer cell line) or MCF7 (epithelial breast cancer cell line) treated with siRNA targeting LSD1 or control siRNA. (B) Nanostring analysis of TMPRSS2 expression in MDA-MB-231 (TNBC breast cancer cell line) MCF7 (epithelial breast cancer cell line), CT26 (Colorectal cancer cell line) or Huh7 (liver cancer cell line). Treated with vehicle control, GSK-LSD1 (catalytic LSD1 inhibitor) or Phenelzine (Dual catalytic/nuclear axis LSD1p inhibitor). (C) CHiP enrichment of TMPRSS2 in MDA-MB-231 (TNBC breast cancer cell line) or Huh7 (liver cancer cell line). The y-AXIS plots the Chip enrichment ratio that is relative to the no-antibody control (mean ± S.E.M., n = 3). *P < 0.05, **P < 0.01 , ***P < 0.001 , ****P < 0.0001 (one-tailed Student’s t-test). Samples were treated with vehicle control, C27 (catalytic PKC0 inhibitor) or phenelzine (dual catalytic/nuclear axis LSD1p inhibitor). (D) Digital microscopy was performed on cells fixed and probed with primary anti- TMPRSS2 antibodies and DAPI. Graph represents the mean fluorescent values for TMPRSS2 measured using ImageJ to select the nucleus minus background (n > 50 individual cells). Data represents 3 separate experiments, Mann-Whitney T-test in PRISM was used to determine significant differences. **P < 0.01 , ***P < 0.001 ,
****P £ 0.0001.
[0048] Figure 2 shows that LSD1 and ACE2 associate as a complex on cell surface in SARS-CoV-2 susceptible cells. (A) Representative images of CaCo2 cells imaged with the ASI digital pathology system. Cells are either permeabilized (intracellular) or not permeabilized (surface) and stained for expression of ACE2, LSD1 and TRMPSS2. Scale bar represents 10 pm. (B) Dot graphs display the nuclear fluorescence intensity in Caco-2 cells for ACE2, LSD1 and TRMPSS2 from (A). > 50 cells counted per group. Data represent mean ± SE. Mann-Whitney-test. **p < 0.01 , ***p < 0.001 , ****p < 0.0001 denote significant differences n.s. denotes non-significant. The PCC(r) was calculated for LSD1 and ACE2, (n = 20 individual cells). -1 = inverse of colocalization; 0 = no colocalization;
+1 = perfect colocalization. (C) Representative FACS plot showing the cell surface and intracellular expression of ACE2 and LSD1 in Caco-2 cells. The numbers in each quadrant indicate the percentage of the total cell population, which also shown in dot plot (D). Data in dot plot represent two independent biological replicates. (E) Representative image of MRC5 cells imaged with the ASI digital pathology system, that are either permeabilized (intracellular) or not permeabilized (surface) and stained for expression of ACE2, LSD1 and TRMPSS2. Scale bar represents 10 pm. (F) Dot graphs displays the nuclear fluorescence intensity in MRC5 cells for ACE2, LSD1 and TRMPSS2 from (E). > 50 cells counted per group. Data represent mean ± SE. Mann-Whitney-test. **p < 0.01 , ***p < 0.001 , ****p < 0.0001 denote significant differences n.s. denotes non-significant.
[0049] Figure 3 provides graphical representation showing LSD1 and ACE2 have increased association on the cell surface in SARS-CoV-2 infected cells. (A, B) Representative image of Caco-2-SARS-CoV-2 infected cells imaged with the ASI digital pathology system (MRC5/Caco-2 uninfected not show) are shown, cells were either (A) permeabilized (intracellular) with 0.5% Triton X-100 for 15 minutes or (B) not permeabilized (surface) and stained for with primary antibodies against ACE2, LSD1 and SARS-CoV-2 nucleocapsid protein. Scale bar represents 12 mm. Dot graphs displays the nuclear fluorescence intensity in Caco-2 cells for ACE2, LSD1 and SARS-CoV-2 from (A). >50 cells counted per group. Data represent mean ± SE. Mann-Whitney-test. **p < 0.01 , ***p < 0.001 , ****p < 0.0001 denote significant differences n.s. denotes non-significant. The PCC(r) was calculated for LSD1 and ACE2 or LSD1 and SARS-CoV-2, (n = 20 individual cells).
-1 = inverse of colocalization; 0 = no colocalization; +1 = perfect colocalization. (C) Representative image of Caco-2 or Caco-2-SARS-CoV-2 infected cells imaged with the ASI digital pathology system are shown, cells were either permeabilized (intracellular) with 0.5% Triton X-100 for 15 minutes or not permeabilized (surface) and stained for with primary antibodies against ACE2, LSD1 and SARS-CoV-2 SPIKE protein. Scale bar represents 12 mm. Dot graphs displays the nuclear fluorescence intensity in Caco-2 cells for ACE2, LSD1 and SARS-CoV-2 from (C). > 50 cells counted per group. Data represent mean ± SE. Mann-Whitney-test. **p < 0.01 , ***p < 0.001 , ****p < 0.0001 denote significant differences n.s. denotes non significant. The PCC(r) was calculated for LSD1 and ACE2 or LSD1 and SARS-CoV-2, (n = 20 individual cells). -1 = inverse of colocalization; 0 = no co-localization; +1 = perfect colocalization. (D) qRT-PCR analysis to detect the growth kinetics of SARS-CoV-2 in Caco-2 and MRC5 culture supernatant at indicated time points after viral infection. The dotted line indicates the limit of detection. (E) FACS analysis of the expression of SARS-CoV-2 nucleocapsid protein, cell surface ACE2 and intracellular LSD1 in Caco-2 cells after 48 hours post infection. The unit of / axis indicates the percentage of the total cell population. Data represent mean ± SD, n = 2. (F) Representative image of Caco-2 or Caco-2-SARS- CoV-2 infected cells imaged with the ASI digital pathology system are shown, cells were permeabilized (intracellular) with 0.5% Triton X-100 for 15 minutes and stained for with primary antibodies against FI3k9me2 and FI3k4me2. Scale bar represents 12 mm. (G) Dot graphs displays the nuclear fluorescence intensity in CaCo2 cells for ACE2, LSD1 and SARS-CoV-2 from (F). >50 cells counted per group. Data represent mean ± SE. Mann-Whitney-test. **p < 0.01 , ***p < 0.001 , ****p < 0.0001 denote significant differences n.s. denotes non-significant. (H, I) Representative image of CaCo2 or CaCo2-SARS-CoV-2 infected cells imaged with the ASI digital, pathology system are shown, cells were either (FI) not permeabilized (surface) or (I) permeabilized (intracellular) with 0.5% Triton X-100 for 15 minutes or and stained for with primary antibodies against SETDB1 , G9A and ACE2. Scale bar represents 12 mm. (J) Dot graphs displays the nuclear fluorescence intensity in Caco-2 cells for ACE2, LSD1 and SARS-CoV-2 from (FI, I). >50 cells counted per group. Data represent mean ± SE. Mann-Whitney-test. **p < 0.01 , ***p < 0.001 , ****p < 0.0001 denote significant differences n.s. denotes non-significant.
[0050] Figure 4 provides graphical representations showing LSD1 directly interacting with the ACE2 cytoplasmic tail that harbours high affinity LSD1 demethylation domain. (A, B) Representative image of Caco-2 cells imaged with the ASI digital pathology system are shown. Caco-2 cells were treated with vehicle control or 200 mM of phenelzine and imaged with the ASI digital pathology system are shown, cells were either (A) permeabilized (intracellular) with 0.5% Triton X-100 for 15 minutes or (B) non-permeabilized (surface) and stained for with primary antibodies against ACE2, LSD1 and SARS-CoV-2 nucleocapsid protein. Scale bar represents 12 mm. (C, D) Dot graphs displays the nuclear fluorescence intensity in Caco-2 cells for ACE2, LSD1 and SARS-CoV-2 from (A, B, respectively). >50 cells counted per group. Data represent mean ± SE. Mann-Whitney-test. **p < 0.01 , ***p
< 0.001 , ****p < 0.0001 denote significant differences n.s. denotes non-significant. The PCC(r) was calculated for LSD1 and ACE2 (n = 20 individual cells). -1 = inverse of colocalization; 0 = no colocalization; +1 = perfect colocalization.
[0051] Figure 5 provides a graphical representation of LSD1 inhibitors abrogating ACE2 expression and inhibiting SARS-CoV-2 expression. (A)
Representative image of Caco-2-SARS-CoV-2 infected cells treated with phenelzine (P400 mM), GSK-LSD1 (G400 mM) or EPI-111 (50 mM) imaged with the ASI digital pathology system are shown, cells were stained for with primary antibodies against ACE2 and LSD1. Scale bar represents 15 mm. (B) Dot graphs displays the nuclear fluorescence intensity in Caco-2 cells for ACE2, LSD1. >50 cells counted per group. Data represent mean ± SE. Mann-Whitney-test. **p < 0.01 , ***p < 0.001 , ****p
< 0.0001 denote significant differences n.s. denotes non-significant. The PCC(r) was calculated for LSD1 and ACE2 (n = 20 individual cells). -1 = inverse of colocalization; 0 = no colocalization; +1 = perfect colocalization. (C) Representative image of CaCo2-SARS-CoV-2 infected cells treated with phenelzine (P400 mM), GSK (G400 mM) or EPI-111 (50 mM) imaged with the ASI digital pathology system are shown, cells were permeabilized with 0.5% Triton X-100 for 15 minutes and stained for with primary antibodies against ACE2, LSD1 and SARS-CoV-2. Scale bar represents 15 mm. (D) Dot graphs displays the nuclear fluorescence intensity in Caco-2 cells for ACE2, LSD1 and SARS-CoV-2. >50 cells counted per group. Data represent mean ± SE. Mann-Whitney-test. **p < 0.01 , ***p < 0.001 , ****p < 0.0001 denote significant differences n.s. denotes non-significant. The PCC(r) was calculated for LSD1 and ACE2 or LSD1 and SARS-CoV-2 (n = 20 individual cells).
-1 = inverse of colocalization; 0 = no colocalization; +1 = perfect colocalization. (E) From the samples from (C) The Plot profile function of ImageJ-Fiji was employed to determine the fluorescent intensity along a line drawn around the surface/cytoplasm of the cell. Comparing the intensities of LSD1 , ACE2 and SARS-CoV-2. This was done for each sample from (C) above. (F) Duolink® proximity ligation assay measurements of protein interactions were performed on Caco-2 cells infected with SASR-CoV-2 and treated with Control or Nardil® 400 mM, cells were not permeabilized and subject to Duolink® Assay which produces a single bright dot per interaction within the cell. Representative images are show for the following pairs: LSD1 & ACE2. PLA signal intensity of the Duolink® assay is shown for average dot intensity (single Duolink® dot) or overall cell intensity for each cell (G) Duolink® proximity ligation assay measurements of protein interactions were performed on Caco-2 cells infected with SARS-CoV-2 and treated with Control, Nardil® 400 mM, or GSK-LSD1 400 mM, permeabilized with 0.5% Triton X-100 and subject to Duolink® assay which produces a single bright dot per interaction within the cell. Representative images are show for the following pairs: LSD1 & ACE2. PLA signal intensity of the Duolink® assay is shown for average dot intensity (single Duolink® dot) or overall cell intensity for each cell. (H) qRT-PCR analysis of ACE2 and TMPRSS2 expression in SARS-CoV-2 infected Caco-2 cells treated with phenelzine (400 mM), GSK-LSD1 (400 pM) and L1 (50 pM) at 48 hour post-infection. Data were normalized to Geomean of GAPDH, HPRT1 and ACTB. Data represent mean ± SD, n = 2. One-way ANOVA, *p < 0.05, **p < 0.01 denote significant differences. (I) qRT-PCR analysis of SARS-Cov-2 infected Caco-2 cells treated with/without Phenelzine (400 pM for 72 hr). One-way ANOVA, * p < 0.05, *** p < 0.001 versus Control. (K) qRT-PCR analysis of SARS-Cov-2 infected Caco-2 cells treated with/without phenelzine, GSK-LSD, L1 (EPI-111 ) or ACE2 peptide inhibitor (P604). One-way ANOVA, * p < 0.05, *** p < 0.001 versus Control. (J) qRT-PCR analysis of IFNa, IFNp, DDX58 (RIG-1), IFIH1 (MDA-5), ISG15, and OASL mRNA expression in uninfected versus SARS-CoV-2-infected control (depicted in Figure 2), phenelzine (400 pM), and GSK (400 pM) treated Caco-2 cells 48 hpi. The graph illustrates the number of gene transcripts from three replicates normalized to the geometric mean of HPRT1 , GAPDFI, and ACTB. Statistical significance was calculated using one-way ANOVA. NS, not significant.
[0052] Figure 6 provides a graphical representation of a global transcript analysis. (A) Caco-2 cells were treated with phenelzine, GSK-LSD1 or L1 and global RNA transcriptome analysis shows that key anti-viral and transcription processes are impacted. The heat map above focuses on a DEGs list related to ISG, IFN-I, cytokine/chemokine activity, and viral entry, nuclear import/RNA synthesis, translation and replication. The heatmap graph depicts the log2 (fold change) of DEGs of inhibition treated compared with control cells. Those selected DEGs have a log2(fold change) of more than 1 and FDR value of less than 0.01. (B) Principal component analysis (PCA) depicting transcriptional profiles for control, GSK-LSD1 , and phenelzine groups after batch effect removal. Experimental batches represented by different shapes (circles, batch 1 ; triangles, batch 2). (C) Euler diagram comparing DEGs from GSK-LSD1 vs. control (green) and phenelzine vs. control (blue). (D) Fleatmap of DEGs belonging to Reactome pathways: R-HSA-913531 interferon signalling (left); MAPK signalling-related pathways (R-FISA-5683057 MAPK family signalling cascades, R-HSA-5673001 RAF/MAP kinase cascade, and R-HSA-5684996 MAPK1/MAPK3 signalling; middle); and translation related pathways (R-HSA-70614 amino acid synthesis and interconversion (transamination), R-HSA-8957275 post-translational protein phosphorylation; right). The heatmap values depict the log2-fold change (logFC) of DEGs from treated cells compared with control cells (GSK-LSD1 vs. control and phenelzine vs. control). (E) Dot plot visualization of the top enriched Reactome pathways in treated cells compared to control cells. The dot colour represents the false discovery rate (FDR) value for each enriched Reactome pathway and size represents the gene ratio. (F) Dot plot visualization of enriched Reactome pathways for GSK-LSD1 vs. control (left) and phenelzine vs. control (right). The dot colour represents the false discovery rate (FDR) value for each enriched Reactome pathway and size represents the gene ratio.
[0053] Figure 7 shows a graphical representation of the interplay of intracellular ACE2 in infected cells. (A) Representative image of Caco-2 or MRC5 SARS-CoV-2 infected cells are depicted. Scale bar represents 15 mm. (B) Cells were permeabilized and imaged with the ASI digital pathology system are shown, cells were stained for with primary antibodies against SARS-CoV-2 (nucleocapsid), ACE2 and LSD1. Dot graphs displays the nuclear fluorescence intensity in Caco-2 cells for ACE2, LSD1. 20 or more cells counted per group. Data represent mean ± SEM. Mann-Whitney-test. **p < 0.01 , ***p < 0.001 , ****p < 0.0001 denote significant differences n.s. denotes non-significant. The nuclear/cytoplasmic fluorescence ratio (Fn/c) using the equation: Fn/c = (Fn- Fb)/(Fc - Fb), where Fn is nuclear fluorescence, Fc is cytoplasmic fluorescence, and the dotted line indicates background fluorescence. The Mann-Whitney nonparametric test (GraphPad Prism, GraphPad Software, San Diego, CA) was used to determine significant differences between datasets. (C) Cells were not permeabilized to track surface expression and stained for with primary antibodies against ACE2 and LSD1. Dot graphs displays the nuclear fluorescence intensity in Caco-2 cells for ACE2, LSD1. 20 or more cells counted per group. Data represent mean ± SEM. Mann-Whitney-test. *p < 0.0181 , **p < 0.01 , ***p < 0.001 , ****p < 0.0001 denote significant differences n.s. denotes non-significant. The nuclear/cytoplasmic fluorescence ratio (Fn/c) using the equation: Fn/c = (Fn- Fb)/(Fc - Fb), where Fn is nuclear fluorescence, Fc is cytoplasmic fluorescence, and the dotted line indicates background fluorescence. The Mann- Whitney nonparametric test (GraphPad Prism, GraphPad Software, San Diego, CA) was used to determine significant differences between datasets.
[0054] Figure 8 shows graphical representations of ACE2 peptide inhibitor effect on nucleocapsid and Spike protein of SARS-Cov-2. (A)
Representative image of Caco-2-SARS-CoV-2 infected cells treated with phenelzine (P400 mM), GSK-LSD1 (G400 mM), L1 (50 mM) or P604 (ACE2 peptide 50 mM) imaged with the ASI digital pathology system are shown, Scale bar represents 15 mm. (B) Cells were permeabilized with 0.5% Triton X-100 for 15 minutes and stained for with primary antibodies against ACE2, TMPRSS2 and SARS-CoV-2 Spike Protein. Dot graphs displays the nuclear fluorescence intensity in Caco-2 cells for ACE2, TMPRSS2 and SARS-CoV-2 Spike Protein. >50 cells counted per group.
Data represent mean ± SEM. Mann-Whitney-test. **p < 0.01 , ***p < 0.001 , ****p < 0.0001 denote significant differences n.s. denotes non-significant. The PCC(r) was calculated for ACE2 and SARS-CoV-2. (C) Representative image of Caco-2-SARS- CoV-2 infected cells treated with phenelzine (P400 mM), GSK-LSD1 (G400 mM), L1 (50 mM) or P604 (ACE2 peptide 50 mM) imaged with the ASI digital pathology system are shown, Scale bar represents 15 mm. (D) Cells were permeabilized with 0.5% T riton X-100 for 15 minutes and stained for with primary antibodies against ACE2, TMPRSS2 and SARS-CoV-2 nucleocapsid protein. Dot graphs displays the nuclear fluorescence intensity in Caco-2 cells for ACE2, TMPRSS2 and SARS- CoV-2 nucleocapsid. >50 cells counted per group. Data represent mean ± SEM. Mann-Whitney-test. **p < 0.01 , ***p < 0.001 , ****p < 0.0001 denote significant differences n.s. denotes non-significant.
[0055] Figure 9 shows a graphical representation showing HBEC ALI cell culture with SARS-CoV-2 infection. HBECs were pre-treated with and without Phenelzine (400 mM) for 24hr, followed by SARS-CoV-2 infection (104 PFU) for 2 hr. Virus was removed by HBSS wash. Infected HEBCs were cultured with and without Phenelzine (400 mM) for 24 hr and 48 hr post-infection. qRT-PCR analysis was performed to detect the replicates of SARS-CoV-2 in infected cells at indicated time points of post-infection. The limit of detection is 100 copies/ml. a Figure 10 provides graphical representations of LSD1-ACE2 interactions and the effect of LSD1 on the spike protein. (A) Dot plot quantification of the fluorescence intensity (cell surface) of SARS-CoV-2 spike protein in SARS-CoV-2- infected Caco-2 cells with phenelzine or GSK treatment. >50 cells were analyzed for each group and were quantified using the digital pathology assay (ASI system).
Mann-Whitney test: *p < 0.05, ****p < 0.0001. (B) Dot plot quantification of the fluorescence intensity (cell surface) of ACE2 and LSD1 in SARS-CoV-2-infected
Caco-2 cells with phenelzine or GSK-LSD1 treatment. >50 cells were analyzed for each group and were quantified with a digital pathology assay (ASI system). Mann-
Whitney-test: **p < 0.01 , ****p < 0.0001. (C) qRT-PCR analysis of LSD1, ACE2,
TMPRSS2 mRNA expression in uninfected versus SARS-CoV-2-infected control, phenelzine (400 mM), and GSK-LSD1 (400 pM) treated Caco-2 cells 48 hpi. The graph illustrates the number of gene transcripts from three replicates normalized to the geometric mean of HPRT1, GAPDH, and ACTB. Statistical significance was calculated using one-way ANOVA. NS, not significant. (D) Schematic of SARS-CoV-2 infection assays. Caco-2 cells were seeded 24 h before the experiment. Then, cells were treated with each drug component for 48 h followed by SARS-CoV-2 infection
(MOI 1.0). After 1 h viral adsorption incubation, the virus inoculum was removed and drug-containing medium was added. Then, cell culture supernatants were harvested at 0, 24, or 48 hpi and infected cells were collected at 48 hpi. Detection of viral genomes in the extracted RNA was performed by qRT-PCR, and viral spike proteins were quantified by a digital pathology assay (ASI system). Duolink® proximity ligation assay measurements of protein interactions were performed on unpermeabilized Caco-2 cells infected with SASR-CoV-2 and treated with control or GSK-LSD1. (E) The Duolink assay produces a single bright dot per interaction within the cell. Representative images (left) are shown for ACE2 and pan-methylation lysine antibody. PLA signal intensity of the Duolink® assay (right) is shown for average dot intensity (single Duolink dot) or overall cell intensity for each cell. Data represent n = 20 cells, with significant differences calculated with the unpaired f-test (****p < 0.0001). Representative images are shown with 10 mM scale bar in orange. (F) Duolink® proximity ligation assay measurements of protein interactions were performed on unpermeabilized Caco-2 cells infected with SARS-CoV-2 and treated with control, or GSK inhibitors. The Duolink assay produces a single bright dot per interaction within the cell. Representative images (left) are shown for ACE2 and SARS-CoV-2 Spike Duolink®. PLA signal intensity of the Duolink® assay (right) is shown for average dot intensity (single Duolink® dot). Data represent n=20 cells, with significant differences calculated with Kruskal-Wallis ANOVA (* p < 0.05, ****p < 0.0001). Representative images are shown with 10 mM scale bar in orange.
DETAILED DESCRIPTION OF THE INVENTION
1. Definitions
[0056] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by those of ordinary skill in the art to which the invention belongs. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, preferred methods and materials are described. For the purposes of the present invention, the following terms are defined below.
[0057] The articles “a” and “an” are used herein to refer to one or to more than one (i.e., to at least one) of the grammatical object of the article. By way of example, “a cell” means one cell or more than one cell.
[0058] The term “about” as used herein refers to the usual error range for the respective value readily known to the skilled person in this technical field. Reference to “about” a value or parameter herein includes (and describes) embodiments that are directed to that value or parameter per se.
[0059] The terms “administration concurrently” or “administering concurrently” or “co-administering” and the like refer to the administration of a single composition containing two or more actives, or the administration of each active as separate compositions and/or delivered by separate routes either contemporaneously or simultaneously or sequentially within a short enough period of time that the effective result is equivalent to that obtained when all such actives are administered as a single composition. By “simultaneously” is meant that the active agents are administered at substantially the same time, and desirably together in the same formulation. By “contemporaneously” it is meant that the active agents are administered closely in time, e.g., one agent is administered within from about one minute to within about one day before or after another. Any contemporaneous time is useful. However, it will often be the case that when not administered simultaneously, the agents will be administered within about one minute to within about eight hours and suitably within less than about one to about four hours. When administered contemporaneously, the agents are suitably administered at the same site on the subject. The term “same site” includes the exact location, but can be within about 0.5 to about 15 cm, preferably from within about 0.5 to about 5 cm. The term “separately” as used herein means that the agents are administered at an interval, for example at an interval of about a day to several weeks or months. The active agents may be administered in either order. The term “sequentially” as used herein means that the agents are administered in sequence, for example at an interval or intervals of minutes, hours, days or weeks. If appropriate the active agents may be administered in a regular repeating cycle. [0060] The term “agent” includes a compound that induces a desired pharmacological and/or physiological effect. The term also encompasses pharmaceutically acceptable and pharmacologically active ingredients of those compounds specifically mentioned herein including but not limited to salts, esters, amides, prodrugs, active metabolites, analogues and the like. When the above term is used, then it is to be understood that this includes the active agent perse as well as pharmaceutically acceptable, pharmacologically active salts, esters, amides, prodrugs, metabolites, analogues, etc. The term “agent” is not to be construed narrowly but extends to small molecules, proteinaceous molecules such as peptides, polypeptides and proteins as well as compositions comprising them and genetic molecules such as RNA, DNA and mimetics and chemical analogues thereof as well as cellular agents. The term “agent” includes a cell that is capable of producing and secreting a polypeptide referred to herein as well as a polynucleotide comprising a nucleotide sequence that encodes that polypeptide. Thus, the term “agent” extends to nucleic acid constructs including vectors such as viral or non-viral vectors, expression vectors and plasmids for expression in and secretion in a range of cells.
[0061] The “amount” or “level” of a biomarker is a detectable level in a sample. These can be measured by methods known to one skilled in the art and also disclosed herein. The expression level or amount of biomarker assessed can be used to determine the response to treatment.
[0062] As used herein, “and/or” refers to and encompasses any and all possible combinations of one or more of the associated listed items, as well as the lack of combinations when interpreted in the alternative (or).
[0063] The term “antagonist” or “inhibitor” refers to a substance that prevents, blocks, inhibits, neutralizes, or reduces a biological activity or effect of another molecule, such as a receptor.
[0064] As use herein, the term “binds”, “specifically binds to” or is “specific for” refers to measurable and reproducible interactions such as binding between a target and a binding molecule, which is determinative of the presence of the target in the presence of a heterogeneous population of molecules including biological molecules. For example, binding molecule that binds to or specifically binds to a target (which can be an epitope) is a molecule that binds this target with greater affinity, avidity, more readily, and/or with greater duration than it binds to other targets. In one embodiment, the extent of binding of a binding molecule to an unrelated target is less than about 10% of the binding of the molecule to the target as measured, e.g., by a radioimmunoassay (RIA). In certain embodiments, a binding molecule that specifically binds to a target has a dissociation constant (Kd) of <1 mM, <100 nM, <10 nM, <1 nM, or <0.1 nM. In certain embodiments, a binding molecule specifically binds to a region on a protein that is conserved among the protein from different species. In another embodiment, specific binding can include, but does not require exclusive binding.
[0065] As used herein, the term “binding agent” refers to an agent that binds to a target antigen and does not significantly bind to unrelated compounds. Examples of binding agents that can be effectively employed in the disclosed methods include, but are not limited to, lectins, proteins, and antibodies, such as monoclonal antibodies, chimeric antibodies, or polyclonal antibodies, or antigen binding fragments thereof, as well as aptamers, Fc domain fusion proteins, and aptamers having or fused to hydrophobic protein domain, e.g., Fc domain, etc. In an embodiment the binding agent is an exogenous antibody. An exogenous antibody is an antibody not naturally produced in a mammal, e.g. in a human, by the mammalian immune system.
[0066] As used herein, the term “complex” refers to an assemblage or aggregate of molecules {e.g., peptides, polypeptides, etc.) in direct or indirect contact with one another. In specific embodiments, “contact”, or more particularly, “direct contact” means two or more molecules are close enough so that attractive noncovalent interactions, such as Van der Waal forces, hydrogen bonding, ionic and hydrophobic interactions, and the like, dominate the interaction of the molecules. In such embodiments, a complex of molecules {e.g., a peptide and polypeptide) is formed under conditions such that the complex is thermodynamically favoured {e.g., compared to a non-aggregated, or non-complexed, state of its component molecules). The term “polypeptide complex” or “protein complex,” as used herein, refers to a trimer, tetramer, pentamer, hexamer, heptamer, octamer, nonamer, decamer, undecamer, dodecamer, or higher order oligomer.
[0067] Throughout this specification, unless the context requires otherwise, the words “comprise”, “comprises” and “comprising” will be understood to imply the inclusion of a stated step or element or group of steps or elements but not the exclusion of any other step or element or group of steps or elements. Thus, use of the term “comprising” and the like indicates that the listed elements are required or mandatory, but that other elements are optional and may or may not be present. By “consisting of” is meant including, and limited to, whatever follows the phrase “consisting of”. Thus, the phrase “consisting of” indicates that the listed elements are required or mandatory, and that no other elements may be present. By “consisting essentially of” is meant including any elements listed after the phrase, and limited to other elements that do not interfere with or contribute to the activity or action specified in the disclosure for the listed elements. Thus, the phrase “consisting essentially of” indicates that the listed elements are required or mandatory, but that other elements are optional and may or may not be present depending upon whether or not they affect the activity or action of the listed elements.
[0068] By “corresponds to” or “corresponding to” is meant an amino acid sequence that displays substantial sequence similarity or identity to a reference amino acid sequence. In general the amino acid sequence will display at least about 70, 71 , 72, 73, 74, 75, 76, 77, 78, 79, 80, 81 , 82, 83, 84, 85, 86, 97, 88, 89, 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99% or even up to 100% sequence similarity or identity to at least a portion of the reference amino acid sequence.
[0069] An “effective amount” is at least the minimum amount required to effect a measurable improvement or prevention of a particular disorder. An effective amount herein may vary according to factors such as the disease state, age, sex, and weight of the patient, and the ability of the antibody to elicit a desired response in the individual. An effective amount is also one in which any toxic or detrimental effects of the treatment are outweighed by the therapeutically beneficial effects. For prophylactic use, beneficial or desired results include results such as eliminating or reducing the risk, lessening the severity, or delaying the onset of the disease, including biochemical, histological and/or behavioural symptoms of the disease, its complications and intermediate pathological phenotypes presenting during development of the disease. For therapeutic use, beneficial or desired results include clinical results such as decreasing one or more symptoms resulting from the disease, increasing the quality of life of those suffering from the disease, decreasing the dose of other medications required to treat the disease, enhancing effect of another medication such as via targeting, delaying the progression of the disease, and/or prolonging survival. In the case of an infection, an effective amount of the drug may have the effect in reducing pathogen (bacterium, virus, etc.) titres in the circulation or tissue; reducing the number of pathogen infected cells; inhibiting (/. e., slow to some extent or desirably stop) pathogen infection of organs; inhibit (/.e., slow to some extent and desirably stop) pathogen growth; and/or relieving to some extent one or more of the symptoms associated with the infection. An effective amount can be administered in one or more administrations. For purposes of this invention, an effective amount of drug, compound, or pharmaceutical composition is an amount sufficient to accomplish prophylactic or therapeutic treatment either directly or indirectly. As is understood in the clinical context, an effective amount of a drug, compound, or pharmaceutical composition may or may not be achieved in conjunction with another drug, compound, or pharmaceutical composition. Thus, an “effective amount” may be considered in the context of administering one or more therapeutic agents, and a single agent may be considered to be given in an effective amount if, in conjunction with one or more other agents, a desirable result may be or is achieved.
[0070] An “effective response” of a patient or a patient’s “responsiveness” to treatment with a medicament and similar wording refers to the clinical or therapeutic benefit imparted to a patient at risk for, or suffering from, a disease or disorder, such as a pathogenic infection. In one embodiment, such benefit includes any one or more of: extending survival (including overall survival and progression free survival); resulting in an objective response (including a complete response or a partial response); or improving signs or symptoms of the pathogenic infection. A patient who “does not have an effective response” to treatment refers to a patient who does not have any one of extending survival (including overall survival and progression free survival); resulting in an objective response (including a complete response or a partial response); or improving signs or symptoms of the pathogenic infection.
[0071] The term “expression” with respect to a gene sequence refers to transcription of the gene to produce a RNA transcript ( e.g ., mRNA, antisense RNA, siRNA, shRNA, miRNA, etc.) and, as appropriate, translation of a resulting mRNA transcript to a protein. Thus, as will be clear from the context, expression of a coding sequence results from transcription and translation of the coding sequence. Conversely, expression of a non-coding sequence results from the transcription of the non-coding sequence.
[0072] The terms “level of expression” or “expression level” in general are used interchangeably and generally refer to the amount of a biomarker in a sample. “Expression” generally refers to the process by which information ( e.g ., gene- encoded and/or epigenetic) is converted into the structures present and operating in the cell. Therefore, as used herein, “expression” may refer to transcription into a polynucleotide, translation into a polypeptide, or even polynucleotide and/or polypeptide modifications {e.g., posttranslational modification of a polypeptide). Fragments of the transcribed polynucleotide, the translated polypeptide, or polynucleotide and/or polypeptide modifications {e.g., post-translational modification of a polypeptide) shall also be regarded as expressed whether they originate from a transcript generated by alternative splicing or a degraded transcript, or from a post translational processing of the polypeptide, e.g., by proteolysis. “Expressed genes” include those that are transcribed into a polynucleotide as mRNA and then translated into a polypeptide, and also those that are transcribed into RNA but not translated into a polypeptide {e.g., transfer and ribosomal RNAs).
[0073] “Elevated expression”, “elevated expression levels”, or “elevated levels” refers to an increased expression or increased levels of a biomarker in an individual or part of an individual {e.g., a cell, tissue or organ) relative to a control, such as an individual or individuals who are not suffering from the disease or disorder {e.g., T cell dysfunctional disorder) or parts thereof {e.g., a cell, tissue or organ) or an internal control {e.g., housekeeping biomarker).
[0074] “Reduced expression”, “reduced expression levels”, or “reduced levels” refers to a decreased expression or decreased levels of a biomarker in an individual or part of an individual {e.g., a cell, tissue or organ) relative to a control, such as an individual or individuals who are not suffering from the disease or disorder {e.g., T-cell dysfunctional disorder) or parts thereof {e.g., a cell, tissue or organ) or an internal control {e.g., housekeeping biomarker). In some embodiments, reduced expression is little or no expression. [0075] The term “infection” refers to invasion of body tissues by disease- causing microorganisms, their multiplication and the reaction of body tissues to these microorganisms and the toxins they produce. “Infection” includes but are not limited to infections by viruses, prions, bacteria, viroids, parasites, protozoans and fungi. In the context of the present invention, however, “infection” generally refers to virus infection of the family Coronnavitidae ( e.g ., coronaviruses);
[0076] As used herein, “instructional material” includes a publication, a recording, a diagram, or any other medium of expression which can be used to communicate the usefulness of the compositions and methods of the invention. The instructional material of the kit of the invention may, for example, be affixed to a container which contains the therapeutic or diagnostic agents of the invention or be shipped together with a container which contains the therapeutic or diagnostic agents of the invention.
[0077] As used herein, the term “LSD1” refers to any form of LSD1 and variants thereof that retain at least part of the activity of LSD1. Unless indicated differently, such as by specific reference to human LSD1 , LSD1 includes all mammalian species of native sequence LSD1, e.g., human, canine, feline, equine, and bovine. One exemplary human LSD1 is found as UniProt Accession Number 060341.
[0078] As used herein, the term “LSD2” refers to any form of LSD2 and variants thereof that retain at least part of the activity of LSD2. Unless indicated differently, such as by specific reference to human LSD2, LSD2 includes all mammalian species of native sequence LSD2, e.g., human, canine, feline, equine, and bovine. One exemplary human wild-type LSD2 peptide sequence is deposited as UniProt Accession Number Q8NB78.
[0079] The term “LSD inhibitor”, “LSD antagonist” and the like, refer to a molecule that decreases, blocks, inhibits, abrogates or interferes with the epigenetic function of an LSD (e.g., LSD1 or LSD2). In some embodiments, the LSD inhibitor is a direct inhibitor of LSD. In some embodiments, the LSD inhibitor is a molecule that inhibits the binding of LSD to one or more of its binding partners. In a specific aspect, the LSD inhibitor inhibits the binding of the LSD binding partner PKC0 to LSD. Alternatively, the LSD inhibitor is an indirect inhibitor of LSD. For example, the LSD inhibitors contemplated include molecules that bind specifically to PKC0, and thus decrease, block, inhibit, abrogate or interfere with the phosphorylation (and therefore subsequent activation) of LSD.
[0080] The terms “patient”, “subject”, “host” or “individual” used interchangeably herein, refer to any subject, particularly a vertebrate subject, and even more particularly a mammalian subject, for whom therapy or prophylaxis is desired. Suitable vertebrate animals that fall within the scope of the invention include, but are not restricted to, any member of the subphylum Chordata including primates ( e.g ., humans, monkeys and apes, and includes species of monkeys such from the genus Macaca {e.g., cynomologus monkeys such as Macaca fascicularis, and/or rhesus monkeys (Macaca mulatta)) and baboon (Papio ursinus), as well as marmosets (species from the genus Callithrix), squirrel monkeys (species from the genus Saimiri) and tamarins (species from the genus Saguinus), as well as species of apes such as chimpanzees (Pan troglodytes)), rodents {e.g., mice rats, guinea pigs), lagomorphs {e.g., rabbits, hares), bovines {e.g., cattle), ovines {e.g., sheep), caprines {e.g., goats), porcines {e.g., pigs), equines {e.g., horses), canines {e.g., dogs), felines {e.g., cats), avians {e.g., chickens, turkeys, ducks, geese, companion birds such as canaries, budgerigars etc.), marine mammals {e.g., dolphins, whales), reptiles (snakes, frogs, lizards etc.), and fish. A preferred subject is a human in need of eliciting an immune response, including an immune response with enhanced T cell activation. However, it will be understood that the aforementioned terms do not imply that symptoms are present.
[0081] The term “pharmaceutical composition” or “pharmaceutical formulation” refers to a preparation which is in such form as to permit the biological activity of the active ingredient(s) to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the composition or formulation would be administered. Such formulations are sterile. “Pharmaceutically acceptable” excipients (vehicles, additives) are those which can reasonably be administered to a subject mammal to provide an effective dose of the active ingredient employed.
[0082] As used herein, the terms “prevent”, “prevented”, or “preventing”, refer to a prophylactic treatment which increases the resistance of a subject to developing the disease or condition or, in other words, decreases the likelihood that the subject will develop the disease or condition as well as a treatment after the disease or condition has begun in order to reduce or eliminate it altogether or prevent it from becoming worse. These terms also include within their scope preventing the disease or condition from occurring in a subject which may be predisposed to the disease or condition but has not yet been diagnosed as having it.
[0083] The term “selective” refers to compounds that inhibit or display antagonism towards a LSD without displaying substantial inhibition or antagonism towards another LSD or another enzyme such as a monoamine oxidase (MAO) ( e.g ., MAO A or MAO B). Accordingly, a compound that is selective for LSD1 exhibits a LSD1 selectivity of greater than about 2-fold, 5-fold, 10-fold, 20-fold, 50-fold or greater than about 100-fold with respect to inhibition or antagonism of another LSD (i.e., a LSD other than LSD1 such as LSD2) or of another enzyme {e.g., a MAO). In some embodiments, selective compounds display at least 50-fold greater inhibition or antagonism towards a specified LSD than towards another LSD or another enzyme {e.g., a MAO). In still other embodiments, selective compounds inhibit or display at least 100-fold greater inhibition or antagonism towards a specified LSD than towards another LSD or another enzyme {e.g., a MAO). In still other embodiments, selective compounds display at least 500-fold greater inhibition or antagonism towards a specified LSD than towards another LSD or another enzyme {e.g., a MAO). In still other embodiments, selective compounds display at least 1000- fold greater inhibition or antagonism towards a specified LSD than towards another LSD or another enzyme {e.g., a MAO).
[0084] The term “sequence identity” as used herein refers to the extent that sequences are identical on a nucleotide-by-nucleotide basis or an amino acid-by- amino acid basis over a window of comparison. Thus, a “percentage of sequence identity” is calculated by comparing two optimally aligned sequences over the window of comparison, determining the number of positions at which the identical nucleic acid base {e.g., A, T, C, G, I) or the identical amino acid residue {e.g., Ala, Pro, Ser, Thr, Gly, Val, Leu, lie, Phe, Tyr, Trp, Lys, Arg, His, Asp, Glu, Asn, Gin, Cys, and Met) occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window of comparison {i.e., the window size), and multiplying the result by 100 to yield the percentage of sequence identity. For the purposes of the present invention, “sequence identity” will be understood to mean the “match percentage” calculated by an appropriate method. For example, sequence identity analysis may be carried out using the DNASIS computer program (Version 2.5 for windows; available from Hitachi Software engineering Co., Ltd., South San Francisco, California, USA) using standard defaults as used in the reference manual accompanying the software.
[0085] As used herein a “small molecule” refers to a compound that has a molecular weight of less than 3 kilodalton (kDa), and typically less than 1.5 kDa, and more preferably less than about 1 kDa. Small molecules may be nucleic acids, peptides, polypeptides, peptidomimetics, carbohydrates, lipids or other organic (carbon-containing) or inorganic molecules. As those skilled in the art will appreciate, based on the present description, extensive libraries of chemical and/or biological mixtures, often fungal, bacterial, or algal extracts, may be screened with any of the assays of the invention to identify compounds that modulate a bioactivity. A “small organic molecule” is an organic compound (or organic compound complexed with an inorganic compound ( e.g ., metal)) that has a molecular weight of less than 3 kDa, less than 1.5 kDa, or even less than about 1 kDa.
[0086] “Stringency” of hybridization reactions is readily determinable by one of ordinary skill in the art, and generally is an empirical calculation dependent upon probe length, washing temperature, and salt concentration. In general, longer probes require higher temperatures for proper annealing, while shorter probes need lower temperatures. Hybridization generally depends on the ability of denatured DNA to reanneal when complementary strands are present in an environment below their melting temperature. The higher the degree of desired homology between the probe and hybridisable sequence, the higher the relative temperature which can be used. As a result, it follows that higher relative temperatures would tend to make the reaction conditions more stringent, while lower temperatures less so. For additional details and explanation of stringency of hybridization reactions, see Ausubel etal., Current Protocols in Molecular Biology, Wiley Interscience Publishers, (1995).
[0087] “Stringent conditions” or “high stringency conditions”, as defined herein, can be identified by those that: (1) employ low ionic strength and high temperature for washing, for example 15 mM sodium chloride/1.5 mM sodium citrate/0.1 % sodium dodecyl sulphate at 50 QC; (2) employ during hybridization a denaturing agent, such as formamide, for example, 50% (v/v) formamide with 0.1% bovine serum albumin/0.1% Ficoll/0.1% polyvinylpyrrolidone/50 mM sodium phosphate buffer at pH 6.5 with 750 mM sodium chloride, 75 mM sodium citrate at 42 QC; or (3) overnight hybridization in a solution that employs 50% formamide, 5 x SSC (0.75 M NaCI, 75 mM sodium citrate), 50 mM sodium phosphate (pH 6.8), 0.1% sodium pyrophosphate, 5 x Denhardt's solution, sonicated salmon sperm DNA (50 pg/mL), 0.1% SDS, and 10% dextran sulphate at 42 QC, with a 10 minute wash at 42 QC in 0.2 x SSC (sodium chloride/sodium citrate) followed by a 10 minute high- stringency wash consisting of 0.1 x SSC containing EDTA at 55 QC.
[0088] As used herein, the term “synergistic” means that the therapeutic effect of a LSD inhibitor ( e.g ., a LSD1 inhibitor or a LSD2 inhibitor) when administered in combination with an antiviral agent (or vice-versa) is greater than the predicted additive therapeutic effects of the LSD inhibitor and the antiviral agent when administered alone. The term “synergistically effective amount” as applied to a LSD inhibitor and an antiviral agent refers to the amount of each component in a composition (generally a pharmaceutical formulation), which is effective for enhancing immune effector function including any one or more of increased recognition of an antigen or an antigen peptide derived from an antigen in the context of MHC class II molecules by T cell receptors, increased release of cytokines and/or the activation of CD8+ lymphocytes (CTLs) and/or B cells, increased recognition of an antigen or an antigen peptide derived from an antigen in the context of MHC class I molecules by T cell receptors, increased elimination of cells presented in the context of MHC class I molecules, i.e., cells characterized by presentation of an antigen with class I MHC, for example, via apoptosis or perforin- mediated cell lysis, increased production of cytokines such as IL-2, IFN-y and TNF-a, and increased specific cytolytic killing of antigen expressing target cells, and which produces an effect which does not intersect, in a dose-response plot of the dose of LSD inhibitor versus a dose of antiviral agent versus enhancing immune effector function as antiviral agent axis. The dose response curve used to determine synergy in the art is described for example by Sande etal., (see, p. 1080-1105 in A. Goodman, The Pharmacological Basis of Therapeutics, MacMillan Publishing Co., Inc., New York (1980)). The optimum synergistic amounts can be determined, using a 95% confidence limit, by varying factors such as dose level, schedule and response, and using a computer-generated model that generates isobolograms from the dose response curves for various combinations of the LSD inhibitor and the antiviral agent. The highest enhancement of immune effector function on the dose response curve correlates with the optimum dosage levels.
[0089] As used herein, the term “treatment” refers to clinical intervention designed to alter the natural course of the individual or cell being treated during the course of clinical pathology. Desirable effects of treatment include decreasing the rate of disease progression, ameliorating or palliating the disease state, and remission or improved prognosis. For example, an individual is successfully “treated” if one or more symptoms associated with a T cell dysfunctional disorder are mitigated or eliminated, including, but are not limited to, reducing the proliferation of (or destroying) cancerous cells, reducing pathogen infection, decreasing symptoms resulting from the disease, increasing the quality of life of those suffering from the disease, decreasing the dose of other medications required to treat the disease, and/or prolonging survival of individuals.
[0090] As used herein, underscoring or italicizing the name of a gene shall indicate the gene, in contrast to its protein product, which is indicated by the name of the gene in the absence of any underscoring or italicizing. For example, “LSD1” shall mean the LSD1 gene, whereas “LSD1” shall indicate the protein product or products generated from transcription and translation and/or alternative splicing of the LSD1 gene.
[0091] Each embodiment described herein is to be applied mutatis mutandis to each and every embodiment unless specifically stated otherwise.
2. Methods of Preventing Virus Host Cell Entry
[0092] The route of host cell entry that is used by the coronaviruses is starting to be elucidated. As such, it is now generally accepted that coronaviruses display spike (S) protein trimers on their cell surface, and these glycosylated proteins accommodate binding to a host cell surface receptor. Upon receptor binding, the viral membrane fuses with that of the host cell, allowing the viral RNA entry into the host cell.
[0093] The coronavirus S protein mediates the first essential step in coronavirus infection, i.e., viral entry into host cells. The S protein contains an N-terminal signal peptide which primes the nascent polypeptide for import into the endoplasmic reticulum (ER). In the ER, the S protein is extensively modified with N-linked glycans, which are thought to provide protection against the neutralising antibodies of the host.
[0094] The S proteins, which are present as trimers on the virus surface, combine two biological functions. First, its surface unit, Si, binds to a specific receptor located at the surface of host cells (e.g., the ACE protein receptor) and thereby determines cellular tropism and, as a consequence, viral pathogenesis. Second, the transmembrane unit, S2, mediates fusion between the viral envelope and a target cell membrane. Priming (the proteolytic separation of the Si and S2 subunits) provides the coronavirus S protein with the structural flexibility required for the subsequent membrane fusion reaction.
[0095] The TMPRSS2 polypeptide has been demonstrated to cleave and activate S protein (Glowacha etal., 2011). Activation by the TMPRSS2 polypeptide is therefore believed to occur at the plasma membrane, likely after S protein binding to a virus cell entry receptor polypeptide (e.g., an ACE2 polypeptide). Notably, TMPRSS2 is reported to bind the virus cell entry polypeptide (e.g., ACE2, as described in Shulla etal., (2011)), and it is likely that the conformational changes in the S protein that are induced upon virus cell entry receptor polypeptide-binding expose the TMPRSS2 polypeptide cleavage site in the S protein.
[0096] The present inventors have determined that post-translational modifications play a significant role in the regulation of functional activity of the TMPRSS2 polypeptide. For example, a plurality of methylation sites have been identified in both (i) nuclear localisation signal domain of the TMPRSS2 polypeptide; and (ii) the catalytic domain of the TMPRSS2 polypeptide. Accordingly, administering an LSD1 inhibitor reduces the demethylation activity asserted on the TMPRSS2, causing a decrease in its functional activity. Accordingly, in some embodiments, an LSD inhibitor {e.g., a LSD1 inhibitor or a LSD2 inhibitor) is administered to a subject to prevent or reduce the ability of the TMPRSS2 polypeptide from transporting the coronavirus into the host cell.
[0097] In some embodiments, the TMPRSS2 polypeptide is a human TMPRSS2 polypeptide, with the amino acid sequence identified as UniProt Accession No. 015393, and set forth below: MALNSGSPPAIGPYYENHGYQPENPYPAQPTVVPTVYEVHPAQYYPSPVP
QYAPRVLTQASNPVVCTQPKSPSGTVCTSKTKKALCITLTLGTFLVGAALAA
GLLWKFMGSKCSNSGIECDSSGTCINPSNWCDGVSHCPGGEDENRCVRL
YGPNFILQVYSSQRKSWHPVCQDDWNENYGRAACRDMGYKNNFYSSQGI
VDDSGSTSFMKLNTSAGNVDIYKKLYHSDACSSKAVVSLRCIACGVNLNSS
RQSRIVGGESALPGAWPWQVSLHVQNVHVCGGSIITPEWIVTAAHCVEKPL
NNPWHWTAFAGILRQSFMFYGAGYQVEKVISHPNYDSKTKNNDIALMKLQ
KPLTFNDLVKPVCLPNPGMMLQPEQLCWISGWGATEEKGKTSEVLNAAKV
LLIETQRCNSRYVYDNLITPAMICAGFLQGNVDSCQGDSGGPLVTSKNNIW
WLIGDTSWGSGCAKAYRPGVYGNVMVFTDWIYRQMRADG.
[0098] Lysine residues K340 and K362 of the TMPRSS2 amino acid sequence set forth above are identified as being key LSD1 -mediated methylation/demethylation residues. Accordingly, in some embodiments the present invention provides methods of antagonising TMPRSS2 polypeptide, the method comprising administering an inhibitor of LSD1 -mediated demethylation of one or both residues K340 and K362.
[0099] The present inventors have also determined that post-translational modifications play a significant role in the regulation of functional activity of the viral cell entry receptor polypeptides ( e.g ., an ACE2 polypeptide). For example, a plurality of methylation sites are identified in both (i) nuclear localisation signal domain of the viral cell entry receptor polypeptide; and (ii) the catalytic domain of the viral cell entry receptor polypeptide. Accordingly, administering an LSD1 inhibitor reduces the demethylation activity asserted on the viral cell entry receptor polypeptide, causing a decrease in its functional activity. Accordingly, in some embodiments, an LSD inhibitor {e.g., a LSD1 inhibitor) is administered to a subject to prevent or reduce the ability of the virus cell entry receptor polypeptide to transporting a coronavirus into the host cell.
[0100] In some embodiments, the virus cell entry polypeptide is a human ACE2 polypeptide, with the amino acid sequence identified as UniProt Accession No. Q9BYF1 , and set forth below:
MSSSSWLLLSLVAVTAAQSTIEEQAKTFLDKFNHEAEDLFYQSSLASWNYNTN
ITEENVQNMNNAGDKWSAFLKEQSTLAQMYPLQEIQNLTVKLQLQALQQNGS
SVLSEDKSKRLNTILNTMSTIYSTGKVCNPDNPQECLLLEPGLNEIMANSLDYN
ERLWAWESWRSEVGKQLRPLYEEYVVLKNEMARANHYEDYGDYWRGDYEV NGVDGYDYSRGQLIEDVEHTFEEIKPLYEHLHAYVRAKLMNAYPSYISPIGCLP
AHLLGDMWGRFWTNLYSLTVPFGQKPNIDVTDAMVDQAWDAQRIFKEAEKFF
VSVGLPNMTQGFWENSMLTDPGNVQKAVCHPTAWDLGKGDFRILMCTKVTM
DDFLTAHHEMGHIQYDMAYAAQPFLLRNGANEGFHEAVGEIMSLSAATPKHL
KSIGLLSPDFQEDNETEINFLLKQALTIVGTLPFTYMLEKWRWMVFKGEIPKDQ
WMKKWWEMKREIVGVVEPVPHDETYCDPASLFHVSNDYSFIRYYTRTLYQFQ
FQEALCQAAKHEGPLHKCDISNSTEAGQKLFNMLRLGKSEPWTLALENVVGA
KNMNVRPLLNYFEPLFTWLKDQNKNSFVGWSTDWSPYADQSIKVRISLKSAL
GDKAYEWNDNEMYLFRSSVAYAMRQYFLKVKNQMILFGEEDVRVANLKPRIS
FNFFVTAPKNVSDIIPRTEVEKAIRMSRSRINDAFRLNDNSLEFLGIQPTLGPPN
QPPVSIWLIVFGVVMGVIVVGIVILIFTGIRDRKKKNKARSGENPYASIDISKGEN
NPGFQNTDDVQTSF.
[0101] Lysine residues K26 and K353 of the ACE2 polypeptide amino acid sequence set forth above are identified as key LSD1 -mediated methylation/ demethylation residues. Accordingly, in some embodiments the present invention provides methods of antagonising an ACE2 polypeptide, the method comprising administering an inhibitor of LSD1 -mediated demethylation. In some embodiments, the LSD1 -mediated demethylation includes demethylation of residues K26 and/or K353 of the ACE2 polypeptide.
[0102] In some alternative embodiments and some of the same embodiments, the present invention provides methods of antagonising an ACE2 polypeptide, the method comprising administering an inhibitor of LSD1 -mediated demethylation of one or both residues K340 and K362.
[0103] As described in the below examples, lysine residue K31 of the ACE2 polypeptide amino acid sequence set forth above is identified as key LSD1 -mediated methylation/demethylation residue for virus entry into the host cell. Accordingly, in some embodiments, the present invention provides methods of preventing entry of a coronavirus into a host cell, the method comprising administering an inhibitor of LSD1 -mediated demethylation of an ACE polypeptide (e.g., a human ACE polypeptide as set forth in UniProt Accession No. Q9BYF1). In some embodiments, the LSD1 -mediated demethylation of the ACE polypeptide includes demethylation of residue K31 of the ACE2 polypeptide [0104] In some alternative embodiments, the viral cell entry receptor polypeptide is selected from the group comprising an ACE2 polypeptide, a dipeptidyl peptidase 4 (DPP4) polypeptide, and a CD13 polypeptide.
[0105] Accordingly, the present invention extends to a method of inhibiting the entry of a coronavirus into a cell of the host, the method comprising administering to the subject an LSD1 inhibitor ( e.g ., a selective LSD1 inhibitor). Without wishing to be bound by any theory or mechanism, by inhibiting the LSD1 demethylation of the TMPRSS2 polypeptide and/or the virus cell entry receptor polypeptide (e.g., an ACE2 polypeptide), the coronavirus cell entry machinery is prevented from transporting the coronavirus into the cell.
[0106] In some of the same embodiments and some other embodiments, post-translational modifications by way of phosphorylation plays a significant role in the regulation of functional activity of the TMPRSS2 polypeptide. For example, at least one PKC0 serine phosphorylation site has been identified in the catalytic domain of the TMPRSS2 polypeptide. Accordingly, administering a PKC0 inhibitor reduces the phosphorylation activity asserted on the TMPRSS2, causing a decrease in its functional activity. Accordingly, in some embodiments, a PKC0 inhibitor is administered to a subject to prevent or reduce the ability of the TMPRSS2 polypeptide from transporting the coronavirus into the host cell. Serine residue S441 of the TMPRSS2 amino acid sequence set forth above is a key PKC0-mediated phosphorylated residue. Accordingly, in some embodiments the present invention provides methods of antagonising TMPRSS2 polypeptide, the method comprising administering an inhibitor of the PKC0-mediated phosphorylation of one or both residues S441.
[0107] The present inventors have also determined that phosphorylation post-translational modifications play a significant role in the regulation of functional activity of the viral cell entry receptor polypeptides {e.g., an ACE2 polypeptide). For example, a plurality of PKC0-mediated phosphorylation sites are present in both (i) the nuclear localisation signal domain of the viral cell entry receptor polypeptide; and (ii) the catalytic domain of the viral cell entry receptor polypeptide. Accordingly, administering a PKC0 inhibitor reduces the phosphorylation activity asserted on the viral cell entry receptor polypeptide, causing a decrease in its functional activity. Accordingly, in some embodiments, a PKC0 inhibitor is administered to a subject to prevent or reduce the ability of the virus cell entry receptor polypeptide to transporting a coronavirus into the host cell.
[0108] Residues S47, S109, S254 and T763 of the ACE2 polypeptide amino acid sequence set forth above are identified as key PKC0-mediated phosphorylation sites. Accordingly, in some embodiments the present invention provides methods of antagonising an ACE2 polypeptide, the method comprising administering an inhibitor of PKC0-mediated phosphorylation of one or more residues S47, S109, S254 and T763 of the ACE2 polypeptide.
[0109] In some of the same embodiments and some other embodiments, post-translational modifications by way of phosphorylation plays a significant role in the regulation of functional activity of the viral cell entry receptor polypeptides ( e.g ., an ACE2 polypeptide). For example, a plurality of methylation sites are identified in both (i) nuclear localisation signal domain of the viral cell entry receptor polypeptide; and (ii) the catalytic domain of the viral cell entry receptor polypeptide. Accordingly, administering an LSD1 inhibitor reduces the demethylation activity asserted on the viral cell entry receptor polypeptide, causing a decrease in its functional activity. Accordingly, in some embodiments, an LSD inhibitor {e.g., a LSD1 inhibitor or a LSD2 inhibitor) is administered to a subject to prevent or reduce the ability of the virus cell entry receptor polypeptide to transporting a coronavirus into the host cell.
[0110] In some particularly important embodiments of the present invention, the coronavirus is a SARS-CoV-2.
3. Compositions and Methods
[0111] The present invention is based in part on the determination that coronavirus infections take advantage of host machinery to enter the host cell, and that these essential host machinery are regulated at the post-translational level and the transcriptional level by methylation, by LSD1. Without wishing to be bound by any theory or mode of operation, it is proposed that LSDs, including LSD1 , play a critical role in two levels: (1) the post-transcriptional methylation of the chromatin domains across the regulatory regions of the cell entry machinery and that prevention of the demethylation by LSD1 prevents the ability of the cell to express the polypeptide expression products of the TMPRSS2 gene and/or the virus cell entry receptor gene; and (2) the post-translational methylation of the active (e.g., catalytic) domains of the TMPRSS2 polypeptide and/or the viral host cell entry polypeptide (e.g., an ACE2 polypeptide).
[0112] Based on this observation, the present inventors propose that LSD1 inhibition will result in reduced ability for the coronavirus to enter into the host cell, and thus provide a novel treatment for coronavirus infections.
[0113] Thus, in accordance with the present invention, methods and compositions are provided that take advantage of an LSD1 inhibitor {e.g., a selective LSD1 inhibitor) to reduce or abrogate the transcription of integral cellular machinery required for a coronavirus entry into a cell. In some embodiments, the LSD1 inhibitor is used in combination with an additional antiviral agent. The methods and compositions of the present invention are thus particularly useful in the treatment or prophylaxis of a coronavirus infection {e.g., a coronavirus infection), as described hereafter.
3J. LSD Inhibitors
[0114] The LSD inhibitor includes and encompasses any active agent that reduces the accumulation, function or stability of a LSD; or decrease expression of a LSD gene, and such inhibitors include without limitation, small molecules and macromolecules such as nucleic acids, peptides, polypeptides, peptidomimetics, carbohydrates, polysaccharides, lipopolysaccharides, lipids or other organic (carbon containing) or inorganic molecules.
[0115] In some embodiments, the LSD inhibitor is an antagonistic nucleic acid molecule that functions to inhibit the transcription or translation of LSD (e.g., LSD1 or LSD2) transcripts. Representative transcripts of this type include nucleotide sequences corresponding to any one the following sequences: (1) human LSD1 nucleotide sequences as set forth for example in GenBank Accession Nos.
NM_01 5013.3, NP_001009999.1 , and NM_001009999.2; human LSD2 nucleotide sequences as set forth for example in GenBank Accession No. NM_153042.3; (2) nucleotide sequences that share at least 70, 71 , 72, 73, 74, 75, 76, 77, 78, 79, 80,
81 , 82, 83, 84, 85, 86, 87, 88, 89, 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99% sequence identity with any one of the sequences referred to in (1); (3) nucleotide sequences that hybridize under at least low, medium or high stringency conditions to the sequences referred to in (1); (4) nucleotide sequences that encode any one of the following amino acid sequences: human LSD1 amino acid sequences as set forth for example in GenPept Accession Nos. NP_055828.2, NP_001009999.1 and 060341.2; human LSD2 amino acid sequences as set forth for example in GenPept Accession Nos. NP_694587.3; (5) nucleotide sequences that encode an amino acid sequence that shares at least 70, 71 , 72, 73, 74, 75, 76, 77, 78, 79, 80, 81 , 82, 83,
84, 85, 86, 87, 88, 89, 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99% sequence similarity with any one of the sequences referred to in (4); and nucleotide sequences that encode an amino acid sequence that shares at least 70, 71 , 72, 73, 74, 75, 76, 77,
78, 79, 80, 81 , 82, 83, 84, 85, 86, 87, 88, 89, 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99% sequence identity with any one of the sequences referred to in (4).
[0116] Illustrative antagonist nucleic acid molecules include antisense molecules, aptamers, ribozymes and triplex forming molecules, RNAi and external guide sequences. The nucleic acid molecules can act as effectors, inhibitors, modulators, and stimulators of a specific activity possessed by a target molecule, or the functional nucleic acid molecules can possess a de novo activity independent of any other molecules.
[0117] Antagonist nucleic acid molecules can interact with any macromolecule, such as DNA, RNA, polypeptides, or carbohydrate chains. Thus, antagonist nucleic acid molecules can interact with LSD (e.g., LSD1) mRNA or the genomic DNA of LSD (e.g., LSD1) or they can interact with a LSD polypeptide (e.g., LSD1). Often antagonist nucleic acid molecules are designed to interact with other nucleic acids based on sequence homology between the target molecule and the antagonist nucleic acid molecule. In other situations, the specific recognition between the antagonist nucleic acid molecule and the target molecule is not based on sequence homology between the antagonist nucleic acid molecule and the target molecule, but rather is based on the formation of tertiary structure that allows specific recognition to take place.
[0118] In some embodiments, anti-sense RNA or DNA molecules are used to directly block the translation of LSD (e.g., LSD1) by binding to targeted mRNA and preventing protein translation. Antisense molecules are designed to interact with a target nucleic acid molecule through either canonical or non-canonical base pairing. The interaction of the antisense molecule and the target molecule may be designed to promote the destruction of the target molecule through, for example, RNAseH mediated RNA-DNA hybrid degradation. Alternatively, the antisense molecule may be designed to interrupt a processing function that normally would take place on the target molecule, such as transcription or replication. Antisense molecules can be designed based on the sequence of the target molecule. Numerous methods for optimization of antisense efficiency by finding the most accessible regions of the target molecule exist. Non-limiting methods include in vitro selection experiments and DNA modification studies using DMS and DEPC. In specific examples, the antisense molecules bind the target molecule with a dissociation constant (Kd) less than or equal to 106, 108, 1010, or 1012. In specific embodiments, antisense oligodeoxyribonucleotides derived from the translation initiation site, e.g., between - 10 and +10 regions are employed.
[0119] Aptamers are molecules that interact with a target molecule, suitably in a specific way. Aptamers are generally small nucleic acids ranging from 15-50 bases in length that fold into defined secondary and tertiary structures, such as stem-loops or G-quartets. Aptamers can bind small molecules, such as ATP and theophiline, as well as large molecules, such as reverse transcriptase and thrombin. Aptamers can bind very tightly with KdS from the target molecule of less than 1012 M. Suitably, the aptamers bind the target molecule with a Kd less than 106, 108, 1010, or 1012 Aptamers can bind the target molecule with a very high degree of specificity. For example, aptamers have been isolated that have greater than a 10,000-fold difference in binding affinities between the target molecule and another molecule that differ at only a single position on the molecule. It is desirable that an aptamer have a Kd with the target molecule at least 10-, 100-, 1000-, 10,000-, or 100,000-fold lower than the Kd with a background-binding molecule. A suitable method for generating an aptamer to a target of interest {e.g., PHD, FIH-1 or vHL) is the “Systematic Evolution of Ligands by Exponential Enrichment” (SELEX™). The SELEX™ method is described in U.S. Pat. Nos. 5,475,096 and 5,270,163 (see also WO 91/19813). Briefly, a mixture of nucleic acids is contacted with the target molecule under conditions favourable for binding. The unbound nucleic acids are partitioned from the bound nucleic acids, and the nucleic acid -target complexes are dissociated. Then the dissociated nucleic acids are amplified to yield a ligand-enriched mixture of nucleic acids, which is subjected to repeated cycles of binding, partitioning, dissociating and amplifying as desired to yield highly specific high affinity nucleic acid ligands to the target molecule.
[0120] In other embodiments, anti-LSD1 ribozymes are used for catalysing the specific cleavage of LSD1 RNA. The mechanism of ribozyme action involves sequence specific hybridization of the ribozyme molecule to complementary target RNA, followed by an endonucleolytic cleavage. There are several different types of ribozymes that catalyse nuclease or nucleic acid polymerase type reactions, which are based on ribozymes found in natural systems, such as hammerhead ribozymes, hairpin ribozymes, and tetrahymena ribozymes. There are also a number of ribozymes that are not found in natural systems, but which have been engineered to catalyse specific reactions de novo. Representative ribozymes cleave RNA or DNA substrates. In some embodiments, ribozymes that cleave RNA substrates are employed. Specific ribozyme cleavage sites within potential RNA targets are initially identified by scanning the target molecule for ribozyme cleavage sites, which include the following sequences, GUA, GUU and GUC. Once identified, short RNA sequences of between 15 and 20 ribonucleotides corresponding to the region of the target gene containing the cleavage site may be evaluated for predicted structural features such as secondary structure that may render the oligonucleotide sequence unsuitable. The suitability of candidate targets may also be evaluated by testing their accessibility to hybridization with complementary oligonucleotides, using ribonuclease protection assays.
[0121] Triplex forming functional nucleic acid molecules are molecules that can interact with either double-stranded or single-stranded nucleic acid. When triplex molecules interact with a target region, a structure called a triplex is formed, in which there are three strands of DNA forming a complex dependent on both Watson-Crick and Hoogsteen base pairing. Triplex molecules are preferred because they can bind target regions with high affinity and specificity. It is generally desirable that the triplex forming molecules bind the target molecule with a Kd less than 106, 108, 1010, or 10-12.
[0122] External guide sequences (EGSs) are molecules that bind a target nucleic acid molecule forming a complex, and this complex is recognized by RNAse P, which cleaves the target molecule. EGSs can be designed to specifically target a RNA molecule of choice. RNAse P aids in processing transfer RNA (tRNA) within a cell.
[0123] Bacterial RNAse P can be recruited to cleave virtually any RNA sequence by using an EGS that causes the target RNA: EGS complex to mimic the natural tRNA substrate. Similarly, eukaryotic EGS/RNAse P-directed cleavage of RNA can be utilized to cleave desired targets within eukaryotic cells.
[0124] In other embodiments, RNA molecules that mediate RNA interference (RNAi) of a LSD1 gene or LSD1 transcript can be used to reduce or abrogate gene expression. RNAi refers to interference with or destruction of the product of a target gene by introducing a single-stranded or usually a double- stranded RNA (dsRNA) that is homologous to the transcript of a target gene. RNAi methods, including double-stranded RNA interference (dsRNAi) or small interfering RNA (siRNA), have been extensively documented in a number of organisms, including mammalian cells and the nematode C. elegans (Fire etal., 1998. Nature 391 , 806-811 ). In mammalian cells, RNAi can be triggered by 21 - to 23-nucleotide (nt) duplexes of small interfering RNA (siRNA) (Chiu etal., 2002 Mol. Cell. 10: 549- 561 ; Elbashir et al., 2001. Nature 411 : 494-498), or by micro-RNAs (miRNA), functional small-hairpin RNA (shRNA), or other dsRNAs which are expressed in vivo using DNA templates with RNA polymerase III promoters (Zeng et al., 2002. Mol.
Cell 9: 1327-1333 ; Paddison et al., 2002. Genes Dev. 16:948-958; Lee et al., 2002. Nature Biotechnol. 20: 500-505; Paul etal., 2002. Nature Biotechnol. 20: 505-508; Tuschl, T., 2002. Nature Biotechnol. 20: 440-448; Yu etal., 2002. Proc. Natl. Acad. Sci. USA 99(9): 6047-6052; McManus etal., 2002. RNA 8: 842-850; Sui et al., 2002. Proc. Natl. Acad. Sci. USA. 99(6): 5515-5520).
[0125] In specific embodiments, dsRNA per se and especially dsRNA- producing constructs corresponding to at least a portion of a LSD1 gene are used to reduce or abrogate its expression. RNAi-mediated inhibition of gene expression may be accomplished using any of the techniques reported in the art, for instance by transfecting a nucleic acid construct encoding a stem-loop or hairpin RNA structure into the genome of the target cell, or by expressing a transfected nucleic acid construct having homology for a LSD1 gene from between convergent promoters, or as a head to head or tail to tail duplication from behind a single promoter. Any similar construct may be used so long as it produces a single RNA having the ability to fold back on itself and produce a dsRNA, or so long as it produces two separate RNA transcripts, which then anneal to form a dsRNA having homology to a target gene.
[0126] Absolute homology is not required for RNAi, with a lower threshold being described at about 85% homology for a dsRNA of about 200 base pairs (Plasterk and Ketting, 2000, Current Opinion in Genetics and Dev. 10: 562-67). Therefore, depending on the length of the dsRNA, the RNAi-encoding nucleic acids can vary in the level of homology they contain toward the target gene transcript, i.e., with dsRNAs of 100 to 200 base pairs having at least about 85% homology with the target gene, and longer dsRNAs, i.e., 300 to 100 base pairs, having at least about 75% homology to the target gene. RNA-encoding constructs that express a single RNA transcript designed to anneal to a separately expressed RNA, or single constructs expressing separate transcripts from convergent promoters, are suitably at least about 100 nucleotides in length. RNA-encoding constructs that express a single RNA designed to form a dsRNA via internal folding are usually at least about 200 nucleotides in length.
[0127] The promoter used to express the dsRNA-forming construct may be any type of promoter if the resulting dsRNA is specific for a gene product in the cell lineage targeted for destruction. Alternatively, the promoter may be lineage specific in that it is only expressed in cells of a particular development lineage. This might be advantageous where some overlap in homology is observed with a gene that is expressed in a non-targeted cell lineage. The promoter may also be inducible by externally controlled factors, or by intracellular environmental factors.
[0128] In some embodiments, RNA molecules of about 21 to about 23 nucleotides, which direct cleavage of specific mRNA to which they correspond, as for example described by in U.S. Pat. Pub. No. 2002/0086356, can be utilized for mediating RNAi. Such 21- to 23-nt RNA molecules can comprise a 3' hydroxyl group, can be single-stranded or double stranded (as two 21 - to 23-nt RNAs) wherein the dsRNA molecules can be blunt ended or comprise overhanging ends ( e.g ., 5', 3').
[0129] In some embodiments, the antagonist nucleic acid molecule is a siRNA. siRNAs can be prepared by any suitable method. For example, reference may be made to International PCT Pat. Pub. No. WO 02/44321 , which discloses siRNAs capable of sequence-specific degradation of target mRNAs when base- paired with 3' overhanging ends, which is incorporated by reference herein. Sequence specific gene silencing can be achieved in mammalian cells using synthetic, short double-stranded RNAs that mimic the siRNAs produced by the enzyme dicer. siRNA can be chemically or in v/ ro-synthesized or can be the result of short double-stranded hairpin-like RNAs (shRNAs) that are processed into siRNAs inside the cell. Synthetic siRNAs are generally designed using algorithms and a conventional DNA/RNA synthesizer. Suppliers include Ambion (Austin, Tex.), ChemGenes (Ashland, Mass.), Dharmacon (Lafayette, Colo.), Glen Research (Sterl ing, Va.), MWB Biotech (Esbersberg, Germany), Proligo (Boulder, Colo.), and Qiagen (Vento, The Netherlands). siRNA can also be synthesized in vitro using kits such as Ambion's SILENCER™ siRNA Construction Kit.
[0130] The production of siRNA from a vector is more commonly done through the transcription of a short hairpin RNAs (shRNAs). Kits for the production of vectors comprising shRNA are available, such as, for example, Imgenex's GENESUPPRESSOR™ Construction Kits and Invitrogen's BLOCK-IT™ inducible RNAi plasmid and lentivirus vectors. In addition, methods for formulation and delivery of siRNAs to a subject are also well known in the art. See, e.g.,
US 2005/0282188; US 2005/0239731 ; US 2005/0234232; US 2005/0176018;
US 2005/0059817; US 2005/0020525; US2004/0192626; US 2003/0073640;
US 2002/0150936; US 2002/0142980; and US 2002/0120129, each of which is incorporated herein by reference.
[0131] Illustrative RNAi molecules {e.g., LSD {e.g., LSD1 or LSD2) siRNA and shRNA) are described in the art {e.g., Yang, etat, 2010. Proc. Natl. Acad. Sci. USA 107: 21499-21504 and He et al., 2012. Transcription 3: 1-16) or available commercially from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA, USA) and OriGene Technologies, Inc. (Rockville, MD, USA) .
[0132] The present invention further contemplates peptide or polypeptide- based inhibitor compounds. For example, BHC80 (also known as PHD finger protein 21 A) forms part of a complex with LSD1 and can inhibit LSD1 demethylase activity. Accordingly, the present invention further contemplates the use of BHC80 or biologically active fragments thereof for inhibiting LSD1 enzymatic activity. Amino acid sequences of BHC80 polypeptides, and nucleotide sequences encoding BHC80 polypeptides, are publicly available. In this regard, reference may be made for example to (1) GenBank Accession No. NP057705 for a Homo sapiens BHC80 amino acid sequence; and GenBank NM016621 for a nucleotide sequence encoding the amino acid sequence set forth in GenBank Accession No. NP057705; (2) GenBank Accession No. NP620094 for a Mus musculus BHC80 amino acid sequence; and GenBank NM138755 for a nucleotide sequence encoding the amino acid sequence set forth in GenBank Accession No. NP620094; (3) GenBank Accession No. NP00118576.1 for a Gallus gallus BHC80 amino acid sequence; and GenBankNMOO1 199647 for a nucleotide sequence encoding the amino acid sequence set forth in GenBank Accession No. NP00118576.1 ; and (4) GenBank Accession No. DAA21793 for a Bos taurus BHC80 amino acid sequence.
[0133] Illustrative BHC80 polypeptides are selected from the group consisting of: (1) a polypeptide comprising an amino acid sequence that shares at least 70, 71 , 72, 73, 74, 75, 76, 77, 78, 79, 80, 81 , 82, 83, 84, 85, 86, 87, 88, 89, 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99% sequence similarity with the amino acid sequence listed in any one of the GenBank BHC80 polypeptide entries noted above; (2) a polypeptide comprising an amino acid sequence that shares at least 70, 71 , 72, 73, 74, 75, 76, 77, 78, 79, 80, 81 , 82, 83, 84, 85, 86, 87, 88, 89, 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99% sequence identity with the amino acid sequence listed in any one of the GenBank BHC80 polypeptide entries noted above; (3) a polypeptide comprising an amino acid sequence that is encoded by a nucleotide sequence that hybridizes under at least low, medium or high stringency conditions to the nucleotide sequence listed in any one of the GenBank BHC80 polynucleotide entries noted above; (4) a polypeptide comprising an amino acid sequence that is encoded by a nucleotide sequence that shares at least 70, 71 , 72, 73, 74, 75, 76, 77, 78, 79, 80, 81 , 82, 83, 84, 85, 86, 87, 88, 89, 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99% sequence identity to the nucleotide sequence listed in any one of the GenBank BHC80 polynucleotide entries noted above; and (5) a fragment of a polypeptide according to any one of (1) to (4), which inhibits LSD1 enzymatic activity.
[0134] A BHC80 polypeptide can be introduced into a cell by delivering a polypeptide perse, or by introducing into the cell a BHC80 nucleic acid comprising a nucleotide sequence encoding a BHC80 polypeptide. In some embodiments, a BHC80 nucleic acid comprises a nucleotide sequence selected from: (1) a BHC80 nucleotide sequence listed in any one of the GenBank BHC80 polynucleotide entries noted above; (2) a nucleotide sequence that shares at least 70, 71 , 72, 73, 74, 75,
76, 77, 78, 79, 80, 81 , 82, 83, 84, 85, 86, 87, 88, 89, 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99% sequence identity with any one of the sequences referred to in (1); (3) a nucleotide sequence that hybridizes under at least low, medium or high stringency conditions to the sequences referred to in (1); (4) a nucleotide sequence that encodes an amino acid sequence listed in any one of the GenBank BHC80 polypeptide entries noted above; (5) a nucleotide sequence that encodes an amino acid sequence that shares at least 70, 71 , 72, 73, 74, 75, 76, 77, 78, 79, 80, 81 , 82, 83, 84, 85, 86, 87, 88, 89, 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99% sequence similarity with any one of the sequences referred to in (4); and a nucleotide sequence that encodes an amino acid sequence that shares at least 70, 71 , 72, 73, 74, 75, 76, 77, 78, 79, 80, 81 , 82, 83, 84, 85, 86, 87, 88, 89, 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99% sequence identity with any one of the sequences referred to in (4).
[0135] The BHC80 nucleic acid can be in the form of a recombinant expression vector. The BHC80 nucleotide sequence can be operably linked to a transcriptional control element(s), e.g., a promoter, in the expression vector. Suitable vectors include, e.g., recombinant retroviruses, lentiviruses, and adenoviruses; retroviral expression vectors, lentiviral expression vectors, nucleic acid expression vectors, and plasmid expression vectors. In some cases, the expression vector is integrated into the genome of a cell. In other cases, the expression vector persists in an episomal state in a cell.
[0136] Suitable expression vectors include, but are not limited to, viral vectors {e.g., viral vectors based on vaccinia virus; poliovirus; adenovirus (see, e.g.,
Li etal., Invest Opthalmol Vis Sci 35: 25432549, 1994; Borras etal., Gene Ther 6: 515524, 1999; Li and Davidson, P/VAS 92: 77007704, 1995; Sakamoto et ai, Gene Ther 5 : 1088 1097, 1999; International PCT Pat. Pub. Nos WO 94/12649,
WO 93/03769; WO 93/19191 ; WO 94/28938; WO 95/11984 and WO 95/00655); adeno-associated virus (see, e.g., AN etal., Hum Gene Ther 9 :8186, 1998,
Flannery et ai, PNAS 94:69166921 , 1997; Bennett et ai, Invest Opthalmol Vis Sci 38: 28572863, 1997; Jomary etal., Gene Ther. 4: 683690, 1997, Rolling etal., Hum Gene Ther. 10 : 641 648, 1999; AN etal. , Hum Mol Genet. 5: 591 594, 1996; International PCT Pat. Pub. No. WO 93/09239, Samulski etal., J. Vir. (1989) 63: 3822-3828; Mendelson etal., Virol. (1988) 166: 154-165; and Flotte etal., PNAS (1993) 90 : 10613-10617); SV40; herpes simplex virus; human immunodeficiency virus (see, e.g., Miyoshi etal., PNAS 94: 1031923, 1997; Takahashi etal., J Virol 73: 78127816, 1999); a retroviral vector {e.g., Murine Leukemia Virus, spleen necrosis virus, and vectors derived from retroviruses such as Rous Sarcoma Virus, Harvey Sarcoma Virus, avian leukosis virus, a lentivirus, human immunodeficiency virus, myeloproliferative sarcoma virus, and mammary tumour virus); and the like.
Small Molecule Inhibitors
[0137] Small molecule agents that reduce enzymatic activity of LSDs {e.g., LSD1)have particular utility for use with the present invention.
[0138] Small molecule agents that reduce enzymatic activity of LSD1 that are suitable for use in the present invention include monoamine oxidase (MAO) inhibitors that also inhibit LSD1 enzymatic activity; polyamine compounds that inhibit LSD1 enzymatic activity; phenylcyclopropylamine derivatives that inhibit LSD1 enzymatic activity; and the like.
[0139] Non-limiting examples of MAO inhibitors include MAO-A-selective inhibitors, MAO-B-selective inhibitors, and MAO non-selective inhibitors. Illustrative examples of MAO inhibitors include reported inhibitors of the MAO-A isoform, which preferentially deaminates 5-hydroxytryptamine (serotonin) (5-HT) and norepinephrine (NE), and/or the MAO-B isoform, which preferentially deaminates phenylethylamine (PEA) and benzylamine (both MAO-A and MAO-B metabolize Dopamine (DA)). In various embodiments, MAO inhibitors may be irreversible or reversible {e.g., reversible inhibitors of MAO-A (RIMA)), and may have varying potencies against MAO-A and/or MAO-B {e.g., non-selective dual inhibitors or isoform-selective inhibitors).
[0140] In some embodiments, the MAO inhibitors are selected from : clorgyline; L-deprenyl; isocarboxazid (MARPLAN™); ayahuasca; nialamide; iproniazide; iproclozide; moclobemide (AURORIX™; 4-chloro-N-(2-morpholin-4- ylethyl)benzamide); phenelzine (NARDIL™; (±)-2-phenylethylhydrazine); tranylcypromine (PARNATE™; (±)-trans-2-phenylcyclopropan-l-amine) (the congeneric of phenelzine); bizine (described in Prusevich et al., ACS Chem. Biol., 2014, 9, 1284-1293); SP-2509 (described in Fiskus et al., Leukemia, (2014), 28, 2155-2164); SP-2577 (Seclidemstat, Salarius Pharmaceuticas, Houston, TX; described in Soldi, et al., PLOS One, 2020, July 10); toloxatone; levo-deprenyl (SELEGILINE™); harmala; RIMAs (e.g., moclobemide, described in Da Prada et al. (1989. J Pharmacol Exp Ther. 248: 400-414); brofaromine; and befloxatone, described in Curet etal. (1998. J Affect Disord. 51 : 287-30), lazabemide (PAKIO™, RO 196327), described in Ann. Neurol., 40(1): 99-107 (1996), and SL25.1131 (described in Aubin et al. (2004. J Pharmacol Exp Ther. 310: 1171 -1182); selegiline hydrochloride (L-deprenyl, ELDEPRYL™, ZELAPAR™); dimethylselegilene; safinamide; rasagiline (AZILECT™); bifemelane; desoxypeganine; harmine (also known as telepathine or banasterine); linezolid (ZYVOX™, ZYVOXID™); pargyiine (EUDATIN™, SUPIRDYL™); dienolide kavapyrone desmethoxyyangonin; 5-(4- Arylmethoxyphenyl)-2-(2-cyanoethyl)tetrazoles; and the like.
[0141] Small molecule LSD1 inhibitors may also be selected from polyamine compounds as described for example in United States Publication No.
US 2007/0208082, which is incorporated herein by reference in its entirety. Illustrative polyamine inhibitors of LSD1 include compounds according to formula (I):
Figure imgf000045_0001
[0142] or a salt, solvate, or hydrate thereof, where n is an integer from 1 to 12; m and p are independently an integer from 1 to 5; q is 0 or 1 ; each Ri is independently selected from the group consisting of C-i-Cs alkyl, C4-C15 cycloalkyl, C3-C15 branched alkyl, C6-C20 aryl, C6-C20 heteroaryl, C7-C24 aralkyl, C7-C24 heteroaralkyl, and
Figure imgf000045_0002
[0143] where R3 is selected from the group consisting of C-i-Cs alkyl, C4-C15 cycloalkyl, C3-C15 branched alkyl, C6-C20 aryl, C6-C20 heteroaryl, C7-C24 aralkyl and C7-C24 heteroaralkyl; and
[0144] each R2 is independently selected from hydrogen or a C-i-Cs alkyl. [0145] A suitable polyamine compound is a compound of Formula (I), wherein one or both Ri is a C6-C20 aryl, such as a single ring aryl, including without limitation, a phenyl. In one embodiment, the compound is of the formula (I) and each Ri is phenyl. In one embodiment, q is 1 , m and p are 3, and n is 4. In another embodiment, q is 1 , m and p are 3, and n is 7.
[0146] A suitable polyamine compound is a compound of Formula (I), where at least one or both Ri is a C8-C12 or a C-i-Cs alkyl, such as a linear alkyl. One or both Ri may be a C-i-Cs linear alkyl, such as methyl or ethyl. In one embodiment, each Ri is methyl. One or both Ri may comprise or be a C4-C15 cycloalkyl group, such as a cycloalkyl group containing a linear alkyl group, where the cycloalkyl group is connected to the molecule either via its alkyl or cycloalkyl moiety. For instance, one or both Ri may be cyclopropylmethyl or cyclohexylmethyl. In one embodiment, one Ri is cyclopropylmethyl or cyclohexyl methyl and the other Ri is a linear alkyl group, such as a linear C-i-Cs unsubstituted alkyl group, including without limitation an ethyl group. In one embodiment, Ri is a C3-C15 branched alkyl group such as isopropyl. When Ri is a C-i-Cs substituted alkyl, the substituted alkyl may be substituted with any substituent, including a primary, secondary, tertiary or quaternary amine. Accordingly, in one embodiment, Ri is a C-i-Cs alkyl group substituted with an amine such that Ri may be e.g., alkyl-NH2 or an alkyl-amine-alkyl moiety such as -(CFl2)yNFI(CFl2)zCFl3 where y and z are independently an integer from 1 to 8. In one embodiment, Ri is -(CFl2)3NFl2.
[0147] In some embodiments, the compound is of the formula (I) where one or both Ri is a C7-C24 substituted or unsubstituted aralkyl, which in one embodiment is an aralkyl connected to the molecule via its alkyl moiety {e.g., benzyl). In some embodiments, both Ri are aralkyl moieties wherein the alkyl portion of the moiety is substituted with two aryl groups and the moiety is connected to the molecule via its alkyl group. For instance, in some embodiments one or both Ri is a C7-C24 aralkyl wherein the alkyl portion is substituted with two phenyl groups, such as when Ri is
2,2-diphenylethyl or 2,2-dibenzylethyl. In one embodiment, both Ri of Formula (I) is
2.2-diphenylethyl and n is 1 , 2 or 5. In some embodiments, each Ri of Formula (I) is
2.2-diphenylethyl, n is 1 , 2 or 5 and m and p are each 1.
[0148] In some embodiments, at least one Ri is hydrogen. When one Ri is hydrogen, the other Ri may be any moiety listed above for Ri, including an aryl group such as benzyl. Any of the compounds of Formula (I) listed above include compounds where at least one or both of R2 is hydrogen or a C-i-Cs substituted or unsubstituted alkyl. In some embodiments, each R2 is an unsubstituted alkyl such as methyl. In some other embodiments, each R2 is hydrogen. Any of the compounds of Formula (I) listed above may be compounds where q is 1 and m and p are the same. Accordingly, the polyaminoguanidines of Formula (I) may be symmetric with reference to the polyaminoguanidine core {e.g., excluding Ri). Alternatively, the compounds of Formula (I) may be asymmetric, e.g., when q is 0. In one embodiment, m and p are 1 . In some embodiments, q is 0. In one embodiment, n is an integer from 1 to 5.
[0149] In some embodiments, the compound is a polyaminobiguanide or N- alkylated polyaminobiguanide. An N-alkylated polyaminobiguanide intends a polyaminobiguanide where at least one imine nitrogen of at least one biguanide is alkylated. In one embodiment, the compound is a polyaminobiguanide of the Formula (I), or a salt, solvate, or hydrate thereof, where q is 1 , and at least one or each R, is of the structure:
Figure imgf000047_0001
[0150] where each R3 is independently selected from the group consisting of C1-C8 alkyl, C6-C20 aryl, C6-C20 heteroaryl, C7-C24 aralkyl, and C7-C24 heteroaralkyl; and each R2 is independently hydrogen or a C-i-Cs alkyl.
[0151] In some embodimenst, in the polyaminobiguanide compound, at least one or each R3 is a C-i-Cs alkyl. For instance, when R is a Ci-C8 alkyl, the alkyl may be substituted with any substituent, including a primary, secondary, tertiary or quaternary amine. Accordingly, in one embodiment, R3 is a C-i-Cs alkyl group substituted with an amine such that R3 may be e.g., alkyl-NFte or an alkyl-amine-alkyl moiety such as -(CH2)yNH(CH2)zCH3 where y and z are independently an integer from 1 to 8. In one embodiment, R3 is -(CH2)3NH2. R3 may also be a C4-C15 cycloalkyl or a C3-C15 branched alkyl. In some embodiments, at least one or each R3 is a C6-C20 aryl. In some embodiments, q is 1 , m and p are 3, and n is 4. In another embodiment, q is 1 , m and p are 3, and n is 7. [0152] In some embodiments, the compound is a polyaminobiguanide of Formula (I) where at least one R3 is a C7-C24 aralkyl, which in one embodiment is an aralkyl connected to the molecule via its alkyl moiety. In some embodiments, each R3 is an aralkyl moiety where the alkyl portion of the moiety is substituted with one or two aryl groups and the moiety is connected to the molecule via its alkyl moiety. For instance, in some embodiments at least one or each R3 is an aralkyl where the alkyl portion is substituted with two phenyl or benzyl groups, such as when R3 is 2,2- diphenylethyl or 2,2-dibenzylethyl. In some embodiments, each R3 is 2,2- diphenylethyl and n is 1 , 2 or 5. In some embodiments, each R3 is 2,2-diphenylethyl and n is 1 , 2 or 5 and m and p are each 1.
[0153] Any of the polyaminobiguanide compounds of Formula (I) listed above include compounds where at least one or both of R2 is hydrogen or a C-i-Cs alkyl. In some embodiments, each R2 is an unsubstituted alkyl, such as methyl. In some other embodiments, each R2 is a hydrogen.
[0154] Any of the polyaminobiguanide compounds of formula (I) listed above include compounds where q is 1 and m and p are the same. Accordingly, the polyaminobiguanides of Formula (I) may be symmetric with reference to the polyaminobiguanide core. Alternatively, the compounds of Formula (I) may be asymmetric. In one embodiment, m and p are 1. In some embodiments, q is 0. In some embodiments, n is an integer from 1 to 5. In some embodiments, q, m and p are each 1 and n is 1 , 2 or 5.
[0155] It is understood and clearly conveyed by this disclosure that each Ri, R2, R3, m, n, p and q disclosed in reference to Formula (I) intends and includes all combinations thereof the same as if each and every combination of R-i, R2, R3, m, n, p and q were specifically and individually listed.
[0156] Representative compounds of the Formula (I) include:
Figure imgf000048_0001
Figure imgf000049_0001
[0157] In certain embodiments, the polyamine compound is represented by the structure according to Formula (II):
Figure imgf000049_0002
[0158] or a salt, solvate or hydrate thereof,
[0159] where n is 1 , 2 or 3;
[0160] each L is independently a linker of from about 2 to 14 carbons in length, for example of about 2, 3, 4, 5, 6, 8, 10, 12 or 14 carbon atoms in length, where the linker backbone atoms may be saturated or unsaturated, usually not more than one, two, three, or four unsaturated atoms will be present in a tether backbone, where each of the backbone atoms may be substituted or unsubstituted (for example with a C1-C8 alkyl), where the linker backbone may include a cyclic group (for example, a cyclohex-1 ,3-diyl group where 3 atoms of the cycle are included in the backbone);
[0161] each R12 is independently selected from hydrogen and a C-i-Cs alkyl; and
[0162] each Rn is independently selected from hydrogen, C2-C8alkenyl, Ci- C8 alkyl or C3-C8 branched alkyl ( e.g ., methyl, ethyl, tert-butyl, isopropyl, pentyl, cyclobutyl, cyclopropylmethyl, 3-methylbutyl, 2-ethylbutyl, 5-NH2-pent-1-yl, propyl-1 - ylmethyl(phenyl)phosphinate, dimethylbicyclo[3.1.1 ]heptyl)ethyl, 2- (decahydronaphthyl)ethyl and the like), C6-C20 aryl or heteroaryl, C1-C23 aralkyl or heteroaralkyl (2-phenylbenzyl, 4-phenylbenzyl, 2-benzylbenzyl, 3-benzylbenzyl, 3,3- diphenylpropyl, 3-(benzoimidazolyl)-propyl, 4-isopropylbenzyl, 4-fluorobenzyl, 4-tert- butylbenzyl, 3-imidazolyl-propyl, 2-phenylethyl and the like), -C(=0)-Ci-C8, alkyl, -C(=0)-Ci-C8 alkenyl, -C(=0)-Ci-C8 alkynyl, an amino-substituted cycloalkyl {e.g., a cycloalkyl group substituted with a primary, secondary, tertiary or quaternary amine, such as 5-NH2-cycloheptyl, 3-NH2-cyclopentyl and the like) and a C2-C8 alkanoyl {e.g., an alkanoyl substituted with a methyl and an alkylazide group) .
[0163] In certain embodiments, each L is independently selected from: -CHRi3-(CH )m-, -CHRi3-(CH )n-CHRi3-, -(CH )mCHRi3-, -CH2-A-CH2- and -(CH2)p- where :
[0164] m is an integer from 1 to 5;
[0165] A is (CH2)m, ethane-1, 1-diyl or cyclohex-1 ,3-diyl;
[0166] p is an integer from 2 to 14, such as 1 , 2, 3, 4 or 5;
[0167] n is an integer from 1 to 12; and
[0168] Ri3 is a C-i-Cs alkyl.
[0169] A substituted aralkyl or heteroaralkyl with reference to Formula (II) intends and includes alkanoyl moieties substituted with an aryl or heteroaryl group, i.e., -C(=0)-aryl, -C(=0)-aralkyl, -C(=0)-heteroaryl, and -C(=0)-heteroaralkyl. In one embodiment, the alkyl portion of the aralkyl or heteroaralkyl moiety is connected to the molecule via its alkyl moiety. For instance at least one or both of Rn may be an aralkyl moiety such as 2-phenylbenzyl, 4-phenylbenzyl, 3,3,-diphenylpropyl, 2-(2- phenylethyl)benzyl, 2-methyl-3-phenylbenzyl, 2-napthylethyl, 4-(pyrenyl)butyl, 2-(3- methylnapthyl)ethyl, 2-(1 ,2-dihydroacenaphth-4-yl)ethyl and the like. In another embodiment, at least one or both of Rn may be a heteroaralkyl moiety such as 3-(benzoimidazolyl)propanoyl, 1 -(benzoimidazolyl)methanoyl, 2-(benzoimidazolyl)ethanoyl, 2-(benzoimidazolyl)ethyl and the like.
[0170] In certain embodiments, the compound of Formula (II) comprises at least one moiety selected from the group consisting of t-butyl, isopropyl, 2-ethylbutyl, 1-methylpropyl, 1-methylbutyl, 3-butenyl, isopent-2-enyl, 2-methylpropan-3-olyl, ethylthiyl, phenylthiyl, propynoyl, 1 -methyl-1 H-pyrrole-2-yl; trifluoromethyl, cyclopropanecarbaldehyde, halo-substituted phenyl, nitro-substituted phenyl, alkyl- substituted phenyl, 2,4,6-trimethylbenzyl, halo-5-substituted phenyl (such as para- (F3S)-phenyl, azido and 2-methylbutyl.
[0171] In certain embodiments, in Formula (II), each Rn is independently selected from hydrogen, n-butyl, ethyl, cyclohexylmethyl, cyclopentylmethyl, cyclopropylmethyl, cycloheptylmethyl, cyclohexyleth-2-yl, and benzyl.
[0172] In certain embodiments, the polyamine compound is of the structure of Formula (II), where n is 3, such that the compound has a structure according to Formula (III):
Figure imgf000051_0001
[0173] where Li, L2 and L3 are independently selected from -CHRi3-(CH )m-, -CHRi3-(CH2)n-CHRi3-, -(CH2)m-CHRi3-, -CH2-A-CH2- and -(CH2)p-;
[0174] where m, A, p, n and R13 are as defined above.
[0175] In certain embodiments, the polyamine compound is of the structure of Formula (III) where: Li is -CHRi3-(CH2)m-; l_2 is -CHRi3-(CH2)n-CHRi3-; and L3 is - (CH2)m-CHRi3-; where Rn, R12, R13, m and n are as defined above. [0176] In certain embodiments, the polyamine compound is of the structure of Formula (III) where: Li, l_2, and L3 are independently -CH2-A-CH2-; and R12 is hydrogen; where R11 and A are as defined above. In particular embodiments, at least one of an A and an Rn comprises an alkenyl moiety.
[0177] In certain embodiments, the polyamine compound is of the structure of Formula (III) where: Li, L2 and l_3are independently -(CFl2)p- where p is as defined above; and R121S hydrogen. In particular embodiments, for Li and L3, p is an integer from 3 to 7, and for L3 p is an integer from 3 to 14.
[0178] In certain embodiments, the polyamine compound is of the structure of Formula (III) where: Li, and L3 are independently -(CH2)P-; L2 is -CFI2-A-CFI2-; and R12 is hydrogen; where R12, p and A are as defined above. In particular embodiments, for Li and L3, p is an integer from 2 to 6, and for L3 A is (CFl2)xwhere x is an integer from 1 to 5, or cyclohex-1 ,3-diyl.
[0179] In certain embodiments, the polyamine compound is of the structure of Formula (II), where n is 2, such that the compound has a structure according to Formula (IV):
Figure imgf000052_0001
[0180] where Li and L2 are independently selected from -CHRi3-(CH2)m- CHRi3-(CH2)n-CHRi3-, -(CH2)n, CHR13-, -CH2-A-CH2- and -(CH2 )P-;
[0181] where m, A, p, n, and R13 are as defined above.
[0182] In certain embodiments, the polyamine compound is of the structure of Formula (IV) where: Li is -(CH2)P-; and L2 is -(CH2)m-CHRi3-; where R13, m and p are as defined above. In particular embodiments, for Li p is an integer from 3 to 10, and for L2 n is an integer from 2 to 9.
[0183] In certain embodiments, the polyamine compound is of the structure of Formula (IV) where: Li and L2 are -(CH2)P-; where p is as defined above. In particular embodiments, p is an integer from 3 to 7. [0184] In certain embodiments, the polyamine compound is of the structure of Formula (II), where n is 1 , such that the compound has a structure according to Formula (V);
Figure imgf000053_0001
[0185] where Li is -(CFl2)p- where p is as defined above. In particular embodiments, p is an integer from 2 to 6.
[0186] In particular embodiments, in formula (V), one Rn is an amino- substituted cycloalkyl ( e.g ., a cycloalkyl group substituted with a primary, secondary, tertiary or quaternary amine) or a C2-C8 alkanoyl (which alkanoyl may be substituted with one or more substituents such as a methyl or an alkylazide group); and the other R11 is a C-i-Cs alkyl or a C7-C24 aralkyl.
[0187] Representative compounds of the Formula (II) include, e.g.\
Figure imgf000053_0002
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
ZQW-16c
Figure imgf000057_0001
[0188] Phenylcyclopropylamine derivatives that are inhibitors of include compounds represented by Formula (VI):
Figure imgf000057_0002
[0189] wherein :
[0190] each of R1-R5 is independently selected from H, halo, alkyl, alkoxy, cycloalkoxy, haloalkyl, haloalkoxy, -L-aryl, -L-heterocyclyl, -L-carbocyclyl, acylamino, acyloxy, alkylthio, cycloalkylthio, alkynyl, amino, alkylamino, aryl, arylalkyl, arylalkenyl, arylalkynyl, arylalkoxy, aryloxy, arylthio, heteroarylthio, cyano, cyanato, haloaryl, hydroxyl, heteroaryloxy, heteroarylalkoxy, isocyanato, isothiocyanate, nitro, sulfinyl, sulfonyl, sulfonamide, thiocarbonyl, thiocyanato, trihalomethanesulfonamido, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, and C-amido;
[0191] R6 is H or alkyl; [0192] R7 is H, alkyl, or cycloalkyl;
[0193] R8 is a -L-heterocyclyl wherein the ring or ring system of the -L-heterocyclyl has from 0 to 3 substituents selected from halo, alkyl, alkoxy, cycloalkoxy, haloalkyl, haloalkoxy, -L-aryl, -L-heterocyclyl, -L-carbocyclyl, acylamino, acyloxy, alkylthio, cycloalkylthio, alkynyl, amino, alkylamino, aryl, arylalkyl, arylalkenyl, arylalkynyl, arylalkoxy, aryloxy, arylthio, heteroarylthio, cyano, cyanato, haloaryl, hydroxyl, heteroaryloxy, heteroarylalkoxy, isocyanato, isothiocyanate, nitro, sulfinyl, sulfonyl, sulfonamide, thiocarbonyl, thiocyanato, trihalomethanesulfonamido, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, and C-amido; or
[0194] R8 is -L-aryl wherein the ring or ring system of the -L-aryl has from 1 to 3 substituents selected from halo, alkyl, alkoxy, cycloalkoxy, haloalkyl, haloalkoxy, -L-aryl, -L-heterocyclyl, -L-carbocyclyl, acylamino, acyloxy, alkylthio, cycloalkylthio, alkynyl, amino, alkylamino, aryl, arylalkyl, arylalkenyl, arylalkynyl, arylalkoxy, aryloxy, arylthio, heteroarylthio, cyano, cyanato, haloaryl, hydroxyl, heteroaryloxy, heteroarylalkoxy, isocyanato, isothiocyanate, nitro, sulfinyl, sulfonyl, sulfonamide, thiocarbonyl, thiocyanato, trihalomethanesulfonamido, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, and C-amido;
[0195] where each L is independently selected from -(CH2)n- (CH2)n-, -(CH2)nNH(CH2)n-, -(CH2)nO(CH2)n-, and -(CH2)nS(CH2)n-, and where each n is independently chosen from 0, 1 , 2, and 3;
[0196] or a pharmaceutically acceptable salt thereof.
[0197] In some cases, L is a covalent bond. In some cases, R6 and R7 are hydro. In some cases, one of R1 -R5 is selected from -L-aryl, -L-heterocyclyl, and -L-carbocyclyl.
[0198] In some embodiments of the compound of Formula VI, the substituent or substituents on the R8 ring or ring system is/are selected from hydroxyl, halo, alkyl, alkoxy, cycloalkoxy, haloalkyl, haloalkoxy, -N(Ci-3 alkyl)2, -NH(Ci-3 alkyl), -C(=0)NH2, -C(=0)NH(Ci-3 alkyl), -C(=0)N(Ci-3 alkyl)2, -S(=0)2(CI-3 alkyl), -S(=0)2NH2, -S(0)2NH2, -S(0)2N(CI-3 alkyl)2, -S(=0)2NH(CI-3 alkyl), -CN, -NH2, and -NO2.
[0199] In certain embodiments, a compound of the invention is of Formula (VI) where: [0200] each R1 -R5 is optionally substituted and independently chosen from -H, halo, alkyl, alkoxy, cycloalkoxy, haloalkyl, haloalkoxy, -L-aryl, -L-heteroaryl, -L- heterocyclyl, -L-carbocyclyl, acylamino, acyloxy, alkylthio, cycloalkylthio, alkynyl, amino, aryl, arylalkyl, arylalkenyl, arylalkynyl, arylalkoxy, aryloxy, arylthio, heteroarylthio, cyano, cyanato, haloaryl, hydroxyl, heteroaryloxy, heteroarylalkoxy, isocyanato, isothiocyanato, nitro, sulfinyl, sulfonyl, sulfonamide, thiocarbonyl, thiocyanato, trihalomethanesulfonamido, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, and C-amido;
[0201] R6 is chosen from -H and alkyl;
[0202] R7 is chosen from -H, alkyl, and cycloalkyl;
[0203] R8 is chosen from -C(=0)NRxRy and -C(=0)Rz;
[0204] Rx when present is chosen from -H, alkyl, alkynyl, alkenyl, -L- carbocyclyl, -L-aryl, and -L-heterocyclyl, all of which are optionally substituted (except -H);
[0205] Ry when present is chosen from -H, alkyl, alkynyl, alkenyl, -L- carbocyclyl, -L-aryl, and -L-heterocyclyl, all of which are optionally substituted (except -H), where Rx and Ry may be cyclically linked;
[0206] Rz when present is chosen from -H, alkoxy, -L-carbocyclyl, -L- heterocyclyl, -L-aryl, wherein the aryl, heterocyclyl, or carbocyclyl are optionally substituted; each L is a linker that links the main scaffold of Formula I to a carbocyclyl, heterocyclyl, or aryl group, wherein the hydrocarbon portion of the linker -L- is saturated, partially saturated, or unsaturated, and is independently chosen from a saturated parent group having a formula of -(CH2)n-(CH2)2-,
-(CH2)nC(=0)(CH2)-, -(CH2)nC(=0)NH(CH2)n-, -(CH2)nNHC(0)0(CH2)n-, -(CH2)nNHC(=0)NH(CH2)n-, -(CH2)nNHC(=S)S(CH2)n-, -(CH)n0C(=0)S(CH2)n-, -(CH2)nNH(CH2)n-, -(CH2)n-0-(CH2)n-, -(CH2)nS(CH2)n-, and - (CH2)nNHC(=S)NH(CH2)n-, where each n is independently chosen from 0, 1 , 2, 3, 4, 5, 6, 7, and 8. According to this embodiment, optionally substituted refers to zero or 1 to 4 optional substituents independently chosen from acylamino, acyloxy, alkenyl, alkoxy, cycloalkoxy, alkyl, alkylthio, cycloalkylthio, alkynyl, amino, aryl, arylalkyl, arylalkenyl, arylalkynyl, arylalkoxy, aryloxy, arylthio, heteroarylthio, carbocyclyl, cyano, cyanato, halo, haloalkyl, haloaryl, hydroxyl, heteroaryl, heteroaryloxy, heterocyclyl, heteroarylalkoxy, isocyanato, isothiocyanato, nitro, sulfinyl, sulfonyl, sulfonamide, thiocarbonyl, thiocyanato, trihalomethanesulphonamido, O-carbamyl, N-carbamyl, O-thiocarbamyl, N- thiocarbamyl, and C-amido. In a more specific aspect of this embodiment, the optional substituents are 1 or 2 optional substituents chosen from halo, alkyl, aryl, and arylalkyl.
[0207] In certain embodiments, in Formula (VI), R8 is -CORz, such that the compound is of the following structure:
Figure imgf000060_0001
[0208] where: R1-R7 are described above; and Rz is -L-heterocyclyl which is optionally substituted with from 1-4 optional substituents independently chosen from acylamino, acyloxy, alkenyl, alkoxy, cycloalkoxy, alkyl, alkylthio, cycloalkylthio, alkynyl, amino, aryl, arylalkyl, arylalkenyl, arylalkynyl, arylalkoxy, aryloxy, arylthio, heteroarylthio, carbocyclyl, cyano, cyanato, halo, haloalkyl, haloaryl, hydroxyl, heteroaryl, heteroaryloxy, heterocyclyl, heteroarylalkoxy, isocyanato, isothiocyanato, nitro, sulfinyl, sulfonyl, sulfonamide, thiocarbonyl, thiocyanato, trihalomethanesulphonamido, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, and C-amido, and wherein said -L- is independently chosen from - (CH2)n-(CH2)n-, -(CH2)nNH(CH2)n-, -(CH2)n-0-(CH2)n-, and -(CH2)nS(CH2)n-, where each n is independently chosen from 0, 1 , 2, and 3.
[0209] In a specific aspect of this embodiment, each L is independently chosen from -(CH2)n-(CH2)n- and -(CH2)n-0-(CH2)n where each n is independently chosen from 0, 1 , 2, and 3. In a more specific aspect of this embodiment, each L is chosen from a bond, -CH2-, -CH2CH2-, -OCH2-, -OCH2CH2-, -CH2OCH2-, -CH2CH2CH2-, -OCH2CH2CH2-, and -CH2OCH2CH2-. In an even more specific aspect, each L is chosen from a bond, -CH2-, -CH2CH2-, OCH2-, and -CH2CH2CH2-. In yet an even more specific aspect, L is chosen from a bond and -CH2-.
[0210] Exemplary compounds of Formula (VI) include:
Figure imgf000061_0001
[0211] Exemplary compounds of Formula VI include: N-cyclopropyl-2- {[(trans)-2-phenylcyclopropyl]amino}acetamide; 2-{[(trans)-2- phenylcyclopropyl]amino acetamide; N-cyclopropyl-2-{[(trans)-2- phenylcyclopropyl]amino}propanamide; 2-{[(trans)-2-phenylcyclopropyl]amino}-N- prop-2-ynylacetamide; N-isopropyl-2-{[(trans)-2-phenylcyclopropyl]amino}acetamide; N-(tert-butyl)-2-{[(trans)-2-phenylcyclopropyl]amino}acetamide; N-(2-morpholin-4-yl- 2-oxoethyl)-N-[(trans)-2-phenylcyclopropyl]amine; 2-{[(trans)-2- phenylcyclopropyl]amino}propanamide; methyl 2-{[(trans)-2- phenylcyclopropyl]amino}propanoate; N-cyclopropyl-2-{methyl[(trans)-2- phenylcyclopropyl]amino}acetamide; 2-{methyl[(trans)-2- phenylcyclopropyl]amino}acetamide; N-methyl-trans-2- (phenylcyclopropylamino)propanamide; 1-(4-methylpiperazin-l-yl)-2-((trans)-2- phenylcyclopropylamino)ethanone; 1-(4-ethylpiperazin-l-yl)-2-((trans)-2- phenylcyclopropylamino)ethanone; 1-(4-benzylpiperazin-l-yl)-2-((trans)-2- phenylcyclopropylamino)-ethanone; 2-((trans)-2-phenylcyclopropylamino)-1-(4- phenylpiperazin-1-yl)ethanone; 2-((trans)-2-(4-(benzyloxy)phenyl)cyclopropylamino)- l-(4-methylpiperazin-1 -yl)ethanone; 2-((trans)-2-(4-
(benzyloxy)phenyl)cyclopropylamino)-N-cyclopropylacetamide; 2-((trans)-2-(4-(3- fluorobenzyloxy)phenyl)cyclopropylamino)-l-(4- -methylpiperazin-l-yl)ethanone; 2- ((trans)-2-(4-(3-chlorobenzyloxy)phenyl)cyclopropylamino)-1 -(4-methylpi- perazin-1 - yl)ethanone; 2-((trans)-2-(biphenyl-4-yl)cyclopropylamino)-1 -(4-methylpiperazin-1 - yl)ethanone; 1 -(4-methylpiperazin-1 -yl)-2-((trans)-2-(4- phenethoxyphenyl)cyclopropylamino)ethanone; 2-((trans)-2-(4-(4- fluorobenzyloxy)phenyl)cyclopropylamino)-1 -(4-methylpiperazin-1 -yl)ethanone; 2- ((trans)-2-(4-(biphenyl-4-ylmethoxy)phenyl)cyclopropylamino)-1 -(4-methylpiperazin- 1 -yl)ethanone; (trans)-N-(4-fluorobenzyl)-2-phenylcyclopropanamine; (trans)-N-(4- fluorobenzyl)-2-phenylcyclopropanaminiurn; 4-(((trans)-2- phenylcyclopropylamino)methyl)benzonitrile; (trans)-N-(4-cyanobenzyl)-2- phenylcyclopropanaminium; (trans)-2-phenyl-N-(4- (trifluoromethyl)benzyl)cyclopropanamine; (trans)-2-phenyl-N-(4- (trifluoromethyl)benzyl)cyclopropanaminium; (trans) -2-phenyl-N-(pyridin-2- ylmethyl)cyclopropanamine; (trans)-2-phenyl-N-(pyridin-3- ylmethyl)cyclopropanamine; (trans)-2-phenyl-N-(pyridin-4- ylmethyl)cyclopropanamine; (trans)-N-((6-methylpyridin-2 yl)methyl)-2- phenylcyclopropanamine; (trans)-2-phenyl-N-(thiazol-2-ylmethyl)cyclopropanamine; (trans)-2-phenyl-N-(thiophen-2-ylmethyl)cyclopropanamine; (trans)-N-((3- bromothiophen-2-yl)methyl)-2-phenylcyclopropanamine; (trans)-N-((4- bromothiophen-2-yl)methyl)-2-phenylcyclopropanamine; (trans)-N-(3,4- dichlorobenzyl)-2-phenylcyclopropanamine; (trans)-N-(3-fluorobenzyl)-2- phenylcyclopropanaminium; (trans)-N-(2-fluorobenzyl)-2-phenylcyclopropanamine; (trans)-2-phenyl-N-(quinolin-4-ylmethyl)cyclopropanaraine; (trans)-N-(3- methoxybenzyl)-2-phenylcyclopropanamine; (trans)-2-phenyl-N-((6- (tnfluoromethyl)pyridin-3-yl)methyl)cyclopropanamine; (trans)-N-((6-chloropyridin-3- yl)methyl)-2-phenylcyclopropanamine; (trans)-N-((4-methylpyndin-2-yl)methyl)-2- phenylcyclopropanamine; (trans)-N-((6-methoxypyridin-2-yl)methyl)-2- phenylcyclopropanamine; 2-(((trans)-2-phenylcyclopropylamino)methyl)pyridin-3-ol; (trans)-N-((6-bromopyridin-2-yl)methyl)-2-phenylcyclopropanamine; 4-(((trans)-2- (4(benzyloxy)phenyl)cyclopropylamino)methyl)benzonitrile; (trans) -N-(4- (benzyloxy)benzyl)-2-phenylcyclopropanamine; (trans)-N-benzyl-2-(4- (benzyloxy)phenyl)cyclopropanamine; (trans)-2-(4-(benzyloxy)phenyl)-N-(4- methoxybenzyl)cyclopropanamine; (trans)-2-(4-(benzyloxy)phenyl)-N-(4- fluorobenzyl)cyclopropanamine; (trans)-2-phenyl-N-(quinolin-2- ylmethyl)cyclopropanamine; (trans)-2-phenyl-N-((5-(trifluoromethyl)pyridin-2- yl)methyl)cyclopropanamine; (trans)-N-((3-fluoropyridin-2-yl)methyl)-2- phenylcyclopropanamine; (trans)-2-phenyl-N-(quinolin-3-ylmethyl)cyclopropanamine; (trans)-N-((6-methoxypyridin-3-yl)methyl)-2-phenylcyclopropanamine; (trans)-N-((5- methoxypyridin-3-yl)methyl)-2-phenylcyclopropanamine; (trans)-N-((2- methoxypyridin-3-yl)methyl)-2-phenylcyclopropanamine; (trans)-N-((3H-indol-3- yl)methyl)-2-phenylcyclopropanamine; 3-(((trans)-2- phenylcyclopropylamino)methyl)benzonitrile; (trans)-N-(2-methoxybenzyl)-2- phenylcyclopropanamine; 3-(((trans)-2-phenylcyclopropylamino)methyl)pyndin-2- amine; (trans)-N-((2-chloropyridin-3-yl)methyl)-2-phenylcyclopropanamine; (trans)-N- (3,4-dimethoxybenzyl)-2-phenylcyclopropanamine; (trans)-N-((2,3- dihydrobenzofuran-5-yl)methyl)-2-phenylcyclopropanamine; (trans)-N- (benzo[d][1 ,3]dioxol-5-ylmethyl)-2-phenylcyclopropanamine; (trans)-N-((2,3- dihydrobenzo[b][1 ,4]dioxin-6-yl)methyl)-2-phenyl-cyclopropanamine; (trans)-N-(2,6- difluoro-4-methoxybenzyl)-2-phenylcyclopropanamine; (trans)-2-phenyl-N-(4- (trifluoromethoxy)benzyl)cyclopropanamine; (trans)-N-(5-fluoro-2-methoxybenzyl)-2- phenylcyclopropanamine; (trans)-N-(2-fluoro-4-methoxybenzyl)-2- phenylcyclopropanamine; (trans)-N-((4-methoxynaphthalen-1-yl)methyl)-2- phenylcyclopropanamine; (trans)-N-(2-fluoro-6-methoxybenzyl)-2- phenylcyclopropanamine; (trans)-N-((2-methoxynaphthalen-1-yl)methyl)-2- phenylcyclopropanamine; (trans)-N-((4,7-dimethoxynaphthalen-1-yl)methyl)-2- phenylcyclopropanamine- ; (trans)-N-(4-methoxy-3-methylbenzyl)-2- phenylcyclopropanamine; (trans)-N-(3-chloro-4-methoxybenzyl)-2- phenylcyclopropanamine; (trans)-N-(3-fluoro-4-methoxybenzyl)-2- phenylcyclopropanamine; (trans)-N-(4-methoxy-2-methylbenzyl)-2- phenylcyclopropanamine; (trans)-N-((3,4-dihydro-2H-benzo[b][1 ,4]dioxepin-6- yl)methyl)-2-phenylcyclopropanamine; (trans)-N-((3,4-dihydro-2H- benzo[b][1 ,4]dioxepin-7-yl)methyl)-2-phenylcyclopropanamine; (trans)-N-((2,2- dimethylchroman-6-yl)methyl)-2-phenylcyclopropanamine; (trans)-N-(4-methoxy-2,3- dimethylbenzyl)-2-phenylcyclopropanamine; (trans)-N-(4-methoxy-2,5- dimethylbenzyl)-2-phenylcyclopropanamine; (trans)-N-(2-fluoro-4,5- dimethoxybenzyl)-2-phenylcyclopropanamine; (trans)-N-(3-chloro-4,5- dimethoxybenzyl)-2-phenylcyclopropanamine; (trans)-N-(2-chloro-3,4- dimethoxybenzyl)-2-phenylcyclopropanamine; (trans)-N-(2,4-dimethoxy-6- methylbenzyl)-2-phenylcyclopropanamine; (trans)-N-(2,5-dimethoxybenzyl)-2- phenylcyclopropanamine; (trans)-N-(2,3-dimethoxybenzyl)-2- phenylcyclopropanamine; (trans)-N-(2-chloro-3-methoxybenzyl)-2- phenylcyclopropanamine; (trans)-N-((1 H-indol-5-yl)methyl)-2- phenylcyclopropanamine; (trans)-2-(4-(benzyloxy)phenyl)-N-(pyridin-2- ylmethyl)cyclopropanamine; (trans)-2-(4-(benzyloxy)phenyl)-N-(2- methoxybenzyl)cyclopropanamine; (trans)-N-(l-(4-methoxyphenyl)ethyl)-2- phenylcyclopropanaraine; (trans)-N-(1-(3,4-dimethoxyphenyl)ethyl)-2- phenylcyclopropanamine; (trans)-N-(l -(2,3-dihydrobenzo[b][1 ,4]dioxin-6-yl)ethyl)-2- phenylcyclopropanamine; (trans)-N-(1-(5-fluoro-2-methoxyphenyl)ethyl)-2- phenylcyclopropanamine; (trans)-N-(1-(3,4-dimethoxyphenyl)propan-2-yl)-2- phenylcyclopropan- amine; (trans)-N-((3-methyl-1 ,2,4-oxadiazol-5-yl)methyl)-2- phenylcyclopropanamine;
[0212] and pharmaceutically acceptable salts thereof.
[0213] Alternative small molecule LSD inhibitor compounds may be selected from selective LSD1 and LSD1/MAOB dual inhibitors disclosed for example in International PCT Patent Pub. No. WO 2010/043,721 , WO 2010/084,160,
WO 2011 /035,942; WO 2011 /042.217; and EP Application No. EP10171345, all of which are explicitly incorporated herein by reference in their entireties to the extent they are not inconsistent with the present disclosure. Representative compounds of this type include phenylcyclopropylamine derivatives or homologs, illustrative examples of which include phenylcyclopropylamine with one or two substitutions on the amine group; phenylcyclopropylamine with zero, one or two substitutions on the amine group and one, two, three, four, or five substitution on the phenyl group; phenylcyclopropylamine with one, two, three, four, or five substitution on the phenyl group; phenylcyclopropylamine with zero, one or two substitutions on the amine group wherein the phenyl group of PCPA is substituted with (exchanged for) another ring system chosen from aryl or heterocyclyl to give an aryl- or heteroaryl- cyclopropylamine having zero, one or two substituents on the amine group; phenylcyclopropylamine wherein the phenyl group of PCPA is substituted with (exchanged for) another ring system chosen from aryl or heterocyclyl to give an aryl- or heterocyclyl-cyclopropylamine wherein said aryl- or heterocyclyl-cyclopropylamine on said aryl or heterocyclyl moiety has zero, one or two substitutions on the amine group and one, two, three, four, or five substitution on the phenyl group; phenylcyclopropylamine with one, two, three, four, or five substitution on the phenyl group; or any of the above described phenylcyclopropylamine analogues or derivatives wherein the cyclopropyl has one, two, three or four additional substituents. Suitably, the heterocyclyl group described above in this paragraph in a heteroaryl.
[0214] Non-limiting embodiments of phenylcyclopropylamine derivatives or analogues include “cyclopropylamine amide” derivatives and “cyclopropylamine” derivatives. Specific examples of “cyclopropylamine acetamide” derivatives include, but are not limited to: N-cyclopropyl-2-{[(trans)-2-phenylcyclopropyl]amino} acetamide; 2-{[(trans)-2-phenylcyclopropyl]amino}acetamide; N-cyclopropyl-2- {[(trans)-2-phenylcyclopropyl]amino}propanamide; 2-{[(trans)-2-phenylcyclopropyl] amino}-N-prop-2-ynylacetamide; N-isopropyl-2-{[(trans)-2-phenylcyclopropyl]amino} acetamide; N-(tert-butyl)-2-{[(trans)-2-phenylcyclopropyl]amino}acetamide; N-(2- morpholin-4-yl-2-oxoethyl)-N-[(trans)-2-phenylcyclopropyl]amine; 2-{[(trans)-2- phenylcyclopropyl]amino}propanamide; Methyl 2-{[(trans)-2-phenylcyclopropyl] aminojpropanoate; 1 -(4-methylpiperazin-1 -yl)-2-((trans)-2-phenylcyclopropylamino) ethanone; 1 -(4-ethylpiperazin-1 -yl)-2-((trans)-2-phenylcyclopropylamino)ethanone; 1 - (4-benzylpiperazin-1 -yl)-2-((trans)-2-phenylcyclopropylamino)ethanone; 2-((trans)-2- phenylcyclopropylamino)-1 -(4-phenylpiperazin-1 -yl)ethanone; 2-((trans)-2-(4- (benzyloxy)phenyl)cyclopropylamino)-1 -(4-methylpiperazin-1 -yl)ethanone; 2-((trans)- 2-(1 ,1 '-biphenyl-4-yl)cyclopropylamino)-1 -(4-methylpiperazin-1 -yl)ethanone; 2- ((trans)-2-(4- (benzyloxy)phenyl)cyclopropylamino)-N-cyclopropylacetamide; 2- ((trans)-2-(4-(3-fluorobenzyloxy)phenyl)cyclopropylamino)-1 -(4-methylpiperazin-1 - yl)ethanone; 2-((trans)-2-(4-(4-fluorobenzyloxy)phenyl)cyclopropylamino)-1 -(4- methylpi- perazin-1 -yl)ethanone; 2-((trans)-2-(4-(3-chlorobenzyloxy)phenyl) cyclopropylamino)-1 -(4-methylpiperazin-1 -yl)ethanone; 1 -(4-methylpiperazin-1 -yl)-2- ((trans)-2-(4-phenethoxyphenyl)cyclopropylamino)ethanone; 2-((trans)-2-(biphenyl-4- yl)cyclopropylamino)-1 -(4-methylpiperazin-1 -yl)ethanone; N-cyclopropyl-2-{[(trans)-2- phenylcyclopropyl]amino}acetamide; N-methyl-trans-2-(Phenylcyclopropylamino) propanamide; 2-{methyl[(trans)-2-phenylcyclopropyl]amino}acetamide; N-[2-(4- methylpiperazin-1 -yl)ethyl]-N-[(trans)-2-phenylcyclopropyl]amine; N-cyclopropyl-N'- [(trans)-2-phenylcyclopropyl]ethane-1 ,2-diamine; N,N-dimethyl-N'-(2-{[(trans)-2- phenylcyclopropyl]amino}ethyl)ethane-1 ,2-diamine; (3R)-1 -(2-{[(trans)-2- phenylcyclopropyl]amino}ethyl)pyrrolidin-3-amine; (3S)-N,N-dimethyl-1-(2-{[(trans)-2- phenylcyclopropyl]amino}ethyl) pyrrolidin-3-amine; (3R)-N,N-dimethyl-1 -(2-{[(trans)- 2-phenylcyclopropyl]amino}ethyl) pyrrolidin-3-amine; N-[(trans)-2-phenylcyclopropyl]- N-(2-piperazin-1 -ylethyl) amine; N,N-diethyl-N'-[(trans)-2-phenylcyclopropyl]ethane- 1 ,2-diamine; N-[(trans)-2-phenylcyclopropyl]-N-(2-piperidin-1 -ylethyl)amine; (trans)- 2-(4-(benzyloxy)phenyl)-N-(2-(4-methylpiperazin-1-yl)ethyl)cyclopropanamine; (trans)-N-(2-(4-methylpiperazin-1-yl)ethyl)-2-(3'-(trifluoromethyl)biphenyl-4- yl)cyclopropanamine; (trans)-2-(3'-chlorobiphenyl-4-yl)-N-(2-(4-methylpiperazin-1- yl)ethyl)cyclopropanamine; (R)-1-(2-((trans)-2-(3'-(trifluoromethyl)biphenyl-4- yl)cyclopropylamino) ethyl)pyrrolidin-3-amine; and N1-cyclopropyl-N2-((trans)-2-(3'- (trifluoromethyl)biphenyl-4-yl-)cyclopropyl)ethane-1 ,2-diamine.
[0215] Specific examples of “cyclopropylamine” derivatives, include, but are not limited to: N-4-fluorobenzyl-N-{(trans)-2-[4-(benzyloxy)phenyl]cyclopropyl}amine, N-4-methoxybenzyl-N-{(trans)-2-[4-(benzyloxy)phenyl]cyclopropyl}amine, N-benzyl- N-{(trans)-2-[4-(benzyloxy)phenyl]cyclopropyl}amine, N-[(trans)-2- phenylcyclopropyl]amino-methyl)pyridin-3-ol, N-[(trans)-2-phenylcyclopropyl]-N-(3- methylpyridin-2-ylmethyl)amine, N-[(trans)-2-phenylcyclopropyl]-N-(4-chloropyridin-3- ylmethyl)amine, N-[(trans)-2-phenylcyclopropyl]-N-(4-trifluoromethylpyridin-3-yl- methyl)amine, N-(3-methoxybenzyl)-N-[(trans)-2-phenylcyclopropyl]amine, N- [(trans)-2-phenylcyclopropyl]-N-(quinolin-4-ylmethyl)amine, N-(2-fluorobenzyl)-N- [(trans)-2-phenylcyclopropyl]amine, N-(3-fluorobenzyl)-N-[(trans)-2- phenylcyclopropyl]amine, N-[(trans)-2-phenylcyclopropyl]-N-(3,4-dichloro-1- phenylmethyl)amine, N-[(trans)-2-phenylcyclopropyl]-N-(5-bromo-thiophen-2- ylmethyl)amine, N-[(trans)-2-phenylcyclopropyl]-N-(3-bromo-thiophen-2-ylmethyl)- amine, N-[(trans)-2-phenylcyclopropyl]-N-(thiophen-2-ylmethyl)amine, N-[(trans)-2- phenylcyclopropyl]-N-(1 ,3-thiazol-2-ylmethyl)amine, N-[(trans)-2-phenylcyclopropyl]- N-(3-methyl-pyridin-2-ylmethyl)amine, N-[(trans)-2-phenylcyclopropyl]-N-(pyridin-4- ylmethyl)amine, N-[(trans)-2-phenylcyclopropyl]-N-(pyridin-3-ylmethyl)amine, N- [(trans)-2-phenylcyclopropyl]-N-(pyridin-2-ylmethyl)amine, [(trans)-2- phenylcyclopropyl]-N-[4-(trifluoromethyl)benzyl]amine, ({[(trans)-2- phenylcyclopropyl]amino}methyl)benzonitrile, N-(4-fluorobenzyl)-N-[(trans)-2- phenylcyclopropyl]amine, N-[(trans)-2-phenylcyclopropyl]-N-(3-bromo-pyridin-2- ylmethyl)amine, N-4-cyanobenzyl-N-{(trans)-2-[4-
(benzyloxy)phenyl]cyclopropyl}amine, N-4-[(benzyloxy)-benzyl]-N-[(trans)-2-(4- phenyl)cyclopropyl]amine; 2-((trans)-2-(4-(4- cyanobenzyloxy)phenyl)cyclopropylamino)acetamide, 2-((trans)-2-(4-(3- cyanobenzyloxy)phenyl)cyclopropylamino)acetamide, 2-((trans)-2-(4- (benzyloxy)phenyl)cyclopropylamino)acetamide, 2-((trans)-2-(4-(4- fluorobenzyloxy)phenyl)cyclopropylamino)acetamide, 2-((trans)-2-(4-(3- fluorobenzyloxy)phenyl)cyclopropylamino)acetamide, 2-((trans)-2-(4-(3- chlorobenzyloxy)phenyl)cyclopropylamino)acetamide, 2-((trans)-2-(4-(4- chlorobenzyloxy)phenyl)cyclopropylamino)acetamide, 2-((trans)-2-(4-(3- bromobenzyloxy)phenyl)cyclopropylamino)acetamide, 2-((trans)-2-(4-(3,5- difluorobenzyloxy)phenyl)cyclopropylamino)acetamide, 2-((trans)-2-(4- phenethoxyphenyl)cyclopropylamino)acetamide, 2-((trans)-2-(3'- (trifluoromethyl)biphenyl-4-yl)cyclopropylamino) acetamide, and 2-((trans)-2-(3'- chlorobiphenyl-4-yl)cyclopropylamino)acetamide.
[0216] [0249] Other examples of LSD1 inhibitors are, e.g., phenelzine or pargyline (propargylamine) or a derivative or analogue thereof. Derivatives and analogues of phenelzine and pargyline (propargylamine) include, but are not limited to, compounds where the phenyl group of the parent compound is replaced with a heteroaryl or optionally substituted cyclic group or the phenyl group of the parent compound is optionally substituted with a cyclic group. In one aspect, the phenelzine or pargyline derivative or analogue thereof has selective LSD1 or dual LSD1/MAOB inhibitory activity as described herein. In some embodiments, the phenelzine derivative or analogue has one, two, three, four or five substituents on the phenyl group. In one aspect, the phenelzine derivative or analogue has the phenyl group substituted with (exchanged for) an aryl or heterocyclyl group wherein said aryl or heterocyclyl group has zero, one, two, three, four or five substituents. In one aspect, the pargyline derivative or analogue has one, two, three, four or five substituents on the phenyl group. In one aspect, the pargyline derivative or analogue has the phenyl group substituted with (exchanged for) an aryl or heterocyclyl group wherein said aryl or heterocyclyl group has zero, one, two, three, four or five substituents.
Methods of preparing such compounds are known to the skilled artisan. By way of an illustrative example, one such derivative of phenelzine that is particularly suitable for use with the present invention is bizine.
[0217] The present invention also contemplates tranylcypromine derivatives as described for example by Binda etal., (2010. J . Am. Chem. Soc. 132: 6827- 6833, which is hereby incorporated by reference herein in its entirety) as inhibitors of LSD ( e.g ., LSD1 and/or LSD2) enzymatic function. Non-limiting example of such compounds include:
Figure imgf000068_0001
[0218] Alternatively, LSD1 inhibitor compounds may be selected from tranylcypromine analogues described by Benelkebir etal. (2011. Bioorg. Med. Chem. doi: 10.1016/j.bmc.2011.02.017, which is hereby incorporated by reference herein in its entirety), Representative analogues of this type, including o-, m-, and p- bromo analogues include: (1 R, 2S)-2-(4-bromophenyl)cyclopropanamine hydrochloride (Compound 4c), (1 R, 2S)-2-(3-bromophenyl)cyclopropanamine hydrochloride (Compound 4d), : (1 R, 2S)-2-(2-bromophenyl)cyclopropanamine hydrochloride (Compound 4e), : (1 R, 2S)-2-(biphenyl-4-yl)cyclopropanamine hydrochloride (Compound 4f). [0219] Reference also may be made to peptide scaffold compounds disclosed by Culhane etal. (2010. J. Am. Chem. Soc. 132: 3164-3176, which is hereby incorporated by reference herein in its entirety), which include chlorovinyl, endo-cyclopropylamine, and hydrazine functionalities. Non-limiting compounds disclosed by Culhane etal., include propargyl-Lys-4, N-methylpropargyl-Lys-4 H3- 21 , c/s-3-chloroallyl-Lys-4 H3-21 , trans- 3-chloroallyl-Lys-4 H3-21 , exo-cyclopropyl- Lys-4 H3-21 , endo- cyclopropyl-Lys-4 H3-21 , enc/o-dimethylcyclopropyl-Lys-4, hydrazino-Lys-4 H3-21 and hydrazino-Lys-4 H3-21.
[0220] Alternative cyclopropylamine compounds that are useful for inhibiting LSD1 include those disclosed by Fyfe etal., in U.S. Pat. Pub. No. 2013/0197013, which is incorporated herein by reference in its entirety. Illustrative cyclopropylamine inhibitors of LSD1 , which are disclosed as being selective for inhibiting LSD1 , include compounds according to Formula VII:
Figure imgf000069_0001
[0221] wherein :
[0222] E is -N(R3)-, -0-, or -S-, or is -X3=X4-;
[0223] X1 and X2 are independently C(R2) or N;
[0224] X3 and X4, when present, are independently C(R2) or N;
[0225] (G) is a cyclyl group (as shown in Formula VII, the cyclyl group (G) has n substituents (R1));
[0226] each (R1) is independently chosen from alkyl, alkenyl, alkynyl, cyclyl, -L1 -cyclyl, -L1 -amino, -L1 -hydroxyl, amino, amido, nitro, halo, haloalkyl, haloalkoxy, cyano, sulfinyl, sulfonyl, sulphonamide, hydroxyl, alkoxy, urea, carbamate, acyl, or carboxyl;
[0227] each (R2) is independently chosen from -H, alkyl, alkenyl, alkynyl, cyclyl, -L1 -cyclyl, -L1 -amino, -L1 -hydroxyl, amino, amido, nitro, halo, haloalkyl, haloalkoxy, cyano, sulfinyl, sulfonyl, sulphonamide, hydroxyl, alkoxy, urea, carbamate, acyl, or carboxyl, wherein each (R2) group has 1 , 2, or 3 independently chosen optional substituents or two (R2) groups can be taken together to form a heterocyclyl or aryl group having 1 , 2, or 3 independently chosen optional substituents, wherein said optional substituents are independently chosen from alkyl, alkanoyl, heteroalkyl, heterocyclyl, haloalkyl, cycloalkyl, carbocyclyl, arylalkoxy, heterocyclylalkoxy, aryl, aryloxy, heterocyclyloxy, alkoxy, haloalkoxy, oxo, acyloxy, carbonyl, carboxyl, carboxamido, cyano, halogen, hydroxyl, amino, aminoalkyl, amidoalkyl, amido, nitro, thiol, alkylthio, arylthio, sulphonamide, sulfinyl, sulfonyl, urea, or carbamate;
[0228] R3 is -H or a (Ci-C6)alkyl group;
[0229] each L1 is independently alkylene or heteroalkylene; and
[0230] n is 0, 1 , 2, 3, 4 or 5,
[0231] or an enantiomer, a diastereomer, or a mixture thereof, or a pharmaceutically acceptable salt or solvate thereof.
[0232] In some embodiments, compounds of Formula (VII) are represented by Formula (VIII):
Figure imgf000070_0001
[0233] wherein:
[0234] X1 is CH or N; (G) is a cyclyl group;
[0235] each (R1) is independently chosen from alkyl, alkenyl, alkynyl, cyclyl, -L1 -cyclyl, -L1 -amino, -L1 -hydroxyl, amino, amido, nitro, halo, haloalkyl, haloalkoxy, cyano, sulfinyl, sulfonyl, sulphonamide, hydroxyl, alkoxy, urea, carbamate, acyl, or carboxyl;
[0236] each (R2) is independently chosen from alkyl, alkenyl, alkynyl, cyclyl, -L1 -cyclyl, -L1 -amino, -L1 -hydroxyl, amino, amido, nitro, halo, haloalkyl, haloalkoxy, cyano, sulfinyl, sulfonyl, sulphonamide, hydroxyl, alkoxy, urea, carbamate, acyl, or carboxyl, wherein each (R2) group has 1 , 2, or 3 optional substituents, wherein said optional substituents are independently chosen from alkyl, alkanoyl, heteroalkyl, heterocyclyl, haloalkyl, cycloalkyl, carbocyclyl, arylalkoxy, heterocyclylalkoxy, aryl, aryloxy, heterocyclyloxy, alkoxy, haloalkoxy, oxo, acyloxy, carbonyl, carboxyl, carboxamido, cyano, halogen, hydroxyl, amino, aminoalkyi, amidoalkyl, amido, nitro, thiol, alkylthio, arylthio, sulfonamide, sulfinyl, sulfonyl, urea, or carbamate;
[0237] each L1 is independently alkylene or heteroalkylene;
[0238] m is 0, 1 , 2 or 3; and n is 0, 1 , 2, 3, 4 or 5, provided that n and m are chosen independently such that n+m is greater than zero when X1 is -CH- and (G) is an aryl,
[0239] or an enantiomer, a diastereomer, or a mixture thereof, or a pharmaceutically acceptable salt or solvate thereof.
[0240] In other embodiments, compounds of Formula (VII) are represented by Formula (IX):
Figure imgf000071_0001
[0241] wherein:
[0242] (G) is a cyclyl group;
[0243] each (R1) is independently chosen from alkyl, alkenyl, alkynyl, cyclyl, -L1 -cyclyl, -L1 -amino, -L1 -hydroxyl, amino, amido, nitro, halo, haloalkyl, haloalkoxy, cyano, sulfinyl, sulfonyl, sulphonamide, hydroxyl, alkoxy, urea, carbamate, acyl, or carboxyl;
[0244] each (R2) is independently chosen from alkyl, alkenyl, alkynyl, cyclyl, -L1 -cyclyl, -L1 -amino, -L1 -hydroxyl, amino, amido, nitro, halo, haloalkyl, haloalkoxy, cyano, sulfinyl, sulfonyl, sulphonamide, hydroxyl, alkoxy, urea, carbamate, acyl, or carboxyl, wherein each (R2) group has 0, 1 , 2, or 3 optional substituents, wherein said optional substituents are independently chosen from alkyl, alkanoyl, heteroalkyl, heterocyclyl, haloalkyl, cycloalkyl, carbocyclyl, arylalkoxy, heterocyclylalkoxy, aryl, aryloxy, heterocyclyloxy, alkoxy, haloalkoxy, oxo, acyloxy, carbonyl, carboxyl, carboxamido, cyano, halogen, hydroxyl, amino, aminoalkyi, amidoalky, amido, nitro, thiol, alkylthio, arylthio, sulfonamide, sulfinyl, sulfonyl, urea, or carbamate;
[0245] each L1 is independently alkylene or heteroalkylene; m is 0, 1 , 2 or 3; and
[0246] n is 0, 1 , 2, 3, 4 or 5,
[0247] or an enantiomer, a diastereomer, or a mixture thereof, or a pharmaceutically acceptable salt or solvate thereof.
[0248] In still other embodiments, compounds of Formula (VII) are represented by Formula (X):
Figure imgf000072_0001
[0249] wherein:
[0250] E is -N(R3)-, -0-, or -S-, or is -X3=X4-;
[0251] X1, X2, X3 and X4 are independently C(R2) or N, provided that at least one of X1, X2, X3and X4is N when E is -X3=X4-;
[0252] (G) is a cyclyl group; each (R1 ) is independently chosen from alkyl, alkenyl, alkynyl, cyclyl, -L1 -cyclyl, -L1 -amino, -L1 -hydroxyl, amino, amido, nitro, halo, haloalkyi, haloalkoxy, cyano, sulphinyl, sulphonyl, sulphonamide, hydroxyl, alkoxy, urea, carbamate, acyl, or carboxyl;
[0253] each (R2) is independently chosen from alkyl, alkenyl, alkynyl, cyclyl, - L1 -cyclyl, -L1 -amino, -L1 -hydroxyl, amino, amido, nitro, halo, haloalkyi, haloalkoxy, cyano, sulphinyl, sulphonyl, sulphonamide, hydroxyl, alkoxy, urea, carbamate, acyl, or carboxyl, wherein each (R2) group has 1 , 2, or 3 optional substituents, wherein said optional substituents are independently chosen from alkyl, alkanoyl, heteroalkyl, heterocyclyl, haloalkyi, cycloalkyi, carbocyclyl, arylalkoxy, heterocyclylalkoxy, aryl, aryloxy, heterocyclyloxy, alkoxy, haloalkoxy, oxo, acyloxy, carbonyl, carboxyl, carboxamido, cyano, halogen, hydroxyl, amino, aminoalkyi, amidoalkyi, amido, nitro, thiol, alkylthio, arylthio, sulfonamide, sulfinyl, sulfonyl, urea, or carbamate;
[0254] R3 is -H or a (Ci-C6)alkyl group; each L1 is alkylene or heteroalkylene; and n is 0, 1 , 2, 3, 4 or 5,
[0255] or an enantiomer, a diastereomer, or a mixture thereof, or a pharmaceutically acceptable salt or solvate thereof.
[0256] In still other embodiments, compounds of Formula (VII) are represented by Formula (XI):
Figure imgf000073_0001
[0257] wherein:
[0258] X1, X2, X3and X4are independently CH or N, provided that at least one of X1, X2, X3 and X4 is N;
[0259] (G) is a cyclyl group; each (R1 ) is independently chosen from alkyl, alkenyl, alkynyl, cyclyl, -L1 -cyclyl, -L1 -amino, -L1 -hydroxyl, amino, amido, nitro, halo, haloalkyl, haloalkoxy, cyano, sulfinyl, sulphonyl, sulphonamide, hydroxyl, alkoxy, urea, carbamate, acyl, or carboxyl;
[0260] each (R2) is independently chosen from alkyl, alkenyl, alkynyl, cyclyl, -L1 -cyclyl, -L1 -amino, -L1 -hydroxyl, amino, amido, nitro, halo, haloalkyl, haloalkoxy, cyano, sulphinyl, sulphonyl, sulphonamide, hydroxyl, alkoxy, urea, carbamate, acyl, or carboxyl, wherein each (R2) group has 1 , 2, or 3 optional substituents, wherein said optional substituents are independently chosen from alkyl, alkanoyl, heteroalkyl, heterocyclyl, haloalkyl, cycloalkyl, carbocyclyl, arylalkoxy, heterocyclylalkoxy, aryl, aryloxy, heterocyclyloxy, alkoxy, haloalkoxy, oxo, acyloxy, carbonyl, carboxyl, carboxamido, cyano, halogen, hydroxyl, amino, aminoalkyl, amidoalkyl, amido, nitro, thiol, alkylthio, arylthio, sulfonamide, sulfinyl, sulfonyl, urea, or carbamate; each L1 is alkylene or heteroalkylene; [0261] m is 0, 1 , 2 or 3; and n is 0, 1 , 2, 3, 4 or 5, or an enantiomer, a diastereomer, or a mixture thereof, or a pharmaceutically acceptable salt or solvate thereof.
[0262] Representative compounds according to Formula VII are suitably selected from: (trans)-2-(3'-(trifluoromethyl)biphenyl-4-yl)cyclopropanamine; (trans)- 2-(terphenyl-4-yl)cyclopropanamine; 4'-((trans)-2-aminocyclopropyl)biphenyl-4-ol; 4'- ((trans)-2-aminocyclopropyl)biphenyl-3-ol; (trans)-2-(6-(3- (trifluoromethyl)phenyl)pyridin-3-yl)cyclopropanamine; (Trans)-2-(6-(3,5- dichlorophenyl)pyridin-3-yl)cyclopropanamine; (trans)-2-(6-(4-chlorophenyl)pyridin-3- yl)cyclopropanamine; (trans)-2-(6-(3-chlorophenyl)pyridin-3-yl)cyclopropanamine; (trans)-2-(6-(4-(trifluoromethyl)phenyl)pyridin-3-yl)cyclopropanamine; (trans)-2-(6-(4- methoxyphenyl)pyridin-3-yl)cyclopropanamine; (trans)-2-(6-(3- methoxyphenyl)pyridin-3-yl)cyclopropanamine; 4-(5-((trans)-2- aminocyclopropyl)pyridin-2-yl)benzonitrile; 3-(5-((trans)-2-aminocyclopropyl)pyridin- 2-yl)benzonitrile; (Trans)-2-(6-p-tolylpyridin-3-yl)cyclopropanamine; (trans)-2-(6-m- tolylpyridin-3-yl)cyclopropanamine; 4-(5-((trans)-2-aminocyclopropyl)pyridin-2- yl)phenol; 3-(5-((trans)-2-aminocyclopropyl)pyridin-2-yl)phenol; 4-(5-((trans)-2- aminocyclopropyl)pyridin-2-yl)benzamide; 3-(5-((trans)-2-aminocyclopropyl)pyridin-2- yl)benzamide; 2-(5-((trans)-2-aminocyclopropyl)pyridin-2-yl)phenol; 3-(5-((trans)-2- aminocyclopropyl)pyridin-2-yl)phenol; (trans)-2-(6-(3-methoxy-4- methylphenyl)pyridin-3-yl)cyclopropanamine; 5-(5-((trans)-2- aminocyclopropyl)pyridin-2-yl)-2-fluorophenol; 3-(5-((trans)-2- aminocyclopropyl)pyridin-2-yl)-5-fluorophenol; 3-(5-((trans)-2- aminocyclopropyl)pyridin-2-yl)-4-fluorophenol; 3-(5-((trans)-2- aminocyclopropyl)pyridin-2-yl)-2-fluorophenol; 3-(5-((trans)-2- aminocyclopropyl)pyridin-2-yl)-2,4-difluorophenol; 3-(5-((trans)-2- aminocyclopropyl)pyndin-2-yl)-2,4,6-trifluorophenol; 3-(5-((trans)-2- aminocyclopropyl)pyridin-2-yl)-5-chlorophenol; (trans)-2-(6-(2-fluoro-3- (tnfluoromethyl)phenyl)pyndin-3-yl)cyclopropanamine; (trans)-2-(6-(5-chlorothiophen- 2- yl)pyridin-3-yl)cyclopropanamine; (trans)-2-(6-(5-methylthiophen-2-yl)pyridin-3- yl)cyclopropanamine; (trans)-2-(6-(1 H-indol-6-yl)pyridin-3-yl)cyclopropanamine; (trans)-2-(6-(benzo[b]thiophen-5-yl)pyridin-3-yl)cyclopropanamine; 3-(5-((trans)-2- aminocyclopropyl)-3-methylpyndin-2-yl)phenol; (trans)-2-(6-(3-chlorophenyl)-5- methylpyndin-3-yl)cyclopropanamine; (trans)-2-(5-methyl-6-(3- (tnfluoromethyl)phenyl)pyndin-3-yl)cyclopropanamine; (trans)-2-(6-(4-fluoro-3- methoxyphenyl)pyridin-3-yl)cyclopropanamine, (trans)-2-(6-(3-fluoro-5- methoxyphenyl)pyndin-3-yl)cyclopropanamine; (trans)-2-(6-(2-fluoro-5- methoxyphenyl)pyndin-3-yl)cyclopropanamine, (trans)-2-(6-(2-fluoro-3- methoxyphenyl)pyndin-3-yl)cyclopropanamine; (trans)-2-(6-(3-chloro-5- methoxyphenyl)pyndin-3-yl)cyclopropanamine; (trans)-2-(6-(2-chloro-5- methoxyphenyl)pyridin-3-yl)cyclopropanamine; (trans)-2-(6-(3-methoxy-5- (tnfluoromethyl)phenyl)pyndin-3-yl)cyclopropanamine; 3-(5-((trans)-2- aminocyclopropyl)pyndin-2-yl)-5-methoxybenzonitri- le; 5-(5-((trans)-2- aminocyclopropyl)pyndin-2-yl)-2-methylphenol; 3-(5-((trans)-2- aminocyclopropyl)pyridin-2-yl)-4-chlorophenol; 3-(5-((trans)-2- aminocyclopropyl)pyndin-2-yl)-5-(tnfluoromethyl)phenol; (trans) -2-(6-(2-fluoro-5- (tnfluoromethyl)phenyl)pyndin-3-yl)cyclopropanamine; (trans)-2-(6-(2-chloro-5- (tnfluoromethyl)phenyl)pyndin-3-yl)cyclopropanamine; (trans)-2-(6-(3,5- bis(tnfluoromethyl)phenyl)pyridin-3-yl)cyclopropanamine; N-(3-(5-((trans)-2- aminocyclopropyl)pyridin-2-yl)phenyl)acetamide; N-(3-(5-((trans)-2- aminocyclopropyl)pyridin-2-yl)phenyl)methanesulfonamide; (trans)-2-(6- (benzo[b]thiophen-2-yl)pyridin-3-yl)cyclopropanamine; (trans)-2-(6- (benzo[b]thiophen- 3- yl)pyridin-3-yl)cyclopropanamine; 5-(5-((trans)-2- aminocyclopropyl)pyridin-2-yl)thiophene-2-carbonitrile; (trans)-2-(6-(4- methylthiophen-3-yl)pyndin-3-yl)cyclopropanamine; (trans)-2-(2-chloro-6-(3- (tnfluoromethyl)phenyl)pyridin-3-yl)cyclopropanamine; (trans)-2-(2-(4-chlorophenyl)- 6-(3- (trifluoromethyl)phenyl)pyndine-3-yl)cyclopropanamine; 4-(3-((trans)-2- aminocyclopropyl)-6-(3-(trifluoromethyl)phenyl)pyridin-2-yl)phenol; 4-(3-((trans)-2- aminocyclopropyl)-6-(3-(trifluoromethyl)phenyl)- pyridin-2-yl)benzamide; (trans)-2-(2- methyl-6-(3-(trifluoromethyl)phenyl)pyndin-3-yl)cyclopropanamine; 3-(5-((trans)-2- aminocyclopropyl)pyndin-2-yl)-5-hydroxybenzonitril- e; (trans)-2-(6-(3,4-difluoro-5- methoxyphenyl)pyndin-3-yl)cyclopropanamine; 5-(5-((trans)-2- aminocyclopropyl)pyridin-2-yl)-2,3-difluorophenol; (trans)-2-(6-(3-chloro-4-fluoro-5- methoxyphenyl)pyndin-3-yl)cyclopropanamine; 5-(5-((trans)-2- aminocyclopropyl)pyndin-2-yl)-3-chloro-2-fluorophenol; (trans)-2-(6-(1 H-indazol-6- yl)pyridin-3-yl)cyclopropanamine; (trans)-2-(6-(9H-carbazol-2-yl)pyridin-3- yl)cyclopropanamine; 6-(5-((trans)-2-aminocyclopropyl)pyridin-2-yl)indolin-2-one; 6- (5-((trans)-2-aminocyclopropyl)pyndin-2-yl)benzofuran-2(3H)-one; 4-(5-((trans)-2- aminocyclopropyl)pyndin-2-yl)pyridin-2(1 H)-one; N-(3-(5-((trans)-2- aminocyclopropyl)pyndin-2-yl)phenyl)benzenesulfonamide; N-(3-(5-((trans)-2- aminocyclopropyl)pyndin-2-yl)phenyl)propane-2-sulfonamide; 4'-((trans)-2- aminocyclopropyl)-4-fluorobiphenyl-3-ol; 4'-((trans)-2-aminocyclopropyl)-5- chlorobiphenyl-3-ol; 4'-((trans)-2-aminocyclopropyl)-5-chloro-4-fluorobiphenyl-3-ol; N-(4'-((trans)-2-aminocyclopropyl)biphenyl-3-yl)benzenesulfonamide; N-(4'-((trans)- 2-aminocyclopropyl)biphenyl-3-yl)propane-2-sulfonamide; N-(4'-((trans)-2- aminocyclopropyl)biphenyl-3-yl)methanesulfonamide; N-(2-(5-((trans)-2- aminocyclopropyl)pyndin-2-yl)phenyl)methanesulfonamide; 3-(5-((trans)-2- aminocyclopropyl)pyndin-2-yl)-4-methoxybenzonitrile; N-(4'-((trans)-2- aminocyclopropyl)biphenyl-2-yl)methanesulfonamide; 4'-((trans)-2- aminocyclopropyl)-6-methoxybiphenyl-3-carbonitnle; N-(4'-((trans)-2- aminocyclopropyl)-6-methoxybiphenyl-3-yl)methanesulfonamide; 4'-((trans)-2- aminocyclopropyl)-6-hydroxybiphenyl-3-carbonitrile; N-(4'-((trans)-2- aminocyclopropyl)-6-hydroxybiphenyl-3-yl)methanesulfonamide; 3-(5-((trans)-2- aminocyclopropyl)pyridin-2-yl)-4-hydroxybenzonitrile; N-(3-(5-((trans)-2- aminocyclopropyl)pyridin-2-yl)-4-hydroxyphenyl)methane-sulfonamide; N-(3-(5- ((trans)-2-aminocyclopropyl)pyridin-2-yl)-5- (trifluoromethyl)phenyl)ethanesulfonamide; N-(3-(5-((trans)-2- aminocyclopropyl)pyndin-2-yl)-5-(tnfluoromethyl)phenyl)methanesulfonamide; 3-(6- ((trans)-2-aminocyclopropyl)pyridin-3-yl)phenol; (Trans)-2-(5-(3- methoxyphenyl)pyridin-2-yl)cyclopropanamine; 4-(6-((trans)-2- aminocyclopropyl)pyridin-3-yl)phenol; 2-(6-((trans)-2-aminocyclopropyl)pyridin-3- yl)phenol; 2-(5-((trans)-2-aminocyclopropyl)thiophen-2-yl)phenol; 3-(5-((trans)-2- aminocyclopropyl)thiophen-2-yl)phenol; 4-(5-((trans)-2-aminocyclopropyl)thiophen-2- yl)phenol; 2-(5-((trans)-2-aminocyclopropyl)thiazol-2-yl)phenol; 3-(5-((trans)-2- aminocyclopropyl)thiazol-2-yl)phenol; 4-(5-((trans)-2-aminocyclopropyl)thiazol-2- yl)phenol; 2-(2-((trans)-2-aminocyclopropyl)thiazol-5-yl)phenol; 3-(2-((trans)-2- aminocyclopropyl)thiazol-5-yl)phenol; 2-(2-((trans)-2-aminocyclopropyl)thiazol-5- yl)phenol; 3-(2-((trans)-2-aminocyclopropyl)thiazol-5-yl)phenol; 3-(5-((trans)-2- aminocyclopropyl)pyrimidin-2-yl)phenol; 4-(5-((trans)-2-aminocyclopropyl)pyrimidin- 2-yl)phenol; N-(3-(5-((trans)-2-aminocyclopropyl)pyridin-2-yl)-4- methoxyphenyl)methanesulfonamide; N-(4'-((trans)-2-aminocyclopropyl)-5-chloro-[1 , -biphenyl]-3-yl)methanesulfonamide; N-(3-(5-((trans)-2-aminocyclopropyl)pyridin-2- yl)-5-chlorophenyl)methanesulfonamide; N-(4'-((trans)-2-aminocyclopropyl)-4-fluoro- [1 ,1 '-biphenyl]-3-yl)methanesulfonamide; N-(5-(5-((trans)-2- aminocyclopropyl)pyridin-2-yl)-2-fluorophenyl)methanesulfonamide; N-(3-(5-((trans)- 2-aminocyclopropyl)pyridin-2-yl)phenyl)ethanesulfonamide; N-(3-(5-((trans)-2- aminocyclopropyl)pyridin-2-yl)phenyl)-4-cyanobenzenesulfonamide; N-(3-(5-((trans)- 2-aminocyclopropyl)pyridin-2-yl)phenyl)-3-cyanobenzenesulfonamide; N-(3-(5- ((trans)-2-aminocyclopropyl)pyridin-2-yl)phenyl)-2-cyanobenzenesulfonamide; N-(3- (5-((trans)-2-aminocyclopropyl)pyridin-2-yl)-5-(trifluoromethyl)phenyl)-4- cyanobenzenesulfonamide; N-(4'-((trans)-2-aminocyclopropyl)-[1 ,-biphenyl]-3-yl)- 1 ,1 ,1 -trifluorom-ethanesulfonamide; 4'-((trans)-2-aminocyclopropyl)-6-hydroxy-[1 ,1 biphenyl]-3-carbonitrile; 4'-((trans)-2-aminocyclopropyl)-[1 ,1 '-biphenyl]-2-ol, 4'- ((trans)-2-aminocyclopropyl)-3'-methoxy-[1 ,1 '-biphenyl]-3-ol; N-(3-(5-((trans)-2- aminocyclopropyl)thiazol-2-yl)phenyl)-2-cyanobenzenesulfonamide; or a pharmaceutically acceptable salt or solvate thereof.
[0263] In other embodiments, LSD1 inhibitor compounds are selected from phenylcyclopropylamine derivatives, as described for example by Ogasawara etal. (2013, Angew. Chem. Int. Ed. 52: 8620-8624, which is hereby incorporated by reference herein in its entirety). Representative compounds of this type are represented by Formula (XII):
Figure imgf000077_0001
[0264] wherein An is a 5 to 7 membered aryl or heteroaryl ring;
[0265] Ar2 and Ar3 are each independently selected from a 5 to 7 membered aryl or heteroaryl ring, optionally substituted with 1 to 3 substituents;
[0266] Ri and R2 are independently selected from hydrogen and hydroxyl or taken together Ri and R2 form =0, =S, or =NR3;
[0267] R3 is selected from hydrogen, -Ci ealkyl or -OH; [0268] m is an integer from 1 to 5; and
[0269] n is an integer from 1 to 3;
[0270] or a pharmaceutically acceptable salt thereof.
[0271] In particular embodiments of Formula (VII), one or more of the following applies:
[0272] An is a six membered aryl or heteroaryl ring, especially phenyl, pyridine, pyrimidine, pyrazine 1 ,3,5-triazine, 1 ,2,4-trazine and 1 ,2,3-triazine, more especially phenyl;
[0273] Ar2 is a six membered aryl or heteroaryl ring, especially phenyl, pyridine, pyrimidine, pyrazine 1 ,3,5-triazine, 1 ,2,4-trazine and 1 ,2,3-triazine, especially phenyl; especially where the six membered aryl or heteroaryl ring is optionally substituted with one optional substituent, especially in the 3 or 4 position;
[0274] Ar3 is a six membered aryl or heteroaryl ring, especially phenyl, pyridine, pyrimidine, pyrazine 1 ,3,5-triazine, 1 ,2,4-trazine and 1 ,2,3-triazine, especially phenyl; especially where the six membered aryl or heteroaryl ring is optionally substituted with one optional substituent, especially in the 3 or 4 position.
[0275] Particular optional substituents for An and Ar2 include -Ci ealkyl, -C2- 6alkenyl, -CH2F, -CFIF2, -CF3, halo, aryl, heteroaryl, -C(0)NFICi-6alkyl, -C(0)NFICI-6 alkylNH2, -C(0)-heterocyclyl, especially methyl, ethyl, propyl, butyl, t-butyl, -CFI2F, - CHF2, -CH3, Cl, F, phenyl, -C(0)NH(CH2)I- NH and -C(0)-heterocyclyl ;
[0276] Ri and R2 taken together form =0, =S or = NR3, especially =0 or =S, more especially =0;
[0277] R3 is FI, -Ci salkyl or -OFI, especially FI, -CFI3 or -OFI.
[0278] m is 2 to 5, especially 3 to 5, more especially 4,
[0279] n is 1 or 2, especially 1.
[0280] In some embodiments the compounds of Formula (XII) are compounds of Formula (Xlla):
Figure imgf000079_0001
(XI la)
[0281] wherein Ar2 and Ar3 are as defined for Formula (XII).
[0282] Non-limiting compounds represented by Formula (XII) include the following:
Figure imgf000079_0002
[0283] The synthesis and inhibitory activity of the compounds of Formula (VII) are described by Ogasawara et al. (2013, supra).
[0284] Other LSD1 inhibitors include, but are not limited to those, e.g., disclosed in Ueda et al. (2009. J. Am. Chem. Soc. 131 (48): 17536-17537) and Mimasu et al., (2010, Biochemistry, 49(30): 6494-503). [0285] Other phenylcyclopropylamine derivatives and analogues are found, e.g., in Kaiser etal. (1962, J. Med. Chem. 5: 1243-1265); Zirkle etal. (1962. J. Med. Chem. 1265-1284; U.S. Pat. Nos. 3,365,458; 3,471 ,522; 3,532,749) and Bolesov et al. (1974. Zhurnal Organicheskoi Khimii 10: 8 1661-1669) and Russian Patent No. 230169 (19681030).
[0286] In some other particularly embodiments, the inhibitory agent is GSK- LSD1 , which has the following molecular structure:
Figure imgf000080_0001
[0287] In some other particularly preferred embodiments, the agent is SP- 2509, which has the following molecular structure:
Figure imgf000080_0002
[0288] In some other particularly preferred embodiments, the agent is SP- 2577, which has the following molecular structure:
Figure imgf000080_0003
[0289] The invention not only encompasses known LSD ( e.g ., LSD1 or LSD2) inhibitors but LSD inhibitors identified by any suitable screening assay. Accordingly, the present invention extends to methods of screening for modulatory agents that are useful for inhibiting a LSD1 and, in turn, for preventing demethylation of an ACE2 polypeptide. In some embodiments, the screening methods comprise (1) contacting a preparation with a test agent, wherein the preparation comprises (i) a polypeptide comprising an amino acid sequence corresponding to at least a biologically active fragment of LSD1 or to a variant or derivative thereof; or (ii) a polynucleotide comprising a nucleotide sequence from which a transcript of a LSD1 gene or portion thereof is producible, or (iii) a polynucleotide comprising at least a portion of a genetic sequence (e.g. , a transcriptional element) that regulates the expression of a LSD1 gene, which is operably linked to a reporter gene; and (2) detecting a change in the level or functional activity of the polypeptide, the polynucleotide or an expression product of the reporter gene, relative to a reference level or functional activity in the absence of the test agent. A detected reduction in the level and/or functional activity of the polypeptide, transcript or transcript portion or an expression product of the reporter gene, relative to a normal or reference level and/or functional activity in the absence of the test agent, indicates that the agent is useful for preventing demethylation of an ACE2 polypeptide. Suitably, this is confirmed by analysing or determining whether the test agent prevents the demethylation of an ACE2 polypeptide, and would therefore be useful as an agent for preventing or treatment a coronavirus infection..
[0290] Modulators falling within the scope of the present invention include inhibitors of the level or functional activity of a LSD (e.g., LSD1 or LSD2), including antagonistic antigen-binding molecules, and inhibitor peptide fragments, antisense molecules, ribozymes, RNAi molecules and co-suppression molecules as well as polysaccharide and lipopolysaccharide inhibitors of a LSD (e.g., LSD1 or LSD2) .
[0291] Candidate agents encompass numerous chemical classes, though typically they are organic molecules, preferably small organic compounds having a molecular weight of more than 50 and less than about 2,500 Dalton. Candidate agents comprise functional groups necessary for structural interaction with proteins, particularly hydrogen bonding, and typically include at least an amine, carbonyl, hydroxyl or carboxyl group, desirably at least two of the functional chemical groups. The candidate agent often comprises cyclical carbon or heterocyclic structures or aromatic or polyaromatic structures substituted with one or more of the above functional groups. Candidate agents are also found among biomolecules including, but not limited to: peptides, saccharides, fatty acids, steroids, purines, pyrimidines, derivatives, structural analogues or combinations thereof.
[0292] Small (non-peptide) molecule modulators of a LSD {e.g., LSD1 or LSD2) are particularly advantageous. In this regard, small molecules are desirable because such molecules are more readily absorbed after oral administration, have fewer potential antigenic determinants, or are more likely to cross the cell membrane than larger, protein-based pharmaceuticals. Small organic molecules may also have the ability to gain entry into an appropriate cell and affect the expression of a gene ( e.g ., by interacting with the regulatory region or transcription factors involved in gene expression); or affect the activity of a gene by inhibiting or enhancing the binding of accessory molecules.
[0293] Alternatively, libraries of natural compounds in the form of bacterial, fungal, plant and animal extracts are available or readily produced. Additionally, natural or synthetically produced libraries and compounds are readily modified through conventional chemical, physical and biochemical means, and may be used to produce combinatorial libraries. Known pharmacological agents may be subjected to directed or random chemical modifications, such as acylation, alkylation, esterification, amidification, etc. to produce structural analogues.
[0294] Screening may also be directed to known pharmacologically active compounds and chemical analogues thereof.
[0295] Screening for modulatory agents according to the invention can be achieved by any suitable method. For example, the method may include contacting a cell expressing a polynucleotide corresponding to a gene that encodes a LSDIwith an agent suspected of having the modulatory activity and screening for the modulation of the level or functional activity of the LSD1 , or the modulation of the level of a transcript encoded by the polynucleotide, or the modulation of the activity or expression of a downstream cellular target of the polypeptide or of the transcript (hereafter referred to as target molecules). Detecting such modulation can be achieved utilizing techniques including, but not restricted to, ELISA, cell-based ELISA, inhibition ELISA, Western blots, immunoprecipitation, slot or dot blot assays, immunostaining, RIA, scintillation proximity assays, fluorescent immunoassays using antigen-binding molecule conjugates or antigen conjugates of fluorescent substances such as fluorescein or rhodamine, Ouchterlony double diffusion analysis, immunoassays employing an avidin-biotin or a streptavidin-biotin detection system, and nucleic acid detection assays including reverse transcriptase polymerase chain reaction (RT-PCR).
[0296] It will be understood that a polynucleotide from which LSD1 is regulated or expressed may be naturally occurring in the cell which is the subject of testing or it may have been introduced into the host cell for the purpose of testing. In addition, the naturally-occurring or introduced polynucleotide may be constitutively expressed - thereby providing a model useful in screening for agents which down- regulate expression of an encoded product of the sequence wherein the down regulation can be at the nucleic acid or expression product level. Further, to the extent that a polynucleotide is introduced into a cell, that polynucleotide may comprise the entire coding sequence that codes for LSD1 or it may comprise a portion of that coding sequence (e.g., the active site of LSD1) or a portion that regulates expression of the corresponding gene that encodes LSD1 (e.g., a LSD1 promoter). For example, the promoter that is naturally associated with the polynucleotide may be introduced into the cell that is the subject of testing. In this instance, where only the promoter is utilized, detecting modulation of the promoter activity can be achieved, for example, by operably linking the promoter to a suitable reporter polynucleotide including, but not restricted to, green fluorescent protein (GFP), luciferase, b-galactosidase and catecholamine acetyl transferase (CAT). Modulation of expression may be determined by measuring the activity associated with the reporter polynucleotide.
[0297] These methods provide a mechanism for performing high throughput screening of putative modulatory agents such as proteinaceous or non- proteinaceous agents comprising synthetic, combinatorial, chemical and natural libraries. These methods will also facilitate the detection of agents which bind either the polynucleotide encoding the target molecule or which modulate the expression of an upstream molecule, which subsequently modulates the expression of the polynucleotide encoding the target molecule. Accordingly, these methods provide a mechanism of detecting agents that either directly or indirectly modulate the expression or activity of a target molecule according to the invention.
[0298] In alternative embodiments, test agents are screened using commercially available assays, illustrative examples of which include EpiQuik Flistone Demethylase LSD1 Inhibitor Screening Assay Kit (Epigentek Group, Brooklyn, NY) or the LSD1 Inhibitor Screening Assay Kit (Cayman Chemical Company, Ann Arbor, Ml).
[0299] Compounds may be further tested in the animal models to identify those compounds having the most potent in vivo effects. These molecules may serve as “lead compounds” for the further development of pharmaceuticals by, for example, subjecting the compounds to sequential modifications, molecular modelling, and other routine procedures employed in rational drug design.
[0300] In some embodiments, the LSD1 inhibitor inhibits the binding between an LSD1 polypeptide and a CoRest polypeptide.
2.1 Indirect LSD Inhibitors
[0301] In some embodiments, the inhibitor specifically binds a polypeptide that is known to play an important role in activating LSD1. For example, it is well known that PKC0 activates the transcription of LDS1 , and accordingly, any inhibitor sufficient to reduce or prevent the phosphorylation of LSD by PKC0 is equally as applicable for use with the present invention.
[0302] Accordingly, in one aspect of the present invention, there is provided an isolated or purified proteinaceous molecule represented by any one of SEQ ID NOs:1 or 2:
[0303] RKEIDPPFRPKVK [SEQ ID NO: 1 ]; or
[0304] RRKRIDWPPRRKPK [SEQ ID NO: 2]
[0305] The proteinaceous molecule of SEQ ID NO: 1 is also referred to herein as "importinib4759" and the proteinaceous molecule of SEQ ID NO: 2 is also referred to herein as "importinib4759_01".
[0306] The present invention also contemplates proteinaceous molecules that are variants of SEQ ID NO: 1 and/or 2. Such “variant” proteinaceous molecules include proteins derived from the native protein by deletion (so-called truncation) or addition of one or more amino acids to the N-terminal and/or C-terminal end of the native protein; deletion or addition of one or more amino acids at one or more sites in the native protein; or substitution of one or more amino acids at one or more sites in the native protein.
[0307] Variant proteins encompassed by the present invention are biologically active, that is, they continue to possess the desired biological activity of the native protein. Such variants may result from, for example, genetic polymorphism or from human manipulation. [0308] The proteinaceous molecules of SEQ ID NO: 1 and/or 2 may be altered in various ways including amino acid substitutions, deletions, truncations, and insertions. Methods for such manipulations are generally known in the art. For example, amino acid sequence variants of SEQ ID NO: 1 and/or 2 can be prepared by mutagenesis of nucleic acids encoding the amino acid sequence of SEQ ID NO: 1 and/or 2. Methods for mutagenesis and nucleotide sequence alterations are well known in the art. See, for example, Kunkel (1985, Proc. Natl. Acad. Sci. USA. 82: 488-492), Kunkel et al., (1987, Methods in Enzymol, 154: 367-382), U.S. Pat. No. 4,873, 192, Watson, J. D. etal., ("Molecular Biology of the Gene", Fourth Edition, Benjamin/Cummings, Menlo Park, Calif, 1987) and the references cited therein. Guidance as to appropriate amino acid substitutions that do not affect biological activity of the protein of interest may be found in the model of Dayhoff etal., (1978) Atlas of Protein Sequence and Structure {Natl. Biomed. Res. Found., Washington, D.C.). Methods for screening gene products of combinatorial libraries made by point mutations or truncation, and for screening cDNA libraries for gene products having a selected property are known in the art. Such methods are adaptable for rapid screening of the gene libraries generated by combinatorial mutagenesis of the proteinaceous molecules of SEQ ID NO: 1 and/or 2. Recursive ensemble mutagenesis (REM), a technique which enhances the frequency of functional mutants in the libraries, can be used in combination with screening assays to identify active variants (Arkin and Yourvan (1992) Proc. Natl. Acad. Sci. USA 89: 7811-7815; Delgrave etal., (1993) Protein Engineering, 6: 327-331). Conservative substitutions, such as exchanging one amino acid with another having similar properties, may be desirable as discussed in more detail below.
[0309] Variant peptides or polypeptides of the invention may contain conservative amino acid substitutions at various locations along their sequence, as compared to a parent ( e.g ., naturally- occurring or reference) amino acid sequence, such as SEQ ID NO: 1 and/or 2. A “conservative amino acid substitution” is one in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art as discussed in detail below. [0310] The amino acid sequence of the proteinaceous molecules of the invention is defined in terms of amino acids of certain characteristics or sub-classes. Amino acid residues are generally sub-classified into major sub-classes as follows:
[0311] Acidic: The residue has a negative charge due to loss of a proton at physiological pH and the residue is attracted by aqueous solution so as to seek the surface positions in the conformation of a peptide in which it is contained when the peptide is in aqueous medium at physiological pH. Amino acids having an acidic side chain include glutamic acid and aspartic acid.
[0312] Basic: The residue has a positive charge due to association with protons at physiological pH or within one or two pH units thereof (e.g. histidine) and the residue is attracted by aqueous solution so as to seek the surface positions in the conformation of a peptide in which it is contained when the peptide is in aqueous medium at physiological pH. Amino acids having a basic side chain include arginine, lysine and histidine.
[0313] Charged: The residue is charged at physiological pH and, therefore, includes amino acids having acidic or basic side chains, such as glutamic acid, aspartic acid, arginine, lysine and histidine.
[0314] Hydrophobic: The residue is not charged at physiological pH and the residue is repelled by aqueous solution so as to seek the inner positions in the conformation of a peptide in which it is contained when the peptide is in aqueous medium at physiological pH. Amino acids having a hydrophobic side chain include tyrosine, valine, isoleucine, leucine, methionine, phenylalanine and tryptophan.
[0315] Neutral/polar: The residues are not charged at physiological pH but the residue is not
[0316] sufficiently repelled by aqueous solutions so that it would seek inner positions in the conformation of a peptide in which it is contained when the peptide is in aqueous medium at physiological pH. Amino acids having a neutral/polar side chain include asparagine, glutamine, cysteine, histidine, serine and threonine.
[0317] This description also characterizes certain amino acids as "small" since their side chains are not sufficiently large, even if polar groups are lacking, to confer hydrophobicity. With the exception of proline, “small” amino acids are those with four carbons or less when at least one polar group is on the side chain and three carbons or less when not. Amino acids having a small side chain include glycine, serine, alanine and threonine. The gene-encoded secondary amino acid proline is a special case due to its known effects on the secondary conformation of peptide chains. The structure of proline differs from all the other naturally-occurring amino acids in that its side chain is bonded to the nitrogen of the a-amino group, as well as the a-carbon. Several amino acid similarity matrices ( e.g ., PAM120 matrix and PAM250 matrix as disclosed for example by Dayhoff etal., (1978), A model of evolutionary change in proteins. Matrices for determining distance relationships In M. O. Dayhoff, (ed.), Atlas of protein sequence and structure, Vol. 5, pp. 345-358, National Biomedical Research Foundation, Washington DC; and by Gonnet etal., (1992), Science, 256(5062): 1443-1445), however, include proline in the same group as glycine, serine, alanine and threonine. Accordingly, for the purposes of the present invention, proline is classified as a “small” amino acid.
[0318] The degree of attraction or repulsion required for classification as polar or non-polar is arbitrary and, therefore, amino acids specifically contemplated by the invention have been classified as one or the other. Most amino acids not specifically named can be classified on the basis of known behaviour .
[0319] Amino acid residues can be further sub-classified as cyclic or non- cyclic, and aromatic or non-aromatic, self-explanatory classifications with respect to the side-chain substituent groups of the residues, and as small or large. The residue is considered small if it contains a total of four carbon atoms or less, inclusive of the carboxyl carbon, provided an additional polar substituent is present; three or less if not. Small amino acid residues are, of course, always non-aromatic.
[0320] Dependent on their structural properties, amino acid residues may fall in two or more classes. For the naturally-occurring protein amino acids, sub classification according to this scheme is presented in Table 1.
TABLE 1
AMINO ACID SUB-CLASSIFICATION
Figure imgf000087_0001
Figure imgf000088_0001
[0321] Conservative amino acid substitution also includes groupings based on side chains. For example, a group of amino acids having aliphatic side chains is glycine, alanine, valine, leucine, and isoleucine; a group of amino acids having aliphatic-hydroxyl side chains is serine and threonine; a group of amino acids having amide-containing side chains is asparagine and glutamine; a group of amino acids having aromatic side chains is phenylalanine, tyrosine, and tryptophan; a group of amino acids having basic side chains is lysine, arginine, and histidine; and a group of amino acids having sulphur-containing side chains is cysteine and methionine. For example, it is reasonable to expect that replacement of a leucine with an isoleucine or valine, an aspartic acid with a glutamic acid, a threonine with a serine, or a similar replacement of an amino acid with a structurally related amino acid will not have a major effect on the properties of the resulting variant peptide useful in the invention. Whether an amino acid change results in a proteinaceous molecule that inhibits PKC-Q can readily be determined by assaying its activity. Conservative substitutions are shown in Table 2 under the heading of exemplary and preferred substitutions. Amino acid substitutions falling within the scope of the invention, are, in general, accomplished by selecting substitutions that do not differ significantly in their effect on maintaining (a) the structure of the peptide backbone in the area of the substitution, (b) the charge or hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain. After the substitutions are introduced, the variants are screened for biological activity.
TABLE 2: EXEMPLARY AND PREFERRED AMINO ACID SUBSTITUTIONS
Figure imgf000089_0001
[0322] Alternatively, similar amino acids for making conservative substitutions can be grouped into three categories based on the identity of the side chains. The first group includes glutamic acid, aspartic acid, arginine, lysine, histidine, which all have charged side chains; the second group includes glycine, serine, threonine, cysteine, tyrosine, glutamine, asparagine; and the third group includes leucine, isoleucine, valine, alanine, proline, phenylalanine, tryptophan, methionine, as described in Zubay, Biochemistry, third edition, Wm.C. Brown Publishers (1993).
[0323] Thus, a predicted non-essential amino acid residue in a peptide of the invention is typically replaced with another amino acid residue from the same side chain family. Alternatively, mutations can be introduced randomly along all or part of the coding sequence of a peptide of the invention, such as by saturation mutagenesis, and the resultant mutants can be screened for an activity of the parent polypeptide, as described for example herein, to identify mutants which retain that activity. Following mutagenesis of the coding sequences, the encoded peptide can be expressed recombinantly and its activity determined. A “non-essential” amino acid residue is a residue that can be altered from the wild-type sequence of an embodiment peptide of the invention without abolishing or substantially altering one or more of its activities. Suitably, the alteration does not substantially alter one of these activities, for example, the activity is at least 20%, 40%, 60%, 70% or 80% of that of the wild-type. By contrast, an “essential” amino acid residue is a residue that, when altered from the wild-type sequence of an embodiment peptide of the invention, results in abolition of an activity of the parent molecule such that less than 20% of the wild-type activity is present.
[0324] Accordingly, the present invention also contemplates variants of the proteinaceous molecules of SEQ ID NO 1 and/or 2 of the invention, wherein the variants are distinguished from the parent sequence by the addition, deletion, or substitution of one or more amino acid residues. In general, variants will display at least about 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% sequence similarity to a parent or reference proteinaceous molecule sequence as, for example, set forth in SEQ ID NO: 1 or 2, as determined by sequence alignment programs described elsewhere herein using default parameters. Desirably, variants will have at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% sequence identity to a parent or reference peptide sequence as, for example, set forth in SEQ ID NO: 1 or 2, as determined by sequence alignment programs described herein using default parameters. Variants of importinib4759 and importinib4759_01 , which fall within the scope of a variant peptide of the invention, may differ from the parent molecule generally by at least 1 , but by less than 5, 4, 3, 2 or 1 amino acid residue(s). In some embodiments, a variant peptide of the invention differs from the corresponding sequence in SEQ ID NO: 1 or 2 by at least 1 , but by less than 5, 4, 3, 2 or 1 amino acid residue(s). In some embodiments, the amino acid sequence of the variant peptide of the invention comprises lie (or modified form thereof) at position 5, Asp (or modified form thereof) at position 6, Pro (or modified form thereof) at position 8 and/or Pro (or modified form thereof) at position 9, relative to the numbering of SEQ ID NO: 2. In particular embodiments, the variant peptide of the invention inhibits PKC- Q nuclear translocation.
4. Therapeutic and Prophylactic Uses
[0325] In accordance with the present invention, it is proposed that agents that inhibit LSD1 ( e.g ., a selective LSD1 inhibitor) function are useful as actives and/or pharmaceutical compositions for treating or preventing a virus infection {e.g., a coronavirus infection). In such embodiments, it is considered that treatment or prevention includes the prevention of incurring a symptom, holding in check such symptoms, or treating existing symptoms associated with the pathogenic infection, when administered to an individual in need thereof.
[0326] The S protein of many betacoronaviruses, including but not limited to SARS-CoV and SARS-CoV-2, have been demonstrated to employ an ACE2 polypeptide for entry into the host cell. Similarly, the virus host cell entry receptor polypeptide used by MERS-CoV is a DPP4 polypeptide, and for hCoV-229, a CD13polypeptide.
[0327] Any LSD1 inhibitor can be used in the compositions and methods of the present invention, provided that the inhibitor is pharmaceutically active. A “pharmaceutically active” LSD1 inhibitor is in a form that results in the treatment and/or prevention of a pathogenic infection, particularly a coronavirus infection, including the prevention of incurring a symptom, holding in check such symptoms or treating existing symptoms associated with the metastatic cancer, when administered to an individual in need thereof.
[0328] Modes of administration, amounts of LSD1 inhibitor administered, and LSD1 inhibitor formulations, for use in the methods of the present invention, are routine and within the skill of practitioners in the art. Whether a pathogenic infection, particularly a coronavirus infection, has been treated is determined by measuring one or more diagnostic parameters indicative of the course of the disease, compared to a suitable control. In the case of an animal experiment, a “suitable control” is an animal not treated with the LSD1 inhibitor, or treated with the pharmaceutical composition without the LSD1 inhibitor. In the case of a human subject, a “suitable control” may be the individual before treatment, or may be a human ( e.g ., an age- matched or similar control) treated with a placebo. In accordance with the present invention, the treatment of a pathogenic infection includes and encompasses without limitation: (1) preventing the uptake of a pathogenic infection {e.g., a coronavirus infection) into a cell of the host; (2) treating a pathogenic infection {e.g., a coronavirus infection) in a subject; (3) preventing a pathogenic infection {e.g., a coronavirus infection) in a subject that has a predisposition to the pathogenic infection but has not yet been diagnosed with the pathogenic infection and, accordingly, the treatment constitutes prophylactic treatment of the pathogenic infection; or (iii) causing regression of a pathogenic infection {e.g., a coronavirus infection).
[0329] The compositions and methods of the present invention are thus suitable for treating an individual who has been diagnosed with a coronavirus infection, who is suspected of having a coronavirus infection, who is known to be susceptible and who is considered likely to develop a coronavirus infection, or who is considered likely to develop a recurrence of a previously treated coronavirus infection. The coronavirus infection may be a SARS-CoV-2 infection. In some embodiments, the coronavirus infection is a SARS-CoV-1 or a SARS-CoV-2 infection.
[0330] In some embodiments, and dependent on the intended mode of administration, the LSD1 inhibitor-containing compositions will generally contain about 0.000001% to 90%, about 0.0001% to 50%, or about 0.01% to about 25%, by weight of LSD inhibitor, the remainder being suitable pharmaceutical carriers or diluents etc. The dosage of the LSD1 inhibitor can depend on a variety of factors, such as mode of administration, the species of the affected subject, age, sex, weight and general health condition, and can be easily determined by a person of skill in the art using standard protocols. The dosages will also take into consideration the binding affinity of the LSD1 inhibitor to its target molecule, its bioavailability and its in vivo and pharmacokinetic properties. In this regard, precise amounts of the agents for administration can also depend on the judgment of the practitioner. In determining the effective amount of the agents to be administered in the treatment or prevention of a pathogenic infection, the physician or veterinarian may evaluate the progression of the disease or condition over time. In any event, those of skill in the art may readily determine suitable dosages of the LSD1 inhibitor without undue experimentation. The dosage of the actives administered to a patient should be sufficient to effect a beneficial response in the patient over time such as impairment, abrogation or prevention in the uptake of the virus into a cell of the host, and/or in the treatment and/or prevention of a pathogenic infection ( e.g ., a coronavirus infection). The dosages may be administered at suitable intervals to ameliorating the symptoms of the hematologic malignancy. Such intervals can be ascertained using routine procedures known to persons of skill in the art and can vary depending on the type of active agent employed and its formulation. For example, the interval may be daily, every other day, weekly, fortnightly, monthly, bimonthly, quarterly, half-yearly or yearly.
[0331] Dosage amount and interval may be adjusted individually to provide plasma levels of the active agent, which are sufficient to maintain LSD-inhibitory effects. Usual patient dosages for systemic administration range from 1 -2000 mg/day, commonly from 1-250 mg/day, and typically from 10-150 mg/day. Stated in terms of patient body weight, usual dosages range from 0.02-25 mg/kg/day, commonly from 0.02-3 mg/kg/day, typically from 0.2-1.5 mg/kg/day. Stated in terms of patient body surface areas, usual dosages range from 0.5-1200 mg/m2/day, commonly from 0.5-150 mg/m2/day, typically from 5-100 mg/m2/day.
[0332] In accordance with the practice of the present invention, inhibition of LSD1 will result in reduced entry of the virus into cells of the host, which will in turn result in fewer virus infected cells. Accordingly, it would be expected that a more effective treatment of the virus infection with an auxiliary cancer therapy or agent would occur. Thus, the present invention further contemplates administering the LSD1 inhibitor concurrently with at least one antiviral agent. The LSD1 inhibitor may be used therapeutically after the antiviral agent or may be used before the antiviral agent is administered or together with the antiviral agent. Accordingly, the present invention contemplates combination therapies, which employ a LSD1 inhibitor and concurrent administration of an antiviral agent, non-limiting examples of which include: broad-spectrum antiviral agents and coronavirus-specific antivirus agents.
[0333] The LSD inhibitors described above or elsewhere herein are particularly effective antiviral agents for monotherapeutic or combined-therapeutic use in treating coronavirus infection. One of the benefits of such combination therapies is that lower doses of the other antiviral agents can be administered while still achieving a similar level of antiviral efficacy. Such lower dosages can be particularly advantageous for drugs known to have genotoxicity and mitochondrial toxicity (for example, some nucleoside analogues). Conversely, greater efficacy might be achieved using therapeutic doses of two drugs than could be achieved using only a single drug.
Other Therapies
[0334] Examples of other cancer therapies include phototherapy, cryotherapy, toxin therapy or pro-apoptosis therapy. One of skill in the art would know that this list is not exhaustive of the types of treatment modalities available for cancer and other hyperplastic lesions.
[0335] The antiviral drug is suitably selected from antimicrobials, which include without limitation compounds that kill or inhibit the growth of microorganisms (including viruses), and antivirals.
[0336] Illustrative antivirals include abacavir sulphate, acyclovir sodium, amantadine hydrochloride, amprenavir, chloroquine, cidofovir, delavirdine mesylate, didanosine, efavirenz, favipiravir, famciclovir, fomivirsen sodium, foscarnet sodium, ganciclovir, hydroxychloroquine, hydroquinone. indinavir sulphate, lamivudine, lamivudine/zidovudine, lopinavir, nelfinavir mesylate, nevirapine, oseltamivir phosphate, ribavirin, remdesivir, rimantadine hydrochloride, ritonavir, saquinavir, saquinavir mesylate, stavudine, valacyclovir hydrochloride, zalcitabine, zanamivir, and zidovudine.
[0337] In some alternative embodiments, the LSD1 inhibitor may be co administered with an antimicrobial agent including chloroquine, hydroxychloroquine and/or hydroquinone. [0338] In some of the same embodiments and some alternative embodiments, the antiviral agent comprises a recombinant IFN-b polypeptide (UniProt Accession No. P01574). In some embodiments of this type, the antiviral agent comprises at least a portion of an IFN-b polypeptide, or a variant of an IFN-b polypeptide.
[0339] As noted above, the present invention encompasses co administration of an LSD1 inhibitor in concert with an additional agent. It will be understood that, in embodiments comprising administration of the LSD1 inhibitor with other agents, the dosages of the actives in the combination may on their own comprise an effective amount and the additional agent(s) may further augment the therapeutic or prophylactic benefit to the patient. Alternatively, the LSD1 inhibitor and the additional agent(s) may together comprise an effective amount for preventing or treating the pathogenic infection. It will also be understood that effective amounts may be defined in the context of particular treatment regimens, including, e.g., timing and number of administrations, modes of administrations, formulations, etc. In some embodiments, the LSD1 inhibitor and optionally the antiviral agent are administered on a routine schedule. Alternatively, the antiviral agent may be administered as symptoms arise. A “routine schedule” as used herein, refers to a predetermined designated period of time. The routine schedule may encompass periods of time which are identical or which differ in length, as long as the schedule is predetermined. For instance, the routine schedule may involve administration of the LSD inhibitor on a daily basis, every two days, every three days, every four days, every five days, every six days, a weekly basis, a monthly basis or any set number of days or weeks there-between, every two months, three months, four months, five months, six months, seven months, eight months, nine months, ten months, eleven months, twelve months, etc. Alternatively, the predetermined routine schedule may involve concurrent administration of the LSD1 inhibitor and the antiviral agent on a daily basis for the first week, followed by a monthly basis for several months, and then every three months after that. Any particular combination would be covered by the routine schedule as long as it is determined ahead of time that the appropriate schedule involves administration on a certain day.
[0340] Additionally, the present invention provides pharmaceutical compositions for reducing or abrogating the uptake a viruses {e.g., a coronavirus) to a cell of the host, the pharmaceutical compositions comprising a LSD inhibitor and optionally an antiviral agent useful for treating malignancies. The formulations of the invention are administered in pharmaceutically acceptable solutions, which may routinely contain pharmaceutically acceptable concentrations of salt, buffering agents, preservatives, compatible carriers, adjuvants, and optionally other therapeutic ingredients. Depending on the specific conditions being treated, the formulations may be administered systemically or locally. Techniques for formulation and administration may be found in “Remington’s Pharmaceutical Sciences”, Mack Publishing Co., Easton, Pa., latest edition. Suitable routes may, for example, include oral, rectal, transmucosal, or intestinal administration; parenteral delivery, including intramuscular, subcutaneous, intramedullary injections, as well as intrathecal, direct intraventricular, intravenous, intraperitoneal, intranasal, or intraocular injections. For injection, the active agents or drugs of the invention may be formulated in aqueous solutions, suitably in physiologically compatible buffers such as Hanks’ solution, Ringer’s solution, or physiological saline buffer. For transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
[0341] The drugs can be formulated readily using pharmaceutically acceptable carriers well known in the art into dosages suitable for oral administration. Such carriers enable the compounds of the invention to be formulated in dosage forms such as tablets, pills, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated. These carriers may be selected from sugars, starches, cellulose and its derivatives, malt, gelatin, talc, calcium sulphate, vegetable oils, synthetic oils, polyols, alginic acid, phosphate buffered solutions, emulsifiers, isotonic saline, and pyrogen-free water.
[0342] Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances, which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents, which increase the solubility of the compounds to allow for the preparation of highly, concentrated solutions.
[0343] Pharmaceutical preparations for oral use can be obtained by combining the active compounds with solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, or polyvinylpyrrolidone (PVP). If desired, disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate. Such compositions may be prepared by any of the methods of pharmacy but all methods include the step of bringing into association one or more drugs as described above with the carrier, which constitutes one or more necessary ingredients. In general, the pharmaceutical compositions of the present invention may be manufactured in a manner that is itself known, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes.
[0344] Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
[0345] Pharmaceutical compositions which can be used orally include push- fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added. [0346] Dosage forms of the drugs of the invention may also include injecting or implanting controlled releasing devices designed specifically for this purpose or other forms of implants modified to act additionally in this fashion. Controlled release of an agent of the invention may be achieved by coating the same, for example, with hydrophobic polymers including acrylic resins, waxes, higher aliphatic alcohols, polylactic and polyglycolic acids and certain cellulose derivatives such as hydroxypropyl methyl cellulose. In addition, controlled release may be achieved by using other polymer matrices, liposomes or microspheres.
[0347] The drugs of the invention may be provided as salts with pharmaceutically compatible counterions. Pharmaceutically compatible salts may be formed with many acids, including but not limited to hydrochloric, sulphuric, acetic, lactic, tartaric, malic, succinic, etc. Salts tend to be more soluble in aqueous or other protonic solvents that are the corresponding free base forms.
[0348] For any compound used in the method of the invention, the therapeutically effective dose can be estimated initially from cell culture assays. For example, a dose can be formulated in animal models to achieve a circulating concentration range that includes the IC50 as determined in cell culture ( e.g ., the concentration of an active agent, which achieves a half-maximal inhibition in activity of a LSD1 polypeptide). Such information can be used to more accurately determine useful doses in humans.
[0349] Toxicity and therapeutic efficacy of such drugs can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50:ED50. Compounds that exhibit large therapeutic indices are preferred.
The data obtained from these cell culture assays and animal studies can be used in formulating a range of dosage for use in human. The dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilised. The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient’s condition (see, for example, Fingl etal., 1975, in “The Pharmacological Basis of Therapeutics”, Ch. 1 ).
[0350] Alternately, one may administer the compound in a local rather than systemic manner, for example, via injection of the compound directly into a tissue, which is preferably subcutaneous or omental tissue, often in a depot or sustained release formulation.
[0351] Furthermore, one may administer the drug in a targeted drug delivery system, for example, in a liposome coated with tissue-specific antibody. The liposomes will be targeted to and taken up selectively by the tissue.
[0352] In cases of local administration or selective uptake, the effective local concentration of the agent may not be related to plasma concentration.
[0353] In order that the invention may be readily understood and put into practical effect, particular preferred embodiments will now be described by way of the following non-limiting examples.
EXAMPLES
[0354] In-depth structural analysis and mining of pre-existing high- throughput clinical and pre-clinical data have shown that: (i) the epigenetic enzyme LSD1 post-translationally regulates ACE2 and TMPRSS2; (ii) LSD1 maintains the dysfunctional T cell phenotype, and (iii) monoamine oxidase inhibitors (MOA inhibitors, which target LSD1 activity) inhibit the ACE2/TMPRSS2 machinery and restore killer T cell function and a durable memory responses in vitro and in vivo. MAO inhibitors such as phenelzine, tranylcypromine, and isocarboxazid are currently used clinically to treat depression and cancer, and Phase 1 b clinical trials are currently in progress for metastatic breast cancer which show that LSD1 inhibitors, including phenelzine (NARDIL), are safe and re-invigorate T cell-mediated immunity in humans.
[0355] Based on these data, we hypothesise that LSD1 inhibitors can be re-purposed for SARS-CoV-2 therapy by targeting viral entry into host cells and enhancing T cell-mediated immunity. Proving the efficacy of pre-existing drugs would rapidly transform clinical practice and outcomes in the current pandemic. Our efficacy, safety, and toxicity data on LSD1 inhibitors and optimised biomarkers in the oncology setting are readily transferrable to SARS-CoV-2 patients and provide not only a drug but also the biomarkers to monitor the exhaustion phenotype and disease progression.
EXPERIMENTAL EXAMPLES EXAMPLE 1
DETERMINATION OF PTM SITES ON TMPRSS2 AND ACE2
[0356] A series of protein domains within both the ACE2 protein and the TMPRSS2 protein were identified as being critical for the entry of the SARS-CoV-2 into the cell. These protein domains are subject to epigenetic post-translational modification (lysine methylation, de-methylation, sumoylation and phosphorylation).
[0357] Accordingly, bioinformatic analysis was used to identify specific post- translational modifications (PTMs) that are unique for LSD1 or PKC0. Multiple studies have now demonstrated that epigenetic PTM is a critical and common mechanism for regulating the dynamic regulation of key proteins, including p53.
[0358] It was therefore hypothesised that these PTMs are critical for interaction with SARS-CoV-2, protease activity (for TMPRSS2) and viral entry to the cell. Part of the replicative process of viruses include trafficking proteins into the nucleus to employ them as transcriptional regulators for more efficient transcription. Accordingly, putative nuclear localisation signal (NLS) peptide sequences within these proteins were identified. These peptides are selective and specific for the targeted proteins as well as being selective for the specific domains within each protein.
[0359] Extensive bioinformatic sequence analysis was employed using software designed to analyse and identify post-translation motifs within the protein sequence (phosphorylation, acetylation, methylation, glycosylation and so on), bioinformatic software to identify nuclear localisation sequences (NLS) were also employed which scored the probability of canonical and non-canonical NLS within the protein sequence, all analysis included cut-off values to reduce false positives and increase stringency. The software employed included NLS-mapper to identify NLS motifs (Kosugi, et al., 2008; Kosugi, et al., 2009a; Kosugi et a., 2009b), PSSMe was used to identify potential sites of methylation/demethylation (Wen et al., 2016), Phosphorylation NetPhos 3.1 Server was used to identify potential phosphorylation motifs (Blom et al., 2004) and Predict- Protein was used for further protein domain analysis (Su et al., 2019; Ofran et al., 2007).
[0360] This information was overlayed with the known protein domains within each analysed protein.
[0361] The present inventors identified within the TMPRSS2 and ACE2 proteins a series of key serine residues that are phosphorylated by PKCq and key lysine residue methylation sites, these methylated lysine residues also represent sites of LSD1 -mediated de-methylation. Other proteins have been demonstrated to be dynamically regulated by lysine methylation and demethylation or phosphorylation include p53. LSD1 regulated p53 at a single lysine residue conferring exquisite regulatory control on p53 (Huang et. al., 2007). Therefore, all such PTM sites have the potential to significantly influence the regulation of these proteins.
EXAMPLE 2
EFFECT OF LSD1 OR PKC0 INHIBITION OF TMPRSS2 AND/OR ACE2
[0362] Given the inventor’s realisation that LSD1 is a key epigenetic regulator, the importance of LSD1 on the direct regulation of TMPRSS2 was investigated. Specifically, an examination was performed on the effect of LSD1p inhibition or LSDIp’s upstream regulator PKC0 on the expression dynamics of TMPRSS2.
[0363] The effect of ablation of LSD1 , by treatment with a LSD1 siRNA, revealed that the effects of inhibition are specific for LSD1 (and not other enzymes). LSD1 siRNA knockouts resulted in significant loss of TMPRSS2 mRNA in both MDA- MB-231 and MCF7 breast cancer cell lines (Figure 1 A).
[0364] Next, the inhibition of two different LSD1 inhibitors (GSK-LSD1 sourced from Cyman Chemical, Product No. 16439; or phenelzine) was analysed by Nanostring. It was determined that both inhibitors were able to significantly abrogate expression of TMPRSS2 message in four different human cell lines. This suggests that targeting the FAD domain as well as the nuclear LSD1 :CoREST complex are able to inhibit TMPRSS2. This data demonstrates the indispensable role of LSD1 in regulating expression of TMPRSS2 (Figure 1B). [0365] In light of the above results, it was examined if LSD1 p or PKC0 tethered to chromatin regions associated with ACE2 or TMPRSS2. Samples were either control or phenelzine (FAD and nuclear LSD1p:CoREST inhibitor) or C27 (PKCq catalytic inhibitor) and CHiP enrichment assay carried out by pulling down chromatin fragments with either LSD1p or PKC0 antibodies. We then monitored the enrichment of TMPRSS2 promoters within each of the pull-down fragments for each antibody the corresponding inhibitor, LSD1p and phenelzine; or PKC0 and C27 followed by inhibition with the corresponding inhibitor. This quantified the enrichment of PKC0 or LSD1p at the promoters of TMPRSS2 within each of the chromatin pull- downs. Analysis revealed that both LSD1p and PKC0 had significant enrichment in either the Huh7 or MDA-MB-231 cell lines and this enrichment was abrogated by treatment with either phenelzine for LSD1p, or C27 for PKC0 (Figure 1C). This provides further evidence that both LSD1 and PKC0 tether to the proximal promoter regions of TMPRSS2 to regulate its transcription. The inventors have therefore elucidated that LSD1 p acts as a transcriptional activator of TMPRSS2 expression by converting FI3k9me2 to FI3k9me1 and FI3k4me2 to FI3k4me1.
[0366] In order to further evidence the mechanism of the biological effects described in the previous examples, a knockdown of TMPRSS2 protein expression in two cell lines was performed. High resolution microscopy using our digital pathology was employed to analyse the TMPRSS2 protein expression level of these samples using the ASkMetagene digital pathology system with automated background controls. These data revealed that LSD1 inhibition, either by inhibiting LSD1 directly with phenelzine, or indirectly by inhibiting the PKC0 protein with the PKC0-specific catalytic inhibitor C27 were able to significantly abrogate expression of TMPRSS2 in both cell lines (Figure 1 D).
Materials & Methods
Nanostring Analysis
[0367] MDA-MB-231 or MCF7 cells were incubated with siRNA targeting LSD1 and cells were then harvested and lysed. Total RNA was hybridized using the human Nanostring custom Panel. The absolute RNA counts were quantified by the nCounter NanoString system (NanoString Technologies, Seattle, WA, USA) and mRNA expression was analysed using the nSolver software and R v.3.3.2 for the Advanced analysis (NanoString Technologies), following the manufacturer’s recommendations and plotted as fold change versus control.
[0368] Human cell lines MDA-MB-231 , MCF7, CT26 and Huh7 cells were incubated with control vehicle, GSK-LSD1 or phenelzine and cells were then harvested and lysed. Total RNA from each treatment group/sample was hybridized using the human Nanostring custom Panel. The absolute RNA counts were quantified by the nCounter NanoString system (NanoString Technologies, Seattle, WA, USA) and mRNA expression was analysed using the nSolver software and R v.3.3.2 for the Advanced analysis (NanoString Technologies), following the manufacturer’s recommendations) and plotted as fold change versus control.
ChIP Assays
[0369] ChIP assays were performed in accordance with the protocol supplied by Upstate Biotechnology. Fixation steps were performed as detailed, and fixed chromatin was sonicated with an Ultrasonic processor (Cole/Parmer) under optimized conditions that gave average DNA fragments of approximately 500 bp, as determined by 2% agarose gel electrophoresis. Prior to antibody addition, samples were precleared with salmon sperm DNA-protein A-agarose and the soluble chromatin fraction was incubated overnight at 4°C with 5 to 10 pg of LSD1p (Merck Millipore) or PKC-0 (Abeam) antibody or without antibody as a control. Immune complexes were bound to salmon sperm DNA-protein A-agarose and then washed and eluted as described. Protein-DNA cross-links were reversed by incubation at 65°C overnight, and the DNA in each sample was recovered by phenol-chloroform extraction and ethanol precipitation. DNA pellets were washed in 70% ethanol, resuspended in Tris (pH 8.0), and subsequently used for SYBR green real-time PCR amplification (Applied Biosystems). Standard curves were generated for each primer set to correct for differences in primer efficiency. ChIP enrichment ratios were calculated for TMPRSS2 as per standard methods.
Figure imgf000103_0001
[0370] MDA-MB-231 mesenchymal cells or MCF7 epithelial cells were treated with either control, phenelzine or GSK-LSD1. Cells were then fixed for IFA Microscopy Analysis. Cells were permeabilised by incubating with 1% Triton X-100 for 20 min and were probed with rabbit TRMPSS2 mouse and visualized with a donkey anti-rabbit AF 488. Cover slips were mounted on glass microscope slides with ProLong Glass Antifade reagent (Life Technologies). Protein targets were localised by confocal laser scanning microscopy. Single 0.5 pm sections were obtained using the Metagene-ASI Digital Pathology platform. The final image was obtained by averaging four sequential images of the same section. Digital images were analysed using ASI proprietary analysis software to determine the mean Fluorescent Intensity (mean FI). Graph represents the mean FI values for TMPRS2 using ImageJ to select the nucleus minus background (n > 50 individual cells).
SUMMARY
[0371] These data show the important effect on transcription and protein expression of two different LSD1 inhibitors (namely, GSK-LSD, which binds to the FAD domain of LSD1 , and phenelzine, which can bind to both the FAD domain of LSD1 and disrupts the LSD1 -CoREST complex). Furthermore, a PKC0 inhibitor which targets the catalytic activity (C27) also exhibited similar effects (as PKC0 is well established as a downstream regulator of LSD1 activity). In summary, LSD1 inhibition significantly inhibits both message and protein expression of TMPRSS2 in a variety of cell lines including in a siLSDI knockdown model. This clearly demonstrates the specificity of LSD1p in regulating TMPRSS2 expression. Flowever, there was a slight decrease on ACE2 transcription in metastatic lung tissue. Despite this it is still likely that LSD1 plays a role in post-translationally modifying ACE2.
[0372] These data therefore suggest that LSD1 inhibition is required to inhibit TMPRSS2, either via the FAD domain or by disrupting the LSD1 :CoREST nuclear complex. Furthermore, targeting the nuclear LSD1 PTM form (decorated with methylation) is likely to be important in this process. The nuclear, phosphorylated (T538p/S636p) PKC0 is critical for TMPRSS2 regulation. Notably, the present inventors have previously shown in metastatic breast cancer and tumour associated macrophages that LSD1 carries out its nuclear role via forming a complex with CoREST.
EXAMPLE 3
LSD1, TMPRSS2 and ACE2 Associate on Cell Surface
[0373] LSD1 is a key eraser enzyme, that demethylates key histone proteins and key proteins such as transcription factors whereby this demethylation/ methylation post-translational modification has resulted in induction, inhibition or stabilization of the expression of the targeted proteins such as p53. Based on these data, the role of LSD1 as a key regulator of the receptor ACE2 and TMPRSS2 responsible for shuttling SARS-CoV-2 into the cell was investigated.
[0374] ASI digital pathology analysis was used to examine non- permeabilized Caco-2 cells which monitor cell surface expression and permeabilized cells that monitor intracellular. Cells were stained positive for the proteins ACE2, TMPRSS2 and LSD1. Strikingly, LSD1 (which is traditionally described as a cytoplasmic or nuclear protein) also stained positive on the cell surface (Figure 2). LSD1 significantly co-localized with ACE2 as demonstrated by the PCC(r) co efficient which adjudicates the degree of co-localisation between two protein targets. This analysis shows strong co-localisation between ACE2 and LSD1 on the cell surface. This was further validated by FACS analysis of Caco-2 cells. MRC5 cells, resistant to SARS-CoV-2 infection do not express ACE2 or TMPRSS2. Flowever, these cells express LSD1 on the cell surface, albeit at 4-fold less compared to SARS-CoV-2 susceptible cell line Caco-2.
[0375] It was then investigated the effect of SARS-CoV-2 infection on LSD1 and ACE2/TMPRSS2 co-expression. High resolution quantitative imaging and FACS analysis was used to examine Caco-2, or Caco-2/MRC5 cells infected with SARS- CoV-2. Cells were stained with ACE2 and the epigenetic enzyme LSD1 ; or LSD1 and antibodies for the nucleocapsid or spike protein of SARS-CoV-2 (Figure 3A-C). LSD1 significantly co-localized higher with ACE2 in cells infected with SARS-CoV-2, as compared to ACE2/LSD1 expression in uninfected CaCo2 and MRC5 cells (which are not susceptible to SARS-CoV-2 infection) as demonstrated by the PCC(r) co efficient which adjudicates the degree of co-localisation between two protein targets, as well as significantly upregulated expression of both LSD1 and ACE2 was demonstrated in infected cells. MRC5 had no expression of ACE2 and no staining of virus proteins (Figure 3D). Interestingly, LSD1 co-localized with both SARS-CoV-2 spike protein and the nucleocapsid proteins. Furthermore, LSD1 nuclear activity was increased as measured by reduction in expression of FI3k9me2 and FI3k4me2 (Figure 3F, G). Strikingly, when the two key methyl transferases (G9A and SETDB1) were compared, there was either little expression on the cell surface or no increase following SARS-CoV-2 infection (Figure 3H-J). [0376] These results clearly demonstrate that LSD1 and ACE2 have increased association on the cell surface.
[0377] Based on this, it was hypothesized that LSD1 complexes with and de-methylates ACE2 to stabilize expression.
[0378] Bioinformatic analysis clearly shows that ACE2 exhibits three high probability lysine residues for post-translation modification by LSD1 and that these lysine residues are part of the C-terminal domain and a novel, putative nuclear localization sequence (NLS). This C-terminal domain of ACE2 is a highly flexible, disordered domain suitable for protein-protein interactions.
[0379] Based upon these data, the inventors then demonstrated that inhibition of LSD1 with phenelzine (a dual targeting demethylase and nuclear LSD1 inhibitor) abrogated expression and co-localization of LSD1 with ACE2 and TMPRSS2 (see, Figure 4). Furthermore, there was minimal effect on ACE2 transcription, however TMPRSS2 transcription was inhibited. These results show that LSD1 inhibition interferes via inhibiting the demethylation destabilizing ACE2 protein expression on the cell surface. This is in contrast to TMPRSS2, which is regulated via the traditional role of LSD1 as an epigenetic regulator.
Materials and Methods
Microscopy Methods
[0380] To examine the signature of LSD1 , ACE2, TMPRSS2 and SARS- CoV-2 in infected cells or uninfected cells (untreated or treated with MAOis or EPI- 111 (myristyl-RRTSRRKRAKV-OFI) Caco-2 or MRC5 cells were permeabilised by incubating with 0.5% Triton X-100 for 15 min, blocked with 1% BSA in PBS and were probed with either LSD1 (Rabbit host), ACE2 (conjugated to AF594), TMPRSS2 (mouse host) and in the case of infected cells SARS-CoV-2 (Mouse Flost) and visualized with a donkey anti-mouse AF 488 or donkey anti-rabbit 647 or the antibodies were primary conjugated to an appropriate AF fluorochrome (AF 594). Cover slips were mounted on glass microscope slides with ProLong Glass Antifade reagent (Life Technologies). Protein targets were localised by digital pathology laser scanning microscopy. Single 0.5 pm sections were obtained using a ASI Digital pathology microscope using 100x oil immersion lens running ASI software. The final image was obtained by averaging four sequential images of the same section. Digital images were analyzed using automated ASI software (Applied Spectral Imaging, Carlsbad, CA) to determine the distribution and intensities automatically with automatic thresholding and background correction of the average nuclear fluorescence intensity (NFI), allowing for the specific targeting of expression of proteins of interest. Ddigital images were also analysed using ImageJ software (ImageJ, NIH, Bethesda, MD, USA) to determine the total cell fluorescence or cell surface only fluorescence for non-permeabilised cells. Digital images were analysed using ImageJ software (ImageJ, NIH, Bethesda, MD, USA) to determine the either the Total Nuclear Fluorescent Intensity (TNFI), the Total Cytoplasmic Fluorescent Intensity (TCFI). ImageJ software with automatic thresholding and manual selection of regions of interest (ROIs) specific for cell nuclei was used to calculate the Pearson’s co-efficient correlation (PCC) for each pair of antibodies. PCC values range from: -1 = inverse of co-localisation, 0 = no co-localisation, +1 = perfect co localisation. The Mann-Whitney nonparametric test (GraphPad Prism, GraphPad Software, San Diego, CA) was used to determine significant differences between datasets.
Microscale thermophoresis methods
[0381] Binding affinity measurements were performed on a Monolith NT.115 (NanoTemper Technologies). The fluorescein-Ahx tagged Ace2 peptide sequence RDRKKKNKARSGEN was manufactured by Genescript. Purified Each reaction consisted of 10 pL of the labelled peptide at 444 nM, mixed with unlabelled LSD1 at the indicated concentrations. All experiments were measured at 25°C with laser off/on/off times of 5/30/5 s. Experiments were conducted at 20% light-emitting diode power and 20-40% MST infra-red laser power. Data from three independently performed experiments were fitted to the single binding model via the NT. Analysis software version 1.5.41 (NanoTemper Technologies) using the signal from Thermophoresis + T-Jump.
EXAMPLE 4
LSD1 INHIBITION EFFECT ON ACE2 AND SARS-COV-2 EXPRESSION
[0382] Given that LSD1 regulates ACE2, the inventors next determined whether SARS-CoV-2 expression will be inhibited by LSD1 inhibition via blocking viral entry by down-regulation of ACE2. [0383] LSD1 expression at the cell surface is inhibited more significantly by phenelzine but not GSK-LSD1 or L1 (EPI-111) (Figure 5). This is explained by GSK- LSD1 being only a FAD domain demethylase inhibitor whereas phenelzine is known to be a FAD domain inhibitor that also causes structural alterations in LSD1. L1 (EPI- 111) is a nuclear translocation inhibitor, with its effect attributed to blocking the shuttling of LSD1 into the nucleus. Consistent with this, phenelzine is superior at inhibiting expression of ACE2, LSD1 and consequently expression of SARS-CoV-2 is also inhibited. Duolink analysis which detects interacting proteins only demonstrated that LSD1 and ACE2 strongly interact and this interaction is significantly inhibited by phenelzine and to a lesser extent, GSK-LSD1. RT-PCR analysis to measure the impact on SARS-CoV-2 RNA transcription showed that the nuclear targeting inhibitor L1 (EPI-111) was the most superior at inhibiting transcription, with both GSK-LSD1 and phenelzine having an effect (Figure 5I). Phenelzine also induced higher levels of SARS-CoV-2 RNA in the supernatant as a consequence of disrupting virus nucleocapsid formation which encapsulates the virus RNA. Furthermore, while LSD1, ACE2, and TMPRSS2 transcript remained unaltered in SARS-CoV-2-infected Caco-2 cells following phenelzine or GSK-LSD1 treatment, a limited type I interferon response was induced in Caco-2 cells following SARS-CoV-2 infection (Figure 5J). Compared to uninfected cells, the transcriptional response to infection including IFNI ¾ RIG-1, MDA-5, ISG15, and OASL was increased at 48 hpi, with !FN( 3 and OASL expression further increased following phenelzine treatment. mRNA levels of RIG1, MDA5, and ISG15 remained unaltered by phenelzine or GSK-LSD1 treatment (Figure 5J).
[0384] Although phenelzine treatment showed a 1.72-fold increase of cellular viral RNA replication compared to control, much less viral replication increase was detected in cell supernatant in phenelzine treatment between 24 hpi and 48 hpi (data not shown). This indicated that the virus can replicate within phenelzine-treated cells but much less virus were released into cell culture supernatant compared to control cells. This potentially suggests that replicated viral RNA was accumulated in phenelzine-treated cells due to a block in viral protein translation and viral assembly processes, such that less intact viral pathogens were released to infect other cells.
Materials & Methods RNA extraction and Quantitative Real-Time PCR
[0385] Total RNA was extracted using the RNeasy Micro kit (Qiagen,
Hilden, Germany) and the Direct-zol™ RNA Miniprep kit (Zymo Research, Irvine,
CA) for uninfected and SARS-Cov-2 studies, respectively. Extracted RNA was quantified using the Nanodrop spectrophotometer (Thermo Fisher Scientific, Waltham, MA) and reverse transcribed to cDNA using the Superscript VILO cDNA synthesis kit (Invitrogen, Waltham, MA), according to the manufacturer’s protocols. TaqMan quantitative real-time PCR was performed using the Applied Biosystems ViiA™7 Real-Time PCR System (Applied Biosystems, Waltham, MA) with the following human TaqMan probes: ACTB (Hs01060665_g1), GAPDH (Hs02786624_g1), HPRT1 (Hs02800695_m1), ACE2 (Hs01085333_m1), TMPRSS2 (Hs01122322_m1), IFNa (Hs03044218_g1 ), /F/V3 (Hs01077958_s1 ), IFNA (Hs00820125_g1), DDX58 (Hs01061436_m1), IFIH1 (Hs00223420_m1), ISG15 (Hs01921425_s1), OASL (Hs00984387_m1 ), IL-6 (Hs00174131_m1), IL-10 (Hs00961622_m1 ) and TNF (Hs00174128_m1 ). qPCR data were normalized to the geometric mean of ACTB, GAPDH and HPRT1.
EXAMPLE 5
GLOBAL T RANSCRIPT ANALYSIS
[0386] RNA sequencing (RNA-seq) was performed to identify global gene expression programs impacted by LSD1 inhibition in SARS-CoV-2-infected Caco-2 cells. LSD1 inhibition albeit at different degrees, impacts on key anti-viral processes, key proteins responsible for viral entry and the transcription and replication of the SARS-CoV-2 virus in the host cell (Figure 6A). Principal component analysis (PCA) demonstrated good separation of samples according to their treatment type and high similarity between biological replicates (Figure 6B). Principal component (PC)1 separated phenelzine-treated samples from all other samples and accounted for 66% of the variance, whilst PC2 further separated GSK-treated samples from the control samples and accounted for 12% of the variance (Figure 6B). Differential expression analysis revealed 1059 differentially expressed genes (DEGs) between phenelzine-treated vs. control samples and 73 DEGs between GSK-LSD1 treated vs. control samples, with 36 of these DEGs shared between the two contrasts (Figure 6C). Taken together, these results suggest that phenelzine and GSK-LSD1 regulate global host responses to prevent SARS-CoV-2 infection via different molecular mechanisms.
[0387] The different degrees of impact on these pathways by the LSD1 inhibitors can be attributed to the different modes of action of each inhibitor, with phenelzine impacting on both catalytic, nuclear and structural functions.
[0388] Next, the present inventors performed over-representation analysis using Reactome to investigate pathway enrichment of the DEGs from our two contrasts, phenelzine vs. control and GSK-LSD1 vs. control. Reactome enrichment analysis revealed that translation and MAPK pathways were enriched in phenelzine- treated samples, whilst GSK-LSD1 treatment downregulated genes enriched in the IFN pathway (Figure 6D-F).
EXAMPLE 6
INTERPLAY OF INTRACELLULAR ACE2 IN INFECTED CELLS
[0389] The signature of intracellular ACE2 was examined in infected cells, including nuclear and cytoplasmic fractions of ACE2 to understand the role of ACE2 in SARS-CoV-2 infection
[0390] In susceptible (e.g., permeabilized) SARS-COV-2 Caco-2 cells ACE2 polypeptide display increased cytoplasmic and nuclear expression cells (Figure 7). The Fn/c ratio (a score > than 1 indicates nuclear bias) of ACE2 also increased significantly upon infection. A similar pattern was demonstrated for LSD1.
Materials & Methods
RNA Sea Analysis
[0391] RNA-seq data were obtained from Caco-2 cell line infected with SARS-CoV-2. Three different treatments were tested (named Phe, GSK, and L1), with each of the treatments targeting the same gene but in different ways. A total of 8 samples from four experimental groups were collected:
[0392] · Caco-2 cells infected with SARS-CoV-2 control (2 x replicates);
[0393] · Caco-2 cells infected with SARS-CoV-2 treated with Phe (2 x replicates); [0394] · Caco-2 cells infected with SARS-CoV-2 treated with GSK-LSD1 (2 x replicates);
[0395] · Caco-2 cells infected with SARS-CoV-2 treated with L1 (2 x replicates).
[0396] The aim was to perform differential expression analysis using edgeR between the control group and each of the treated groups to find the differentially expressed genes. We then want to compare the genes between the treated groups to find common and unique genes, and also perform pathway analysis.
[0397] RNA-seq data were generated, fastq data were downloaded to the QIMR Berghofer server, and then archived to the HSM by Scott Wood. Sequence reads were trimmed for adapter sequences using Cutadapt (version 1.9; Martin (2011 )) and aligned using STAR (version 2.5.2a; Dobin et al. (2013)) to the GRCh37 assembly with the gene, transcript, and exon features of Ensembl (release 89) gene model, and the SARS-CoV-2 RefSeq accession NC_045512. Quality control metrics were computed using RNA-SeQC (version 1.1.8; DeLuca et al. (2012)) and expression was estimated using RSEM (version 1.2.30; Li and Dewey (2011)).
Quality control
[0398] The quality control of RNA-seq samples is an important step to guarantee quality and reproducible analytical results. RNA-SeQC was run for this purpose, the results of which can be found on the HPC cluster. Another common quality metric is whether the RNA sample is contaminated with mitochondrial DNA (mtDNA) or whether there is a high amount of ribosomal RNA (rRNA) in the sample. We determined the number of reads which mapped to Ensembl biotypes, including protein-coding genes, rRNA, and mitochondria. Given we use a threshold of 95% of reads mapping to protein coding regions, 7 samples passed this QC measure.
Normalisation
[0399] The aim of normalisation is to remove differences between samples based on systematic technical effects to warrant that these technical biases have a minimal effect on the results. The library size is important to correct for as differences in the initial RNA quantity sequenced will have an impact on the number of reads sequenced. Differences in RNA sequence composition occurs when RNAs are over-represented in one sample compared to others. In these samples, other RNAs will be under-sampled which will lead to higher false-positive rates when predicting differentially expressed genes.
Normalisation methods
[0400] In our analysis, we corrected for library size by dividing each sample’s gene count by million reads mapped. This procedure is a common approach known as counts per million (CPM). We further corrected for differences in RNA composition using a method proposed by Robinson and Oshlack (2010a) called trimmed mean of M values (TMM). We used the function calcNormFactors() from the edgeR package (Robinson, McCarthy, and Smyth (2010b)) to obtain TMM factors and used these to correct for differences in RNA composition.
Differential expression analysis
[0401] Differential expression (DE) analysis was performed using the R package edgeR (Robinson, McCarthy, and Smyth (2010b)). Note that the inputs for DE analysis are the filtered but not normalised read counts, since edgeR performs normalisation (library size and RNA composition) internally. The glmQLFit() function was used to fit a quasi-likelihood negative binomial generalised log-linear model to the read counts for each gene. Using the glmQLFTest() function, we conducted gene-wise empirical Bayes quasi-likelihood F-tests for a given contrast. As per the edgeR user’s guide, “the quasi-likelihood method is highly recommended for differential expression analyses of bulk RNA-seq data [versus the likelihood ratio test] as it gives stricter error rate control by accounting for the uncertainty in dispersion estimation.”
EXAMPLE 7
HBEC ALI CELL CULTURE WITH SARS-COV-2 INFECTION
[0402] Further increase of SARS-CoV-2 was detected at 48 hpi in control FIBECs from patient #549138 compared to 24 hpi samples (Figure 8). Interestingly, control HBECs from patient #501936 showed decreased SARS-CoV-2 replicates at 48 hpi compared to 24 hpi. Such changes can be explained by the individual variance response to SARS-CoV-2 infection. [0403] Strikingly, phenelzine treatment reduced the SARS-CoV-2 replicates in both infected HBECs at 24 hpi and 48 hpi as compared to the control group (Figure 8).
Materials & Methods
Air-liquid interface culture of HBECs
[0404] HBECs were obtained from two healthy donor (#501936 and #549138) and cultured in PneumaCult™-ALI Medium on collagen-coated transwell inserts with a 0.4-micron pore size (Costar, Corning, Tewksbury, MA, USA) and inserted into 24 well culture plates according to manufacturer instructions (StemCell Technologies, Cambridge, MA, USA). HBECs were maintained at air-liquid interface allowing them to differentiate. Medium in the basal chamber was changed every 2-3 days (500 mI).
Infection with SARS-CoV-2 in the presence of inhibitor
[0405] 24 h prior to infection, HBECs were treated with Phenelzine (400 mM; Sigma) in the basal chamber. All infection experiments were performed in a PC3 facility. At the time of infection, the inhibitor-containing media was removed and replaced with fresh media in the basal chamber. 104 plaque forming units (PFU) of SARS-CoV-2 virus inoculum in 100 mI was added to the apical compartment. After 2hr of virus adsorption at 37 °C, 5% C02, the unbound virus inoculum was removed, and cells were cultured with an air-liquid interface with inhibitor-containing media in the basal chamber for 24 hr and 48 hr post infection (24 hpi and 48 hpi). The cells were collected at 24 hpi and 48 hpi in TRIzol reagent, and RNA was extracted by using the Direct-zol RNA miniprep kit (Zymo Research, CA) following the manufacturer’s instructions. For ASI digital pathology, cells were fixed with 4% formaldehyde for 30 min at room temperature
Determination of virus titers using gRT-PCR
[0406] Viral titers (TCID50 equivalents per ml) in the extracted RNA was determined by qRT-PCR, using Real-time fluorescent RT-PCR kit for detecting 2019- nCoV (BGI, China) following the manufacturer’s instructions. Positive control (mix of pseudo-virus with target virus genes and internal reference) and blank control (DNase/RNase free water) were used as quality control. Limit of detection is 100 copies/ml. The quantity of viral genomes was calculated by normalizing to a viral stock with a known viral titer.
EXAMPLE 8
COLLECTION AND SAFE STORAGE OF SARS-COV-2 PATIENT CELLS [0407] Bronchoalveolar lavage cells (BALCs) and peripheral blood mononuclear cells (PBMCs) collection and storage; plasma collection, SARS-CoV2 virus detection, and secure storage.
Materials & Methods
[0408] Patients with SARS-CoV-2 infection only (cohort 1 , n = 5); early and advanced solid tumour cancer patients with SARS-CoV-2 infection (cohort 2, n = 5); and healthy donors (cohort 3, n = 5) are obtained. Written consent is provided for study participation and patients will be followed up as per standard national/local guidelines with regular clinical examination. Blood samples (40 ml total) are collected by a clinical trial nurse as part of standard blood collection. Clinicopathological/ virological data is collected by the clinical team. PBMCs are isolated according to well established protocols and stored in liquid nitrogen. Plasma is collected for SARS-CoV-2 detection by RT-PCR and stored at -80°C for the virus infection assay. BALF (20 ml/patient) is obtained and processed within 2 hours in a BSL-3 laboratory. BALCs are isolated by filtering and centrifugation before being resuspended in medium for future use.
Materials & Methods
[0409] Patients with SARS-CoV-2 infection only (cohort 1 , n = 5); early and advanced solid tumour cancer patients with SARS-CoV-2 infection (cohort 2, n = 5); and healthy donors (cohort 3, n = 5). Written consent will be obtained for study participation and patients will be followed up as per standard national/local guidelines with regular clinical examination. Blood samples (40 ml total) will be collected by a clinical trial nurse as part of standard blood collection. Clinicopathological/virological data will be collected by the clinical team. PBMCs will be isolated according to our established protocols and stored in liquid nitrogen. Plasma will be collected for SARS-CoV-2 detection by RT-PCR and stored at -80°C for the virus infection assay. BALF (20 ml/patient) will be obtained and processed within 2 hours in a BSL-3 laboratory. BALCs will be isolated by filtering and centrifugation before being resuspended in medium for future use.
EXAMPLE 9
INHIBITION OF VIRUS CELL ENTRY VIA THE ACE2/TMMSR2 MACHINERY
[0410] To address whether LSD1 demethylase activity is required for ACE2-spike protein interactions, Caco-2 cells were pre-treated with either phenelzine or GSK-LSD1 prior to and following SARS-CoV-2 infection. Phenelzine, and to a greater extent GSK-LSD1 treatment, reduced spike protein expression at the cell surface of infected cells at 48 hpi (Figure 9A, 9B). Furthermore, LSD1, ACE2, and TMPRSS2 mRNA transcripts remained largely unaltered in SARS-CoV- 2-infected Caco-2 cells following phenelzine or GSK-LSD1 treatment (Figure 9C).
[0411] After examining the effect of three MOA inhibitors on SARS-CoV-2 viral replication, viral entry, and T cell-mediated killing of SARS-CoV-2-infected cell, it was hypothesized that blocking LSD1 nuclear or catalytic activity will both block viral entry into the host cell and separately viral replication as well as inducing increased efficacy in T-cell mediated killing SARS-CoV-2-infected cells.
[0412] To determine whether GSK-LSD1 inhibited ACE2 demethylation at the cell surface of Caco-2 cells, cells were treated with inhibitor for 48 h followed by SARS-CoV-2 infection (MOI 1.0) for 1 hour and culture in inhibitor-containing medium for up to 48 hours (Figure 9D). Using a proximity ligation assay to assess co-localization, GSK-LSD1 treatment indeed increased the association between ACE2 and pan-methylation lysine antibody in infected cells; suggesting that LSD1 modulates the methylation status of ACE2 (Figure 9E).
[0413] A proximity ligation assay was used to assess the co-localization of ACE2 and spike protein at the surface of SARS-CoV-2 infected Caco-2 cells. GSK- LSD1 treatment significantly decreased interaction between ACE2 spike protein Figure 9F. This suggests that methylation of ACE2 via inhibition of LSD1 activity contributes to blocking access to ACE2 by the SARS-CoV-2 spike protein.
Materials & Methods
[0414] Caco-2 cells were tested negative for Mycoplasma contamination prior to experiments. Caco-2 cells were seeded at 2 x 105 cells per well in six-well plates in DEME (10% FBS) and incubated overnight at 37°C, 5% CO2. 48 h prior to infection, cells were treated with either 400 mM phenelzine or 400 mM GSK-LSD1. At the time of infection, plates were transferred to the BSL3 facility and inhibitor- containing medium was removed and replaced with SARS-CoV-2 virus inoculum (MOI 1) containing DEME (5% FBS). After 1 h incubation at 37°C, 5% CO2, the virus inoculum was removed and cells were washed three times with PBS to remove unbound virus prior to adding inhibitor-containing medium. 48 h post infection (48 hpi), the cell culture supernatant and cells were collected separately in TRIzol reagent and RNA was extracted using the Direct-zol RNA miniprep kit (Zymo Research, Irvine, CA) following the manufacturer’s instructions. For ASI digital pathology and FACS, cells were fixed with 4% formaldehyde for 30 min at room temperature.
Determination of viral titers by gRT-PCR
[0415] Viral titers (TCID50 equivalents per ml) in the extracted RNA were determined by qRT-PCR using a real-time fluorescent RT-PCR kit for detecting 2019-nCoV (BGI Genomics, China) following the manufacturer’s instructions.
Positive control (mix of pseudo-virus with target virus genes and internal reference) and blank control (DNase/RNase free water) were used as quality control. The limit of detection was 100 copies/ml. The quantity of viral genomes was calculated by normalizing to a viral stock with a known viral titre.
[0416] PBMCs are pre-treated with inhibitors and killing assays performed using the xCELLigence® Real Time Cell Analyzer.
Proximity ligation assay
[0417] The Duolink proximity ligation assay was employed using PLA probe anti-mouse PLUS (DU092001), PLA probe anti-rabbit MINUS (DU092005), and Duolink In Situ Detection Reagent Red Kit (DU092008) (Sigma Aldrich). Cells were fixed, permeabilized, and incubated with primary antibodies targeting LSD1 and ACE2. Cells were processed according to the manufacturer’s recommendations. Finally, coverslips were mounted onto slides and examined as above
[0418] Time-of-addition of inhibitors will be optimized based on SARS-CoV- 2 virus infection status: “entry” treatment, “post-entry” treatment, and “full-time” treatment. Viral RNA will be extracted from cell supernatants and quantified by qRT- PCR (Thevarajan etai, 2020) and inhibition of viral replication assayed using a modified plaque reduction assay and EC50s. The SARS-CoV-2 highly susceptible cell lines (Floffman etal., 2020) Calu-3, H1299, FlepG2, and Caco- 2 will be virus infected (MRC-5 cells as negative control).
[0419] SARS-CoV-2-infected cells with/without inhibitor treatments will be assayed by qRT-PCR for ACE2 and TMPRSS2 and flow cytometry and digital pathology using antibodies targeting ACE2 and TMPRSS2.
[0420] PBMCs will be pre-treated with inhibitors and killing assays performed using the xCELLigence® Real Time Cell Analyzer.
EXAMPLE 9
T CELL EXHAUSTION BIOMARKER AS A PREDICTIVE TOOL FOR DISEASE PROGRESSION AND IMMUNE EXHAUSTION IN SARS-CoV-2 PATIENTS
[0421] Establish the exhaustion signature in SARS-CoV-2 -infected patients at the protein, transcriptome, and epigenetic level with MOA inhibitors; Use our exhaustion biomarkers to track/predict patient outcomes. We hypothesize that our exhaustion biomarker signature will track increased exhaustion of patient immune cells preceding severe disease in that patient or when the patient has severe disease. This will allow us to predict which patients will have severe disease or mild disease. Treatment with MOAi inhibitors will enable us to track if we can reverse this exhaustion signature in pre-clinical patient immune cell samples.
Materials & Methods
[0422] Enriched BALCs and PBMCs will be treated in vitro with 3 MOAi and FACS and ASI’s mIF digital analysis used to: (i) phenotype immune cells; (ii) quantify T cell exhaustion, proliferation, and effector markers. High-resolution single-cell RNA, ATAC-seq, and LSD1p CFIIP-seq will be performed with our bioinformatics pipeline we will identify predictive biomarkers of SARS-CoV-2 progression related to immune exhaustion.
[0423] Enriched BALCs and PBMCs will be profiled with our established liquid biopsy digital pathology platform for our novel EOMES exhaustion biomarker in samples taken within 48 hours of presentation and matched samples taken 7-9 and 20 days later and related to clinical progression.
EXAMPLE 10
ESTABLISH PHARMACODYNAMIC PROFILE OF PHENELZINE IN AN NHP MODEL
[0424] In order to establish pharmacodynamic profiles, MOA inhibitors are assessed in vivo at two doses with four animals per dose. Parameters include: Haematological, Clinical follow up (body weight, temperature), Xray scanner, blood chemistry, local and systemic reaction. Inflammation parameters, Full pathology after necropsy in case of dose lethality. This will track both the safety profile of our MOAi inhibitors as well as their effect on effect on the NHP immunological model and if MOAi inhibition is able to improve recovery from SARS-CoV-2 challenge.
Materials & Methods
[0425] MOA inhibitors were assessed at two doses for each. Four animals per dose are tested (8 animals per group), challenged with SARS-CoV-2. A control group with no drugs will be included. In total the study will include 4 groups with 8 animals for 3 of them and 4 animals for the control group (28 animals). We will test multiple parameters will be followed and studied at the IDMIT, to check safety and efficacy. These include: (1) Haematological parameters, (2) Clinical follow up (body weight, temperature, repeated clinical examination), (3) Food and drinking consumption, (4) Xray scanner, (5) Blood chemistry, (6) Local and systemic reaction (if drug injected), (7) Inflammation parameters (CRP, cytokine), This work will be carried out in collaboration with the IDMIT, which has a unique infrastructure in Europe for health and biology science, supporting highly demanding and costing animal facilities for NHP models that includes PET-CT in vivo imaging, two-photon and confocal microscopy, flow and mass cytometry, BSL2 and BSL3 labs. It is important to note that the facility is equipped with state-of-the-art instruments for in vivo imaging technologies with the following objectives: (1) track antigens and therapeutics in infected hosts; (2) visualize host responses to vaccination and/or treatment, with a specific focus on dynamics of immune effectors: (3) track microbes dissemination in the host; (4) refine and reduce using high numbers of animal models for preclinical research; (5) translate part of the technology to the clinical practice in humans.
REFERENCES
Kosugi, S., Hasebe, M., Entani, T., Takayama, S., Tomita, M., and Yanagawa, H. (2008). Design of peptide inhibitors for the importin alpha/beta nuclear import pathway by activity-based profiling. Chemistry & biology 15, 940-949.
Kosugi, S., Hasebe, M., Matsumura, N., Takashima, H., Miyamoto-Sato, E., Tomita, M., and Yanagawa, H. (2009). Six classes of nuclear localization signals specific to different binding grooves of importin alpha. The Journal of biological chemistry 284, 478-485.
Kosugi, S., Hasebe, M., Tomita, M., and Yanagawa, H. (2009). Systematic identification of cell cycle-dependent yeast nucleocytoplasmic shuttling proteins by prediction of composite motifs. Proceedings of the National Academy of Sciences of the United States of America 106, 10171 -10176.
Wen, P. P„ Shi, S. P„ Xu, H. D„ Wang, L. N„ and Qiu, J. D. (2016). Accurate in silico prediction of species-specific methylation sites based on information gain feature optimization. Bioinformatics (Oxford, England) 32, 3107-3115.
Blom, N., Sicheritz-Ponten, T., Gupta, R., Gammeltoft, S., and Brunak, S. (2004). Prediction of post-translational glycosylation and phosphorylation of proteins from the amino acid sequence. Proteomics 4, 1633-1649.
Su, H., Liu, M., Sun, S., Peng, Z., and Yang, J. (2019). Improving the prediction of protein-nucleic acids binding residues via multiple sequence profiles and the consensus of complementary methods. Bioinformatics (Oxford, England) 35, 930- 936.
Ofran, Y., and Rost, B. (2007). ISIS: interaction sites identified from sequence. Bioinformatics (Oxford, England) 23, e13-16.
Huang, J., Sengupta, R., Espejo, A. B., Lee, M. G., Dorsey, J. A., Richter, M.,
Opravil, S., Shiekhattar, R., Bedford, M. T., Jenuwein, T., and Berger, S. L. (2007). p53 is regulated by the lysine demethylase LSD1. Nature 449, 105-108.
Wang, H. et al. SARS coronavirus entry into host cells through a novel clathrin-and caveolae-independent endocytic pathway. Cell Research 18, 290-301 (2008). Yang, J. etal. Reversible methylation of promoter-bound STAT3 by histone-modifying enzymes. Proceedings of the National Academy of Sciences of the United States of America 107, 21499-21504, doi:10.1073/pnas.1016147107 (2010).
Sakane, N. et al. Activation of HIV transcription by the viral Tat protein requires a demethylation step mediated by lysine-specific demethylase 1 (LSD1/KDM1). PLoS Pathogens 7, e1002184, doi:10.1371/journal.ppat.1002184 (2011).
Tan, A. H. Y. et al. Lysine-Specific Histone Demethylase 1A Regulates Macrophage Polarization and Checkpoint Molecules in the Tumor Microenvironment of Triple- Negative Breast Cancer. Frontiers in Immunology 10, 1351 , doi :10.3389/fimmu.2019.01351 (2019).

Claims

THE CLAIMS DEFINING THE INVENTION ARE AS FOLLOWS:
1. A method of treating a coronavirus infection in a subject having a coronavirus infection, the method comprising administering to the subject an agent that inhibits or reduces an activity of a lysine-specific demethylase 1 (LSD1) polypeptide, to thereby treat the coronavirus infection in the subject.
2. A method of preventing a coronavirus from entering a cell, the method comprising, exposing the cell to an agent that inhibits or reduces an activity of a LSD1 polypeptide, to thereby reduce or prevent the coronavirus from entering the cell.
3. The method of claim 2, wherein the agent that inhibits or reduces an activity of a LSD1 polypeptide is exposed to the cell for a time and under conditions sufficient to antagonise a TMPRSS2 polypeptide.
4. The method of claim 2, wherein the agent that inhibits or reduces an activity of a LSD1 polypeptide is exposed to the cell for a time and under conditions sufficient to antagonise a virus cell entry receptor polypeptide.
5. The method of claim 4, wherein the virus cell entry receptor polypeptide is an ACE2 polypeptide.
6. The method of any one of claim 4 or claim 5, wherein the activity of the LSD1 polypeptide is the demethylation of ACE2 residue K31.
7. A method of reducing or preventing entry of a coronavirus into a cell, the method comprising exposing the cell to an agent that inhibits an activity of a lysine- specific demethylase 1 (LSD1) polypeptide, to thereby antagonise, or otherwise reduce the level or amount of TMPRSS2 polypeptide present on the surface of the cell.
8. A method of reducing or preventing entry of a coronavirus into a cell, the method comprising exposing the cell to an agent that inhibits an activity of a lysine- specific demethylase 1 (LSD1) polypeptide, to thereby antagonise, or otherwise reduce the level or amount of a virus cell entry receptor polypeptide present on the surface of the cell.
9. The method of any one of claims 1 -8, wherein the agent that inhibits or reduces an activity of a LSD1 polypeptide is selected from the group comprising: a small molecule; a polypeptide; and an antigen-binding molecule.
10. The method of any one of claims 1 -9, wherein the agent is a monoamine oxidase (MAO) inhibitor.
11. The method of claim 10, wherein the MAO inhibitor is selected from the group comprising phenelzine, bizine, tranylcypromine, pargyline, selegiline, selegiline hydrochloride, dimethylselegilene, brofaromine, moclobemide, beflozatone, safinamide, isocarboxazid, nialamide, rasagiline, iproniazide, iproclozide, toloxatone, bifemelane, desoxypeganine, harmine, harmaline, and linezolid.
12. The method of any one of claims 1-11, wherein the agent is bizine and has the molecular structure of:
Figure imgf000122_0001
13. The method of any one of claims 1 -9, wherein the agent is a selective LSD1 inhibitor.
14. The method of any one of claims 1 -9, wherein the agent is GSK-LSD, which comprises the molecular structure:
Figure imgf000122_0002
15. The method of any one of claims 1 -9, wherein the agent is SP-2509, which comprises the molecular structure:
Figure imgf000123_0001
16. The method of any one of claims 1 -9, wherein the agent is SP-2577, which comprises the molecular structure:
Figure imgf000123_0002
17. The method of any one of claims 1 -9, wherein the agent is an isolated or purified proteinaceous molecule comprising, consisting or consisting essentially of sequence corresponding to residues 108 to 118 of LSD1.
18. The method of claim 17, wherein the isolated or purified proteinaceous molecule is an isolated or purified proteinaceous molecule represented by Formula (I):
Z1RRTX1RRKRAKVZ2 (I) wherein:
Zi and Z2 are independently absent or are independently selected from at least one of a proteinaceous moiety comprising from about 1 to about 50 amino acid residues (and all integers in between), and a protecting moiety; and
Xi is selected from small amino acid residues, including S, T, A, G, and modified forms thereof.
19. The method of claim 18, wherein Xi is selected from S and A.
20. The method of claim 18 or claim 19, wherein Zi is a proteinaceous molecule represented by Formula (II):
X2X3X4 (II) wherein:
X2 is absent or is a protecting moiety;
X3 is absent or is selected from any amino acid residue; and X4 is selected from any amino acid residue.
21. The method of claim 20, wherein X3 is selected from basis amino acid residues including R, K, and modified forms thereof.
22. The method of claim 20 or claim 21 , wherein X4 is selected from aromatic amino acid residues, including F, Y, W, and modified forms thereof.
23. The method of any one of claim 18-22, wherein Z2 is absent.
24. The method according to any one of claims 18-23, wherein the isolated or purified proteinaceous molecule of Formula (I) comprises, consists or consists essentially of an amino acid sequence represented by SEQ ID NO: 1 , 2, or 3:
RRTSRRKRAKV [SEQ ID NO: 1];
RRTARRKRAKV [SEQ ID NO: 2]; or RWRRTARRKRAKV [SEQ ID NO: 3]
25. The method of any one of claims 18-24, wherein the proteinaceous molecule of Formula I further comprises at least one membrane permeating moiety.
26. The method of claim 25, wherein the membrane permeating moiety is a lipid moiety.
27. The method of claim 25 or claim 26, wherein the membrane permeating moiety is a myristoyl group.
28. The method of any one of claims 1 -27, wherein the coronavirus is selected from the group comprising SARS-CoV, SARS-CoV-2, and MERS-CoV.
29. The method of any one of claims 1 -28, wherein the coronavirus is selected from SARS-CoV-2.
30. The method of any one of claims 1 -8, wherein the agent that inhibits or reduces an activity of a LSD1 polypeptide prevents the phosphorylation of the LSD polypeptide.
31. The method of any one of claims 1 -8, wherein the agent that inhibits or reduces an activity of a LSD1 polypeptide prevents the demethylation of the ACE2 polypeptide.
32. The method of claim 31 , wherein the agent that inhibits or reduces an activity of a LSD polypeptide prevents the interaction between an LSD polypeptide and a PKC0 polypeptide.
33. The method of claim 31 or claim 32, wherein the agent that inhibits or reduces an activity of a LSD polypeptide binds to a PKC0 polypeptide.
34. A method of treating a coronavirus infection in a subject having a coronavirus infection, the method comprising administering to the subject an agent that reduces the level of a lysine-specific demethylase 1 (LSD1) polypeptide, to thereby treat the coronavirus infection in the subject.
35. The method of claim 34, wherein the agent is an interfering nucleic acid.
36. A method of treating a coronavirus infection in a subject having a coronavirus infection, the method comprising administering to the subject an agent that reduces the level or activity of a CoREST polypeptide, to thereby treat the coronavirus infection in the subject.
37. Use of a LSD1 inhibitor for the treatment of a coronavirus infection.
38. A composition comprising (i) an agent that inhibits or reduces an activity of a lysine-specific demethylase-1 (LSD1) polypeptide; and (ii) an antiviral agent.
39. The composition of claim 38, wherein the agent that inhibits or reduces an activity of a LSD1 polypeptide is a MAO inhibitor.
40. The composition of claim 39, wherein the MAO inhibitor is selected from the group comprising phenelzine, bizine, tranylcypromine, pargyline, selegiline, selegiline hydrochloride, dimethylselegilene, brofaromine, moclobemide, beflozatone, safinamide, isocarboxazid, nialamide, rasagiline, iproniazide, iproclozide, toloxatone, bifemelane, desoxypeganine, harmine, harmaline, and linezolid.
41. The method of any one of claims 34-38, wherein the agent that inhibits or reduces an activity of a LSD1 polypeptide is GSK-LSD, which comprises the molecular structure:
Figure imgf000126_0001
42. method of any one of claims 34-38, wherein the agent that inhibits or reduces an activity of a LSD1 polypeptide is SP-2509, which comprises the molecular structure:
Figure imgf000126_0002
43. The method of any one of claims 34-38, wherein the agent that inhibits or reduces an activity of a LSD1 polypeptide is SP-2577, which comprises the molecular structure:
Figure imgf000127_0001
44. The composition of any one of claims 38-43, wherein the antiviral agent is selected from the group comprising hydroxychloroquine, chloroquine, lopinavir, ritonavir, favipiravir, and remdesivir.
45. The composition of claim 44, wherein the antiviral agent comprises an IFN-b polypeptide.
46. A method of inhibiting the phosphorylating activity of a protein kinase C (PKC), comprising contacting a cell infected with a coronavirus with an isolated or purified proteinaceous molecule comprising, consisting or consisting essentially of a sequence corresponding to residues 108 to 118 of LSD1.
47. A method of preventing S protein priming by a coronavirus, the method comprising administering a TMPRSS2 antagonist to a cell infected by the coronavirus, wherein the TMPRSS2 antagonist is an inhibitor of the interaction between an LSD1 polypeptide and a PKC0 polypeptide.
48. A method of inhibiting coronavirus entry into a cell, the method comprising exposing the cell to an LSD1 inhibitor, wherein the LSD1 inhibitor reduces the expression of ACE2 by the cell, to thereby inhibit coronavirus entry into the cell.
49. A method of preventing coronavirus entry into a cell, the method comprising exposing the cell to an LSD1 inhibitor, wherein the LSD1 inhibitor reduces the translocation of an ACE2 polypeptide from the cell surface to the cell nucleus, to thereby prevent coronavirus entry into the cell.
50. The method of one of claims 34-36, or 44-49, wherein the coronavirus is a betacoronavirus.
51. The method of claim 50, wherein the betacoronavirus is a SARS-CoV.
52. The method of claim 51 , wherein the SARS-CoV is SARS-CoV-2.
PCT/AU2021/050317 2020-04-03 2021-04-06 "methods for treatment of coronavirus infections" WO2021195723A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
AU2020901054 2020-04-03
AU2020901054A AU2020901054A0 (en) 2020-04-03 Methods for treatment of virus infections
AU2020902492 2020-07-17
AU2020902492A AU2020902492A0 (en) 2020-07-17 Novel methods for treating virus infections

Publications (1)

Publication Number Publication Date
WO2021195723A1 true WO2021195723A1 (en) 2021-10-07

Family

ID=77926849

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/AU2021/050317 WO2021195723A1 (en) 2020-04-03 2021-04-06 "methods for treatment of coronavirus infections"

Country Status (1)

Country Link
WO (1) WO2021195723A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022221920A1 (en) * 2021-04-20 2022-10-27 The Council Of The Queensland Institute Of Medical Research Novel compositions and methods for treating coronavirus infections
WO2024020646A1 (en) * 2022-07-27 2024-02-01 The Council Of The Queensland Institute Of Medical Research Methylation biomarker for infection resistance

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010011845A2 (en) * 2008-07-24 2010-01-28 The United States Of America, As Represented By The Secretary, Department Of Health & Human Services Methods of preventing or treating viral infection or reactivation after latency in a host using inhibitors of the lsd1 protein
WO2011106105A2 (en) * 2010-02-24 2011-09-01 Oryzon Genomics, S.A. Inhibitors for antiviral use
WO2019104381A1 (en) * 2017-11-29 2019-06-06 University Of Canberra Enhancing t-cell function and treating a t-cell dysfunctional disorder with a combination of an lsd inhibitor and a pd1 binding antagonist

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010011845A2 (en) * 2008-07-24 2010-01-28 The United States Of America, As Represented By The Secretary, Department Of Health & Human Services Methods of preventing or treating viral infection or reactivation after latency in a host using inhibitors of the lsd1 protein
WO2011106105A2 (en) * 2010-02-24 2011-09-01 Oryzon Genomics, S.A. Inhibitors for antiviral use
WO2019104381A1 (en) * 2017-11-29 2019-06-06 University Of Canberra Enhancing t-cell function and treating a t-cell dysfunctional disorder with a combination of an lsd inhibitor and a pd1 binding antagonist

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
ALSAQER SAQER F., TASHKANDI MUSTAFA M., KARTHA VINAY K., YANG YA-TING, ALKHERIJI YAZEED, SALAMA ANDREW, VARELAS XARALABOS, KUKURUZ: "Inhibition of LSD1 epigenetically attenuates oral cancer growth and metastasis", ONCOTARGET, vol. 8, no. 43, 26 September 2017 (2017-09-26), pages 73372 - 73386, XP055925847, DOI: 10.18632/oncotarget.19637 *
PRUSEVICH POLINA ET AL: "A Selective Phenelzine Analogue Inhibitor of Histone Demethylase LSD1", ACS CHEMICAL BIOLOGY, vol. 9, no. 6, 1 June 2014 (2014-06-01), pages 1284 - 1293, XP002773269, DOI: 10.1021/cb500018s *
SAMUEL CHARLES E.: "Antiviral Actions of Interferons", CLINICAL MICROBIOLOGY REVIEW, WASHINGTON, DC., US, vol. 14, no. 4, 1 October 2001 (2001-10-01), US , pages 778 - 809, XP055925848, ISSN: 0893-8512, DOI: 10.1128/CMR.14.4.778-809.2001 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022221920A1 (en) * 2021-04-20 2022-10-27 The Council Of The Queensland Institute Of Medical Research Novel compositions and methods for treating coronavirus infections
WO2024020646A1 (en) * 2022-07-27 2024-02-01 The Council Of The Queensland Institute Of Medical Research Methylation biomarker for infection resistance

Similar Documents

Publication Publication Date Title
US20210121496A1 (en) Methods and compositions for modulating cancer stem cells
AU2018377852B2 (en) Enhancing T-cell function and treating a T-cell dysfunctional disorder with a combination of an LSD inhibitor and a PD1 binding antagonist
US20200289437A1 (en) Compositions for Modulating Cancer Stem Cells and Uses Therefor
WO2021195723A1 (en) &#34;methods for treatment of coronavirus infections&#34;
US20220160710A1 (en) Compositions and methods for treating or preventing alzheimer&#39;s disease
US10736892B2 (en) Stem cell modulation II
WO2017189856A2 (en) Compositions and methods for treating cancer
WO2007140263A9 (en) Macrophage migration inhibitory factor antagonists and methods of using same
JP2013510832A (en) PAR-1 antagonist for the treatment or prevention of influenza A virus infection
US10106797B2 (en) Methods for suppressing adipocyte differentiation
WO2021195088A1 (en) TGF-Bβ1 INHIBITORS FOR PREVENTING AND TREATING SARS-COV-2
US20120231015A1 (en) Fragile x mental retardation protein (fmrp), compositions, and methods related thereto
WO2019067468A1 (en) Modulation of immune response through stimulator of interferon genes
Li et al. Duck karyopherin α4 (duKPNA4) is involved in type I interferon expression and the antiviral response against Japanese encephalitis virus
EP4257146A1 (en) Cystic lymphangioma treatment drug
US20190336579A1 (en) Compositions and methods to induce targeted apoptosis by modulating protein tyrosine phosphatase 13 (ptpn13)
US20200282010A1 (en) Immunogenic compositions and uses therefor

Legal Events

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

Ref document number: 21781296

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21781296

Country of ref document: EP

Kind code of ref document: A1