WO2017066459A1 - INHIBITION OF NEDDYLATION USING GLYCYL-tRNA SYNTHETASE INHIBITORS - Google Patents

INHIBITION OF NEDDYLATION USING GLYCYL-tRNA SYNTHETASE INHIBITORS Download PDF

Info

Publication number
WO2017066459A1
WO2017066459A1 PCT/US2016/056862 US2016056862W WO2017066459A1 WO 2017066459 A1 WO2017066459 A1 WO 2017066459A1 US 2016056862 W US2016056862 W US 2016056862W WO 2017066459 A1 WO2017066459 A1 WO 2017066459A1
Authority
WO
WIPO (PCT)
Prior art keywords
glyrs
inhibitor
cell
protein
gene
Prior art date
Application number
PCT/US2016/056862
Other languages
French (fr)
Inventor
Xiang-Lei Yang
Zhongying MO
Paul Schimmel
Original Assignee
The Scripps Research Institute
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The Scripps Research Institute filed Critical The Scripps Research Institute
Priority to EP16856197.5A priority Critical patent/EP3362086A1/en
Priority to US15/767,627 priority patent/US20190381086A1/en
Publication of WO2017066459A1 publication Critical patent/WO2017066459A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/25Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving enzymes not classifiable in groups C12Q1/26 - C12Q1/66
    • 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
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7076Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines containing purines, e.g. adenosine, adenylic acid
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • 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
    • C12N2310/12Type of nucleic acid catalytic nucleic acids, e.g. ribozymes
    • C12N2310/122Hairpin
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2440/00Post-translational modifications [PTMs] in chemical analysis of biological material
    • G01N2440/36Post-translational modifications [PTMs] in chemical analysis of biological material addition of addition of other proteins or peptides, e.g. SUMOylation, ubiquitination

Definitions

  • compositions and methods for inhibiting neddylation using Glycyl-tRNA synthase (GlyRS) inhibitors and related compositions and methods for treating diseases such as cancer.
  • GlyRS Glycyl-tRNA synthase
  • the NEDD8 pathway plays a critical role in the activation of the ubiquitin E3 ligase activity of cullin-RING ligase (CRL) E3s via the covalent attachment of NEDD8 to the core cullin protein of these enzyme complexes.
  • CRL cullin-RING ligase
  • This process of neddylation has been shown to be essential for the E3 ligase activity of CRLs.
  • CRLs are a large superfamily of E3s that are responsible for the ubiquitination of multiple substrate proteins, including several that are involved in the regulation of normal cellular function as well as some that have been shown to be associated with cancer.
  • Some embodiments disclosed herein relate to methods of reducing neddylation in a cell.
  • the methods comprise: contacting a cell with a composition comprising a Glycyl-tRNA synthetase (GlyRS) inhibitor, wherein the level of neddylation is decreased in the cell.
  • GlyRS Glycyl-tRNA synthetase
  • Some embodiments relate to methods of reducing neddylation in a cell population.
  • the methods comprise: acquiring knowledge of the level of neddylation in a cell population; and contacting the cell population with a composition comprising a Glycyl-tRNA synthetase (GlyRS) inhibitor to thereby decrease the level of neddylation in the cell population.
  • Some embodiments relate to methods of reducing neddylation in a cell population.
  • the methods comprise: identifying a cell population having undesirable level of neddylation; and contacting the ceil population with a composition comprising a Glycyl-tRNA synthetase (GlyRS) inhibitor to thereby decrease the level of neddylation in the cell population.
  • GlyRS Glycyl-tRNA synthetase
  • the composition can be, for example, a pharmaceutical composition.
  • the GlyRS inhibitor inhibits GlyRS functions in aminoacylation and neddylation. In some embodiments, the GlyRS inhibitor does not significantly inhibit GlyRS function in aminoacylation.
  • the GlyRS inhibitor is an inhibitor for a mammalian GlyRS protein. In some embodiments, the GlyRS inhibitor is an inhibitor for a human GlyRS protein. In some embodiments, the human GlyRS protein comprises an amino acid sequence having at least 90% identity to the amino acid sequence set forth in SEQ ID NO: 2. In some embodiments, the GlyRS inhibitor is an inhibitor for a plant GlyRS protein.
  • the GlyRS inhibitor can be, for example, a protein, a nucleic acid, a small molecule compound, or a combination thereof.
  • the GlyRS inhibitor is an RNA molecule capable of silencing the expression of a GlyRS gene.
  • the GlyRS inhibitor is an RNA molecule that binds to an mRNA encoded by a GlyRS gene.
  • the GlyRS gene is a mammalian GlyRS gene, for example a human GARS gene.
  • the human GARS gene consists of or comprises a nucleotide sequence having at least 90% sequence identity to the nucleotide sequence set forth in SEQ ID NO: 1.
  • the GlyRS inhibitor is a short- hairpin RNA (shRNA) consisting of or comprising a nucleotide sequence having at least 90% sequence identity to the nucleic acid sequence of SEQ ID NOs: 3-35.
  • shRNA short- hairpin RNA
  • the GlyRS inhibitor is a RNA consisting of or comprising the nucleic acid sequence of SEQ ID NOs:3 ⁇ 35
  • the GlyRS inhibitor is GlySA or a derivative thereof.
  • the GlyRS inhibitor interferes with the binding between the GlyRS protein and NEDD8 in the cell or the cell population. In some embodiments, the GlyRS inhibitor interferes with the binding between the GlyRS protein and Ubcl2 in the cell or the cell population. In some embodiments, the GlyRS inhibitor interferes with the binding between the GlyRS protein and NEDD8-conjugated Ubcl2 in the cell or the cell population. In some embodiments, the GlyRS inhibitor binds to one or more of amino acids 84-93 of SEQ ID NO: 2. In some embodiments, the GlyRS inhibitor binds to one or more of amino acids 232-238 of SEQ ID NO: 2.
  • the GlyRS inhibitor binds to one or more of amino acids Arg277, Glu279, Val289, Glu296, Ue404, and Glu552 of SEQ ID NO: 2.
  • the GlyRS inhibitor is a GlySA derivative.
  • the GlyRS inhibitor decreases the amount of NEDD8-conjugated Ubcl2 in the cell or the cell population.
  • the GlyRS inhibitor decreases Ubcl2 activity in the cell or the cell population.
  • the GlyRS inhibitor increases Ubcl2 degradation in the ceil or the cell population.
  • the GlyRS inhibitor interferes with the binding between a GlyRS protein and heterodimeiic El enzyme for neddylation (APPBP1/UBA3) in the cell or the cell population. In some embodiments, the GlyRS inhibitor decreases neddylation of a cuilin protein in the cell or the cell population.
  • the cell is a mammalian cell.
  • the contacting is performed in vitro, ex vivo, or in vivo.
  • the ceil or the ceil population is present in a tissue or in a body of a subject.
  • the level of neddylation in the cell or the cell population is reduced by at least [0012]
  • Some embodiments disclosed herein relate to isolated double-stranded ribonucleic acid (dsRNA) molecules that inhibit expression of a Glycyl-tRNA synthetase (GlyRS) gene, wherein a first strand of the dsRNA is substantially identical to at least 19 consecutive nucleotides of the GlyRS gene, and a second strand of the dsRNA is substantially complementary to the first strand.
  • the GlyRS gene is a mammalian GlyRS gene, for example a human GARS gene.
  • the human GARS gene comprises or consists of a nucleotide sequence having at least 90% identity to the nucleotide sequence set forth in SEQ ID NO: 1.
  • the GlyRS gene is a plant GlyRS gene.
  • the dsRNA is encoded by a polynucleotide, wherein the first strand and the second strand of the dsRNA are transcribed from said polynucleotide and form a hairpin loop.
  • GlyRS Glycyl-tRNA synthetase
  • the GlyRS gene is a mammalian GlyRS gene, for example a human GARS gene.
  • the human GARS gene comprises or consists of a nucleotide sequence having at least 90% identity to the nucleotide sequence set forth in SEQ ID NO: I .
  • the GlyRS gene is a plant GlyRS gene.
  • Some embodiments disclosed herein relate to methods of identifying an inhibitor of neddylation.
  • the methods comprise: providing a test compound; testing the testcompound for its ability to reduce or inhibit the binding between a Glycyl-tRNA synthetase (GlyRS) protein and NEDDS-conjugated Ubcl2; and identifying the compound as an inhibitor of neddylation if the compound has the ability to reduce or inhibit the binding between the GlyRS protein and NEDDS-conjugated Ubcl2.
  • the inhibitors bind to the catalytic domain of the GlyRS protein.
  • the GlyRS protein can be a mammalian GlyRS protein, for example a human GlyRS protein.
  • the human GlyRS protein comprises or consists of an amino acid sequence having at least 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:2.
  • the methods comprise testing the test compound for its ability to reduce or inhibit the aminoacyiation activity of the GlyRS protein.
  • the methods comprise testing one or more additional test compounds for their ability to reduce or inhibit the binding between the GlyRS protein and NEDD8-conjugated Ubcl2.
  • the methods comprise testing the one or more additional test compounds for their ability to reduce or inhibit the aminoacylation activity of the GlyRS protein.
  • compositions comprising a Glycyl-tRNA synthetase (GlyRS) inhibitor, for example pharmaceutical compositions comprising one or more pharmaceutically acceptable excipients.
  • GlyRS inhibitor is an isolated siR A. molecule that binds to an mRNA of the GlyRS protein.
  • the GlyRS inhibitor is a molecule that inhibits binding between the GlyRS protein and NEDD8-conjugated Ubcl2.
  • the GlyRS inhibitor is GlySA or a derivative thereof.
  • the GlyRS inhibitor inhibits GlyRS functions in aminoacylation and neddylation. In some embodiments, the GlyRS inhibitor does not significantly inhibit GlyRS function in aminoacylation.
  • Some embodiments disclosed herein relate to methods of reducing cell proliferation.
  • the methods comprise: contacting a cell with a composition comprising a Glycyl-tRNA synthetase (GlyRS) inhibitor, whereby the proliferation of the cell is reduced.
  • GlyRS Glycyl-tRNA synthetase
  • the activit of the CRLl (cullinl - RING) ubiquitin ligases is inhibited in the cell.
  • the activity of a substrate of the CRLl ubiquitin ligase is increased in the cell.
  • the substrate of the CRLl ubiquitin ligase is selected from the group consisting of c-Myc, c-Jun, cyclin E, Emi l , Cdt-1, ⁇ , NRF2, HIF-!a, ⁇ -catenin, Cdc25A, mTOR, BimEL and p27.
  • the methods comprise providing MLN4924 to the ceil.
  • the cell can be, for example, a mammalian cell, a plant cell.
  • the proliferation of the cell is reduced by at least 50%
  • the GlyRS inhibitor inhibits GlyRS functions in aminoacylation and neddylation. In some embodiments, the GlyRS inhibitor does not significantly inhibit GlyRS function in aminoacylation.
  • the methods comprise: administering a therapeutically effective amount of a pharmaceutical composition comprising a Glycyl-tRNA synthetase (GlyRS) inhibitor to a subject in need thereof.
  • the pharmaceutical composition further comprises one or more of additional therapeutic agents.
  • the methods comprise administering one or more additional pharmaceutical compositions comprising one or more of additional therapeutic agents.
  • the cancer is breast cancer, ovarian cancer, lung cancer, breast duct carcinoma, colorectal adenocarcinoma and lung squamous cell carcinoma, or a combination thereof.
  • the cancer is selected from the group consisting of breast cancer, cervical cancer, colon cancer, liver cancer, prostate cancer, melanoma, ovarian cancer, lung cancer, renal cell carcinoma, Schwannoma, mesothelioma, acute myeloid leukemia, multiple myeloma, non-Hodgkin lymphoma, and a combination thereof.
  • the cancer is a solid tumor.
  • the cancer is a hematological malignancy.
  • the GlyRS inhibitor is GlySA or a GlySA derivative.
  • the GlyRS inhibitor inhibits GlyRS functions in aminoacyiation and neddylation.
  • the GlyRS inhibitor does not significantly inhibit GlyRS function in aminoacyiation.
  • the GlyRS inhibitor is an inhibitor for a human GlyRS
  • FIG. 1A A schematic flowchart of the neddylation pathway.
  • Fig. IB The domain composition of human GlyRS.
  • Fig. 1C NEDD8, but not ubiquitin and SUMO I , specifically binds to His- tagged GlyRS, but not His-tagged SerRS and TrpRS.
  • Fig. ID GlyRS binds to NEDD8 in vivo.
  • FIEK293 cells were transfected with Myc-NEDD8 constructs. 48 hours after transfection ceils were harvested and lysed with acid lysis buffer and used for immunoprecipitation assay.
  • Fig. IE Domain mapping by His-Tag pull-down assay suggests that NEDD8 binds to catalytic domain of GlyRS,
  • FIG. 2A shows that GlyRS plays critical role in neddylation.
  • Fig. 2A Over-expression of human GlyRS specifically leads to increase of Ubcl 2 N8 in HEK293 ceils. The cells were transfected with pcDNA6V5 vectors containing the indicated gene fragment with a V5-tag.
  • Figs. 2B-D Ubcl2 N8 and substrates neddylation decrease specifically upon GlyRS knock-down. Knockdown of the expression of GlyRS, but not SerRS, specifically decreases the levels of Ubcl2 N8 , and NEDD8-modified cullin proteins in HeLa cells.
  • HeLa cells were transfected by pLenti vectors containing either a scramble sequence, SerRS or GlyRS specific sequences respectively. Ceils were harvested 48 hours after transfection and iysed with acid lysis buffer and subjected to SDS-PAGE.
  • FIGS 3A-H show that GlyRS preferentially binds to and promote Ubcl2 N8 .
  • Fig. 3A Co-immunoprecipitation indicates that GlyRS prefers binding to Ubel2 N8 than to the apo Ubcl2.
  • V5 -tagged GlyRS and Flag-tagged Ubcl2-Cl l l S genes were co- transfected to HEK293 cell for the assay.
  • Fig, 3B Strong binding of Ubcl2 N8 to GlyRS as determined by biolayer interferometry.
  • A.U. indicates Arbitrary Unit.
  • Fig. 3C Biolayer interferometry analysis confirms that GlyRS binding to Ubcl2 N8 .
  • Binding analysis to immobilized Ubcl2 N8 was carried out with a range of concentrations of full-length (8.07 nM-129 nM) GlyRS.
  • Fig. 3D Structural model of the GiyRS-Ubcl2 N8 interaction generated using Patchdock (a server for molecular docking). Figs.
  • 3E-G Biolayer interferometry analysis confirms that F84-L93 and I232-M238 regions, but not Insertion 1, are important for GlyRS binding to Ubcl2 N8 . Binding analysis to immobilized Ubcl2 N8 was carried out with a range of concentrations (1.25-20 ⁇ ' ⁇ ) of AF84-L93 (8.16-130 nM), ⁇ 232- ⁇ 238 (8.13-130 nM), and Assertion I (62,5-125 nM) GlyRS.
  • Fig. 3H Over-expression of ⁇ 232- ⁇ 238 GlyRS in HEK293 ceils cannot promote NEDD8 conjugation of Ubcl 2.
  • HE 293 cells were transfected with pcDNA6V5 vectors containing the indicated gene fragment with a V5-tag.
  • FIGS 4A-E show that GlyRS captures Ubcl2 N8 and escorts it to substrates.
  • Fig. 4A GlyRS binds to APPBP1 in HEK293 cells. Ceils were lysed with lysis buffer and used for immunoprecipitation assay.
  • Fig. 43B Structural model for how GlyRS protects UbcI 2 N8 during the neddylation cascade. The interaction between GlyRS and El (APPBPl subunit) is modeled by using the Patchdock server. The interaction between E1 N8 and Ubcl2 is adapted from a crystal structure (PDB 2NVU). The crystal structure of Ubcl2 N8 is from PDB 4P50.
  • the black dotted line indicates the calculated sum of the binding curves for Ubcl2 N8 and GlyRS to El .
  • Fig. 4D The ABD domain alone lacks the synergistic effect with Ubcl 2 NS for binding to El .
  • Binding analysis to immobilized El was card ed out with 5 .ug/mL of Ubel2 N8 (1 19 nM), 5 ⁇ g/mL of ABD GlyRS (289 nM), or the mixture of Ubcl2 N8 and ABD each at 5 _ug mL.
  • the black dotted line indicates the calculated sum of the binding curves for Ubcl2 N8 and ABD to El, separately.
  • Fig, 4E Cullinl competes off GlyRS for Ubcl2 N8 interaction. Binding analysis to immobilized Ubcl 2 NS was carried out with 20 .ug/ ' mL of GlyRS (128 nM), 20 g/mL of cullinl ctd /Rbxl (385 nM), or the mixture of cullinl Ctd /Rbxl and GlyRS each at 20 ⁇ ig/niL. The black dotted line indicates the calculated sum of the binding curves for cullinl Ctd /R.bxl and GlyRS to Ubel2 N8 .
  • FIG. 5A A schematic figure showing how cell cycle kinase inhibitor p27 kip half-life is tightly regulated for proper cell cycle progression. p27 kip undergoes fast turn over through neddylation activated poly-ubiquitination directed degradation and NEDD8 specific inhibitor MLN 924 could disrupt this and result in abnormal accumulation of p27 klp .
  • Fig. SB Knock-down of GARS extends p27 kip half-life. As indicated, HeLa cells were transfected by pLenti vectors containing either a scramble sequence, or GlyRS specific sequences respectively.
  • Fig. 5C FACS analyses of cell cycle confirmed GlyRS involved in cell cycle regulation. Briefly, HeLa cells were transfected with indicated constructs and 24 hours after transfection cells were treated with either 0.2 ⁇ MLN4924 or DMSO for another 24 hours. Cells were then collected, fixed and stained with PI and analyzed by flow cytometry. Cells treated with MLN4924 or GARS knock-down showed significant drop of the 2N peak and sequestered in the 4N population, indicating cell cycle arrest.
  • FIG. 6 shows that GlyRS binds to NEDD8 via its catalytic domain.
  • Hydrogen-deuterium exchange (HDX) analysis shows that NEDD8 bind mainly to the catalytic domain of GlyRS. Changes in deuterium incorporation resulting from the GlyRS- NEDD8 interaction are mapped to the protein sequence and the crystal structure of GlyRS (PDB 2PME).
  • FIG. 7A shows that GlyRS knockdown does not affect Ube2F conjugation.
  • Fig. 7A Ube2F N8 remains unchanged upon GlyRS knockdown.
  • HeLa cells were transfected by pLenti vectors containing either a scramble sequence, SerRS or GlyRS specific sequences respectively. Cells were harvested 48 hours after transfection and lysed with acid lysis buffer and subjected to SDS-PAGE.
  • Fig. 7B Ube2F binds to GlyRS much weaker compared to that of Ubcl2. Bioiayer interferometry analysis confirms that Ube2F does not bind to GlyRS compared to that of UbcI 2. Binding analysis to immobilized GST- GlyRS was carried out with 1.0 ⁇ of either Ubc l2 or Ube2F.
  • FIG. 8A shows that GlyRS catalytic domain mediates interaction with Ubcl2.
  • FIG. 8A Domain mapping by GST pull-down assay suggests that Ubcl2 also binds to the catalytic domain of GlyRS.
  • Fig. SB Hydrogen-deuterium exchange (FIDX) analysis confirms that Ubcl2 binds to the catalytic domain of GlyRS. Changes in deuterium incorporation resulting from the GlyRS-Ubcl2 interaction are mapped to the protein sequence and the crystal structure of GlyRS (PDB 2PME).
  • Fig. 8C GlyRS but not BSA significantly extends the half-life of Ubcl2 N8 i vitro.
  • FIGS 9A-F show that GlyRS binds to APPBP1 and facilitates cullin neddylation.
  • Fig. 9A Domain mapping by GST pull-down suggests that El (APPBP1/UBA3) binds to the anti-codon binding domain (ABD) of GlyRS, as ABD alone can be pulled-down by GST-APPBP1.
  • Fig. 9B Bioiayer interferometry analysis confirms that ABD alone is sufficient for El interaction, as full-length, AWHEP, and ABD GlyRS bind to El (APPBP1/UBA3) with similar affinity.
  • Binding analysis to immobilized El was carried out with a range of concentrations (62.5-500 nM) of full-length, AWHEP, or ABD GlyRS.
  • Fig. 9C Bioiayer interferometry analysis confirms that ABD alone binds to El regulatory subunit APPBPI, as ABD GlyRS binds to APPBPl with similar affinity to that of APPBP1 UBA3.
  • Binding analysis to immobilized APPBP1 was carried out with a range of concentrations (0,58-4,6 ⁇ ) of ABD GlvRS
  • Fig. 9C Bioiayer interferometry analysis confirms that ABD alone binds to El regulatory subunit APPBPI, as ABD GlyRS binds to APPBPl with similar affinity to that of APPBP1 UBA
  • Figure 10 shows a non-limiting schematic illustration showing that GlyRS is associated with cell proliferation via dual cel lular functions.
  • the schematic summarizes the dual function of GlyRS in aminoacylation as an enzyme and in neddylation as a chaperone that supports protein synthesis and cell-cycle progression.
  • FIG 11 shows bioinformatic data demonstrating that high level of GlyRS is associated with rapid breast cancer progression.
  • the expression of all cytoplamic human tRNA synthetases in breast cancer was analyzed by Kaplan-Meier plots and hazard ratio (HR).
  • HR Kaplan-Meier plots and hazard ratio
  • Figere 12 shows bioinformatic data demonstrating that high level of GlyRS is associated with rapid ovarian cancer progression.
  • Figure 13 shows bioinformatic data demonstrating that high level of GlyRS is associated with rapid lung cancer progression.
  • Figures 14A-B show higher level of GlyRS staining in most malignant patient cancer tissue samples.
  • Fig. 14A shows that high level staining of GlyRS is observed in patient tissue samples of breast duct carcinoma, colorectal adenocarcinoma and lung squamous cell carcinoma.
  • Fig, 14B shows that most malignant patient cancer tissue samples have higher level of GlyRS expression compared to normal tissue.
  • FIG 15 is a non-limiting schematic illustration of GlySA binding to GlyRS active site (PDB 2ZT8), GlySA is an analog of Gly-AMP, reaction intermediate of GlyRS.
  • Figure 16 is a plot showing that GlySA (but not MLN4924) inhibits GlyRS aminoacylation.
  • the aminoacylation assay was performed using recombinant human GlyRS (200 nM) proteins at room temperature.
  • MLN4924 is an inhibitor of neddylation currently used in clinical trials for multiple solid and hematopoietic cancers.
  • MLN4924 targets the El enzyme (UBA3) of neddylation.
  • FIG 17 shows experimental data demonstrating that GlySA decreases GlyRS binding to activated EDD8 E2 (Ubcl2 N8 ).
  • the interactions of GlyRS (0.25 ⁇ ) to that of Ubcl2 NS (100 nM; immobilized to the sensor tips) were compared in the presence of DMSO or GlySA at 30°C by biolayer interferometry (Octet).
  • Figure 18 shows experimental data demonstrating that unlike MLN4924, GlySA does not affect NEDD8 El (LIB A3) activation.
  • NEDD8 activation assay was performed with recombinant human APPBP1-UBA3 (2.7 ⁇ ) protein and fluorescein - labeled NEDD8 proteins at 37°C for 1 hour.
  • the concentration of GlySA and MLN4924 was 300 ⁇ .
  • FIG 19 shows experimental data demonstrating that GlySA, but not SerSA, TyrSA, inhibits cullin neddylation in MDA-MB-231 cells.
  • MDA-MB-231 cells at 80% confluence were treated overnight with each compound and then the cells were harvested and lysed with the acid lysis buffer and subjected to non-reducing SDS-PAGE.
  • SerSA and TyrSA are analogs of Ser-AMP and Tyr-AMP, reaction intermediate of SerRS and TyrRS, respectively.
  • MLN4924 was used as a positive control for the experiment.
  • Figure 20 shows experimental data determining iC 50 of GlySA for inhibiting cullin neddylation in MDA-MB-231 cells.
  • MD A-MB-231 cells at 80% confluence were treated overnight with GlySA and then the cells were harvested and lysed with the acid lysis buffer and subjected to non-reducing SDS-PAGE.
  • Figure 21 shows experimental data on time course of GlySA in inhibiting neddylation in MDA-MB-231 cells.
  • MDA-MB-231 ceils at 80% confluence were treated with 200 nM GlySA and then the cells were harvested at different time points and lysed with the acid lysis buffer and subjected to non-reducing SDS-PAGE.
  • Figure 22 shows experimental data on GlySA effect over a range of concentrations on key components and substrates of the neddylation pathway in MDA-MB- 231 cells.
  • MDA-MB-231 cells at 80% confluence were treated overnight with GlySA and then the cells were harvested and lysed with the acid lysis buffer and subjected to non- reducing SDS-PAGE.
  • Figure 23 shows experimental data on GlySA effect over a range of concentrations on key components and substrates of neddylation in MDA-MB-468 cells.
  • MDA-MB-468 ceils at 80% confluence were treated overnight with GlySA and then the ceils were harvested and lysed with the acid lysis buffer and subjected to non-reducing SDS- PAGE.
  • Figure 24 shows experimental data on GlySA effect over a range of concentrations on key components and substrates of neddylation in MCF7 cells.
  • MCF7 cells at 80% confluence were treated overnight with GlySA and then the ceils were harvested and lysed with the acid lysis buffer and subjected to non-reducing SDS-PAGE.
  • FIG. 25 shows a schematic illustration of a non-limiting exemplary maximum tolerant dosage assay of GlySA.
  • GlySA DMSO stock solution diluted by saline
  • mice mice via tail vein injections. Mice after four injections were evaluated and all were alive.
  • FIG. 26 shows a schematic illustration of non-limiting exemplary lung metastasis assay methods.
  • 1 x 10 5 MDA-MB-231 cells were injected via tail vein to NOD.Cg- Prkdc scld I12rg mice. Then mice were separated into 3 groups.
  • group B GlySA (4 mg/kg; 100 ⁇ )
  • group C MLN4924 GlySA (4,4 mg/kg; 100 ⁇ ) were administrated via tail vein injections twice per week.
  • N T0 for each group.
  • Figure 27 shows experimental data demonstrating that GlySA treatment reduces lung metastasis in mice.
  • Top panel mice lungs 14 days after tumor cells (MDA- MB-231) injection.
  • White dots show the surface tumor colonies.
  • Bottom panel numbers of
  • Figure 28 shows a non-limiting schematic illustration of a working model of GlySA on inhibiting both protein synthesis and cell-cycle regulation.
  • Figure 29 shows GlySA and several GlySA derivatives.
  • the top panel shows chemical structures of GlySA and several GlySA derivatives.
  • the bottom panel shows the key interacting residues on GlyRS with GlySA based on a co-crystal structure of GlySA bound GlyRS (PDB: 2ZT8).
  • polypeptide oligopeptide
  • peptide and “protein” are used interchangeably herein to refer to polymers of amino acids of any length, e.g., at least 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 100, 200, 300, 400, 500, 1,000 or more amino acids.
  • the polymer may be linear or branched, it may include, for example, modified amino acids, and it may be interrupted by non-ami no acids.
  • the terms also encompass an amino acid polymer that has been modified naturally or by intervention; for example, disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation or modification, such as conjugation with a labeling component.
  • polypeptides containing one or more analogs of an amino acid including, for example, unnatural amino acids, etc.
  • polynucleotide oligonucleotide
  • nucleic acid nucleic acid molecule
  • polymeric form of nucleotides of any length, e.g., at least 8, 9, 10, 20, 30, 40, 50, 100, 200, 300, 400, 500, 1 ,000 or more nucleotides, and may include ribonucleotides, deoxyribonucleotides, analogs thereof, or mixtures thereof. This term refers only to the primary structure of the molecule.
  • the term includes triple-, double- and single-stranded deoxyribonucleic acid ("DNA”), as well as triple-, double- and single-stranded ribonucleic acid (“RNA”). It also includes modified, for example by alkylation, and/or by capping, and unmodified forms of the polynucleotide.
  • polynucleotide examples include polydeoxyribonucleotides (containing 2-deoxy-D-ribose), polyribonucleotides (containing D-ribose), including tRNA, rRNA, hRNA, and mRNA, whether spliced or unspliced, any other type of polynucleotide which is an N- or C-glycoside of a purine or pyrimidine base, and other polymers containing normucleotidic backbones, for example, polyamide (e.g., peptide nucleic acids (“PNAs”)) and polymorpholino (commercially available from the Anti-Virais, Inc., Corvallis, OR., as Neugene) polymers, and other synthetic sequence-specific nucleic acid polymers providing that the polymers contain nucleobases in a configuration which allows for base
  • PNAs peptide nucleic acids
  • polymorpholino commercially available from the Anti-Virais, Inc.,
  • these terms include, for example, 3'-deoxy-2',5'-DNA, oligodeoxyribonucleotide N3' to P5' phosphoramidates, 2'-0-aikyi-substituted RNA, hybrids between DNA and RNA or between PNAs and DNA or RNA, and also include known types of modifications, for example, labels, alkylation, "caps, ' " substitution of one or more of the nucleotides with an analog, intemucleotide modifications such as, for example, those with uncharged linkages (e.g., methyl phosphonates, phosphotriesters, phosphoramidates, carbamates, etc.), with negatively charged linkages (e.g., phosphorothioates, phosphorodithioates, etc.), and with positively charged linkages (e.g., aminoalkylphosphoramidates, aminoalkylphosphotriesters), those containing pendant moieties, such as, for example, proteins (including enzymes
  • sequence identity or “identity” or “homology” in the context of two protein sequences (or nucleotide sequences) includes reference to the residues in the two sequences which are the same when aligned for maximum correspondence over a specified comparison window.
  • the portion of the amino acid sequence or nucleotide sequence in the comparison window may comprise additions or deletions (i.e., gaps) as compared to the reference sequence for optimal alignment of the two sequences.
  • percentage of sequence identity is used in reference to proteins it is recognized that residue positions which are not identical often differ by conservative amino acid substitutions, where amino acids are substituted for other amino acid residues with similar chemical properties (e.g.
  • sequences differ in conservative substitutions
  • the percentage sequence identity may be adjusted upwards to correct for the conservative nature of the substitutions. Sequences, which differ by such conservative substitutions are said to have "sequence similarity" or “similarity”. Means for making these adjustments are well known to persons skilled in the art. The percentage is calculated by determining the number of positions at which the identical amino acid or nucleic acid base residue 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 and multiplying the result by 100 to yield the percentage of sequence identity.
  • the term "homologue” is used to refer to a nucleic acid which differs from a naturally occurring nucleic acid (i.e., the "prototype” or “wild-type” nucleic acid) by minor modifications to the naturally occurring nucleic acid, but which maintains the basic nucleotide structure of the naturally occurring form. Such changes include, but are not limited to: changes in one or a few nucleotides, including deletions (e.g., a truncated version of the nucleic acid) insertions and/or substitutions.
  • a homologue can have enhanced, decreased, or substantially similar properties as compared to the naturally occurring nucleic acid.
  • a homologue can be complementary or matched to the naturally occurring nucleic acid, Homologues can be produced using techniques known in the art for the production of nucleic acids including, but not limited to, recombinant DNA techniques, chemical synthesis, or any combination thereof.
  • complementary or matched means that two nucleic acid sequences have at least 50% sequence identity.
  • the two nucleic acid sequences can have at least 60%, 70%, 80%, 90%, 95%, 96%, 97%, 98%, 99% or 100% of sequence identity.
  • “Complementary or matched” also means that two nucleic acid sequences can hybridize under low, middle and/or high stringency condition(s).
  • substantially complementary or substantially matched means that two nucleic acid sequences have at least 90% sequence identity.
  • the two nucleic acid sequences can have at least 95%, 96%, 97%, 98%, 99% or 100% of sequence identity.
  • substantially complementary or substantially matched means that two nucleic acid sequences can hybridize under high stringency condition(s).
  • a "subject” refers to an animal that is the object of treatment, observation or experiment.
  • Animal includes cold- and warm-blooded vertebrates and invertebrates such as fish, shellfish, reptiles and, in particular, mammals.
  • mice includes, without limitation, mice, rats; rabbits; guinea pigs; dogs, cats; sheep; goats; cows; horses; primates, such as monkeys, chimpanzees, and apes, and, in particular, humans.
  • a "patient” refers to a subject that is being treated by a medical professional, such as a Medical Doctor (i.e. Doctor of Allopathic medicine or Doctor of Osteopathic medicine) or a Doctor of Veterinary Medicine, to attempt to cure, or at least ameliorate the effects of, a particular disease or disorder or to prevent the disease or disorder from occurring in the first place.
  • a medical professional such as a Medical Doctor (i.e. Doctor of Allopathic medicine or Doctor of Osteopathic medicine) or a Doctor of Veterinary Medicine, to attempt to cure, or at least ameliorate the effects of, a particular disease or disorder or to prevent the disease or disorder from occurring in the first place.
  • administering refers to a method of giving a dosage of a pharmaceutically active ingredient to a vertebrate.
  • a "dosage” refers to an amount of therapeutic agent administered to a patient.
  • a "daily dosage” refers to the total amount of therapeutic agent administered to a patient in a day.
  • terapéutica agent means a substance that is effective in the treatment of a disease or condition.
  • terapéuticaally effective amount or “pharmaceutically effective amount” is meant an amount of therapeutic agent, which has a therapeutic effect.
  • the dosages of a pharmaceutically active ingredient which are useful in treatment are therapeutically effective amounts.
  • a therapeutically effective amount refers to an amount of therapeutic agent which produces the desired therapeutic effect as judged by clinical trial results and/or model animal studies.
  • a "therapeutic effect” relieves, to some extent, one or more of the symptoms of a disease or disorder.
  • a therapeutic effect may be observed by a reduction of the subjective discomfort that is communicated by a subject (e.g., reduced discomfort noted in self-administered patient questionnaire).
  • treatment refers to a clinical intervention made in response to a disease, disorder or physiological condition manifested by a patient, particularly a patient suffering from cancer.
  • the aim of treatment may include, but is not limited to, one or more of the alleviation or prevention of symptoms, slowing or stopping the progression or worsening of a disease, disorder, or condition and the remission of the disease, disorder or condition.
  • treatment refers to both therapeutic treatment and prophylactic or preventative measures.
  • Those in need of treatment include those already- affected by a disease or disorder or undesired physiological condition as well as those in which the disease or disorder or undesired physiological condition is to be prevented.
  • prevention refers to any activity that reduces the burden of the individual later expressing the symptoms. This takes place at primary, secondary and tertiary prevention levels, wherein: a) primary prevention avoids the development of symptoms/disorder/condition; b) secondary prevention activities are aimed at early stages of the condition/disorder/symptom treatment, thereby increasing opportunities for interventions to prevent progression of the condition/disorder/symptom and emergence of symptoms; and c) tertiary prevention reduces the negative impact of an already established condition/disorder/symptom by, for example, restoring function and/or reducing any condition/disorder/symptom or related complications.
  • a therapeutic agent or a protective agent may comprise a "drug,"
  • a “drug” refers to a therapeutic agent or a diagnostic agent and includes any substance, other than food, used in the prevention, diagnosis, alleviation, treatment, or cure of a disease.
  • the drug can include any substance disclosed in at least one of: The Merck Index, 12th Edition (1996); Pei-Show Juo, Concise Dictionary of Biomedicine and Molecular Biology, (1996); U.S. Pharmacopeia Dictionary, 2000 Edition, and Physician's Desk Reference, 2001 Edition.
  • the therapeutic agent is one of the embodiments of the compositions described herein.
  • the drug used in the therapeutic system will often be placed on, embedded, encapsulated or otherwise incorporated into a deliver ⁇ ' matrix.
  • the delivery matrix may be included in or on either the first skeletal structure or the second cushioning structure, or both.
  • the delivery matrix in turn, comprises either a biodegradable or a non-biodegradable material.
  • the delivery matrix may include, although it is not limited to, a polymer.
  • biodegradable polymers include protein, hydrogel, polyglycolic acid (PGA), polyiactic acid (PLA), poly(L-lactic acid) (PLLA), poiy(L-giycolic acid) (PLGA), polyglycolide, poly-L-lactide, poly-D-lactide, poly(amino acids), polydioxanone, polycaprolactone, polygluconate, polylactic acid-polyethylene oxide copolymers, modified cellulose, collagen, polyorthoesters, polyhydroxybutyrate, polyanhydride, polyphosphoester, poly(alpha-hydroxy acid), and combinations thereof.
  • Non-biodegradable polymers may comprise silicone, acrylates, polyethylenes, polyurethane, polyurethane, hydrogel, polyester (e.g., DACRON® from E. I. Du Pont de Nemours and Company, Wilmington, Del.), polypropylene, polytetrafluoroethylene (PTFE), expanded PTFE (ePTFE), polyether ether ketone (PEEK), nylon, extruded collagen, polymer foam, silicone rubber, polyethylene terephthalate, ultra-high molecular weight polyethylene, polycarbonate urethane, polyurethane, polyimides, stainless steel, nickel-titanium alloy (e.g., Nitinol), titanium, stainless steel, cobalt-chrome alloy (e.g., ELGILOY® from Elgin Specialty Metals, Elgin, 111.; CONICHROME® from Carpenter Metals Corp., Wyomissing, Pa.).
  • the hydrogel may comprise poly(alkyleneoxides), such
  • range format is merely for convenience and brevity and should not be constmed as an inflexible limitation on the scope of the disclosure. Accordingly, the description of a range should be considered to have specifically disclosed all the possible sub-ranges as well as individual numerical values within that range. For example, description of a range such as from 1 to 6 should be considered to have specifically disclosed sub-ranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual numbers within that range, for example, 1, 2, 3, 4, 5, and 6. This applies regardless of the breadth of the range.
  • Glycyl-tRNA synthetase (GlyRS; also known as glycine-tRNA ligase) is an enzyme that belongs to the aminoacyi tRNA synthetase (aaRS) family.
  • aaRS is an enzyme that attaches the appropriate amino acid onto its tRNA. It does so by catalyzing the esterification of a specific cognate amino acid or its precursor to one of ail its compatible cognate tRNAs to form an aminoacyi -tRNA.
  • GlyRS is an enzyme that catalyzes the chemical reaction:
  • the three substrates of the GlyRS enzyme are ATP, glycine, and tRNA(Gly), whereas the three products are AMP, diphosphate and glycyi-tRNA(Gly).
  • Human GlyRS is encoded by the GARS gene, and is composed of three distinct domains: the N-terminal metazoan-specific WHEP domain, catalytic domain, and the C-terminal anticodon-binding domain (ABD).
  • a GlyRS in some embodiments, can function in aminoacylation as an enzyme and in neddylation (for example as a chaperon that supports protein synthesis and cell-cycle progress).
  • the GlyRS proteins disclosed herein are capable of interaction with one or more components of the neddylation pathway, including NEDD8, El and E2.
  • the GlyRS proteins may capable of binding to the APPBPl subunit of El and activated E2 (NEDD8-conjugated Ubcl2).
  • the GlyRS proteins are capable of increasing the level of neddylation in a ceil, for example, neddylation of El, E2, and neddylation substrates
  • Neddylation substrates include, but are not limited to, members of the cullin protein family, e.g., culiin 1, cullin 2, cullin 3, culiin 4A, cuilin 4B, cullin 5, cullin 7, and cullin 9.
  • the substrates are human cullin proteins.
  • the GlyRS proteins are capable of increasing the level of cullin-RTNG ubiquitin ligases (CRLs) in a cell.
  • CTLs cullin-RTNG ubiquitin ligases
  • the GlyRS proteins disclosed herein may increase the level of neddyiation through interacting with NEDDS-conjugation Ubcl 2 and protecting it from degradation.
  • the GlyRS proteins do not interact with other ubiquitin or ubiquitin-like proteins such as SUMOl .
  • the coding sequence of a human GARB gene is shown below (SEQ ID NO: 1). Also contemplated herein are GlyRS nucleotide sequences that have at least 70%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or more sequence identity to SEQ ID NO: 1.
  • GlyRS proteins having sequences that have at least
  • expression vectors capable of expressing eukaryotic proteins may be used to express the recombinant GlyRS proteins.
  • the host cells can be bacterial, fungal, plant, yeast, insect or mammalian cells.
  • the term host cell includes both the cells, progeny of the ceils and protoplasts created from the cells that are used to produce a GlyRS according to the disclosure.
  • the host cells are prokaryotic ceils, for example bacteria host ceils.
  • a DNA construct comprising nucleic acid encoding the amino acid sequence of the designated GlyRS can be constmcted and transferred into, for example, an E. coli host cell.
  • the vector may be any vector which when introduced into an E. coli host ceil can be integrated into the host cell genome and can be replicated.
  • the nucleic acid encoding the GlyRS can be operably linked to a suitable promoter, which shows transcriptional activity in E. coli host ceil.
  • the promoter may be derived from genes encoding proteins either homologous or heterologous to the host cell.
  • an "inducible promoter" may refer to a promoter that is active under environmental or d evel opmental regul ati on .
  • the GlyRS coding sequence can be operably linked to a signal sequence.
  • the expression vector may also include a termination sequence.
  • the termination sequence and the promoter sequence can be derived from the same source.
  • the termination sequence can be homologous to the host cell ,
  • the expression vector may include one or more selectable markers.
  • selectable markers include ones that confer antimicrobial resistance (e.g., hygromycin and phleomycin).
  • nutritional selective markers including those markers known in the art as amdS, argB, and pyr4, can be used as the selectable marker.
  • An expression vector comprising a DNA construct with a polynucleotide encoding the GlyRS may be any vector which is capable of replicating autonomously in a given host organism or of integrating into the DNA of the host.
  • the expression vector can be a plasmid or a viral construct.
  • the first expression vector may comprise DNA sequences in which the promoter, GlyRS-coding region, and terminator all originate from the gene to be expressed.
  • gene truncation can be obtained by deleting undesired DNA sequences (e.g., DNA encoding unwanted domains) to leave the domain to be expressed under control of its own transcriptional and translational regulatory sequences.
  • the second type of expression vector may be preassembled and contains sequences needed for high-level transcription and a selectable marker.
  • the coding region for the GARS gene or part thereof can be inserted into this general-purpose expression vector such that it is under the transcriptional control of the expression construct promoter and terminator sequences.
  • genes or part thereof may be inserted downstream of a strong promoter.
  • Methods used to ligate the DNA construct comprising a polynucleotide encoding the GlyRS, a promoter, a terminator and other sequences and to insert them into a suitable vector are well known in the art. Linking can be generally accomplished by ligation at convenient restriction sites. If such sites do not exist, the synthetic oligonucleotide linkers are used in accordance with conventional practice (Bennett & Lasure, More Gene Manipulations In Fungi, Academic Press, San Diego (1991) pp 70-76), Additionally, vectors can be constructed using known recombination techniques (e.g., Invitrogen Life Technologies, Gateway Technology).
  • Introduction of a DNA construct or vector into a host cell includes techniques such as transformation; electroporation; nuclear microinjection; transduction; transfection, (e.g., lipofection mediated and DEAE-Dextrin mediated transfection); incubation with calcium phosphate DNA precipitate; high velocity bombardment with DNA- coated microprojectiles; and protoplast fusion.
  • General transformation techniques are known in the art (see, e.g., Campbell et al., (1989) Curr. Genet 1.6:53-56).
  • genetically stable transformants can be constructed with vector systems whereby the nucleic acid encoding GlyRS is stably integrated into a host strain chromosome. Transformants can then be purified by known techniques. Methods of Inhibiting Neddylation
  • NEDD8 is an 81-amino acid protein with 9 kDa relative molecular mass and is 60% identical and 80% homologous to ubiquitin.
  • NEDD8 has a dedicated El- activating enzyme (AppBpl/UBA3, or NAE) and E2-conjugating enzymes (UBC12, UBE2F) and is essential for the enzymatic activity of the CRL family of E3 ligases, through conjugation to the cullin scaffold.
  • Other components of the neddvlation pathway include DEN1 which processes NEDD8 to its mature, 76-amino acid form, and the COP9 signalosome complex, which is responsible for removing NEDD8 from cullin proteins.
  • CANDl cullin-associated and neddylation-dissociated
  • CANDl is an additional component that regulates CRL complex assembly by binding to the cullin in the absence of NEDD8 activation.
  • Neddylation is a posttranslational modification that controls cell cycle and proliferation by conjugating the ubiquitin-like protein NEDD8 to specific targets. It is hereby disclosed that GlyRS plays a critical role in neddylation. In human ceils, knockdown of GlyRS, but not a different tRNA synthetase, decreases the global level of neddylation and delays cell cycle progression. This function of GlyRS is achieved through direct interactions with multiple components of the neddylation pathway, including NEDD8, El , and E2. GlyRS can bind to the APPBP1 subunit of El to capture and protect the activated E2 (NEDD8 ⁇ conjugated Ubcl2) before it reaches a downstream target.
  • Some embodiments disclosed herein provide methods of reducing or inhibiting neddylation in a cell.
  • inhibition of neddylation includes partially or fully blocks or abolished neddylation in a cell or a cell population.
  • the inhibition can reduce 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 60%, 70%, 80%, 90%, 95%, 99%o, or a range between any two of these values, of neddylation in the cell or the cell population.
  • the inhibition can reduce about 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, or a range between any two of these values, of neddylation in the cell or the ceil population.
  • the neddylation in the cell or the cell population is completely abolished.
  • the methods comprise contacting the cell with a GlyRS inhibitor, wherein the level of neddylation is decreased in the cell.
  • the level of neddylation can be decreased to, or to about 99%, 95%, 90%, 80%, 70%, 60%, 50%, 45%, 40%, 35%, 30%, 25%, 20%, 15%, 10%, 5%, 1%, or a range between any two of these values, of the level of neddylation in cell(s) not treated with the GlyRS inhibitor.
  • Neddylation in some embodiments, refers to the conjugation of NEDD 8 to components of the neddylation pathway, e.g., the El enzyme, the E2 enzyme, or the E3 ligases.
  • neddylation may refer to the conjugation of NEDD 8 to a cullin protein in the E3 ligases, e.g., a CRL.
  • the decrease of neddylation may occur to individual components of the neddylation pathway, or at a global level.
  • the level of neddylation may be reduced by 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98%, 99%, or more.
  • the GlyRS inhibitor may decrease the level of neddylation in the cell through a variety of mechanisms, for example, by interfering the binding between the GlyRS protein and NEDD 8, by interfering the binding between the GlyRS protein and an E2 enzyme, such as Ubcl2, by interfering the binding between the GlyRS protein and NEDD8-conjugated Ubc 2, by interfering the binding between the GlyRS protein and an El enzyme, such as APPBP1/UBA3 heterodimer, by directly targeting El enzyme (given the similarity in the first step reaction between GlyRS and El ), or any combination thereof.
  • E2 enzyme such as Ubcl2
  • El enzyme such as APPBP1/UBA3 heterodimer
  • the GlyRS inhibitor may inhibit the activity or decrease the level of a component of the neddylation pathway, for example, the level of NEDD 8 -conjugated Ubcl2, the activity of the Ubcl2 enzyme, the protein level of the Ubcl2 enzyme, or any combination thereof.
  • the GlyRS inhibitor binds to both GlyRS and El enzyme to decrease neddylation.
  • inhibiting neddylation by the methods disclosed herein may result in the inhibition of ubiquitination of one or more of the E3 substrates in a cell, such as ATF4, CCNEl, CDC25, CDKN1A, CDKN1B, CTNNB 1 , DAPK1 , Emil, FancM, HIF2A, IRS1 , JUN, MCL1 , NRF2, ORC1 L, PDCD4, POLR2A, SETD8, SNAI3, USP18, etc.
  • a cell such as ATF4, CCNEl, CDC25, CDKN1A, CDKN1B, CTNNB 1 , DAPK1 , Emil, FancM, HIF2A, IRS1 , JUN, MCL1 , NRF2, ORC1 L, PDCD4, POLR2A, SETD8, SNAI3, USP18, etc.
  • GlyRS inhibitor is used herein in a broad sense and includes any molecule that partially or fully blocks, inhibits or neutralizes a biological activity mediated by GlyRS. In some embodiments, it can prevent the activation of GlyRS.
  • GlyRS inhibitor also includes any molecule that abolishes or reduces the function or expression of GlyRS.
  • the GlyRS inhibitor can act directly on GlyRS, for example by binding to GlyRS, to prevent or reduce activation of GlyRS.
  • the GlyRS inhibitor can interfere, preferably abolish or reduce, GlyRS from interacting with a binding partner or a substrate, such a component of the neddylation pathway .
  • the GlyRS inhibitor can modulate the level of GlyRS gene expression, for example, inhibiting or reducing the transcription of GlyRS gene.
  • the GlyRS inhibitor can modulate the levels of GlyRS protein in cells by, for example, inhibiting or reducing the translation of GlyRS mRNA, or increasing the degradation of GlyRS mRNA or GiyRS protein.
  • the GlyRS inhibitor can block the interaction of GlyRS with EDD8 and/or NEDD8-conjugated Ubcl2.
  • a GlyRS can perform function in various biological processes, for example aminoacyiation and neddylation.
  • a compound is considered to be a GlyRS inhibitor if the compound can reduce or inhibit one or more biological activities of a GlyRS.
  • a GiyRS inhibitor may reduce or inhibit GlyRS functions in both aminoacyiation and neddylation.
  • the GlyRS inhibitor only reduce or inhibit GlyRS function in neddylation.
  • the GlyRS inhibitor does not significantly inhibit GlyRS function in aminoacyiation.
  • a GiyRS inhibitor does not significantly inhibit GlyRS function in aminoacyiation if the GlyRS inhibitor can at most reduce the activity of the GlyRS function in aminoacyiation by, or by about, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 10%, 50%, 1%, or a range between any two of these values.
  • the GlyRS inhibitor reduces the activity of the GlyRS function in aminoacyiation by, or by about, 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 10%, 50%, 1%, or a range between any two of these values.
  • GlyRS inhibitors include, for example, small molecules, nucleic acids, antibodies, peptides, or any combination thereof.
  • the GlyRS inhibitor can be a small molecule that binds to GiyRS.
  • the GlyRS inhibitor can be a molecule that blocks interaction of GlyRS and it binding partner.
  • the GlyRS inhibitor is a nucleic acid, for example, an anti-GlyRS small-hairpin RNA (shRNA) or an GlyRS anti-sense RNA.
  • Some embodiments of the present disclosure therefore include, for example, inhibitors of GlyRS function, for example, its interactions with components of the neddvlation pathway.
  • the GlyRS inhibitors can be used, for example, in any of the methods described herein. Any agent that may prevent or reduce the interaction between the GlyRS protein and NEDD8, El , and E2, or eliminate or reduce the level of GlyRS protein expression, is contemplated by the present disclosure.
  • a reduction refers to at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99% or more of the interaction between the GlyRS protein and NEDD8, El, and E2, or of the GlyRS protein expression level in a cell.
  • Interaction refers the binding of NEDD8, El, and E2 with the GlyRS protein, which may lead to a conformational change to the GlyRS protein and/or the NEDD8, El, and E2 protein.
  • GlyRS inhibitors can be identified using biolayer interferometry. Briefly, in biolayer interferometry, a molecule can be examined for the ability to affect the binding between a GlyRS protein and its binding partner. For example, GlyRS proteins are immobilized on either anti-GST or Ni-NTA sensor tips in 96-weil plates. A binding partner, e.g., NEDD8, is added to the buffer. Candidate GlyRS molecules are added to the buffer and the dissociation constants are measured. GlyRS inhibitors that interfere with the binding between the GlyRS protein and NEDD8 can be identified based on the change in the dissociation constants One of skill in the art will be able to select the appropriate assays and reaction conditions based on the particular circumstances.
  • Some embodiments disclosed herein provide small molecule compounds that inhibit the function of a GlyRS protein.
  • Non-limiting examples of inhibitory small molecule compounds include ones that exhibit binding specificity for at least one region of the GlyRS protein that is involved in its interaction with a component of the neddvlation pathway, and/or its stability/degradation in a cell.
  • small molecule refers to an organic compound that is of synthetic or biological origin (biomolecule), but is typically not a polymer.
  • the small molecule compounds disclosed herein may bind to a region of the GlyRS protein involved in its interaction with a component of the neddylation pathway, e.g., the catalytic domain, the ABD domain, or a combination thereof.
  • the small molecule compounds may interfere with the interaction between GlyRS and NEDD8, El, and/or E2.
  • the small molecule compounds bind to a sequence comprising amino acids 84-93 of SEQ ID NO: 2.
  • the small molecule compounds bind to one or more of amino acids 84-93 of SEQ ID NO: 2. In some embodiments, the small molecule compounds bind to a sequence comprising amino acids 232-238 of SEQ ID NO: 2. In some embodiments, the small molecule compounds bind to one or more of amino acids 232-238 of SEQ ID NO: 2.
  • GlyRS inhibitor is glycylsulfamoyladenosin ly-SA.
  • the structure of this compound is as follows.
  • Gly-SA is an analogue of the Gly-AMP reaction intermediate and inhibits GlyRS catalytic activity. Estimates of the potency of inhibition are obtained by performing enzyme assays in the presence of a range of inhibitor concentrations, and fitting the effect of inhibitor concentration on enzyme velocity to a four parameter logistic function that allows calculation of an IC 50 (the inhibitor concentration at which GlyRS activity is reduced by half). This parameter is directly related to the dissociation constant for inhibitor binding (Kj or K d ) and has a value of around 2.4 mM for Gly-SA when tested against the S. aureus GlyRS. Binding of Gly-SA to GlyRS can also be measured directly using stopped-fiow fluorescence techniques because enzyme:inhibitor binary complex has around 5% higher tryptophan fluorescence than the free enzyme.
  • Gly-SA derivatives that function as GlyRS inhibitors.
  • Gly-SA derivatives include Compound-l, Compound-2, and Compound-3 (shown below and in Figure 29) and the analogues described for aaSA in Van de Vijver et al. (2008) J. Med. Chem. 51 :3020-3029 (the content of which is incorporated by reference herein in its entirety).
  • chemical modifications for various aaRS inhibitors having similar chemical structure with Gly-SA have been described in, for example, Brown et al. (2000) Biochemistry 39(20):6003-6011, Lee et al.
  • dsRNA Double- stranded RNA
  • RNAi RNA interference
  • the process is known to occur in a wide variety of organisms, including embryos of mammals and other vertebrates.
  • the use of these dsRNAs (or recombinant! y produced or chemically synthesized oligonucleotides of the same or similar nature) enables the targeting of the GlyRS mR As, for example, GARS in humans, for degradation in mammalian cells.
  • RNA interfering RNA small interfering RNA sequences, RNA interfering vectors, and RNA interfering lentiviruses that are directed at a GlyRS gene, e.g., the human GARS gene.
  • dsRNA double-stranded ribonucleic acid molecules that inhibit expression of a GlyRS protein, wherein a first strand of the dsRNA is substantially identical to at least 19 consecutive nucleotides of the GlyRS gene, and a second strand of the dsRNA is substantially complementary to the first strand.
  • the dsRNA molecules are small hairpin RNA (shRNA) molecules.
  • shRNA small hairpin RNA
  • shGARS2277 5'-GCATGGAGTATCTCACAAAGT (SEQ ID NO:3)
  • shGARS-696 5' GCCCAAAGATGATATTGTAGA (SEQ ID NO: 4)
  • shGARS-1324 5' GCTGCCCAGATTGGAAATTCT (SEQ ID NO:5)
  • the shRNA may comprise a sense fragment, which comprises a nucleotide sequence substantially identical to a target sequence in the GlyRS gene, and an antisense fragment, wherein the sense and antisense fragments are separated by a loop fragment, wherein the loop fragments may comprise a sequence selected from the group consisting of UUCAAGAGA, AUG, CCC, UUCG, CCACC, CTCGAG, AAGCUU and CCACACC.
  • an siRNA target sequence may be designed on the basis of the human GARS gene, preferably 15 to 27, more preferably 19 to 23, and optimally 19, 20 or 21, consecutive bases.
  • siRNA molecules that mediate RNAi.
  • the siRNA molecules disclosed herein can also comprise a 3' hydroxyl group.
  • the siRNA molecules can be single-stranded or double stranded, such molecules can be blunt ended or comprise overhanging ends (e.g., 5', 3').
  • the siRNA molecule is double stranded and either blunt ended or comprises overhanging ends.
  • at least one strand of the siRNA molecule has a 3 ! overhang from about 1 to about 6 nucleotides (e.g., pyrimidine nucleotides, purine nucleotides) in length.
  • the siRNA molecule is double stranded, one strand has a 3 ! overhang and the other strand can be blunt-ended or have an overhang.
  • the length of the overhangs may be the same or different for each strand.
  • the siRNA of the present invention comprises 21 nucleotide strands which are paired and which have overhangs of from about 1 to about 3, particularly about 2, nucleotides on both 3' ends of the siRNA.
  • the 3' overhangs can be stabilized against degradation.
  • the RNA is stabilized by including purine nucleotides, such as adenosine or guanosine nucleotides.
  • purine nucleotides such as adenosine or guanosine nucleotides.
  • substitution of pyrimidine nucleotides by modified analogues e.g., substitution of uridine 2 nucleotide 3' overhangs by 2'-deoxythymidine is tolerated and does not affect the efficiency of RNAi.
  • the absence of a 2' hydroxyl significantly enhances the nuclease resistance of the overhang in tissue culture medium.
  • siRNA molecules disclosed herein can be obtained using a number of techniques known to those of skill in the art.
  • the siRNA can be chemically synthesized or recombinant! y produced using methods known in the art.
  • the siRNA can also be obtained using an in vitro system.
  • the in vitro system can also be used to obtain siRNA of about 19 to about 23 nucleotides in length which mediates RNA interference of the mRNA of the GlyRS gene.
  • the method of obtaining the siRNA sequence using the in vitro system can further comprise isolating the RNA sequence from the combination.
  • the siRNA molecules can be isolated using a number of techniques known to those of skill in the art. For example, gel electrophoresis can be used to separate siRNA from the combination, gel slices comprising the RNA sequences removed and RNAs eluted from the gel slices. Alternatively, non-denaturing methods, such as non-denaturing column chromatography, can be used to isolate the RNA produced. In addition, chromatography (e.g., size exclusion chromatography), glycerol gradient centrifugation, affinity purification with antibody can be used to isolate siRNAs.
  • the RNA-protein complex isolated from the in vitro system can also be used directly in the methods described herein (e.g., method of mediating RNAi of mRNA of the GiyRS gene).
  • the siRNAs described herein can be used in a variety of ways.
  • the siRNA molecules can be used to mediate RNA interference of mRNA of a gene in a cell or organism.
  • the siRNA is introduced into human cells or a human in order to mediate RNA interference in the cells or in cells in the individual, such as to prevent or treat a disease or undesirable condition, hi this method, a gene (or genes) that cause or contribute to the disease or undesirable condition is targeted and the corresponding mRNA (the transcriptional product of the targeted gene) is degraded by RNAi.
  • an siRNA that targets the corresponding mRNA (the mRNA of the targeted gene) for degradation is introduced into the cell or organism.
  • the ceil or organism is maintained under conditions under which degradation of the corresponding mRN A occurs, thereby mediating RNA interference of the mRNA of the gene in the cell or organism.
  • the factors needed to mediate RNAi are introduced into such a cell or the expression of the needed factors is induced in such a cell.
  • siRNA produced by other methods e.g., chemical synthesis, recombinant DNA production
  • Such siRNAs can be altered by addition, deletion, substitution or modification of one or more nucleotides and/or can comprise non-nucleotide materials.
  • a nucleic acid construct that expresses the above- described siRNA may be constructed by means of gene cloning and packaged with a lentivirus that expresses the above-described siRNA, Cell experiments prove that the above- described siRNA sequence can specifically silence the expression of endogenous GlyRS genes in cells.
  • a DNA sequence encoding the above-described siRNA may be contained in a lentivirus vector.
  • the lentivirus vector may further comprise a promoter sequence.
  • the lentivirus vector may further comprise a nucleotide sequence encoding a detectable marker in the ceil, wherein the detectable marker may be a green fluorescent protein (GFP).
  • the lentivirus vector may be selected from the group consisting of pLentiLox 3,7, pLKO. l -puro, pL O. l-CMV-tGFP, pL O. l-puro-CMV-tGFP, pLKO.
  • the siRNA lentiviruses designed for GlyRS stably and specifically lower GlyRS expression and effectively inhibit neddylation.
  • the method comprises: providing one or more compounds; testing the one or more compounds for their ability to reduce or inhibit neddylation.
  • the compounds can be tested for their ability to reduce the binding between a GlyRS protein and NEDD8-conjugated Ubcl2.
  • the methods comprise identifying one or more compounds that have the ability to reduce or inhibit the binding between the GlyRS protein and NEDD8-conjugated Ubcl 2 as inhibitors of neddylation.
  • the method can further comprise testing the one or more compounds for their ability to reduce or inhibit aminoacylation activity of the GlyRS protein.
  • the potential inhibitory or binding effect of a chemical compound on GlyRS may be analyzed prior to its actual synthesis and testing by the use of computer modelling techniques. If the theoretical structure of the given compound suggests insufficient interaction and association between it and GlyRS, synthesis and testing of the compound is obviated. However, if computer modelling indicates a strong interaction, the molecule may then be synthesized and tested for its ability to bind to GlyRS and inhibit using a suitable assay. In this manner, synthesis of inoperative compounds may be avoided.
  • An inhibitory or other binding compound of GlyRS may be computationally evaluated and designed by means of a series of steps in which chemical entities or fragments are screened and selected for their ability to associate with the individual binding pockets or other areas of GlyRS.
  • small molecule inhibitors of neddyiation may be identified using standard techniques.
  • candidate compounds may be used in binding assays using conventional formats to screen inhibitors of interaction between GlyRS and a component of the neddyiation pathway.
  • One particularly suitable assay format includes the enzyme-linked immunosorbent assay (ELISA).
  • ELISA enzyme-linked immunosorbent assay
  • Other assay formats may be used; these assay formats are not a limitation on the present disclosure.
  • the structure of the GlyRS protein permits the design and identification of synthetic compounds and/or other molecules which are characterized by the conformation of the GlyRS protein.
  • the coordinates of the GlyRS protein staicture may be provided in machine readable form, the test compounds designed and/or screened and their conformations superimposed on the structure of the GlyRS protein.
  • suitable candidates identified as above may be screened for the desired GlyRS protein inhibitors bioactivity, stability, and the like. Once identified and screened for biological activity, these inhibitors may be used therapeutically or prophylacticaily to block GlyRS protein activity, and thus, neddyiation in a cell.
  • the identified inhibitor of neddyiation binds to the catalytic domain of the GlyRS protein.
  • the GlyRS protein can be, for example, a mammalian GlyRS protein, particularly a human GlyRS protein.
  • the human GlyRS protein comprises an amino acid sequence having at least 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 2.
  • the compound that inhibits binding between the GlyRS protein and NEDD8-conjugated Ubcl2 binds to a sequence comprising amino acids 84-93 of SEQ ID NO: 2. In some embodiments, the compound that inhibits binding between the GlyRS protein and NEDD8-conjugated Ubcl 2 binds to one or more of amino acids 84-93 of SEQ ID NO: 2. In some embodiments, the compound that inhibits binding between the GlyRS protein and NEDD8-conjugated Ubcl2 binds to a sequence comprising amino acids 232-238 of SEQ ID NO: 2. In some embodiments, the compound that inhibits binding between the GlyRS protein and NEDD 8 -conjugated Ubc-12 binds to one or more of amino acids 232-238 of SEQ ID NO: 2,
  • GlyRS inhibitors can be any of the GlyRS inhibitors disclosed herein.
  • the GlyRS inhibitors may be small molecules, nucleic acids, antibodies, peptides, or any combination thereof.
  • the GlyRS inhibitors may be small molecule compounds that inhibit or reduce the interaction between a GlyRS protein and components of the neddvlation pathway, or isolated double- stranded dsRNA molecules that inhibit or reduce expression of a GlyRS protein.
  • the GlyRS inhibitor reduces or inhibits GlyRS functions in aminoacylation and neddvlation. In some embodiments, the GlyRS inhibitor does not significantly reduce or inhibit GlyRS function in aminoacylation. In some embodiments, the GlyRS inhibitor only reduces or inhibits GlyRS function in neddvlation.
  • the pharmaceutical compositions disclosed herein can comprise one or more therapeutic agents.
  • therapeutic agents include chemotherapeutic agents, cancer drugs, or prodrugs or pharmaceutically acceptable salts thereof.
  • the chemotherapeutic agents can be, for example, AZ-23, BMS-754807, bosutinib, cabozantinib, ceritinib, crizotinib, entrectinib, foretmib, GNF 5837, GW441756, imatinib mesylate, K252a, l .OXO- 10 i , MGCD516, nilotinib hydrochloride monohydrate, NVP-TAE684, PF-06463922, rebastinib, staurosporine, sorafenib tosylate, sunitinib malate, and TSR-01 1.
  • prodrugs of the pharmaceutical compositions and treatment methods employing such pharmaceutically acceptable prodrugs.
  • prodrug means a precursor of a designated compound that, following administration to a subject, yields the compound in vivo via a chemical or physiological process such as solvolysis or enzymatic cleavage, or under physiological conditions (e.g., a prodaig on being brought to physiological pH is converted to the agent).
  • a "pharmaceutically acceptable prodrug” is a prodrug that is non-toxic, biologically tolerable, and otherwise biologically suitable for administration to the subject. Illustrative procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in Bundgaard, Design of Prodrugs (Elsevier Press, 1985).
  • a "pharmaceutically active metabolite” means a pharmacologically active product of metabolism in the body of a compound or salt thereof.
  • Prodaigs and active metabolites of a compound may be determined using routine techniques known or available in the art. See, e.g., Bertolini et al., J. Med. Chem. 1997, 40, 2011-2016; Shan et al., J. Pharm. Sci. 1997, 86 (7), 765-767; Bagshawe, Drug Dev. Res. 1995, 34, 220-230; Bodor, Adv. Drug Res.
  • Any suitable formulation of the compounds described herein can be prepared. See, generally, Remington's Pharmaceutical Sciences, (2000) Hoover, J. E. editor, 20th edition, Lippincott Williams and Wilkins Publishing Company, Easton, Pa., pages 780- 857.
  • a formulation is selected to be suitable for an appropriate route of administration. Some routes of administration are oral, parenteral, by inhalation, topical, rectal, nasal, buccal, vaginal, via an implanted reservoir, or other drug administration methods. In cases where compounds are sufficiently basic or acidic to form stable nontoxic acid or base salts, administration of the compounds as salts may be appropriate.
  • Examples of pharmaceutically acceptable salts are organic acid addition salts formed with acids that form a physiological acceptable anion, for example, tosylate, methanesulfonate, acetate, citrate, malonate, tartarate, succinate, benzoate, ascorbate, a-ketoglutarate, and ⁇ -glycerophosphate.
  • Suitable inorganic salts may also be formed, including hydrochloride, sulfate, nitrate, bicarbonate, and carbonate salts.
  • Pharmaceutically acceptable salts are obtained using standard procedures well known in the art, for example, by a sufficiently basic compound such as an amine with a suitable acid, affording a physiologically acceptable anion.
  • Alkali metal e.g., sodium, potassium or lithium
  • alkaline earth metal e.g., calcium
  • contemplated compounds are administered in a pharmacological composition
  • the compounds can be formulated in admixture with a pharmaceutically acceptable excipient and/or carrier.
  • contemplated compounds can be administered orally as neutral compounds or as pharmaceutically acceptable salts, or intravenously in a physiological saline solution.
  • Conventional buffers such as phosphates, bicarbonates or citrates can be used for this purpose.
  • one of ordinary skill in the art may modify the formulations within the teachings of the specification to provide numerous formulations for a particular route of administration.
  • contemplated compounds may be modified to render them more soluble in water or other vehicle, which for example, may be easily accomplished with minor modifications (salt formulation, esterification, etc.) that are well within the ordinary skill in the art. It is also well within the ordinary skill of the art to modify the route of administration and dosage regimen of a particular compound in order to manage the pharmacokinetics of the present compounds for maximum beneficial effect in a patient.
  • compositions as described herein are generally soluble in organic solvents such as chloroform, dichloromethane, ethyl acetate, ethanol, methanol, isopropanol, acetonitrile, glycerol, N,N-dimethylformamide, AvA r -dimetheylaceatmide, dimethylsulfoxide, or any combination thereof.
  • organic solvents such as chloroform, dichloromethane, ethyl acetate, ethanol, methanol, isopropanol, acetonitrile, glycerol, N,N-dimethylformamide, AvA r -dimetheylaceatmide, dimethylsulfoxide, or any combination thereof.
  • the present invention provides formulations prepared by mixing an agent with a pharmaceutically acceptable carrier.
  • the formulation may be prepared using a method comprising: a) dissolving a described agent in a water-soluble organic solvent, a non-ionic solvent, a water- soluble lipid, a cyclodextrin, a vitamin such as tocopherol, a fatty acid, a fatty acid ester, a phospholipid, or a combination thereof, to provide a solution; and b) adding saline or a buffer containing 1-10% carbohydrate solution.
  • the carbohydrate comprises dextrose.
  • water soluble organic solvents for use in the present methods include and are not limited to polyethylene glycol (PEG), alcohols, acetonitrile, N-methyl-2-pyrrolidone, N,N-dimethylformarnide, A V-dimethylacetamide, dimethyl sulfoxide, or a combination thereof.
  • PEG polyethylene glycol
  • alcohols include but are not limited to methanol, ethanol, isopropanol, glycerol, or propylene glycol.
  • Illustrative examples of water soluble non-ionic surfactants for use in the present methods include and are not limited to CREMOPHOR* EL, polyethylene glycol modified CREMOPHOR* (polyoxyethyleneglyceroltriricinoleat 35), hydrogenated CREMOPHOR ® RH40, hydrogenated CREMOPHOR ® RH60, PEG-succinate, polysorbate 20, polysorbate 80, SOLUTOL*' HS (polyethylene glycol 660 12-hydroxystearate), sorbitan monooleate, poloxamer, LABRAFIL* (ethoxylated persic oil), LABRASQL ® (capryl- caproyl macrogol-8-glyceride), GELUCIRE ® (glycerol ester), SOFTIGEN* (PEG 6 caprylic glyceride), glycerin, glycol-polysorbate, or a combination thereof.
  • CREMOPHOR* EL polyethylene glycol modified CREMOPHOR
  • Illustrative examples of water soluble lipids for use in the present methods include but are not limited to vegetable oils, triglycerides, plant oils, or a combination thereof
  • lipid oils include but are not limited to castor oil, polyoxvl castor oil, corn oil, olive oil, cottonseed oil, peanut oil, peppermint oil, safflower oil, sesame oil, soybean oil, hydrogenated vegetable oil, hydrogenated soybean oil, a triglyceride of coconut oil, palm seed oil, and hydrogenated forms thereof, or a combination thereof.
  • Illustrative examples of fatty acids and fatty acid esters for use in the present methods include but are not limited to oleic acid, monoglycerides, diglycerides, a mono- or di -fatty acid ester of PEG, or a combination thereof.
  • cyclodextrins for use in the present methods include but are not limited to alpha-cyclodextrin, beta-cyclodextrin, hydroxypropyl-beta- cyclodextrin, or sulfobutyl ether-beta-cyclodextrin.
  • Illustrative examples of phospholipids for use in the present methods include but are not limited to soy phosphatidylcholine, or distearoyl phosphatidylglycerol, and hydrogenated forms thereof, or a combination thereof.
  • One of ordinary skill in the art may modify the formulations within the teachings of the specification to provide numerous formulations for a particular route of administration.
  • the compounds may be modified to render them more soluble in water or other vehicle. It is also well within the ordinary skill of the art to modify the route of admini stration and dosage regimen of a particular compound in order to manage the pharmacokinetics of the present compounds for maximum beneficial effect in a patient.
  • compositions comprising GiyRS inhibitors and disclosed herein can be used in combination with a pharmaceutical composition comprising one or more therapeutic agents.
  • the terms “combination” and “in combination with” mean the administration of a therapeutic agent described herein together with at least one additional pharmaceutical or medicinal agent (e.g., an anti-cancer agent), either sequentially or simultaneously.
  • the term encompasses dosing simultaneously, or within minutes or hours of each other, or on the same day, or on alternating days, or dosing the therapeutic agent described herein on a daily basis, or multiple days per week, or weekly basis, for example, while administering another compound such as a chemotherapeutic agent on the same day or alternating days or weeks or on a periodic basis during a time simultaneous therewith or concurrent therewith, or at least a part of the time during which the therapeutic agent described herein is dosed.
  • compositions for use in accordance with the present disclosure can be manufactured and/or formulated in conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen. Any of the well- known techniques, carriers, and excipients may be used as suitable and as understood in the art, e.g., in Remington's Pharmaceutical Sciences, above.
  • Suitable routes of admini stration of the pharmaceutical compositions disclosed herein include, for example, oral, rectal, transmucosal, topical, or intestinal administration, parenteral delivery, including intramuscular, subcutaneous, intravenous, intramedullary injections, as well as intrathecal, direct intraventricular, intraperitoneal, intranasal, or intraocular injections.
  • the compound or combination of compounds disclosed herein can also be administered in sustained or controlled release dosage forms, including depot injections, osmotic pumps, pills, transdermal (including electrotransport) patches, and the like, for prolonged and/or timed, pulsed administration at a predetermined rate.
  • Injectables can be prepared in conventional forms, either as liquid solutions or suspensions, solid forms suitable for solution or suspension in liquid prior to injection, or as emulsions.
  • Suitable excipients are, for example, water, saline, dextrose, mannitol, lactose, lecithin, albumin, sodium glutamate, cysteine hydrochloride, and the like.
  • the injectable pharmaceutical compositions may contain minor amounts of nontoxic auxiliary substances, such as wetting agents, pH buffering agents, and the like.
  • Physiologically compatible buffers include, but are not limited to, Hanks's solution. Ringer's solution, or physiological saline buffer. If desired, absorption enhancing preparations (for example, liposomes), may be utilized.
  • penetrants appropriate to the barrier to be permeated may be used in the formulation.
  • 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 other organic oils such as soybean, grapefruit or almond oils, 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.
  • the suspension may also contain suitable stabilizers or agents that increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative.
  • the compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • the compound(s) or combination of compounds disclosed herein can be formulated readily by combining the active compound with pharmaceutically acceptable carriers well known in the art.
  • Such carriers enable the compound or combination of compounds disclosed herein to be formulated as tablets, film coated tablets, pills, dragees, capsules, liquids, gels, get caps, pellets, beads, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated.
  • Pharmaceutical preparations for oral use can be obtained by combining the active compound 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, and/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.
  • Dragee cores are provided with suitable coatings.
  • concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/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.
  • concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/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.
  • stabilizers can be added. All formulations for oral administration should be in dosages suitable for such administration.
  • formulations of the compound(s) or combination of compounds disclosed herein with an acceptable immediate release dissolution profile and a robust, scalable method of manufacture are disclosed,
  • 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, and/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. All formulations for oral administration should be in dosages suitable for such administration.
  • compositions may take the form of tablets or lozenges formulated in conventional manner.
  • the compound or combination of compounds disclosed herein is conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofiuoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofiuoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofiuoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlor
  • compositions for intraocular delivery include aqueous ophthalmic solutions of the active compounds in water-soluble form, such as eyedrops, or in geilan gum or hydrogels; ophthalmic ointments; ophthalmic suspensions, such as microparticulat.es, drug- containing small polymeric particles that are suspended in a liquid carrier medium, lipid- soluble formulations, microspheres, and ocular inserts.
  • Suitable pharmaceutical formulations are most often and preferably formulated to be sterile, isotonic and buffered for stability and comfort.
  • Pharmaceutical compositions for intranasal delviery may also include drops and sprays often prepared to simulate in many respects nasal secretions to ensure maintenance of normal ciliary action.
  • suitable formulations are often and preferably isotonic, slightly buffered to maintain a pH of 5.5 to 6.5, and most often and preferably include antimicrobial preservatives and appropriate drug stabilizers.
  • compositions for intraauricular delivery include suspensions and ointments for topical application in the ear.
  • Common solvents for such aural formulations include glycerin and water.
  • the compound(s) or combination of compounds disclosed herein may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other giycerides.
  • the compound or combination of compounds disclosed herein may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneousiy or intramuscularly) or by intramuscular injection.
  • the compound or combination of compounds disclosed herein may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • a suitable pharmaceutical carrier may be a cosumble system comprising benzyl alcohol, a nonpoiar surfactant, a water-miscible organic polymer, and an aqueous phase.
  • a common cosumble system used is the VPD co-solvent system, which is a solution of 3% w/v benzyl alcohol, 8% w/v of the nonpoiar surfactant Polysorbate 80. TM., and 65% w/v polyethylene glycol 300, made up to volume in absolute ethanol.
  • VPD co-solvent system is a solution of 3% w/v benzyl alcohol, 8% w/v of the nonpoiar surfactant Polysorbate 80. TM., and 65% w/v polyethylene glycol 300, made up to volume in absolute ethanol.
  • the proportions of a co-solvent system may be varied considerably without destroying its solubility and toxicity characteristics.
  • co-solvent components may be varied: for example, other low-toxicity nonpoiar surfactants may be used instead of POLYSORBATE 80. TM.; the fraction size of polyethylene glycol may be varied, other biocompatible polymers may replace polyethylene glycol, e.g., polyvinyl pyrrolidone; and other sugars or polysaccharides may substitute for dextrose.
  • hydrophobic pharmaceutical compounds may ⁇ be employed. Liposomes and emulsions are well known examples of deliver ⁇ ' vehicles or carriers for hydrophobic drugs. Certain organic solvents such as dimethylsulfoxide also may ⁇ be employed, although usually at the cost of greater toxicity. Additionally, the compounds may be delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent. Various sustained-release materials have been established and are well known by those skilled in the art. Sustained-release capsules may, depending on their chemical nature, release the compounds for a few weeks up to over 100 days. Depending on the chemical nature and the biological stability of the therapeutic reagent, additional strategies for protein stabilization may be employed.
  • Agents intended to be administered intracellularly may be administered using techniques well known to those of ordinary skill in the art.
  • such agents may be encapsulated into liposomes. All molecules present in an aqueous solution at the time of liposome formation are incorporated into the aqueous interior.
  • the liposomal contents are both protected from the external micro-environment and, because liposomes fuse with cell membranes, are efficiently delivered into the cell cytoplasm.
  • the liposome may be coated with a tissue-specific antibody.
  • the liposomes will be targeted to and taken up selectively by the desired organ.
  • small hydrophobic organic molecules may be directly administered intracellularly.
  • Additional therapeutic or diagnostic agents may be incorporated into the pharmaceutical compositions.
  • pharmaceutical compositions may be combined with other compositions that contain other therapeutic or diagnostic agents.
  • Some embodiments disclosed herein provide methods of reducing neddylation in a cell, in a cell population, and/or in a subject.
  • the methods comprise contacting a ceil, a ceil population, and/or a subject with a composition comprising a GlyRS inhibitor, wherein the level of neddylation is decreased in the cell, the ceil population, and/or the subject.
  • the methods comprise acquiring knowledge of the level of neddylation in the cell, the cell population, and/or the subject before and/or after contacting the cell, the cell population, and/or the subject with the composition.
  • the methods comprise identifying a cell, a cell population, and/or a subject having undesired level of neddylation before and/or after contacting the cell, the cell population, and/or the subject with the composition.
  • the composition can be, for example, a pharmaceutical composition.
  • the GlyRS inhibitor can be any of the GlyRS inhibitor disclosed herein.
  • the GlyRS inhibitor may inhibit GlyRS functions in aminoacylation and neddylation. In some embodiments, the GlyRS inhibitor does not significantly inhibit GlyRS function in aminoacylation.
  • the GlyRS inhibitor is an inhibitor for a mammalian GlyRS protein (e.g., a human GlyRS protein).
  • the GlyRS inhibitor is GlySA or a derivative thereof.
  • the cell can be, for example a mammalian cell (e.g., a human cell).
  • the cell population can comprise, or consist of, mammalian cells (e.g., human cells).
  • the cell or the cell population can be present in cell culture, a tissue, in an organ, or in a body of a subject. Contacting the cell or the cell population with the composition can be performed in vitro, ex vivo, in vivo, or a combination thereof.
  • Some embodiments disclosed herein provide methods of inhibiting or reducing cell proliferation.
  • the methods comprise: contacting a cell, a ceil population, or a subject with a pharmaceutical composition comprising a GlyRS inhibitor, wherein the proliferation of the cell, one or more cells present in the cell population or the subject is inhibited.
  • the inhibition of cell proliferation can be complete or partial.
  • a method of reducing cell proliferation is provided.
  • the methods comprise acquiring knowledge of the level of cell proliferation in the cell, the cell population, and/or the subject before and/or after contacting the ceil, the ceil population, and/or the subject with the composition. In some embodiments, the methods comprise identifying a cell, a cell population, and/or a subject having undesired level of cell proliferation before and/or after contacting the cell, the cell population, and/or the subject with the composition,
  • the NEDD8 pathway has been shown to be essential for cellular function, through its critical role in mediating the ubiquitination by CRLs of numerous proteins involved in cell cycle progression and cell growth and survival.
  • the relevance of the NEDD8 conjugation pathway in various cancer therapies has been discussed in Soucy et ai., The NEDD8 Conjugation Pathway and Its Relevance in Cancer Biology and Therapy. Genes Cancer 1, 708-16 (2010), the content of which is hereby expressly incorporated by reference in its entirety.
  • Table 1 of Soucy et al. summarizes various substrate proteins of the cullin- RTNG ligases and their associations with cancer, and is reproduced below. Additional substrates that are involved in tumorigenesis include: tumor suppressor NF2, which encodes Merlin, p53, Mdm2, epidermal growth factor receptor, VHL tumor suppressor protein, Ll l, or any combination thereof .
  • Neddylation of the cullin proteins activates the E3 ligases for ubiquitination and promotes the degradation of their downstream targets, including key regulators of ceil cycle.
  • E3 ligases for ubiquitination and promotes the degradation of their downstream targets, including key regulators of ceil cycle.
  • Cell proliferation can be inhibited in various types of cells, including animal cells and plant cells.
  • the cell is a mammalian ceil.
  • the extent by which cell proliferation is reduced can vary.
  • the proliferation of the cell is reduced by at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 99%.
  • the composition can be, for example, a pharmaceutical composition.
  • the GlyRS inhibitor can be any of the GlyRS inhibitor disclosed herein.
  • the GlyRS inhibitor may inhibit GlyRS functions in aminoacylation and neddylation. In some embodiments, the GlyRS inhibitor does not significantly inhibit GlyRS function in aminoacylation.
  • the GlyRS inhibitor is an inhibitor for a mammalian GlyRS protein (e.g., a human GlyRS protein). In some embodiments, the GlyRS inhibitor is GlySA or a derivative thereof.
  • the cell can be, for example a mammalian cell (e.g., a human cell).
  • the ceil population can comprise, or consist of, mammalian cells (e.g., human cells).
  • the cell or the cell population can be present in cell culture, a tissue, in an organ, or in a body of a subject. Contacting the cell or the cell population with the composition can be performed in vitro, ex vivo, in vivo, or a combination thereof.
  • the GlyRS inhibitor may be capable of inhibiting or reducing interaction between the GlyRS protein and the components of the neddylation pathway, thereby reduce proliferation of the cancer cells.
  • the methods may be used for treating or ameliorating a solid tumor or a hematological malignancy, for example, a cancer that is selected from the group consisting of breast cancer, cervical cancer, colon cancer, liver cancer, prostate cancer, melanoma, ovarian cancer, lung cancer, renal cell carcinoma, Schwannoma, mesothelioma, acute myeloid leukemia, multiple myeloma, non-Hodgkin lymphoma, and a combination thereof.
  • a cancer that is selected from the group consisting of breast cancer, cervical cancer, colon cancer, liver cancer, prostate cancer, melanoma, ovarian cancer, lung cancer, renal cell carcinoma, Schwannoma, mesothelioma, acute myeloid leukemia, multiple myeloma, non-Hodgkin lymphoma, and a combination thereof.
  • the GlyRS inhibitor can be any of the GlyRS inhibitor disclosed herein.
  • the GlyRS inhibitor may inhibit GlyRS functions in aminoacylation and neddylation. In some embodiments, the GlyRS inhibitor does not significantly inhibit GlyRS function in aminoacylation.
  • the GlyRS inhibitor is an inhibitor for a mammalian GlyRS protein (e.g., a human GlyRS protein). In some embodiments, the GlyRS inhibitor is GiySA or a derivative thereof.
  • a GlyRS inhibitor, and/or an agent that prevents or reduces interaction between a GlyRS protein and a component of the neddylation pathway, and pharmaceutical compositions thereof may be administered orally, parenteral ly, by inhalation, topically, rectal ly, nasally, buccally, vaginally, via an implanted reservoir, or other drug administration methods.
  • parenteral as used herein includes subcutaneous, intracutaneous, intravenous, intramuscular, intraarticular, intraarterial, intrasynovial, intrastemal, intrathecal, intralesional and intracranial injection or infusion techniques,
  • a sterile injectable composition such as a sterile injectable aqueous or oleaginous suspension, may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent.
  • acceptable vehicles and solvents include mannitol, water. Ringer's solution and isotonic sodium chloride solution.
  • Suitable carriers and other pharmaceutical composition components are typically sterile.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium (e.g., synthetic mono- or diglycerides).
  • Fatty acids such as oleic acid and its giyceride derivatives, are useful in the preparation of injectables, as are pharmaceutically acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • These oil solutions or suspensions can also contain a long-chain alcohol diluent or dispersant, or carboxymethyl cellulose or similar dispersing agents.
  • Various emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms can also be used for the purpose of formulation.
  • a composition for oral administration may be any orally acceptable dosage form including, but not limited to, tablets, capsules, emulsions and aqueous suspensions, dispersions and solutions.
  • commonly used carriers include lactose and corn starch.
  • Lubricating agents such as magnesium stearate, can also be added.
  • useful diluents include lactose and dried com starch.
  • a nasal aerosol or inhalation compositions can be prepared according to techniques well-known in the art of pharmaceutical formulation and can be prepared as solutions in, for example saline, employing suitable preservatives (for example, benzyl alcohol), absorption promoters to enhance bioavailability, and/or other solubilizing or dispersing agents known in the art.
  • suitable preservatives for example, benzyl alcohol
  • absorption promoters to enhance bioavailability
  • other solubilizing or dispersing agents known in the art.
  • Glvcyl-tRNA synthetase is critical for neddylation by protecting activated E2
  • Ubiquitination and ubiquitin-like modifications are post-translational modifications in eukaryotes playing key roles in protein homeostasis and functions 1 .
  • neddylation conjugating the ubiquitin-like protein NEDD8 to its target proteins— is an essential biological process in organisms from yeast to mammals to critically regulate cell cycle progression.
  • the modification is achieved through a sequential enzymatic cascade involving an activating enzyme (El), a conjugating enzyme (E2), and a ligase (E3) (Fig. la).
  • Ubiquitin and most ubiquitin-like modifier proteins have a conserved C-terminal glycine that is used to activate, conjugate, and finally attach the modifiers to their targets.
  • the glycine residue is located at the tip of a flexible 'tail' protruding out from the central ubiquitin fold to provide with the accessibility.
  • GlyRS is a member of the evoiutionarily ancient aminoacyl-tRNA synthetase family essential for all living organisms. GlyRS catalyzes the aminoacylation reaction to attach glycine to the 3 ' end of the cognate tRNAs to provide substrates for protein synthesis in the ribosome.
  • Aminoacylation of tRNA is a two-step reaction: first, GlyRS activates glycine with ATP to generate Gly-AMP; then the glycyi moiety is transferred from Gly-AMP to the tRNA to make the 'charged' tRNA.
  • the first step of this aminoacylation reaction is chemically equivalent to the first step activation reaction catalyzed by an E l enzyme.
  • the specific amino acid binding pocket of a tRNA synthetase can be exploited for binding to a cognate amino acid residue on a protein to develop new functions.
  • GlyRS plays an important role in neddylation through direct interactions with multiple components of the neddylation pathway.
  • GlyRS cannot function as an El, it can bind to the APPBP1 subunit of El to capture and protect the activated E2 (NEDD8-conjugated Ubcl2) to critically enhance neddylation.
  • E2 NEDD8-conjugated Ubcl2
  • Knockdown of GlyRS, but not a different tRNA synthetase decreases the global level of neddylation and delays cell cycle progression. This provides the first example of a translation factor directly functioning in posttranslational modification.
  • GST-tagged or untagged APPBP1-UBA3, Ubcl2, and EDD8 proteins were purified as previously reported.
  • GST-APPBP1 was obtained by injecting the purified GST-APPBP1/UBA3 into the Superdex 200 column and collected the fractions that contained only GST-APPBP1.
  • N-terminal His-SUMO-tagged NEDD8 construct was generated by subcloning the pGEX2TK-NEDD8 into a modified pET28a vector and the protein was purified using a Nickel -NT A column followed by MonoQ ion exchange column.
  • Ubiquitin and SUMOl proteins R&D were purchased. His-tagged full-length and AWHEP GlyRS, SerRS, and TrpRS were purified as reported earlier.
  • the ABD (V541 -E685) of GlyRS was cloned, expressed, and purified in a similar way as for the full-length GlyRS. (The yield for ABD alone is higher than that for the full-length GlyRS).
  • Tag-free full-length GlyRS, AF84-L93 (F84-L93 replaced by GGG), ⁇ 232- ⁇ 238 ( ⁇ 232- ⁇ 238 replaced by GG), Alnsertionl (F 147-F224 replaced by GSGSGG) and G526R GlyRS proteins were similarly prepared and had the N-terminal His-SUMO-tag removed by Ulpl protease.
  • the conjugated Ubcl2 was prepared by mixing APPBP1/UBA3 (2 ⁇ ), Ubcl2 (C I 1 I S; the mutant would form a stable ester linkage to NEDD8) (70 ⁇ ), and His- SUMO-tagged NEDD8 ( ⁇ ⁇ ). The mixture was incubated at 25°C for 16 h in 50 mM NaOAc pH 5.5, 50mM NaCl, ImM MgC12, and 2mM ATP. Ubcl2 N8 was then purified by- gel filtration chromatography using a Superdex 200 column. The fractions containing only Libel 2 N8 were concentrated and used for the study. The purity of proteins was examined by SDS-PAGE to be above 95%.
  • Ubcl2 conjugation was performed in a similar manner except using wild type Ubcl2 instead of Ubcl2 el l Is at 25°C for 16 h in 50 mM Tris pH 7.4, 50mM NaCl, ImM MgC12, and 2mM ATP.
  • Ubcl2 N8 was then purified by gel filtration chromatography using a Superdex 200 column. The fractions containing only Libel 2 N8 were concentrated and used for the study. The purity of proteins was examined by SDS-PAGE to be above 95%.
  • the stability assay was carried out by incubating Ubcl2 N8 (5uM) with GivRS, BSA or same volume of PBS buffer at 37° for indicated time in the PBS buffer (PH7.4 supplemented with 5mM: DTT). Samples were then subjected to SDS-PAGE and stained with commassie blue.
  • HEK293 and HeLa cells were cultured in DMEM media supplemented with 10% FBS.
  • Short-hairpin RNA (shRNA) sequences targeting the human GlyRS (5'- GCATGGAGTATCTCACAAAGT-3', SEQ ID NO: 3) or human SerRS (5'- GGCATAGGGACCCATCATTGA-3', SEQ ID NO: 36) were inserted into the pLentiLox- hHl piasmid, modified from the pLentiLox 3.7 plasmid to contain a HI promoter (between Xba I and Xho I sites) to drive shRNA expression. All transfections were done with Lipofectamine 2000 (Invitrogen) and cells were harvested 48 hours after transtection. Cycloheximide chasing assay
  • HeLa cells at 80% confluence were transfected with pLentiLox-hHl vectors containing either a scramble sequence or GlyRS specific sequence using Lipofectamine 2000. 48 hours after transfection, medium were replaced with that containing 3C ⁇ g/mL cycloheximide (Celisignaliing) or 20uM MG-132 (#508338, Fisher) as indicated.
  • MLN4924 (1-502, Bostonbiochem) samples are prepared by treating the ceils with 0.2 uM MLN4924 for 24 hours and then followed by cycloheximide or MG132. Samples are collected and lysed with acid lysis buffer and later subjected to SDS-PAGE.
  • Anti-NEDD8 (#2745), anti-Ubiquitin (#3936), anti-SUMOl (#4930), anti-Ubcl 2 (#5641 ), anti-Ubc9 (#4918), anti-Flag (#2908), anti-UBAf (#4891), anti-UbcH7 (#3848), anti-UBA2 (#8688), anti-p27kip (#3698) and anti- a-Tubulin (#3873) are all from Cell Signaling, Immunoblots are quantified and analyzed using Image J. The integrated areas of the bands were normalized to that of the corresponding a-tubulin level. Error bars indicate standard deviation with n>3. P values are calculated by one-tailed Student's t test.
  • HeLa cells were transfected with either pCDNA6-V5c vectors containing either GARS or G526R mutant using Lipofectaming 2000. 48 hours after transefection, cells were treated with medium containing 0.5 uM MLN4924 or same amount of DMSO. 24 hours after treatment, cells were washed once with sorting buffer (PBS supplemented with 1 %FBS and 5mM EDTA) and collected using 0.05% Trypsin. The mixture was then spinned at 500g for 5min to spin cells down to the pellets. Cells were then washed twice with sorting buffer and then suspended and fixed with 70% EtOH at 4 for 2 hours. After fixation, cells were washed twice with sorting buffer and suspended with the PI staining solution. Samples were later analyzed by flow cytometry (BD FACS Canto).
  • sorting buffer PBS supplemented with 1 %FBS and 5mM EDTA
  • Glutathione sepharose beads (GE Healthcare) were equilibrated with TEE buffer (50mM Tris pH 7.9), ImM EDTA, and ImM EGTA). GST-fusion proteins were mixed with 50 ul of glutathione sepharose beads and incubated for 2 h at 4°C and then washed with TEE buffer twice. Aliquots of the protein-bound beads were then incubated together with different forms of GlyRS for 2 h at 4°C.
  • the beads were washed 5 times with washing buffer (20 mM Hepes pH 7.9, 150 mM NaCi, 0.5 mM EDTA, 10% Glycerol, 0.1% Triton X- 100, and ImM DTT) and proteins were eluted with SDS sample buffer and analyzed by immunoblotting. Control experiments were performed with GST- coated beads. His-tag pull-down assay were carried out in a similar manner using purified his-tagged proteins and incubated with nickel-NT A beads (Qiagen).
  • Solution-phase amide HDX was carried out with a fully automated system as described previously. Briefly, 4 iL of protein was diluted to 20 ⁇ , ⁇ with D20-comtaining buffer and incubated at 4°C for 10, 30, 60, 900, or 3600 s. Samples were diluted to 50 ⁇ with 3M urea, 1% TFA. at 1°C to denature the proteins and minimize back-exchange. Samples were then passed across an immobilized pepsin column at 50 ⁇ in 0.1% TFA at 15°C. Resulting peptides were trapped on a C8 cartridge (Hypersil Gold, Thermo Fisher).
  • Peptides were then gradient eluted (4% CH3CN to 40% CH3CN, 0.3% formic acid) at 1°C across a lmm x 50mm C 18 reversed phase HPLC column Hypersil Gold, Thermo Fisher) and electrosprayed directly into an orbitrap mass spectrometer (either LTQ Orbitrap or Q- Exactive, Thermo Fisher). Data were processed with in-house software and visualized with PyMol (Schrodinger, LLC). To measure the difference in exchange rates, the average percent deuterium uptake for unbound GlyRS protein was calculated at all time points. From this value, the average percent deuterium uptake for GlyRS protein bound to NEDD8 was subtracted.
  • Negative perturbation values indicate exchange rates are slower for GlyRS bound to NEDD8, which suggests the region is less accessible to amide exchange due to structural alteration or direct contact between GlyRS and NEDD8.
  • GlyRS-Ubcl2 interaction was analyzed in a similar way.
  • the dissociation constants (K d ) were obtained with biolayer interferometry by using an Octet QK system (ForteBio, Menio Park, Calif, USA). Samples or buffer were dispensed into 96-well plates (Millipore, Bil led ca, MA) at 200
  • the Ubcl 2 N8 ⁇ GlyRS interaction is obtained by using the Patchdock server.
  • GlyRS (PDB: 2PME) is assigned as the receptor and Ubcl 2 8 (PDB: 4P50 chain G&H) is assigned as the ligand.
  • Clustering RMSD is set at 4.0.
  • the GlyRS- APPBP1 interaction is modeled using similar settings with APPBP1/UBA3 (PDB: 2NVU chain A&B) assigned as the receptor and GlyRS assigned as the ligand.
  • Molecular visualization and analysis were performed using PyMOL (The PyMOL Molecular Graphics System, Version 1.2rl, Schrodinger, LLC).
  • GlyRS specifically binds to NEDD8 through the catalytic domain
  • Human GlyRS is composed of three distinct domains: the N-terminal metazoan-specific WHEP domain, catalytic domain, and the C -terminal anticodon-hinding domain (ABD) (Fig. lb).
  • GlyRS interaction with ubiquitin or ubiquitin-like proteins such as NEDD8 and SUMOl were investigated.
  • purified human GlyRS protein can specifically bind to NEDD8, but not ubiquitin or SUMOl .
  • Two other human tRNA synthetases were tested side-by-side (SerRS and TrpRS) and showed no interaction with NEDD8, ubiquitin or SUMOl (Fig. lc).
  • the GlyRS-NEDD8 interaction was verified in HEK293 cells by co-immunoprecipitation (Fig. Id). Furthermore, by using truncated recombinant proteins, the interaction was mapped to the catalytic domain of GlyRS (Fig. le). Hydrogen-deuterium exchange (HDX) analysis (monitored by mass spectrometry) further confirmed that the catalytic domain is the site for interaction with NEDD8 (Fig. 6).
  • HDX Hydrogen-deuterium exchange
  • GlyRS plays a critical role in neddylation
  • GlyRS was ectopicaliy expressed in HEK293 cells.
  • the overexpression of GlyRS (but not TrpRS) increased the amount of NEDD8-conjugated Ubcl2 (an E2 for neddylation), but not the ubiquitin- conjugated UbcH7 (an E2 for ubiquitination) and SUMO-conjugated Ubc9 (an E2 for sumoylation) (Fig. 2a).
  • the core synthetase (AWHEP) was still active, while ABD domain alone did not have the effect (Fig. 2a), further highlighting the importance of the catalytic domain.
  • GlvRS strongly binds to NEDD8-conj ugation Ubcl2 and protects it from degradation
  • Ubcl2 exists in two forms in the cell, the apo form and the conjugated form Ubcl2 N8 .
  • the latter form is achieved by linking a NEDD8 molecule, transferred from the activating enzyme APPBP1/UBA3, to the catalytic cysteine residue (Cysl l l) of Ubcl 2 via a thioester bond.
  • GlyRS can interact with both Ubcl2 and EDD8, it was plausible to test if GlyRS could bind to Libe l 2 8 and protect it from degradation.
  • Thioester bonds like the one that links NEDD8 to Ubcl 2, are common intermediates in biological reactions and are highly labile.
  • GlyRS can bind to Ubcl 2 8 in HEK293 cells as detected by co- immunoprecipitation (Fig. 3a). Moreover, relative to the input, more Ubc l 2 NS than i. be 12 was bound to GlyRS, indicating that GlyRS preferably binds to NEDD8-conjugated Ubcl2. To quantify the difference, Uhe l2 NS was enzymatieally made and purified, and its binding affinity to GlyRS was measured using biolayer interferometry.
  • Ubcl2 N8 binds to immobi lized GlyRS with a K& of 4.09 ⁇ 0.30 nM, which is 100-fold and 30-fold stronger the ⁇ ⁇ for Ubcl2 alone (488+73 nM) and for NEDD8 alone (126 ⁇ 19 nM), respectively (Fig. 3b).
  • the GlyRS-Ubcl 2 N8 interaction was also analyzed reversely by immobilizing Ubcl2 N8 to give a similar K ⁇ L of 3.21 ⁇ 0.17 nM (Fig. 3c).
  • Ubcl2 N8 was significantly more stable than in the absence of GlyRS (Fig, 8 ⁇ ). Therefore, it is possible that GlyRS enhances neddylation by protecting reaction intermediate Ubcl2 N8 .
  • GiyRS could al so bind to the heterodimeric El enzyme for neddylation (APPBP1/UBA3) (Fig. 9a & b).
  • EI binds to the anticodon-binding domain (ABD) of GiyRS, as revealed by both GST-pull down and bioiayer interferometry analysis (Fig. 9a,b,c).
  • GlyRS the role of GlyRS might be to capture and protect the conjugated E2 after it is released from the El and before it finds a correct E3 and/or substrate.
  • GlyRS By binding to the APPBP1 subunit of El through the ABD domain, GlyRS is in proximity to capture the released Ubcl2 N8 (Fig. 4b).
  • GlyRS does not interfere with transferring Ubcl2 N8 to substrate
  • GlyRS promotes cell cycle progression
  • neddylation of the cullin activates the CRLl (cull in 1 -RING) ubiquitin ligases and facilitates the degradation of their downstream targets, including ceil cycle inhibitor p27 klp (Fig. 5a), p27 kip induces cell cycle arrest by binding to cyclin-CDK (cyclin-dependent kinase) complexes to inhibit their catalytic activity. Therefore, degradation of p27 kip through cullin neddylation promotes cell cycle progression and cell proliferation. Because this is a well-established pathway, p27 kip degradation and ceil cycle progression were focused on to study the biological role of GlyRS in neddylation,
  • a neddylation specific inhibitor MLN4924 was used, which binds to the ATP -binding site of LIB A3 to block neddylation 21 .
  • MLN4924 treatment completely abolished cullin neddylation and blocked p27 kip degradation (Fig. 5b),
  • Results from biochemical and structural analysis support the model, in which GlyRS, by docking on the APPBP1 subunit of El through the ABD domain, is in proximity to capture, by the catalytic domain, the NEDD8-conjugated E2 (Ubcl2 N8 ) after it is released from the El and before it finds the correct E3 and/or substrate to transfer the NEDD8 modifier.
  • the confinement provided by the synthetase would protect the conjugated E2 from random hydrolysis and thereby enhance the overall efficiency of the neddylation pathway.
  • a synergy may exist between the aminoacylation function of GlyRS to support new protein synthesis and its neddylation enhancer function to stimulate cell proliferation.
  • MLN4924 The selective NEDD8 El inhibitor MLN4924 is currently being tested in several clinical trials for hematological malignancies and solid tumors. As was shown here in Fig 5, a partial knockdown of GlyRS expression in HeLa cells had a similar effect as MLN4924 in causing cell cycle arrest, suggesting that inhibition of GlyRS may be considered for cancer treatment as well. Importantly, inhibition of GlyRS would not only suppress neddylation but also impede protein synthesis, both of which are undesirable for tumor growth.
  • tRNA synthetases are predominantly cytoplasmic proteins for their role in protein synthesis; however, a large repertoire of regulator ⁇ ' functions of tRNA synthetases beyond their enzymatic role in protein synthesis has been reported and many tRNA synthetases are found in the nucleus to carry out important biological functions, such as regulating vascular development, activating p53 signaling, promoting DNA damage response, and regulating gene expression under immunological challenge. Although a nuclear localization signal sequence cannot be readily identified in GlyRS, given the role of GlyRS in neddylation, GlyRS may also reside in the nucleus.
  • the association between GlyRS activity and progression of cancer is accessed by analyzing the expression level of GlyRS in multiple cancer patient samples.
  • the results demonstrate that GlyRS is associated with progression of various cancer types, including breast cancer, ovarian cancer, lung cancer, breast duct carcinoma, colorectal adenocarcinoma and lung squamous cell carcinoma.
  • high level of GlyRS is associated with rapid breast cancer progression.
  • Figure 13 shows experimental data demonstrating that high level of GlyRS is associated with rapid lung cancer progression.
  • FIG. 14 A Also shown in Figures ⁇ 4 ⁇ and 1.4B, higher level of GlyRS staining in most malignant patient cancer tissue samples.
  • Figure 14 B As shown in Figure 14 A, high level staining of GlyRS is observed in patient tissue samples of breast duct carcinoma, colorectal adenocarcinoma and lung squamous cell carcinoma.
  • Figure 14B most malignant patient cancer tissue samples show higher level of GlyRS expression compared to normal tissue.
  • GlySA targets GlyRS with dual mechani sm
  • FIG. 15 shows a non-limiting schematic illustration of GlySA binding to GlyRS active site.
  • GlySA is an analog of Gly-AMP, reaction intermediate of GlyRS.
  • the inhibitor ⁇ ' effects of GlySA on GlyRS aminoacylation and neddylation were tested in both cell lines and mice models.
  • MLN4924 is a known inhibitor of neddylation currently used in clinical trials for multiple solid and hematopoietic cancers. ML.N4924 targets the E l enzyme (UBA3) of neddylation.
  • GlySA (but not MLN4924) inhibits GlyRS aminoacylation.
  • GlySA decreases GlyRS binding to activated NEDD8 E2 (Ubcl2 N8 ).
  • the interactions of GlyRS to that of Ubcl2 N8 were compared in the presence of DMSO or GlySA at 30°C by biolayer interferometry (Octet).
  • NEDD8 activation assay was performed with recombinant human APPBP1-UBA3 (2,7 ⁇ ) protein and fluorescein -labeled NEDD8 proteins in the reaction buffer at 37°C for 1 hour.
  • the concentration of GlySA and MLN4924 was 300 ⁇ .
  • Figisre 18 shows that unlike MLN4924, GlySA does not affect NEDD8 El (UBA3) activation.
  • MDA-MB-231 cells at 80% confluence were treated with 200 nM GlySA and then the cells were harvested at different time points and lysed with the acid lysis buffer and subjected to non-reducing SDS-PAGE,
  • Figure 21 shows experimental data on time course of GlySA in inhibiting neddylation in MDA-MB-231 cells.
  • Figure 23 shows experimental data on GlySA effect on key components and substrates of neddylation in MDA-MB-468 cells.
  • MCF ' 7 cells at 80% confluence were also treated with compounds for overnight and then the cells were harvested and lysed with the acid lysis buffer and subjected to non-reducing SDS- PAGE.
  • Figure 24 shows experimental data on GlySA effect on key components and substrates of neddylation in MCF7 cells.
  • the One-dose data will be reported as a mean graph of the percent growth of treated cells and will be similar in appearance to mean graphs from the 5-dose assay.
  • the number reported for the One-dose assay is growth relative to the no-drug control, and relative to the time zero number of cells. This allows detection of both growth inhibition (values between 0 and 100) and lethality (values less than 0). This is the same as for the 5-dose assay, described below. For example, a value of 100 means no growth inhibition, A value of 40 would mean 60% growth inhibition. A value of 0 means no net growth over the course of the experiment. A value of -40 would mean 40% lethality. A value of -100 means all cells are dead. Information from the One-dose mean graph is available for COMPARE analysis.
  • the plates are incubated for an additional 48 h at 37°C, 5 % C02, 95 % air, and 100 % relative humidity.
  • the assay is terminated by the addition of cold TCA.
  • Cells are fixed in situ by the gentle addition of 50 ⁇ of cold 50 % (w/v) TCA (final concentration, 10 % TCA) and incubated for 60 minutes at 4°C. The supernatant is discarded, and the plates are washed five times with tap water and air dried.
  • Sulforhodamine B (SRB) solution (100 ⁇ ) at 0,4 % (w/v) in I % acetic acid is added to each well, and plates are incubated for 10 minutes at room temperature.
  • Table 2 List of cancer cell lines sorted according to the GlySA growth inhibition effect.
  • Non-Small Cell Lung Cancer A549/ATCC 2.344 19.05 8.1
  • GlySA DMSO stock solution diluted by saline
  • mice mice via tail vein injections. Mice after four injections were evaluated and ail were alive.
  • a schematic illustration of the assay is shown in Figure 25.

Abstract

Disclosed herein are methods and compositions for inhibiting neddylation using Glycyl-tRNA synthase (GlyRS) inhibitors. Also disclosed are related compositions and methods for treating diseases such as cancer.

Description

INHIBITION OF NEDDYLATION USING G L YC YL-tR A SYNTHETASE
INHIBITORS
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] The present application claims the benefit of priority to U.S. Provisional Patent Application No. 62/241386, filed October 14, 2015, the disclosure of which is incorporated by reference herein in its entirety.
STATEMENT REGARDING FEDERALLY SPONSORED R&D
[0002] This invention was made with government support, under National Institutes of Health grant R01GM088278. The U.S. Government has certain rights in this invention.
REFERENCE TO SEQUENCE LISTING
[0003] The present application is being filed along with a Sequence Listing in electronic format. The Sequence Listing is provided as a file entitled SEQLISTING.TXT, created October 7, 2015, which is 15.2 Kb in size. The information in the electronic format of the Sequence Listing is incorporated herein by reference in its entirety
BACKGROUND
Field of the Disclosure
[0004] The present disclosure relates to the fields of molecular biology and medicine. In particular, disclosed herein are compositions and methods for inhibiting neddylation using Glycyl-tRNA synthase (GlyRS) inhibitors, and related compositions and methods for treating diseases such as cancer.
Description of the Related Art
[0005] The NEDD8 pathway plays a critical role in the activation of the ubiquitin E3 ligase activity of cullin-RING ligase (CRL) E3s via the covalent attachment of NEDD8 to the core cullin protein of these enzyme complexes. This process of neddylation has been shown to be essential for the E3 ligase activity of CRLs. CRLs are a large superfamily of E3s that are responsible for the ubiquitination of multiple substrate proteins, including several that are involved in the regulation of normal cellular function as well as some that have been shown to be associated with cancer.
SUMMARY
[0006] Some embodiments disclosed herein relate to methods of reducing neddylation in a cell. In some embodiments, the methods comprise: contacting a cell with a composition comprising a Glycyl-tRNA synthetase (GlyRS) inhibitor, wherein the level of neddylation is decreased in the cell. Some embodiments relate to methods of reducing neddylation in a cell population. In some embodiments, the methods comprise: acquiring knowledge of the level of neddylation in a cell population; and contacting the cell population with a composition comprising a Glycyl-tRNA synthetase (GlyRS) inhibitor to thereby decrease the level of neddylation in the cell population. Some embodiments relate to methods of reducing neddylation in a cell population. In some embodiments, the methods comprise: identifying a cell population having undesirable level of neddylation; and contacting the ceil population with a composition comprising a Glycyl-tRNA synthetase (GlyRS) inhibitor to thereby decrease the level of neddylation in the cell population.
[0007] In the methods of reducing neddylation, the composition can be, for example, a pharmaceutical composition. In some embodiments, the GlyRS inhibitor inhibits GlyRS functions in aminoacylation and neddylation. In some embodiments, the GlyRS inhibitor does not significantly inhibit GlyRS function in aminoacylation.
[0008] In some embodiments, the GlyRS inhibitor is an inhibitor for a mammalian GlyRS protein. In some embodiments, the GlyRS inhibitor is an inhibitor for a human GlyRS protein. In some embodiments, the human GlyRS protein comprises an amino acid sequence having at least 90% identity to the amino acid sequence set forth in SEQ ID NO: 2. In some embodiments, the GlyRS inhibitor is an inhibitor for a plant GlyRS protein.
[0009] The GlyRS inhibitor can be, for example, a protein, a nucleic acid, a small molecule compound, or a combination thereof. In some embodiments, the GlyRS inhibitor is an RNA molecule capable of silencing the expression of a GlyRS gene. In some embodiments, the GlyRS inhibitor is an RNA molecule that binds to an mRNA encoded by a GlyRS gene. In some embodiments, the GlyRS gene is a mammalian GlyRS gene, for example a human GARS gene. In some embodiments, the human GARS gene consists of or comprises a nucleotide sequence having at least 90% sequence identity to the nucleotide sequence set forth in SEQ ID NO: 1. In some embodiments, the GlyRS inhibitor is a short- hairpin RNA (shRNA) consisting of or comprising a nucleotide sequence having at least 90% sequence identity to the nucleic acid sequence of SEQ ID NOs: 3-35. For example, the GlyRS inhibitor is a RNA consisting of or comprising the nucleic acid sequence of SEQ ID NOs:3~35, In some embodiments, the GlyRS inhibitor is GlySA or a derivative thereof.
[0010] In some embodiments, the GlyRS inhibitor interferes with the binding between the GlyRS protein and NEDD8 in the cell or the cell population. In some embodiments, the GlyRS inhibitor interferes with the binding between the GlyRS protein and Ubcl2 in the cell or the cell population. In some embodiments, the GlyRS inhibitor interferes with the binding between the GlyRS protein and NEDD8-conjugated Ubcl2 in the cell or the cell population. In some embodiments, the GlyRS inhibitor binds to one or more of amino acids 84-93 of SEQ ID NO: 2. In some embodiments, the GlyRS inhibitor binds to one or more of amino acids 232-238 of SEQ ID NO: 2. In some embodiments, the GlyRS inhibitor binds to one or more of amino acids Arg277, Glu279, Val289, Glu296, Ue404, and Glu552 of SEQ ID NO: 2. In some embodiments, the GlyRS inhibitor is a GlySA derivative. In some embodiments, the GlyRS inhibitor decreases the amount of NEDD8-conjugated Ubcl2 in the cell or the cell population. In some embodiments, the GlyRS inhibitor decreases Ubcl2 activity in the cell or the cell population. In some embodiments, the GlyRS inhibitor increases Ubcl2 degradation in the ceil or the cell population. In some embodiments, the GlyRS inhibitor interferes with the binding between a GlyRS protein and heterodimeiic El enzyme for neddylation (APPBP1/UBA3) in the cell or the cell population. In some embodiments, the GlyRS inhibitor decreases neddylation of a cuilin protein in the cell or the cell population.
[0011] In some embodiments, the cell is a mammalian cell. In some embodiments, the contacting is performed in vitro, ex vivo, or in vivo. In some embodiments, the ceil or the ceil population is present in a tissue or in a body of a subject. In some embodiments, the level of neddylation in the cell or the cell population is reduced by at least [0012] Some embodiments disclosed herein relate to isolated double-stranded ribonucleic acid (dsRNA) molecules that inhibit expression of a Glycyl-tRNA synthetase (GlyRS) gene, wherein a first strand of the dsRNA is substantially identical to at least 19 consecutive nucleotides of the GlyRS gene, and a second strand of the dsRNA is substantially complementary to the first strand. In some embodiments, the GlyRS gene is a mammalian GlyRS gene, for example a human GARS gene. In some embodiments, the human GARS gene comprises or consists of a nucleotide sequence having at least 90% identity to the nucleotide sequence set forth in SEQ ID NO: 1. In some embodiments, the GlyRS gene is a plant GlyRS gene. In some embodiments, the dsRNA is encoded by a polynucleotide, wherein the first strand and the second strand of the dsRNA are transcribed from said polynucleotide and form a hairpin loop.
[0013] Some embodiments disclosed herein relate to isolated single stranded oligonucleotides that are complementary to a portion of a Glycyl-tRNA synthetase (GlyRS) gene of at least 10 consecutive nucleotides. In some embodiments, the GlyRS gene is a mammalian GlyRS gene, for example a human GARS gene. In some embodiments, the human GARS gene comprises or consists of a nucleotide sequence having at least 90% identity to the nucleotide sequence set forth in SEQ ID NO: I . In some embodiments, the GlyRS gene is a plant GlyRS gene.
[0014] Some embodiments disclosed herein relate to methods of identifying an inhibitor of neddylation. The methods, in some embodiments, comprise: providing a test compound; testing the testcompound for its ability to reduce or inhibit the binding between a Glycyl-tRNA synthetase (GlyRS) protein and NEDDS-conjugated Ubcl2; and identifying the compound as an inhibitor of neddylation if the compound has the ability to reduce or inhibit the binding between the GlyRS protein and NEDDS-conjugated Ubcl2. In some embodiments, the inhibitors bind to the catalytic domain of the GlyRS protein. The GlyRS protein can be a mammalian GlyRS protein, for example a human GlyRS protein. In some embodiments, the human GlyRS protein comprises or consists of an amino acid sequence having at least 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:2. In some embodiments, the methods comprise testing the test compound for its ability to reduce or inhibit the aminoacyiation activity of the GlyRS protein. In some embodiments, the methods comprise testing one or more additional test compounds for their ability to reduce or inhibit the binding between the GlyRS protein and NEDD8-conjugated Ubcl2. In some embodiments, the methods comprise testing the one or more additional test compounds for their ability to reduce or inhibit the aminoacylation activity of the GlyRS protein.
[0015] Also disclosed herein are compositions comprising a Glycyl-tRNA synthetase (GlyRS) inhibitor, for example pharmaceutical compositions comprising one or more pharmaceutically acceptable excipients. In some embodiments, the GlyRS inhibitor is an isolated siR A. molecule that binds to an mRNA of the GlyRS protein. In some embodiments, the GlyRS inhibitor is a molecule that inhibits binding between the GlyRS protein and NEDD8-conjugated Ubcl2. In some embodiments, the GlyRS inhibitor is GlySA or a derivative thereof. In some embodiments, the GlyRS inhibitor inhibits GlyRS functions in aminoacylation and neddylation. In some embodiments, the GlyRS inhibitor does not significantly inhibit GlyRS function in aminoacylation.
[0016] Some embodiments disclosed herein relate to methods of reducing cell proliferation. In some embodiments, the methods comprise: contacting a cell with a composition comprising a Glycyl-tRNA synthetase (GlyRS) inhibitor, whereby the proliferation of the cell is reduced. In some embodiments, the activit of the CRLl (cullinl - RING) ubiquitin ligases is inhibited in the cell. In some embodiments, the activity of a substrate of the CRLl ubiquitin ligase is increased in the cell. In some embodiments, the substrate of the CRLl ubiquitin ligase is selected from the group consisting of c-Myc, c-Jun, cyclin E, Emi l , Cdt-1, ρΙκΒα, NRF2, HIF-!a, β-catenin, Cdc25A, mTOR, BimEL and p27. In some embodiments, the methods comprise providing MLN4924 to the ceil. The cell can be, for example, a mammalian cell, a plant cell. In some embodiments, the proliferation of the cell is reduced by at least 50%, In some embodiments, the GlyRS inhibitor inhibits GlyRS functions in aminoacylation and neddylation. In some embodiments, the GlyRS inhibitor does not significantly inhibit GlyRS function in aminoacylation.
[0017] Also disclosed herein are methods of treating or ameliorating cancer in a subject. In some embodiments, the methods comprise: administering a therapeutically effective amount of a pharmaceutical composition comprising a Glycyl-tRNA synthetase (GlyRS) inhibitor to a subject in need thereof. In some embodiments, the pharmaceutical composition further comprises one or more of additional therapeutic agents. In some embodiments, the methods comprise administering one or more additional pharmaceutical compositions comprising one or more of additional therapeutic agents. In some embodiments, the cancer is breast cancer, ovarian cancer, lung cancer, breast duct carcinoma, colorectal adenocarcinoma and lung squamous cell carcinoma, or a combination thereof. In some embodiments, the cancer is selected from the group consisting of breast cancer, cervical cancer, colon cancer, liver cancer, prostate cancer, melanoma, ovarian cancer, lung cancer, renal cell carcinoma, Schwannoma, mesothelioma, acute myeloid leukemia, multiple myeloma, non-Hodgkin lymphoma, and a combination thereof. In some embodiments, the cancer is a solid tumor. In some embodiments, the cancer is a hematological malignancy. In some embodiments, the GlyRS inhibitor is GlySA or a GlySA derivative. In some embodiments, the GlyRS inhibitor inhibits GlyRS functions in aminoacyiation and neddylation. In some embodiments, the GlyRS inhibitor does not significantly inhibit GlyRS function in aminoacyiation. In some embodiments, the GlyRS inhibitor is an inhibitor for a human GlyRS
BRIEF DESCRIPTION OF THE DRAWINGS
[0018] Figures 1.A-E show specific binding between GlyRS and NEDD8. Fig. 1A: A schematic flowchart of the neddylation pathway. Fig. IB: The domain composition of human GlyRS. Fig. 1C: NEDD8, but not ubiquitin and SUMO I , specifically binds to His- tagged GlyRS, but not His-tagged SerRS and TrpRS. Fig. ID: GlyRS binds to NEDD8 in vivo. FIEK293 cells were transfected with Myc-NEDD8 constructs. 48 hours after transfection ceils were harvested and lysed with acid lysis buffer and used for immunoprecipitation assay. Fig. IE: Domain mapping by His-Tag pull-down assay suggests that NEDD8 binds to catalytic domain of GlyRS,
[0019] Figures 2A-D shows that GlyRS plays critical role in neddylation. Fig. 2A: Over-expression of human GlyRS specifically leads to increase of Ubcl 2N8 in HEK293 ceils. The cells were transfected with pcDNA6V5 vectors containing the indicated gene fragment with a V5-tag. Figs. 2B-D: Ubcl2N8 and substrates neddylation decrease specifically upon GlyRS knock-down. Knockdown of the expression of GlyRS, but not SerRS, specifically decreases the levels of Ubcl2N8, and NEDD8-modified cullin proteins in HeLa cells. HeLa cells were transfected by pLenti vectors containing either a scramble sequence, SerRS or GlyRS specific sequences respectively. Ceils were harvested 48 hours after transfection and iysed with acid lysis buffer and subjected to SDS-PAGE.
[0020] Figures 3A-H show that GlyRS preferentially binds to and promote Ubcl2N8. Fig. 3A: Co-immunoprecipitation indicates that GlyRS prefers binding to Ubel2N8 than to the apo Ubcl2. V5 -tagged GlyRS and Flag-tagged Ubcl2-Cl l l S genes were co- transfected to HEK293 cell for the assay. Fig, 3B: Strong binding of Ubcl2N8 to GlyRS as determined by biolayer interferometry. A.U. indicates Arbitrary Unit. Binding analysis to immobilized GST-GlyRS was carried out with the same concentration (62.5nM) of NEDD8, Ubcl 2, or Uhcl2N8 at 30°, Fig. 3C; Biolayer interferometry analysis confirms that GlyRS binding to Ubcl2N8. Binding analysis to immobilized Ubcl2N8 was carried out with a range of concentrations of full-length (8.07 nM-129 nM) GlyRS. Fig. 3D: Structural model of the GiyRS-Ubcl2N8 interaction generated using Patchdock (a server for molecular docking). Figs. 3E-G: Biolayer interferometry analysis confirms that F84-L93 and I232-M238 regions, but not Insertion 1, are important for GlyRS binding to Ubcl2N8. Binding analysis to immobilized Ubcl2N8 was carried out with a range of concentrations (1.25-20 μ^'ΊηΕ) of AF84-L93 (8.16-130 nM), ΔΙ232-Μ238 (8.13-130 nM), and Assertion I (62,5-125 nM) GlyRS. Fig. 3H: Over-expression of ΔΙ232-Μ238 GlyRS in HEK293 ceils cannot promote NEDD8 conjugation of Ubcl 2. HE 293 cells were transfected with pcDNA6V5 vectors containing the indicated gene fragment with a V5-tag.
[0021] Figures 4A-E show that GlyRS captures Ubcl2N8 and escorts it to substrates. Fig. 4A: GlyRS binds to APPBP1 in HEK293 cells. Ceils were lysed with lysis buffer and used for immunoprecipitation assay. Fig. 43B: Structural model for how GlyRS protects UbcI 2N8 during the neddylation cascade. The interaction between GlyRS and El (APPBPl subunit) is modeled by using the Patchdock server. The interaction between E1N8 and Ubcl2 is adapted from a crystal structure (PDB 2NVU). The crystal structure of Ubcl2N8 is from PDB 4P50. The transient interaction between El and Ubcl2N8 is modeled by aligning the NEDD8 molecule from the above two crystal structures. Fig. 4C: Synergistic effect between GlyRS and Ubcl2N8 for interacting with El suggesting the capture of Ubcl2N8 by GlyRS as the conjugated E2 is released from El . Binding analysis to immobilized E l was carried out with 5 .ug/mL of Ubcl2N8 (119 nM), 5 μg/mL of GlyRS (32 nM), or the mixture of Ubcl2N8 and GlyRS each at 5 μΰ/mL. The black dotted line indicates the calculated sum of the binding curves for Ubcl2N8 and GlyRS to El . Fig. 4D: The ABD domain alone lacks the synergistic effect with Ubcl 2NS for binding to El . Binding analysis to immobilized El was card ed out with 5 .ug/mL of Ubel2N8 (1 19 nM), 5 μg/mL of ABD GlyRS (289 nM), or the mixture of Ubcl2N8 and ABD each at 5 _ug mL. The black dotted line indicates the calculated sum of the binding curves for Ubcl2N8 and ABD to El, separately. Fig, 4E: Cullinl competes off GlyRS for Ubcl2N8 interaction. Binding analysis to immobilized Ubcl 2NS was carried out with 20 .ug/'mL of GlyRS (128 nM), 20 g/mL of cullinl ctd/Rbxl (385 nM), or the mixture of cullinl Ctd/Rbxl and GlyRS each at 20 ^ig/niL. The black dotted line indicates the calculated sum of the binding curves for cullinl Ctd/R.bxl and GlyRS to Ubel2N8.
[0022] Figures 5A-C show that GlyRS is involved in cell cycle regulation via neddylation. Fig. 5A: A schematic figure showing how cell cycle kinase inhibitor p27kip half-life is tightly regulated for proper cell cycle progression. p27kip undergoes fast turn over through neddylation activated poly-ubiquitination directed degradation and NEDD8 specific inhibitor MLN 924 could disrupt this and result in abnormal accumulation of p27klp. Fig. SB: Knock-down of GARS extends p27kip half-life. As indicated, HeLa cells were transfected by pLenti vectors containing either a scramble sequence, or GlyRS specific sequences respectively. 24 hours after transfection, cells were treated with fresh medium containing either 0.2μΜ MLN4924 or DMSO for 24 hours. Cell medium were then replaced with that containing cycloheximide (30 g/mL) at indicated time. Meanwhile some ceils were treated with 20 uM MG132 and harvested after 7hours. Cells were harvested and lysed using acid lysis buffer and then subjected to SDS-PAGE. Fig. 5C: FACS analyses of cell cycle confirmed GlyRS involved in cell cycle regulation. Briefly, HeLa cells were transfected with indicated constructs and 24 hours after transfection cells were treated with either 0.2 μΜ MLN4924 or DMSO for another 24 hours. Cells were then collected, fixed and stained with PI and analyzed by flow cytometry. Cells treated with MLN4924 or GARS knock-down showed significant drop of the 2N peak and sequestered in the 4N population, indicating cell cycle arrest.
[0023] Figure 6 shows that GlyRS binds to NEDD8 via its catalytic domain. Hydrogen-deuterium exchange (HDX) analysis shows that NEDD8 bind mainly to the catalytic domain of GlyRS. Changes in deuterium incorporation resulting from the GlyRS- NEDD8 interaction are mapped to the protein sequence and the crystal structure of GlyRS (PDB 2PME).
[0024] Figures 7A-B shows that GlyRS knockdown does not affect Ube2F conjugation. Fig. 7A: Ube2FN8 remains unchanged upon GlyRS knockdown. HeLa cells were transfected by pLenti vectors containing either a scramble sequence, SerRS or GlyRS specific sequences respectively. Cells were harvested 48 hours after transfection and lysed with acid lysis buffer and subjected to SDS-PAGE. Fig. 7B: Ube2F binds to GlyRS much weaker compared to that of Ubcl2. Bioiayer interferometry analysis confirms that Ube2F does not bind to GlyRS compared to that of UbcI 2. Binding analysis to immobilized GST- GlyRS was carried out with 1.0 μΜ of either Ubc l2 or Ube2F.
[0025] Figures 8A-C show that GlyRS catalytic domain mediates interaction with Ubcl2. Fig. 8A: Domain mapping by GST pull-down assay suggests that Ubcl2 also binds to the catalytic domain of GlyRS. Fig. SB: Hydrogen-deuterium exchange (FIDX) analysis confirms that Ubcl2 binds to the catalytic domain of GlyRS. Changes in deuterium incorporation resulting from the GlyRS-Ubcl2 interaction are mapped to the protein sequence and the crystal structure of GlyRS (PDB 2PME). Fig. 8C: GlyRS but not BSA significantly extends the half-life of Ubcl2N8 i vitro. Ubcl2N8 (5μΜ) were incubated with GivRS, BSA or same volume of PBS buffer at 37° for indicated time in the PBS buffer (PH7.4 supplemented with 5mM DTT). Samples were then subjected to SDS-PAGE and stained with commassie blue. The images of the gels were then quantified by Image J and plotted against the time. Error bars represent standard deviations for the image quantification (n=3).
[0026] Figures 9A-F show that GlyRS binds to APPBP1 and facilitates cullin neddylation. Fig. 9A: Domain mapping by GST pull-down suggests that El (APPBP1/UBA3) binds to the anti-codon binding domain (ABD) of GlyRS, as ABD alone can be pulled-down by GST-APPBP1. Fig. 9B: Bioiayer interferometry analysis confirms that ABD alone is sufficient for El interaction, as full-length, AWHEP, and ABD GlyRS bind to El (APPBP1/UBA3) with similar affinity. Binding analysis to immobilized El was carried out with a range of concentrations (62.5-500 nM) of full-length, AWHEP, or ABD GlyRS. Fig. 9C: Bioiayer interferometry analysis confirms that ABD alone binds to El regulatory subunit APPBPI, as ABD GlyRS binds to APPBPl with similar affinity to that of APPBP1 UBA3. Binding analysis to immobilized APPBP1 was carried out with a range of concentrations (0,58-4,6 μΜ) of ABD GlvRS Fig, 91): Molecular docking of GlyRS (PDB 2PME) and ΛΡΡΒΡ Ι - ί ΒΛ3 (PDB 2NVU) by using Patchdock. Fig. 9E: Modeling analysis suggesting that GlyRS is unlikely to interfere with NEDD8 transferring from Ubcl2 to cullin. The complex structure of cullinl-Rbxl -Ubcl2N8 is adapted from PDB 4P50. Thioester bond formed between NEDD8 and Ubcl2 is exposed outside while bound to GlyRS and would not interfere with its transfer to cullin. Fig. 9F: In vitro neddylation assay shows GlyRS facilitate cul lin neddylation ,
[0027] Figure 10 shows a non-limiting schematic illustration showing that GlyRS is associated with cell proliferation via dual cel lular functions. The schematic summarizes the dual function of GlyRS in aminoacylation as an enzyme and in neddylation as a chaperone that supports protein synthesis and cell-cycle progression.
[0028] Figure 11 shows bioinformatic data demonstrating that high level of GlyRS is associated with rapid breast cancer progression. The expression of all cytoplamic human tRNA synthetases in breast cancer was analyzed by Kaplan-Meier plots and hazard ratio (HR). Patient samples were divided in halves as low-expression and high-expression sets for each tRNA synthetase in the analysis, n =;: 3557 patients, P values were calculated with two-sided log-rank tests.
[0029] Figere 12 shows bioinformatic data demonstrating that high level of GlyRS is associated with rapid ovarian cancer progression. Kaplan-Meier plots and hazard ratio (HR) of the expression of human GlyRS in stage 2 ovarian cancer were analyzed. Patient samples were divided in halves as low-expression and high-expression sets for GlyRS in the analysis, n := 60 patients. P values were calculated with two-sided log-rank tests.
[0030] Figure 13 shows bioinformatic data demonstrating that high level of GlyRS is associated with rapid lung cancer progression. Kaplan-Meier plots and hazard ratio (HR) of the expression of human GlyRS in lung squamous cell carcinoma were analyzed. Patient samples were divided in halves as low-expression and high-expression sets for GlyRS in the analysis, n = 524 patients, P values were calculated with two-sided log-rank tests,
[0031] Figures 14A-B show higher level of GlyRS staining in most malignant patient cancer tissue samples. Fig. 14A shows that high level staining of GlyRS is observed in patient tissue samples of breast duct carcinoma, colorectal adenocarcinoma and lung squamous cell carcinoma. Fig, 14B shows that most malignant patient cancer tissue samples have higher level of GlyRS expression compared to normal tissue.
[0032] Figure 15 is a non-limiting schematic illustration of GlySA binding to GlyRS active site (PDB 2ZT8), GlySA is an analog of Gly-AMP, reaction intermediate of GlyRS.
[0033] Figure 16 is a plot showing that GlySA (but not MLN4924) inhibits GlyRS aminoacylation. The aminoacylation assay was performed using recombinant human GlyRS (200 nM) proteins at room temperature. MLN4924 is an inhibitor of neddylation currently used in clinical trials for multiple solid and hematopoietic cancers. MLN4924 targets the El enzyme (UBA3) of neddylation.
[0034] Figure 17 shows experimental data demonstrating that GlySA decreases GlyRS binding to activated EDD8 E2 (Ubcl2N8). The interactions of GlyRS (0.25 μΜ) to that of Ubcl2NS (100 nM; immobilized to the sensor tips) were compared in the presence of DMSO or GlySA at 30°C by biolayer interferometry (Octet).
[0035] Figure 18 shows experimental data demonstrating that unlike MLN4924, GlySA does not affect NEDD8 El (LIB A3) activation. In vitro NEDD8 activation assay was performed with recombinant human APPBP1-UBA3 (2.7 μΜ) protein and fluorescein - labeled NEDD8 proteins at 37°C for 1 hour. The concentration of GlySA and MLN4924 was 300 μ\Ι.
[0036] Figure 19 shows experimental data demonstrating that GlySA, but not SerSA, TyrSA, inhibits cullin neddylation in MDA-MB-231 cells. MDA-MB-231 cells at 80% confluence were treated overnight with each compound and then the cells were harvested and lysed with the acid lysis buffer and subjected to non-reducing SDS-PAGE. SerSA and TyrSA are analogs of Ser-AMP and Tyr-AMP, reaction intermediate of SerRS and TyrRS, respectively. MLN4924 was used as a positive control for the experiment.
[0037] Figure 20 shows experimental data determining iC50 of GlySA for inhibiting cullin neddylation in MDA-MB-231 cells. MD A-MB-231 cells at 80% confluence were treated overnight with GlySA and then the cells were harvested and lysed with the acid lysis buffer and subjected to non-reducing SDS-PAGE.
[0038] Figure 21 shows experimental data on time course of GlySA in inhibiting neddylation in MDA-MB-231 cells. MDA-MB-231 ceils at 80% confluence were treated with 200 nM GlySA and then the cells were harvested at different time points and lysed with the acid lysis buffer and subjected to non-reducing SDS-PAGE.
[0039] Figure 22 shows experimental data on GlySA effect over a range of concentrations on key components and substrates of the neddylation pathway in MDA-MB- 231 cells. MDA-MB-231 cells at 80% confluence were treated overnight with GlySA and then the cells were harvested and lysed with the acid lysis buffer and subjected to non- reducing SDS-PAGE.
[0040] Figure 23 shows experimental data on GlySA effect over a range of concentrations on key components and substrates of neddylation in MDA-MB-468 cells. MDA-MB-468 ceils at 80% confluence were treated overnight with GlySA and then the ceils were harvested and lysed with the acid lysis buffer and subjected to non-reducing SDS- PAGE.
[0041] Figure 24 shows experimental data on GlySA effect over a range of concentrations on key components and substrates of neddylation in MCF7 cells. MCF7 cells at 80% confluence were treated overnight with GlySA and then the ceils were harvested and lysed with the acid lysis buffer and subjected to non-reducing SDS-PAGE.
[0042] Figure 25 shows a schematic illustration of a non-limiting exemplary maximum tolerant dosage assay of GlySA. GlySA (DMSO stock solution diluted by saline) were administrated to three month old female BALB CJ mice via tail vein injections. Mice after four injections were evaluated and all were alive. The GlySA concentration tested were 0.4 mg kg (10 μΜ), 2.0 mg/kg (50 uM), 4.0 mg/kg (100 μΜ). N=3 for each group.
[0043] Figure 26 shows a schematic illustration of non-limiting exemplary lung metastasis assay methods. 1 x 105 MDA-MB-231 cells were injected via tail vein to NOD.Cg- Prkdcscld I12rg mice. Then mice were separated into 3 groups. Group A: vehicle alone (PBS with 1% DMSO), group B: GlySA (4 mg/kg; 100 μΜ), group C: MLN4924 GlySA (4,4 mg/kg; 100 μΜ) were administrated via tail vein injections twice per week. N=T0 for each group.
[0044] Figure 27 shows experimental data demonstrating that GlySA treatment reduces lung metastasis in mice. Top panel: mice lungs 14 days after tumor cells (MDA- MB-231) injection. White dots show the surface tumor colonies. Bottom panel: numbers of
1 ^ lung metastasis colonies are analyzed by two tails unpaired T test. The error bars represent SEM in 8- 1 0}
[0045] Figure 28 shows a non-limiting schematic illustration of a working model of GlySA on inhibiting both protein synthesis and cell-cycle regulation.
[0046] Figure 29 shows GlySA and several GlySA derivatives. The top panel shows chemical structures of GlySA and several GlySA derivatives. The bottom panel shows the key interacting residues on GlyRS with GlySA based on a co-crystal structure of GlySA bound GlyRS (PDB: 2ZT8).
DETAILED DESCRIPTION
[0047] In the following detailed description, reference is made to the accompanying drawings, which form a part hereof. In the drawings, similar symbols typically identify similar components, unless context dictates otherwise. The illustrative embodiments described in the detailed description, drawings, and claims are not meant to be limiting. Other embodiments may be utilized, and other changes may be made, without departing from the spirit or scope of the subject matter presented herein. It will be readily understood that the aspects of the present disclosure, as generally described herein, and il lustrated in the Figures, can be arranged, substituted, combined, separated, and designed in a wide variety of different configurations, all of which are explicitly contemplated herein.
General Techniques
[0048] The practice of the techniques described herein may employ, unless otherwise indicated, conventional techniques and descriptions of organic chemistry, polymer technology, molecular biology (including recombinant techniques), ceil biology, biochemistry, sequencing technology, and micro- and nano-fabrication which are within the skill of those who practice in the art. Such conventional techniques include polymer array synthesis, hybridization and ligation of polynucleotides, and detection of hybridization using a label. Specific illustrations of suitable techniques can be had by reference to the examples herein. However, other equivalent conventional procedures can, of course, also be used. Such conventional techniques and descriptions can be found in standard laboratory manuals such as Green, et al., Eds., Genome Analysis: A Laboratory' Manual Series (Vols. I-IV) (1999); Weiner, Gabriel, Stephens, Eds., Genetic Variation: A Laboratory Manual (2007); Dieffenbach, Dveksler, Eds., PGR Primer: A Laboratory Manual (2003); Bowtell and Sambrook, DNA Microarrays: A Molecular Cloning Manual (2003); Mount, Bioinformatics: Sequence and Genome Analysis (2004); Sambrook and Russell, Condensed Protocols from Molecular Cloning: A Laboratory Manual (2006); and Sambrook and Russell, Molecular Cloning: A Laboratory Manual (2002) (all from Cold Spring Harbor Laboratory Press); Stryer, Biochemistry (4th Ed.) (1995) W.H. Freeman, New York N.Y.; Gait, Oligonucleotide Synthesis: A Practical Approach (2002) IRL Press, London; Nelson and Cox, Lehninger, Principles of Biochemistry (2000) 3rd Ed,, W. H. Freeman Pub., New York, N.Y.; Berg, et ah, Biochemistry (2002) 5th Ed., W.H. Freeman Pub., New York, N.Y., Jaeger, Introduction to Microelectronic Fabrication (2002) 2nd Ed., Prentice Hall, and Madou, Fundamentals of Microfabrication (2002) all of which are herein incorporated in their entireties by reference for all purposes.
Definitions
[0049] Unless defined otherwise, technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the present disclosure belongs. See, e.g.. Singleton et al ,, Dictionary of Microbiology and Molecular Biology 2nd ed., J. Wiley & Sons (New York, NY 1994). All publications mentioned herein are incorporated by reference for the puipose of describing and disclosing devices, formulations and methodologies that may be used in connection with the presently described methods and disclosures.
[0050J For purposes of the present disclosure, the following terms are defined below.
[0051] In this application, the use of the singular can include the plural unless specifically stated otherwise or unless, as will be understood by one of skill in the art in light of the present disclosure, the singular is the only functional embodiment. Thus, for example, "a" can mean more than one, and "one embodiment" can mean that the description applies to multiple embodiments. Additionally, in this application, "and/or" denotes that bot the inclusive meaning of "and" and, alternatively, the exclusive meaning of "or" applies to the list. Thus, the listing should be read to include ail possible combinations of the items of the list and to also include each item, exclusively, from the other items. The addition of this term is not meant to denote any particular meaning to the use of the terms "and" or "or" alone. The meaning of such terms will be evident to one of skill in the art upon reading the particular disclosure.
[0052] The terms "polypeptide", "oligopeptide", "peptide," and "protein" are used interchangeably herein to refer to polymers of amino acids of any length, e.g., at least 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 100, 200, 300, 400, 500, 1,000 or more amino acids. The polymer may be linear or branched, it may include, for example, modified amino acids, and it may be interrupted by non-ami no acids. The terms also encompass an amino acid polymer that has been modified naturally or by intervention; for example, disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation or modification, such as conjugation with a labeling component. Also included within the definition are, for example, polypeptides containing one or more analogs of an amino acid (including, for example, unnatural amino acids, etc. ), as well as other modifications known in the art.
[0053] The terms "polynucleotide," "oligonucleotide," "nucleic acid" and "nucleic acid molecule" are used interchangeably herein to refer to a polymeric form of nucleotides of any length, e.g., at least 8, 9, 10, 20, 30, 40, 50, 100, 200, 300, 400, 500, 1 ,000 or more nucleotides, and may include ribonucleotides, deoxyribonucleotides, analogs thereof, or mixtures thereof. This term refers only to the primary structure of the molecule. Thus, the term includes triple-, double- and single-stranded deoxyribonucleic acid ("DNA"), as well as triple-, double- and single-stranded ribonucleic acid ("RNA"). It also includes modified, for example by alkylation, and/or by capping, and unmodified forms of the polynucleotide. More particularly, the terms "polynucleotide," "oligonucleotide," "nucleic acid" and "nucleic acid molecule" include polydeoxyribonucleotides (containing 2-deoxy-D-ribose), polyribonucleotides (containing D-ribose), including tRNA, rRNA, hRNA, and mRNA, whether spliced or unspliced, any other type of polynucleotide which is an N- or C-glycoside of a purine or pyrimidine base, and other polymers containing normucleotidic backbones, for example, polyamide (e.g., peptide nucleic acids ("PNAs")) and polymorpholino (commercially available from the Anti-Virais, Inc., Corvallis, OR., as Neugene) polymers, and other synthetic sequence-specific nucleic acid polymers providing that the polymers contain nucleobases in a configuration which allows for base pairing and base stacking, such as is found in DNA and RNA. Thus, these terms include, for example, 3'-deoxy-2',5'-DNA, oligodeoxyribonucleotide N3' to P5' phosphoramidates, 2'-0-aikyi-substituted RNA, hybrids between DNA and RNA or between PNAs and DNA or RNA, and also include known types of modifications, for example, labels, alkylation, "caps,'" substitution of one or more of the nucleotides with an analog, intemucleotide modifications such as, for example, those with uncharged linkages (e.g., methyl phosphonates, phosphotriesters, phosphoramidates, carbamates, etc.), with negatively charged linkages (e.g., phosphorothioates, phosphorodithioates, etc.), and with positively charged linkages (e.g., aminoalkylphosphoramidates, aminoalkylphosphotriesters), those containing pendant moieties, such as, for example, proteins (including enzymes (e.g., nucleases), toxins, antibodies, signal peptides, poly-L-lysine, etc.), those with intercalators (e.g., acridine, psoralen, etc.), those containing chelates (of e.g., metals, radioactive metals, boron, oxidative metals, etc.), those containing alkylators, those with modified linkages (e.g., alpha anomeric nucleic acids, etc.), as well as unmodified forms of the polynucleotide or oligonucleotide.
[0054] As used herein, "sequence identity" or "identity" or "homology" in the context of two protein sequences (or nucleotide sequences) includes reference to the residues in the two sequences which are the same when aligned for maximum correspondence over a specified comparison window. The portion of the amino acid sequence or nucleotide sequence in the comparison window may comprise additions or deletions (i.e., gaps) as compared to the reference sequence for optimal alignment of the two sequences. When percentage of sequence identity is used in reference to proteins it is recognized that residue positions which are not identical often differ by conservative amino acid substitutions, where amino acids are substituted for other amino acid residues with similar chemical properties (e.g. charge or hydrophobicity) and therefore do not change the functional properties of the molecule. Where sequences differ in conservative substitutions, the percentage sequence identity may be adjusted upwards to correct for the conservative nature of the substitutions. Sequences, which differ by such conservative substitutions are said to have "sequence similarity" or "similarity". Means for making these adjustments are well known to persons skilled in the art. The percentage is calculated by determining the number of positions at which the identical amino acid or nucleic acid base residue 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 and multiplying the result by 100 to yield the percentage of sequence identity. Typically this involves scoring a conservative substitution as a partial rather than a full mismatch, thereby increasing the percentage sequence identity. Thus, for example, where an identical amino acid is given a score of 1 and a non-conservative substitution is give a score of zero, a conservative substitution is given a score between 0 and 1. The scoring of conservative substitutions is calculated, e.g. according to the algorithm of Meyers and Miller (Computer Applic. Biol. Sci., 1998, 4, 11-17).
[0055] As used herein, the term "homologue" is used to refer to a nucleic acid which differs from a naturally occurring nucleic acid (i.e., the "prototype" or "wild-type" nucleic acid) by minor modifications to the naturally occurring nucleic acid, but which maintains the basic nucleotide structure of the naturally occurring form. Such changes include, but are not limited to: changes in one or a few nucleotides, including deletions (e.g., a truncated version of the nucleic acid) insertions and/or substitutions. A homologue can have enhanced, decreased, or substantially similar properties as compared to the naturally occurring nucleic acid. A homologue can be complementary or matched to the naturally occurring nucleic acid, Homologues can be produced using techniques known in the art for the production of nucleic acids including, but not limited to, recombinant DNA techniques, chemical synthesis, or any combination thereof.
[0056] As used herein, "complementary or matched" means that two nucleic acid sequences have at least 50% sequence identity. For example, the two nucleic acid sequences can have at least 60%, 70%, 80%, 90%, 95%, 96%, 97%, 98%, 99% or 100% of sequence identity. "Complementary or matched" also means that two nucleic acid sequences can hybridize under low, middle and/or high stringency condition(s).
[0057] As used herein, "substantially complementary or substantially matched" means that two nucleic acid sequences have at least 90% sequence identity. For example, the two nucleic acid sequences can have at least 95%, 96%, 97%, 98%, 99% or 100% of sequence identity. Alternatively, "substantially complementary or substantially matched" means that two nucleic acid sequences can hybridize under high stringency condition(s). [0058] As used herein, a "subject" refers to an animal that is the object of treatment, observation or experiment. "Animal " includes cold- and warm-blooded vertebrates and invertebrates such as fish, shellfish, reptiles and, in particular, mammals. "Mammal" includes, without limitation, mice, rats; rabbits; guinea pigs; dogs, cats; sheep; goats; cows; horses; primates, such as monkeys, chimpanzees, and apes, and, in particular, humans.
[0059] As used herein, a "patient" refers to a subject that is being treated by a medical professional, such as a Medical Doctor (i.e. Doctor of Allopathic medicine or Doctor of Osteopathic medicine) or a Doctor of Veterinary Medicine, to attempt to cure, or at least ameliorate the effects of, a particular disease or disorder or to prevent the disease or disorder from occurring in the first place.
[0060] As used herein, "administration" or "administering" refers to a method of giving a dosage of a pharmaceutically active ingredient to a vertebrate.
[0061] As used herein, a "dosage" refers to an amount of therapeutic agent administered to a patient.
[0062] As used herein, a "daily dosage" refers to the total amount of therapeutic agent administered to a patient in a day.
[0063] As used herein, the term "therapeutic agent" means a substance that is effective in the treatment of a disease or condition.
[0064] As used herein, "therapeutically effective amount" or "pharmaceutically effective amount" is meant an amount of therapeutic agent, which has a therapeutic effect. The dosages of a pharmaceutically active ingredient which are useful in treatment are therapeutically effective amounts. Thus, as used herein, a therapeutically effective amount refers to an amount of therapeutic agent which produces the desired therapeutic effect as judged by clinical trial results and/or model animal studies.
[0065] As used herein, a "therapeutic effect" relieves, to some extent, one or more of the symptoms of a disease or disorder. For example, a therapeutic effect may be observed by a reduction of the subjective discomfort that is communicated by a subject (e.g., reduced discomfort noted in self-administered patient questionnaire).
[0066] As used herein, the term "treatment" refers to a clinical intervention made in response to a disease, disorder or physiological condition manifested by a patient, particularly a patient suffering from cancer. The aim of treatment may include, but is not limited to, one or more of the alleviation or prevention of symptoms, slowing or stopping the progression or worsening of a disease, disorder, or condition and the remission of the disease, disorder or condition. In some embodiments, "treatment" refers to both therapeutic treatment and prophylactic or preventative measures. Those in need of treatment include those already- affected by a disease or disorder or undesired physiological condition as well as those in which the disease or disorder or undesired physiological condition is to be prevented. As used herein, the term "prevention" refers to any activity that reduces the burden of the individual later expressing the symptoms. This takes place at primary, secondary and tertiary prevention levels, wherein: a) primary prevention avoids the development of symptoms/disorder/condition; b) secondary prevention activities are aimed at early stages of the condition/disorder/symptom treatment, thereby increasing opportunities for interventions to prevent progression of the condition/disorder/symptom and emergence of symptoms; and c) tertiary prevention reduces the negative impact of an already established condition/disorder/symptom by, for example, restoring function and/or reducing any condition/disorder/symptom or related complications.
[0067] A therapeutic agent or a protective agent may comprise a "drug," As used herein, a "drug" refers to a therapeutic agent or a diagnostic agent and includes any substance, other than food, used in the prevention, diagnosis, alleviation, treatment, or cure of a disease. Stedman's Medical Dictionary, 25th Edition (1990). The drug can include any substance disclosed in at least one of: The Merck Index, 12th Edition (1996); Pei-Show Juo, Concise Dictionary of Biomedicine and Molecular Biology, (1996); U.S. Pharmacopeia Dictionary, 2000 Edition, and Physician's Desk Reference, 2001 Edition. In some embodiments, the therapeutic agent is one of the embodiments of the compositions described herein.
[0068] In some embodiments, the drug used in the therapeutic system will often be placed on, embedded, encapsulated or otherwise incorporated into a deliver}' matrix. The delivery matrix may be included in or on either the first skeletal structure or the second cushioning structure, or both. The delivery matrix, in turn, comprises either a biodegradable or a non-biodegradable material. The delivery matrix may include, although it is not limited to, a polymer. Examples of biodegradable polymers include protein, hydrogel, polyglycolic acid (PGA), polyiactic acid (PLA), poly(L-lactic acid) (PLLA), poiy(L-giycolic acid) (PLGA), polyglycolide, poly-L-lactide, poly-D-lactide, poly(amino acids), polydioxanone, polycaprolactone, polygluconate, polylactic acid-polyethylene oxide copolymers, modified cellulose, collagen, polyorthoesters, polyhydroxybutyrate, polyanhydride, polyphosphoester, poly(alpha-hydroxy acid), and combinations thereof. Non-biodegradable polymers may comprise silicone, acrylates, polyethylenes, polyurethane, polyurethane, hydrogel, polyester (e.g., DACRON® from E. I. Du Pont de Nemours and Company, Wilmington, Del.), polypropylene, polytetrafluoroethylene (PTFE), expanded PTFE (ePTFE), polyether ether ketone (PEEK), nylon, extruded collagen, polymer foam, silicone rubber, polyethylene terephthalate, ultra-high molecular weight polyethylene, polycarbonate urethane, polyurethane, polyimides, stainless steel, nickel-titanium alloy (e.g., Nitinol), titanium, stainless steel, cobalt-chrome alloy (e.g., ELGILOY® from Elgin Specialty Metals, Elgin, 111.; CONICHROME® from Carpenter Metals Corp., Wyomissing, Pa.). In one embodiment, the hydrogel may comprise poly(alkyleneoxides), such as poly(ethyleneoxide), also known as polyethyleneglycols or PEGs.
[0069] The term "comprising" as used herein is synonymous with "including," "containing," or "characterized by," and is inclusive or open-ended and does not exclude additional, unrecited elements or method steps.
[0070] Throughout this disclosure, various aspects are presented in a range format. It should be understood that the description in range format is merely for convenience and brevity and should not be constmed as an inflexible limitation on the scope of the disclosure. Accordingly, the description of a range should be considered to have specifically disclosed all the possible sub-ranges as well as individual numerical values within that range. For example, description of a range such as from 1 to 6 should be considered to have specifically disclosed sub-ranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual numbers within that range, for example, 1, 2, 3, 4, 5, and 6. This applies regardless of the breadth of the range.
[0071] Other objects, advantages and features of the present disclosure will become apparent from the following specification taken in conjunction with the accompanying drawings.
[0072] In the following description, numerous specific details are set forth to provide a more thorough understanding of the present disclosure. However, it will be apparent to one of skill in the art that the methods of the present disclosure may be practiced without one or more of these specific details. In other instances, well-known features and procedures well known to those skilled in the art have not been described in order to avoid obscuring the disclosure.
GlvRS Proteins and Polynucleotides
[0073] Glycyl-tRNA synthetase (GlyRS; also known as glycine-tRNA ligase) is an enzyme that belongs to the aminoacyi tRNA synthetase (aaRS) family. aaRS is an enzyme that attaches the appropriate amino acid onto its tRNA. It does so by catalyzing the esterification of a specific cognate amino acid or its precursor to one of ail its compatible cognate tRNAs to form an aminoacyi -tRNA. GlyRS is an enzyme that catalyzes the chemical reaction:
ATP + glycine + tRNAGly AMP + diphosphate + glycyl-tRNAGly
[0074] The three substrates of the GlyRS enzyme are ATP, glycine, and tRNA(Gly), whereas the three products are AMP, diphosphate and glycyi-tRNA(Gly). Human GlyRS is encoded by the GARS gene, and is composed of three distinct domains: the N-terminal metazoan-specific WHEP domain, catalytic domain, and the C-terminal anticodon-binding domain (ABD). As described herein (for example shown in Figure 10), a GlyRS, in some embodiments, can function in aminoacylation as an enzyme and in neddylation (for example as a chaperon that supports protein synthesis and cell-cycle progress).
[0075] In some embodiments, the GlyRS proteins disclosed herein are capable of interaction with one or more components of the neddylation pathway, including NEDD8, El and E2. For example, the GlyRS proteins may capable of binding to the APPBPl subunit of El and activated E2 (NEDD8-conjugated Ubcl2). In some embodiments, the GlyRS proteins are capable of increasing the level of neddylation in a ceil, for example, neddylation of El, E2, and neddylation substrates, Neddylation substrates include, but are not limited to, members of the cullin protein family, e.g., culiin 1, cullin 2, cullin 3, culiin 4A, cuilin 4B, cullin 5, cullin 7, and cullin 9. In some embodiments, the substrates are human cullin proteins. In some embodiments, the GlyRS proteins are capable of increasing the level of cullin-RTNG ubiquitin ligases (CRLs) in a cell. Without being bound by any particular theory, it is believed that the GlyRS proteins disclosed herein may increase the level of neddyiation through interacting with NEDDS-conjugation Ubcl 2 and protecting it from degradation. In some embodiments, the GlyRS proteins do not interact with other ubiquitin or ubiquitin-like proteins such as SUMOl .
[0076] The coding sequence of a human GARB gene is shown below (SEQ ID NO: 1). Also contemplated herein are GlyRS nucleotide sequences that have at least 70%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or more sequence identity to SEQ ID NO: 1.
atg
361 ccctctccgc gtccagtgct gcttagaggt gctcgcgccg ctctgctgct
gctgctgccg
421 ccccggctct tagcccgacc ctcgctcctg ctccgccggt ccctcagcgc
ggcctcctgc
481 cccccgatct ccttgcccgc cgccgcctcc cggagcagca tggacggcgc
gggggctgag
541 gaggtgctgg cacctct;gag gctagcagtg cgccagcagg gagatc1.1gt
gcgaaaactc
601 aaagaagata aagcacccca agtagacgta gacaaagcag tggctgagct
caaagcccgc
661 aagagggttc tggaagcaaa ggagctggcg ttacagccca aagatgatat
tgtagaccga
721 gcaaaaatgg aagataccct gaagaggagg tttttctatg atcaagcttt
tgctatttat
781 ggaggtgtta gtggtctgta tgact.ttggg ccagttggct. gtgcttt.gaa
gaacaatatt
841 attcagacct. ggaggcagca ctttatccaa gaggaacaga tcctggagat
cgattgcacc
901 atgctcaccc ctgagccagt tttaaagacc tctggccatg tagacaaatt
tgctgact.tc
961 atgg gaaag acgtaaaaaa tggagaatgt tttcgtgctg accatctatt
aaaagctcat
1021 ttacagaaat tgatgtctga taagaag gt tctgtcgaaa agaaatcaga
aatggaaagt
1081 gt.tttggccc agcttgataa ctatggacag caagaacttg cggatct.ttt
tgtgaactat
1141 aatgtaaaat ctxccattac tggaaatgat ctatcccctc cagtgtcttt
taacttaatg
1201 ttcaagactt tcattgggcc tggaggaaac atgcctgggt acttgagacc
agaaactgca
1261 cagggga111 tct;t.gaa111 caaacgac1.1 11ggag11ca accaaggaaa
gttgcctttt
1321 gctgctgccc agattggaaa ttcttttaga aatgagatct cccctcgatc
tggactgatc
1381 agagtcagag aattcacaat ggcagaaatt gagcactttg tagatcccag
tgagaaagac
1441 caccccaagt tccagaatgt ggcagacctt caccttta t. tgtattcagc
aaaagcccag
1501 gtcagcggac agtccgctcg gaaaatgcgc ctgggagatg ctgttgaaca
gggtgtgatt tattaggcta tttcattggc cgcatctacc tctacctcac
gaaggttgga
1621 i ataaactccg cttccggcag cacatggaga atgagatggc
ccattatgcc
1681 t gggatgcaga atccaaaaca tcctacggtt ggattgagat
tgttggatgt
1741 c cctgttatga cctctcctgt catgcacgag ccaccaaagt
cccacttgta
1801 c c ctgaaaga acccaaaaca gtcaat.gttg ttcagtttga
acccagtaag
1861 c gtaaggcata taagaaggat gcaaaactgg tgatggagta
tcttgccatt
1921 t gctacattac agaaatggag atgctgctga atgagaaagg
ggaattcaca
1981 i aagggaaaac atttcagtta acaaaagaca tgatcaatgt
gaagagattc
2041 c tatatgtgga agaagttgtt ccgaatgtaa ttgaaccttc
cttcggcctg
2101 c tgtatacggt atttgaacat acattccatg tacgagaagg
agatgaacag
2161 a tcagtttccc tgctgtagtt getccattea aatgttccgt
cctcccactg
2221 i aggagttcat gccatttgtc aaggaattat cggaagccct
gaccaggcat
2281 acaaagtaga cgattcctct gggtcaatcg gaaggegcta
tgccaggact
2341 c gcgtggcttt tggtgtcacc attgactttg acacagtgaa
caagaccccc
2401 c ctctgaggga ccgtgact ca atgeggcaga taagagcaga
gatctctgag
2461 c tagtccaaga cctagccaat ggcaacatca catgggctga
tgtggaggcc
tgtttgaagg gcaagagact ggtaaaaaag agacaatcga
The amino acid sequence of a human GiyRS is shown below (SEQ ID
NO: 2), Also contemplated herein are GlyRS proteins having sequences that have at least
70%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or more sequence identity to SEQ ID NO: 2.
MDGAGAEEVLAPLRLAVRQQGDLVRKLKEDKAPQVDVDKAVAELKARKRVLEAKELA LQPKDDIVDRAKMEDTLKRRFFYDQAFAIYGGVSGLYDFGPVGCALKNNIIQTWRQH FIQEEQILEIDCTMLTPEPVLKTSGHVDKFADFMVKDVKNGECFRADHLLKAHLQKL MSDKKCSYEKKSEMESVLAQLDKYGQQELADLFVKYNVKSPITGNDLSPPVSFNLMF KTFIGPGGNMPGYLRPETAQGIFLNFKRLLEFNQGKLPFAAAQIGNSFRNEISPRSG LTRVREFTMAEIEHFVDPSEKDHPKFQNVADLHLYLYSAKA.QVSGQSARKMRLGD.AV EQGVINNTVLGYFIGRIYLYLTKVGISPDKLRFRQHMENEMAHYACDCWDAESK SY GWIEIVGCADRSCYDLSCHARATKVPLVAEKPLKEPKTVNWQFEPSKGAIGKAYKK DAKLVMEYLAICDECYITEMEMLLKEKGEFTIETEGKTFQLTKDMINVKRFQKTLYV EEWPNVIEPSFGLGRIMYTVFEHTFHVREGDEQRTFFSFPAWAPFKCSVLPLSQN QEFMPFVKELSEALTRHGVSHKVDDSSGSIGRRYARTDEIGVAFGVTIDFDTVNKTP HTATLRDRDSMRQIRAEISELPSTVQDLANGNITWADVEARYPLFEGQETGKKE IE E [0078] GlyRS proteins suitable for the embodiments of the present disclosure may be produced with recombinant DNA technology in various host cells. For example, expression vectors capable of expressing eukaryotic proteins (e.g., plasmid pcDNA6) may be used to express the recombinant GlyRS proteins. In some embodiments, the host cells can be bacterial, fungal, plant, yeast, insect or mammalian cells. The term host cell includes both the cells, progeny of the ceils and protoplasts created from the cells that are used to produce a GlyRS according to the disclosure. In some embodiments, the host cells are prokaryotic ceils, for example bacteria host ceils.
[0079] As a non-limiting example, to produce the GlyRS protein with the recombinant DNA technology, a DNA construct comprising nucleic acid encoding the amino acid sequence of the designated GlyRS can be constmcted and transferred into, for example, an E. coli host cell. The vector may be any vector which when introduced into an E. coli host ceil can be integrated into the host cell genome and can be replicated. The nucleic acid encoding the GlyRS can be operably linked to a suitable promoter, which shows transcriptional activity in E. coli host ceil. The promoter may be derived from genes encoding proteins either homologous or heterologous to the host cell. As used herein, an "inducible promoter" may refer to a promoter that is active under environmental or d evel opmental regul ati on .
[0080] In some embodiments, the GlyRS coding sequence can be operably linked to a signal sequence. In some embodiments, the expression vector may also include a termination sequence. In one embodiment, the termination sequence and the promoter sequence can be derived from the same source. In another embodiment, the termination sequence can be homologous to the host cell ,
[0081] In some embodiments, the expression vector may include one or more selectable markers. Examples of representative selectable markers include ones that confer antimicrobial resistance (e.g., hygromycin and phleomycin). In some embodiments, nutritional selective markers including those markers known in the art as amdS, argB, and pyr4, can be used as the selectable marker.
[0082] An expression vector comprising a DNA construct with a polynucleotide encoding the GlyRS may be any vector which is capable of replicating autonomously in a given host organism or of integrating into the DNA of the host. In some embodiments, the expression vector can be a plasmid or a viral construct.
[0083] In some embodiments, two types of expression vectors for obtaining expression of genes are contemplated. For example, the first expression vector may comprise DNA sequences in which the promoter, GlyRS-coding region, and terminator all originate from the gene to be expressed. In some embodiments, gene truncation can be obtained by deleting undesired DNA sequences (e.g., DNA encoding unwanted domains) to leave the domain to be expressed under control of its own transcriptional and translational regulatory sequences. The second type of expression vector may be preassembled and contains sequences needed for high-level transcription and a selectable marker. In some embodiments, the coding region for the GARS gene or part thereof can be inserted into this general-purpose expression vector such that it is under the transcriptional control of the expression construct promoter and terminator sequences. In some embodiments, genes or part thereof may be inserted downstream of a strong promoter.
[0084] Methods used to ligate the DNA construct comprising a polynucleotide encoding the GlyRS, a promoter, a terminator and other sequences and to insert them into a suitable vector are well known in the art. Linking can be generally accomplished by ligation at convenient restriction sites. If such sites do not exist, the synthetic oligonucleotide linkers are used in accordance with conventional practice (Bennett & Lasure, More Gene Manipulations In Fungi, Academic Press, San Diego (1991) pp 70-76), Additionally, vectors can be constructed using known recombination techniques (e.g., Invitrogen Life Technologies, Gateway Technology).
[0085] Introduction of a DNA construct or vector into a host cell includes techniques such as transformation; electroporation; nuclear microinjection; transduction; transfection, (e.g., lipofection mediated and DEAE-Dextrin mediated transfection); incubation with calcium phosphate DNA precipitate; high velocity bombardment with DNA- coated microprojectiles; and protoplast fusion. General transformation techniques are known in the art (see, e.g., Campbell et al., (1989) Curr. Genet 1.6:53-56).
[0086] In some embodiments, genetically stable transformants can be constructed with vector systems whereby the nucleic acid encoding GlyRS is stably integrated into a host strain chromosome. Transformants can then be purified by known techniques. Methods of Inhibiting Neddylation
[0087] NEDD8 is an 81-amino acid protein with 9 kDa relative molecular mass and is 60% identical and 80% homologous to ubiquitin. NEDD8 has a dedicated El- activating enzyme (AppBpl/UBA3, or NAE) and E2-conjugating enzymes (UBC12, UBE2F) and is essential for the enzymatic activity of the CRL family of E3 ligases, through conjugation to the cullin scaffold. Other components of the neddvlation pathway include DEN1 which processes NEDD8 to its mature, 76-amino acid form, and the COP9 signalosome complex, which is responsible for removing NEDD8 from cullin proteins. CANDl (cullin-associated and neddylation-dissociated) is an additional component that regulates CRL complex assembly by binding to the cullin in the absence of NEDD8 activation.
[0088] Neddylation is a posttranslational modification that controls cell cycle and proliferation by conjugating the ubiquitin-like protein NEDD8 to specific targets. It is hereby disclosed that GlyRS plays a critical role in neddylation. In human ceils, knockdown of GlyRS, but not a different tRNA synthetase, decreases the global level of neddylation and delays cell cycle progression. This function of GlyRS is achieved through direct interactions with multiple components of the neddylation pathway, including NEDD8, El , and E2. GlyRS can bind to the APPBP1 subunit of El to capture and protect the activated E2 (NEDD8~conjugated Ubcl2) before it reaches a downstream target.
[0089] Some embodiments disclosed herein provide methods of reducing or inhibiting neddylation in a cell. As used herein, inhibition of neddylation includes partially or fully blocks or abolished neddylation in a cell or a cell population. For example, the inhibition can reduce 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 60%, 70%, 80%, 90%, 95%, 99%o, or a range between any two of these values, of neddylation in the cell or the cell population. In some embodiments, the inhibition can reduce about 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, or a range between any two of these values, of neddylation in the cell or the ceil population. In some embodiments, the neddylation in the cell or the cell population is completely abolished. In some embodiments, the methods comprise contacting the cell with a GlyRS inhibitor, wherein the level of neddylation is decreased in the cell. For example, the level of neddylation can be decreased to, or to about 99%, 95%, 90%, 80%, 70%, 60%, 50%, 45%, 40%, 35%, 30%, 25%, 20%, 15%, 10%, 5%, 1%, or a range between any two of these values, of the level of neddylation in cell(s) not treated with the GlyRS inhibitor. Neddylation, in some embodiments, refers to the conjugation of NEDD 8 to components of the neddylation pathway, e.g., the El enzyme, the E2 enzyme, or the E3 ligases. In some embodiments, neddylation may refer to the conjugation of NEDD 8 to a cullin protein in the E3 ligases, e.g., a CRL. The decrease of neddylation may occur to individual components of the neddylation pathway, or at a global level. In some embodiments, the level of neddylation may be reduced by 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98%, 99%, or more.
[0090] It would be appreciated that the GlyRS inhibitor may decrease the level of neddylation in the cell through a variety of mechanisms, for example, by interfering the binding between the GlyRS protein and NEDD 8, by interfering the binding between the GlyRS protein and an E2 enzyme, such as Ubcl2, by interfering the binding between the GlyRS protein and NEDD8-conjugated Ubc 2, by interfering the binding between the GlyRS protein and an El enzyme, such as APPBP1/UBA3 heterodimer, by directly targeting El enzyme (given the similarity in the first step reaction between GlyRS and El ), or any combination thereof. In some embodiments, the GlyRS inhibitor may inhibit the activity or decrease the level of a component of the neddylation pathway, for example, the level of NEDD 8 -conjugated Ubcl2, the activity of the Ubcl2 enzyme, the protein level of the Ubcl2 enzyme, or any combination thereof. In some embodiments, the GlyRS inhibitor binds to both GlyRS and El enzyme to decrease neddylation.
[0091] It will also be appreciated by one of skill in the art that inhibiting neddylation by the methods disclosed herein may result in the inhibition of ubiquitination of one or more of the E3 substrates in a cell, such as ATF4, CCNEl, CDC25, CDKN1A, CDKN1B, CTNNB 1 , DAPK1 , Emil, FancM, HIF2A, IRS1 , JUN, MCL1 , NRF2, ORC1 L, PDCD4, POLR2A, SETD8, SNAI3, USP18, etc.
GlyRS Inhibitors
[0092] As discussed above, the term "GlyRS inhibitor" is used herein in a broad sense and includes any molecule that partially or fully blocks, inhibits or neutralizes a biological activity mediated by GlyRS. In some embodiments, it can prevent the activation of GlyRS. The term "GlyRS inhibitor" also includes any molecule that abolishes or reduces the function or expression of GlyRS.
[0093] The method by which GlyRS is inhibited is not limited in any way. In some embodiments, the GlyRS inhibitor can act directly on GlyRS, for example by binding to GlyRS, to prevent or reduce activation of GlyRS. In some embodiments, the GlyRS inhibitor can interfere, preferably abolish or reduce, GlyRS from interacting with a binding partner or a substrate, such a component of the neddylation pathway . In some embodiments, the GlyRS inhibitor can modulate the level of GlyRS gene expression, for example, inhibiting or reducing the transcription of GlyRS gene. In some embodiments, the GlyRS inhibitor can modulate the levels of GlyRS protein in cells by, for example, inhibiting or reducing the translation of GlyRS mRNA, or increasing the degradation of GlyRS mRNA or GiyRS protein. In some embodiments, the GlyRS inhibitor can block the interaction of GlyRS with EDD8 and/or NEDD8-conjugated Ubcl2.
[0094] As disclosed herein, a GlyRS can perform function in various biological processes, for example aminoacyiation and neddylation. As used herein, a compound is considered to be a GlyRS inhibitor if the compound can reduce or inhibit one or more biological activities of a GlyRS. For example, a GiyRS inhibitor may reduce or inhibit GlyRS functions in both aminoacyiation and neddylation. In some embodiments, the GlyRS inhibitor only reduce or inhibit GlyRS function in neddylation. In some embodiments, the GlyRS inhibitor does not significantly inhibit GlyRS function in aminoacyiation. For example, a GiyRS inhibitor does not significantly inhibit GlyRS function in aminoacyiation if the GlyRS inhibitor can at most reduce the activity of the GlyRS function in aminoacyiation by, or by about, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 10%, 50%, 1%, or a range between any two of these values. In some embodiments, the GlyRS inhibitor reduces the activity of the GlyRS function in aminoacyiation by, or by about, 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 10%, 50%, 1%, or a range between any two of these values.
[0095] The types of GlyRS inhibitor are not limited in any way. GlyRS inhibitors include, for example, small molecules, nucleic acids, antibodies, peptides, or any combination thereof. In some embodiments, the GlyRS inhibitor can be a small molecule that binds to GiyRS. In some embodiments, the GlyRS inhibitor can be a molecule that blocks interaction of GlyRS and it binding partner. In some embodiments, the GlyRS inhibitor is a nucleic acid, for example, an anti-GlyRS small-hairpin RNA (shRNA) or an GlyRS anti-sense RNA.
[0096] Some embodiments of the present disclosure therefore include, for example, inhibitors of GlyRS function, for example, its interactions with components of the neddvlation pathway. The GlyRS inhibitors can be used, for example, in any of the methods described herein. Any agent that may prevent or reduce the interaction between the GlyRS protein and NEDD8, El , and E2, or eliminate or reduce the level of GlyRS protein expression, is contemplated by the present disclosure. A reduction refers to at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99% or more of the interaction between the GlyRS protein and NEDD8, El, and E2, or of the GlyRS protein expression level in a cell. Interaction refers the binding of NEDD8, El, and E2 with the GlyRS protein, which may lead to a conformational change to the GlyRS protein and/or the NEDD8, El, and E2 protein.
[0097] The ability of a molecule to inhibit GlyRS activity can be measured using assays that are known in the art. For example and without limitation, GlyRS inhibitors can be identified using biolayer interferometry. Briefly, in biolayer interferometry, a molecule can be examined for the ability to affect the binding between a GlyRS protein and its binding partner. For example, GlyRS proteins are immobilized on either anti-GST or Ni-NTA sensor tips in 96-weil plates. A binding partner, e.g., NEDD8, is added to the buffer. Candidate GlyRS molecules are added to the buffer and the dissociation constants are measured. GlyRS inhibitors that interfere with the binding between the GlyRS protein and NEDD8 can be identified based on the change in the dissociation constants One of skill in the art will be able to select the appropriate assays and reaction conditions based on the particular circumstances.
[0098] Some embodiments disclosed herein provide small molecule compounds that inhibit the function of a GlyRS protein. Non-limiting examples of inhibitory small molecule compounds include ones that exhibit binding specificity for at least one region of the GlyRS protein that is involved in its interaction with a component of the neddvlation pathway, and/or its stability/degradation in a cell.
[0099] As used herein, the term "small molecule" refers to an organic compound that is of synthetic or biological origin (biomolecule), but is typically not a polymer. [0100] The small molecule compounds disclosed herein may bind to a region of the GlyRS protein involved in its interaction with a component of the neddylation pathway, e.g., the catalytic domain, the ABD domain, or a combination thereof. For example, the small molecule compounds may interfere with the interaction between GlyRS and NEDD8, El, and/or E2. In some embodiments, the small molecule compounds bind to a sequence comprising amino acids 84-93 of SEQ ID NO: 2. In some embodiments, the small molecule compounds bind to one or more of amino acids 84-93 of SEQ ID NO: 2. In some embodiments, the small molecule compounds bind to a sequence comprising amino acids 232-238 of SEQ ID NO: 2. In some embodiments, the small molecule compounds bind to one or more of amino acids 232-238 of SEQ ID NO: 2.
[0101] One non-limiting example of the GlyRS inhibitor is glycylsulfamoyladenosin ly-SA). The structure of this compound is as follows.
Figure imgf000031_0001
[0102] Gly-SA is an analogue of the Gly-AMP reaction intermediate and inhibits GlyRS catalytic activity. Estimates of the potency of inhibition are obtained by performing enzyme assays in the presence of a range of inhibitor concentrations, and fitting the effect of inhibitor concentration on enzyme velocity to a four parameter logistic function that allows calculation of an IC50 (the inhibitor concentration at which GlyRS activity is reduced by half). This parameter is directly related to the dissociation constant for inhibitor binding (Kj or Kd) and has a value of around 2.4 mM for Gly-SA when tested against the S. aureus GlyRS. Binding of Gly-SA to GlyRS can also be measured directly using stopped-fiow fluorescence techniques because enzyme:inhibitor binary complex has around 5% higher tryptophan fluorescence than the free enzyme.
[0103] Also disclosed herein are Gly-SA derivatives that function as GlyRS inhibitors. Non-limiting examples of Gly-SA derivatives include Compound-l, Compound-2, and Compound-3 (shown below and in Figure 29) and the analogues described for aaSA in Van de Vijver et al. (2008) J. Med. Chem. 51 :3020-3029 (the content of which is incorporated by reference herein in its entirety). Moreover, chemical modifications for various aaRS inhibitors having similar chemical structure with Gly-SA have been described in, for example, Brown et al. (2000) Biochemistry 39(20):6003-6011, Lee et al. (2003) Bioorganic & Medicinal Chemistry Letters 12: 1087-1092, Bernier et al. (2005) Bioorganic & Medicinal Chemistry 13 :69-75, and Balg et al. (2007) Bioorganic & Medicinal Chemistry 15:295-304. One of skill in the art will appreciate that those chemical modifications can be introduced to Gly-SA in some embodiments to produce Gly-SA derivatives,
Compound^
Figure imgf000032_0001
Oompottmi-S
Figure imgf000032_0002
[0104] Also disclosed herein are si NA and shRNA against GlyRS. Double- stranded RNA (dsRNA) directs the sequence-specific degradation of niRNA through a process known as RNA interference (RNAi). The process is known to occur in a wide variety of organisms, including embryos of mammals and other vertebrates. The use of these dsRNAs (or recombinant! y produced or chemically synthesized oligonucleotides of the same or similar nature) enables the targeting of the GlyRS mR As, for example, GARS in humans, for degradation in mammalian cells. Use of long dsRNAs in mammalian cells to elicit RNAi is not desired in some embodiments because of the deleterious effects of the interferon response. Specific targeting of a particular gene function, which is possible with short oligonucleotides (e.g., 19-23 nt RNA), is useful in functional genomic and therapeutic applications. [0105] Some embodiments disclosed herein provide small interfering RNA (siRNA) sequences, RNA interfering vectors, and RNA interfering lentiviruses that are directed at a GlyRS gene, e.g., the human GARS gene. In some embodiments, provided are isolated double-stranded ribonucleic acid (dsRNA) molecules that inhibit expression of a GlyRS protein, wherein a first strand of the dsRNA is substantially identical to at least 19 consecutive nucleotides of the GlyRS gene, and a second strand of the dsRNA is substantially complementary to the first strand.
[0106] In some embodiments, the dsRNA molecules are small hairpin RNA (shRNA) molecules. The following is a list of exemplar}' shRNAs targeting human GARS gene:
shGARS2277 5'-GCATGGAGTATCTCACAAAGT (SEQ ID NO:3) shGARS-696 5' GCCCAAAGATGATATTGTAGA (SEQ ID NO: 4) shGARS-1324 5' GCTGCCCAGATTGGAAATTCT (SEQ ID NO:5)
[0107] In some embodiments, the shRNA may comprise a sense fragment, which comprises a nucleotide sequence substantially identical to a target sequence in the GlyRS gene, and an antisense fragment, wherein the sense and antisense fragments are separated by a loop fragment, wherein the loop fragments may comprise a sequence selected from the group consisting of UUCAAGAGA, AUG, CCC, UUCG, CCACC, CTCGAG, AAGCUU and CCACACC.
[0108] In some embodiments, an siRNA target sequence may be designed on the basis of the human GARS gene, preferably 15 to 27, more preferably 19 to 23, and optimally 19, 20 or 21, consecutive bases.
[0109] The following is a list of non-limiting exemplary siRNAs targeting human GARS gene:
GGCGT TACAGCCCAAAGAT (SEQ ID O:6)
G C C C AAAGAT G T AT T G T A (SEQ ID NO:7)
C C T G GAG G GAG C AC T T T AT (SEQ ID NO: 8)
GCTGCCCAGAT TGGAAAT T (SEQ ID NO: 9)
GGAGCAAT T GG TAAG GCAT (SEQ ID NO: 10)
G CAAT T G G T AAG G CAT AT A (SEQ ID NO: 11) C C G AAT G AA GAAC C T (SEQ ID NO: 12)
G C C T G G G TAG GAT CAT G T A (SEQ ID NO: 13)
GGAGAT GAACAGAGAACAT (SEQ ID NO : 14)
G CAT G GAG TAT C T C AC AAA (SEQ ID NO: 15)
C CAGAAT G T G G C AGAC C T T (SEQ ID NO: 16)
C C T G G G TAG GAT C AT G T AT (SEQ ID NO: 17)
G GC C C AG C T T G AT AAC TAT (SEQ ID NO: 18)
G GG T AC T T GAGAC C AGAAA (SI ) ID NO: 1 0)
G G C AG AAA.T T GAG C AC T T T (SEsQ ID NO:20)
G GAAGAAG T T G T T C C GAAT (SEQ ID NO:21)
G GAT CAT G T AT AC G G T AT T (SEQ ID NO:22)
G G CA G GAG TAT C T C AC AA (SEsQ ID NO:23)
CCTATGCTTTGAAGGTTCT (SEQ ID NO:24)
GCTTTGAAGGTTCTCGTGT (SEQ ID NO:25)
T CAGAG C T G T G T C C C T G A (SEQ ID NO:26)
GCAAATCTGTTCGCTCGCA (SEQ ID NO: 27)
GCG GCGAT T T C T CAT GC T (SEQ ID NO:28)
G C GAT T T CAT CAT G C T C C G (SEQ ID NO: 29)
C C C AAAGA.T GAT AT T G T AG (SEQ ID NO: 30)
GCTGTGCTTT G AAGAAC AA (SEQ ID NO:31)
GCACTTTATCCAAGAGGAA (SEQ ID NO:32)
T CCCAT TA.C T GGAAAT G T (SEsQ IE) NO: 3 )
G C T AT T T CAT T G G C C G CAT (SEQ ID NO: 34)
G CAT C T AC C T C T AC C T C AC (SEQ ID NO:35)
[0110] Some embodiments disclosed herein provide siRNA molecules that mediate RNAi. The siRNA molecules disclosed herein can also comprise a 3' hydroxyl group. The siRNA molecules can be single-stranded or double stranded, such molecules can be blunt ended or comprise overhanging ends (e.g., 5', 3'). In some embodiments, the siRNA molecule is double stranded and either blunt ended or comprises overhanging ends. [0111] In some embodiments, at least one strand of the siRNA molecule has a 3! overhang from about 1 to about 6 nucleotides (e.g., pyrimidine nucleotides, purine nucleotides) in length. In some embodiments, the 3! overhang is from about 1 to about 5 nucleotides, from about J to about 3 nucleotides and from about 2 to about 4 nucleotides in length. In one embodiment, the siRNA molecule is double stranded, one strand has a 3! overhang and the other strand can be blunt-ended or have an overhang. In the embodiment in which the siRNA molecule can be double stranded and both strands comprise an overhang, the length of the overhangs may be the same or different for each strand. In a particular embodiment, the siRNA of the present invention comprises 21 nucleotide strands which are paired and which have overhangs of from about 1 to about 3, particularly about 2, nucleotides on both 3' ends of the siRNA. In order to further enhance the stability of the RNA of the present invention, the 3' overhangs can be stabilized against degradation. In one embodiment, the RNA is stabilized by including purine nucleotides, such as adenosine or guanosine nucleotides. Alternatively, substitution of pyrimidine nucleotides by modified analogues, e.g., substitution of uridine 2 nucleotide 3' overhangs by 2'-deoxythymidine is tolerated and does not affect the efficiency of RNAi. The absence of a 2' hydroxyl significantly enhances the nuclease resistance of the overhang in tissue culture medium.
[0112] The siRNA molecules disclosed herein can be obtained using a number of techniques known to those of skill in the art. For example, the siRNA can be chemically synthesized or recombinant! y produced using methods known in the art. The siRNA can also be obtained using an in vitro system. The in vitro system can also be used to obtain siRNA of about 19 to about 23 nucleotides in length which mediates RNA interference of the mRNA of the GlyRS gene.
[0113] The method of obtaining the siRNA sequence using the in vitro system can further comprise isolating the RNA sequence from the combination. The siRNA molecules can be isolated using a number of techniques known to those of skill in the art. For example, gel electrophoresis can be used to separate siRNA from the combination, gel slices comprising the RNA sequences removed and RNAs eluted from the gel slices. Alternatively, non-denaturing methods, such as non-denaturing column chromatography, can be used to isolate the RNA produced. In addition, chromatography (e.g., size exclusion chromatography), glycerol gradient centrifugation, affinity purification with antibody can be used to isolate siRNAs. The RNA-protein complex isolated from the in vitro system can also be used directly in the methods described herein (e.g., method of mediating RNAi of mRNA of the GiyRS gene).
[0114] The siRNAs described herein can be used in a variety of ways. For example, the siRNA molecules can be used to mediate RNA interference of mRNA of a gene in a cell or organism. In a specific embodiment, the siRNA is introduced into human cells or a human in order to mediate RNA interference in the cells or in cells in the individual, such as to prevent or treat a disease or undesirable condition, hi this method, a gene (or genes) that cause or contribute to the disease or undesirable condition is targeted and the corresponding mRNA (the transcriptional product of the targeted gene) is degraded by RNAi. In this embodiment, an siRNA that targets the corresponding mRNA (the mRNA of the targeted gene) for degradation is introduced into the cell or organism. The ceil or organism is maintained under conditions under which degradation of the corresponding mRN A occurs, thereby mediating RNA interference of the mRNA of the gene in the cell or organism. In the event that the siRNA is introduced into a cell in which RNAi does not normally occur, the factors needed to mediate RNAi are introduced into such a cell or the expression of the needed factors is induced in such a cell. Alternatively, siRNA produced by other methods (e.g., chemical synthesis, recombinant DNA production) to have a composition the same as or sufficiently similar to an siRNA known to mediate RNAi can be similarly used to mediate RNAi. Such siRNAs can be altered by addition, deletion, substitution or modification of one or more nucleotides and/or can comprise non-nucleotide materials.
[0115] At the same time, disclosed herein is a GlyRS RNAi lentivirus and the preparation and application thereof. A nucleic acid construct that expresses the above- described siRNA may be constructed by means of gene cloning and packaged with a lentivirus that expresses the above-described siRNA, Cell experiments prove that the above- described siRNA sequence can specifically silence the expression of endogenous GlyRS genes in cells.
[0116] In some embodiments, a DNA sequence encoding the above-described siRNA may be contained in a lentivirus vector. In some embodiments, the lentivirus vector may further comprise a promoter sequence. In some embodiments, the lentivirus vector may further comprise a nucleotide sequence encoding a detectable marker in the ceil, wherein the detectable marker may be a green fluorescent protein (GFP). In some embodiments, the lentivirus vector may be selected from the group consisting of pLentiLox 3,7, pLKO. l -puro, pL O. l-CMV-tGFP, pL O. l-puro-CMV-tGFP, pLKO. l-CMV-Neo, pLKO. l -Neo, pLKO. 1 -Neo-CMV-tGFP, pLKO.1-puro-CMV-TagCFP, pLKO. 1 -puro-C V-TagYFP, pLKO.1 -puro-CMV-TagRFP, pLKO.1 -puro-CMV-TagFP635, pLKO.1 -puro-UbC- TurboGFP, pLKO. l-puro-UbC-TagFP635, pLKO-puro-IPTG-lxLacO, pLKO-puro-IPTG- 3xLacO, pLPl , pLP2, pLP/VSV-G, pENTR/U6, pLenti6/BLOCK-iT-DEST, pLenti6- GW./U6 aminshrna, pcDNAl ,2/V5-GW/iacZ, pLenti6.2/N-Lumio/V5-DEST, pGCSIL-GFP and Lenti6.2/N-Lumio/V5-GW/lacZ. In some embodiments, the siRNA lentiviruses designed for GlyRS stably and specifically lower GlyRS expression and effectively inhibit neddylation.
Methods of Identifying Inhibitors of Neddylation
[0117] Disclosed herein are methods of identifying an inhibitor of neddylation. The method, in some embodiments, comprises: providing one or more compounds; testing the one or more compounds for their ability to reduce or inhibit neddylation. In some embodiments, the compounds can be tested for their ability to reduce the binding between a GlyRS protein and NEDD8-conjugated Ubcl2. In some embodiments, the methods comprise identifying one or more compounds that have the ability to reduce or inhibit the binding between the GlyRS protein and NEDD8-conjugated Ubcl 2 as inhibitors of neddylation. The method can further comprise testing the one or more compounds for their ability to reduce or inhibit aminoacylation activity of the GlyRS protein.
[0118] The potential inhibitory or binding effect of a chemical compound on GlyRS may be analyzed prior to its actual synthesis and testing by the use of computer modelling techniques. If the theoretical structure of the given compound suggests insufficient interaction and association between it and GlyRS, synthesis and testing of the compound is obviated. However, if computer modelling indicates a strong interaction, the molecule may then be synthesized and tested for its ability to bind to GlyRS and inhibit using a suitable assay. In this manner, synthesis of inoperative compounds may be avoided. An inhibitory or other binding compound of GlyRS may be computationally evaluated and designed by means of a series of steps in which chemical entities or fragments are screened and selected for their ability to associate with the individual binding pockets or other areas of GlyRS.
[0119] One of skill in the art may use various methods to test chemical entities or fragments for their ability to associate with GlyRS and more particularly with the individual binding pockets of the GlyRS active site or accessory binding site. PCT Patent Publication No, WO 2000058345 Al describes design of GlyRS binding compounds using computer modeling, the content of which is hereby incorporated by reference in its entirety. In some embodiments, a known GlyRS inhibitor, such as Gly-SA, may be used as a starting point for designing derivative compounds that inhibit GlyRS.
[0120] In some embodiments, small molecule inhibitors of neddyiation may be identified using standard techniques. For example, candidate compounds may be used in binding assays using conventional formats to screen inhibitors of interaction between GlyRS and a component of the neddyiation pathway. One particularly suitable assay format includes the enzyme-linked immunosorbent assay (ELISA). Other assay formats may be used; these assay formats are not a limitation on the present disclosure.
[0121] In another aspect, the structure of the GlyRS protein permits the design and identification of synthetic compounds and/or other molecules which are characterized by the conformation of the GlyRS protein. Using known computer systems, the coordinates of the GlyRS protein staicture may be provided in machine readable form, the test compounds designed and/or screened and their conformations superimposed on the structure of the GlyRS protein. Subsequently, suitable candidates identified as above may be screened for the desired GlyRS protein inhibitors bioactivity, stability, and the like. Once identified and screened for biological activity, these inhibitors may be used therapeutically or prophylacticaily to block GlyRS protein activity, and thus, neddyiation in a cell.
[0122] In some embodiments, the identified inhibitor of neddyiation binds to the catalytic domain of the GlyRS protein. The GlyRS protein can be, for example, a mammalian GlyRS protein, particularly a human GlyRS protein. In some embodiments, the human GlyRS protein comprises an amino acid sequence having at least 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 2.
[0123] In some embodiments, the compound that inhibits binding between the GlyRS protein and NEDD8-conjugated Ubcl2 binds to a sequence comprising amino acids 84-93 of SEQ ID NO: 2. In some embodiments, the compound that inhibits binding between the GlyRS protein and NEDD8-conjugated Ubcl 2 binds to one or more of amino acids 84-93 of SEQ ID NO: 2. In some embodiments, the compound that inhibits binding between the GlyRS protein and NEDD8-conjugated Ubcl2 binds to a sequence comprising amino acids 232-238 of SEQ ID NO: 2. In some embodiments, the compound that inhibits binding between the GlyRS protein and NEDD 8 -conjugated Ubc-12 binds to one or more of amino acids 232-238 of SEQ ID NO: 2,
Pharmaceutical Compositions and Methods of Admi istration
[0124] Some embodiments disclosed herein provide pharmaceutical compositions comprising one or more GlyRS inhibitors and a pharmaceutically acceptable excipient for the treatment of cancers. The GlyRS inhibitors can be any of the GlyRS inhibitors disclosed herein. As disclosed herein, the GlyRS inhibitors may be small molecules, nucleic acids, antibodies, peptides, or any combination thereof. In some embodiments, the GlyRS inhibitors may be small molecule compounds that inhibit or reduce the interaction between a GlyRS protein and components of the neddvlation pathway, or isolated double- stranded dsRNA molecules that inhibit or reduce expression of a GlyRS protein. In some embodiments, the GlyRS inhibitor reduces or inhibits GlyRS functions in aminoacylation and neddvlation. In some embodiments, the GlyRS inhibitor does not significantly reduce or inhibit GlyRS function in aminoacylation. In some embodiments, the GlyRS inhibitor only reduces or inhibits GlyRS function in neddvlation.
[0125] In addition to the one or more GlyRS inhibitors, the pharmaceutical compositions disclosed herein can comprise one or more therapeutic agents. Non-limiting examples of therapeutic agents include chemotherapeutic agents, cancer drugs, or prodrugs or pharmaceutically acceptable salts thereof. The chemotherapeutic agents can be, for example, AZ-23, BMS-754807, bosutinib, cabozantinib, ceritinib, crizotinib, entrectinib, foretmib, GNF 5837, GW441756, imatinib mesylate, K252a, l .OXO- 10 i , MGCD516, nilotinib hydrochloride monohydrate, NVP-TAE684, PF-06463922, rebastinib, staurosporine, sorafenib tosylate, sunitinib malate, and TSR-01 1. [0089] Also provided are pharmaceutically acceptable prodrugs of the pharmaceutical compositions, and treatment methods employing such pharmaceutically acceptable prodrugs. The term "prodrug" means a precursor of a designated compound that, following administration to a subject, yields the compound in vivo via a chemical or physiological process such as solvolysis or enzymatic cleavage, or under physiological conditions (e.g., a prodaig on being brought to physiological pH is converted to the agent). A "pharmaceutically acceptable prodrug" is a prodrug that is non-toxic, biologically tolerable, and otherwise biologically suitable for administration to the subject. Illustrative procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in Bundgaard, Design of Prodrugs (Elsevier Press, 1985).
[0126] Also provided are pharmaceutically active metabolites of the pharmaceutical compositions, and uses of such metabolites in the methods of the invention. A "pharmaceutically active metabolite" means a pharmacologically active product of metabolism in the body of a compound or salt thereof. Prodaigs and active metabolites of a compound may be determined using routine techniques known or available in the art. See, e.g., Bertolini et al., J. Med. Chem. 1997, 40, 2011-2016; Shan et al., J. Pharm. Sci. 1997, 86 (7), 765-767; Bagshawe, Drug Dev. Res. 1995, 34, 220-230; Bodor, Adv. Drug Res. 1984, 13, 255-331 ; Bundgaard, Design of Prodrugs (Elsevier Press, 1985), and Larsen, Design and Application of Prodrugs, Drug Design and Development ( rogsgaard-Larsen et al., eds., Harwood Academic Publishers, 1991).
[0127] Any suitable formulation of the compounds described herein can be prepared. See, generally, Remington's Pharmaceutical Sciences, (2000) Hoover, J. E. editor, 20th edition, Lippincott Williams and Wilkins Publishing Company, Easton, Pa., pages 780- 857. A formulation is selected to be suitable for an appropriate route of administration. Some routes of administration are oral, parenteral, by inhalation, topical, rectal, nasal, buccal, vaginal, via an implanted reservoir, or other drug administration methods. In cases where compounds are sufficiently basic or acidic to form stable nontoxic acid or base salts, administration of the compounds as salts may be appropriate. Examples of pharmaceutically acceptable salts are organic acid addition salts formed with acids that form a physiological acceptable anion, for example, tosylate, methanesulfonate, acetate, citrate, malonate, tartarate, succinate, benzoate, ascorbate, a-ketoglutarate, and α-glycerophosphate. Suitable inorganic salts may also be formed, including hydrochloride, sulfate, nitrate, bicarbonate, and carbonate salts. Pharmaceutically acceptable salts are obtained using standard procedures well known in the art, for example, by a sufficiently basic compound such as an amine with a suitable acid, affording a physiologically acceptable anion. Alkali metal (e.g., sodium, potassium or lithium) or alkaline earth metal (e.g., calcium) salts of carboxylic acids also are made.
[0128] Where contemplated compounds are administered in a pharmacological composition, it is contemplated that the compounds can be formulated in admixture with a pharmaceutically acceptable excipient and/or carrier. For example, contemplated compounds can be administered orally as neutral compounds or as pharmaceutically acceptable salts, or intravenously in a physiological saline solution. Conventional buffers such as phosphates, bicarbonates or citrates can be used for this purpose. Of course, one of ordinary skill in the art may modify the formulations within the teachings of the specification to provide numerous formulations for a particular route of administration. In particular, contemplated compounds may be modified to render them more soluble in water or other vehicle, which for example, may be easily accomplished with minor modifications (salt formulation, esterification, etc.) that are well within the ordinary skill in the art. It is also well within the ordinary skill of the art to modify the route of administration and dosage regimen of a particular compound in order to manage the pharmacokinetics of the present compounds for maximum beneficial effect in a patient.
[0129] The pharmaceutical compositions as described herein are generally soluble in organic solvents such as chloroform, dichloromethane, ethyl acetate, ethanol, methanol, isopropanol, acetonitrile, glycerol, N,N-dimethylformamide, AvAr-dimetheylaceatmide, dimethylsulfoxide, or any combination thereof. In one embodiment, the present invention provides formulations prepared by mixing an agent with a pharmaceutically acceptable carrier. In one aspect, the formulation may be prepared using a method comprising: a) dissolving a described agent in a water-soluble organic solvent, a non-ionic solvent, a water- soluble lipid, a cyclodextrin, a vitamin such as tocopherol, a fatty acid, a fatty acid ester, a phospholipid, or a combination thereof, to provide a solution; and b) adding saline or a buffer containing 1-10% carbohydrate solution. In one example, the carbohydrate comprises dextrose. The pharmaceutical compositions obtained using the present methods are stable and useful for animal and clinical applications. [0130] Illustrative examples of water soluble organic solvents for use in the present methods include and are not limited to polyethylene glycol (PEG), alcohols, acetonitrile, N-methyl-2-pyrrolidone, N,N-dimethylformarnide, A V-dimethylacetamide, dimethyl sulfoxide, or a combination thereof. Examples of alcohols include but are not limited to methanol, ethanol, isopropanol, glycerol, or propylene glycol.
[0131] Illustrative examples of water soluble non-ionic surfactants for use in the present methods include and are not limited to CREMOPHOR* EL, polyethylene glycol modified CREMOPHOR* (polyoxyethyleneglyceroltriricinoleat 35), hydrogenated CREMOPHOR® RH40, hydrogenated CREMOPHOR® RH60, PEG-succinate, polysorbate 20, polysorbate 80, SOLUTOL*' HS (polyethylene glycol 660 12-hydroxystearate), sorbitan monooleate, poloxamer, LABRAFIL* (ethoxylated persic oil), LABRASQL® (capryl- caproyl macrogol-8-glyceride), GELUCIRE® (glycerol ester), SOFTIGEN* (PEG 6 caprylic glyceride), glycerin, glycol-polysorbate, or a combination thereof.
[0132] Illustrative examples of water soluble lipids for use in the present methods include but are not limited to vegetable oils, triglycerides, plant oils, or a combination thereof Examples of lipid oils include but are not limited to castor oil, polyoxvl castor oil, corn oil, olive oil, cottonseed oil, peanut oil, peppermint oil, safflower oil, sesame oil, soybean oil, hydrogenated vegetable oil, hydrogenated soybean oil, a triglyceride of coconut oil, palm seed oil, and hydrogenated forms thereof, or a combination thereof.
[0133] Illustrative examples of fatty acids and fatty acid esters for use in the present methods include but are not limited to oleic acid, monoglycerides, diglycerides, a mono- or di -fatty acid ester of PEG, or a combination thereof.
[0134] Illustrative examples of cyclodextrins for use in the present methods include but are not limited to alpha-cyclodextrin, beta-cyclodextrin, hydroxypropyl-beta- cyclodextrin, or sulfobutyl ether-beta-cyclodextrin.
[0135] Illustrative examples of phospholipids for use in the present methods include but are not limited to soy phosphatidylcholine, or distearoyl phosphatidylglycerol, and hydrogenated forms thereof, or a combination thereof.
[0136] One of ordinary skill in the art may modify the formulations within the teachings of the specification to provide numerous formulations for a particular route of administration. In particular, the compounds may be modified to render them more soluble in water or other vehicle. It is also well within the ordinary skill of the art to modify the route of admini stration and dosage regimen of a particular compound in order to manage the pharmacokinetics of the present compounds for maximum beneficial effect in a patient.
[0137] The pharmaceutical compositions comprising GiyRS inhibitors and disclosed herein can be used in combination with a pharmaceutical composition comprising one or more therapeutic agents. As used herein the terms "combination" and "in combination with" mean the administration of a therapeutic agent described herein together with at least one additional pharmaceutical or medicinal agent (e.g., an anti-cancer agent), either sequentially or simultaneously. For example, the term encompasses dosing simultaneously, or within minutes or hours of each other, or on the same day, or on alternating days, or dosing the therapeutic agent described herein on a daily basis, or multiple days per week, or weekly basis, for example, while administering another compound such as a chemotherapeutic agent on the same day or alternating days or weeks or on a periodic basis during a time simultaneous therewith or concurrent therewith, or at least a part of the time during which the therapeutic agent described herein is dosed.
[0138] Pharmaceutical compositions for use in accordance with the present disclosure can be manufactured and/or formulated in conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen. Any of the well- known techniques, carriers, and excipients may be used as suitable and as understood in the art, e.g., in Remington's Pharmaceutical Sciences, above.
[0139] Suitable routes of admini stration of the pharmaceutical compositions disclosed herein include, for example, oral, rectal, transmucosal, topical, or intestinal administration, parenteral delivery, including intramuscular, subcutaneous, intravenous, intramedullary injections, as well as intrathecal, direct intraventricular, intraperitoneal, intranasal, or intraocular injections. The compound or combination of compounds disclosed herein can also be administered in sustained or controlled release dosage forms, including depot injections, osmotic pumps, pills, transdermal (including electrotransport) patches, and the like, for prolonged and/or timed, pulsed administration at a predetermined rate. [0140] Injectables can be prepared in conventional forms, either as liquid solutions or suspensions, solid forms suitable for solution or suspension in liquid prior to injection, or as emulsions. Suitable excipients are, for example, water, saline, dextrose, mannitol, lactose, lecithin, albumin, sodium glutamate, cysteine hydrochloride, and the like. In addition, if desired, the injectable pharmaceutical compositions may contain minor amounts of nontoxic auxiliary substances, such as wetting agents, pH buffering agents, and the like. Physiologically compatible buffers include, but are not limited to, Hanks's solution. Ringer's solution, or physiological saline buffer. If desired, absorption enhancing preparations (for example, liposomes), may be utilized. For transmucosal administration, penetrants appropriate to the barrier to be permeated may be used in the formulation.
[0141] Pharmaceutical formulations for parenteral administration, e.g., by bolus injection or continuous infusion, 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 other organic oils such as soybean, grapefruit or almond oils, 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 that increase the solubility of the compounds to allow for the preparation of highly concentrated solutions. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
[0142] For oral administration, the compound(s) or combination of compounds disclosed herein can be formulated readily by combining the active compound with pharmaceutically acceptable carriers well known in the art. Such carriers enable the compound or combination of compounds disclosed herein to be formulated as tablets, film coated tablets, pills, dragees, capsules, liquids, gels, get caps, pellets, beads, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated. Pharmaceutical preparations for oral use can be obtained by combining the active compound 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, and/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. 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, and/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. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/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. In addition, stabilizers can be added. All formulations for oral administration should be in dosages suitable for such administration. In some embodiments, formulations of the compound(s) or combination of compounds disclosed herein with an acceptable immediate release dissolution profile and a robust, scalable method of manufacture are disclosed,
[0143] Pharmaceutical preparations 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, and/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. All formulations for oral administration should be in dosages suitable for such administration. [0144] For buccal administration, the compositions may take the form of tablets or lozenges formulated in conventional manner. For administration by inhalation, the compound or combination of compounds disclosed herein is conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofiuoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of, e.g., gelatin for use in an inhaler or insufflator may be formulated contaimiig a powder mix of the compound and a suitable powder base such as lactose or starch.
[0145] Further disclosed herein are various pharmaceutical compositions well known in the pharmaceutical art for uses that include intraocular, intranasal, and intraauricular delivery. Suitable penetrants for these uses are generally known in the art. Pharmaceutical compositions for intraocular delivery include aqueous ophthalmic solutions of the active compounds in water-soluble form, such as eyedrops, or in geilan gum or hydrogels; ophthalmic ointments; ophthalmic suspensions, such as microparticulat.es, drug- containing small polymeric particles that are suspended in a liquid carrier medium, lipid- soluble formulations, microspheres, and ocular inserts. Suitable pharmaceutical formulations are most often and preferably formulated to be sterile, isotonic and buffered for stability and comfort. Pharmaceutical compositions for intranasal delviery may also include drops and sprays often prepared to simulate in many respects nasal secretions to ensure maintenance of normal ciliary action. As disclosed in Remington's Phamiaceutical Sciences, 18th Ed., Mack Publishing Co., Easton, Pa. (1990), which is incorporated herein by reference in its entirety, and well-known to those skilled in the art, suitable formulations are often and preferably isotonic, slightly buffered to maintain a pH of 5.5 to 6.5, and most often and preferably include antimicrobial preservatives and appropriate drug stabilizers. Pharmaceutical formulations for intraauricular delivery include suspensions and ointments for topical application in the ear. Common solvents for such aural formulations include glycerin and water. [0146] The compound(s) or combination of compounds disclosed herein may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other giycerides.
[0147] In addition to the formulations described previously, the compound or combination of compounds disclosed herein may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneousiy or intramuscularly) or by intramuscular injection. Thus, for example, the compound or combination of compounds disclosed herein may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
[0148] For hydrophobic compounds, a suitable pharmaceutical carrier may be a cosoivent system comprising benzyl alcohol, a nonpoiar surfactant, a water-miscible organic polymer, and an aqueous phase. A common cosoivent system used is the VPD co-solvent system, which is a solution of 3% w/v benzyl alcohol, 8% w/v of the nonpoiar surfactant Polysorbate 80. TM., and 65% w/v polyethylene glycol 300, made up to volume in absolute ethanol. Naturally, the proportions of a co-solvent system may be varied considerably without destroying its solubility and toxicity characteristics. Furthermore, the identity of the co-solvent components may be varied: for example, other low-toxicity nonpoiar surfactants may be used instead of POLYSORBATE 80. TM.; the fraction size of polyethylene glycol may be varied, other biocompatible polymers may replace polyethylene glycol, e.g., polyvinyl pyrrolidone; and other sugars or polysaccharides may substitute for dextrose.
[0149] Other delivery systems for hydrophobic pharmaceutical compounds may¬ be employed. Liposomes and emulsions are well known examples of deliver}' vehicles or carriers for hydrophobic drugs. Certain organic solvents such as dimethylsulfoxide also may¬ be employed, although usually at the cost of greater toxicity. Additionally, the compounds may be delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent. Various sustained-release materials have been established and are well known by those skilled in the art. Sustained-release capsules may, depending on their chemical nature, release the compounds for a few weeks up to over 100 days. Depending on the chemical nature and the biological stability of the therapeutic reagent, additional strategies for protein stabilization may be employed. [0150] Agents intended to be administered intracellularly may be administered using techniques well known to those of ordinary skill in the art. For example, such agents may be encapsulated into liposomes. All molecules present in an aqueous solution at the time of liposome formation are incorporated into the aqueous interior. The liposomal contents are both protected from the external micro-environment and, because liposomes fuse with cell membranes, are efficiently delivered into the cell cytoplasm. The liposome may be coated with a tissue-specific antibody. The liposomes will be targeted to and taken up selectively by the desired organ. In some embdoimetns, small hydrophobic organic molecules may be directly administered intracellularly. Additional therapeutic or diagnostic agents may be incorporated into the pharmaceutical compositions. Alternatively or additionally, pharmaceutical compositions may be combined with other compositions that contain other therapeutic or diagnostic agents.
Methods of Reducing Neddylation
[0151] Some embodiments disclosed herein provide methods of reducing neddylation in a cell, in a cell population, and/or in a subject. In some embodiments, the methods comprise contacting a ceil, a ceil population, and/or a subject with a composition comprising a GlyRS inhibitor, wherein the level of neddylation is decreased in the cell, the ceil population, and/or the subject. In some embodiments, the methods comprise acquiring knowledge of the level of neddylation in the cell, the cell population, and/or the subject before and/or after contacting the cell, the cell population, and/or the subject with the composition. In some embodiments, the methods comprise identifying a cell, a cell population, and/or a subject having undesired level of neddylation before and/or after contacting the cell, the cell population, and/or the subject with the composition.
[0152] The composition can be, for example, a pharmaceutical composition. The GlyRS inhibitor can be any of the GlyRS inhibitor disclosed herein. The GlyRS inhibitor may inhibit GlyRS functions in aminoacylation and neddylation. In some embodiments, the GlyRS inhibitor does not significantly inhibit GlyRS function in aminoacylation. In some embodiments, the GlyRS inhibitor is an inhibitor for a mammalian GlyRS protein (e.g., a human GlyRS protein). In some embodiments, the GlyRS inhibitor is GlySA or a derivative thereof. [0153] The cell can be, for example a mammalian cell (e.g., a human cell). The cell population can comprise, or consist of, mammalian cells (e.g., human cells). The cell or the cell population can be present in cell culture, a tissue, in an organ, or in a body of a subject. Contacting the cell or the cell population with the composition can be performed in vitro, ex vivo, in vivo, or a combination thereof.
Methods of Inhibiting Cell Proliferation
[0154] Some embodiments disclosed herein provide methods of inhibiting or reducing cell proliferation. The methods, in some embodiments, comprise: contacting a cell, a ceil population, or a subject with a pharmaceutical composition comprising a GlyRS inhibitor, wherein the proliferation of the cell, one or more cells present in the cell population or the subject is inhibited. As described herein, the inhibition of cell proliferation can be complete or partial. In some embodiments, a method of reducing cell proliferation is provided.
[0155] In some embodiments, the methods comprise acquiring knowledge of the level of cell proliferation in the cell, the cell population, and/or the subject before and/or after contacting the ceil, the ceil population, and/or the subject with the composition. In some embodiments, the methods comprise identifying a cell, a cell population, and/or a subject having undesired level of cell proliferation before and/or after contacting the cell, the cell population, and/or the subject with the composition,
[0156] The NEDD8 pathway has been shown to be essential for cellular function, through its critical role in mediating the ubiquitination by CRLs of numerous proteins involved in cell cycle progression and cell growth and survival. The relevance of the NEDD8 conjugation pathway in various cancer therapies has been discussed in Soucy et ai., The NEDD8 Conjugation Pathway and Its Relevance in Cancer Biology and Therapy. Genes Cancer 1, 708-16 (2010), the content of which is hereby expressly incorporated by reference in its entirety. Table 1 of Soucy et al. summarizes various substrate proteins of the cullin- RTNG ligases and their associations with cancer, and is reproduced below. Additional substrates that are involved in tumorigenesis include: tumor suppressor NF2, which encodes Merlin, p53, Mdm2, epidermal growth factor receptor, VHL tumor suppressor protein, Ll l, or any combination thereof .
Figure imgf000050_0001
[0157] Neddylation of the cullin proteins activates the E3 ligases for ubiquitination and promotes the degradation of their downstream targets, including key regulators of ceil cycle. For example, c-Myc, c-Jun, cyclin E, Emi l, Cdt-1, ρΙκΒα, F2, HIF-l a, β-catenin, Cdc25A, mTOR, BimEL and p27.
[0158] Cell proliferation can be inhibited in various types of cells, including animal cells and plant cells. In some embodiments, the cell is a mammalian ceil. The extent by which cell proliferation is reduced can vary. In some embodiments, the proliferation of the cell is reduced by at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 99%.
[0159] The composition can be, for example, a pharmaceutical composition. The GlyRS inhibitor can be any of the GlyRS inhibitor disclosed herein. The GlyRS inhibitor may inhibit GlyRS functions in aminoacylation and neddylation. In some embodiments, the GlyRS inhibitor does not significantly inhibit GlyRS function in aminoacylation. In some embodiments, the GlyRS inhibitor is an inhibitor for a mammalian GlyRS protein (e.g., a human GlyRS protein). In some embodiments, the GlyRS inhibitor is GlySA or a derivative thereof.
[0160] The cell can be, for example a mammalian cell (e.g., a human cell). The ceil population can comprise, or consist of, mammalian cells (e.g., human cells). The cell or the cell population can be present in cell culture, a tissue, in an organ, or in a body of a subject. Contacting the cell or the cell population with the composition can be performed in vitro, ex vivo, in vivo, or a combination thereof.
Methods of Treating Cancer
[0161] Also disclosed herein are methods of treating or ameliorating cancer in a subject by administering a therapeutically effective amount of a pharmaceutical composition comprising a GlyRS inhibitor to the subject. As disclosed herein, the GlyRS inhibitor may be capable of inhibiting or reducing interaction between the GlyRS protein and the components of the neddylation pathway, thereby reduce proliferation of the cancer cells. For example, the methods may be used for treating or ameliorating a solid tumor or a hematological malignancy, for example, a cancer that is selected from the group consisting of breast cancer, cervical cancer, colon cancer, liver cancer, prostate cancer, melanoma, ovarian cancer, lung cancer, renal cell carcinoma, Schwannoma, mesothelioma, acute myeloid leukemia, multiple myeloma, non-Hodgkin lymphoma, and a combination thereof.
[0162] The GlyRS inhibitor can be any of the GlyRS inhibitor disclosed herein. The GlyRS inhibitor may inhibit GlyRS functions in aminoacylation and neddylation. In some embodiments, the GlyRS inhibitor does not significantly inhibit GlyRS function in aminoacylation. In some embodiments, the GlyRS inhibitor is an inhibitor for a mammalian GlyRS protein (e.g., a human GlyRS protein). In some embodiments, the GlyRS inhibitor is GiySA or a derivative thereof.
[0163] To practice the methods disclosed herein, a GlyRS inhibitor, and/or an agent that prevents or reduces interaction between a GlyRS protein and a component of the neddylation pathway, and pharmaceutical compositions thereof may be administered orally, parenteral ly, by inhalation, topically, rectal ly, nasally, buccally, vaginally, via an implanted reservoir, or other drug administration methods. The term "parenteral" as used herein includes subcutaneous, intracutaneous, intravenous, intramuscular, intraarticular, intraarterial, intrasynovial, intrastemal, intrathecal, intralesional and intracranial injection or infusion techniques,
[0164] A sterile injectable composition, such as a sterile injectable aqueous or oleaginous suspension, may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent. Among the acceptable vehicles and solvents that may be employed include mannitol, water. Ringer's solution and isotonic sodium chloride solution. Suitable carriers and other pharmaceutical composition components are typically sterile.
[0165] In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium (e.g., synthetic mono- or diglycerides). Fatty acids, such as oleic acid and its giyceride derivatives, are useful in the preparation of injectables, as are pharmaceutically acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions can also contain a long-chain alcohol diluent or dispersant, or carboxymethyl cellulose or similar dispersing agents. Various emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms can also be used for the purpose of formulation.
[0166] A composition for oral administration may be any orally acceptable dosage form including, but not limited to, tablets, capsules, emulsions and aqueous suspensions, dispersions and solutions. In the case of tablets for oral use, commonly used carriers include lactose and corn starch. Lubricating agents, such as magnesium stearate, can also be added. For oral administration in a capsule form, useful diluents include lactose and dried com starch. When aqueous suspensions or emulsions are administered orally, the active ingredient can be suspended or dissolved in an oily phase combined with emulsifying or suspending agents. If needed, certain sweetening, flavoring, or coloring agents can be added. A nasal aerosol or inhalation compositions can be prepared according to techniques well-known in the art of pharmaceutical formulation and can be prepared as solutions in, for example saline, employing suitable preservatives (for example, benzyl alcohol), absorption promoters to enhance bioavailability, and/or other solubilizing or dispersing agents known in the art. EXAMPLES
[0167] Embodiments of the present application are disclosed in further detail in the following examples, which are not in any way intended to limit the scope of the present disclosure.
Example 1
Glvcyl-tRNA synthetase is critical for neddylation by protecting activated E2
[0168] Ubiquitination and ubiquitin-like modifications are post-translational modifications in eukaryotes playing key roles in protein homeostasis and functions1. Among them, neddylation— conjugating the ubiquitin-like protein NEDD8 to its target proteins— is an essential biological process in organisms from yeast to mammals to critically regulate cell cycle progression. Like ubiquitination, the modification is achieved through a sequential enzymatic cascade involving an activating enzyme (El), a conjugating enzyme (E2), and a ligase (E3) (Fig. la). To date, one El ( APPB P 1 /UB A3 ), two E2 (Ubcl2 and Ubc2F), and several E3 ligases have been found for neddylation (Fig. la). Although many NEDD8 targets were reported8, the biological functions of neddylation so far have been primarily characterized in the context of its main target— the cullin family, a critical component of the ubiquitin E3 ligase family of culiin-RING ligases. Neddylation of the cuilin proteins activates the E3 ligases for ubiquitination and promotes the degradation of their downstream targets, including key regulators of cell cycle.
[0169] Ubiquitin and most ubiquitin-like modifier proteins, including NEDD8, have a conserved C-terminal glycine that is used to activate, conjugate, and finally attach the modifiers to their targets. Structurally, the glycine residue is located at the tip of a flexible 'tail' protruding out from the central ubiquitin fold to provide with the accessibility. On the other hand, GlyRS is a member of the evoiutionarily ancient aminoacyl-tRNA synthetase family essential for all living organisms. GlyRS catalyzes the aminoacylation reaction to attach glycine to the 3 ' end of the cognate tRNAs to provide substrates for protein synthesis in the ribosome. Aminoacylation of tRNA is a two-step reaction: first, GlyRS activates glycine with ATP to generate Gly-AMP; then the glycyi moiety is transferred from Gly-AMP to the tRNA to make the 'charged' tRNA. Interestingly, the first step of this aminoacylation reaction is chemically equivalent to the first step activation reaction catalyzed by an E l enzyme. Moreover, the specific amino acid binding pocket of a tRNA synthetase can be exploited for binding to a cognate amino acid residue on a protein to develop new functions.
[0170] By using biochemical and cell-based functional assays, a function connection between GlyRS and neddylation in human cells have been revealed. It turns out that GlyRS plays an important role in neddylation through direct interactions with multiple components of the neddylation pathway. Although it appears that GlyRS cannot function as an El, it can bind to the APPBP1 subunit of El to capture and protect the activated E2 (NEDD8-conjugated Ubcl2) to critically enhance neddylation. Knockdown of GlyRS, but not a different tRNA synthetase, decreases the global level of neddylation and delays cell cycle progression. This provides the first example of a translation factor directly functioning in posttranslational modification.
Methods and materials
Plasmid constructs and protein purification
[0171] GST-tagged or untagged APPBP1-UBA3, Ubcl2, and EDD8 proteins were purified as previously reported. GST-APPBP1 was obtained by injecting the purified GST-APPBP1/UBA3 into the Superdex 200 column and collected the fractions that contained only GST-APPBP1. N-terminal His-SUMO-tagged NEDD8 construct was generated by subcloning the pGEX2TK-NEDD8 into a modified pET28a vector and the protein was purified using a Nickel -NT A column followed by MonoQ ion exchange column. Ubiquitin and SUMOl proteins (R&D) were purchased. His-tagged full-length and AWHEP GlyRS, SerRS, and TrpRS were purified as reported earlier. The ABD (V541 -E685) of GlyRS was cloned, expressed, and purified in a similar way as for the full-length GlyRS. (The yield for ABD alone is higher than that for the full-length GlyRS). Tag-free full-length GlyRS, AF84-L93 (F84-L93 replaced by GGG), ΔΙ232-Μ238 (Γ232-Μ238 replaced by GG), Alnsertionl (F 147-F224 replaced by GSGSGG) and G526R GlyRS proteins were similarly prepared and had the N-terminal His-SUMO-tag removed by Ulpl protease. Inserting the GlyRS sequence into a modified pET28a vector generated N-terminal GST-tagged GlyRS and the protein was purified by glutathione sepharose chromatography. GST-Cuilinlcld/Rbxl was purified by glutathione sepharose chromatography as well. Ube2F protein was purified by a Nickel-NTA column. The purity of proteins was examined by SDS-PAGE to be above 95%.
Libel 2' preparation and stability assay
[0172] The conjugated Ubcl2 was prepared by mixing APPBP1/UBA3 (2μΜ), Ubcl2 (C I 1 I S; the mutant would form a stable ester linkage to NEDD8) (70μΜ), and His- SUMO-tagged NEDD8 (Ι ΟΟμΜ). The mixture was incubated at 25°C for 16 h in 50 mM NaOAc pH 5.5, 50mM NaCl, ImM MgC12, and 2mM ATP. Ubcl2N8 was then purified by- gel filtration chromatography using a Superdex 200 column. The fractions containing only Libel 2N8 were concentrated and used for the study. The purity of proteins was examined by SDS-PAGE to be above 95%.
[0173] The wild type Ubcl2 conjugation was performed in a similar manner except using wild type Ubcl2 instead of Ubcl2 el l Is at 25°C for 16 h in 50 mM Tris pH 7.4, 50mM NaCl, ImM MgC12, and 2mM ATP. Ubcl2N8 was then purified by gel filtration chromatography using a Superdex 200 column. The fractions containing only Libel 2N8 were concentrated and used for the study. The purity of proteins was examined by SDS-PAGE to be above 95%. The stability assay was carried out by incubating Ubcl2N8 (5uM) with GivRS, BSA or same volume of PBS buffer at 37° for indicated time in the PBS buffer (PH7.4 supplemented with 5mM: DTT). Samples were then subjected to SDS-PAGE and stained with commassie blue.
Cell culture and shRNA knockdown
[0174] HEK293 and HeLa cells were cultured in DMEM media supplemented with 10% FBS. Short-hairpin RNA (shRNA) sequences targeting the human GlyRS (5'- GCATGGAGTATCTCACAAAGT-3', SEQ ID NO: 3) or human SerRS (5'- GGCATAGGGACCCATCATTGA-3', SEQ ID NO: 36) were inserted into the pLentiLox- hHl piasmid, modified from the pLentiLox 3.7 plasmid to contain a HI promoter (between Xba I and Xho I sites) to drive shRNA expression. All transfections were done with Lipofectamine 2000 (Invitrogen) and cells were harvested 48 hours after transtection. Cycloheximide chasing assay
[0175] HeLa cells at 80% confluence were transfected with pLentiLox-hHl vectors containing either a scramble sequence or GlyRS specific sequence using Lipofectamine 2000. 48 hours after transfection, medium were replaced with that containing 3C^g/mL cycloheximide (Celisignaliing) or 20uM MG-132 (#508338, Fisher) as indicated. MLN4924 (1-502, Bostonbiochem) samples are prepared by treating the ceils with 0.2 uM MLN4924 for 24 hours and then followed by cycloheximide or MG132. Samples are collected and lysed with acid lysis buffer and later subjected to SDS-PAGE.
Immunobloting and antibodies
[0176] Cells were washed with PBS and then lysed with either the lysis buffer (#4930; Ceil Signaling Technology) or the acid lysis buffer (50 mM HEPES, 150 mM NaCi, 0.1% (w/v) SDS, pH 6.0) supplemented with protease cocktail (Roche). The supernatant of the lysates was used for Western blotting. The antibodies used in this studies include anti- cullinl (H213; Santa cruz) anti -UB A3 I - 10; Santa cruz), anti-GlyRS (B01P; Abnova), anti- SerRS (homemade), anti-V5 (R96-CUS; Invitrogen). Anti-NEDD8 (#2745), anti-Ubiquitin (#3936), anti-SUMOl (#4930), anti-Ubcl 2 (#5641 ), anti-Ubc9 (#4918), anti-Flag (#2908), anti-UBAf (#4891), anti-UbcH7 (#3848), anti-UBA2 (#8688), anti-p27kip (#3698) and anti- a-Tubulin (#3873) are all from Cell Signaling, Immunoblots are quantified and analyzed using Image J. The integrated areas of the bands were normalized to that of the corresponding a-tubulin level. Error bars indicate standard deviation with n>3. P values are calculated by one-tailed Student's t test.
Immunoprecipitation
[0177] 2 μg of anti-V5 (R96-CUS; Invitrogen), anti-GlyRS (D-10; Santa Cruz), anti-myc (9E10; Santa Cruz) antibodies or mouse IgG (#5415, Cell Signaling) were coupled to 30 μΐ of protein G-sepharose (Amersham Biosciences) beads and used for immunoprecipitations. Supernatant of HEK293 cells lysates were then added and incubated with the antibodies for 3 h or overnight at 4°C. The G-sepharose beads were then washed four times with 1 mL of cold PBS buffer (pH 7.4). The beads-bound proteins were eluted and denatured with SDS-ioading buffer and subjected to SDS-PAGE and Western blotting. FACS analysis
[0178] HeLa cells were transfected with either pCDNA6-V5c vectors containing either GARS or G526R mutant using Lipofectaming 2000. 48 hours after transefection, cells were treated with medium containing 0.5 uM MLN4924 or same amount of DMSO. 24 hours after treatment, cells were washed once with sorting buffer (PBS supplemented with 1 %FBS and 5mM EDTA) and collected using 0.05% Trypsin. The mixture was then spinned at 500g for 5min to spin cells down to the pellets. Cells were then washed twice with sorting buffer and then suspended and fixed with 70% EtOH at 4 for 2 hours. After fixation, cells were washed twice with sorting buffer and suspended with the PI staining solution. Samples were later analyzed by flow cytometry (BD FACS Canto).
Affinity pull-down assay
[0179] Glutathione sepharose beads (GE Healthcare) were equilibrated with TEE buffer (50mM Tris pH 7.9), ImM EDTA, and ImM EGTA). GST-fusion proteins were mixed with 50 ul of glutathione sepharose beads and incubated for 2 h at 4°C and then washed with TEE buffer twice. Aliquots of the protein-bound beads were then incubated together with different forms of GlyRS for 2 h at 4°C. Finally, the beads were washed 5 times with washing buffer (20 mM Hepes pH 7.9, 150 mM NaCi, 0.5 mM EDTA, 10% Glycerol, 0.1% Triton X- 100, and ImM DTT) and proteins were eluted with SDS sample buffer and analyzed by immunoblotting. Control experiments were performed with GST- coated beads. His-tag pull-down assay were carried out in a similar manner using purified his-tagged proteins and incubated with nickel-NT A beads (Qiagen).
Hydrogen-Deuterium exchange mass spectrometry
[0180] Solution-phase amide HDX was carried out with a fully automated system as described previously. Briefly, 4 iL of protein was diluted to 20 μ,Τ with D20-comtaining buffer and incubated at 4°C for 10, 30, 60, 900, or 3600 s. Samples were diluted to 50 Ε with 3M urea, 1% TFA. at 1°C to denature the proteins and minimize back-exchange. Samples were then passed across an immobilized pepsin column at 50 μΙΛηίη in 0.1% TFA at 15°C. Resulting peptides were trapped on a C8 cartridge (Hypersil Gold, Thermo Fisher). Peptides were then gradient eluted (4% CH3CN to 40% CH3CN, 0.3% formic acid) at 1°C across a lmm x 50mm C 18 reversed phase HPLC column Hypersil Gold, Thermo Fisher) and electrosprayed directly into an orbitrap mass spectrometer (either LTQ Orbitrap or Q- Exactive, Thermo Fisher). Data were processed with in-house software and visualized with PyMol (Schrodinger, LLC). To measure the difference in exchange rates, the average percent deuterium uptake for unbound GlyRS protein was calculated at all time points. From this value, the average percent deuterium uptake for GlyRS protein bound to NEDD8 was subtracted. Negative perturbation values indicate exchange rates are slower for GlyRS bound to NEDD8, which suggests the region is less accessible to amide exchange due to structural alteration or direct contact between GlyRS and NEDD8. GlyRS-Ubcl2 interaction was analyzed in a similar way.
Biolaver interferometry
[0181 ] The dissociation constants (Kd) were obtained with biolayer interferometry by using an Octet QK system (ForteBio, Menio Park, Calif, USA). Samples or buffer were dispensed into 96-well plates (Millipore, Bil led ca, MA) at 200 |iL per well . Operating temperature was maintained at 30 °C. Proteins were diluted into kinetic buffer (PBS with 0.1 % BSA and 0.002% Tween-20) and immobilized on either anti-GST or Ni-NTA sensor tips. The other proteins were diluted using the same buffer into a range of different concentrations. Assays with Ubcl2N& were carried out using the acid kinetic buffer (50 rnM NaOAc pH 5.5, 50 mM NaCl, 0.1% BSA, and 0.002% Tween-20). The raw data were processed by subtraction to reference cells and then aligned with baselines. The dissociation constants J¾ were obtained by fitting the processed data using the 1 : 1 model in the Octet analysis software with R2 >0.99.
Molecular modeling
[0182] The Ubcl 2N8~GlyRS interaction is obtained by using the Patchdock server. GlyRS (PDB: 2PME) is assigned as the receptor and Ubcl 2 8 (PDB: 4P50 chain G&H) is assigned as the ligand. Clustering RMSD is set at 4.0. The GlyRS- APPBP1 interaction is modeled using similar settings with APPBP1/UBA3 (PDB: 2NVU chain A&B) assigned as the receptor and GlyRS assigned as the ligand. Molecular visualization and analysis were performed using PyMOL (The PyMOL Molecular Graphics System, Version 1.2rl, Schrodinger, LLC).
Results
GlyRS specifically binds to NEDD8 through the catalytic domain
[0183] Human GlyRS is composed of three distinct domains: the N-terminal metazoan-specific WHEP domain, catalytic domain, and the C -terminal anticodon-hinding domain (ABD) (Fig. lb). GlyRS interaction with ubiquitin or ubiquitin-like proteins such as NEDD8 and SUMOl were investigated. As shown in Figure lc, purified human GlyRS protein can specifically bind to NEDD8, but not ubiquitin or SUMOl . Two other human tRNA synthetases were tested side-by-side (SerRS and TrpRS) and showed no interaction with NEDD8, ubiquitin or SUMOl (Fig. lc). The GlyRS-NEDD8 interaction was verified in HEK293 cells by co-immunoprecipitation (Fig. Id). Furthermore, by using truncated recombinant proteins, the interaction was mapped to the catalytic domain of GlyRS (Fig. le). Hydrogen-deuterium exchange (HDX) analysis (monitored by mass spectrometry) further confirmed that the catalytic domain is the site for interaction with NEDD8 (Fig. 6).
GlyRS plays a critical role in neddylation
[0184] To test the effect of GlyRS in neddylation, GlyRS was ectopicaliy expressed in HEK293 cells. The overexpression of GlyRS (but not TrpRS) increased the amount of NEDD8-conjugated Ubcl2 (an E2 for neddylation), but not the ubiquitin- conjugated UbcH7 (an E2 for ubiquitination) and SUMO-conjugated Ubc9 (an E2 for sumoylation) (Fig. 2a). The core synthetase (AWHEP) was still active, while ABD domain alone did not have the effect (Fig. 2a), further highlighting the importance of the catalytic domain. Consistently, knockdown of GlyRS in HeLa cells led to significant decrease of NEDD8-conjugated Ubcl2 (Ubcl2 8) (Fig. 2b), but not of the conjugated UbcH7 and Ubc9 (UbcH7u and Ubc9Sumo) (Fig. 2c,d). It was moted that the GlyRS knockdown had no effect on E l neddylation (UBA3N8) or the level of free NEDD8 (Fig. 2b). Also, GlyRS knockdown did not affect Ube2F (Fig. 7a), the other less frequently used E2 for neddylation8. Knockdown of a different tRNA synthetase (SerRS) had no effect on all E2s was tested (Fig. 2b-d and Fig. 7a). Importantly, knockdown of GlyRS, but not SerRS, also decreased the global level of neddylation, including neddylation of culiin proteins (Fig. 2b). These observations indicate that GlyRS plays an important role in the neddylation process, most likely through mechanism that enhances Ubcl 2 activity and/or protects the NEDD8- conjugated Ubc l2.
GlvRS strongly binds to NEDD8-conj ugation Ubcl2 and protects it from degradation
[0185] To understand how GlyRS promotes the cellular level of Ubcl2N8, whether GlyRS could interact with Ubcl2 was tested. GST pull-down assay together with HDX analysis showed that GlyRS could bind to Ubcl2 and again the catalytic domain is primarily responsible for this interaction (Fig. 8a & b). Consistent with the lack of effect on Ube2F conjugation when GlyRS was knocked down (Fig. 7a), there was very weak binding between Ube2F and GlyRS, if any (Fig. 7b).
[0186] As a conjugating enzyme, Ubcl2 exists in two forms in the cell, the apo form and the conjugated form Ubcl2N8. The latter form is achieved by linking a NEDD8 molecule, transferred from the activating enzyme APPBP1/UBA3, to the catalytic cysteine residue (Cysl l l) of Ubcl 2 via a thioester bond. Because GlyRS can interact with both Ubcl2 and EDD8, it was tempting to test if GlyRS could bind to Libe l 2 8 and protect it from degradation. Thioester bonds, like the one that links NEDD8 to Ubcl 2, are common intermediates in biological reactions and are highly labile.
[0187] Indeed, GlyRS can bind to Ubcl 2 8 in HEK293 cells as detected by co- immunoprecipitation (Fig. 3a). Moreover, relative to the input, more Ubc l 2NS than i. be 12 was bound to GlyRS, indicating that GlyRS preferably binds to NEDD8-conjugated Ubcl2. To quantify the difference, Uhe l2NS was enzymatieally made and purified, and its binding affinity to GlyRS was measured using biolayer interferometry. Remarkably, Ubcl2N8 binds to immobi lized GlyRS with a K& of 4.09±0.30 nM, which is 100-fold and 30-fold stronger the Κά for Ubcl2 alone (488+73 nM) and for NEDD8 alone (126±19 nM), respectively (Fig. 3b). The GlyRS-Ubcl 2N8 interaction was also analyzed reversely by immobilizing Ubcl2N8 to give a similar K<L of 3.21±0.17 nM (Fig. 3c). In the presence of GlyRS, but not BSA, Ubcl2N8 was significantly more stable than in the absence of GlyRS (Fig, 8ε). Therefore, it is possible that GlyRS enhances neddylation by protecting reaction intermediate Ubcl2N8. Model of GlvRS-Ubcl2N8 complex
[0188] Using the crystal structure of human GiyRS 16 and of Ubcl 2N8 (adapted from its complex with substrate cuilin and E3), a model was generated for the GlyRS- Ubcl2 8 interaction using the Patchdock algorithm (http ://bioinfo3 d. cs.tau. ac.il/PatchDock/). The top solution places Ubcl2N8 right on top of the dimerization interface of GiyRS, with Ubcl2 wedged against motifs 1 and 2 that are conserved for class II tRNA synthetases, and with EDD8 snuggled in between two β-hai in loops (F84-L93 and I232-M238) (Fig. 3d). This model is in general consistency with the results obtained from hydrogen-deuterium exchange analysis (Fig. 6 and Fig. 8b). For example, motif 2 and part of motif 1 of GiyRS had decreased levels of deuterium incorporation as a result of Ubcl 2 binding, the two β- hairpin loops (F84-L93 and I232-M238) also have reduced deuterium incorporation when binding to EDD8 (Fig. 6). To further validate the model, two deletion mutants of the β- hairpin loops (Δ84-93 and Δ232-238) were created and Δ84-93 decreased binding of GiyRS to Ubc l 2 8 by 9 fold (A' a 27 0 · 0.40 nM) and Δ232-238 abolished the binding completely (Fig. 3e & f). Seemingly, Δ232-238 not only disrupts the NEDD8-binding site, but also creates potential conformational changes that affect UbcI2 binding. In contrast, deletion of an insertion domain unique to GiyRS (Insertion 1), which shows no involvement in the GlyRS-Ubc l2N8 interaction according to the model (Fig. 3d), had no effect on the binding (A.;.- 4.32 . 0. 1 2 nM) (Fig. 3g). Consi stent with its loss of binding to Ubcl 2N8, the Δ232-238 GiyRS was completely inactive for promoting Ubcl 2 conjugation (Fig. 3h), suggesting the function of GiyRS in promoting neddylation is tightly linked to its ability to bind and protect Ubcl 2 8.
GlvRS captures Libel 2NS released from El
[0189] It was found that GiyRS could al so bind to the heterodimeric El enzyme for neddylation (APPBP1/UBA3) (Fig. 9a & b). However, unlike NEDD8 and Ubc l2, EI binds to the anticodon-binding domain (ABD) of GiyRS, as revealed by both GST-pull down and bioiayer interferometry analysis (Fig. 9a,b,c). The APPBP 1/UBA3 heterodimer has similar binding affinities to full-length GiyRS (J¾ :=: 159± 12 nM) and the ABD domain alone (Κά = 187±32 nM) (Fig. 9b). Furthermore, similar binding affinity was determined for APPBP1 subunit alone with the ABD of GiyRS (A',; 144±3.5 nM) (Fig. 9c), suggesting that the APPBPl subunit of El is responsible for the GlyRS interaction. The APPBPl-GlyRS interaction was further validated by co-immunoprecipitation in HEK293 cells between endogenous proteins (Fig. 4a). A modeling study through Patchdock also suggested that the ABD of GlyRS interacts with the APPBP1 subunit of the El (Fig, 9d).
[0190] The fact that GlyRS uses different domains to interact with E l and Ubcl2N8 interaction suggests that GlyRS may be able to simultaneously bind to both El and Ubcl2N8. However, this is not possible if El and Ubcl 2N8 are in complex with each other (Fig. 4b). It is important to note that the El-Ubcl2N8 interaction is weak, because once NEDD8 is transferred from El to E2 during the enzymatic cascade, the association is weakened through conformational changes to facilitate the release of Ubcl 2N8 for the next event. Therefore, the role of GlyRS might be to capture and protect the conjugated E2 after it is released from the El and before it finds a correct E3 and/or substrate. By binding to the APPBP1 subunit of El through the ABD domain, GlyRS is in proximity to capture the released Ubcl2N8 (Fig. 4b).
[0191] An experiment was designed to test this concept using biolayer interferon! etry analysis. El was immobilized to detect its interaction with Ubcl 2N8 and GlyRS, separately and simultaneously. The binding of Ubcl2N8 to El was weak and the disassociation was faster (Fig. 4c). In contrast, the binding of Ubcl 2N8 to GlyRS was much stronger (Fig. 4c). Interestingly, when both Ubcl2N8 and GlyRS are present, the overall binding was stronger than the sum of the individual bindings (Fig. 4c), suggesting that a ternary complex of El-GlyRS-Ubcl2N8 is formed that prevents Ubcl2N8 from releasing to the solvent.
[0192] The ABD domain of GlyRS was used as a control for this experiment. Although ABD domain alone interacted with the El as strongly as the full length GlyRS (Fig. 9a&b), the absence of the catalytic domain of GlyRS for capturing Ubcl 2N8 would not allow the formation of a ternary complex. Indeed, the ternary complex with ABD domain was not detected (Fig. 4d).
GlyRS does not interfere with transferring Ubcl2N8 to substrate
[0193] Because there is a tight binding between GlyRS and Ubcl2N8, it is important to confirm that the binding does not impede Ubcl2N8 from passing NEDD8 down to its downstream targets such as cullin. Notably, the protection of Ubcl2N8 by GlyRS does not seem to require burying the thioester bond between Ubcl 2 and NEDD8. In the model of the GlyRS-Ubcl2N8 complex (Fig. 3d), the thioester bond between the C -terminal glycine residue Gly76 of NEDD8 and Cysl i l of Ubcl2 is facing outward rather than buried inside (Fig. 9e). The positioning of this thioester bond is fully compatible with transferring the NEDD8 from Ubcl2 to the acceptor residue Lys720 of cullin 1 (Fig. 9e). Using cullinl-Rbxl (RING-box protein 1) as a substrate, it was further tested if cullinl -Rbxl can bind to Ubcl2N8 in the presence of GlyRS. It was found that not only cullinl-Rbxl could bind to Ubcl 2N8 in the presence of GlyRS, the binding seems to release GlyRS from Ubcl 2NS (Fig. 4e), presumably to allow GlyRS to turnover. Using an in vitro neddvlation assay, it was demonstrated that the presence of GlyRS did not decrease but rather increased the neddylation of cullin 1 (Fig. 9f).
GlyRS promotes cell cycle progression
[0194] The biological functions of neddylation are best known in the context of cullin proteins as the target. eddylation of the cullin activates the CRLl (cull in 1 -RING) ubiquitin ligases and facilitates the degradation of their downstream targets, including ceil cycle inhibitor p27klp (Fig. 5a), p27kip induces cell cycle arrest by binding to cyclin-CDK (cyclin-dependent kinase) complexes to inhibit their catalytic activity. Therefore, degradation of p27kip through cullin neddylation promotes cell cycle progression and cell proliferation. Because this is a well-established pathway, p27kip degradation and ceil cycle progression were focused on to study the biological role of GlyRS in neddylation,
[0195] To evaluate p27kip degradation in cells, cycloheximide, an inhibitor of translation, was used to block new protein synthesis. In control ceils (untreated cells or ceils transfected with a control shRNA (shCtrl)), the level of p27kip decreased rapidly upon cycloheximide treatment (half-life = 2.5h) (Fig. 5b). (Proteasome inhibitor MG132 was used as a control to confirm the decrease of p27kip level was due to ubiquitin-dependent proteolysis.) However, in cells transfected with an shRNA to knock down GlyRS (shGARS), the half-life of p27kip was significantly extended (~5h) (Fig, 5b). And the stabilization of p27kip was concurrent with a decreased level of culiinNS (Fig. 5b). To further confirm that this effect is related to neddylation, a neddylation specific inhibitor MLN4924 was used, which binds to the ATP -binding site of LIB A3 to block neddylation21. MLN4924 treatment completely abolished cullin neddylation and blocked p27kip degradation (Fig. 5b),
[0196] It was expected the stabilization of p27kip would lead to ceil cycle arrest. To test flow cytometry cell cycle analysis was used. Indeed, compared with the control cells, HeLa cells transfected with shGARS or treated with MLN4924 showed significantly decreased number of diploid (2N) cells and increased population of tetraploid (4N) cells (Fig. 5c), indicating cell cycle arrest. Therefore, the role of GlyRS in neddylation was linked to ceil cycle progression.
[0197] The study described in this example provides the first example that a 'translation factor' can act in post-translational modification. In fact multiple components of the translation machinery including tRNA synthetases have been reported as substrates of neddylation, however, there was no indication that they can function directly to influence the post-translational modification pathway. Despite no El -like activity was found in GlyRS an enhancer role for neddylation has been revealed. This role is achieved through the two essential domains of GlyRS for aminoacylation— the catalytic domain and the anticodon- binding domain (ABD). Results from biochemical and structural analysis support the model, in which GlyRS, by docking on the APPBP1 subunit of El through the ABD domain, is in proximity to capture, by the catalytic domain, the NEDD8-conjugated E2 (Ubcl2N8) after it is released from the El and before it finds the correct E3 and/or substrate to transfer the NEDD8 modifier. The confinement provided by the synthetase would protect the conjugated E2 from random hydrolysis and thereby enhance the overall efficiency of the neddylation pathway. As neddylation is well established for its role in promoting cell cycle progression and proliferation, a synergy may exist between the aminoacylation function of GlyRS to support new protein synthesis and its neddylation enhancer function to stimulate cell proliferation.
[0198] The selective NEDD8 El inhibitor MLN4924 is currently being tested in several clinical trials for hematological malignancies and solid tumors. As was shown here in Fig 5, a partial knockdown of GlyRS expression in HeLa cells had a similar effect as MLN4924 in causing cell cycle arrest, suggesting that inhibition of GlyRS may be considered for cancer treatment as well. Importantly, inhibition of GlyRS would not only suppress neddylation but also impede protein synthesis, both of which are undesirable for tumor growth.
[0199] According to the online Human Protein Atlas, all components of the neddylation pathway are predominantly localized in the nucleus, consistent with their prominent role in cell cycle regulation. However, many reports suggested that Ubcl2, and neddylation substrates are also localized in the cytoplasm, indicative of the existence of other cytoplasmic targets for neddylation. On the other hand, tRNA synthetases are predominantly cytoplasmic proteins for their role in protein synthesis; however, a large repertoire of regulator}' functions of tRNA synthetases beyond their enzymatic role in protein synthesis has been reported and many tRNA synthetases are found in the nucleus to carry out important biological functions, such as regulating vascular development, activating p53 signaling, promoting DNA damage response, and regulating gene expression under immunological challenge. Although a nuclear localization signal sequence cannot be readily identified in GlyRS, given the role of GlyRS in neddylation, GlyRS may also reside in the nucleus.
[0200] It is interesting to note that often time the regulatory functions of tRNA synthetase are linked to the new domains that were added to the catalytic core during evolution. In GlyRS, this new domain is the WHEP domain, which was added to the synthetase in metazoans. The role of the WHEP domain in regulating neddylation remains to be further characterized, however, based on binding and functional results (Fig.le, Fig.2a and Fig. 9a&b), it appears that the WHEP domain is not directly involved, suggesting that the role of GlyRS in neddylation should not be limited to animals and may exist in all eukaryotes where the modification occurs.
Example 2
GlyRS association with progression of multiple cancer types
[0201] In this example, the association between GlyRS activity and progression of cancer is accessed by analyzing the expression level of GlyRS in multiple cancer patient samples. The results demonstrate that GlyRS is associated with progression of various cancer types, including breast cancer, ovarian cancer, lung cancer, breast duct carcinoma, colorectal adenocarcinoma and lung squamous cell carcinoma. [0202] As shown in Figure 11, high level of GlyRS is associated with rapid breast cancer progression. The Kaplan-Meier plots and hazard ratio (HR) analysis of human tRNA synthetases in breast cancer patients are shown in Figure 1 1. Patient samples were divided in halves as low-expression and high-expression sets for each tRNA synthetase in the analysis, n = 3,557 patients. P values were calculated with two-sided log-rank tests.
[0203] High level of GlyRS was also found to be associated with rapid ovarian cancer progression (see Figure 12), The Kaplan-Meier plots and hazard ratio (HR) analysis of human GlyRS in stage 2 ovarian cancer patients are shown in Figure 12. Patient samples were divided in halves as low-expression and high-expression sets for GlyRS in the analysis, n = 60 patients. P values were calculated with two-sided log-rank tests.
[0204] Figure 13 shows experimental data demonstrating that high level of GlyRS is associated with rapid lung cancer progression. The Kaplan-Meier plots and hazard ratio (HR) analysis of human GlyRS in lung squamous cell carcinoma patients are shown. Patient samples were divided in halves as low-expression and high-expression sets for GlyRS in the analysis, n = 524 patients. P values were calculated with two-sided log-rank tests.
[0205] Also shown in Figures Ί 4Α and 1.4B, higher level of GlyRS staining in most malignant patient cancer tissue samples. As shown in Figure 14 A, high level staining of GlyRS is observed in patient tissue samples of breast duct carcinoma, colorectal adenocarcinoma and lung squamous cell carcinoma. As shown in Figure 14B, most malignant patient cancer tissue samples show higher level of GlyRS expression compared to normal tissue.
Example 3
GlySA targets GlyRS with dual mechani sm
[0206] In this example, the mechanism under which GlySA, a GlyRS inhibitor, inhibits GlyRS, was investigated. Figure 15 shows a non-limiting schematic illustration of GlySA binding to GlyRS active site. GlySA is an analog of Gly-AMP, reaction intermediate of GlyRS. The inhibitor}' effects of GlySA on GlyRS aminoacylation and neddylation were tested in both cell lines and mice models.
Biochemical experiments
[0207] Biochemical experiments were performed to study the inhibiting activity of GlySA to GlyRS. Aminoacylation assay was performed using recombinant human GlyRS (200 nM) proteins at room temperature (RT). MLN4924 is a known inhibitor of neddylation currently used in clinical trials for multiple solid and hematopoietic cancers. ML.N4924 targets the E l enzyme (UBA3) of neddylation.
[0208] As shown in Figisre 16, GlySA (but not MLN4924) inhibits GlyRS aminoacylation. As shown in Figure 17, GlySA decreases GlyRS binding to activated NEDD8 E2 (Ubcl2N8). The interactions of GlyRS to that of Ubcl2N8 were compared in the presence of DMSO or GlySA at 30°C by biolayer interferometry (Octet).
[0209] In vitro NEDD8 activation assay was performed with recombinant human APPBP1-UBA3 (2,7 μΜ) protein and fluorescein -labeled NEDD8 proteins in the reaction buffer at 37°C for 1 hour. The concentration of GlySA and MLN4924 was 300 μΜ. Figisre 18 shows that unlike MLN4924, GlySA does not affect NEDD8 El (UBA3) activation.
Cell experiments
[0210] Cell experiments were also performed to study the inhibiting activity of GlySA to GlyRS. MDA-MB-231 cells at 80% confluence were treated with compounds for overnight and then the ceils were harvested and lysed with the acid lysis buffer and subjected to non-reducing SDS-PAGE. SerSA and TyrSA are analogs of Ser-AMP and Tyr-AMP, reaction intermediate of SerRS and TyrRS, respectively. It was found that GlySA, but not SerSA, TyrSA, inhibits neddylation in MDA-MB-231 cells (Figure 19). Concentration range of GlySA in inhibiting neddylation in MDA-MB-231 cells were also evaluated and the results are shown in Figure 20.
[0211] MDA-MB-231 cells at 80% confluence were treated with 200 nM GlySA and then the cells were harvested at different time points and lysed with the acid lysis buffer and subjected to non-reducing SDS-PAGE, Figure 21 shows experimental data on time course of GlySA in inhibiting neddylation in MDA-MB-231 cells.
[0212] The inhibitory effect of GlySA on key components and substrates of the neddylation pathways were studied using various cancer cell lines. MDA-MB-231 ceils at 80% confluence were treated with compounds for overnight and then the cells were harvested and lysed with the acid lysis buffer and subjected to non-reducing SDS-PAGE. Figure 22 shows GlySA" s effect on key components and substrates of the neddylation pathway in MDA-MB-231 cells. MDA-MB-468 cells at 80% confluence were treated with compounds for overnight and then the cells were harvested and lysed with the acid lysis buffer and subjected to non-reducing SDS-PAGE. Figure 23 shows experimental data on GlySA effect on key components and substrates of neddylation in MDA-MB-468 cells. MCF'7 cells at 80% confluence were also treated with compounds for overnight and then the cells were harvested and lysed with the acid lysis buffer and subjected to non-reducing SDS- PAGE. Figure 24 shows experimental data on GlySA effect on key components and substrates of neddylation in MCF7 cells.
NCI 60 Cell One-Dose Screen
General Description:
[0213] As of early 2007 all compounds submitted to the NCI 60 Cell screen are tested initially at a single high dose (10-5 M) in the full NCI 60 cell panel. Only compounds which satisfy pre-determined threshold inhibition criteria in a minimum number of cell lines will progress to the full 5-dose assay. The threshold inhibition criteria for progression to the 5-dose screen was selected to efficiently capture compounds with anti-proliferative activity based on careful analysis of historical DTP screening data. The threshold criteria may be updated as additional data becomes available.
Interpretation of One-Dose Data:
[0214] The One-dose data will be reported as a mean graph of the percent growth of treated cells and will be similar in appearance to mean graphs from the 5-dose assay. The number reported for the One-dose assay is growth relative to the no-drug control, and relative to the time zero number of cells. This allows detection of both growth inhibition (values between 0 and 100) and lethality (values less than 0). This is the same as for the 5-dose assay, described below. For example, a value of 100 means no growth inhibition, A value of 40 would mean 60% growth inhibition. A value of 0 means no net growth over the course of the experiment. A value of -40 would mean 40% lethality. A value of -100 means all cells are dead. Information from the One-dose mean graph is available for COMPARE analysis.
NCI 60 Cell Five-Dose Screen
General Description:
[0215] Compounds which exhibit significant growth inhibition in the One-Dose Screen are evaluated against the 60 cell panel at five concentration levels,
[0216] The human tumor cell lines of the cancer screening panel grown in RPMI 1640 medium containing 5% fetal bovine serum and 2 mM L-glutamine. For a typical screening experiment, ceils are inoculated into 96 well microtiter plates in 100 uL at plating densities ranging from 5,000 to 40,000 cells/well depending on the doubling time of individual cell lines. After cell inoculation, the microtiter plates are incubated at 37° C, 5 % C02, 95 % air and 100 % relative humidity for 24 h prior to addition of experimental drugs.
[0217] After 24 hours, two plates of each cell line fixed in situ with TCA, to represent a measurement of the cell population for each cell line at the time of drug addition (Tz). Experimental drugs are solubiiized in dimethyl sulfoxide at 400-fold the desired final niaximum test concentration and stored frozen prior to use. At the time of drug addition, an aliquot of frozen concentrate is thawed and diluted to twice the desired final maximum test concentration with complete medium containing 50 ^ig/ml gentamicin. Additional four, 10- fold or ½ log serial dilutions made to provide a total of five drug concentrations plus control. Aiiquots of 100 μΐ of these different drug dilutions are added to the appropriate microtiter wells already containing 100 μΐ of medium, resulting in the required final drug concentrations.
[0218] Following drug addition, the plates are incubated for an additional 48 h at 37°C, 5 % C02, 95 % air, and 100 % relative humidity. For adherent cells, the assay is terminated by the addition of cold TCA. Cells are fixed in situ by the gentle addition of 50 μΐ of cold 50 % (w/v) TCA (final concentration, 10 % TCA) and incubated for 60 minutes at 4°C. The supernatant is discarded, and the plates are washed five times with tap water and air dried. Sulforhodamine B (SRB) solution (100 μΐ) at 0,4 % (w/v) in I % acetic acid is added to each well, and plates are incubated for 10 minutes at room temperature. After staining, unbound dye is removed by washing five times with 1 % acetic acid and the plates are air dried. Bound stain is subsequently solubiiized with 10 mM trizma base, and the absorbance is read on an automated plate reader at a wavelength of 515 nm. For suspension cells, the methodology is the same except that the assay is terminated by fixing settled cells at the bottom of the wells by gently adding 50 μΐ of 80 % TCA (final concentration, 16 % TCA). Using the seven absorbance measurements [time zero, (Tz), control growth, (C), and test growth in the presence of drug at the five concentration levels (Ti)], the percentage growth is calculated at each of the drug concentrations levels. Percentage growth inhibition is calculated as: [(Ti-Tz)/(C-Tz)] x 100 for concentrations for which Τ{>/~Τζ
[(Ti-Tz)/Tz] x 100 for concentrations for which Ti<Tz.
[0219] Three dose response parameters are calculated for each experimental agent. Growth inhibition of 50 % (GI50) is calculated from [(Ti-Tz)/(C-Tz)] x 100 = 50, which is the drug concentration resulting in a 50% reduction in the net protein increase (as measured by SRB staining) in control cells during the dmg incubation. The drug concentration resulting in total growth inhibition (TGI) is calculated from Ti = Tz. The LC50 (concentration of drug resulting in a 50% reduction in the measured protein at the end of the drug treatment as compared to that at the beginning) indicating a net loss of cells following treatment is calculated from [(Ti-Tz)/Tz] x 100 = -50. Values are calculated for each of these three parameters if the level of activity is reached; however, if the effect is not reached or is exceeded, the value for that parameter is expressed as greater or less than the maximum or minimum concentration tested.
[0220] The general procedures of NCI 60 Cell One-Dose Screen and NCI 60 Cell Five-Dose Screen were used to evaluate the inhibitor}' effect of GlySA on the growth of various cancer ceil lines. The results are shown in Table 2 (one-dose screen) and Table 3 (five-dose screen) below.
Table 2. List of cancer cell lines sorted according to the GlySA growth inhibition effect.
Panel name Cell name Growth inhibition count (%}
Renal Cancer A498 -58.80134788
Non-Small Cell Lung Cancer NCI-H226 -54.57056308
!Nion-Smali Cell Lung Cancer NCI-H522 -51.58866995
Melanoma MDA-MB-435 -49.81617647
Melanoma LOX IMVI -45.56709265
CNS Cancer U251 -39.33823529
Renal Cancer RXF 393 -36.59850484
Melanoma S -MEL-2 -34.37833511
Breast Cancer MDA-MB-468 -34.302595
CMS Cancer SF-295 -33.68333333
Non-Small Cell Lung Cancer HOP-62 -32.11206897
Prostate Cancer DU-145 -31.72348485
Melanoma M14 -23.10222672
Ovarian Cancer OVCAR-4 -22.10026212
Ovarian Cancer OVCA -8 -11.91763727 CNS Cancer SF-! -9.360885276
Leukemia HL~ SG(TB) -9.284776903
Renal Cancer 786 -0 -7.021837349
CNS Cancer S!SSE ,-75 -6.717877095
CNS Cancer s ,-19 -6.057336621
Melanoma MA LME-3IV1 -2.822580645
Non-Small Cell Lung Cancer NO -H322M -1.738410596
Melanoma S -! V1EL-5 -0.3413371
Ovarian Cancer OV< :AR-S 0.291560931
Colon Cancer KM 12 0.734685773
Renal Cancer SU1 2C 1.029729281
Leukemia MO LT-4 1.145822073
Ovarian Cancer iMC! /ADR-RES 2.072589755
Breast Cancer MD A-MB-231/ATCC 2.607853364
Non-Small Cell Lung Cancer NO -H460 3.350093377
Colon Cancer SW- 620 3.712061898
CNS Cancer SF-2 568 3.776035626
Renal Cancer CAK If-1 5.110152835
Non-Small Cell Lung Cancer HCi H23 5.695418902
Colon Cancer HT2 9 6.002098636
Renal Cancer UO- 31 6.083971978
Ovarian Cancer S -i 3V-3 6.281150543
Colon Cancer HO -116 6.566078895
Leukemia -5 52 7.376219586
Breast Cancer T-4" 7D 7.655948339
Breast Cancer MC F7 8.271239384
Melanoma S -! V!EL-28 8.357448779
Prostate Cancer PC- 3 9.662392044
Non-Small Cell Lung Cancer 9/ATCC 9.701463415
Leukemia S 9.925740754
Ovarian Cancer OVi :AR-3 10.04947249
Colon Cancer HO -15 11.41530732
Ovarian Cancer IGR OVI 11.70363841
Renal Cancer ACh \u 11.7925978
Breast Cancer HS ! 578T 16.07429649
Non-Small Cell Lung Cancer HO! 3-92 17.26373251
Leukemia
Figure imgf000071_0001
18.03788521
Breast Cancer ΒΪ- 549 21.05943912
Leukemia CC F-CEM 21.67622168
Melanoma UA X-257 27.43273074
Melanoma UA< -62 30.70756508
Colon Cancer HCC :-2998 41.35051734
Non-Small Cell Lung Cancer E V X 44.07083614 Colon Cancer 48.50851164
3. List of cancer cell lines sorted according to the GlySA (LC50/GI50).
Pane! name Ceil name GIso (μΜ) LCso (μΜ) LCSa G!so
Non-Small Cell Lung Cancer HOP-62 0.372 >100 >269.2
CN5 Cancer SF-268 0.501 >100 >199.5
Renal Cancer SN12C 0.562 95.50 169.8
Prostate Cancer DU-145 0.692 >100 >144.5
Ovarian Cancer OVCAR-5 0.741 >100 > 134.9
Leukemia SR 0.933 >100 >107.2
Ovarian Cancer OVCA -4 0.389 23.99 61.7
Breast Cancer T-47D 2,344 >100 >42.7
Ovarian Cancer OVCAR-8 2.455 >100 >40.7
Ovarian Cancer G/ADR-F IES 2.512 >100 >39.8
Non-Small Cell Lung Cancer E VX 2.570 >100 >38.9
Non-Small Cell Lung Cancer !MG-H23 2.570 >100 >38.9
Leukemia MOLT-4 2,630 >100 >38.0
Leukemia HL-60{TB) 2.754 >100 >36.3
Breast Cancer IV1CF7 2.818 >100 >35.5
Breast Cancer HS 578T 2.884 >100 >34.7
Breast Cancer IVIDA-MB- 231/ATCC 0.525 18.20 34.7
Colon Cancer HT29 2.951 >100 >33.9
Colon Cancer HCT-116 3.162 >100 >31.6
Colon Cancer KM12 3.236 >100 >30.9
Non-Small Cell Lung Cancer O-H46G 3.236 >100 >30.9
Leukemia RFM!-S22i 3.311 >100 >30.2
Colon Cancer COLO 205 2.291 64.57 28.2
Colon Cancer HCT-15 2,951 81.28 27.5
Leukemia CCRF-CEIVl 3.715 >100 >26.9
Leukemia -562 3.715 >100 >26.9
Colon Cancer SW-620 3.715 >100 >26.9
Prostate Cancer PC-3 2.630 56.23 21.4
CMS Cancer SNB-19 0,380 6.92 18.2
Ovarian Cancer fGROVl 3.020 47.86 15.8
Non-Small Cell Lung Cancer O-H322l¾ A 1.514 22.91 15.1
Ovarian Cancer S -OV-3 2.884 37.15 12.9
Renal Cancer ACH!M 2.884 33.88 11.7
Breast Cancer BT-549 2.344 25.12 10.7
CMS Cancer U251 0.550 5.75 10.5
Breast Cancer IVIDA-MB- $68 0.741 7.59 10.2
Non-Small Cell Lung Cancer HOP-92 2.951 27.54 9.3 Renal Cancer T -10 2,754 23.44 8.5
CN5 Cancer SNB-75 1.148 9.55 8.3
Non-Small Cell Lung Cancer A549/ATCC 2.344 19.05 8.1
CMS Cancer 0.871 6.92 7.9
CMS Cancer SF-539 1.445 8.91 6.2
Ovarian Cancer OVCA -3 1,995 10.00 5.0
Non-Small Cell Lung Cancer rra~H522 1,738 8.51 4.9
Colon Cancer HCC-2998 2.042 9.33 4.6
Melanoma S -MEL-28 1.950 8.91 4.6
Melanoma S -MEL-2 2.239 10.00 4,5
Melanoma LOX !MV! 1,698 7.41 4.4
Melanoma UACC-62 1,862 7.76 4.2
Renal Cancer A498 1.585 6.46 4.1
Melanoma fVlALfVlE-3!Vl 1.905 7.76 4.1
Melanoma IV114 1.820 7.24 4.0
Renal Cancer RXF 393 2.138 8.32 3.9
Renal Cancer 786-0 1,778 6.76 3.8
Melanoma MDA-MB-435 1,820 6.61 3.6
Renal Cancer UO-31 1.698 6.17 3.6
Non-Small Cell Lung Cancer O-H226 1.995 7.08 3.5
Melanoma S -MEL-5 1.862 6.03 3.2
Mice experiments
[0221] Animal experiments were also performed to study the use of GlySA to treat cancer. A maximum tolerant dosage assay of GlySA was conducted. GlySA (DMSO stock solution diluted by saline) were administrated to three month old female BALB CJ mice via tail vein injections. Mice after four injections were evaluated and ail were alive. The GlySA concentration tested were 0.4 mg/kg (10 μΜ), 2.0 mg/kg (50 μΜ), 4.0 mg/kg (100 uM). N=3 for each group. A schematic illustration of the assay is shown in Figure 25.
[0222] Figure 26 shows a schematic illustration of lung metastasis assay methods conducted. 1 χ 105 MDA-MB-231 cells were injected via tail vein to NOD.Cg-Prkdcwid I12rg mice. Then mice were separated into 3 groups. Group A: vehicle alone (PBS withl% DMSO), group B: GlySA (4 mg/kg; 100 μΜ), and group C: MLN4924 GlySA (4.4 mg/kg, 100 uM) were administrated via tail vein injections twice per week. N=10 for each group.
[0223] It was found that GlySA treatment reduces lung metastasis in mice (Figure 27). Top panel of Figure 27 shows mice lungs 14 days after tumor cells (MDA-MB- 231) injection. White dots show the surface tumor colonies. Bottom panel of Figure 27 shows numbers of lung metastasis colonies are analyzed by two tails unpaired T test. The error bars represent SEM (ι =;:8-10),
[0224] In at least some of the previously described embodiments, one or more elements used in an embodiment can interchangeably be used in another embodiment unless such a replacement is not technically feasible. It will be appreciated by those skilled in the art that various other omissions, additions and modifications may be made to the methods and structures described above without departing from the scope of the claimed subject matter. All such modifications and changes are intended to fall within the scope of the subject matter, as defined by the appended claims.
[0225] With respect to the use of substantially any plural and/or singular terms herein, those having skill in the art can translate from the plural to the singular and/or from the singular to the plural as is appropriate to the context and/or application. The various singular/plural permutations may be expressly set forth herein for sake of clarity.
[0226] It will be understood by those within the art that, in general, terms used herein, and especially in the appended claims (e.g., bodies of the appended claims) are generally intended as "open" terms (e.g., the term "including" should be interpreted as "including but not limited to," the term "having" should be interpreted as "having at least," the term "includes" should be interpreted as "includes but is not limited to," etc.). It will be further understood by those within the art that if a specific number of an introduced claim recitation is intended, such an intent will be explicitly recited in the claim, and in the absence of such recitation no such intent is present. For example, as an aid to understanding, the following appended claims may contain usage of the introductory phrases "at least one" and "one or more" to introduce claim recitations. However, the use of such phrases should not be construed to imply that the introduction of a claim recitation by the indefinite articles "a" or "an" limits any particular claim containing such introduced claim recitation to embodiments containing only one such recitation, even when the same claim includes the introductory phrases "one or more" or "at least one" and indefinite articles such as "a" or "an" (e.g., "a" and/or "an" should be interpreted to mean "at least one" or "one or more"); the same holds taie for the use of definite articles used to introduce claim recitations. In addition, even if a specific number of an introduced claim recitation is explicitly recited, those skilled in the art will recognize that such recitation should be interpreted to mean at least the recited number (e.g., the bare recitation of "two recitations," without other modifiers, means at least two recitations, or two or more recitations). Furthermore, in those instances where a convention analogous to "at least one of A, B, and C, etc." is used, in general such a construction is intended in the sense one having skill in the art would understand the convention (e.g., " a system having at least one of A, B, and C" would include but not be limited to systems that have A alone, B alone, C alone, A and B together, A and C together, B and C together, and/or A, B, and C together, etc.). In those instances where a convention analogous to "at least one of A, B, or C, etc." is used, in general such a construction is intended in the sense one having skill in the art would understand the convention (e.g., " a system having at least one of A, B, or C" would include but not be limited to systems that have A alone, B alone, C alone, A and B together, A and C together, B and C together, and/or A, B, and C together, etc.). It will be further understood by those within the art that virtually any disjunctive word and/or phrase presenting two or more alternative terms, whether in the description, claims, or drawings, should be understood to contemplate the possibilities of including one of the terms, either of the terms, or both terms. For example, the phrase "A or B" wil l be understood to include the possibilities of "A" or "B" or "A and B."
[0227] In addition, where features or aspects of the disclosure are described in terms of Markush groups, those skilled in the art will recognize that the disclosure is also thereby described in terms of any individual member or subgroup of members of the Markush group.
[0228] As will be understood by one of skill in the art, for any and all puiposes, such as in terms of providing a written description, all ranges disclosed herein also encompass any and all possible sub-ranges and combinations of sub-ranges thereof. Any listed range can be easily recognized as sufficiently describing and enabling the same range being broken down into at least equal halves, thirds, quarters, fifths, tenths, etc. As a non- limiting example, each range discussed herein can be readily broken down into a lower third, middle third and upper third, etc. As will also be understood by one skilled in the art all language such as "up to," "at least," "greater than," "less than," and the like include the number recited and refer to ranges which can be subsequently broken down into sub-ranges as discussed above. Finally, as will be understood by one skilled in the art, a range includes each individual member. Thus, for example, a group having 1-3 articles refers to groups having 1, 2, or 3 articles. Similarly, a group having 1 -5 articles refers to groups having 1 , 2, 3, 4, or 5 articles, and so forth.

Claims

WHAT IS CLAIMED IS:
1. A method of reducing neddyiation in a cell , the method comprises:
contacting a cell with a composition comprising a Glycyl-tRNA synthetase
(GlyRS) inhibitor,
wherein the level of neddyiation is decreased in the ceil.
2. A method of reducing neddyiation in a cell population, the method comprises: acquiring knowledge of the level of neddyiation in a cell population; and contacting the cell population with a composition comprising a Glycyl-tRNA synthetase (GlyRS) inhibitor to thereby decrease the level of neddyiation in the cell population.
3. A method of reducing neddyiation in a cell population, the method comprises: identifying a cell population having undesirable level of neddyiation; and contacting the cell population with a composition comprising a Glycyl-tRNA synthetase (GlyRS) inhibitor to thereby decrease the level of neddyiation in the cell population.
4. The method of any one of Claims 1-3, wherein the composition is a pharmaceutical composition.
5. The method of any one of Claims 1-4, wherein the GlyRS inhibitor inhibits GlyRS functions in aminoacylation and neddyiation.
6. The method of any one of Claims 1-4, wherein the GlyRS inhibitor does not significantly inhibit GlyRS function in aminoacylation.
7. The method of any one of Claims 1-6, wherein the GlyRS inhibitor is an inhibitor for a mammalian GlyRS gene product.
8. The method of any one of Claims 1-6, wherein the GlyRS inhibitor is an inhibitor for a human GlyRS gene product.
9. The method of Claim 8, wherein the human GlyRS gene product comprises an amino acid sequence having at least 90% identity to the amino acid sequence set forth in SEQ ID NO: 2.
10. The method of any one of Claims 1-6, wherein the GlyRS inhibitor is an inhibitor for a plant GlyRS gene product.
1 1. The method of any one of Claims 1-10, wherein the GlyRS inhibitor is a protein, a nucleic acid, a small molecule compound, or a combination thereof.
12. The method of Claim 11, wherein the GlyRS inhibitor is an RNA molecule capable of silencing the expression of a GlyRS gene.
13. The method of Claim 1 1, wherein the GlyRS inhibitor is an RNA molecule that binds to an mRNA encoded by a GlyRS gene.
14. The method of Claims 12 or 13, wherein the GlyRS gene is a mammalian GlyRS gene.
15. The method of Claims 12 or 13, wherein the GlyRS gene is the human GARS gene.
16. The method of Claim 15, wherein the human GARS gene comprises a nucleotide sequence having at least 90% sequence identity to the nucleotide sequence set forth in SEQ ID NO: I .
17. The method of Claim 16, wherein the GlyRS inhibitor is a short-hairpin RNA (shRNA) comprising a nucleotide sequence having at least 90% sequence identity to the nucleic acid sequence of SEQ ID NOs:3-35.
18. The method of any one of Claims 1-4, wherein the GlyRS inhibitor is GiySA or a derivative thereof.
19. The method of any one of Claims 1-18, wherein the GlyRS inhibitor interferes with the binding between the GlyRS protein and NEDD8 in the cell.
20. The method of any one of Claims 1-18, wherein the GlyRS inhibitor interferes with the binding between the GlyRS protein and Ubcl2 in the cell.
21. The method of any one of Claims 1-18, wherein the GlyRS inhibitor interferes with the binding between the GlyRS protein and NEDDS-conjugated Ubcl2 in the ceil.
22. The method of Claim 21 , wherein the GlyRS inhibitor binds to one or more of amino acids 84-93 of SEQ ID NO: 2.
23. The method of Claim 21, wherein the GlyRS inhibitor binds to one or more of amino acids 232-238 of SEQ ID NO: 2.
24. The method of Claim 21 , wherein the GlyRS inhibitor binds to one or more of amino acids Arg277, Glu279, Vai289, Glu296, Ile404, and Glu552 of SEQ ID NO: 2.
25. The method of Claim 21, wherein the GlyRS inhibitor is a GlySA derivative.
26. The method of any one of Claims 1-25, wherein the GlyRS inhibitor decreases the amount of NEDD8-conjugated Ubcl 2 in the cell,
27. The method of any one of Claims 1 -26, wherein the GlyRS inhibitor decreases Ubcl2 activity in the cell.
28. The method of any one of Claims 1-27, wherein the GlyRS inhibitor increases Ubcl2 degradation in the cell.
29. The method of any one of Claims 1-28, wherein the GlyRS inhibitor interferes with the binding between a GlyRS protein and heterodimeric El enzyme for neddylation (APPBP1/UBA3) in the cell.
30. The method of any one of Claims 1-29, wherein the GlyRS inhibitor decreases neddylation of a cullin protein in the cell.
31. The method of any one of Claims 1 -30, wherein the cell is a mammalian cell.
32. The method of any one of Claims 1-31, wherein the contacting is performed in vitro, ex vivo, or in vivo.
33. The method of any one of Claims 1-31, wherein the ceil or the ceil population is present in a tissue or in a body of a subject.
34. The method of any one of Claims 1-33, wherein the level of neddylation in the cell or the cell population is reduced by at least 50%.
35. An isolated double-stranded ribonucleic acid (dsRNA) molecule that inhibits expression of a Glycyl-tRNA synthetase (GlyRS) gene, wherein a first strand of the dsRNA is substantially identical to at least 19 consecutive nucleotides of the GlyRS gene, and a second strand of the dsRNA is substantially complementary to the first strand.
36. The isolated dsRNA molecule of Claim 35, wherein the GlyRS gene is a mammalian GlyRS gene.
37. The isolated dsRNA molecule of Claim 36, wherein the GlyRS gene is the human GARS gene.
38. The isolated dsRNA molecule of Claim 37, wherein the human GARS gene comprises a nucleotide sequence having at least 90% identity to the nucleotide sequence set forth in SEQ ID NO: l .
39. The isolated dsRNA molecule of Claim 35, wherein the GlyRS gene is a plant GlyRS gene.
40. The isolated dsRNA molecule of any one of Claims 35-39, wherein the dsRNA is encoded by a polynucleotide, wherein the first strand and the second strand of the dsRNA are transcribed from said poiynucleotide and form a hairpin loop.
41. An isolated single stranded oligonucleotide that is complementary to a portion of a Glycyl-tRNA synthetase (GlyRS) gene of at least 10 consecutive nucleotides.
42. The isolated single stranded oligonucleotide of Claim 41, wherein the GlyRS gene is a mammalian GlyRS gene.
43. The isolated single stranded oligonucleotide of Claim 42, wherein the GlyRS gene is the human GARS gene.
44. The isolated single stranded oligonucleotide of Claim 43, wherein the human GARS gene comprises a nucleotide sequence having at least 90% identity to the nucleotide sequence set forth in SEQ ID NO: 1.
45. The isolated single stranded oligonucleotide of Claim 41, wherein the GlyRS gene is a plant GlyRS gene.
46. A method of identifying an inhibitor of neddylation comprising:
providing a test compound;
testing the testcompound for its ability to reduce or inhibit the binding between a Glycyl-tRNA synthetase (GlyRS) protein and NEDD8-conjugated Ubcl 2; and
identifying the compound as an inhibitor of neddylation if the compound has the ability to reduce or inhibit the binding between the GlyRS protein and NEDD8- conjugated Ubcl2.
47. The method of Claim 46, wherein the inhibitors bind to the catalytic domain of the GlyRS protein.
48. The method of Claim 46 or 47, wherein the GlyRS protein is a mammalian GlyRS protein.
49. The method of Claim 46 or 47, wherein the GlyRS protein is a human GlyRS protein.
50. The method of Claim 49, wherein the human GlyRS protein comprises an amino acid sequence having at least 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:2.
51. The method of any one of Claims 46-50, further comprising testing the test compound for its ability to reduce or inhibit the aminoacylation activity of the GlyRS protein.
52. The method of any one of Claims 46-51, further comprising testing one or more additional test compounds for their ability to reduce or inhibit the binding between the GlyRS protein and NEDDS-conjugated Ubcl2.
53. The method of Claim 52, further comprising testing the one or more additional test compounds for their ability to reduce or inhibit the aminoacylation activity of the GlyRS protein.
54. A pharmaceutical composition comprising a Glycyl-tRNA synthetase (GlyRS) inhibitor and a pharmaceutically acceptable excipient.
55. The pharmaceutical composition of Claim 54, wherein the GlyRS inhibitor is an isolated siRNA molecule that binds to an mRNA of the GlyRS protein.
56. The pharmaceutical composition of Claim 54, wherein the GlyRS inhibitor is a molecule that inhibits binding between the GlyRS protein and NEDDS-conjugated Ubcl2.
57. The pharmaceutical composition of Claim 54, wherein the GlyRS inhibitor is GiySA or a derivative thereof.
58. The method of Claim 54, wherein the GlyRS inhibitor inhibits GlyRS functions in aminoacylation and neddylation.
59. The method of Claim 54, wherein the GlyRS inhibitor does not significantly inhibit GlyRS function in aminoacylation.
60. A method of reducing cell proliferation, comprising:
contacting a cell with a composition comprising a Glycyl-tRNA synthetase (GlyRS) inhibitor,
whereby the proliferation of the cell is reduced,
61. The method of Claim 60, wherein the activity of the CRL1 (cullinl-RTNG) ubiquitin ligases is inhibited in the cell.
62. The method of Claim 61, wherein the activity of a substrate of the CRL1 ubiquitin ligase is increased in the cell.
63. The method of Claim 62, wherein the substrate of the CRL1 ubiquitin ligase is selected from the group consisting of c-Myc, c-Jun, cyclin E, Emi l, Cdt-1, ρϊκΒα, NRF2, 1 11!··· l , β-catenin, Cdc25A, mTOR, BimEL and p27.
64. The method of any one of Claims 60-63, further comprising providing MLN4924 to the cell.
65. The method of any one of Claims 60-64, wherein the cell is a mammalian cell.
66. The method of any one of Claims 60-64, wherein the cell is a plant cell.
67. The method of any one of Claims 60-66, wherein the proliferation of the cell is reduced by at least 50%.
68. The method of Claim 60, wherein the GlyRS inhibitor inhibits GlyRS functions in aminoacylation and neddylation.
69. The method of Claim 60, wherein the GlyRS inhibitor does not significantly inhibit GlyRS function in aminoacylation.
70. A method of treating or ameliorating cancer in a subject, comprising:
administering a therapeutically effective amount of a pharmaceutical composition comprising a Glycyl-tRNA synthetase (GlyRS) inhibitor to a subject in need thereof.
71. The method of Claim 70, wherein the pharmaceutical composition further comprises one or more of additional therapeutic agents.
72. The method of Claim 70, further comprising administering one or more additional pharmaceutical compositions comprising one or more of additional therapeutic agents.
73. The method of any one of Claims 70-72, wherein the cancer is breast cancer, ovarian cancer, lung cancer, breast duct carcinoma, colorectal adenocarcinoma and lung squamous cell carcinoma, or a combination thereof.
74. The method of any one of Claims 7072, wherein the cancer is selected from the group consisting of breast cancer, cervical cancer, colon cancer, liver cancer, prostate cancer, melanoma, ovarian cancer, lung cancer, renal cell carcinoma, Schwannoma, mesothelioma, acute myeloid leukemia, multiple myeloma, non-Hodgkin lymphoma, and a combination thereof.
75. The method of Claim 70, wherein the cancer is a solid tumor.
76. The method of Claim 70, wherein the cancer is a hematological malignancy.
77. The method of any one of Claims 70-75, wherein the GlyRS inhibitor is GiySA or a GiySA derivative.
78. The method of any one of Claims 70-75, wherein the GlyRS inhibitor inhibits GiyRS functions in aminoacylation and neddylation.
79. The method of any one of Claims 70-75, wherein the GlyRS inhibitor does not significantly inhibit GlyRS function in aminoacylation.
80. The method of any one of Claims 70-75, wherein the GiyRS inhibitor is an inhibitor for a human GlyRS.
PCT/US2016/056862 2015-10-14 2016-10-13 INHIBITION OF NEDDYLATION USING GLYCYL-tRNA SYNTHETASE INHIBITORS WO2017066459A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP16856197.5A EP3362086A1 (en) 2015-10-14 2016-10-13 INHIBITION OF NEDDYLATION USING GLYCYL-tRNA SYNTHETASE INHIBITORS
US15/767,627 US20190381086A1 (en) 2015-10-14 2016-10-13 INHIBITION OF NEDDYLATION USING GLYCYL-tRNA SYNTHETASE INHIBITORS

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201562241386P 2015-10-14 2015-10-14
US62/241,386 2015-10-14

Publications (1)

Publication Number Publication Date
WO2017066459A1 true WO2017066459A1 (en) 2017-04-20

Family

ID=58518556

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2016/056862 WO2017066459A1 (en) 2015-10-14 2016-10-13 INHIBITION OF NEDDYLATION USING GLYCYL-tRNA SYNTHETASE INHIBITORS

Country Status (3)

Country Link
US (1) US20190381086A1 (en)
EP (1) EP3362086A1 (en)
WO (1) WO2017066459A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111066727A (en) * 2019-12-20 2020-04-28 中国人民解放军陆军军医大学 Method for constructing mouse model of action mechanism in permeability of hypoxic blood testis barrier

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023218306A1 (en) * 2022-05-09 2023-11-16 Giam Pharma International sarl Antitumor activity of meso-(p-acetamidophenyl)-calix[4]pyrrole by inhibition of the enzymatic activity of the protein glycyl-trna synthase 1 (gars1)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060204508A1 (en) * 2003-01-23 2006-09-14 Champion Brian R Treatment of autoimmune diseases using an activator for the notch signalling pathway
WO2012158945A2 (en) * 2011-05-19 2012-11-22 The Scripps Research Institute Compositions and methods for treating charcot-marie-tooth diseases and related neuronal diseases
US20140220596A1 (en) * 2011-10-10 2014-08-07 Medicinal Bioconvergence Research Center Method for screening an agent preventing or treating cancer using glycyl-trna synthetase and cadherin
US20140363415A1 (en) * 2008-06-26 2014-12-11 Atyr Pharma, Inc. Compositions and methods comprising glycyl-trna synthetases having non-canonical biological activities

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060204508A1 (en) * 2003-01-23 2006-09-14 Champion Brian R Treatment of autoimmune diseases using an activator for the notch signalling pathway
US20140363415A1 (en) * 2008-06-26 2014-12-11 Atyr Pharma, Inc. Compositions and methods comprising glycyl-trna synthetases having non-canonical biological activities
WO2012158945A2 (en) * 2011-05-19 2012-11-22 The Scripps Research Institute Compositions and methods for treating charcot-marie-tooth diseases and related neuronal diseases
US20140220596A1 (en) * 2011-10-10 2014-08-07 Medicinal Bioconvergence Research Center Method for screening an agent preventing or treating cancer using glycyl-trna synthetase and cadherin

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
BROWNELL ET AL.: "Substrate-Assisted Inhibition of Ubiquitin-Like Protein-Activating Enzymes: the NEDD8 E1 Inhibitor MLN4024 Forms a NEDDO-AMP Mimetic in Situ''.", MULECULAR CELL, vol. 37, no. 1, 15 January 2010 (2010-01-15), pages 102 - 11, XP055376930 *
GUO ET AL.: "Architecture and Metamorphosis''.", IN AMINOACYL-TRNA SYNTHETASES IN BIOLOGY AND MEDICINE, vol. 344, 2013, pages 89 - 118, XP055376940 *
MO ET AL.: "Neddylation Requires Glycyl-tRNA Synthetase to Protect Activated E2''.", NATURE STRUCTURAL & MOLOOULOF DIALOGY., vol. 23, 27 June 2016 (2016-06-27), pages 730 - 737, XP055376935 *
MOCIBOB ET AL.: "Homologs of Aminoacyl-tRNA Synthetases Acylate Carrier Proteins and Provide a Link between Ribosomal and Nonribosomal Peptide Synthesis.", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, vol. 107, no. 33, 17 August 2010 (2010-08-17), pages 14585 - 14590, XP055376934 *
SOUCY ET AL.: "The NEDD8 Conjugation Pathway and its Relevance in Cancer Biology and Therapy''.", GENES & CANCER., vol. 1, no. 7, 1 July 2010 (2010-07-01), pages 708 - 716, XP055151062 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111066727A (en) * 2019-12-20 2020-04-28 中国人民解放军陆军军医大学 Method for constructing mouse model of action mechanism in permeability of hypoxic blood testis barrier
CN111066727B (en) * 2019-12-20 2021-08-27 中国人民解放军陆军军医大学 Method for constructing mouse model of action mechanism in permeability of hypoxic blood testis barrier

Also Published As

Publication number Publication date
EP3362086A1 (en) 2018-08-22
US20190381086A1 (en) 2019-12-19

Similar Documents

Publication Publication Date Title
KR102061353B1 (en) Inhibitors of human ezh2, and methods of use thereof
US20100267626A1 (en) Methods and compositions for measuring wnt activation and for treating wnt-related cancers
US20190269761A1 (en) Methods, compositions and screens for therapeutics for the treatment of synovial sarcoma
Diviani et al. Small-molecule protein-protein interaction inhibitor of oncogenic Rho signaling
CN102770767A (en) Methods and compounds for muscle growth
US9493813B2 (en) Modulation of phosphatidylinositol-5-phosphate-4-kinase activity
Lamothe et al. Chapter five-ubiquitination of ion channels and transporters
Luo et al. Human ClpP protease, a promising therapy target for diseases of mitochondrial dysfunction
JP2020527569A (en) Compositions and Methods for Preventing and Treating Radiation-Induced Bystander Effects Caused by Radiation or Radiation Therapy
Konovalova et al. Exposure to arginine analog canavanine induces aberrant mitochondrial translation products, mitoribosome stalling, and instability of the mitochondrial proteome
Zhang et al. Epigenetic regulation of Wnt signaling by carboxamide-substituted benzhydryl amines that function as histone demethylase inhibitors
US20190381086A1 (en) INHIBITION OF NEDDYLATION USING GLYCYL-tRNA SYNTHETASE INHIBITORS
WO2013063412A2 (en) Methods of diagnosis and treatment of endoplasmic reticulum (er) stress-related conditions
Göricke et al. Discovery and characterization of BAY-805, a potent and selective inhibitor of ubiquitin-specific protease USP21
US20160052918A1 (en) Small compounds targeting tacc3
Makovski et al. Down-regulation of Fer induces ROS levels accompanied by ATM and p53 activation in colon carcinoma cells
US9382330B2 (en) Pdia4, target of cytopiloyne derivatives, for tumor diagnosis and treatment
US8980820B2 (en) Fatty acid binding proteins as drug targets for endocannabinoids
US9752151B2 (en) Composition for treatment or metastasis suppression of cancers which includes p34 expression inhibitor or activity inhibitor as active ingredient
Lin et al. RTN1-C is involved in high glucose-aggravated neuronal cell subjected to oxygen-glucose deprivation and reoxygenation injury via endoplasmic reticulum stress
US20190167771A1 (en) CONTROL ANGIOGENESIS BY REGULATING PHOSPHORYLATION OF SERYL-tRNA SYNTHETASE (SerRS)
Zhang et al. Tgm2 alleviates LPS-induced apoptosis by inhibiting JNK/BCL-2 signaling pathway through interacting with Aga in macrophages
WO2020165570A1 (en) Methods relating to disrupting the binding of af9 partner proteins to af9 and/or enl
US20230280334A1 (en) Cdc20 variants resistant to anti-mitotic drugs and related methods and compositions
WO2022197978A1 (en) Compositions and methods for modulating mitochondrial function and biogenesis

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: 16856197

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2016856197

Country of ref document: EP