WO2019195854A1 - Compositions and methods for treating phenylketonuria - Google Patents

Compositions and methods for treating phenylketonuria Download PDF

Info

Publication number
WO2019195854A1
WO2019195854A1 PCT/US2019/026399 US2019026399W WO2019195854A1 WO 2019195854 A1 WO2019195854 A1 WO 2019195854A1 US 2019026399 W US2019026399 W US 2019026399W WO 2019195854 A1 WO2019195854 A1 WO 2019195854A1
Authority
WO
WIPO (PCT)
Prior art keywords
pathway
gene
signaling
expression
cell
Prior art date
Application number
PCT/US2019/026399
Other languages
French (fr)
Inventor
David A. Bumcrot
Alfica Sehgal
Alla SIGOVA
Gavin WHISSELL
Vaishnavi RAJAGOPAL
Original Assignee
Camp4 Therapeutics Corporation
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 Camp4 Therapeutics Corporation filed Critical Camp4 Therapeutics Corporation
Publication of WO2019195854A1 publication Critical patent/WO2019195854A1/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
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/20Carboxylic acids, e.g. valproic acid having a carboxyl group bound to a chain of seven or more carbon atoms, e.g. stearic, palmitic, arachidic acids
    • A61K31/203Retinoic acids ; Salts thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1858Platelet-derived growth factor [PDGF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1875Bone morphogenic factor; Osteogenins; Osteogenic factor; Bone-inducing factor
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/191Tumor necrosis factors [TNF], e.g. lymphotoxin [LT], i.e. TNF-beta
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/30Insulin-like growth factors (Somatomedins), e.g. IGF-1, IGF-2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • 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
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers

Definitions

  • the present invention provides compositions and methods for the treatment of phenylalanine hydroxylase deficiency, such as phenylketonuria, in humans.
  • Phenylketonuria is caused by a defect in the Phenylalanine Hydroxylase (PAH) gene and due to a buildup of phenylalanine in the blood.
  • Phenylalanine hydroxylase is an enzyme that catalyzes the hydroxylation of phenylalanine to generate tyrosine. Mutations in the PAH gene reduce the activity of phenylalanine hydroxylase and prevent the breakdown of phenylalanine. As a result, phenylalanine builds up to toxic levels in the blood and other tissues, causing brain damage and other serious medical problems.
  • Symptoms include behavioral problems, seizures, mental and growth retardation. The most severe form of this disorder is known as classic PKU, which occurs when phenylalanine hydroxylase activity is severely reduced or absent. Several milder versions of PKU and non-PKU hyperphenylalaninemia are often due to partial deficiency of the PAH enzyme.
  • KUVAN® distalin dihydrochloride
  • KUVAN® is the only prescription medication for PKU.
  • KUVAN® works by adding more tetrahydrobiopterin (BH4), an essential cofactor of PAH, and stimulating the PAH enzyme to process phenylalanine in people with PKU.
  • BH4 tetrahydrobiopterin
  • PAH PAH
  • the present invention provides novel targets, compositions and treatment methods for PKU.
  • the present invention discloses the mapping and identification of gene signaling network(s) associated with the Phenylalanine Hydroxylase (PAH) gene, which has been linked to diseases of phenylalanine hydroxylase deficiency such as phenylketonuria.
  • PAH Phenylalanine Hydroxylase
  • the inventors By perturbing the components of the gene signaling network(s), the inventors have identified novel targets, compounds and/or methods that could be utilized to modulate PAH expression.
  • Such methods and compositions may be used to develop various therapies for a PAH-related disorder (e.g., phenylketonuria) to prevent and/or alleviate the symptoms of such a disease.
  • a method of treating a subject with phenylalanine hydroxylase deficiency by administering to the subject an effective amount of a compound capable of modulating the expression of the PAH gene.
  • a compound capable of modulating the expression of the PAH gene may be a small molecule, a polypeptide, an antibody, a hybridizing oligonucleotide, and a genome editing agent.
  • the compound administered to the subject may include an inhibitor of the JAK/STAT pathway.
  • Such compound may include at least one of Ruxolitinib, Oclacitinib, Baricitinib, Filgotinib, Gandotinib, Lestaurtinib, PF-04965842, Upadacitinib, Cucurbitacin I, CHZ868, Fedratinib, AC430, AT9283, ati-5000l and ati- 50002, AZ 960, AZD1480, BMS-911543, CEP-33779, Cerdulatinib (PRT062070, PRT2070), Curcumol, Decernotinib (VX-509), Fedratinib (SAR302503, TG101348), FLLL32, FM-381, GLPG0634 analogue, Go6976, JANEX-l (WHI-P131), Momelotinib (CYT387), NVP-
  • the compound administered to the subject may include an inhibitor of the Tyrosine kinase/MAPK pathway.
  • Such compound may include at least one of Amuvatinib, Bosutinib, Cediranib, Ceritinib, CP-673451, Dasatinib, GZD824 Dimesylate, Merestinib, R788 (fostamatinib disodium hexahydrate), or a derivative or an analog thereof.
  • the compound administered to the subject may include 17- AAG (Tanespimycin), Amlodipine Besylate, ATRA (all-trans retinoic acid), Chloroquine phosphate, Deoxycorticosterone, Darapladib, Echinomycin, Enzastaurin, Epinephrine, EVP-6124 (hydrochloride) (encenicline), EW-7197, FRAX597, Ibrutinib, Perphenazine, Phenformin, PND-1186, Rifampicin, Semagacestat, Thalidomide, WAY600, WYE-125132 (WYE-132), Zibotentan, Activin, Anti mullerian hormone, GDF10 (BMP3b), IGF-l,
  • the compound administered to the subject may include one or more RNAi agents against a signaling molecule identified to regulate PAH expression.
  • the compound includes one or more small interfering RNA (siRNA) targeting one or more genes selected from the group consisting of JAK1, JAK2, PDGFRA, PDGFRB, SRC and ABL.
  • the compound increases the expression of the PAH gene in the subject.
  • the expression of the PAH gene is increased by at least about 40% over baseline or over levels measured following administration of a control.
  • the expression of the PAH gene is increased in the liver of the subject.
  • the subject may have at least one mutated allele of the PAH gene.
  • the mutation may occur within or near the PAH gene.
  • the mutation may decrease the activity of phenylalanine hydroxylases or reduce the expression of PAH in the subject as compared to activity or expression associated with a canonical wild-type sequence.
  • the phenylalanine hydroxylase deficiency is mild hyperphenylalaninemia.
  • the phenylalanine hydroxylase deficiency is mild phenylketonuria (PKU).
  • PKU phenylketonuria
  • the phenylalanine hydroxylase deficiency is classic phenylketonuria (PKU).
  • Also provided herein is a method of modulating the expression of a PAH gene in a cell by introducing to the cell an effective amount of a compound capable of altering one or more signaling molecules associated with the regulatory sequence regions (RSRs) or portion thereof of the PAH gene.
  • a compound capable of altering one or more signaling molecules associated with the regulatory sequence regions (RSRs) or portion thereof of the PAH gene.
  • RSRs regulatory sequence regions
  • Such compound may be a small molecule, a polypeptide, an antibody, a hybridizing oligonucleotide, or a genome editing agent.
  • the compound introduced to the cell may include an inhibitor of the JAK/STAT pathway.
  • Such compound may include at least one of
  • Ruxolitinib Oclacitinib, Baricitinib, Filgotinib, Gandotinib, Lestaurtinib, PF-04965842, Upadacitinib, Cucurbitacin I, CHZ868, Fedratinib, AC430, AT9283, ati-5000l and ati- 50002, AZ 960, AZD1480, BMS-911543, CEP-33779, Cerdulatinib (PRT062070, PRT2070), Curcumol, Decemotinib (VX-509), Fedratinib (SAR302503, TG101348), FLLL32, FM-381, GLPG0634 analogue, Go6976, JANEX-l (WHI-P131), Momelotinib (CYT387), NVP-BSK805, Pacritinib (SB1518), Peficitinib (ASP015K, JNJ-54781532),
  • the compound introduced to the cell may include an inhibitor of the Tyrosine kinase/MAPK pathway.
  • Such compound may include at least one of Amuvatinib, Bosutinib, Cediranib, Ceritinib, CP-673451, Dasatinib, GZD824
  • Dimesylate Merestinib, R788 (fostamatinib disodium hexahydrate), or a derivative or an analog thereof
  • the compound introduced to the cell may include 17-AAG (Tanespimycin), Amlodipine Besylate, ATRA (all-trans retinoic acid), Chloroquine phosphate, Deoxycorticosterone, Darapladib, Echinomycin, Enzastaurin, Epinephrine, EVP-6124 (hydrochloride) (encenicline), EW-7197, FRAX597, Ibrutinib, Perphenazine, Phenformin, PND-1186, Rifampicin, Semagacestat, Thalidomide, WAY600, WYE-125132 (WYE-132), Zibotentan, Activin, Anti mullerian hormone, GDF10 (BMP3b), IGF-l,
  • the compound introduced to the cell may include one or more RNAi agents against a signaling molecule identified to regulate PAH expression.
  • the compound includes one or more small interfering RNA (siRNA) targeting one or more genes selected from the group consisting of JAK1 , JAK2, PDGFRA, PDGFRB, SRC and ABL.
  • siRNA small interfering RNA
  • the compound increases the expression of the PAH gene.
  • the expression of the PAH gene is increased by at least about 40% over baseline or over levels measured following administration of a control.
  • the cell may have at least one mutation within or near the PAH gene. The mutation(s) may decrease the activity of phenylalanine hydroxylases or reduce the expression of PAH in the cell as compared to activity or expression associated with a canonical wild-type sequence.
  • the cell may be a mammalian cell.
  • the cell is a human cell.
  • the cell is a mouse cell.
  • the cell is a hepatocyte.
  • a method of modulating the expression of a PAH gene in a cell by comprising introducing to the cell one or more compounds that alter expression of one or more of the upstream or downstream neighborhood genes or its RSRs of the insulated neighborhood comprising the PAH gene.
  • the insulated neighborhood may comprise the region on chromosome 12 at position 102,882,556 to 103,550,727 (human CRCH38/hg38 genome assembly).
  • the one or more downstream neighborhood gene includes at least one of ASCL1 and C120RF42.
  • FIG. 1 illustrates the packaging of chromosomes in a nucleus, the localized topological domains into which chromosomes are organized, insulated neighborhoods in TADs and finally an example of an arrangement of a signaling center(s) around a particular disease gene.
  • FIG. 2A and FIG. 2B illustrate a linear and 3D arrangement of the CTCF boundaries of an insulated neighborhood.
  • FIG. 3A and FIG. 3B illustrate tandem insulated neighborhoods and gene loops formed in such insulated neighborhoods.
  • FIG. 4 illustrates the concept of an insulated neighborhood contained within a larger insulated neighborhood and the signaling which may occur in each.
  • FIG. 5 illustrates the components of a signaling center; including transcriptional factors, signaling proteins, and/or chromatin regulators.
  • FIG. 6 is a plot showing the dose response curve of Momelotinib in primary human hepatocytes.
  • FIG. 7 is a bar chart showing the effect of JAK1 and JAK2 knock-down on PAH mRNA levels. Plotted values are the relative fold changes of target mRNA level with respect to the non-targeting control.
  • FIG. 8A and FIG. 8B are bar charts showing the effect of selected compound treatments on PAH expression in mouse liver.
  • FIG. 9 is a diagram of the BH4 pathway.
  • FIG. 10A-C show (A) a diagram of the conversion of phenylalanine to tyrosine using phenylalanine hydroxylase, (B) an exemplary figure of the production concentration over time, and (C) a gel showing the separation of tyrosine and phenylalanine.
  • FIG. 11 shows a schematic of the phenylalanine hydroxylase activity assay.
  • FIG. 12 shows the PAH enzyme activity in cytosolic extracts from primary human hepatocytes.
  • FIG. 13 shows the PAH enzyme activity in mouse livers after treatment with dasatinib.
  • FIG. 14 shows the effect of selected compound treatments on PAH enzyme levels in mouse liver.
  • FIG. 15 shows the fold increase in expression of PAH and GCH1 upon targeting of select gene regulatory pathways by select perturbagens in primary hepatocytes.
  • FIG. 16A shows the relative expression of PAH in mice, 6 hours post treatment with select compounds.
  • FIG. 16B shows the relative expression of GCH1 in mice, 6 hours post treatment with select compounds.
  • FIG. 17 is a western blot showing relative expression of PAH protein and Bactin protein as a loading control in mouse liver lysates from mice treated with select compounds.
  • FIG. 18A is a diagram of the dosing schedule in mice for the treatment of Dasatinib for analysis of PAH activity in mouse livers.
  • FIG. 18B shows the increase in PAH activity and mRNA in mouse livers after treatment with Dasatinib.
  • FIG. 19 is a diagram of the dosing schedule for measurement of PAH activity in mouse livers treated with XL228, PF-562271, SIS3.
  • FIG. 20 is a graph that shows PAH activity of mouse livers treated with XL228, PF-562271, SIS3 and vehicle control.
  • FIG. 21 is a graph that shows the specific activity of PAH of mouse livers treated with XL228, PF-562271 and vehicle control.
  • FIG. 22 is a western blot showing amount of PAH protein produced by cells expressing the select constructs.
  • FIG. 23 is a graph showing increased activity of PAH198 R243Q with increased lysate.
  • the present disclosure provides compositions and methods for the treatment of phenylalanine hydroxylase deficiency, such as phenylketonuria, in mammals, including in humans.
  • the disclosure provides compounds and related use for the modulation of the Phenylalanine Hydroxylase (PAH) gene.
  • PAH Phenylalanine Hydroxylase
  • phenylalanine hydroxylase deficiency or“PAH deficiency”, as used herein, refers to any condition or disorder that is manifested in an elevated phenylalanine level in the blood. Phenylalanine hydroxylase deficiency includes mild
  • “Mild hyperphenylalaninemia” or “non-PKU hyperphenylalaninemia” is defined as the presence of plasma phenylalanine levels that exceed the limits of the upper reference range (120 pmol/L or 2 mg/dL) without treatment but that are below the level found in patients with PKU.
  • “Mild PKU” or “moderate PKU” refers to conditions with plasma phenylalanine levels over 600 pmol/L (10 mg/dL) but lower than 1200 pmol/L (20 mg/dL) without treatment.
  • “Classic PKU” is defined as plasma phenylalanine levels that exceed 1200 pmol/L (20 mg/dL).“Mild PKU” or“classic PKU” are herein broadly referred to as“PKU.”
  • GSNs gene signaling networks
  • Such gene signaling networks include genomic signaling centers found within insulated neighborhoods of the genomes of biological systems.
  • Compounds modulating gene expression may act through modulating one or more gene signaling networks.
  • compositions and methods for perturbation of genomic signaling centers (GSCs) or entire gene signaling networks (GSNs) for the treatment of phenylalanine hydroxylase deficiency, such as phenylketonuria are set forth in the accompanying description below.
  • a“gene signaling network” or“GSN” comprises the set of biomolecules associated with any or all of the signaling events from a particular gene, e.g., a gene-centric network. As there are over 20,000 protein-coding genes in the human genome, there are at least this many gene signaling networks. And to the extent some genes are non-coding genes, the number increases greatly. Gene signaling networks differ from canonical signaling pathways which are mapped as standard protein cascades and feedback loops.
  • biochemical techniques and, for the most part, are linear cascades with one protein product signaling the next protein product-driven event in the cascade. While these pathways may bifurcate or have feedback loops, the focus has been almost exclusively at the protein level.
  • GSNs Gene signaling networks of the present disclosure represent a different paradigm to defining biological signaling— taking into account protein-coding and nonprotein-coding signaling molecules, genomic structure, chromosomal occupancy, chromosomal remodeling, the status of the biological system and the range of outcomes associated with the perturbation of any biological systems comprising such gene signaling networks.
  • Genomic architecture while not static, plays an important role in defining the framework of the GSNs of the present disclosure.
  • Such architecture includes the concepts of chromosomal organization and modification, topologically associated domains (TADs), insulated neighborhoods (INs), genomic signaling centers (GSCs), signaling molecules and their binding motifs or sites, and of course, the genes encoded within the genomic architecture.
  • the present disclosure by elucidating a more definitive set of connectivities of the GSNs associated with the PAH gene, provides a fine-tuned mechanism to address phenylalanine hydroxylase deficiency such as phenylketonuria.
  • Genomic system architecture includes regions of DNA, RNA transcripts, chromatin remodelers, and signaling molecules.
  • Chromosomes are the largest subunit of genome architecture that contain most of the DNA in humans. Specific chromosome structures have been observed to play important roles in gene control, as described in Hnisz et al., Cell 167, November 17, 2016, which is hereby incorporated by reference in its entirety.
  • the introns (“non-coding regions”) provide protein binding sites and other regulatory structures, while the exons encode for signaling molecules, such as transcription factors, that interact with the non-coding regions to regulate gene expression.
  • DNA sites within non-coding regions on the chromosome also interact with each other to form looped structures. These interactions form a chromosome scaffold that is preserved through development and plays an important role in gene activation and repression. Interactions rarely occur among chromosomes and are usually within the same domain of a chromosome.
  • TADs Topologically associating domains
  • topologically associating domains refers to structures that represent a modular organization of the chromatin and have boundaries that are shared by the different cell types of an organism.
  • TADs alternatively known as topological domains, are hierarchical units that are subunits of the mammalian
  • TADs are megabase-sized chromosomal regions that demarcate a microenvironment that allows genes and regulatory elements to make productive DNA-DNA contacts. TADs are defined by DNA-DNA interaction frequencies.
  • TADs represent structural chromosomal units that function as gene expression regulators.
  • TADs may contain about 7 or more protein-coding genes and have boundaries that are shared by the different cell types. See, Smallwood et al., Current Opinion in Cell Biology, 25(3):387-94, 2013, which is hereby incorporated by reference in its entirety. Some TADs contain active genes and others contain repressed genes, as the expression of genes within a single TAD is usually correlated. See, Cavalli et al., Nature Structural & Molecular Biology, 20(3):290-9, 2013, which is hereby incorporated by reference in its entirety. Sequences within a TAD find each other with high frequency and have concerted, TAD-wide histone chromatin signatures, expression levels, DNA replication timing, lamina association, and chromocenter association. See, Dixon et al., Nature,
  • TFs transcription factors
  • CCF 11 -zinc finger protein
  • TADs include cohesin-associated enhancer-promoter loops that are produced when enhancer-bound TFs bind cofactors, for example Mediator, that, in turn, bind RNA polymerase II at promoter sites.
  • NIPBL cohesin-loading factor Nipped-B-like protein
  • TADs have similar boundaries in all human cell types examined and constrain enhancer-gene interactions. See, Dixon et al., Nature, 518:331-336, 2015; Dixon et al., Nature, 485:376-380, 2012, which are hereby incorporated by reference in their entirety. This architecture of the genome helps explain why most DNA contacts occur within the TADs and enhancer-gene interactions rarely occur between chromosomes. However, TADs provide only partial insight into the molecular mechanisms that influence specific enhancer-gene interactions within TADs.
  • the methods of the present disclosure are used to alter gene expression from genes located in a TAD.
  • TAD regions are modified to alter gene expression of a non-canonical pathway as defined herein or as definable using the methods described herein.
  • insulated neighborhood refers to chromosome structure formed by the looping of two interacting sites in the chromosome sequence that may comprise CCCTC-Binding factor (CTCF) co-occupied by cohesin and affect the expression of genes in the insulated neighborhood as well as those genes in the vicinity of the insulated neighborhoods.
  • CCCTC-Binding factor CCCTC-Binding factor
  • CTCF CCCTC-Binding factor
  • Insulated neighborhood architecture is defined by at least two boundaries which come together, directly or indirectly, to form a DNA loop.
  • the boundaries of any insulated neighborhood comprise a primary upstream boundary and a primary downstream boundary. Such boundaries are the outermost boundaries of any insulated neighborhood.
  • secondary loops may be formed. Such secondary loops, when present, are defined by secondary upstream boundaries and secondary downstream boundaries, relative to the primary insulated neighborhood.
  • the loops are numbered relative to the primary upstream boundary of the primary loop, e.g., the secondary loop (first loop within the primary loop), the tertiary loop (second loop within the primary loop), the quaternary loop (the third loop within the primary loop) and so on.
  • Insulated neighborhoods may be located within topologically associated domains (TADs) and other gene loops.
  • TADs are defined by DNA-DNA interaction frequencies, and average 0.8 Mb, contain approximately 7 protein-coding genes and have boundaries that are shared by the different cell types of an organism. According to Dowen, the expression of genes within a TAD is somewhat correlated, and thus some TADs tend to have active genes and others tend to have repressed genes. Dowen et al., Cell. 2014 Oct 9; 159(2): 374-387.
  • Insulated neighborhoods may exist as contiguous entities along a chromosome or may be separated by non-insulated neighborhood sequence regions. Insulated
  • insulated neighborhoods may overlap linearly only to be defined once the DNA looping regions have been joined. While insulated neighborhoods may comprise 3-12 genes, they may contain,
  • A“minimal insulated neighborhood” is an insulated neighborhood having at least one neighborhood gene and associated regulatory sequence region or regions (RSRs) which facilitate the expression or repression of the neighborhood gene such as a promoter and/or enhancer and/or repressor region, and the like. It is contemplated that regulatory sequence regions may coincide or even overlap with an insulated neighborhood boundary. Regulatory sequence regions, as used herein, include but are not limited to regions, sections, sites or zones along a chromosome whereby interactions with signaling molecules occur in order to alter expression of a neighborhood gene.
  • a“signaling molecule” is any entity, whether protein, nucleic acid (DNA or RNA), organic small molecule, lipid, sugar or other biomolecule, which interacts directly, or indirectly, with a regulatory sequence region on a chromosome. Regulatory sequence regions (RSRs) may also be referred to as“genomic signaling centers” or“GSCs.”
  • transcription factors One category of specialized signaling molecules are transcription factors.
  • Transcription factors are those signaling molecules which alter, whether to increase or decrease, the transcription of a target gene, e.g., a neighborhood gene.
  • neighborhood genes may have any number of upstream or downstream genes along the chromosome.
  • there may be one or more, e.g., one, two, three, four or more, upstream and/or downstream neighborhood genes relative to the primary neighborhood gene.
  • a “primary neighborhood gene” is a gene which is most commonly found within a specific insulated neighborhood along a chromosome.
  • An upstream neighborhood gene of a primary neighborhood gene may be located within the same insulated neighborhood as the primary neighborhood gene.
  • a downstream neighborhood gene of a primary neighborhood gene may be located within the same insulated neighborhood as the primary neighborhood gene.
  • penetrance is the proportion of individuals carrying a particular variant of a gene (e.g., mutation, allele or generally a genotype, whether wild type or not) that also exhibits an associated trait (phenotype) of that variant gene.
  • penetrance of a disease-causing mutation measured as the proportion of individuals with the mutation who exhibit clinical symptoms. Consequently, penetrance of any gene or gene variant exists on a continuum.
  • Insulated neighborhoods are functional units that group genes under the same control mechanism, which are described in Dowen et al., Cell, 159: 374-387 (2014), which is hereby incorporated by reference in its entirety. Insulated neighborhoods provide the mechanistic background for higher-order chromosome structures, such as TADs which are shown in FIG. 1. Insulated neighborhoods are chromosome structures formed by the looping of the two interacting CTCF sites co-occupied by cohesin as shown in FIG. 1. The integrity of these structures is important for proper expression of local genes. Generally, 1 to 10 genes are clustered in each neighborhood with a median number of 3 genes within each one. The genes controlled by the same insulated neighborhood are not readily apparent from a two-dimensional view of DNA.
  • TADs can consist of a single IN, or one IN and one NIN and two NINs as shown in FIG. 2B.
  • an“insulated neighborhood boundary” refers to a boundary that delimits an insulated neighborhood on a chromosome.
  • an insulated neighborhood is defined by at least two insulated neighborhood boundaries, a primary upstream boundary and a primary downstream boundary.
  • The“primary upstream boundary” refers to the insulated neighborhood boundary located upstream of a primary neighborhood gene.
  • The“primary downstream boundary” refers to the insulated neighborhood boundary located downstream of a primary neighborhood gene.
  • a “secondary upstream boundary” is the upstream boundary of a secondary loop within a primary insulated neighborhood
  • a“secondary downstream boundary” is the downstream boundary of a secondary loop within a primary insulated neighborhood. The directionality of the secondary boundaries follows that of the primary insulated neighborhood boundaries.
  • Components of an insulated neighborhood boundary may comprise the DNA sequences at the anchor regions and associated factors (e.g., CTCF, cohesin) that facilitate the looping of the two boundaries.
  • the DNA sequences at the anchor regions may contain at least one CTCF binding site. Experiments using the ChIP-exo technique revealed a 52 bp CTCF binding motif containing four CTCF binding modules (see Fig 1, Ong and Corces, Nature reviews Genetics, 12:283-293, 2011, which is incorporated herein by reference in its entirety).
  • the DNA sequences at the insulated neighborhood boundaries may contain insulators. In some cases, insulated neighborhood boundaries may also coincide or overlap with regulatory sequence regions, such as enhancer-promoter interaction sites.
  • disrupting or altering an insulated neighborhood boundary may be accomplished by altering specific DNA sequences (e.g., CTCF binding sites) at the boundaries.
  • CTCF binding sites e.g., CTCF binding sites
  • existing CTCF binding sites at insulated neighborhood boundaries may be deleted, mutated, or inverted.
  • new CTCF binding sites may be introduced to form new insulated neighborhoods.
  • disrupting or altering an insulated neighborhood boundary may be accomplished by altering the histone modification (e.g., methylation, demethylation) at the boundaries.
  • disrupting or altering an insulated neighborhood boundary may be accomplished by altering (e.g., blocking) the binding of CTCF and/or cohesin to the boundaries.
  • RSR regulatory sequence regions
  • the term“signaling center” has been used to describe a group of cells responding to changes in the cellular environment. See, Guger et al., Developmental Biology 172: 115-125 (1995), which is incorporated by reference herein in its entirety.
  • the term“signaling center”, as used herein refers to a defined region of DNA in a living cell that interacts with a defined set of biomolecules, such as signaling proteins or signaling molecules (e.g., transcription factors) to regulate gene expression in a context- specific manner.
  • a“signaling center” refers to regions within insulated neighborhoods that include regions capable of binding context-specific combinatorial assemblies of signaling molecules/signaling proteins that participate in the regulation of the genes within that insulated neighborhood or among more than one insulated neighborhood.
  • Signaling centers have been discovered to regulate the activity of insulated neighborhoods. These regions control which genes are expressed and the level of expression in the human genome. Loss of the structural integrity of signaling centers contributes to deregulation of gene expression and potentially causing disease.
  • Signaling centers include enhancers bound by a highly context-specific combinatorial assemblies of transcription factors. These factors are recruited to the site through cellular signaling. Signaling centers include multiple genes that interact to form a three-dimensional transcription factor hub macrocomplex. Signaling centers are generally associated with one to four genes in a loop organized by biological function.
  • compositions of each signaling center has a unique composition including the assemblies of transcription factors, the transcription apparatus, and chromatin regulators.
  • Signaling centers are highly context specific, permitting drugs to control response by targeting signaling pathways.
  • a series of consensus binding sites, or binding motifs for binding sites, for signaling molecules has been identified by the present inventors. These consensus sequences reflect binding sites along a chromosome, gene, or polynucleotide for signaling molecules or for complexes which include one or more signaling molecules.
  • binding sites are associated with more than one signaling molecule or complex of molecules.
  • the term“enhancer”, as used herein, refers to regulatory DNA sequences that, when bound by transcription factors, enhance the transcription of an associated gene.
  • Enhancers are gene regulatory elements that control cell type specific gene expression programs in humans. See, Buecker and Wysocka, Trends in genetics: TIG 28, 276-284, 2012; Heinz et ak, Nature reviews Molecular Cell Biology, 16: 144-154, 2015; Levine et ak, Cell, 157:13-25, 2014; Ong and Corces, Nature reviews Genetics, 12:283- 293, 2011; Ren and Yue, Cold Spring Harbor symposia on quantitative biology, 80: 17-26, 2015, which are hereby incorporated by reference in their entireties. Enhancers are segments of DNA that are generally a few hundred base pairs in length and are occupied by multiple transcription factors that recruit co-activators and RNA polymerase II to target genes.
  • Enhancer RNA molecules transcribed from these regions of DNA also“trap” transcription factors capable of binding DNA and RNA.
  • a region with more than one enhancer is a“super-enhancer.”
  • the term“super enhancers”, as used herein, refers to clusters of transcriptional enhancers that drive expression of genes that define cell identity.
  • Insulated neighborhoods provide a microenvironment for specific enhancer-gene interactions that are vital for both normal gene activation and repression.
  • Transcriptional enhancers control over 20,000 protein-coding genes to maintain cell type-specific gene expression programs in all human cells. Tens of thousands of enhancers are estimated to be active in any given human cell type. See, ENCODE Project Consortium et ak, Nature, 489, 57- 74, 2012; Roadmap Epigenomics et ak, Nature, 518, 317-330, 2015, which are hereby incorporated by reference in their entirety.
  • Enhancers and their associated factors can regulate expression of genes located upstream or downstream by looping to the promoters of these genes.
  • DNA sequences in enhancers and in promoter-proximal regions bind to a variety of transcription factors expressed in a single cell. Diverse factors bound at these two sites interact with large cofactor complexes and interact with one another to produce enhancer-gene specificity. See, Zabidi et al., Nature, 518:556-559, 2015, which is hereby incorporated by reference in its entirety.
  • enhancer regions may be targeted to alter or elucidate gene signaling networks (GSNs).
  • GSNs gene signaling networks
  • Insulator refers to regulatory elements that block the ability of an enhancer to activate a gene when located between them and contribute to specific enhancer-gene interactions. See, Chung et al., Cell 74:505-514, 1993; Geyer and Corces, Genes & Development 6:1865-1873, 1992; Kellum and Schedl, Cell 64:941-950, 1991; Udvardy et al., Journal of molecular biology 185:341-358, 1985, which are hereby incorporated by reference in their entirety. Insulators are bound by the transcription factor CTCF but not all CTCF sites function as insulators. See, Bell et al., Cell 98: 387-396,
  • Enhancer-bound proteins are constrained such that they tend to interact only with genes within these CTCF-CTCF loops.
  • the subset of CTCF sites that form these loop anchors thus function to insulate enhancers and genes within the loop from enhancers and genes outside the loop, as shown in FIG. 2B.
  • insulator regions may be targeted to alter or elucidate gene signaling networks (GSNs).
  • CTCF interactions link sites on the same chromosome forming loops, which are generally less than 1 Mb in length. Transcription occurs both within and outside the loops, but the nature of this transcription differs between the two regions. Studies show that enhancer-associated transcription is more prominent within the loops. Thus, the insulator state is enriched specifically at the CTCF loop anchors. CTCF loops thus either enclose gene poor regions, with a tendency for genes to be centered within the loops or leave out gene dense regions outside the CTCF loops. CTCF loops exhibit reduced exon density relative to their flanking regions. Gene ontology analysis reveals that genes located within CTCF loops are enriched for response to stimuli and for extracellular, plasma membrane and vesicle cellular localizations.
  • genes present within the flanking regions just outside the loops exhibit an expression pattern similar to housekeeping genes i.e. these genes are on average more highly expressed than the loop-enclosed genes, are less cell-line specific in their expression pattern, and have less variation in their expression levels across cell lines. See Oti et al., BMC Genomics, 17:252, 2016, which is incorporated by reference in its entirety.
  • Anchor regions are binding sites for CTCF that influence conformation of an insulated neighborhood. Deletion of anchor sites may result in activation of genes that are usually transcriptionally silent, thereby resulting in a disease phenotype. In fact, somatic mutations are common in loop anchor sites of oncogene-associated insulated
  • CTCF DNA-binding motif of the loop anchor region has been observed to be the most altered human transcription-factor binding sequence of cancer cells. See, Hnisz et al., Cell 167, November 17, 2016, which is incorporated by reference in its entirety.
  • Anchor regions have been observed to be largely maintained during cell development, and are especially conserved in the germline of humans and primates. In fact, the DNA sequence of anchor regions are more conserved in CTCF anchor regions than at CTCF binding sites that are not part of an insulated neighborhood. Therefore, cohesin may be used as a target for ChIA-RET to identify locations of both.
  • Cohesin also becomes associated with CTCF-bound regions of the genome, and some of these cohesin-associated CTCF sites facilitate gene activation while others may function as insulators. See, Dixon et al.. Nature. 485(73981:376-80, 2012: Parelho et al.. Cell, 132(31:422-33, 2008: Phillips -Cremins and Corces, Molecular Cell, 50(41:461-74, 2013); Seitan et al.. Genome Research, 23(121:2066-77, 2013; Wendt et al.. Nature, 45l(7l80):796-80l, 2008), which are hereby incorporated by reference in their entireties.
  • Cohesin and CTCF are associated with large loop substructures within TAPs, and cohesin and Mediator are associated with smaller loop structures that form within CTCF-bounded regions. See, de Wit et al.. Nature. 501 (74661:227- 31. 2013: Cremins et ah. Cell. 153(61:1281-95. 2013: Sofueva et al.. EMBO, 32(24):3l 19-29, 2013, which are hereby incorporated by reference in their entireties.
  • cohesin and CTCF associated loops and anchor sites/regions may be targeted to alter or elucidate gene signaling networks (GSNs).
  • GSNs gene signaling networks
  • SNPs Most disease associated SNPs are located in the proximity of signaling centers. For example, 94.2% of SNPs occur in non-coding regions, which include signaling centers. In some embodiments, SNPs are altered in order to study and/or alter the signaling from one or more GSN.
  • Signaling molecules include any protein that functions in cellular signaling pathways, whether canonical or the gene signaling network pathways defined herein or capable of being defined using the methods described herein. Transcription factors are a subset of signaling molecules. Certain combinations of signaling and master transcription factors associate to an enhancer region to influence expression of a gene. Master regulator factors direct transcription factors in specific tissues. For example, in blood, GATA transcription factors are master regulators that direct TCF7L2 of the Wnt cellular signaling pathway. In the liver, HNG4 is a master regulator to direct SMAD in lineage tissues and patterns.
  • Transcriptional regulation allows controlling how often a given gene is transcribed. Transcription factors alter the rate at which transcripts are produced by making conditions for transcription initiation more or less favorable. A transcription factor selectively alters a signaling pathway which in turn affects the genes expressed by a signaling center. Signaling centers are components of transcriptional regulators. In some embodiments, signaling molecules may be used, targeted in order to elucidate or alter the signaling of gene signaling networks of the present disclosure.
  • Table 18 of U.S. 62/501,795, which is hereby incorporated by reference in its entirety, provides a list of signaling molecules including those which act as transcription factors (TF) and/or chromatin remodeling factors (CR) that function in various cellular signaling pathways.
  • the methods described herein may be used to inhibit or activate the expression of one or more signaling molecules associated with the regulatory sequence region of the primary neighborhood gene encoded within an insulated neighborhood. The methods may thus alter the signaling signature of one or more primary neighborhood genes which are differentially expressed upon treatment with the therapeutic agent compared to an untreated control.
  • Transcription factors generally regulate gene expression by binding to enhancers and recruiting coactivators and RNA polymerase II to target genes. See Whyte et al., Cell, 153(2): 307-319, 2013, which is incorporated by reference in its entirety. Transcription factors bind“enhancers” to stimulate cell-specific transcriptional program by binding regulatory elements distributed throughout the genome.
  • transcription factors there are about 1800 known transcription factors in the human genome. There are epitopes on the DNA of the chromosomes that provide binding sites for proteins or nucleic acid molecules such as ribosomal RNA complexes. Master regulators direct a combination of transcription factors through cell signaling above and DNA below. These characteristics allow for determination of the location of the next signaling center. In some embodiments, transcription factors may be used or targeted, to alter or elucidate the gene signaling networks of the present disclosure.
  • Master transcription factors recruit additional signaling proteins, such as other transcription factors, to enhancers to form signaling centers.
  • An atlas of candidate master TFs for 233 human cell types and tissues is described in D’Alessio et al., Stem Cell Reports 5, 763-775 (2015), which is hereby incorporated by reference in its entirety.
  • master transcription factors may be used or targeted, to alter or elucidate the gene signaling networks of the present invention.
  • Signaling transcription factors are transcription factors, such as homeoproteins, that travel between cells as they contain protein domains that allow them to do the so.
  • Homeoproteins such as Engrailed, Hoxa5, Hoxb4, Hoxc8, Emxl, Emx2, Otx2 and Pax6 are able to act as signaling transcription factors.
  • the homeoprotein Engrailed possesses internalization and secretion signals that are believed to be present in other homeoproteins as well. This property allows homeoproteins to act as signaling molecules in addition to being transcription factors.
  • Homeoproteins lack characterized extracellular functions leading to the perception that their paracrine targets are intracellular. The ability of homeoproteins to regulate transcription and, in some cases, translation is most likely to affect paracrine action. See Prochiantz and Joliot, Nature Reviews Molecular Cell Biology, 2003.
  • signaling transcription factors may be used or targeted, to alter or elucidate the gene signaling networks of the present disclosure.
  • Chromatin remodeling is regulated by over a thousand proteins that are associated with histone modification. See, Ji et al., PNAS, 112(12):3841-3846(2015), which is hereby incorporated by reference in its entirety.
  • Chromatin regulators are specific sets of proteins associated with genomic regions marked with modified histones. For example, histones may be modified at certain lysine residues: H3K20me3, H3K27ac, H3K4me3, H3K79me2, H3K36me3, H3K9me2, and H3K9me3. Certain histone modifications mark regions of the genome that are available for binding by signaling molecules.
  • ChIP-MS may be performed to identify chromatin regulator proteins associated with specific histone modification. ChIP-seq with antibodies specific to certain modified histones may also be used to identify regions of the genome that are bound by signaling molecules. In some embodiments, chromatin modifying enzymes or proteins may be used or targeted, to alter or elucidate the gene signaling networks of the present disclosure.
  • RSRs active regulatory sequence regions
  • eRNAs enhancer-associated RNAs
  • RNAs derived from regulatory sequence regions of the PAH gene may be used or targeted to alter or elucidate the gene signaling networks of the present disclosure.
  • RNAs derived from regulatory sequence regions may be an enhancer-associated RNA (eRNA).
  • RNAs derived from regulatory sequence regions may be a promoter-associated RNA, including but not limited to, a promoter upstream transcript (PROMPT), a promoter-associated long RNA (PALR), and a promoter-associated small RNA (PASR).
  • RNAs derived from regulatory sequence regions may include but are not limited to transcription start sites (TSS)-associated RNAs (TSSa-RNAs), transcription initiation RNAs (tiRNAs), and terminator-associated small RNAs (TASRs).
  • TSS transcription start sites
  • tiRNAs transcription initiation RNAs
  • TASRs terminator-associated small RNAs
  • RNAs derived from regulatory sequence regions may be long non-coding RNAs (lncRNAs) (i.e., >200 nucleotides). In some embodiments, RNAs derived from regulatory sequence regions may be intermediate non-coding RNAs. (i.e., about 50 to 200 nucleotides). In some embodiments, RNAs derived from regulatory sequence regions may be short non-coding RNAs (i.e., about 20 to 50 nucleotides).
  • eRNAs that may be modulated by methods and compounds described herein may be characterized by one or more of the following features: (1) transcribed from regions with high levels of monomethylation on lysine 4 of histone 3 (H3K4mel) and low levels of trimethylation on lysine 4 of histone 3
  • H3K4me3 (2) transcribed from genomic regions with high levels of acetylation on lysine 27 of histone 3 (H3K27ac); (3) transcribed from genomic regions with low levels of trimethylation on lysine 36 of histone 3 (H3K36me3); (4) transcribed from genomic regions enriched for RNA polymerase II (Pol II); (5) transcribed from genomic regions enriched for transcriptional co-regulators, such as the p300 co-activator; (6) transcribed from genomic regions with low density of CpG island; (7) their transcription is initiated from Pol II-binding sites and elongated bidirectionally; (8) evolutionarily conserved DNA sequences encoding eRNAs; (9) short half-life; (10) reduced levels of splicing and polyadenylation, (11) dynamically regulated upon signaling; (12) positively correlated to levels of nearby mRNA expression; (13) extremely high tissue specificity; (14) preferentially nuclear and chromatin-bound; and/or (15
  • Non-limiting examples of eRNAs that may be modulated by methods and compounds described herein include those described in Djebali et al., Nature. 2012 Sep 6;489(74l4) (for example, Supplementary data file for Figure 5a) and Andersson et al., Nature. 2014 Mar 27;507(7493):455-46l (for example, Supplementary Tables S3, S12, S13, S15, and 16), which are herein incorporated by reference in their entirety.
  • promoter-associated RNAs that may be modulated by methods or compounds described herein may be characterized by one or more of the following features: (1) transcribed from regions with high levels of H3K4mel and low to medium levels of H3K4me3; (2) transcribed from genomic regions with high levels of H3K27ac; (3) transcribed from genomic regions with no or low levels of H3K36me3; (4) transcribed from genomic regions enriched for RNA polymerase II (Pol II); (5) transcribed from genomic regions with high density of CpG island; (6) their transcription is initiated from Pol II-binding sites and elongated in the opposite direction from the sense strand (that is, mRNAs) or bidirectionally; (7) short half-life; (8) reduced levels of splicing and polyadenylation; (9) preferentially nuclear and chromatin-bound; and/or (10) degraded by the exosome.
  • RNA polymerase II RNA polymerase II
  • Methods and compositions described herein may be used to modulate RNAs derived from regulatory sequence regions to alter or elucidate the gene signaling networks of the present disclosure.
  • methods and compounds described herein may be used to inhibit the production and/or function of an RNA derived from regulatory sequence regions.
  • a hybridizing oligonucleotide such as an siRNA or an antisense oligonucleotide may be used to inhibit the activity of the RNA of interest via RNA interference (RNAi), or RNase H-mediated cleavage, or physically block binding of various signaling molecules to the RNA.
  • RNAi RNA interference
  • Exemplary hybridizing oligonucleotide may include those described in U.S.
  • the hybridizing oligonucleotide may be provided as a chemically modified or unmodified RNA, DNA, locked nucleic acids (LNA), or a combination of RNA and DNA, a nucleic acid vector encoding the hybridizing oligonucleotide, or a vims carrying such vector.
  • genome editing tools such as CRISPR/Cas9 may be used to delete specific DNA elements in the regulatory sequence regions that control the transcription of the RNA or degrade the RNA itself.
  • genome editing tools such as a catalytically inactive CRISPR/Cas9 may be used to bind to specific elements in the regulatory sequence regions and block the transcription of the RNA of interest.
  • bromodomain and extra terminal domain (BET) inhibitors e.g., JQ1, 1-BET may be used to reduce RNA transcription through inhibition of histone acetylation by BET protein Brd4.
  • methods and compounds described herein may be used to increase the production and/or function of an RNA derived from regulatory sequence regions.
  • an exogenous synthetic RNA that mimic the RNA of interest may be introduced into the cell.
  • the synthetic RNA may be provided as an RNA, a nucleic acid vector encoding the RNA, or a vims carrying such vector.
  • genome editing tools such as CRISPR/Cas9 may be used to tether an exogenous synthetic RNA to specific sites in the regulatory sequence regions.
  • Such RNA may be fused to the guide RNA of the CRISPR/Cas9 complex.
  • modulation of RNAs derived from regulatory sequence regions increases the expression of the PAH gene. In some embodiments, modulation of RNAs derived from regulatory sequence regions reduces the expression of the PAH gene.
  • RNAs modulated by compounds described herein include RNAs derived from regulatory sequence regions of the PAH gene in a liver cell (e.g., hepatocytes).
  • GSNs gene signaling networks
  • GSCs genomic signaling centers
  • INs insulated neighborhoods
  • Potential stimuli may include exogenous biomolecules such as small molecules, antibodies, proteins, peptides, lipids, fats, nucleic acids, and the like or environmental stimuli such as radiation, pH, temperature, ionic strength, sound, light and the like.
  • the present disclosure serves, not only as a discovery tool for the elucidation of better defined gene signaling networks (GSNs) and consequently a better understanding of biological systems.
  • GSNs gene signaling networks
  • the present disclosure allows the ability to properly define gene signaling for PAH at the gene level in a manner which allows the prediction, a priori, of potential treatment outcomes, the identification of novel compounds or targets which may have never been implicated in the treatment of a PAH-related disease or condition, reduction or removal of one or more treatment liabilities associated with new or known drugs such as toxicity, poor half-life, poor bioavailability, lack of or loss of efficacy or pharmacokinetic or pharmacodynamic risks.
  • a method of treating a disease may include modifying a signaling center that is involved in a gene associated with that disease. Such genes may not presently be associated with the disease except as is elucidated using the methods described herein.
  • a perturbation stimulus may be a small molecule, a known drug, a biological, a vaccine, an herbal preparation, a hybridizing oligonucleotide (e.g., siRNA and antisense oligonucleotide), a gene or cell therapy product, or other treatment product.
  • a hybridizing oligonucleotide e.g., siRNA and antisense oligonucleotide
  • methods of the present disclosure include applying a perturbation stimulus to perturb GSNs, genomic signaling centers, and/or insulated neighborhoods associated with the PAH gene.
  • Perturbation stimuli that cause changes in PAH gene expression may inform the connectivities of the associated GSNs and provide potential targets and/or treatments for phenylalanine hydroxylase deficiency such as phenylketonuria.
  • a stimulus is administered that targets a downstream product of a gene of a gene signaling network.
  • the stimulus disrupts a gene signaling network that affects downstream expression of at least one downstream target.
  • the gene is PAH.
  • Perturbation of a single or multiple gene signaling network (GSN) associated with a single insulated neighborhood or across multiple insulated neighborhoods can affect the transcription of a single gene or a multiple set of genes by altering the boundaries of the insulated neighborhood due to loss of anchor sites comprising cohesins.
  • Perturbation stimuli may result in the modification of the RNA expression and/or the sequences in the primary transcript within the mRNA, i.e. the exons or the RNA sequences between the exons that are removed by splicing, i.e. the introns.
  • Such changes may consequently alter the members of the set of signaling molecules within the gene signaling network of a gene, thereby defining a variant of the gene signaling network.
  • Perturbation of a single or multiple gene signaling networks associated with a single insulated neighborhood or across multiple insulated neighborhoods can affect the translation of a single gene or a multiple set of genes that are part of the genomic signaling center, as well as those downstream to the genomic signaling center. Perturbation might result in the inhibition of the translated protein.
  • Perturbation stimuli may cause interactions with signaling molecules to occur in order to alter expression of the nearest primary neighborhood gene that may be located upstream or downstream of the primary neighborhood gene.
  • Neighborhood genes may have any number of upstream or downstream genes along the chromosome. Within any insulated neighborhood, there may be one or more, e.g., one, two, three, four or more, upstream and/or downstream neighborhood genes relative to the primary neighborhood gene.
  • A“primary neighborhood gene” is a gene which is most commonly found within a specific insulated neighborhood along a chromosome.
  • An upstream neighborhood gene of a primary neighborhood gene may be located within the same insulated neighborhood as the primary neighborhood gene.
  • a downstream neighborhood gene of a primary neighborhood gene may be located within the same insulated neighborhood as the primary neighborhood gene.
  • GSNs gene signaling networks
  • GSN gene signaling networks of the disclosure are defined at the gene level and characterized based on any number of stimuli or perturbation to the cell, tissue, organ or organ system expressing that gene.
  • the nature of a GSN is both structurally (e.g., the gene) and situationally (e.g., the function, e.g., expression profile) defined.
  • two different gene signaling networks may share members, they are still unique in that the nature of the perturbation can distinguish them.
  • the value of gene signaling networks in the elucidation of the function of biological systems in support of therapeutic research and development.
  • methods of the present disclosure involve altering the Janus kinases (JAK)/signal transducers and activators of transcription (STAT) pathway.
  • JAK/STAT pathway is the major mediator for a wide array of cytokines and growth factors.
  • Cytokines are regulatory molecules that coordinate immune responses.
  • JAKs are a family of intracellular, nonreceptor tyrosine kinases that are typically associated with cell surface receptors such as cytokine receptors. Mammals are known to have 4 JAKs: JAK1, JAK2, JAK3, and Tyrosine kinase 2 (TYK2).
  • STATs are latent transcription factors that reside in the cytoplasm until activated.
  • STAT1 There are seven mammalian STATs: STAT1, STAT2, STAT3, STAT4, STAT5 (STAT5A and STAT5B), and STAT6.
  • STAT5A and STAT5B STAT5A and STAT5B
  • STAT6 STAT6A and STAT6B
  • Activated STATs translocate to the nucleus where they complex with other nuclear proteins and bind to specific sequences to regulate the expression of target genes.
  • the JAK/STAT pathway provides a direct mechanism to translate an extracellular signal into a transcriptional response.
  • JAK/STAT pathway Target genes regulated by JAK/STAT pathway are involved in immunity, proliferation, differentiation, apoptosis and oncogenesis. Activation of JAKs may also activate the phosphatidylinositol 3 -kinase (PI3K) and mitogen- activated protein kinase (MAPK) pathways.
  • PI3K phosphatidylinositol 3 -kinase
  • MAPK mitogen- activated protein kinase
  • methods of the present disclosure involve altering the mitogen-activated protein kinase (MAPK) signaling pathway.
  • the MAPK pathway involves a chain of signaling molecules (e.g., Ras, Raf, MEK, and ERK) in the cell that communicates a signal from a receptor at the cell membrane to the nucleus. This pathway can be activated by receptor- linked tyrosine kinases such as epidermal growth factor receptor (EGFR), Trk A/B, Fibroblast growth factor receptor (FGFR) and PDGFR.
  • EGFR epidermal growth factor receptor
  • Trk A/B Trk A/B
  • FGFR Fibroblast growth factor receptor
  • PDGFR receptor- linked tyrosine kinases
  • the MAPK signaling pathway is essential in regulating numerous cellular processes including cell stress response, cell differentiation, cell division, cell proliferation, inflammation, metabolism, motility and apoptosis.
  • MAPK interacts with major pathway targets: ERK1/2, ERK5, JNK, and p38 kinase. MAPK regulates the activities of several transcription factors including C-myc, CREB and C-Fos. MAPK also interacts with other pathways such as the PI3K networks, NF-kB and JAK/STAT pathways.
  • methods of the present disclosure involve altering the Platelet-derived Growth Factor Receptor (PDGFR)-mediated signal pathway.
  • PDGFRs are cell surface tyrosine kinase receptors for members of the platelet-derived growth factor (PDGF) family. There are two isoforms of PDGFRs, PDGFRa and PDGFRp. The two receptor isoforms dimerize upon binding the PDGF dimer, leading to the activation of the kinase. PDGFRs mediate a number of signaling pathways that are important for regulating cell proliferation, cellular differentiation, cell growth and development.
  • PDGFR-mediated signaling pathway has been correlated with reduced expression of PDGF, angl/2, and VEGF mRNA. Since PDGF is a known stimulus for PI3-K activation, inhibiting PDGFR may lead to decreased activation of the PI3-K signaling cascade.
  • PDGFs and PDGFRs in physiology and medicine is reviewed in Andrae et ak,
  • canonical pathways which may also be altered according to the present disclosure include, but are not limited to the 2-arachidonoylglycerol biosynthesis pathway, 2-oxocarboxylic acid metabolism pathway, 5HT1 type receptor mediated signaling pathway, 5HT2 type receptor mediated signaling pathway, 5HT3 type receptor mediated signaling pathway, 5HT4 type receptor mediated signaling pathway, 5 -hydroxy tryptamine biosynthesis pathway, 5 -hydroxy tryptamine degradation pathway, abacavir transport and metabolism pathway, ABC transporters pathway, ABC-family proteins mediated transport pathway, ACE inhibitor pathway, acetate utilization pathway, acetylcholine synthesis pathway, activation of camp-dependent PKA pathway, activin beta signaling pathway, adenine and hypoxanthine salvage pathway, adherens junction pathway, adipocytokine signaling pathway, adipogenesis pathway, adrenaline and noradrenaline biosynthesis pathway, adrenergic signaling in cardiomyocytes pathway, advanced glycation end
  • RNA polymerase-II initiation complex pathway ATM pathway, ATP synthesis pathway, axon guidance pathway, axon guidance mediated by netrin pathway, axon guidance mediated by semaphorins pathway, axon guidance mediated by slit/robo pathway, B cell activation pathway, B cell receptor (BCR) pathway, B cell receptor signaling pathway, bacterial invasion of epithelial cells pathway, basal transcription factors pathway, base excision repair pathway, B-cell development pathway, B-cell receptor pathway, B-cell receptor complex pathway, benzo pathway, betal adrenergic receptor signaling pathway, beta2 adrenergic receptor signaling pathway, beta3 adrenergic receptor signaling pathway, beta- alanine metabolism pathway, bile acid and bile salt metabolism pathway, bile secretion pathway, binding and uptake of ligands by scavenger receptors pathway, biogenic amine synthesis pathway, biogenic amine synthesis pathway, biogenic amine synthesis pathway, biogenic
  • glycerophospholipid biosynthetic pathway glycerophospholipid metabolism pathway, glycine metabolism pathway, glycogen metabolism pathway, glycolysis/gluconeogenesis pathway, glycosaminoglycan biosynthesis -heparan sulfate / heparin pathway,
  • glycosaminoglycan biosynthesis-keratan sulfate pathway glycosaminoglycan degradation pathway, glycosaminoglycan metabolism pathway, glycosphingolipid biosynthesis - ganglio series pathway, glycosphingolipid biosynthesis-globo series pathway,
  • glycosphingolipid biosynthesis - lacto and neolacto series pathway glycosphingolipid biosynthesis - lacto and neolacto series pathway, glyoxylate and dicarboxylate metabolism pathway, gonadotropin-releasing hormone receptor pathway, GP1B-IX-V activation signaling pathway, GPCR pathway, GPCR downstream signaling pathway, GPCR ligand binding pathway, GPVI-mediated activation cascade pathway, granulocyte adhesion and diapedesis pathway, granzyme pathway, growth hormone signaling pathway, GSK 3 signaling pathway, hedgehog signaling pathway, hematopoiesis from pluripotent stem cells pathway, hematopoietic cell lineage pathway, hematopoietic stem cell differentiation pathway, heme biosynthesis pathway, hepatitis B pathway, hepatitis C pathway, heterotrimeric G-protein signaling-Gi alpha and Gs alpha mediated pathway, heterotrimeric g-protein signaling -rod outer segment phototransduction pathway
  • tetracosanoyl-coA pathway peroxisomal lipid metabolism pathway, pertussis pathway, phagosome pathway, phase 1 -functionalization of compounds pathway, phase I biotransformations pathway, phase II conjugation pathway, phenylacetate degradation pathway, phenylalanine biosynthesis pathway, phenylalanine metabolism pathway, phenylethylamine degradation pathway, phenylpropionate degradation pathway, phosphatidylinositol signaling system pathway, phospholipase D signaling pathway, phototransduction pathway, PI3 kinase pathway, PI3K signaling in B -lymphocytes pathway, PI3K-AKT signaling pathway, PIP3 activates AKT signaling pathway, plasminogen activating cascade pathway, platelet activation pathway, platelet adhesion to exposed collagen pathway, platelet aggregation pathway, platelet homeostasis pathway, polyol pathway, porphyrin and chlorophyll metabolism pathway, PPAR signaling pathway,
  • PRPP biosynthesis pathway PTEN pathway, purine metabolism pathway, pyridoxal phosphate salvage pathway, pyridoxal-5 -phosphate biosynthesis pathway, pyrimidine metabolism pathway, pyruvate metabolism pathway, racl pathway, rank signaling in osteoclast pathway, rankl/rank pathway, rapl signaling pathway, ras signaling pathway, Ras-RAF-MEK-ERK pathway, receptor activator of nuclear factor kappa-b ligand
  • RNKL RANKL pathway
  • actin cytoskeleton pathway regulation of actin cytoskeleton pathway
  • apoptosis pathway regulation of autophagy pathway, regulation of DNA replication pathway, regulation of lipolysis in adipocytes pathway, regulation of microtubule cytoskeleton pathway, regulation of toll-like receptor signaling pathway, remodeling of adherens junctions pathway, renin secretion pathway, renin-angiotensin system pathway, respiratory electron transport pathway, retinol metabolism pathway, retrograde
  • endocannabinoid signaling pathway Rho family GTPase pathway, Rhoa pathway, ribosome biogenesis in eukaryotes pathway, RIG-I-like receptor signaling pathway, RNA degradation pathway, RNA polymerase I pathway, RNA polymerase II transcription pathway, RNA transport pathway, RNAi pathway, s-adenosylmethionine biosynthesis pathway, salivary secretion pathway, salvage pyrimidine deoxyribonucleotides pathway, salvage pyrimidine ribonucleotides pathway, SCW signaling pathway, selenium metabolism and selenoproteins pathway, selenium micronutrient network pathway, selenocompound metabolism pathway, semaphorin interactions pathway, serine and threonine metabolism pathway, serine glycine biosynthesis pathway, serotonergic synapse pathway, serotonin htrl group and fos pathway, serotonin receptor 2 and ELK-SRF/gata4 signaling pathway, seroton
  • TGF- beta transforming growth factor beta
  • translation factors pathway transmission across electrical synapses pathway
  • transport of glucose and other sugars pathway transport of glycerol from adipocytes to the liver by aquaporins pathway
  • transport of vitamins pathway trans- sulfuration pathway, trans-sulfuration and one carbon metabolism pathway
  • analog refers to a compound that is structurally related to the reference compound and shares a common functional activity with the reference compound.
  • biological refers to a medical product made from a variety of natural sources such as micro-organism, plant, animal, or human cells.
  • boundary refers to a point, limit, or range indicating where a feature, element, or property ends or begins.
  • the term“derivative”, as used herein, refers to a compound that differs in structure from the reference compound, but retains the essential properties of the reference molecule.
  • the term“downstream neighborhood gene”, as used herein, refers to a gene downstream of primary neighborhood gene that may be located within the same insulated neighborhood as the primary neighborhood gene.
  • the term“gene”, as used herein, refers to a unit or segment of the genomic architecture of an organism, e.g., a chromosome. Genes may be coding or non-coding. Genes may be encoded as contiguous or non-contiguous polynucleotides. Genes may be DNA or RNA.
  • genomic system architecture refers to the organization of an individual’s genome and includes chromosomes, topologically associating domains (TADs), and insulated neighborhoods.
  • master transcription factor refers to signaling molecules which alter, whether to increase or decrease, the transcription of a target gene, e.g., a neighborhood gene and establish cell-type specific enhancers. Master transcription factors recruit additional signaling proteins, such as other transcription factors to enhancers to form signaling centers.
  • modulate refers to an alteration (e.g., increase or decrease) in the expression of the target gene and/or activity of the gene product.
  • penetrance refers to the proportion of individuals carrying a particular variant of a gene (e.g., mutation, allele or generally a genotype, whether wild type or not) that also exhibits an associated trait (phenotype) of that variant gene and in some situations is measured as the proportion of individuals with the mutation who exhibit clinical symptoms thus existing on a continuum.
  • polypeptide refers to a polymer of amino acid residues (natural or unnatural) linked together most often by peptide bonds.
  • primary neighborhood gene refers to a gene which is most commonly found within a specific insulated neighborhood along a chromosome.
  • the term“primary downstream boundary”, as used herein, refers to the insulated neighborhood boundary located downstream of a primary neighborhood gene.
  • the term“primary upstream boundary”, as used herein, refers to the insulated neighborhood boundary located upstream of a primary neighborhood gene.
  • promoter refers to a DNA sequence that defines where transcription of a gene by RNA polymerase begins and defines the direction of
  • regulatory sequence regions include but are not limited to regions, sections or zones along a chromosome whereby interactions with signaling molecules occur in order to alter expression of a neighborhood gene.
  • repressor refers to any protein that binds to DNA and therefore regulates the expression of genes by decreasing the rate of transcription.
  • second downstream boundary refers to the downstream boundary of a secondary loop within a primary insulated neighborhood.
  • second upstream boundary refers to the upstream boundary of a secondary loop within a primary insulated neighborhood.
  • signaling molecule refers to any entity, whether protein, nucleic acid (DNA or RNA), organic small molecule, lipid, sugar or other biomolecule, which interacts directly, or indirectly, with a regulatory sequence region on a chromosome.
  • signaling transcription factor refers to signaling molecules which alter, whether to increase or decrease, the transcription of a target gene, e.g., a neighborhood gene and also act as cell-cell signaling molecules.
  • small molecule refers to a low molecular weight drug, i.e. ⁇ 5000 Daltons organic compound that may help regulate a biological process.
  • therapeutic agent refers to a substance that has the ability to cure a disease or ameliorate the symptoms of the disease.
  • therapeutic or treatment outcome refers to any result or effect (whether positive, negative or null) which arises as a consequence of the perturbation of a GSC or GSN.
  • therapeutic outcomes include, but are not limited to, improvement or amelioration of the unwanted or negative conditions associated with a disease or disorder, lessening of side effects or symptoms, cure of a disease or disorder, or any improvement associated with the perturbation of a GSC or GSN.
  • therapeutic or treatment liability refers to a feature or characteristic associated with a treatment or treatment regime which is unwanted, harmful or which mitigates the therapies positive outcomes.
  • treatment liabilities include for example toxicity, poor half-life, poor bioavailability, lack of or loss of efficacy or pharmacokinetic or pharmacodynamic risks.
  • upstream neighborhood gene refers to a gene upstream of a primary neighborhood gene that may be located within the same insulated neighborhood as the primary neighborhood gene.
  • methods of the present disclosure involve modulating the expression of PAH gene.
  • the PAH gene encodes phenylalanine hydroxylase which is a metabolic enzyme that converts of phenylalanine to tyrosine.
  • PAH may also be referred to as Phenylalaninase, Phenylalanine 4-Monooxygenase, Phe-4-Monooxygenase, EC
  • PAH gene has a cytogenetic location of l2q23.2 and is located on Chromosome 12 at position 102,836,885-102,958,410 on the reverse strand.
  • PAH has a NCBI gene ID of 5053, Uniprot ID of P00439 and Ensembl Gene ID of ENSG00000171759.
  • PAH is primarily expressed in the liver, kidney, nervous system, gall bladder, and blood. More than 900 pathogenic variants have been described in PAH. Information on the pathogenic variants, associated phenotypes, gene structure, and enzyme structure can be found on the Phenylalanine Hydroxylase Locus Knowledgebase (PAHdb).
  • PAH deficiency refers to any condition or disorder that is manifested in an elevated phenylalanine level in the blood. PAH deficiency can be detected in newborn screening using methods well known in the art (e.g., Guthrie microbial inhibition assay) based on the presence of hyperphenylalaninemia on a blood spot obtained from a heel prick.
  • phenylalanine hydroxylase results in a spectrum of disorders including mild hyperphenylalaninemia, mild phenylketonuria, and classic phenylketonuria.
  • the term“mild hyperphenylalaninemia” or“non-PKU hyperphenylalaninemia” is defined as the presence of plasma phenylalanine levels that exceed the limits of the upper reference range (120 pmol/L or 2 mg/dL) without treatment but that are below the level found in patients with PKU.
  • “Mild PKU” or“moderate PKU” refers to conditions with plasma phenylalanine levels over 600 pmol/L (10 mg/dL) but lower than 1200 pmol/L (20 mg/dL) without treatment.
  • Plasma phenylalanine levels that exceed 1200 pmol/L (20 mg/dL) are classified as“classic PKU”, which is the most severe form of PKU.“Mild PKU” or“classic PKU” are herein broadly referred to as“PKU.”
  • methods and compositions of the present disclosure increase the levels of partially functional PAH protein to breakdown phenylalanine and prevent or treat PKU.
  • methods of the present disclosure involve altering the composition and/or the structure of the insulated neighborhood containing the PAH gene.
  • the present inventors have identified the insulated neighborhood containing the PAH gene in primary human hepatocytes.
  • the insulated neighborhood containing the PAH gene is approximately 668 kb in length and contains 14 signaling centers.
  • the insulated neighborhood contains PAH and 2 other genes, namely ASCL1 and C120RF42, both of which are located downstream of PAH.
  • the chromatin marks and/or chromatin-associated proteins include H3K27Ac, BRD4, p300, and SMC1.
  • the transcription factors include FOXA2, HNF4A, ONECUT1, ONECUT2, YY1, and HNF1A.
  • the signaling proteins include TCF7L2, ESR1, HIF1A, FOS, NR3C1, JUN, NF-kB, RBPJ, RXR, STAT3, NR1I1, SMAD2/3, SMAD1, SMAD4, STAT1, TEAD1, and TP53. Any components of these signaling centers and/or signaling molecules, or any regions within or near the insulated neighborhood, may be targeted or altered to change the composition and/or structure of the insulated neighborhood, thereby modulating the expression of PAH.
  • GCH1 is the rate limiting enzyme in BH4 synthesis. This patheway is subject to allosteric feed-forward activation by L- phenylalanine and feedback inhibition by BH4. GCH1 is not liver specific, rather it is expressed in multiple cell types. An advantage of targeting GCH1 is a concomitant increase in PAH activity by increasing the BH4 levels. This will lead to increase in intracellular BH4.
  • BH4 pathway Modulating the BH4 pathway will result in greater treatment opportunities to rare PKU patients with mutations in BH4 pathway such as 6-pyruvoyl- tetrahydropterin synthase deficiency (PTPS), GTP cyclohydrolase I (GTPCH), and Dihydropteridine reductase deficiency (DHPR).
  • PTPS 6-pyruvoyl- tetrahydropterin synthase deficiency
  • GTPCH GTP cyclohydrolase I
  • DHPR Dihydropteridine reductase deficiency
  • upregulation of GCH1 could be beneficial for a wide range of cardiovascular disorders (Cunnington, Heart 96
  • Methods to increase intracellular BH4 include administering Kuvan, a synthetic BH4 which stabilizes and increases the activity of some PAH mutants; enzyme substitution therapy using pegValiase which converts phenylalanine to a non-toxic metabolite; and gene therapy.
  • Kuvan a synthetic BH4 which stabilizes and increases the activity of some PAH mutants
  • enzyme substitution therapy using pegValiase which converts phenylalanine to a non-toxic metabolite
  • gene therapy Gene therapy.
  • compositions and methods for modulating the expression of the PAH gene Any one of the compositions and methods described herein may be used to treat or prevent a PAH-related disorder such as phenylketonuria. In some embodiments, a combination of the compositions and methods described herein may be used to treat a PAH-related disorder.
  • the terms“subject” and“patient” are used interchangeably herein and refer to an animal to whom treatment with the compositions according to the present disclosure is provided.
  • the subject is a mammal.
  • the subject is a human being.
  • subjects may have been diagnosed with or have symptoms for phenylalanine hydroxylase deficiency, e.g., mild hyperphenylalaninemia, mild PKU, or classic PKU.
  • subjects may be susceptible to or at risk for phenylalanine hydroxylase deficiency, e.g., mild hyperphenylalaninemia, mild PKU, or classic PKU.
  • subjects may carry one or more mutations within or near the PAH gene. In some embodiment, subjects may carry one functional allele and one mutated allele of the PAH gene. In some embodiment, subjects may carry two mutated alleles of the PAH gene.
  • subjects may have dysregulated expression of the PAH gene.
  • subjects may have a deficiency of the phenylalanine hydroxylase enzyme.
  • subjects may have a partially functional phenylalanine hydroxylase.
  • compositions and methods may be used to increase the expression of the PAH gene in a cell or a subject.
  • Changes in gene expression may be assessed at the RNA level or protein level by various techniques known in the art and described herein, such as RNA-seq, qRT-PCR, Western Blot, or enzyme-linked immunosorbent assay (ELISA). Changes in gene expression may be determined by comparing the level of target gene expression in the treated cell or subject to the level of expression in an untreated or control cell or subject. In some embodiments, the
  • compositions and methods cause an increase in the expression of the PAH gene by at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 100%, at least about 125%, at least about 150%, at least about 175%, at least about 200%, at least about 250%, at least about 300%, at least about 400%, at least about 500%, from about 25% to about 50%, from about 40% to about 60%, from about 50% to about 70%, from about 60% to about 80%, from about 80% to about 100%, from about 100% to about 125%, from about 100 to about 150%, from about 150% to about 200%, from about 200% to about 300%, from about 300% to about 400%, from about 400% to about 500% as compared to baseline or to administration of a control, or more than 500% as compared to baseline or to administration of a control.
  • compositions and methods cause a fold change in the expression of the PAH gene by about 2 fold, about 3 fold, about 4 fold, about 5 fold, about 6 fold, about 7 fold, about 8 fold, about 9 fold, about 10 fold, about 12 fold, about 15 fold, about 18 fold, about 20 fold, about 25 fold, or more than 30 fold as compared to baseline or to administration of a control.
  • the increase in the expression of the PAH gene induced by compositions and methods of the present disclosure may be sufficient to prevent or alleviate one or more signs or symptoms of phenylalanine hydroxylase deficiency in a subject.
  • administering to a subject with phenylalanine hydroxylase deficiency compositions and methods of the present disclosure may result in reduction of blood phenylalanine levels in the subject to below 120 pmol/L, below 240 pmol/L, below 360 pmol/L, below 480 pmol/L, below 600 pmol/L, below 720 pmol/L, from about 120 pmol/L to about 360 pmol/L, from about 240 pmol/L to about 480 pmol/L, from about 360 mihoI/L to about 600 mpioI/L, from about 480 pmol/L to about 720 pmol/L, from about 600 pmol/L to about 840 pmol/
  • the compounds may be used in combination with other drugs, such as KUVAN® (sapropterin dihydrochloride), to treat phenylalanine hydroxylase deficiency (e.g., PKU).
  • KUVAN® sipropterin dihydrochloride
  • compounds used to modulate the expression of the PAH gene may include small molecules.
  • small molecule refers a low molecular weight drug, i.e. ⁇ 5000 Daltons organic compound that may help regulate a biological process.
  • small molecule compounds described herein are applied to a genomic system to interfere with components (e.g., transcription factor, signaling proteins) of the gene signaling networks associated with the PAH gene, thereby modulating the expression of PAH.
  • small molecule compounds described herein are applied to a genomic system to alter the boundaries of an insulated neighborhood and/or disrupt signaling centers associated with the PAH gene, thereby modulating the expression of PAH.
  • a small molecule screen may be performed to identify small molecules that act through signaling centers of an insulated neighborhood to alter gene signaling networks which may modulate expression of the PAH gene. For example, known signaling agonists/antagonists may be administered. Credible hits are identified and validated by the small molecules that are known to work through a signaling center and modulate expression of the target gene.
  • small molecule compounds capable of modulating expression of the PAH gene include compounds that modulate the JAK/STAT signaling pathway.
  • Such compounds may be JAK inhibitors, including but not limited to Ruxolitinib, Oclacitinib, Baricitinib, Filgotinib, Gandotinib, Lestaurtinib, PF-04965842, Upadacitinib, Cucurbitacin I, CHZ868, Fedratinib, AC430, AT9283, ati-5000l and ati-50002, AZ 960, AZD1480, BMS-911543, CEP-33779, Cerdulatinib (PRT062070, PRT2070), Curcumol, Decemotinib (VX-509), Fedratinib (SAR302503, TG101348), FLLL32, FM-381,
  • GLPG0634 analogue Go6976, JANEX-l (WHI-P131), Momelotinib (CYT387), NVP- BSK805, Pacritinib (SB1518), Peficitinib (ASP015K, JNJ-54781532), PF-06651600, PF- 06700841, R256 (AZD0449), Solcitinib (GSK2586184 or GLPG0778), S-Ruxolitinib (INCB018424), TG101209, Tofacitinib (CP-690550), WHI-P154, WP1066, XL019, ZM 39923 HC1, and those described herein.
  • compounds capable of modulating the expression of the PAH gene may include Pacritinib (SB 1518), or a derivative or an analog thereof.
  • Pacritinib also known as SB 1518, is a potent and selective inhibitor JAK2 and
  • compounds capable of modulating the expression of the PAH gene may include Momelotinib, or a derivative or an analog thereof.
  • Momelotinib also known as CYT387, is an ATP-competitive inhibitor of JAK1/JAK2 with IC50 of 11 hM/18 nM and approximately lO-fold selectivity versus JAK3.
  • compounds capable of modulating the expression of the PAH gene may include Tofacitinib, or a derivative or an analog thereof.
  • Tofacitinib also known as CP-690550, is an inhibitor of JAK1 and JAK3. It is currently approved for the treatment of rheumatoid arthritis (RA) in the United States and other countries.
  • compounds capable of modulating the expression of the PAH gene may include Ruxolitinib, or a derivative or an analog thereof.
  • Ruxolitinib is an oral bioavailable JAK inhibitor with selectivity for JAK1 and JAK2. It is used in the treatment of intermediate or high risk myelofibrosis.
  • compounds capable of modulating the expression of the PAH gene may include Cerdulatinib, or a derivative or an analog thereof.
  • Cerdulatinib is an oral, dual-JAK and Syk inhibitor with IC50 of 12 nM/6 nM/8 nM/0.5 nM and 32 nM for JAK1/JAK2/JAK3/TYK2 and Syk, respectively.
  • compounds capable of modulating the expression of the PAH gene may include JANEX-l (WHI-P131), or a derivative or an analog thereof.
  • JANEX-l is a cell-permeable JAK3 inhibitor and does not inhibit JAK1, JAK2, or Zap/S yk or Src tyrosine kinases.
  • compounds capable of modulating the expression of the PAH gene may include Oclacitinib, or a derivative or an analog thereof.
  • Oclacitinib is an orally bioavailable, broad spectrum JAK inhibitor with IC50S ranging from 10 to 99 nM. It is used as a veterinary medication in the control of pruritus (itching) associated with allergic dermatitis and atopic dermatitis in dogs.
  • small molecule compounds capable of modulating expression of the PAH gene include compounds that modulate the Tyrosine Kinase/MAPK signaling pathway.
  • Such compounds may be Tyrosine kinase inhibitors, including but not limited to Amuvatinib, Bosutinib, Cediranib, Ceritinib, CP-673451, Dasatinib, GZD824 Dimesylate, Merestinib, R788 (fostamatinib disodium hexahydrate), and those described herein.
  • compounds capable of modulating the expression of the PAH gene may include Amuvatinib, or a derivative or an analog thereof.
  • Amuvatinib also known as MP-470, is a potent and multi-targeted inhibitor of c-Kit, PDGFRa and FLT3 with IC50 of 10 nM, 40 nM and 81 nM, respectively. Amuvatinib also suppresses c-Met and c-Ret.
  • compounds capable of modulating the expression of the PAH gene may include Bosutinib, or a derivative or an analog thereof.
  • Bosutinib also known as SKI-606, is a novel, dual Src/Abl inhibitor with IC50 of 1.2 nM and 1 nM, respectively.
  • compounds capable of modulating the expression of the PAH gene may include Cediranib, or a derivative or an analog thereof.
  • Cediranib is a potent inhibitor of vascular endothelial growth factor (VEGF) receptor tyrosine kinases.
  • VEGF vascular endothelial growth factor
  • compounds capable of modulating the expression of the PAH gene may include Ceritinib, or a derivative or an analog thereof.
  • Ceritinib also known as LDK378, is potent inhibitor against ALK with IC50 of 0.2 nM, exhibiting 40- and 35-fold selectivity against IGF-1R and InsR, respectively.
  • compounds capable of modulating the expression of the PAH gene may include CP-673451, or a derivative or an analog thereof.
  • CP-673451 is a selective inhibitor of PDGFRa/b with IC50 of 10 nM/l nM, exhibiting >450-fold selectivity over other angiogenic receptors.
  • CP-673451 also has antiangiogenic and antitumor activity.
  • compounds capable of modulating the expression of the PAH gene may include Dasatinib, or a derivative or an analog thereof.
  • Dasatinib is a novel, potent and multi-targeted inhibitor that targets Abl, Src, and c-Kit, with IC50 of ⁇ 1 nM,
  • compounds capable of modulating the expression of the PAH gene may include GZD824 Dimesylate, or a derivative or an analog thereof.
  • GZD824 is a novel orally bioavailable Bcr-Abl inhibitor for Bcr-Abl (wildtype) and Bcr-Abl (T315I) with IC50 of 0.34 nM and 0.68 nM, respectively.
  • compounds capable of modulating the expression of the PAH gene may include Merestinib, or a derivative or an analog thereof.
  • Merestinib also known as LY2801653, is a type-II ATP competitive, slow-off inhibitor of MET tyrosine kinase with a Kd of 2 nM, a pharmacodynamic residence time (Koff) of 0.00132 min-l and half life (tl/2) of 525 min.
  • compounds capable of modulating the expression of the PAH gene may include R788 (fostamatinib disodium hexahydrate), or a derivative or an analog thereof.
  • R788 sodium salt hydrate (fostamatinib), a prodrug of the active metabolite R406, is a potent Syk inhibitor with IC50 of 41 nM.
  • small molecule compounds capable of modulating expression of the PAH gene include, but are not limited to, 17-AAG (Tanespimycin), Amlodipine Besylate, ATRA (all-trans retinoic acid), Chloroquine phosphate,
  • compounds capable of modulating the expression of the PAH gene may include 17-AAG (Tanespimycin), or a derivative or an analog thereof.
  • 17- AAG also known as NSC 330507 or CP 127374, is a potent HSP90 inhibitor with half-maximal inhibitory concentration (IC50) of 5 nM, a lOO-fold higher binding affinity for HSP90 derived from tumor cells than HSP90 from normal cells.
  • compounds capable of modulating the expression of the PAH gene may include Amlodipine Besylate, or a derivative or an analog thereof.
  • Amlodipine also known as Norvasc, is a long-acting calcium channel blocker with an IC50 of 1.9 nM.
  • compounds capable of modulating the expression of the PAH gene may include ATRA (all-trans retinoic acid), or a derivative or an analog thereof.
  • ATRA all-trans retinoic acid
  • ATRA is an active metabolite of vitamin A under the family retinoid, which exert potent effects on cell growth, differentiation and apoptosis through their cognate nuclear receptors.
  • compounds capable of modulating the expression of the PAH gene may include Chloroquine phosphate, or a derivative or an analog thereof.
  • Chloroquine phosphate is an aminoquinoline antimaiarial compound.
  • compounds capable of modulating the expression of the PAH gene may include Deoxycorticosterone, or a derivative or an analog thereof.
  • Deoxycorticosterone acetate is a steroid hormone used for intramuscular injection for replacement therapy of the adrenocortical steroid. 1 I b-hydroxylation of
  • deoxycorticosterone leads to corticosterone.
  • compounds capable of modulating the expression of the PAH gene may include Darapladib, or a derivative or an analog thereof.
  • Darapladib is a selective and orally active inhibitor of lipoprotein-associated phospholipase A2 (Lp-PLA2) with IC50 of 270 pM.
  • Lp-PLA2 may link lipid metabolism with inflammation, leading to the increased stability of atherosclerotic plaques present in the major arteries.
  • Darapladib is being studied as a possible add-on treatment for atherosclerosis.
  • compounds capable of modulating the expression of the PAH gene may include Echinomycin, or a derivative or an analog thereof.
  • Hypoxia- inducible factor- 1 (HIF-l) is a transcription factor that controls genes involved in glycolysis, angiogenesis, migration, and invasion.
  • Echinomycin is a cell-permeable inhibitor of HIF-l -mediated gene transcription. It acts by intercalating into DNA in a sequence- specific manner, blocking the binding of either HIF-l a or HIF-l b to the hypoxia- responsive element.
  • compounds capable of modulating the expression of the PAH gene may include Enzastaurin, or a derivative or an analog thereof.
  • Enzastaurin also known as LY317615, is a potent RKC ⁇ b selective inhibitor with IC50 of 6 nM, exhibiting 6- to 20-fold selectivity against PKCa, PKCy and PKCs.
  • compounds capable of modulating the expression of the PAH gene may include Epinephrine, or a derivative or an analog thereof.
  • Epinephrine HC1 is a hormone and a neurotransmitter ⁇
  • compounds capable of modulating the expression of the PAH gene may include EVP-6124 (hydrochloride) (encenicline), or a derivative or an analog thereof.
  • EVP-6124 hydrochloride also known as encenicline, is a novel partial agonist of a7 neuronal nicotinic acetylcholine receptors (nAChRs).
  • nAChRs neuronal nicotinic acetylcholine receptors
  • EVP- 6124 shows selectivity for a7 nAChRs and does not activate or inhibit heteromeric a4b2 nAChRs.
  • compounds capable of modulating the expression of the PAH gene may include EW-7197.
  • EW-7197 is a highly potent, selective, and orally bioavailable TGF-b receptor ALK4/ALK5 inhibitor with IC50 of 13 nM and 11 nM, respectively.
  • compounds capable of modulating the expression of the PAH gene may include FRAX597, or a derivative or an analog thereof.
  • FRAX597 is a potent, ATP-competitive inhibitor of group I PAKs with IC50 of 8 nM, 13 nM, and 19 nM for PAK1, PAK2, and PAK3, respectively.
  • compounds capable of modulating the expression of the PAH gene may include Ibrutinib, or a derivative or an analog thereof.
  • Ibrutinib is a Tec family kinase inhibitor that irreversibly inhibits Bruton tyrosine kinase (BTK) and IL-2 Inducible T-cell Kinase (ITK).
  • BTK and ITK are enzymes responsible for the
  • BCR B-cell receptor
  • TCR T cell receptor
  • compounds capable of modulating the expression of the PAH gene may include Perphenazine, or a derivative or an analog thereof.
  • Perphenazine is a phenothiazine derivative that binds with high affinity to a wide variety of receptors, including dopamine, serotonin (5-HT), histamine, and a-adrenergic receptors.
  • Perphenazine is used as an antipsychotic for the symptomatic management of psychotic disorders (e.g., schizophrenia).
  • compounds capable of modulating the expression of the PAH gene may include Phenformin, or a derivative or an analog thereof.
  • Phenformin hydrochloride is a hydrochloride salt of phenformin that is an anti-diabetic drug from the biguanide class.
  • compounds capable of modulating the expression of the PAH gene may include PND-1186, or a derivative or an analog thereof.
  • PND-1186, VS- 4718, is a reversible and selective focal adhesion kinase (FAK) inhibitor with IC50 of 1.5 nM.
  • FAK focal adhesion kinase
  • compounds capable of modulating the expression of the PAH gene may include Rifampicin, or a derivative or an analog thereof.
  • rifampicin Access of rifampicin to the nuclear receptor PXR requires its import into the cell viaorganic anion transporters (OATs) in the OAT polypeptide (OATP) family. By acting as a transporter substrate, rifampicin inhibits OATPs with K1/IC50 values ranging from 0.58-18 mM.
  • compounds capable of modulating the expression of the PAH gene may include Semagacestat, or a derivative or an analog thereof.
  • Semagacestat also known as LY-450139, is a g-secretase inhibitor for Ab42, Ab40 and Ab38 with IC50 of 10.9 nM, 12.1 nM and 12.0 nM, respectively. Semagacestat also inhibits Notch signaling with IC50 of 14.1 nM.
  • compounds capable of modulating the expression of the PAH gene may include Thalidomide, or a derivative or an analog thereof.
  • Thalidomide was introduced as a sedative drug, immunomodulatory agent and also is investigated for treating symptoms of many cancers.
  • Thalidomide inhibits an E3 ubiquitin ligase, which is a CRBN-DDB l-Cul4A complex.
  • compounds capable of modulating the expression of the PAH gene may include WAY600, or a derivative or an analog thereof.
  • WAY600 is a potent, ATP-competitive and selective inhibitor of mTOR with IC50 of 9 nM.
  • compounds capable of modulating the expression of the PAH gene may include WYE-125132 (WYE-132), or a derivative or an analog thereof.
  • WYE-125132 also known as WYE-132, is a highly potent, ATP-competitive mTOR inhibitor with IC50 of 0.19 nM. It is highly selective for mTOR versus PI3Ks or PI3K- related kinases hSMGl and ATR.
  • compounds capable of modulating the expression of the PAH gene may include Zibotentan, or a derivative or an analog thereof.
  • Zibotentan also known as ZD4054, is an orally administered, potent and specific endothelin A receptor (ETA) -receptor antagonist with IC50 of 21 nM.
  • ETA potent and specific endothelin A receptor
  • compounds for altering expression of the PAH gene comprise a polypeptide.
  • polypeptide refers to a polymer of amino acid residues (natural or unnatural) linked together most often by peptide bonds.
  • the term, as used herein, refers to proteins, polypeptides, and peptides of any size, structure, or function.
  • the polypeptide encoded is smaller than about 50 amino acids and the polypeptide is then termed a peptide. If the polypeptide is a peptide, it will be at least about 2, 3, 4, or at least 5 amino acid residues long.
  • polypeptides include gene products, naturally occurring polypeptides, synthetic polypeptides, homologs, orthologs, paralogs, fragments and other equivalents, variants, and analogs of the foregoing.
  • a polypeptide may be a single molecule or may be a multi-molecular complex such as a dimer, trimer or tetramer. They may also comprise single chain or multichain polypeptides and may be associated or linked.
  • the term polypeptide may also apply to amino acid polymers in which one or more amino acid residues are an artificial chemical analog of a corresponding naturally occurring amino acid.
  • polypeptide compounds capable of modulating expression of the PAH gene include, but are not limited to, Activin, Anti mullerian hormone, GDF10 (BMP3b), IGF-l, Nodal, PDGF, TNF-a, Wnt3a, or derivatives or analogs thereof. Any one of these compounds or a combination thereof may be administered to a subject to treat phenylalanine hydroxylase deficiency.
  • compounds capable of modulating the expression of the PAH gene may include Activin, or a derivative or an analog thereof.
  • Activins are homodimers or heterodimers of the different b subunit isoforms, part of the transforming growth factor-beta (TGF-B) family.
  • Mature Activin A has two 116 amino acids residues bA subunits (bA-bA).
  • Activin displays an extensive variety of biological activities, including mesoderm induction, neural cell differentiation, bone remodeling, hematopoiesis, and reproductive physiology. Activins takes part in the production and regulation of hormones such as FSH, LH, GnRH and ACTH.
  • Cells that are identified to express Activin A include fibroblasts, endothelial cells, hepatocytes, vascular smooth muscle cells, macrophages, keratinocytes, osteoclasts, bone marrow monocytes, prostatic epithelium, neurons, chondrocytes, osteoblasts, Leydig cells, Sertoli cells, and ovarian granulosa cells.
  • compounds capable of modulating the expression of the PAH gene may include anti Mullerian hormone, or a derivative or an analog thereof.
  • Anti Mullerian hormone is a member of the TGF-B gene family which mediates male sexual differentiation.
  • Anti Mullerian hormone causes the regression of Mullerian ducts which would otherwise differentiate into the uterus and fallopian tubes.
  • Some mutations in the anti-Mullerian hormone result in persistent Mullerian duct syndrome.
  • compounds capable of modulating the expression of the PAH gene may include GDF10 (BMP3b), or a derivative or an analog thereof.
  • GDF10 also known as BMP3b, is a member of the BMP family and the TGF-B superfamily.
  • GDF10 is expressed in femur, brain, lung, skeletal, muscle, pancreas and testis, and has a role in head formation and possibly multiple roles in skeletal morphogenesis.
  • GDF10 mRNA is found in the cochlea and lung of fetuses, and in testis, retina, pineal gland, and other neural tissues of adults. These proteins are characterized by a polybasic proteolytic processing site which is cleaved to produce a mature protein containing 7 conserved cysteine residues.
  • compounds capable of modulating the expression of the PAH gene may include IGF-l, or a derivative or an analog thereof.
  • Insulin-like growth factor I also known as Somatamedin C is a hormone similar in molecular structure to insulin.
  • Human IGF-I has two isoforms (IGF-IA and IGF-IB) which is differentially expressed by various tissues. Mature human IGF-I respectively shares 94% and 96% aa sequence identity with mouse and rat IGF-I.
  • IGF-I and IGF-II can signal through the IGF-I receptor (IGF1R), but IGF-II can alone bind the IGF-II receptor (IGFIIR/Mannose-6-phosphate receptor). IGF-I plays an important role in childhood growth and continues to have anabolic effects in adults.
  • compounds capable of modulating the expression of the PAH gene may include Nodal, or a derivative or an analog thereof.
  • Nodal is a 13 kDa member of the TGF-B superfamily of molecules. In human, it is synthesized as a 347 amino acid preproprecursor that contains a 26 amino acid signal sequence, a 211 amino acid prodomain, and a 110 amino acid mature region. Consistent with its TGF-B superfamily membership, it exists as a disulfide-linked homodimer and would be expected to demonstrate a cysteine-knot motif. Mature human Nodal is 99%, 98%, 96% and 98% amino acid identical to mature canine, rat, bovine and mouse Nodal, respectively. Nodal signals through two receptor complexes, both of which contain members of the TGF-beta family of Ser/Thr kinase receptors.
  • compounds capable of modulating the expression of the PAH gene may include PDGF, or a derivative or an analog thereof the Platelet-derived growth factor (PDGF) is a disulfide-linked dimer consisting of two peptides -chain A and chain B.
  • PDGF has three subforms: PDGF-AA, PDGF-BB, PDGF-AB. It is involved in a number of biological processes, including hyperplasia, embryonic neuron development, chemotaxis, and respiratory tubule epithelial cell development. The function of PDGF is mediated by two receptors (PDGFRa and PDGFR ).
  • compounds capable of modulating the expression of the PAH gene may include TNF-a, or a derivative or an analog thereof.
  • TNF-a the prototypical member of the TNF protein superfamily, is a homotrim eric type-II membrane protein.
  • Membrane bound TNF-a is cleaved by the metalloprotease TACE/ADAM17 to generate a soluble homotrimer. Both membrane and soluble forms of TNF-a are biologically active.
  • TNF-a is produced by a variety of immune cells including T cells, B cells, NK cells and macrophages.
  • TNF-a Activation of kinase pathways (including JNK, ERK (p44/42), p38 MAPK and NF-kB) promotes the survival of cells, while TNF-a mediated activation of caspase-8 leads to programmed cell death.
  • TNF-a plays a key regulatory role in inflammation and host defense against bacterial infection, notably Mycobacterium tuberculosis. The role of TNF- a in autoimmunity is underscored by blocking TNF-a action to treat rheumatoid arthritis and Crohn’ s disease.
  • compounds capable of modulating the expression of the PAH gene may include Wnt3a, or a derivative or an analog thereof.
  • the WNT gene family consists of structurally related genes which encode secreted signaling proteins. These proteins have been implicated in oncogenesis and in several developmental processes, including regulation of cell fate and patterning during embryogenesis. This gene is a member of the WNT gene family. It encodes a protein which shows 96% amino acid identity to mouse Wnt3a protein, and 84% to human WNT3 protein, another WNT gene product. This gene is clustered with WNT14 gene, another family member, in chromosome lq42 region.
  • compounds for altering expression of the PAH gene comprise an antibody.
  • antibodies described herein comprise antibodies, antibody fragments, their variants or derivatives that are specifically immunoreaetive with at least one component of the gene signaling networks associated with the PAH gene.
  • antibody is used in the broadest sense and specifically covers various embodiments including, but not limited to monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g. bispecific antibodies formed from at least two intact antibodies), and antibody fragments such as diabodies so long as they exhibit a desired biological activity.
  • Antibodies are primarily amino-acid based molecules but may also comprise one or more modifications such as with sugar moieties.
  • Antibody fragments comprise a portion of an intact antibody, preferably comprising an antigen binding region thereof.
  • antibody fragments include Fab, Fab', F(ab') 2 , and Fv fragments; diabodies; linear antibodies; single-chain antibody molecules; and multispecific antibodies formed from antibody fragments.
  • Papain digestion of antibodies produces two identical antigen-binding fragments, called “Fab” fragments, each with a single antigen-binding site. Also produced is a residual "Fc” fragment, whose name reflects its ability to crystallize readily. Pepsin treatment yields an F(ab')2 fragment that has two antigen-binding sites and is still capable of cross-linking antigen.
  • Antibodies of the present disclosure may comprise one or more of these fragments.
  • an "antibody” may comprise a heavy and light variable domain as well as an Fc region.
  • Native antibodies are usually heterotetrameric glycoproteins of about 150,000 Daltons, composed of two identical light (L) chains and two identical heavy (H) chains. Each light chain is linked to a heavy chain by one covalent disulfide bond, while the number of disulfide linkages varies among the heavy chains of different immunoglobulin isotypes. Each heavy and light chain also has regularly spaced intrachain disulfide bridges. Each heavy chain has at one end a variable domain (VH) followed by a number of constant domains.
  • VH variable domain
  • Each light chain has a variable domain at one end (VL) and a constant domain at its other end; the constant domain of the light chain is aligned with the first constant domain of the heavy chain, and the light chain variable domain is aligned with the variable domain of the heavy chain.
  • variable domain refers to specific antibody domains that differ extensively in sequence among antibodies and are used in the binding and specificity of each particular antibody for its particular antigen.
  • Fv refers to antibody fragments which contain a complete antigen-recognition and antigen-binding site. This region consists of a dimer of one heavy chain and one light chain variable domain in tight, non-covalent association.
  • Antibody "light chains" from any vertebrate species can be assigned to one of two clearly distinct types, called kappa and lambda based on amino acid sequences of their constant domains. Depending on the amino acid sequence of the constant domain of their heavy chains, antibodies can be assigned to different classes. There are five major classes of intact antibodies: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgGl, IgG2, IgG3, IgG4, IgA, and IgA2.
  • Single-chain Fv or “scFv” as used herein, refers to a fusion protein of VH and VL antibody domains, wherein these domains are linked together into a single polypeptide chain.
  • the Fv polypeptide linker enables the scFv to form the desired structure for antigen binding.
  • diabodies refers to small antibody fragments with two antigen-binding sites, which fragments comprise a heavy chain variable domain V H connected to a light chain variable domain V L in the same polypeptide chain.
  • linker that is too short to allow pairing between the two domains on the same chain, the domains are forced to pair with the complementary domains of another chain and create two antigen-binding sites.
  • Diabodies are described more fully in, for example, EP 404,097; WO 93/11161; and Hollinger et al., Proc. Natl. Acad. Sci. USA, 90:6444-6448 (1993), the contents of each of which are incorporated herein by reference in their entirety.
  • Antibodies of the present disclosure may be polyclonal or monoclonal or recombinant, produced by methods known in the art or as described in this application.
  • the term "monoclonal antibody” as used herein refers to an antibody obtained from a population of substantially homogeneous cells (or clones), i.e., the individual antibodies comprising the population are identical and/or bind the same epitope, except for possible variants that may arise during production of the monoclonal antibody, such variants generally being present in minor amounts.
  • each monoclonal antibody is directed against a single determinant on the antigen.
  • the modifier "monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies herein include "chimeric" antibodies (immunoglobulins) in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies.
  • Humanized forms of non-human (e.g., murine) antibodies are chimeric antibodies that contain minimal sequence derived from non-human immunoglobulin.
  • humanized antibodies are human immunoglobulins (recipient antibody) in which residues from the hypervariable region from an antibody of the recipient are replaced by residues from the hypervariable region from an antibody of a non-human species (donor antibody) such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity, and capacity.
  • hypervariable region when used herein in reference to antibodies refers to regions within the antigen binding domain of an antibody comprising the amino acid residues that are responsible for antigen binding.
  • the amino acids present within the hypervariable regions determine the structure of the complementarity determining region (CDR).
  • CDR complementarity determining region
  • the“CDR” refers to the region of an antibody that comprises a structure that is complimentary to its target antigen or epitope.
  • compositions of the present disclosure may be antibody mimetics.
  • antibody mimetic refers to any molecule which mimics the function or effect of an antibody and which binds specifically and with high affinity to their molecular targets.
  • antibody mimics include nanobodies and the like.
  • antibody mimetics may be those known in the art including, but are not limited to affibody molecules, affilins, affitins, anticalins, avimers, DARPins, Fynomers and Kunitz and domain peptides. In other embodiments, antibody mimetics may include one or more non-peptide region.
  • antibody variant refers to a biomolecule resembling an antibody in structure and/or function comprising some differences in their amino acid sequence, composition or structure as compared to a native antibody.
  • Antibodies of the present disclosure may be characterized by their target molecule(s), by the antigens used to generate them, by their function (whether as agonists or antagonists) and/or by the cell niche in which they function. [00165] Measures of antibody function may be made relative to a standard under normal physiologic conditions, in vitro or in vivo. Measurements may also be made relative to the presence or absence of the antibodies.
  • Such methods of measuring include standard measurement in tissue or fluids such as serum or blood such as Western blot, enzyme- linked immunosorbent assay (ELISA), activity assays, reporter assays, luciferase assays, polymerase chain reaction (PCR) arrays, gene arrays, Real Time reverse transcriptase (RT) PCR and the like.
  • Antibodies exert their effects via binding (reversibly or irreversibly) to one or more target sites. While not wishing to be bound by theory, target sites which represent a binding site for an antibody, are most often formed by proteins or protein domains or regions. However, target sites may also include biomolecules such as sugars, lipids, nucleic acid molecules or any other form of binding epitope.
  • antibodies of the present disclosure may function as ligand mimetics or nontraditional payload carriers, acting to deliver or ferry bound or conjugated drug payloads to specific target sites.
  • neomorphic change is a change or alteration that is new or different. Such changes include extracellular, intracellular and cross cellular signaling.
  • compounds or agents act to alter or control proteolytic events. Such events may be intracellular or extracellular.
  • Antibodies as well as antigens used to generate them, are primarily amino acid- based molecules. These molecules may be "peptides,” “polypeptides,” or “proteins.”
  • the term“peptide” refers to an amino-acid based molecule having from 2 to 50 or more amino acids. Special designators apply to the smaller peptides with “dipeptide” referring to a two amino acid molecule and“tripeptide” referring to a three amino acid molecule. Amino acid based molecules having more than 50 contiguous amino acids are considered polypeptides or proteins.
  • amino acid and “amino acids” refer to all naturally occurring L- alpha-amino acids as well as non-naturally occurring amino acids.
  • Amino acids are identified by either the one-letter or three-letter designations as follows: aspartic acid (Asp:D), isoleucine (He : I) , threonine (Thr:T), leucine (Leu:L), serine (Ser:S), tyrosine (Tyr:Y), glutamic acid (Glu:E), phenylalanine (Phe:F), proline (Pro:P), histidine (His:H), glycine (Gly:G), lysine (Lys:K), alanine (Ala:A), arginine (Arg:R), cysteine (Cys:C), tryptophan (Trp:W), valine (Val:V), glutamine (Gln:Q) methionine (Met:M), asparagines
  • oligonucleotides including those which function via a hybridization mechanism, whether single of double stranded such as antisense molecules, RNAi constructs (including siRNA, saRNA, microRNA, etc.), aptamers and ribozymes may be used to alter or as perturbation stimuli of the gene signaling networks associated with the PAH gene.
  • hybridizing oligonucleotides may be used to knock down signaling molecules involved in the pathways regulating PAH expression such that PAH expression is enhanced in the absence of the signaling molecule.
  • a component of the pathway e.g., a receptor, a protein kinase, a transcription factor
  • a RNAi agent e.g., siRNA
  • the pathway targeted with a hybridizing oligonucleotide (e.g., siRNA) of the present disclosure to enhance PAH expression is the JAK/STAT pathway.
  • the hybridizing oligonucleotide e.g., siRNA
  • the hybridizing oligonucleotide is used to knock down JAK1.
  • the hybridizing oligonucleotide e.g., siRNA is used to knock down JAK2.
  • the pathway targeted with a hybridizing oligonucleotide (e.g., siRNA) of the present disclosure to enhance PAH expression is the PDGFR pathway.
  • the hybridizing oligonucleotide e.g., siRNA
  • the hybridizing oligonucleotide is used to knock down PDGFRA.
  • the hybridizing oligonucleotide e.g., siRNA
  • the pathway targeted with a hybridizing oligonucleotide (e.g., siRNA) of the present disclosure to enhance PAH expression is the Src/Abl pathway.
  • the hybridizing oligonucleotide e.g., siRNA
  • the hybridizing oligonucleotide is used to knock-down SRC.
  • the hybridizing oligonucleotide e.g., siRNA
  • a hybridizing oligonucleotide as described above may be used together with another hybridizing oligonucleotide to target more than one components in the same pathway, or more than one components from different pathways, to enhance PAH expression.
  • Such combination therapies may achieve additive or synergetic effects by simultaneously blocking multiple signaling molecules and/or pathways that negatively regulate PAH expression.
  • oligonucleotides may also serve as therapeutics, their therapeutic liabilities and treatment outcomes may be ameliorated or predicted, respectively by interrogating the gene signaling networks of the disclosure.
  • expression of the PAH gene may be modulated by altering the chromosomal regions defining the insulated neighborhood(s) and/or genome signaling center(s) associated with the PAH gene.
  • protein production may be increased by targeting a component of the gene signaling network that functions to repress the expression of the PAH gene.
  • Methods of altering the gene expression attendant to an insulated neighborhood include altering the signaling center (e.g. using CRISPR/Cas to change the signaling center binding site or repair/replace if mutated). These alterations may result in a variety of results including: activation of cell death pathways prematurely/inappropriately (key to many immune disorders), production of too little/much gene product (also known as the rheostat hypothesis), production of too little/much extracellular secretion of enzymes, prevention of lineage differentiation, switch of lineage pathways, promotion of sternness, initiation or interference with auto regulatory feedback loops, initiation of errors in cell metabolism, inappropriate imprinting/gene silencing, and formation of flawed chromatin states.
  • CRISPR/Cas to change the signaling center binding site or repair/replace if mutated
  • genome editing approaches including those well-known in the art may be used to create new signaling centers by altering the cohesin necklace or moving genes and enhancers.
  • genome editing approaches describe herein may include methods of using site-specific nucleases to introduce single-strand or double-strand DNA breaks at particular locations within the genome. Such breaks can be and regularly are repaired by endogenous cellular processes, such as homology-directed repair (HDR) and non-homologous end joining (NHEJ).
  • HDR is essentially an error- free mechanism that repairs double-strand DNA breaks in the presence of a homologous DNA sequence.
  • the most common form of HDR is homologous recombination. It utilizes a homologous sequence as a template for inserting or replacing a specific DNA sequence at the break point.
  • the template for the homologous DNA sequence can be an endogenous sequence (e.g., a sister chromatid), or an exogenous or supplied sequence (e.g., plasmid or an oligonucleotide).
  • HDR may be utilized to introduce precise alterations such as replacement or insertion at desired regions.
  • NHEJ is an error-prone repair mechanism that directly joins the DNA ends resulting from a double-strand break with the possibility of losing, adding or mutating a few nucleotides at the cleavage site.
  • the resulting small deletions or insertions (termed“Indels”) or mutations may disrupt or enhance gene expression.
  • NHEJ may be utilized to introduce insertions, deletions or mutations at the cleavage site.
  • a CRISPR/Cas system may be used to delete CTCF anchor sites to modulate gene expression within the insulated neighborhood associated with that anchor site. See, Hnisz et ak, Cell 167, November 17, 2016, which is hereby incorporated by reference in its entirety. Disruption of the boundaries of insulated neighborhood prevents the interactions necessary for proper function of the associated signaling centers. Changes in the expression genes that are immediately adjacent to the deleted neighborhood boundary have also been observed due to such disruptions.
  • a CRISPR/Cas system may be used to modify existing CTCF anchor sites.
  • existing CTCF anchor sites may be mutated or inverted by inducing NHEJ with a CRISPR/Cas nuclease and one or more guide RNAs, or masked by targeted binding with a catalytically inactive CRISPR/Cas enzyme and one or more guide RNAs.
  • Alteration of existing CTCF anchor sites may disrupt the formation of existing insulated neighborhoods and alter the expression of genes located within these insulated neighborhoods.
  • a CRISPR/Cas system may be used to introduce new CTCF anchor sites.
  • CTCF anchor sites may be introduced by inducing HDR at a selected site with a CRISPR/Cas nuclease, one or more guide RNAs and a donor template containing the sequence of a CTCF anchor site.
  • Introduction of new CTCF anchor sites may create new insulated neighborhoods and/or alter existing insulated neighborhoods, which may affect expression of genes that are located adjacent to these insulated neighborhoods.
  • a CRISPR/Cas system may be used to alter signaling centers by changing signaling center binding sites. For example, if a signaling center binding site contains a mutation that affects the assembly of the signaling center with associated transcription factors, the mutated site may be repaired by inducing a double strand DNA break at or near the mutation using a CRISPR/Cas nuclease and one or more guide RNAs in the presence of a supplied corrected donor template.
  • a CRISPR/Cas system may be used to modulate expression of neighborhood genes by binding to a region within an insulated neighborhood (e.g., enhancer) and block transcription. Such binding may prevent recruitment of transcription factors to signaling centers and initiation of transcription.
  • the CRISPR/Cas system may be a catalytically inactive CRISPR/Cas system that do not cleave DNA.
  • a CRISPR/Cas system may be used to knockdown expression of neighborhood genes via introduction of short deletions in coding regions of these genes. When repaired, such deletions would result in frame shifts and/or introduce premature stop codons in mRNA produced by the genes followed by the mRNA
  • a CRISPR/Cas system may also be used to alter cohesion necklace or moving genes and enhancers.
  • CRISPR/Cas systems are bacterial adaptive immune systems that utilize RNA- guided endonucleases to target specific sequences and degrade target nucleic acids. They have been adapted for use in various applications in the field of genome editing and/or transcription modulation. Any of the enzymes or orthologs known in the art or disclosed herein may be utilized in the methods herein for genome editing.
  • the CRISPR/Cas system may be a Type II CRISPR/Cas9 system.
  • Cas9 is an endonuclease that functions together with a trans-activating CRISPR RNA (tracrRNA) and a CRISPR RNA (crRNA) to cleave double stranded DNAs.
  • the two RNAs can be engineered to form a single-molecule guide RNA by connecting the 3’ end of the crRNA to the 5’ end of tracrRNA with a linker loop. Jinek et ak, Science,
  • CRISPR/Cas9 systems include those derived from Streptococcus pyogenes, Streptococcus thermophilus, Neisseria meningitidis, Treponema denticola, Streptococcus aureas, and Francisella tularensis.
  • the CRISPR/Cas system may be a Type V CRISPR/Cpfl system.
  • Cpfl is a single RNA-guided endonuclease that, in contrast to Type II systems, lacks tracrRNA. Cpfl produces staggered DNA double-stranded break with a 4 or 5 nucleotide 5’ overhang.
  • Zetsche et al. Cell. 2015 Oct 22;l63(3):759-7l provides examples of Cpfl endonuclease that can be used in genome editing applications, which is incorporated herein by reference in its entirety.
  • Exemplary CRISPR/Cpfl systems include those derived from Francisella tularensis, Acidaminococcus sp., and Lachnospiraceae bacterium.
  • nickase variants of the CRISPR/Cas endonucleases that have one or the other nuclease domain inactivated may be used to increase the specificity of CRISPR-mediated genome editing.
  • Nickases have been shown to promote HDR versus NHEJ. HDR can be directed from individual Cas nickases or using pairs of nickases that flank the target area.
  • catalytically inactive CRISPR/Cas systems may be used to bind to target regions (e.g., CTCF anchor sites or enhancers) and interfere with their function.
  • Cas nucleases such as Cas9 and Cpfl encompass two nuclease domains. Mutating critical residues at the catalytic sites creates variants that only bind to target sites but do not result in cleavage. Binding to chromosomal regions (e.g., CTCF anchor sites or enhancers) may disrupt proper formation of insulated neighborhoods or signaling centers and therefore lead to altered expression of genes located adjacent to the target region.
  • a CRISPR/Cas system may include additional functional domain(s) fused to the CRISPR/Cas enzyme.
  • the functional domains may be involved in processes including but not limited to transcription activation, transcription repression, DNA methylation, histone modification, and/or chromatin remodeling.
  • Such functional domains include but are not limited to a transcriptional activation domain (e.g., VP64 or KRAB, SID or SID4X), a transcriptional repressor, a recombinase, a transposase, a histone remodeler, a DNA methyltransferase, a cryptochrome, a light inducible/controllable domain or a chemically inducible/controllable domain.
  • a CRISPR/Cas enzyme may be administered to a cell or a patient as one or a combination of the following: one or more polypeptides, one or more mRNAs encoding the polypeptide, or one or more DNAs encoding the polypeptide.
  • guide nucleic acids may be used to direct the activities of an associated CRISPR/Cas enzymes to a specific target sequence within a target nucleic acid.
  • Guide nucleic acids provide target specificity to the guide nucleic acid and
  • CRISPR/Cas complexes by virtue of their association with the CRISPR/Cas enzymes, and the guide nucleic acids thus can direct the activity of the CRISPR/Cas enzymes.
  • guide nucleic acids may be RNA molecules.
  • guide RNAs may be single-molecule guide RNAs.
  • guide RNAs may be chemically modified.
  • more than one guide RNAs may be provided to mediate multiple CRISPR/Cas-mediated activities at different sites within the genome.
  • guide RNAs may be administered to a cell or a patient as one or more RNA molecules or one or more DNAs encoding the RNA sequences.
  • RNPs Ribonucleoprotein complexes
  • the CRISPR/Cas enzyme and guide nucleic acid may each be administered separately to a cell or a patient.
  • the CRISPR/Cas enzyme may be pre-complexed with one or more guide nucleic acids.
  • the pre-complexed material may then be administered to a cell or a patient.
  • Such pre-complexed material is known as a ribonucleoprotein particle (RNP).
  • Zinc finger nucleases are modular proteins comprised of an engineered zinc finger DNA binding domain linked to a DNA- cleavage domain.
  • a typical DNA-cleavage domain is the catalytic domain of the type II endonuclease Fokl. Because Fokl functions only as a dimer, a pair of ZFNs must are required to be engineered to bind to cognate target“half-site” sequences on opposite DNA strands and with precise spacing between them to allow the two enable the catalytically active Fokl domains to dimerize.
  • Upon dimerization of the Fokl domain, which itself has no sequence specificity per se a DNA double-strand break is generated between the ZFN half-sites as the initiating step in genome editing.
  • TALENs Transcription Activator-Like Effector Nucleases
  • genome editing approaches of the present disclosure involve the use of Transcription Activator-Like Effector Nucleases (TALENs).
  • TALENs represent another format of modular nucleases which, similarly to ZFNs, are generated by fusing an engineered DNA binding domain to a nuclease domain, and operate in tandem to achieve targeted DNA cleavage. While the DNA binding domain in ZFN consists of Zinc finger motifs, the TALEN DNA binding domain is derived from transcription activator-like effector (TALE) proteins, which were originally described in the plant bacterial pathogen Xanthomonas sp.
  • TALE transcription activator-like effector
  • TALEs are comprised of tandem arrays of 33-35 amino acid repeats, with each repeat recognizing a single basepair in the target DNA sequence that is typically up to 20 bp in length, giving a total target sequence length of up to 40 bp.
  • Nucleotide specificity of each repeat is determined by the repeat variable diresidue (RVD), which includes just two amino acids at positions 12 and 13.
  • RVD repeat variable diresidue
  • the bases guanine, adenine, cytosine and thymine are predominantly recognized by the four RVDs: Asn-Asn, Asn-Ile, His- Asp and Asn-Gly, respectively.
  • RVD repeat variable diresidue
  • compositions of the present disclosure may be prepared as pharmaceutical compositions. It will be understood that such compositions necessarily comprise one or more active ingredients and, most often, a pharmaceutically acceptable excipient.
  • Relative amounts of the active ingredient, a pharmaceutically acceptable excipient, and/or any additional ingredients in a pharmaceutical composition in accordance with the present disclosure may vary, depending upon the identity, size, and/or condition of the subject being treated and further depending upon the route by which the composition is to be administered.
  • the composition may comprise between 0.1% and 99% (w/w) of the active ingredient.
  • the composition may comprise between 0.1% and 100%, e.g., between .5 and 50%, between 1-30%, between 5-80%, at least 80% (w/w) active ingredient.
  • the pharmaceutical compositions described herein may comprise at least one payload.
  • the pharmaceutical compositions may contain 1, 2, 3, 4 or 5 payloads.
  • compositions are principally directed to pharmaceutical compositions which are suitable for administration to humans, it will be understood by the skilled artisan that such compositions are generally suitable for administration to any other animal, e.g., to non-human animals, e.g. non human mammals. Modification of pharmaceutical compositions suitable for administration to humans in order to render the compositions suitable for administration to various animals is well understood, and the ordinarily skilled veterinary pharmacologist can design and/or perform such modification with merely ordinary, if any, experimentation.
  • Subjects to which administration of the pharmaceutical compositions is contemplated include, but are not limited to, humans and/or other primates; mammals, including commercially relevant mammals such as cattle, pigs, horses, sheep, cats, dogs, mice, rats, birds, including commercially relevant birds such as poultry, chickens, ducks, geese, and/or turkeys.
  • compositions are administered to humans, human patients or subjects.
  • Formulations can include, without limitation, saline, liposomes, lipid
  • nanoparticles polymers, peptides, proteins, cells transfected with viral vectors (e.g., for transfer or transplantation into a subject) and combinations thereof.
  • Formulations of the pharmaceutical compositions described herein may be prepared by any method known or hereafter developed in the art of pharmacology.
  • pharmaceutical composition refers to compositions comprising at least one active ingredient and optionally one or more pharmaceutically acceptable excipients.
  • such preparatory methods include the step of associating the active ingredient with an excipient and/or one or more other accessory ingredients.
  • Formulations of the compositions described herein may be prepared by any method known or hereafter developed in the art of pharmacology. In general, such preparatory methods include the step of bringing the active ingredient into association with an excipient and/or one or more other accessory ingredients, and then, if necessary and/or desirable, dividing, shaping and/or packaging the product into a desired single- or multi dose unit.
  • a pharmaceutical composition in accordance with the present disclosure may be prepared, packaged, and/or sold in bulk, as a single unit dose, and/or as a plurality of single unit doses.
  • a“unit dose” refers to a discrete amount of the pharmaceutical composition comprising a predetermined amount of the active ingredient.
  • the amount of the active ingredient is generally equal to the dosage of the active ingredient which would be administered to a subject and/or a convenient fraction of such a dosage such as, for example, one-half or one-third of such a dosage.
  • Relative amounts of the active ingredient, the pharmaceutically acceptable excipient, and/or any additional ingredients in a pharmaceutical composition in accordance with the present disclosure may vary, depending upon the identity, size, and/or condition of the subject being treated and further depending upon the route by which the composition is to be administered.
  • the composition may comprise between 0.1% and 99% (w/w) of the active ingredient.
  • the composition may comprise between 0.1% and 100%, e.g., between 0.5 and 50%, between 1-30%, between 5-80%, at least 80% (w/w) active ingredient.
  • a pharmaceutically acceptable excipient may be at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% pure.
  • an excipient is approved for use for humans and for veterinary use.
  • an excipient may be approved by United States Food and Drug
  • an excipient may be of pharmaceutical grade. In some embodiments, an excipient may meet the standards of the United States
  • USP European Pharmacopoeia
  • EP European Pharmacopoeia
  • British Pharmacopoeia British Pharmacopoeia
  • International Pharmacopoeia International Pharmacopoeia
  • Excipients include, but are not limited to, any and all solvents, dispersion media, diluents, or other liquid vehicles, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, and the like, as suited to the particular dosage form desired.
  • Various excipients for formulating pharmaceutical compositions and techniques for preparing the composition are known in the art (see Remington: The Science and Practice of Pharmacy, 2lst Edition, A. R.
  • Exemplary diluents include, but are not limited to, calcium carbonate, sodium carbonate, calcium phosphate, dicalcium phosphate, calcium sulfate, calcium hydrogen phosphate, sodium phosphate lactose, sucrose, cellulose, microcrystalline cellulose, kaolin, mannitol, sorbitol, inositol, sodium chloride, dry starch, cornstarch, powdered sugar, etc., and/or combinations thereof.
  • the pharmaceutical compositions formulations may comprise at least one inactive ingredient.
  • the term“inactive ingredient” refers to one or more agents that do not contribute to the activity of the active ingredient of the pharmaceutical composition included in formulations.
  • all, none or some of the inactive ingredients which may be used in the formulations of the present disclosure may be approved by the US Food and Drug Administration (FDA).
  • the pharmaceutical compositions comprise at least one inactive ingredient such as, but not limited to, l,2,6-Hexanetriol; l,2-Dimyristoyl-Sn- Glycero-3-(Phospho-S-(l-Glycerol)); l,2-Dimyristoyl-Sn-Glycero-3-Phosphocholine; 1,2- Dioleoyl-Sn-Glycero-3-Phosphocholine; l,2-Dipalmitoyl-Sn-Glycero-3-(Phospho-Rac-(l- Glycerol)); l,2-Distearoyl-Sn-Glycero-3-(Phospho-Rac-(l-Glycerol)); l,2-Distearoyl-Sn- Glycero-3-Phosphocholine; l-O-Tolylbiguanide; 2-Ethyl- l,6-Hexanetriol; l,2-
  • Ammonium Sulfonic Acid Betaine Alkyl Aryl Sodium Sulfonate; Allantoin; Allyl .Alpha.-Ionone; Almond Oil; Alpha-Terpineol; Alpha-Tocopherol; Alpha-Tocopherol Acetate, D1-; Alpha-Tocopherol, D1-; Aluminum Acetate; Aluminum Chlorhydroxy Allantoinate; Aluminum Hydroxide; Aluminum Hydroxide - Sucrose, Hydrated; Aluminum Hydroxide Gel; Aluminum Hydroxide Gel F 500; Aluminum Hydroxide Gel F 5000; Aluminum Monostearate; Aluminum Oxide; Aluminum Polyester; Aluminum Silicate; Aluminum Starch Octenylsuccinate; Aluminum Stearate; Aluminum Subacetate; Aluminum Sulfate Anhydrous; Amerchol C; Amerchol-Cab; Aminomethylpropanol;
  • Benzenesulfonic Acid Benzethonium Chloride; Benzododecinium Bromide; Benzoic Acid; Benzyl Alcohol; Benzyl Benzoate; Benzyl Chloride; Betadex; Bibapcitide; Bismuth Subgallate; Boric Acid; Brocrinat; Butane; Butyl Alcohol; Butyl Ester Of Vinyl Methyl Ether/Maleic Anhydride Copolymer (125000 Mw); Butyl Stearate; Butylated
  • Caprylocaprate Cola Nitida Seed Extract; Collagen; Coloring Suspension; Corn Oil;
  • Cottonseed Oil Cream Base; Creatine; Creatinine; Cresol; Croscarmellose Sodium;
  • Crospovidone Cupric Sulfate; Cupric Sulfate Anhydrous; Cyclomethicone;
  • Cyclomethicone/Dimethicone Copolyol Cysteine; Cysteine Hydrochloride; Cysteine Hydrochloride Anhydrous; Cysteine, D1-; D&C Red No. 28; D&C Red No. 33; D&C Red No. 36; D&C Red No. 39; D&C Yellow No. 10; Dalfampridine; Daubert 1-5 Pestr (Matte) l64z; Decyl Methyl Sulfoxide; Dehydag Wax Sx; Dehydroacetic Acid; Dehymuls E; Denatonium Benzoate; Deoxycholic Acid; Dextran; Dextran 40; Dextrin; Dextrose;
  • Dichlorobenzyl Alcohol Dichlorodifluorome thane; Dichlorotetrafluoroethane;
  • Cocoamphodiacetate Disodium Laureth Sulfosuccinate; Disodium Lauryl Sulfosuccinate; Disodium Sulfosalicylate; Disofenin; Divinylbenzene Styrene Copolymer; Dmdm
  • Ethylcelluloses Ethylene Glycol; Ethylene Vinyl Acetate Copolymer; Ethylenediamine; Ethylenediamine Dihydrochloride; Ethylene-Propylene Copolymer; Ethylene- Vinyl Acetate Copolymer (28% Vinyl Acetate); Ethylene- Vinyl Acetate Copolymer (9%
  • Fragrance Bouquet 10328 Fragrance Chemoderm 6401-B; Fragrance Chemoderm 6411; Fragrance Cream No. 73457; Fragrance Cs-28l97; Fragrance Felton 066m;
  • Fragrance Firmenich 47373; Fragrance Givaudan Ess 9090/lc; Fragrance H-6540;
  • Octoxynol-9 Octyldodecanol
  • Octylphenol Polymethylene Oleic Acid; Oleth-lO/Oleth-5; Oleth-2; Oleth-20; Oleyl Alcohol; Oleyl Oleate; Olive Oil; Oxidronate Disodium;
  • Phenylethyl Alcohol Phenylmercuric Acetate; Phenylmercuric Nitrate; Phosphatidyl Glycerol, Egg; Phospholipid; Phospholipid, Egg; Phospholipon 90g; Phosphoric Acid; Pine Needle Oil (Pinus Sylvestris); Piperazine Hexahydrate; Plastibase-50w; Polacrilin;
  • Polidronium Chloride Polidronium Chloride; Poloxamer 124; Poloxamer 181; Poloxamer 182; Poloxamer 188; Poloxamer 237; Poloxamer 407; Poly(Bis(P-Carboxyphenoxy)Propane
  • Polyethylene Glycol 1500 Polyethylene Glycol 1540; Polyethylene Glycol 200;
  • Polyethylene Glycol 300 Polyethylene Glycol 300-1600; Polyethylene Glycol 3350; Polyethylene Glycol 400; Polyethylene Glycol 4000; Polyethylene Glycol 540;
  • Polyethylene Glycol 900 Polyethylene High Density Containing Ferric Oxide Black ( ⁇ l%); Polyethylene Low Density Containing Barium Sulfate (20-24%); Polyethylene T; Polyethylene Terephthalates; Polyglactin; Poly glyceryl-3 Oleate; Poly glyceryl-4 Oleate; Polyhydroxyethyl Methacrylate; Polyisobutylene; Polyisobutylene (1100000 Mw);
  • Polyisobutylene (35000 Mw); Polyisobutylene 178-236; Polyisobutylene 241-294;
  • Polyoxyethylene - Polyoxypropylene 1800 Polyoxyethylene Alcohols; Polyoxyethylene Fatty Acid Esters; Polyoxyethylene Propylene; Polyoxyl 20 Cetostearyl Ether; Polyoxyl 35 Castor Oil; Polyoxyl 40 Hydrogenated Castor Oil; Polyoxyl 40 Stearate; Polyoxyl 400 Stearate; Polyoxyl 6 And Polyoxyl 32 Palmitostearate; Polyoxyl Distearate; Polyoxyl Glyceryl Stearate; Polyoxyl Lanolin; Polyoxyl Palmitate; Polyoxyl Stearate;
  • Monoalky lolamide Sodium Tartrate; Sodium Thioglycolate; Sodium Thiomalate; Sodium Thiosulfate; Sodium Thiosulfate Anhydrous; Sodium Trimetaphosphate; Sodium
  • Somay 44 Sorbic Acid; Sorbitan; Sorbitan Isostearate; Sorbitan
  • Styrene/Isoprene/Styrene Block Copolymer Succimer; Succinic Acid; Sucralose; Sucrose; Sucrose Distearate; Sucrose Polyesters; Sulfacetamide Sodium; Sulfobutylether .Beta.- Cyclodextrin; Sulfur Dioxide; Sulfuric Acid; Sulfurous Acid; Surfactol Qs; Tagatose, D-; Talc; Tall Oil; Tallow Glycerides; Tartaric Acid; Tartaric Acid, D1-; Tenox; Tenox-2; Tert- Butyl Alcohol; Tert-Butyl Hydroperoxide; Tert-Butylhydroquinone; Tetrakis(2- Methoxyisobutylisocyanide)Copper(I) Tetrafluoroborate; Tetrapropyl Orthosilicate;
  • Tetrofosmin Theophylline; Thimerosal; Threonine; Thymol; Tin; Titanium Dioxide; Tocopherol; Tocophersolan; Total parenteral nutrition, lipid emulsion; Triacetin;
  • Tricaprylin Trichloromonofluoromethane; Trideceth-lO; Triethanolamine Lauryl Sulfate; Trifluoroacetic Acid; Triglycerides, Medium Chain; Trihydroxystearin; Trilaneth-4 Phosphate; Trilaureth-4 Phosphate; Trisodium Citrate Dihydrate; Trisodium Hedta; Triton 720; Triton X-200; Trolamine; Tromantadine; Tromethamine (TRIS); Tryptophan;
  • Tyloxapol Tyrosine; Undecylenic Acid; Union 76 Amsco-Res 6038; Urea; Valine;
  • Vegetable Oil Vegetable Oil Glyceride, Hydrogenated; Vegetable Oil, Hydrogenated; Versetamide; Viscarin; Viscose/Cotton; Vitamin E; Wax, Emulsifying; Wecobee Fs; White Ceresin Wax; White Wax; Xanthan Gum; Zinc; Zinc Acetate; Zinc Carbonate; Zinc Chloride; and Zinc Oxide.
  • composition formulations disclosed herein may include cations or anions.
  • the formulations include metal cations such as, but not limited to, Zn2+, Ca2+, Cu2+, Mn2+, Mg+ and combinations thereof.
  • formulations may include polymers and complexes with a metal cation ( See e.g. , U.S. Pat. Nos. 6,265,389 and 6,555,525, each of which is herein incorporated by reference in its entirety).
  • Formulations may also include one or more pharmaceutically acceptable salts.
  • “pharmaceutically acceptable salts” refers to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or base moiety to its salt form (e.g., by reacting the free base group with a suitable organic acid).
  • suitable organic acid examples include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like.
  • Representative acid addition salts include acetate, acetic acid, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzene sulfonic acid, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate, heptonate, hexanoate, hydrobromide, hydrochloride, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate
  • alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like, as well as nontoxic ammonium, quaternary ammonium, and amine cations, including, but not limited to ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, and the like.
  • the pharmaceutically acceptable salts of the present disclosure include the conventional non-toxic salts of the parent compound formed, for example, from non-toxic inorganic or organic acids.
  • Solvates may be prepared by crystallization, recrystallization, or precipitation from a solution that includes organic solvents, water, or a mixture thereof.
  • suitable solvents are ethanol, water (for example, mono-, di-, and trl -hydrates), N- methylpyrrolidinone (NMP), dimethyl sulfoxide (DMSO), NN '-di methyl formamide (DMF), 7V,7V’-dimethylacetamide (DMAC), l,3-dimethyl-2-imidazolidinone (DMEU), 1,3- dimethyl-3,4,5,6-tetrahydro-2-(lH)-pyrimidinone (DMPU), acetonitrile (ACN), propylene glycol, ethyl acetate, benzyl alcohol, 2-pyrrolidone, benzyl benzoate, and the like.
  • water is the solvent
  • the solvate is referred to as a“hydrate.”
  • administering and "introducing” are used interchangeable herein and refer to the delivery of the pharmaceutical composition into a cell or a subject.
  • the pharmaceutical composition is delivered by a method or route that results in at least partial localization of the introduced cells at a desired site, such as hepatocytes, such that a desired effect(s) is produced.
  • the pharmaceutical composition may be any pharmaceutical composition.
  • a route such as, but not limited to, enteral (into the intestine), gastroenteral, epidural (into the dura matter), oral (by way of the mouth), transdermal, peridural, intracerebral (into the cerebrum), intracerebro ventricular (into the cerebral ventricles), epicutaneous (application onto the skin), intradermal, (into the skin itself), subcutaneous (under the skin), nasal administration (through the nose), intravenous (into a vein), intravenous bolus, intravenous drip, intraarterial (into an artery), intramuscular (into a muscle), intracardiac (into the heart), intraosseous infusion (into the bone marrow), intrathecal (into the spinal canal), intraperitoneal, (infusion or injection into the peritoneum), intravesical infusion, intravitreal, (through the eye), intracavernous injection (into a pathologic cavity) intracavitary (into the base of the penis),
  • intrauterine, extra- amniotic administration transdermal (diffusion through the intact skin for systemic distribution), transmucosal (diffusion through a mucous membrane), transvaginal, insufflation (snorting), sublingual, sublabial, enema, eye drops (onto the conjunctiva), in ear drops, auricular (in or by way of the ear), buccal (directed toward the cheek), conjunctival, cutaneous, dental (to a tooth or teeth), electro-osmosis, endocervical, endosinusial, endotracheal, extracorporeal, hemodialysis, infiltration, interstitial, intra-abdominal, intra-amniotic, intra-articular, intrabiliary, intrabronchial, intrabursal, intracartilaginous (within a cartilage), intracaudal (within the cauda equine), intracistemal (within the cistem
  • Modes of administration include injection, infusion, instillation, and/or ingestion.
  • injection includes, without limitation, intravenous, intramuscular, intra-arterial, intrathecal, intraventricular, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, sub capsular, subarachnoid, intraspinal, intracerebro spinal, and intrasternal injection and infusion.
  • the route is intravenous.
  • administration by injection or infusion can be made.
  • the cells can be administered systemically.
  • administered peripherally refer to the administration other than directly into a target site, tissue, or organ, such that it enters, instead, the subject's circulatory system and, thus, is subject to metabolism and other like processes.
  • the term "effective amount” refers to the amount of the active ingredient needed to prevent or alleviate at least one or more signs or symptoms of a specific disease and/or condition, and relates to a sufficient amount of a composition to provide the desired effect.
  • the term "therapeutically effective amount” therefore refers to an amount of active ingredient or a composition comprising the active ingredient that is sufficient to promote a particular effect when administered to a typical subject.
  • An effective amount would also include an amount sufficient to prevent or delay the development of a symptom of the disease, alter the course of a symptom of the disease (for example but not limited to, slow the progression of a symptom of the disease), or reverse a symptom of the disease. It is understood that for any given case, an appropriate "effective amount” can be determined by one of ordinary skill in the art using routine experimentation.
  • compositions of the present disclosure may be administered to a subject using any amount and any route of administration effective for preventing, treating, managing, or diagnosing diseases, disorders and/or conditions.
  • the exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the disease, the particular composition, its mode of administration, its mode of activity, and the like.
  • the subject may be a human, a mammal, or an animal.
  • Compositions in accordance with the disclosure are typically formulated in unit dosage form for ease of administration and uniformity of dosage. It will be understood, however, that the total daily usage of the compositions of the present disclosure may be decided by the attending physician within the scope of sound medical judgment.
  • prophylactically effective, or appropriate diagnostic dose level for any particular individual will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific payload employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, and route of administration; the duration of the treatment; drugs used in combination or coincidental with the active ingredient; and like factors well known in the medical arts.
  • compositions in accordance with the present disclosure may be administered at dosage levels sufficient to deliver from about 0.01 mg/kg to about 100 mg/kg, from about 0.01 mg/kg to about 0.05 mg/kg, from about 0.05 mg/kg to about 0.5 mg/kg, from about 0.01 mg/kg to about 50 mg/kg, from about 0.1 mg/kg to about 40 mg/kg, from about 0.5 mg/kg to about 30 mg/kg, from about 0.01 mg/kg to about 10 mg/kg, from about 0.1 mg/kg to about 10 mg/kg, or from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic, diagnostic, or prophylactic, effect.
  • the desired dosage of the composition present disclosure may be delivered only once, three times a day, two times a day, once a day, every other day, every third day, every week, every two weeks, every three weeks, or every four weeks.
  • the desired dosage may be delivered using multiple administrations (e.g. , two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, or more administrations).
  • split dosing regimens such as those described herein may be used.
  • a“split dose” is the division of “single unit dose” or total daily dose into two or more doses, e.g., two or more
  • a“single unit dose” is a dose of any therapeutic administered in one dose/at one time/single route/single point of contact, i.e., single administration event.
  • Human hepatocytes were obtained from two donors from Massachusetts General Hospital, namely MGH54 and MGH63, and one donor from Lonza, namely HUM4111B. Cryopreserved hepatocytes were cultured in plating media for 16 hours, transferred to maintenance media for 4 hours. Cultured on serum-free media for 2 hours, then a compound was added. The hepatocytes were maintained on the serum-free media for 16 hours prior to gene expression analysis. Primary human hepatocytes were stored in the vapor phase of a liquid nitrogen freezer (about -l30°C).
  • vials of cells were retrieved from the LN2 freezer, thawed in a 37°C water bath, and swirled gently until only a sliver of ice remains.
  • cells were gently pipetted out of the vial and gently pipetted down the side of 50mL conical tube containing 20mL cold thaw medium.
  • the vial was rinsed with about lmL of thaw medium, and the rinse was added to the conical tube. Up to 2 vials were added to one tube of 20mL thaw medium.
  • the conical tube(s) were gently inverted 2-3 times and centrifuged at 100 g for 10 minutes at 4°C with reduced braking (e.g. 4 out of 9).
  • the thaw medium slowly was slowly aspirated to avoid the pellet.
  • 4 mL cold plating medium was added slowly down the side (8 mL if combined 2 vials to 1 tube), and the vial was inverted gently several times to resuspend cells.
  • the plate was transferred to an incubator (37°C, 5% CC about 90% humidity) and rocked forwards and backwards, then side to side several times each to distribute cells evenly across the plate or wells.
  • the plate(s) were rocked again every 15 minutes for the first hour post-plating.
  • About 4 hours post-plating (or first thing the morning if cells were plated in the evening), cells were washed once with PBS and complete maintenance medium was added.
  • the primary human hepatocytes were maintained in the maintenance medium and transferred to fresh medium daily.
  • mice Female C57BL/6 mouse hepatocytes (F005152-cry opreserved) were purchased from BioreclamationIVT as a pool of 45 donors. Cells were plated in InvitroGRO CP Rodent Medium (Z990028) and Torpedo Rodent Antibiotic Mix (Z99027) on Collagen- coated 24-well plates for 24 hours at 200K cells/well in 0.5 mL media. Compound stocks in 10 mM DMSO, were diluted to 10 uM (with final concentration of 1% DMSO), and applied on cells in biological triplicates. Medium was removed after 20 hours and cells processed for further analysis, e.g. qRT-PCR.
  • the thaw medium contained 6 mL isotonic percoll and 14 mL high glucose DMEM (Invitrogen #11965 or similar).
  • the plating medium contained 100 mL Williams E medium (Invitrogen #A1217601, without phenol red) and the supplement pack #CM3000 from ThermoFisher Plating medium containing 5mL FBS, 10m1 dexamethasone, and 3.6 mL plating/maintenance cocktail.
  • Stock trypan blue (0.4%, Invitrogen #15250) was diluted 1:5 in PBS. Normocin was added at 1:500 to both the thaw medium and the plating medium.
  • ThermoFisher complete maintenance medium contained supplement pack #CM4000 (1 pl dexamethasone and 4 mL maintenance cocktail) and 100 mL Williams E (Invitrogen #A1217601, without phenol red).
  • the modified maintenance media had no stimulating factors (dexamethasone, insulin, etc.), and contained lOOmL Williams E (Invitrogen #A1217601, without phenol red), lmL L-Glutamine (Sigma #G75l3) to 2 mM, 1.5 mL HEPES (VWR #J848) to 15 mM, and 0.5 mL penicillin/streptomycin (Invitrogen #15140) to a final concentration of 50 U/mL each.
  • the cell lysate was layered on top of 2.5 volumes of sucrose cushion made up of 24% (wt/vol) sucrose in Nonidet P-40 lysis buffer. This sample was centrifuged at 18,000 g for 10 minutes at 4°C to isolate the nuclei pellet (the supernatant represented the cytoplasmic fraction). The nuclei pellet was washed once with PBS/l mM EDTA. The nuclei pellet was resuspended gently with 0.5 mL glycerol buffer followed by incubation for 2 minutes on ice with an equal volume of nuclei lysis buffer. The sample was centrifuged at 16,000 g for 2 minutes at 4°C to isolate the chromatin pellet (the supernatant represented the nuclear soluble fraction). The chromatin pellet was washed twice with PBS/l mM EDTA. The chromatin pellet was stored at -80°C.
  • the Nonidet P-40 lysis buffer contained 10 mM Tris-HCl (pH 7.5), 150 mM NaCl, and 0.05% Nonidet P-40.
  • the glycerol buffer contained 20 mM Tris-HCl (pH 7.9), 75 mM NaCl, 0.5 mM EDTA, 0.85 mM DTT, and 50% (vol/vol) glycerol.
  • the nuclei lysis buffer contained 10 mM Hepes (pH 7.6), 1 mM DTT, 7.5 mM MgCK 0.2 mM EDTA, 0.3 M NaCl, 1 M urea, and 1% Nonidet P-40.
  • ChIP-seq was performed using the following protocol for primary hepatocytes and HepG2 cells to determine the composition and confirm the location of signaling centers.
  • the cells were transferred to 15 ml conical tubes, and the tubes were placed on ice. Plates were washed with an additional 4 ml of PBS and combined with cells in 15 ml tubes. Tubes were centrifuged for 5 minutes at 1,500 rpm at 4°C in a tabletop centrifuge. PBS was aspirated, and the cells were flash frozen in liquid nitrogen. Pellets were stored at -80°C until ready to use.
  • COMPLETE® protease inhibitor cocktail was added to lysis buffer 1 (LB1) before use.
  • One tablet was dissolved in 1 ml of fLO for a 50x solution.
  • the cocktail was stored in aliquots at -20°C.
  • Cells were resuspended in each tube in 8 ml of LB1 and incubated on a rotator at 4°C for 10 minutes.
  • Nuclei were spun down at 1,350 g for 5 minutes at 4°C.
  • LB1 was aspirated, and cells were resuspended in each tube in 8 ml of LB2 and incubated on a rotator at 4°C for 10 minutes.
  • a COVARIS ® E220E VOLUTION TM ultrasonicator was programmed per the manufacturer’s recommendations for high cell numbers. HepG2 cells were sonicated for 12 minutes, and primary hepatocyte samples were sonicated for 10 minutes. Lysates were transferred to clean l.5ml Eppendorf tubes, and the tubes were centrifuged at 20,000 g for 10 minutes at 4°C to pellet debris. The supernatant was transferred to a 2 ml Protein LoBind Eppendorf tube containing pre-blocked Protein G beads with pre -bound antibodies. Fifty pl of the supernatant was saved as input. Input material was kept at -80°C until ready to use. Tubes were rotated with beads overnight at 4°C.
  • IP samples were transferred to fresh tubes, and 300 m ⁇ of TE buffer was added to IP and Input samples to dilute SDS. RNase A (20 mg/ml) was added to the tubes, and the tubes were incubated at 37°C for 30 minutes.
  • MaXtract High Density 2 ml gel tubes (Qiagen) were prepared by centrifugation at full speed for 30 seconds at RT. Six hundred m ⁇ of phenol/chloroform/isoamyl alcohol was added to each proteinase K reaction and transferred in about 1.2 ml mixtures to the MaXtract tubes. Tubes were spun at 16,000 g for 5 minutes at RT.
  • the aqueous phase was transferred to two clean DNA LoBind tubes (300 m ⁇ in each tube), and 1.5 m ⁇ glycogen, 30 m ⁇ of 3M sodium acetate, and 900 m ⁇ ethanol were added.
  • the mixture was precipitated overnight at -20°C or for 1 hour at -80°C, and spun down at maximum speed for 20 minutes at 4°C.
  • the ethanol was removed, and pellets were washed with 1 ml of 75% ethanol by spinning tubes down at maximum speed for 5 minutes at 4°C. Remnants of ethanol were removed, and pellets were dried for 5 min at RT. Twenty-five m ⁇ of 3 ⁇ 40 was added to each immunoprecipitant (IP) and input pellet, left standing for 5 minutes, and vortexed briefly.
  • IP immunoprecipitant
  • DNA from both tubes was combined to obtain 50 m ⁇ of IP and 50 m ⁇ of input DNA for each sample.
  • One m ⁇ of this DNA was used to measure the amount of pulled down DNA using Qubit dsDNA HS assay (ThermoFisher, #Q32854).
  • the total amount of immunoprecipitated material ranged from several ng (for TFs) to several hundred ng (for chromatin modifications).
  • Six m ⁇ of DNA was analyzed using qRT-PCR to determine enrichment. The DNA was diluted if necessary. If enrichment was satisfactory, the rest was used for library preparation for DNA sequencing.
  • PCR product was eluted with 17 m ⁇ of 0.1X TE buffer, and the amount of PCT product was measured using Qubit dsDNA HS assay. An additional 4 cycles of PCR were run for the second half of samples with less than 5 ng of PCR product, DNA was purified using 22.5 m ⁇ of AMPure XP beads. The concentration was measured to determine whether there was an increased yield. Both halves were combined, and the volume was brought up to 50 m ⁇ using fTO.
  • Sensitivity Bio Analyzer DNA kit (Agilent, #5067-4626) based on manufacturer’s recommendations
  • Formaldehyde Solution contained l4.9ml of 37% formaldehyde (final cone. 11%), 1 ml of 5 M NaCl (final cone. 0.1 M), IOOmI of 0.5M EDTA (pH 8)
  • Block Solution contained 0.5% BSA (w/v) in PBS and 500 mg BSA in 100 ml PBS. Block solution may be prepared up to about 4 days prior to use.
  • Lysis buffer 1 (LB1) (500 ml) contained 25 ml of 1 M Hepes-KOH, pH 7.5; 14 ml of 5 M NaCl; 1 ml of 0.5 M EDTA, pH 8.0; 50 ml of 100% Glycerol solution; 25 ml of 10% NP-40; and 12.5 ml of 10% Triton X-100. The pH was adjusted to 7.5. The buffer was sterile-filtered, and stored at 4 °C. The pH was re-checked immediately prior to use.
  • Lysis buffer 2 (LB2) (lOOOml) contained 10 ml of 1 M Tris-HCL, pH 8.0; 40 ml of 5 M NaCl; 2 ml of 0.5 M EDTA, pH 8.0; and 2 ml of 0.5 M EGTA, pH 8.0. The pH was adjusted to 8.0. The buffer was sterile-filtered, and stored at 4 °C. The pH was re-checked immediately prior to use.
  • Sonication buffer 500 ml contained 25 ml of 1 M Hepes-KOH, pH 7.5; 14 ml of 5 M NaCl; 1 ml of 0.5 M EDTA, pH 8.0; 50 ml of 10% Triton X-100; 10 ml of 5% Na- deoxycholate; and 5 ml of 10% SDS. The pH was adjusted to 7.5. The buffer was sterile- filtered, and stored at 4 °C. The pH was re-checked immediately prior to use.
  • Proteinase inhibitors were included in the LB1, LB2, and Sonication buffer.
  • Wash Buffer 2 (500 ml) contained 25 ml of 1 M Hepes-KOH, pH 7.5; 35 ml of 5 M NaCl; 1 ml of 0.5 M EDTA, pH 8.0; 50 ml of 10% Triton X-100; 10 ml of 5% Na- deoxycholate; and 5 ml of 10% SDS. The pH was adjusted to 7.5. The buffer was sterile- filtered, and stored at 4 °C. The pH was re-checked immediately prior to use.
  • Wash Buffer 3 (500ml) contained 10ml of 1 M Tris-HCL, pH 8.0; 1 ml of 0.5 M EDTA, pH 8.0; 125 ml of 1 M LiCl solution; 25 ml of 10% NP-40; and 50 ml of 5% Na- deoxycholate. The pH was adjusted to 8.0. The buffer was sterile-filtered, and stored at 4 °C. The pH was re-checked immediately prior to use.
  • ChIP elution Buffer (500ml) contained 25ml of 1 M Tris-HCL, pH 8.0; 10 ml of 0.5 M EDTA, pH 8.0; 50 ml of 10% SDS; and 415 ml of ddH 2 0. The pH was adjusted to 7.5. The buffer was sterile-filtered, and stored at 4 °C. The pH was re-checked immediately prior to use.
  • ChIP-seq signals were also normalized by read depth and visualized using the UCSC browser.
  • This protocol is a modified version of the following protocols: MagMAX m/rVana Total RNA Isolation Kit User Guide (Applied Biosystems #MAN00ll l3l Rev B.0), NEBNext Poly(A) mRNA Magnetic Isolation Module (E7490), and NEBNext Ultra Directional RNA Library Prep Kit for Illumina (E7420) (New England Biosystems #E7490l).
  • the MagMAX m/rVana kit instructions (the section titled“Isolate RNA from cells” on pages 14-17) were used for isolation of total RNA from cells in culture. Two hundred pl of Lysis Binding Mix was used per well of the multiwell plate containing adherent cells (usually a 24-well plate).
  • RNA isolation and library prep For mRNA isolation and library prep, the NEBNext Poly(A) mRNA Magnetic Isolation Module and Directional Prep kit was used. RNA isolated from cells above was quantified, and prepared in 500 pg of each sample in 50 pl of nuclease-free water. This protocol may be run in microfuge tubes or in a 96-well plate.
  • the libraries were quantified using the Qubit DNA High Sensitivity Kit. 1 m ⁇ of each sample were diluted to 1-2 ng/pl to ran on the Bioanalyzer (DNA High Sensitivity Kit, Agilent # 5067-4626). If Bioanalyzer peaks were not clean (one narrow peak around 300bp), the AMPure XP bead cleanup step was repeated using a 0.9X or 1.0X beads: sample ratio. Then, the samples were quantified again with the Qubit, and ran again on the Bioanalyzer (1-2 ng/pl).
  • RNA from INTACT -purified nuclei or whole neocortical nuclei was converted to cDNA and amplified with the Nugen Ovation RNA-seq System V2. Libraries were sequenced using the Illumina HiSeq 2500.
  • RNA-seq signals were also normalized by read depth and visualized using the UCSC browser.
  • qRT-PCR was performed as described in North et ak, PNAS, 107(40) 17315- 17320 (2010), which is hereby incorporated by reference in its entirety.
  • cell medium Prior to qRT-PCR analysis, cell medium was removed and replaced with RLT Buffer for RNA extraction (Qiagen RNeasy 96 QIAcube HT Kit Cat#74l7l). Cells were processed for RNA extraction using RNeasy 96 kit (Qiagen Cat#74l82).
  • cDNA was synthesized using High-Capacity cDNA Reverse Transcription Kit (ThermoFisher Scientific cat:43688l3 or 4368814) according to manufacturer instructions.
  • qRT-PCR was performed with cDNA using the iQ5 Multicolor rtPCR Detection system from BioRad with 60°C annealing.
  • Samples were amplified using the following Taqman probes from ThermoFisher: Hs00609359_ml (human PAH), Mm005009l8_ml (mouse PAH); Hs0l026983_ml (JAK1); Hs0l078l36_ml (JAK2); 4352341E (ACTB); 4326320E (GUSB); 4326319E (B2M); and 4326317E (GAPDH).
  • RQ Min and RQ Max values are also reported.
  • ChlA-PET was performed as previously described in Chepelev et al. (2012) Cell Res. 22, 490-503; Fullwood et al. (2009) Nature 462, 58-64; Goh et al. (2012) J. Vis. Exp., http://dx.doi.org/l0.379l/3770; Li et al. (2012) Cell 148, 84-98; and Dowen et al. (2014) Cell 159, 374-387, which are each hereby incorporated by reference in their entireties. Briefly, embryonic stem (ES) cells (up to lxlO 8 cells) were treated with 1% formaldehyde at room temperature for 20 minutes and then neutralized using 0.2M glycine.
  • ES embryonic stem
  • the crosslinked chromatin was fragmented by sonication to size lengths of 300-700 bp.
  • the anti-SMCl antibody (Bethyl, A300-055A) was used to enrich SMCl-bound chromatin fragments.
  • a portion of ChIP DNA was eluted from antibody-coated beads for concentration quantification and for enrichment analysis using quantitative PCR.
  • ChIP DNA fragments were end-repaired using T4 DNA polymerase (NEB). ChIP DNA fragments were divided into two aliquots and either linker A or linker B were ligated to the fragment ends.
  • the two linkers differ by two nucleotides which were used as a nucleotide barcode (Linker A with CG; I .inker B with AT). After linker ligation, the two samples were combined and prepared for proximity ligation by diluting in a 20 ml volume to minimize ligations between different DNA-protein complexes. The proximity ligation reaction was performed with T4 DNA ligase
  • the cells were crosslinked as described for ChIP. Frozen cell pellets are stored in the -80°C freezer until ready to use. This protocol required at least 3xl0 8 cells frozen in six l5ml Falcon tubes (50 million cells per tube). Six 100 pl Protein G Dynabeads (for each ChIA-RET sample) are added to six l.5ml Eppendorf tubes on ice. Beads were washed three times with 1.5 ml Block solution, and incubated end over end at 4°C for 10 minutes between each washing step to allow for efficient blocking.
  • Protein G Dynabeads were resuspended in 250 m ⁇ of Block solution in each of six tubes and 10 mg of SMC1 antibody (Bethyl A300- 055A) was added to each tube. The bead-antibody mixes were incubated at 4°C end-over- end overnight.
  • Supernatant (SNE) was pooled into a new pre-cooled 50 ml Falcon tube, and brought to a volume of 18 ml with sonication buffer. Two tubes of 50 pl were taken as input and to check the size of fragments. 250 pl of ChIP elution buffer was added and reverse crosslinking occured at 65°C overnight in the oven. After reversal of crosslinking, the size of sonication fragments wa determined on a gel.
  • ChIP-DNA was quantified using the following protocol. Ten percent of beads (by volume), or 100 m ⁇ , were transferred into a new 1.5 ml tube, using a magnet. Beads were resuspended in 300 m ⁇ of ChIP elution buffer and the tube was rotated with beads for 1 hour at 65°C. The tube with beads was placed on a magnet and the eluate was transferred to a fresh DNA LoBind Eppendorf tube. The eluate was incubated overnight at 65°C oven without rotating. Immuno-precipitated samples were transferred to fresh tubes, and 300 pl of TE buffer was added to the immuno-precipitants and Input samples to dilute. Five pl of RNase A (20mg/ml) was added, and the tube was incubated at 37°C for 30 minutes.
  • the aqueous phase was transferred to two clean DNA LoBind tubes (300 m ⁇ in each tube), and 1 pl glycogen, 30 pl of 3 M sodium acetate, and 900 m ⁇ ethanol was added. The mixture was allowed to precipitate overnight at -20°C or for 1 hour at -80°C.
  • On-Bead A-tailing was performed by preparing Klenow (3 To 5'exo-) master mix as stated below: 70 m ⁇ 10X NEB buffer 2, 7 pl 10 mM dATP, 616 m ⁇ dH20, and 7 pl of 3U/pl Klenow (3 "to 5"exo-) (NEB, M0212L). The mixture was incubated at 37°C with rotation for 50 minutes. Beads were collected with a magnet, then beads were washed 3 times with 1 ml of ice-cold ChIA-RET Wash Buffer (30 seconds per each wash).
  • Tinkers were thawed gently on ice. T nkers were mixed weii with water gentiy by pipetting, then with PEG buffer, then gentiy vortexed. Then, T394 pi of master mix and 6 pi of ligase was added per tube and mixed by inversion. Parafilm was put on the tube, and the tube was incubated at 16°C with rotation overnight (at least 16 hours).
  • the biotinylated linker was ligated to ChIP-DNA on beads by setting up the following reaction mix and adding reagents in order: 1110 pi d!TO, 4 pi 200 ng/pl biotinylated bridge linker, 280 pi 5X T4 DNA ligase buffer with PEG (Invitrogen), and 6 pi 30 U/pl T4 DNA ligase (Fermentas).
  • Exonuclease lambda/Exonuclease I On-Bead digestion was performed using the following protocol. Beads were collected with a magnet and washed 3 times with 1ml of ice- cold ChIA-RET Wash Buffer (30 seconds per each wash). The Wash buffer was removed from beads, then resuspended in the following reaction mix: 70 pi 10X lambda nuclease buffer (NEB, M0262L), 618 pi nuclease-free dH20, 6 pi 5 U/pl Lambda Exonuclease (NEB, M0262L), and 6 pi Exonuclease I (NEB, M0293L). The reaction was incubated at 37°C with rotation for 1 hour. Beads were collected with a magnet, and beads are washed 3 times with lml ice-cold ChIA-RET Wash Buffer (30 seconds per each wash).
  • Chromatin complexes were eluted off the beads by removing all residual buffer and resuspending the beads in 300 pi of ChIP elution buffer. The tube with beads was rotated 1 hour at 65°C. The tube was placed on a magnet and the eluate was transferred to a fresh DNA LoBind Eppendorf tube. The eluate was incubated overnight at 65°C in an oven without rotating.
  • the eluted sample was transferred to a fresh tube and 300 pi of TE buffer was added to dilute the SDS. Three pi of RNase A (30 mg/ml) was added to the tube, and the mixture was incubated at 37°C for 30 minutes. Following incubation, 3pl of 1M CaCE and 7 pi of 20 mg/ml Proteinase K was added, and the tube was incubated again for 1.5 hours at 55°C.
  • MaXtract High Density 2 ml gel tubes (Qiagen) are precipitated by centrifuging them at full speed for 30 seconds at RT. Six hundred pi of phenol/chloroform/isoamyl alcohol was added to each proteinase K reaction, and about 1.2 ml of the mixture was transferred to the MaXtract tubes. Tubes were spun at 16,000 g for 5 minutes at RT. [00296] The aqueous phase was transferred to two clean DNA LoBind tubes (300 m ⁇ in each tube), and 1 m ⁇ glycogen, 30 m ⁇ of 3M sodium acetate, and 900 m ⁇ ethanol was added. The mixture was precipitated for 1 hour at -80°C.
  • the tubes were spun down at maximum speed for 30 minutes at 4°C, and the ethanol was removed.
  • the pellets were washed with 1 ml of 75% ethanol by spinning tubes down at maximum speed for 5 minutes at 4°C. Remnants of ethanol were removed, and the pellets were dried for 5 minutes at RT. Thirty m ⁇ of fTO was added to the pellet and allowed to stand for 5 minutes. The pellet mixture was vortexed briefly, and spun down to collect the DNA.
  • ChlA-PETs were immobilized on Streptavidin beads using the following steps.
  • 2X B&W Buffer (40 ml) was prepared as follows for coupling of nucleic acids: 400 m ⁇ 1M Tris- HC1 pH 8.0 (10 mM final), 80 m ⁇ 1M EDTA (1 mM final), 16 ml 5 M NaCl (2 M final), and 23.52 ml dH 2 0.
  • IX B&W Buffer (40 ml total) was prepared by adding 20 ml dfTO to 20 ml of the 2X B&W Buffer.
  • MyOne Streptavidin Dynabeads M-280 were allowed to come to room temperature for 30 minutes, and 30 m ⁇ of beads were transferred to a new 1.5 ml tube. Beads were washed with 150 m ⁇ of 2X B&W Buffer twice. Beads were resuspended in 100 m ⁇ of iBlock buffer (Applied Biosystems), and mixed. The mixture was incubated at RT for 45 minutes on a rotator.
  • I-BLOCK Reagent was prepared to contain: 0.2% I-Block reagent (0.2 g), IX PBS or IX TBS (10 ml 10X PBS or 10X TBS), 0.05% Tween-20 (50 pl), and H 2 0 to 100 ml. 10X PBS and I-BLOCK reagent was added to H 2 0, and the mixture was microwaved for 40 seconds (not allowed to boil), then stirred. Tween-20 was added after the solution was cooled. The solution remained opaque, but particles were dissolved. The solution was cooled to RT for use.
  • the beads were washed 5 times with 500 m ⁇ of 2xSSC/0.5% SDS buffer (30 seconds each time) followed by 2 washes with 500 ml of IX B&W Buffer and incubating each after wash for 5 minutes at RT with rotation.
  • the beads were washed once with 100 m ⁇ elution buffer (EB) from a Qiagen Kit by resuspending beads gently and putting the tube on a magnet. The supernatant was removed from the beads, and they were resuspended in 30 m ⁇ of EB.
  • EB elution buffer
  • a paired end sequencing library was constructed on beads using the following protocol. Ten m ⁇ of beads are tested by PCR with 10 cycles of amplification. The 50 m ⁇ of the PCR mixture contained: 10 m ⁇ of bead DNA, 15 m ⁇ NPM mix (from Illumina Nextera kit), 5 m ⁇ of PPC PCR primer, 5 m ⁇ of Index Primer 1 (i7), 5 m ⁇ of Index Primer 2 (i5), and 10 m ⁇ of H 2 0. PCR was performed using the following cycle conditions: denaturing the DNA at 72°C for 3 minutes, then 10-12 cycles of 98°C for 10 seconds, 63°C for 30 seconds, and 72°C for 50 seconds, and a final extension of 72°C for 5 minutes. The number of cycles was adjusted to obtain about 300 ng of DNA total with four 25 m ⁇ reactions. The PCR product was held at 4°C for an indefinite amount of time.
  • PCR product was cleaned-up using AMPure beads. Beads were allowed to come to RT for 30 minutes before using. Fifty m ⁇ of the PCR reaction was transferred to a new Low- Bind Tube and (l.8x volume) 90 m ⁇ of AMPure beads was added. The mixture was pipetted well and incubated at RT for 5 minutes. A magnet was used for 3 minutes to collect beads and remove the supernatant. Three hundred m ⁇ of freshly prepared 80% ethanol was added to the beads on the magnet, and the ethanol was carefully dicarded. The wash was repeated, and then all ethanol was removed. The beads were dried on the magnet rack for 10 minutes. Ten m ⁇ EB was added to the beads, mixed well, and incubated for 5 minutes at RT. The eluate was collected, and 1 m ⁇ of eluate was used for Qubit and Bioanalyzer.
  • the library was cloned to verify complexity using the following protocol.
  • One m ⁇ of the library was diluted at 1:10.
  • the PCR reaction mixture (total volume: 50 m ⁇ ) contained the following: 10 m ⁇ of 5X GoTaq buffer, 1 m ⁇ of 10 mM dNTP, 5 m ⁇ of 10 mM primer mix, 0.25 m ⁇ of GoTaq polymerase, 1 m ⁇ of diluted template DNA, and 32.75 m ⁇ of H 2 0.
  • PCR was performed using the following cycle conditions: denaturing the DNA at 95°C for 2 minutes and 20 cycles at the following conditions: 95°C for 60 seconds, 50°C for 60 seconds, and 72°C for 30 seconds with a final extension at 72°C for 5 minutes.
  • the PCR product was held at 4°C for an indefinite amount of time.
  • the PCR product was ligated with the pGEM® T-Easy vector (Promega) protocol. Five m ⁇ of 2X T4 Quick ligase buffer, Im ⁇ of pGEM® T-Easy vector, 1 m ⁇ of T4 ligase, 1 m ⁇ of PCR product, and 2 m ⁇ of H 2 0 were combined to a total volume of 10 m ⁇ . The product was incubated for 1 hour at RT and 2 m ⁇ was used to transform Stellar competent cells. Two hundred m ⁇ of 500 m ⁇ of cells were plated in SOC media. The next day, 20 colonies were selected for Sanger sequencing using a T7 promoter primer. 60% clones had a full adapter, and 15% had a partial adapter
  • Protein G Dynabeads for 10 samples were from Invitrogen Dynal, Cat# 10003D.
  • Block solution 50 ml
  • ddH20 0.25g BSA dissolved in 50 ml of ddH20 (0.5% BSA, w/v)
  • Lysis buffer 1 (LB1) (500 ml) contained 25 ml of 1 M Hepes-KOH, pH 7.5; 14 ml of 5 M NaCl; 1 ml of 0.5 M EDTA, pH 8.0; 50 ml of 100% Glycerol solution; 25 ml of 10% NP- 40; and 12.5 ml of 10% Triton X-100. The pH was adjusted to 7.5. The buffer was sterile - filtered, and stored at 4°C. The pH was re-checked immediately prior to use.
  • Lysis buffer 2 (LB2) (1000 ml) contained 10 ml of 1 M Tis-HCL, pH 8.0; 40 ml of 5 M NaCl; 2 ml of 0.5 M EDTA, pH 8.0; and 2 ml of 0.5 M EGTA, pH 8.0. The pH was adjusted to 8.0. The buffer was sterile-filtered, and stored at 4 °C. The pH was re-checked immediately prior to use.
  • Sonication buffer 500 ml contained 25 ml of 1 M Hepes-KOH, pH 7.5; 14 ml of 5M NaCl; 1 ml of 0.5 M EDTA, pH 8.0; 50 ml of 10% Triton X-100; 10 ml of 5% Na-deoxycholate; and 5 ml of 10% SDS.
  • the buffer was sterile-filtered, and stored at 4 °C. The pH was re-checked immediately prior to use.
  • High-salt sonication buffer (500ml) contained 25 ml of 1 M Hepes- KOH, pH 7.5; 35 ml of 5M NaCl; 1 ml of 0.5 M EDTA, pH 8.0; 50 ml of 10% Triton X-100; 10 ml of 5% Na-deoxycholate; and 5 ml of 10% SDS.
  • the buffer was sterile-filtered, and stored at 4 °C. The pH was re-checked immediately prior to use.
  • LiCl wash buffer (500 ml) contained 10 ml of 1 M Tris-HCL, pH 8.0; 1 ml of 0.5M EDTA, pH 8.0; 125 ml of 1M LiCl solution; 25 ml of 10% NP-40; and 50 ml of 5% Na- deoxycholate. The pH was adjusted to 8.0. The buffer was sterile-filtered, and stored at 4 °C. The pH was re-checked immediately prior to use.
  • Elution buffer used to quantify the amount of ChIP DNA contained 25 ml of 1M Tris-HCL, pH 8.0; 10 ml of 0.5M EDTA, pH 8.0; 50 ml of 10% SDS; and 415 ml of ddH 2 0. The pH was adjusted to 8.0. The buffer was sterile-filtered, and stored at 4 °C. The pH was re checked immediately prior to use.
  • ChIA-RET Wash Buffer (50 ml) contained 500 pl of 1 M Tris-HCl, pH 8.0 (final 10 mM); 100 pl of 0.5 M EDTA, pH 8.0 (final 1 mM); 5 ml of 5 M NaCl (final 500 mM); and 44.4 ml of dhkO.
  • HiChIP was used to analyze chromatin interactions and conformation. HiChIP requires fewer cells than ChlA-PET.
  • the resuspension was incubated at 62°C for 10 minutes, and then 285 pL of H 2 0 and 50 pL of 10% Triton X-100 were added to quench the SDS. The resuspension was mixed well, and incubated at 37°C for 15 minutes. Fifty pL of 10X NEB Buffer 2 and 375 U of Mbol restriction enzyme (NEB, R0147) was added to the mixture to digest chromatin for 2 hours at 37°C with rotation. For lower starting material, less restriction enzyme is used: 15 pL was used for 10-15 million cells, 8 pL for 5 million cells, and 4 pL for 1 million cells. Heat (62°C for 20 minutes) was used to inactivate Mbol.
  • Ligation Master Mix contains 150 pL of
  • the pellet was brought up to 1000 pL in Nuclear Lysis Buffer.
  • the sample was transferred to a Covaris millitube, and the DNA was sheared using a Covaris ® E220Evolution TM with the manufacturer recommended parameters.
  • Each tube (15 million cells) was sonicated for 4 minutes under the following conditions: Fill Level 5; Duty Cycle 5%; PIP 140; and Cycles/Burst 200.
  • the sample was clarified for 15 minutes at l6,l00g at 4°C.
  • the sample is split into 2 tubes of about 400 pL each and 750 pL of ChIP Dilution Buffer is added.
  • the sample is diluted 1:2 in ChIP Dilution Buffer to achieve an SDS concentration of 0.33%.
  • 60 pL of Protein G beads were washed for every 10 million cells in ChIP Dilution Buffer. Amounts of beads (for preclearing and capture) and antibodies were adjusted linearly for different amounts of cell starting material. Protein G beads were resuspended in 50 pL of Dilution Buffer per tube (100 pL per HiChIP).
  • the sample was rotated at 4°C for 1 hour.
  • the samples were put on a magnet, and the supernatant was transferred into new tubes.
  • 7.5 pg of antibody was added for every 10 million cells, and the mixture was incubated at 4°C overnight with rotation.
  • Another 60 pL of Protein G beads for every 10 million cells in ChIP Dilution Buffer was added.
  • Protein G beads were resuspended in 50 pL of Dilution Buffer (100 pL per HiChIP), added to the sample, and rotated at 4°C for 2 hours.
  • the beads were washed three times each with Low Salt Wash Buffer, High Salt Wash Buffer, and LiCl Wash Buffer. Washing was performed at room temperature on a magnet by adding 500 pL of a wash buffer, swishing the beads back and forth twice by moving the sample relative to the magnet, and then removing the supernatant
  • ChIP sample beads were resuspended in 100 pL of fresh DNA Elution Buffer. The sample beads were incubated at RT for 10 minutes with rotation, followed by 3 minutes at 37°C with shaking. ChIP samples were placed on a magnet, and the supernatant was removed to a fresh tube. Another 100 pL of DNA Elution Buffer was added to ChIP samples and incubations were repeated. ChIP sample supernatants were removed again and transferred to a new tube. There was about 200 pL of ChIP sample. Ten pL of Proteinase K (20 mg/ml) was added to each sample and incubated at 55°C for 45 minutes with shaking.
  • the beads were washed in 100 pL of IX (diluted from 2X) TD Buffer.
  • the beads were resuspended in 25 pL of 2X TD Buffer, 2.5 pL of Tn5 for each 50 ng of post-ChIP DNA, and water to a volume of 50 pL.
  • the Tn5 had a maximum amount of 4 pL. For example, for 25 ng of DNA transpose, 1.25 pL of Tn5 was added, while for 125 ng of DNA transpose, 4 pL of Tn5 was used. Using the correct amount of Tn5 resulted in proper size distribution. An over-transposed sample had shorter fragments and exhibited lower alignment rates (when the junction was close to a fragment end). An undertransposed sample has fragments that are too large to cluster properly on an Illumina sequencer. The library was amplified in 5 cycles and had enough complexity to be sequenced deeply and achieve proper size distribution regardless of the level of transposition of the library.
  • the beads were incubated at 55°C with interval shaking for 10 minutes. Samples were placed on a magnet, and the supernatant was removed. Fifty mM EDTA was added to samples and incubated at 50°C for 30 minutes. The samples were then quickly placed on a magnet, and the supernatant was removed. The samples were washed twice with 50 mM EDTA at 50°C for 3 minutes, then were removed quickly from the magnet. Samples were washed twice in Tween Wash Buffer for 2 minutes at 55°C, then were removed quickly from the magnet. The samples were washed with 10 mM Tris-HCl, pH8.0.
  • the beads were resuspended in 50 pL of PCR master mix (use Nextera XT DNA library preparation kit from Illumina, #15028212 with dual-index adapters # 15055289). PCR was performed using the following program. The cycle number was estimated using one of two methods: (1) A first run of 5 cycles (72°C for 5 minutes, 98°C for 1 minute, 98°C for 15 seconds, 63°C for 30 seconds, 72°C for 1 minute) was performed on a regular PCR and then the product was removed from the beads. Then, 0.25X SYBR green was added, and the sample was ran on a qPCR.
  • Libraries were placed on a magnet and eluted into new tubes.
  • the libraries were purified using a kit form Zymo Research and eluted into 10 pL of water. A two-sided size selection was performed with AMPure XP beads. After PCR, the libraries were placed on a magnet and eluted into new tubes. Then, 25 pL of AMPure XP beads were added, and the supernatant was kept to capture fragments less than 700 bp. The supernatant was transferred to a new tube, and 15 pL of fresh beads were added to capture fragments greater than 300 bp. A final elution was performed from the Ampure XP beads into 10 pL of water. The library quality was verified using a Bioanalyzer.
  • Hi-C Lysis Buffer (10 mL) contains 100 pL of 1 M Tris-HCl pH 8.0; 20 pL of 5 M NaCl; 200 pL of 10% NP-40; 200 pL of 50X protease inhibitors; and 9.68 mL of water.
  • Nuclear Lysis Buffer (10 mL) contains 500 pL of 1 M Tris-HCl pH 7.5; 200 pL of 0.5 M EDTA; 1 mL of 10% SDS; 200 pL of 50X Protease Inhibitor; and 8.3mL of water.
  • ChIP Dilution Buffer (10 mL) contains 10 pL of 10% SDS; 1.1 mL of 10% Triton X-100; 24 pL of 500 mM EDTA; 167 pL of 1 M Tris pH 7.5; 334 pL of 5 M NaCl; and 8.36 5mL of water.
  • Low Salt Wash Buffer (lOmL) contains 100 pL of 10% SDS; 1 mL of 10% Triton X-100; 40 pL of 0.5 M EDTA; 200 pL of 1 M Tris-HCl pH 7.5; 300 pL of 5 M NaCl; and 8.36 mL of water.
  • High Salt Wash Buffer (10 mL) contains 100 pL of 10% SDS; 1 mL of 10% Triton X-100; 40 pL of 0.5 M EDTA; 200 pL of 1 M Tris-HCl pH 7.5; 1 mL of 5 M NaCl; and 7.66 mL of water.
  • DNA Elution Buffer contains 250 pL of fresh 1 M NaHC03; 500 pL of 10% SDS; and 4.25 mL of water.
  • Tween Wash Buffer (50 mL) contains 250 pL of 1 M Tris-HCl pH 7.5; 50 pL of 0.5 M EDTA; 10 mL of 5 M NaCl; 250 pL of 10% Tween-20; and 39.45 mL of water.
  • 2X Biotin Binding Buffer (10 mL) contains 100 pL 1 M Tris-HCl pH 7.5; 20 pL of 0.5 M; 4 mL of 5 M NaCl; and 5.88 mL of water.
  • 2X TD Buffer (lmL) contains 20 pL of 1 M Tris- HCl pH 7.5; 10 pL of 1 M M MgCL; 200 pL of 100% Dimethylformamide; and 770 pL of water.
  • Bioactive compounds were also administered to hepatocytes.
  • lOOOx stock of the bioactive compounds in 1 ml DMSO 0.1 ml of IO,OOOC stock was combined with 0.9 ml DMSO.
  • RNAiMAX Reagent ThermoFisher Cat#l3778030
  • modified maintenance medium for an additional 24 hours.
  • RLT Buffer for RNA extraction Qiagen RNeasy 96 QIAcube HT Kit Cat#74l7l
  • siRNAs were obtained from Dharmacon and are a pool of four siRNA duplex all designed to target distinct sites whitin the specific gene of interest (“SMARTpool”). The following siRNAs were used for each target: D-001206-13-05 (non- targeting); M-003145-02- 0005 (JAK1); and M-003146-02-0005 (JAK2).
  • mice C57BL/6J strain
  • 3 male and 3 female were administered with a candidate compound once daily via oral gavage for four consecutive days.
  • Mice were sacrificed 4 hours post-last dose on the fourth day.
  • Organs including liver, spleen, kidney, adipose, plasma were collected.
  • Mouse liver tissues were pulverized in liquid nitrogen and aliquoted into small microtubes.
  • TRIzol Invitrogen Cat# 15596026
  • the TRIzol solution containing the disrupted tissue was then centrifuged and the supernatant phase was collected.
  • Total RNA was extracted from the supernatant using Qiagen RNA Extraction Kit (Qiagen Cat#74l82) and the target mRNA levels were analyzed using qRT-PCR.
  • RNA-seq was performed to determine the effects of the compounds on the expression of PAH in hepatocytes. Fold change was calculated by dividing the level of expression in the cell system that had been perturbed by the level of expression in an unperturbed system. Changes in expression having a p-value ⁇ 0.05 were considered significant.
  • Compounds used to perturb the signaling centers of hepatocytes include at least one compound listed in Table 1. In the table, compounds are listed with their ID, target, pathway, and pharmaceutical action. Most compounds chosen as perturbation signals are known in the art to modulate at least one canonical cellular pathway. Some compounds were selected from compounds that failed in Phase III clinical evaluation due to lack of efficacy.
  • RNA-seq results for compounds that significantly increase expression of PAH are shown in Table 2.
  • tyrosine kinase inhibitors were among the identified compounds that upregulate PAH expression (including R788, Dasatinib, Bosutinib, CP-673451, Merestinib, Pacritinib, Cediranib, GZD824, Amuvatinib, and Ceritinib), suggesting that PAH expression may be strongly regulated via one or more tyrosine kinase-mediated signaling pathways.
  • R788 (fostamatinib disodium hexahydrate) is a known Syk inhibitor. Dasatinib inhibits BCR/ABL and the Src kinase family.
  • Bosutinib is a dual Src/Abl inhibitor.
  • CP-673451 is a selective inhibitor of PDGFRa/b.
  • Merestinib selectively inhibits c-MET and several other receptor tyrosine kinases such as MST1R, FLT3, AXL, MERTK, TEK, ROS1, NTRK1/2/3, and DDR1/2.
  • Pacritinib selectively inhibits JAK2 and FLT3.
  • PND- 1186 is a reversible and selective FAK inhibitor.
  • Cediranib is a potent inhibitor of vascular endothelial growth factor (VEGF) receptor tyrosine kinases.
  • VEGF vascular endothelial growth factor
  • GZD824 is known to inhibit a broad spectrum of Bcr/Abl tyrosine kinase mutants.
  • Amuvatinib is a multi-targeted tyrosine kinase inhibitor with potent activity against mutant c-Met, c-Kit, PDGFRa, Flt3, and c-Ret.
  • Ceritinib is a potent inhibitor against ALK.
  • These tyrosine kinases are associated with the JAK/STAT pathway and MAPK pathway. Pathways associated with these tyrosine kinases may be manipulated to increase PAH expression.
  • ChIP-seq was used to determine the genomic position and composition of signaling centers.
  • ChIP-seq targets for primary human hepatocytes
  • the insulated neighborhood that contains the PAH gene was identified to be on chromosome 12 with a size of approximately 668 kb. 14 signaling centers were found within the insulated neighborhood.
  • the chromatin marks or chromatin- associated proteins, transcription factors, and signaling proteins/or factors that were found in the insulated neighborhood are presented in Table 4.
  • the ChIP-seq profile indicates that the insulated neighborhood containing PAH may be regulated by JAK/STAT signaling, TGF signaling, WNT signaling, nuclear receptor signaling, BMP signaling, NF-kB signaling, MAPK signaling, and/or Hippo signaling pathways.
  • STAT1 and STAT3 both associated with the JAK/STAT pathway, were observed to bind to the signaling centers within the insulated neighborhood.
  • the insulated neighborhood is also enriched with NF-kB and AP- 1 transcription factors, which are associated with tyrosine kinase/MAPK pathway. Targeting these pathways can upregulate PAH expression.
  • Example 5 Determining genome architecture in hepatocytes
  • Hl-ChIP was performed as described in Example 1 to decipher genome architecture.
  • ChIA-RET for SMC1 structural protein was used for the same purpose.
  • the insulated neighborhood containing the PAH gene was identified to be on chromosome 12 at position 102,882,556 to 103,550,727 (human CRCH38/hg38 genome assembly) with a size of approximately 668 kb.
  • the insulated neighborhood contains PAH and 2 other genes, which are positioned in the following order with respect to PAH: (PAH), ASCL1 and C120RF42.
  • qRT-PCR was performed on samples of primary human hepatocytes from two donors to validate the identified compounds for disease associated targets such as PAH. Cryopreserved hepatocytes were thawed and cultured in plating media for 16 hours, and transferred to complete maintenance media for 2 hours. Cells were then either maintained in the complete maintenance media or transferred to modified maintenance media for another 2 hours, and a compound was added. Compounds were added at concentrations ranging from 0.01 mM to 50 mM. Cells were maintained in respective media for 16 hours prior to qRT-PCR analysis. qRT-PCR was performed as described in Example 1. Gene expression level was determined as the relative mRNA level of the gene to an internal control, GAPDH.
  • Echinomycin is an inhibitor of hypoxia- inducible factor- 1 DNA- binding activity.
  • Dasatinib and CP-673451 are both tyrosine kinase inhibitors.
  • Relative PAH mRNA expression in human hepatocytes treated with Echinomycin, Dasatinib, and CP-673451 are presented in Table 5. The data demonstrated that the compounds caused a dose-dependent increase in PAH levels. The effect of the compounds on PAH expression was reproducible in multiple human donor and culture conditions. This experiment confirms that Dasatinib, CP- 67345! and Echinomycin are capable of increasing expression of PAH in primary human hepatocytes.
  • the aim of this example was to confirm the role of the identified signaling pathways (e.g., JAK/STAT) that are controlling PAH expression.
  • the end component of the JAK/STAT pathway was targeted via siRNA-mediated knock-down.
  • Primary human hepatocytes were reverse transfected with 10 nM siRNA targeting JAK1, JAK2 or both.
  • Levels of the target mRNA were measured via qRT-PCR and normalized to a non-targeting siRNA control to evaluate the known-down efficiency. PAH mRNA levels were then assayed via qRT-PCR to assess the effect of JAK1 or JAK2 knock-down on PAH expression.
  • results of JAK1/JAK2 knock-down and the corresponding effect on PAH mRNA are shown in FIG. 7.
  • Plotted values are the relative fold changes of target mRNA level with repect to the non-targeting control.
  • JAK1 and JAK2 were specifically knocked-down by siRNA at an efficiency of ⁇ 95% and 75%, respectively. While knocking down of only JAK1 or JAK2 led to a slight increase in PAH expression, JAK1 + JAK2 knock-down resulted in a notable (-40%) increase in PAH mRNA, confirming the role of JAK/STAT pathway in PAH expression regulation.
  • Example 8 Compound testing in mouse hepatocvtes
  • ATRA all-trans retinoic acid
  • WYE- 125132 WYE- 132
  • WAY600 is highly potent mTOR inhibitors.
  • Ibrutinib is a Syk pathway inhibitor that inhibits Tec family kinases (e.g., BTK). These compounds were chosed for further characterization in the in vivo experiments.
  • Example 9 In vivo compound testing in mice
  • candidate compounds are evaluated in kidney cells to confirm their efficacy. Changes in target gene expression in kidney cells are analyzed with qRT-PCR. Results are compared with that from the primary hepatocytes. Compounds that show consistent induction of PAH expression are selected for further analysis.
  • Candidate compounds are evaluated in patient derived induced pluripotent stem (iPS)-hepatoblast cells to confirm their efficacy. Selected patients have a deficiency in PAH. Changes in target gene expression in iPS-hepatoblast cells are analyzed with qRT-PCR. Results are used to confirm if the pathway is similarly functional in patient cells and if the compound has the same impact.
  • iPS patient derived induced pluripotent stem
  • Example 12 Compound testing in a mouse model
  • Candidate compounds are evaluated in a mouse model of phenylketonuria for in vivo activity and safety.
  • Example 13 Phenylalanine hydroxylase (PAH) enzyme assay
  • a biochemical reaction assay in a cell free system was developed to monitor the rate and amount of tyrosine product made by PAH as a function of enzyme concentration.
  • PAH enzyme was collected from hepatocyte lysate and mixed with a phenylalanine substrate, tetrahydrobiopterin (BH4) coenzyme, catalase, and a ferrous ammonium sulfate non-heme Fe(II) cofactor.
  • the reaction is shown in FIG. 10A, 10B, and 10C; and a schematic of the assay is shown in FIG. 11.
  • the samples were processed via Thin Layer Chromatography to resolve the substrate phenylalanine from the tyrosine product.
  • the chromatography assay used Silica Gel 60 plates.
  • the mobile phase was Isopropanol: Acetone: Ammonium Hydroxide: in a 25:25:13 ratio; and detection was via Ninhydrin Spray. This allowed separation of the phenylalanine from the tyrosine.
  • the tyrosine product was detected via fluorometric assays using a 274 nm wavelength light source.
  • reaction rates and the amount of tyrosine generated increase linearly with the amount of lysate (amount of PAH enzyme) used in the reaction.
  • PAH enzyme levels in the mouse liver samples from Example 9 were next assessed using the PAH enzyme assay. As shown in FIG. 13, the treatment induced increase in the liver PAH mRNA upregulation correlates with an increase in the PAH enzyme activity.
  • Example 15 PAH and GCH1 in vitro and in vivo compound testing [00363] Next, additional compounds were tested in vitro in two different primary human hepatocyte cell lines and one primary mouse hepatocyte cell line for PAH and GCH1 mRNA expression upregulation. Table 11 lists the compounds used in the study.
  • Table 12 shows the PAH and GCH1 mRNA results for the HH4178 human primary hepatocytes
  • Table 14 shows the PAH and GCH1 mRNA results for the mouse primary hepatocytes.
  • Table 15 shows the PAH and GCH1 mRNA results for the in vivo samples.
  • Example 16 Perturbogen upregulation of PAH and GCH1 in primary human hepatocvtes
  • Example 17 Compounds tested in mice in vivo for upregulation of PAH and GCH1 expression.
  • mice were tested per group.
  • Select compounds (Table 17) were administered by gavage daily for 4 consequtive days. Animals were sacraficed 6 h after admini station of final dose. Table 17 shows compund names, dosing and signaling pathway that is the target of each compound tested.
  • FIG. 16A shows the fold change in PAH mRNA in treated mice relative to mice administered vehicle alone for select compounds.
  • FIG. 16B shows the fold change in GCH1 mRNA in treated mice relative to mice administered vehicle alone for select compounds.
  • FIG 17 shows the relative increase in PAH protein in livers of treated mice.
  • Table 18 summarizes the results and shows fold change in mRNA for PAH and GCH1 and PAH protein.
  • Mouse liver lysates were prepared for western blotting using a lysis buffer comprising M- PER/HALT. Lysis buffer was prepared by mixing 10 ml of M-PER and 100 uL HALT
  • Protease/phosphatase cocktail HALT cocktail 100 uL M-PER/HALT mix was added to each tissue powder sample. The samples were mixed well and then placed at 4°C for 30 min. Tubes were spun at 4°C for 10 min at 5 K rpm. Supernatant was removed ( ⁇ l00uL) and transfered to ice. Protein was quantified, mixed appropriate amount with Lamelli’s buffer, heated at 95°C for 10 min, and loaded on a polyacrylamide gel.
  • mice were treated with Dasatinib by oral gavage for 4 days and mice were sacraficed on the fouth day of treatment (FIG. 18 A).
  • Liver lysates were prepared for in vitro analysis of PAH activity as described above.
  • the relative increase in tyrosine in the livers from mice treated with Dasatinib versus livers from mice treated with vehicle alone is shown in FIG. 18B.
  • Three additional compounds, XL228, SIS3 and PF-562271 were tested in mice following the dosing schedule as shown in FIG. 19.
  • PAH activity (FIG. 20) and PAH specific activity (FIG. 21) was increased in mice treated with PF-562271 and XL228.
  • Example 19 Increased amount of PAH protein and GCH1 protein increases activity in vitro for select PAH mutants.
  • Mutants of PAH were cloned into plasmids in frame with a Renilla luciferase reporter gene and transfected into 293XT cells that do not express endogenous PAH.
  • the human and mouse PAH constructs are l356bp and l359bp in length, respectively and are based on the Consensus Coding Sequences (CCDS) attained from NCBI. All constructs include a minimal Kozak sequence for optimal expression (GCCGCCACC) located directly upstream of the start codon (ATG) of PAH. Additionally, all PAH sequences include a C-terminal FLAG tag
  • pCDNA3.l was modified by incorporating a viral self-cleaving peptide P2A cleavage sequence(GCAACAAACTTCTCTCTGCTGAAACAAGCCGGAGATGTCGAAGAGAATCC TGGACCG), which can induce the cleaving of the recombinant protein in cells followed by Renilla Luciferase for quantifying the transfection efficiency of the constructs.
  • P2A and Renilla Luciferase are located downstream of the Mlul restriction site in the pCDN3.l vector. All PAH constructs were cloned upstream of the P2A-Renilla Luciferase. The bicistronic expression of both PAH and Renilla luciferase was achieved using a minimal CMV promoter in the pCDNA3.l vector.
  • Table 19 shows the PAH activity as percent WT activity measured for different PAH constructs harboring different mutations of PAH.
  • FIG. 22 shows the relative protein expression of PAH in lystaes transfected with the different constructs harboroing PAH mutations.
  • FIG. 23 shows increased amonts of cell lysate from cells transfected with a construct harboring PAH 198R243Q lead to increases in PAH activity.
  • GCH1 is also cloned into plasmids and transfected into cells. PAH activity in vitro is increased in cells overexpressing GCH1 or in cells where GCH1 expression is increased by treatment of cells with select compounds as described above.
  • the disclosure includes embodiments in which exactly one member of the group is present in, employed in, or otherwise relevant to a given product or process.
  • the disclosure includes embodiments in which more than one, or the entire group members are present in, employed in, or otherwise relevant to a given product or process.
  • the term“comprising” is intended to be open and permits but does not require the inclusion of additional elements or steps. When the term“comprising” is used herein, the term“consisting of’ is thus also encompassed and disclosed. Where ranges are given, endpoints are included.
  • compositions of the disclosure e.g., any antibiotic, therapeutic or active ingredient; any method of production; any method of use; etc.
  • any particular embodiment of the compositions of the disclosure can be excluded from any one or more claims, for any reason, whether or not related to the existence of prior art. It is to be understood that the words which have been used are words of description rather than limitation, and that changes may be made within the purview of the appended claims without departing from the true scope and spirit of the disclosure in its broader aspects.

Abstract

The present invention provides methods and compositions for the treating a patient with phenylalanine hydroxylase (PAH) deficiency, such as phenylketonuria. Methods and compositions are also provided for modulating the PAH gene by altering gene signaling networks.

Description

COMPOSITIONS AND METHODS FOR TREATING PHENYLKETONURIA
CROSS REFERENCE TO RELATED APPLICATIONS
[0001] The present application claims priority to, and the benefits of U.S. Provisional Patent Application Serial No. 62/653,748, filed April 6, 2018, and U.S. Provisional Patent Application Serial No. 62/789,477, filed January 7, 2019, both of which are incorporated herein by reference in their entireties.
FIELD OF THE INVENTION
[0002] The present invention provides compositions and methods for the treatment of phenylalanine hydroxylase deficiency, such as phenylketonuria, in humans.
BACKGROUND
[0003] Phenylketonuria (PKU) is caused by a defect in the Phenylalanine Hydroxylase (PAH) gene and due to a buildup of phenylalanine in the blood. Phenylalanine hydroxylase is an enzyme that catalyzes the hydroxylation of phenylalanine to generate tyrosine. Mutations in the PAH gene reduce the activity of phenylalanine hydroxylase and prevent the breakdown of phenylalanine. As a result, phenylalanine builds up to toxic levels in the blood and other tissues, causing brain damage and other serious medical problems.
[0004] The incidence of phenylketonuria was estimated to be approximately 1 in 15,000 in the United States. The signs and symptoms of PKU vary from mild to severe.
Symptoms include behavioral problems, seizures, mental and growth retardation. The most severe form of this disorder is known as classic PKU, which occurs when phenylalanine hydroxylase activity is severely reduced or absent. Several milder versions of PKU and non-PKU hyperphenylalaninemia are often due to partial deficiency of the PAH enzyme.
[0005] Managing PKU is difficult, complex, and life-long. Standard of care usually includes restriction of dietary phenylalanine, symptom management, and the use of KUVAN® (sapropterin dihydrochloride). KUVAN® is the only prescription medication for PKU. KUVAN® works by adding more tetrahydrobiopterin (BH4), an essential cofactor of PAH, and stimulating the PAH enzyme to process phenylalanine in people with PKU. However, only 30 percent to 50 percent of PKU patients respond to KUVAN® and the response can be inconsistent or unpredictable. Consequently, there is an unmet medical need to provide effective treatment for people with PKU.
[0006] The present invention provides novel targets, compositions and treatment methods for PKU.
SUMMARY
[0007] The present invention discloses the mapping and identification of gene signaling network(s) associated with the Phenylalanine Hydroxylase (PAH) gene, which has been linked to diseases of phenylalanine hydroxylase deficiency such as phenylketonuria. By perturbing the components of the gene signaling network(s), the inventors have identified novel targets, compounds and/or methods that could be utilized to modulate PAH expression. Such methods and compositions may be used to develop various therapies for a PAH-related disorder (e.g., phenylketonuria) to prevent and/or alleviate the symptoms of such a disease.
[0008] Accordingly, provided herein is a method of treating a subject with phenylalanine hydroxylase deficiency by administering to the subject an effective amount of a compound capable of modulating the expression of the PAH gene. Such compound may be a small molecule, a polypeptide, an antibody, a hybridizing oligonucleotide, and a genome editing agent.
[0009] In some embodiments, the compound administered to the subject may include an inhibitor of the JAK/STAT pathway. Such compound may include at least one of Ruxolitinib, Oclacitinib, Baricitinib, Filgotinib, Gandotinib, Lestaurtinib, PF-04965842, Upadacitinib, Cucurbitacin I, CHZ868, Fedratinib, AC430, AT9283, ati-5000l and ati- 50002, AZ 960, AZD1480, BMS-911543, CEP-33779, Cerdulatinib (PRT062070, PRT2070), Curcumol, Decernotinib (VX-509), Fedratinib (SAR302503, TG101348), FLLL32, FM-381, GLPG0634 analogue, Go6976, JANEX-l (WHI-P131), Momelotinib (CYT387), NVP-BSK805, Pacritinib (SB1518), Peficitinib (ASP015K, JNJ-54781532), PF-06651600, PF-06700841, R256 (AZD0449), Solcitinib (GSK2586184 or GLPG0778), S-Ruxolitinib (INCB018424), TG101209, Tofacitinib (CP-690550), WHI-P154, WP1066, XL019, ZM 39923 HC1, or a derivative or an analog thereof.
[0010] In some embodiments, the compound administered to the subject may include an inhibitor of the Tyrosine kinase/MAPK pathway. Such compound may include at least one of Amuvatinib, Bosutinib, Cediranib, Ceritinib, CP-673451, Dasatinib, GZD824 Dimesylate, Merestinib, R788 (fostamatinib disodium hexahydrate), or a derivative or an analog thereof.
[0010] In some embodiments, the compound administered to the subject may include 17- AAG (Tanespimycin), Amlodipine Besylate, ATRA (all-trans retinoic acid), Chloroquine phosphate, Deoxycorticosterone, Darapladib, Echinomycin, Enzastaurin, Epinephrine, EVP-6124 (hydrochloride) (encenicline), EW-7197, FRAX597, Ibrutinib, Perphenazine, Phenformin, PND-1186, Rifampicin, Semagacestat, Thalidomide, WAY600, WYE-125132 (WYE-132), Zibotentan, Activin, Anti mullerian hormone, GDF10 (BMP3b), IGF-l,
Nodal, PDGF, TNF-a, Wnt3a, or a derivative or an analog thereof.
[0011] In alternative embodiments, the compound administered to the subject may include one or more RNAi agents against a signaling molecule identified to regulate PAH expression. In some embodiments, the compound includes one or more small interfering RNA (siRNA) targeting one or more genes selected from the group consisting of JAK1, JAK2, PDGFRA, PDGFRB, SRC and ABL.
[0012] In any one of the embodiments disclosed above, the compound increases the expression of the PAH gene in the subject. In some embodiments, the expression of the PAH gene is increased by at least about 40% over baseline or over levels measured following administration of a control. In some embodiments, the expression of the PAH gene is increased in the liver of the subject. The subject may have at least one mutated allele of the PAH gene. The mutation may occur within or near the PAH gene. The mutation may decrease the activity of phenylalanine hydroxylases or reduce the expression of PAH in the subject as compared to activity or expression associated with a canonical wild-type sequence.
[0013] In any one of the embodiments disclosed above, the phenylalanine hydroxylase deficiency is mild hyperphenylalaninemia. In some embodiments, the phenylalanine hydroxylase deficiency is mild phenylketonuria (PKU). In other embodiments, the phenylalanine hydroxylase deficiency is classic phenylketonuria (PKU).
[0014] Also provided herein is a method of modulating the expression of a PAH gene in a cell by introducing to the cell an effective amount of a compound capable of altering one or more signaling molecules associated with the regulatory sequence regions (RSRs) or portion thereof of the PAH gene. Such compound may be a small molecule, a polypeptide, an antibody, a hybridizing oligonucleotide, or a genome editing agent. [0015] In some embodiments, the compound introduced to the cell may include an inhibitor of the JAK/STAT pathway. Such compound may include at least one of
Ruxolitinib, Oclacitinib, Baricitinib, Filgotinib, Gandotinib, Lestaurtinib, PF-04965842, Upadacitinib, Cucurbitacin I, CHZ868, Fedratinib, AC430, AT9283, ati-5000l and ati- 50002, AZ 960, AZD1480, BMS-911543, CEP-33779, Cerdulatinib (PRT062070, PRT2070), Curcumol, Decemotinib (VX-509), Fedratinib (SAR302503, TG101348), FLLL32, FM-381, GLPG0634 analogue, Go6976, JANEX-l (WHI-P131), Momelotinib (CYT387), NVP-BSK805, Pacritinib (SB1518), Peficitinib (ASP015K, JNJ-54781532), PF-06651600, PF-06700841, R256 (AZD0449), Solcitinib (GSK2586184 or GLPG0778), S-Ruxolitinib (INCB018424), TG101209, Tofacitinib (CP-690550), WHI-P154, WP1066, XL019, ZM 39923 HC1, or a derivative or an analog thereof.
[0016] In some embodiments, the compound introduced to the cell may include an inhibitor of the Tyrosine kinase/MAPK pathway. Such compound may include at least one of Amuvatinib, Bosutinib, Cediranib, Ceritinib, CP-673451, Dasatinib, GZD824
Dimesylate, Merestinib, R788 (fostamatinib disodium hexahydrate), or a derivative or an analog thereof
[0017] In some embodiments, the compound introduced to the cell may include 17-AAG (Tanespimycin), Amlodipine Besylate, ATRA (all-trans retinoic acid), Chloroquine phosphate, Deoxycorticosterone, Darapladib, Echinomycin, Enzastaurin, Epinephrine, EVP-6124 (hydrochloride) (encenicline), EW-7197, FRAX597, Ibrutinib, Perphenazine, Phenformin, PND-1186, Rifampicin, Semagacestat, Thalidomide, WAY600, WYE-125132 (WYE-132), Zibotentan, Activin, Anti mullerian hormone, GDF10 (BMP3b), IGF-l,
Nodal, PDGF, TNF-a, Wnt3a, or a derivative or an analog thereof.
[0018] In alternative embodiments, the compound introduced to the cell may include one or more RNAi agents against a signaling molecule identified to regulate PAH expression.
In some embodiments, the compound includes one or more small interfering RNA (siRNA) targeting one or more genes selected from the group consisting of JAK1 , JAK2, PDGFRA, PDGFRB, SRC and ABL.
[0019] In any one of the cellular methods disclosed above, the compound increases the expression of the PAH gene. In some embodiments, the expression of the PAH gene is increased by at least about 40% over baseline or over levels measured following administration of a control. The cell may have at least one mutation within or near the PAH gene. The mutation(s) may decrease the activity of phenylalanine hydroxylases or reduce the expression of PAH in the cell as compared to activity or expression associated with a canonical wild-type sequence.
[0020] In any one of the methods disclosed above, the cell may be a mammalian cell. In some embodiments, the cell is a human cell. In some embodiments, the cell is a mouse cell. In some embodiments, the cell is a hepatocyte.
[0021] Further provided herein is a method of modulating the expression of a PAH gene in a cell by comprising introducing to the cell one or more compounds that alter expression of one or more of the upstream or downstream neighborhood genes or its RSRs of the insulated neighborhood comprising the PAH gene. The insulated neighborhood may comprise the region on chromosome 12 at position 102,882,556 to 103,550,727 (human CRCH38/hg38 genome assembly). In some embodiments, the one or more downstream neighborhood gene includes at least one of ASCL1 and C120RF42.
BRIEF DESCRIPTION OF THE DRAWINGS
[0022] The foregoing and other objects, features and advantages will be apparent from the following description of particular embodiments of the invention, as illustrated in the accompanying drawings. The drawings are not necessarily to scale; emphasis instead being placed upon illustrating the principles of various embodiments of the invention.
[0023] FIG. 1 illustrates the packaging of chromosomes in a nucleus, the localized topological domains into which chromosomes are organized, insulated neighborhoods in TADs and finally an example of an arrangement of a signaling center(s) around a particular disease gene.
[0024] FIG. 2A and FIG. 2B illustrate a linear and 3D arrangement of the CTCF boundaries of an insulated neighborhood.
[0025] FIG. 3A and FIG. 3B illustrate tandem insulated neighborhoods and gene loops formed in such insulated neighborhoods.
[0026] FIG. 4 illustrates the concept of an insulated neighborhood contained within a larger insulated neighborhood and the signaling which may occur in each.
[0027] FIG. 5 illustrates the components of a signaling center; including transcriptional factors, signaling proteins, and/or chromatin regulators.
[0028] FIG. 6 is a plot showing the dose response curve of Momelotinib in primary human hepatocytes. [0029] FIG. 7 is a bar chart showing the effect of JAK1 and JAK2 knock-down on PAH mRNA levels. Plotted values are the relative fold changes of target mRNA level with respect to the non-targeting control.
[0030] FIG. 8A and FIG. 8B are bar charts showing the effect of selected compound treatments on PAH expression in mouse liver.
[0031] FIG. 9 is a diagram of the BH4 pathway.
[0032] FIG. 10A-C show (A) a diagram of the conversion of phenylalanine to tyrosine using phenylalanine hydroxylase, (B) an exemplary figure of the production concentration over time, and (C) a gel showing the separation of tyrosine and phenylalanine.
[0033] FIG. 11 shows a schematic of the phenylalanine hydroxylase activity assay.
[0034] FIG. 12 shows the PAH enzyme activity in cytosolic extracts from primary human hepatocytes.
[0035] FIG. 13 shows the PAH enzyme activity in mouse livers after treatment with dasatinib.
[0036] FIG. 14 shows the effect of selected compound treatments on PAH enzyme levels in mouse liver.
[0037] FIG. 15 shows the fold increase in expression of PAH and GCH1 upon targeting of select gene regulatory pathways by select perturbagens in primary hepatocytes.
[0038] FIG. 16A shows the relative expression of PAH in mice, 6 hours post treatment with select compounds.
[0039] FIG. 16B shows the relative expression of GCH1 in mice, 6 hours post treatment with select compounds.
[0040] FIG. 17 is a western blot showing relative expression of PAH protein and Bactin protein as a loading control in mouse liver lysates from mice treated with select compounds.
[0041] FIG. 18A is a diagram of the dosing schedule in mice for the treatment of Dasatinib for analysis of PAH activity in mouse livers.
[0042] FIG. 18B shows the increase in PAH activity and mRNA in mouse livers after treatment with Dasatinib.
[0043] FIG. 19 is a diagram of the dosing schedule for measurement of PAH activity in mouse livers treated with XL228, PF-562271, SIS3.
[0044] FIG. 20 is a graph that shows PAH activity of mouse livers treated with XL228, PF-562271, SIS3 and vehicle control. [0045] FIG. 21 is a graph that shows the specific activity of PAH of mouse livers treated with XL228, PF-562271 and vehicle control.
[0046] FIG. 22 is a western blot showing amount of PAH protein produced by cells expressing the select constructs.
[0047] FIG. 23 is a graph showing increased activity of PAH198 R243Q with increased lysate.
DETAILED DESCRIPTION
I. INTRODUCTION AND DEFINTIONS
[0048] The present disclosure provides compositions and methods for the treatment of phenylalanine hydroxylase deficiency, such as phenylketonuria, in mammals, including in humans. In particular, the disclosure provides compounds and related use for the modulation of the Phenylalanine Hydroxylase (PAH) gene.
[0049] The term“phenylalanine hydroxylase deficiency” or“PAH deficiency”, as used herein, refers to any condition or disorder that is manifested in an elevated phenylalanine level in the blood. Phenylalanine hydroxylase deficiency includes mild
hyperphenylalaninemia, mild PKU, and classic PKU.“Mild hyperphenylalaninemia” or “non-PKU hyperphenylalaninemia” is defined as the presence of plasma phenylalanine levels that exceed the limits of the upper reference range (120 pmol/L or 2 mg/dL) without treatment but that are below the level found in patients with PKU.“Mild PKU” or “moderate PKU” refers to conditions with plasma phenylalanine levels over 600 pmol/L (10 mg/dL) but lower than 1200 pmol/L (20 mg/dL) without treatment.“Classic PKU” is defined as plasma phenylalanine levels that exceed 1200 pmol/L (20 mg/dL).“Mild PKU” or“classic PKU” are herein broadly referred to as“PKU.”
[0050] The present disclosure also embraces the alteration, perturbation and ultimate regulated control of gene signaling networks (GSNs). Such gene signaling networks include genomic signaling centers found within insulated neighborhoods of the genomes of biological systems. Compounds modulating gene expression may act through modulating one or more gene signaling networks.
[0051] Described herein are compositions and methods for perturbation of genomic signaling centers (GSCs) or entire gene signaling networks (GSNs) for the treatment of phenylalanine hydroxylase deficiency, such as phenylketonuria. The details of one or more embodiments of the invention are set forth in the accompanying description below.
Although any materials and methods similar or equivalent to those described herein can be used in the practice or testing of the present invention, the preferred materials and methods are now described. Other features, objects and advantages of the invention will be apparent from the description. In the description, the singular forms also include the plural unless the context clearly dictates otherwise. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. In the case of conflict, the present description will control.
[0052] As used herein, a“gene signaling network” or“GSN” comprises the set of biomolecules associated with any or all of the signaling events from a particular gene, e.g., a gene-centric network. As there are over 20,000 protein-coding genes in the human genome, there are at least this many gene signaling networks. And to the extent some genes are non-coding genes, the number increases greatly. Gene signaling networks differ from canonical signaling pathways which are mapped as standard protein cascades and feedback loops.
[0053] Traditionally, signaling pathways have been identified using standard
biochemical techniques and, for the most part, are linear cascades with one protein product signaling the next protein product-driven event in the cascade. While these pathways may bifurcate or have feedback loops, the focus has been almost exclusively at the protein level.
[0054] Gene signaling networks (GSNs) of the present disclosure represent a different paradigm to defining biological signaling— taking into account protein-coding and nonprotein-coding signaling molecules, genomic structure, chromosomal occupancy, chromosomal remodeling, the status of the biological system and the range of outcomes associated with the perturbation of any biological systems comprising such gene signaling networks.
[0055] Genomic architecture, while not static, plays an important role in defining the framework of the GSNs of the present disclosure. Such architecture includes the concepts of chromosomal organization and modification, topologically associated domains (TADs), insulated neighborhoods (INs), genomic signaling centers (GSCs), signaling molecules and their binding motifs or sites, and of course, the genes encoded within the genomic architecture.
[0056] The present disclosure, by elucidating a more definitive set of connectivities of the GSNs associated with the PAH gene, provides a fine-tuned mechanism to address phenylalanine hydroxylase deficiency such as phenylketonuria. Genomic architecture
[0057] Cells control gene expression using thousands of elements that link cellular signaling to the architecture of the genome. Genomic system architecture includes regions of DNA, RNA transcripts, chromatin remodelers, and signaling molecules.
Chromosomes
[0058] Chromosomes are the largest subunit of genome architecture that contain most of the DNA in humans. Specific chromosome structures have been observed to play important roles in gene control, as described in Hnisz et al., Cell 167, November 17, 2016, which is hereby incorporated by reference in its entirety. The introns (“non-coding regions”) provide protein binding sites and other regulatory structures, while the exons encode for signaling molecules, such as transcription factors, that interact with the non-coding regions to regulate gene expression. DNA sites within non-coding regions on the chromosome also interact with each other to form looped structures. These interactions form a chromosome scaffold that is preserved through development and plays an important role in gene activation and repression. Interactions rarely occur among chromosomes and are usually within the same domain of a chromosome.
[0059] In situ hybridization techniques and microscopy have revealed that individual interphase chromosomes tend to occupy small portions of the nucleus and do not spread throughout this organelle. See, Cremer and Cremer, Cold Spring Harbor Perspectives in Biology 2, a003889, 2010, which is hereby incorporated by reference in its entirety. This small surface area occupancy might reduce interactions between chromosomes.
Topologically associating domains (TADs)
[0060] The term“topologically associating domains” (TADs), as used herein, refers to structures that represent a modular organization of the chromatin and have boundaries that are shared by the different cell types of an organism. TADs, alternatively known as topological domains, are hierarchical units that are subunits of the mammalian
chromosome structure. See, Dixon et al., Nature, 485(7398):376-80, 2012; Filippova et ak, Algorithms for Molecular Biology, 9:14, 2014; Gibcus and Dekker Molecular Cell, 49(5):773-82, 2013; Naumova et al., Science, 42(6l6l):948-53, 2013; which are hereby incorporated by reference in their entireties. TADs are megabase-sized chromosomal regions that demarcate a microenvironment that allows genes and regulatory elements to make productive DNA-DNA contacts. TADs are defined by DNA-DNA interaction frequencies. The boundaries of TADs consist of regions where relatively fewer DNA-DNA interactions occur, as described in Dixon et al., Nature, 485(7398):376-80, 2012; Nora et al., Nature, 485(7398):38l-5, 2012; which are hereby incorporated by reference in their entirety. TADs represent structural chromosomal units that function as gene expression regulators.
[0061] TADs may contain about 7 or more protein-coding genes and have boundaries that are shared by the different cell types. See, Smallwood et al., Current Opinion in Cell Biology, 25(3):387-94, 2013, which is hereby incorporated by reference in its entirety. Some TADs contain active genes and others contain repressed genes, as the expression of genes within a single TAD is usually correlated. See, Cavalli et al., Nature Structural & Molecular Biology, 20(3):290-9, 2013, which is hereby incorporated by reference in its entirety. Sequences within a TAD find each other with high frequency and have concerted, TAD-wide histone chromatin signatures, expression levels, DNA replication timing, lamina association, and chromocenter association. See, Dixon et al., Nature,
485(7398):376-80, 2012; Le Dily et al., Genes Development, 28:2151-62, 2014; Dixon et al., Nature, 485(7398):376-80, 2012; Wijchers, Genome Research, 25:958-69, 2015, which are hereby incorporated by reference in their entireties.
[0062] Gene loops and other structures within TADs influence the activities of transcription factors (TFs), cohesin, and 11 -zinc finger protein (CTCF), a transcriptional repressor. See, Baranello et al., Proceedings of the National Academy of Sciences,
111(3):889-9, 2014, which is hereby incorporated by reference in its entirety. The structures within TADs include cohesin-associated enhancer-promoter loops that are produced when enhancer-bound TFs bind cofactors, for example Mediator, that, in turn, bind RNA polymerase II at promoter sites. See, Lee and Young, Cell, 152(6):1237-51, 2013; Lelli et al., 2012; Roeder, Annual Reviews Genetics 46:43-68, 2005; Spitz and Furlong, Nature Reviews Genetics, 13(9):613-26, 2012; Dowen et al., Cell, 159(2): 374- 387, 2014; Lelli et al., Annual Review of Genetics, 46:43-68, 2012, which are hereby incorporated by reference in their entireties. The cohesin-loading factor Nipped-B-like protein (NIPBL) binds Mediator and loads cohesin at these enhancer-promoter loops. See, Kagey et al., Nature, 467(73l4):430-5, 2010, which is hereby incorporated by reference in its entirety.
[0063] TADs have similar boundaries in all human cell types examined and constrain enhancer-gene interactions. See, Dixon et al., Nature, 518:331-336, 2015; Dixon et al., Nature, 485:376-380, 2012, which are hereby incorporated by reference in their entirety. This architecture of the genome helps explain why most DNA contacts occur within the TADs and enhancer-gene interactions rarely occur between chromosomes. However, TADs provide only partial insight into the molecular mechanisms that influence specific enhancer-gene interactions within TADs.
[0064] Long-range genomic contacts segregate TADs into an active and inactive compartment. See, Lieberman-Aiden et al., Science, 326:289-93, 2009, which is hereby incorporated by reference in its entirety. The loops formed between TAD boundaries seem to represent the longest-range contacts that are stably and reproducibly formed between specific pairs of sequences. See, Dixon et al., Nature, 485(7398):376-80, 2012, which is hereby incorporated by reference in its entirety.
[0065] In some embodiments, the methods of the present disclosure are used to alter gene expression from genes located in a TAD. In some embodiments, TAD regions are modified to alter gene expression of a non-canonical pathway as defined herein or as definable using the methods described herein.
Insulated neighborhoods
[0010] The term“insulated neighborhood” (IN), as used herein, refers to chromosome structure formed by the looping of two interacting sites in the chromosome sequence that may comprise CCCTC-Binding factor (CTCF) co-occupied by cohesin and affect the expression of genes in the insulated neighborhood as well as those genes in the vicinity of the insulated neighborhoods.
[0011] These interacting sites may comprise CCCTC-Binding factor (CTCF). These CTCF sites are often co-occupied by cohesin. The integrity of these cohesin-associated chromosome structures affects the expression of genes in the insulated neighborhood as well as those genes in the vicinity of the insulated neighborhoods. A“neighborhood gene” is a gene localized within an insulated neighborhood. Neighborhood genes may be coding or non-coding.
[0012] Insulated neighborhood architecture is defined by at least two boundaries which come together, directly or indirectly, to form a DNA loop. The boundaries of any insulated neighborhood comprise a primary upstream boundary and a primary downstream boundary. Such boundaries are the outermost boundaries of any insulated neighborhood. Within any insulated neighborhood loop, however, secondary loops may be formed. Such secondary loops, when present, are defined by secondary upstream boundaries and secondary downstream boundaries, relative to the primary insulated neighborhood. Where a primary insulated neighborhood contains more than one internal loop, the loops are numbered relative to the primary upstream boundary of the primary loop, e.g., the secondary loop (first loop within the primary loop), the tertiary loop (second loop within the primary loop), the quaternary loop (the third loop within the primary loop) and so on.
[0013] Insulated neighborhoods may be located within topologically associated domains (TADs) and other gene loops. TADs are defined by DNA-DNA interaction frequencies, and average 0.8 Mb, contain approximately 7 protein-coding genes and have boundaries that are shared by the different cell types of an organism. According to Dowen, the expression of genes within a TAD is somewhat correlated, and thus some TADs tend to have active genes and others tend to have repressed genes. Dowen et al., Cell. 2014 Oct 9; 159(2): 374-387.
[0014] Insulated neighborhoods may exist as contiguous entities along a chromosome or may be separated by non-insulated neighborhood sequence regions. Insulated
neighborhoods may overlap linearly only to be defined once the DNA looping regions have been joined. While insulated neighborhoods may comprise 3-12 genes, they may contain,
1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or more genes.
[0015] A“minimal insulated neighborhood” is an insulated neighborhood having at least one neighborhood gene and associated regulatory sequence region or regions (RSRs) which facilitate the expression or repression of the neighborhood gene such as a promoter and/or enhancer and/or repressor region, and the like. It is contemplated that regulatory sequence regions may coincide or even overlap with an insulated neighborhood boundary. Regulatory sequence regions, as used herein, include but are not limited to regions, sections, sites or zones along a chromosome whereby interactions with signaling molecules occur in order to alter expression of a neighborhood gene. As used herein, a“signaling molecule” is any entity, whether protein, nucleic acid (DNA or RNA), organic small molecule, lipid, sugar or other biomolecule, which interacts directly, or indirectly, with a regulatory sequence region on a chromosome. Regulatory sequence regions (RSRs) may also be referred to as“genomic signaling centers” or“GSCs.”
[0016] One category of specialized signaling molecules are transcription factors.
“Transcription factors” are those signaling molecules which alter, whether to increase or decrease, the transcription of a target gene, e.g., a neighborhood gene.
[0017] According to the present disclosure, neighborhood genes may have any number of upstream or downstream genes along the chromosome. Within any insulated neighborhood, there may be one or more, e.g., one, two, three, four or more, upstream and/or downstream neighborhood genes relative to the primary neighborhood gene. A “primary neighborhood gene” is a gene which is most commonly found within a specific insulated neighborhood along a chromosome. An upstream neighborhood gene of a primary neighborhood gene may be located within the same insulated neighborhood as the primary neighborhood gene. A downstream neighborhood gene of a primary neighborhood gene may be located within the same insulated neighborhood as the primary neighborhood gene.
[0018] The present disclosure provides methods of altering the penetrance of a gene or gene variant. As used herein,“penetrance” is the proportion of individuals carrying a particular variant of a gene (e.g., mutation, allele or generally a genotype, whether wild type or not) that also exhibits an associated trait (phenotype) of that variant gene. In some situations of disease, penetrance of a disease-causing mutation measured as the proportion of individuals with the mutation who exhibit clinical symptoms. Consequently, penetrance of any gene or gene variant exists on a continuum.
[0019] Insulated neighborhoods are functional units that group genes under the same control mechanism, which are described in Dowen et al., Cell, 159: 374-387 (2014), which is hereby incorporated by reference in its entirety. Insulated neighborhoods provide the mechanistic background for higher-order chromosome structures, such as TADs which are shown in FIG. 1. Insulated neighborhoods are chromosome structures formed by the looping of the two interacting CTCF sites co-occupied by cohesin as shown in FIG. 1. The integrity of these structures is important for proper expression of local genes. Generally, 1 to 10 genes are clustered in each neighborhood with a median number of 3 genes within each one. The genes controlled by the same insulated neighborhood are not readily apparent from a two-dimensional view of DNA. In humans, there are about 13,801 insulated neighborhoods in a size range of 25 kb-940 kb with a median size of 186 kb. Insulated neighborhoods are conserved among different cell types. Smaller INs that occur within a bigger IN are referred to as nested insulated neighborhoods (NINs). TADs can consist of a single IN, or one IN and one NIN and two NINs as shown in FIG. 2B.
[0020] As used herein, the term“boundary” refers to a point, limit, or range indicating where a feature, element, or property ends or begins. Accordingly, an“insulated neighborhood boundary” refers to a boundary that delimits an insulated neighborhood on a chromosome. According to the present disclosure, an insulated neighborhood is defined by at least two insulated neighborhood boundaries, a primary upstream boundary and a primary downstream boundary. The“primary upstream boundary” refers to the insulated neighborhood boundary located upstream of a primary neighborhood gene. The“primary downstream boundary” refers to the insulated neighborhood boundary located downstream of a primary neighborhood gene. Similarly, when secondary loops are present as shown in FIG. 2B, they are defined by secondary upstream and downstream boundaries. A “secondary upstream boundary” is the upstream boundary of a secondary loop within a primary insulated neighborhood, and a“secondary downstream boundary” is the downstream boundary of a secondary loop within a primary insulated neighborhood. The directionality of the secondary boundaries follows that of the primary insulated neighborhood boundaries.
[0021] Components of an insulated neighborhood boundary may comprise the DNA sequences at the anchor regions and associated factors (e.g., CTCF, cohesin) that facilitate the looping of the two boundaries. The DNA sequences at the anchor regions may contain at least one CTCF binding site. Experiments using the ChIP-exo technique revealed a 52 bp CTCF binding motif containing four CTCF binding modules (see Fig 1, Ong and Corces, Nature reviews Genetics, 12:283-293, 2011, which is incorporated herein by reference in its entirety). The DNA sequences at the insulated neighborhood boundaries may contain insulators. In some cases, insulated neighborhood boundaries may also coincide or overlap with regulatory sequence regions, such as enhancer-promoter interaction sites.
[0022] In some embodiments of the present disclosure, disrupting or altering an insulated neighborhood boundary may be accomplished by altering specific DNA sequences (e.g., CTCF binding sites) at the boundaries. For example, existing CTCF binding sites at insulated neighborhood boundaries may be deleted, mutated, or inverted. Alternatively, new CTCF binding sites may be introduced to form new insulated neighborhoods. In other embodiments, disrupting or altering an insulated neighborhood boundary may be accomplished by altering the histone modification (e.g., methylation, demethylation) at the boundaries. In other embodiments, disrupting or altering an insulated neighborhood boundary may be accomplished by altering (e.g., blocking) the binding of CTCF and/or cohesin to the boundaries. In cases where insulated neighborhood boundaries coincide or overlap with regulatory sequence regions, disrupting or altering an insulated neighborhood boundary may be accomplished by altering the regulatory sequence regions (RSR) or the binding of the RSR-associated signaling molecules.
Controlling expression from insulated neighborhoods: Signaling centers
[0023] Historically, the term“signaling center” has been used to describe a group of cells responding to changes in the cellular environment. See, Guger et al., Developmental Biology 172: 115-125 (1995), which is incorporated by reference herein in its entirety. Similarly, the term“signaling center”, as used herein, refers to a defined region of DNA in a living cell that interacts with a defined set of biomolecules, such as signaling proteins or signaling molecules (e.g., transcription factors) to regulate gene expression in a context- specific manner.
[0024] Specifically, the term“genomic signaling center”, i.e., a“signaling center”, as used herein, refers to regions within insulated neighborhoods that include regions capable of binding context-specific combinatorial assemblies of signaling molecules/signaling proteins that participate in the regulation of the genes within that insulated neighborhood or among more than one insulated neighborhood.
[0025] Signaling centers have been discovered to regulate the activity of insulated neighborhoods. These regions control which genes are expressed and the level of expression in the human genome. Loss of the structural integrity of signaling centers contributes to deregulation of gene expression and potentially causing disease.
[0026] Signaling centers include enhancers bound by a highly context-specific combinatorial assemblies of transcription factors. These factors are recruited to the site through cellular signaling. Signaling centers include multiple genes that interact to form a three-dimensional transcription factor hub macrocomplex. Signaling centers are generally associated with one to four genes in a loop organized by biological function.
[0027] The compositions of each signaling center has a unique composition including the assemblies of transcription factors, the transcription apparatus, and chromatin regulators. Signaling centers are highly context specific, permitting drugs to control response by targeting signaling pathways.
[0028] Multiple signaling centers may interact to control the different combinations of genes within the same insulated neighborhood.
Binding sites for signaling molecules
[0029] A series of consensus binding sites, or binding motifs for binding sites, for signaling molecules has been identified by the present inventors. These consensus sequences reflect binding sites along a chromosome, gene, or polynucleotide for signaling molecules or for complexes which include one or more signaling molecules.
[0030] In some embodiments, binding sites are associated with more than one signaling molecule or complex of molecules.
Enhancers
[0031] The term“enhancer”, as used herein, refers to regulatory DNA sequences that, when bound by transcription factors, enhance the transcription of an associated gene.
[0032] Enhancers are gene regulatory elements that control cell type specific gene expression programs in humans. See, Buecker and Wysocka, Trends in genetics: TIG 28, 276-284, 2012; Heinz et ak, Nature reviews Molecular Cell Biology, 16: 144-154, 2015; Levine et ak, Cell, 157:13-25, 2014; Ong and Corces, Nature reviews Genetics, 12:283- 293, 2011; Ren and Yue, Cold Spring Harbor symposia on quantitative biology, 80: 17-26, 2015, which are hereby incorporated by reference in their entireties. Enhancers are segments of DNA that are generally a few hundred base pairs in length and are occupied by multiple transcription factors that recruit co-activators and RNA polymerase II to target genes. See, Bulger and Groudine, Cell, 144:327-339, 2011; Spitz and Furlong, Nature reviews Genetics, 13:613-626, 2012; Tjian and Maniatis, Cell, 77:5-8, 1994, which are hereby incorporated by reference in their entireties. Enhancer RNA molecules transcribed from these regions of DNA also“trap” transcription factors capable of binding DNA and RNA. A region with more than one enhancer is a“super-enhancer.” The term“super enhancers”, as used herein, refers to clusters of transcriptional enhancers that drive expression of genes that define cell identity.
[0033] Insulated neighborhoods provide a microenvironment for specific enhancer-gene interactions that are vital for both normal gene activation and repression. Transcriptional enhancers control over 20,000 protein-coding genes to maintain cell type-specific gene expression programs in all human cells. Tens of thousands of enhancers are estimated to be active in any given human cell type. See, ENCODE Project Consortium et ak, Nature, 489, 57- 74, 2012; Roadmap Epigenomics et ak, Nature, 518, 317-330, 2015, which are hereby incorporated by reference in their entirety. Enhancers and their associated factors can regulate expression of genes located upstream or downstream by looping to the promoters of these genes. Cohesin ChIA-RET studies carried out to gain insight into the relationship between transcriptional control of cell identity and control of chromosome structure reveal that majority of the super-enhancers and their associated genes occur within large loops that are connected through interacting CTCF-sites co-occupied by cohesin. Such super enhancer domains (SD) usually contain one super-enhancer that loops to one gene within the SD and the SDs appear to restrict super-enhancer activity to genes within the SD. The correct association of super-enhancers and their target genes in insulated neighborhoods is highly vital because the mis-targeting of a single super-enhancer is sufficient to cause disease. See Groschel et al., Cell, 157(2):369-81, 2014.
[0034] Most of the disease-associated non-coding variation occurs in the vicinity of enhancers and hence might impact these enhancer target genes. Therefore, deciphering the features conferring specificity to enhancers is important for modulatory gene expression. See, Ernst et al., Nature, 473, 43-49, 2011; Farh et al., Nature, 518, 337-343,2015; Hnisz et al., Cell, 155, 934-947, 2013; Maurano et al., Science, 337, 1190-1195, 2012, which are hereby incorporated by reference in their entirety. Studies suggest that some of the specificity of enhancer-gene interactions may be due to the interaction of DNA binding transcription factors at enhancers with specific partner transcription factors at promoters. See, Butler and Kadonaga, Genes & Development, 15, 2515-2519, 2001; Choi and Engel, Cell, 55, 17- 26, 1988; Ohtsuki et al., Genes & Development, 12, 547-556, 1998, which are hereby incorporated by reference in their entireties. DNA sequences in enhancers and in promoter-proximal regions bind to a variety of transcription factors expressed in a single cell. Diverse factors bound at these two sites interact with large cofactor complexes and interact with one another to produce enhancer-gene specificity. See, Zabidi et al., Nature, 518:556-559, 2015, which is hereby incorporated by reference in its entirety.
[0035] In some embodiments, enhancer regions may be targeted to alter or elucidate gene signaling networks (GSNs).
Insulators
[0010] The term“insulator”, as used herein, refers to regulatory elements that block the ability of an enhancer to activate a gene when located between them and contribute to specific enhancer-gene interactions. See, Chung et al., Cell 74:505-514, 1993; Geyer and Corces, Genes & Development 6:1865-1873, 1992; Kellum and Schedl, Cell 64:941-950, 1991; Udvardy et al., Journal of molecular biology 185:341-358, 1985, which are hereby incorporated by reference in their entirety. Insulators are bound by the transcription factor CTCF but not all CTCF sites function as insulators. See, Bell et al., Cell 98: 387-396,
1999; Liu et al., Nature biotechnology 33:198-203, 2015, which are hereby incorporated by reference in their entireties. The features that distinguish the subset of CTCF sites that function as insulators have not been previously understood.
[0011] Genome-wide maps of the proteins that bind enhancers, promoters and insulators, together with knowledge of the physical contacts that occur between these elements provide further insight into understanding of the mechanisms that generate specific enhancer-gene interactions. See, Chepelev et al., Cell research, 22:490-503, 2012; DeMare et al., Genome Research, 23:1224-1234, 2013; Dowen et al., Cell, 159:374-387, 2014; Fullwood et al., Genes & Development 6:1865-1873, 2009; Handoko et al., Nature genetics 43:630-638, 2011; Phillips -Cremins et al., Cell, 153:1281-1295, 2013; Tang et al., Cell 163: 1611-1627, 2015, which are hereby incorporated by reference in their entirety. Enhancer-bound proteins are constrained such that they tend to interact only with genes within these CTCF-CTCF loops. The subset of CTCF sites that form these loop anchors thus function to insulate enhancers and genes within the loop from enhancers and genes outside the loop, as shown in FIG. 2B. In some embodiments, insulator regions may be targeted to alter or elucidate gene signaling networks (GSNs).
Cohesin and CTCF associated loops and anchor sites/regions
[0012] CTCF interactions link sites on the same chromosome forming loops, which are generally less than 1 Mb in length. Transcription occurs both within and outside the loops, but the nature of this transcription differs between the two regions. Studies show that enhancer-associated transcription is more prominent within the loops. Thus, the insulator state is enriched specifically at the CTCF loop anchors. CTCF loops thus either enclose gene poor regions, with a tendency for genes to be centered within the loops or leave out gene dense regions outside the CTCF loops. CTCF loops exhibit reduced exon density relative to their flanking regions. Gene ontology analysis reveals that genes located within CTCF loops are enriched for response to stimuli and for extracellular, plasma membrane and vesicle cellular localizations. On the other hand, genes present within the flanking regions just outside the loops exhibit an expression pattern similar to housekeeping genes i.e. these genes are on average more highly expressed than the loop-enclosed genes, are less cell-line specific in their expression pattern, and have less variation in their expression levels across cell lines. See Oti et al., BMC Genomics, 17:252, 2016, which is incorporated by reference in its entirety.
[0013] Anchor regions are binding sites for CTCF that influence conformation of an insulated neighborhood. Deletion of anchor sites may result in activation of genes that are usually transcriptionally silent, thereby resulting in a disease phenotype. In fact, somatic mutations are common in loop anchor sites of oncogene-associated insulated
neighborhoods. The CTCF DNA-binding motif of the loop anchor region has been observed to be the most altered human transcription-factor binding sequence of cancer cells. See, Hnisz et al., Cell 167, November 17, 2016, which is incorporated by reference in its entirety.
[0014] Anchor regions have been observed to be largely maintained during cell development, and are especially conserved in the germline of humans and primates. In fact, the DNA sequence of anchor regions are more conserved in CTCF anchor regions than at CTCF binding sites that are not part of an insulated neighborhood. Therefore, cohesin may be used as a target for ChIA-RET to identify locations of both.
Cohesin also becomes associated with CTCF-bound regions of the genome, and some of these cohesin-associated CTCF sites facilitate gene activation while others may function as insulators. See, Dixon et al.. Nature. 485(73981:376-80, 2012: Parelho et al.. Cell, 132(31:422-33, 2008: Phillips -Cremins and Corces, Molecular Cell, 50(41:461-74, 2013); Seitan et al.. Genome Research, 23(121:2066-77, 2013; Wendt et al.. Nature, 45l(7l80):796-80l, 2008), which are hereby incorporated by reference in their entireties. Cohesin and CTCF are associated with large loop substructures within TAPs, and cohesin and Mediator are associated with smaller loop structures that form within CTCF-bounded regions. See, de Wit et al.. Nature. 501 (74661:227- 31. 2013: Cremins et ah. Cell. 153(61:1281-95. 2013: Sofueva et al.. EMBO, 32(24):3l 19-29, 2013, which are hereby incorporated by reference in their entireties. In some embodiments, cohesin and CTCF associated loops and anchor sites/regions may be targeted to alter or elucidate gene signaling networks (GSNs).
Genetic variants
[0015] Genetic variations within signaling centers are known to contribute to disease by disrupting protein binding on chromosomes, such as described in Hnisz et al., Cell 167, November 17, 2016. Variations of the sequence of CTCF anchor regions that interfere with formation of insulated neighborhoods are observed to result in dysregulation of gene activation and repression. CTCF malfunctions caused by various genetic and epigenetic mechanisms may lead to pathogenesis. Therefore, in some embodiments, it is beneficial to alter any one or more gene signaling networks (GSNs) associated with such variant-driven etiology in order to effect one or more positive treatment outcomes. Single nucleotide polymorphisms ( SNPs )
[0016] Most disease associated SNPs are located in the proximity of signaling centers. For example, 94.2% of SNPs occur in non-coding regions, which include signaling centers. In some embodiments, SNPs are altered in order to study and/or alter the signaling from one or more GSN.
Signaling molecules
[0017] Signaling molecules include any protein that functions in cellular signaling pathways, whether canonical or the gene signaling network pathways defined herein or capable of being defined using the methods described herein. Transcription factors are a subset of signaling molecules. Certain combinations of signaling and master transcription factors associate to an enhancer region to influence expression of a gene. Master regulator factors direct transcription factors in specific tissues. For example, in blood, GATA transcription factors are master regulators that direct TCF7L2 of the Wnt cellular signaling pathway. In the liver, HNG4 is a master regulator to direct SMAD in lineage tissues and patterns.
[0018] Transcriptional regulation allows controlling how often a given gene is transcribed. Transcription factors alter the rate at which transcripts are produced by making conditions for transcription initiation more or less favorable. A transcription factor selectively alters a signaling pathway which in turn affects the genes expressed by a signaling center. Signaling centers are components of transcriptional regulators. In some embodiments, signaling molecules may be used, targeted in order to elucidate or alter the signaling of gene signaling networks of the present disclosure.
[0019] Table 18 of U.S. 62/501,795, which is hereby incorporated by reference in its entirety, provides a list of signaling molecules including those which act as transcription factors (TF) and/or chromatin remodeling factors (CR) that function in various cellular signaling pathways. The methods described herein may be used to inhibit or activate the expression of one or more signaling molecules associated with the regulatory sequence region of the primary neighborhood gene encoded within an insulated neighborhood. The methods may thus alter the signaling signature of one or more primary neighborhood genes which are differentially expressed upon treatment with the therapeutic agent compared to an untreated control. Transcription factors
[0020] Transcription factors generally regulate gene expression by binding to enhancers and recruiting coactivators and RNA polymerase II to target genes. See Whyte et al., Cell, 153(2): 307-319, 2013, which is incorporated by reference in its entirety. Transcription factors bind“enhancers” to stimulate cell-specific transcriptional program by binding regulatory elements distributed throughout the genome.
[0021] There are about 1800 known transcription factors in the human genome. There are epitopes on the DNA of the chromosomes that provide binding sites for proteins or nucleic acid molecules such as ribosomal RNA complexes. Master regulators direct a combination of transcription factors through cell signaling above and DNA below. These characteristics allow for determination of the location of the next signaling center. In some embodiments, transcription factors may be used or targeted, to alter or elucidate the gene signaling networks of the present disclosure.
Master transcription factors
[0022] Master transcription factors bind and establish cell-type specific enhancers.
Master transcription factors recruit additional signaling proteins, such as other transcription factors, to enhancers to form signaling centers. An atlas of candidate master TFs for 233 human cell types and tissues is described in D’Alessio et al., Stem Cell Reports 5, 763-775 (2015), which is hereby incorporated by reference in its entirety. In some embodiments, master transcription factors may be used or targeted, to alter or elucidate the gene signaling networks of the present invention.
Signaling transcription factors
[0023] Signaling transcription factors are transcription factors, such as homeoproteins, that travel between cells as they contain protein domains that allow them to do the so. Homeoproteins such as Engrailed, Hoxa5, Hoxb4, Hoxc8, Emxl, Emx2, Otx2 and Pax6 are able to act as signaling transcription factors. The homeoprotein Engrailed possesses internalization and secretion signals that are believed to be present in other homeoproteins as well. This property allows homeoproteins to act as signaling molecules in addition to being transcription factors. Homeoproteins lack characterized extracellular functions leading to the perception that their paracrine targets are intracellular. The ability of homeoproteins to regulate transcription and, in some cases, translation is most likely to affect paracrine action. See Prochiantz and Joliot, Nature Reviews Molecular Cell Biology, 2003. In some embodiments, signaling transcription factors may be used or targeted, to alter or elucidate the gene signaling networks of the present disclosure.
Chromatin modifications
[0024] Chromatin remodeling is regulated by over a thousand proteins that are associated with histone modification. See, Ji et al., PNAS, 112(12):3841-3846(2015), which is hereby incorporated by reference in its entirety. Chromatin regulators are specific sets of proteins associated with genomic regions marked with modified histones. For example, histones may be modified at certain lysine residues: H3K20me3, H3K27ac, H3K4me3, H3K79me2, H3K36me3, H3K9me2, and H3K9me3. Certain histone modifications mark regions of the genome that are available for binding by signaling molecules. For example, previous studies have observed that active enhancer regions include nucleosomes with H3K27ac, and active promoters include nucleosomes with H3K27ac. Further, transcribed genes include nucleosomes with H3K79me2. ChIP-MS may be performed to identify chromatin regulator proteins associated with specific histone modification. ChIP-seq with antibodies specific to certain modified histones may also be used to identify regions of the genome that are bound by signaling molecules. In some embodiments, chromatin modifying enzymes or proteins may be used or targeted, to alter or elucidate the gene signaling networks of the present disclosure.
RNAs derived from regulatory sequence regions
[0025] Many active regulatory sequence regions (RSRs), such as enhancers, signaling centers, and promoters of protein-coding genes, are known to produce non-coding RNAs. Transcripts produced at or in the vicinity of active regulatory sequence regions have been implicated in transcription regulation of nearby genes. Recent reports have demonstrated that enhancer-associated RNAs (eRNAs) are strong indicators of enhancer activity (See Li et ak, Nat Rev Genet. 2016 Apr;l7(4):207-23, which is hereby incorporated by reference in its entirety). Further, non-coding RNAs from active regulatory sequence regions have been shown to be involved in facilitating the binding of transcription factors to these regions (Sigova et ak, Science. 2015 Nov 20;350(6263):978-8l, which is hereby incorporated by reference in its entirety). This suggests that such RNAs may be important for the assembly of signaling centers and regulation of neighborhood genes. In some embodiments, RNAs derived from regulatory sequence regions of the PAH gene may be used or targeted to alter or elucidate the gene signaling networks of the present disclosure. [0026] In some embodiments, RNAs derived from regulatory sequence regions may be an enhancer-associated RNA (eRNA). In some embodiments, RNAs derived from regulatory sequence regions may be a promoter-associated RNA, including but not limited to, a promoter upstream transcript (PROMPT), a promoter-associated long RNA (PALR), and a promoter-associated small RNA (PASR). In further embodiments, RNAs derived from regulatory sequence regions may include but are not limited to transcription start sites (TSS)-associated RNAs (TSSa-RNAs), transcription initiation RNAs (tiRNAs), and terminator-associated small RNAs (TASRs).
[0027] In some embodiments, RNAs derived from regulatory sequence regions may be long non-coding RNAs (lncRNAs) (i.e., >200 nucleotides). In some embodiments, RNAs derived from regulatory sequence regions may be intermediate non-coding RNAs. (i.e., about 50 to 200 nucleotides). In some embodiments, RNAs derived from regulatory sequence regions may be short non-coding RNAs (i.e., about 20 to 50 nucleotides).
[0028] In some embodiments, eRNAs that may be modulated by methods and compounds described herein may be characterized by one or more of the following features: (1) transcribed from regions with high levels of monomethylation on lysine 4 of histone 3 (H3K4mel) and low levels of trimethylation on lysine 4 of histone 3
(H3K4me3); (2) transcribed from genomic regions with high levels of acetylation on lysine 27 of histone 3 (H3K27ac); (3) transcribed from genomic regions with low levels of trimethylation on lysine 36 of histone 3 (H3K36me3); (4) transcribed from genomic regions enriched for RNA polymerase II (Pol II); (5) transcribed from genomic regions enriched for transcriptional co-regulators, such as the p300 co-activator; (6) transcribed from genomic regions with low density of CpG island; (7) their transcription is initiated from Pol II-binding sites and elongated bidirectionally; (8) evolutionarily conserved DNA sequences encoding eRNAs; (9) short half-life; (10) reduced levels of splicing and polyadenylation, (11) dynamically regulated upon signaling; (12) positively correlated to levels of nearby mRNA expression; (13) extremely high tissue specificity; (14) preferentially nuclear and chromatin-bound; and/or (15) degraded by the exosome.
[0029] Non-limiting examples of eRNAs that may be modulated by methods and compounds described herein include those described in Djebali et al., Nature. 2012 Sep 6;489(74l4) (for example, Supplementary data file for Figure 5a) and Andersson et al., Nature. 2014 Mar 27;507(7493):455-46l (for example, Supplementary Tables S3, S12, S13, S15, and 16), which are herein incorporated by reference in their entirety. [0030] In some embodiments, promoter-associated RNAs that may be modulated by methods or compounds described herein may be characterized by one or more of the following features: (1) transcribed from regions with high levels of H3K4mel and low to medium levels of H3K4me3; (2) transcribed from genomic regions with high levels of H3K27ac; (3) transcribed from genomic regions with no or low levels of H3K36me3; (4) transcribed from genomic regions enriched for RNA polymerase II (Pol II); (5) transcribed from genomic regions with high density of CpG island; (6) their transcription is initiated from Pol II-binding sites and elongated in the opposite direction from the sense strand (that is, mRNAs) or bidirectionally; (7) short half-life; (8) reduced levels of splicing and polyadenylation; (9) preferentially nuclear and chromatin-bound; and/or (10) degraded by the exosome.
[0031] Methods and compositions described herein may be used to modulate RNAs derived from regulatory sequence regions to alter or elucidate the gene signaling networks of the present disclosure. In some embodiments, methods and compounds described herein may be used to inhibit the production and/or function of an RNA derived from regulatory sequence regions. In some embodiments, a hybridizing oligonucleotide such as an siRNA or an antisense oligonucleotide may be used to inhibit the activity of the RNA of interest via RNA interference (RNAi), or RNase H-mediated cleavage, or physically block binding of various signaling molecules to the RNA. Exemplary hybridizing oligonucleotide may include those described in U.S. 9,518,261 and WO2014/040742, which are hereby incorporated by reference in their entirety. The hybridizing oligonucleotide may be provided as a chemically modified or unmodified RNA, DNA, locked nucleic acids (LNA), or a combination of RNA and DNA, a nucleic acid vector encoding the hybridizing oligonucleotide, or a vims carrying such vector. In other embodiments, genome editing tools such as CRISPR/Cas9 may be used to delete specific DNA elements in the regulatory sequence regions that control the transcription of the RNA or degrade the RNA itself. In other embodiments, genome editing tools such as a catalytically inactive CRISPR/Cas9 may be used to bind to specific elements in the regulatory sequence regions and block the transcription of the RNA of interest. In further embodiments, bromodomain and extra terminal domain (BET) inhibitors (e.g., JQ1, 1-BET) may be used to reduce RNA transcription through inhibition of histone acetylation by BET protein Brd4.
[0032] In some embodiments, methods and compounds described herein may be used to increase the production and/or function of an RNA derived from regulatory sequence regions. In some embodiments, an exogenous synthetic RNA that mimic the RNA of interest may be introduced into the cell. The synthetic RNA may be provided as an RNA, a nucleic acid vector encoding the RNA, or a vims carrying such vector. In other
embodiments, genome editing tools such as CRISPR/Cas9 may be used to tether an exogenous synthetic RNA to specific sites in the regulatory sequence regions. Such RNA may be fused to the guide RNA of the CRISPR/Cas9 complex.
[0033] In some embodiments, modulation of RNAs derived from regulatory sequence regions increases the expression of the PAH gene. In some embodiments, modulation of RNAs derived from regulatory sequence regions reduces the expression of the PAH gene.
[0034] In some embodiments, RNAs modulated by compounds described herein include RNAs derived from regulatory sequence regions of the PAH gene in a liver cell (e.g., hepatocytes).
Perturbation of genomic systems
[0035] Behavior of one or more components of the gene signaling networks (GSNs), genomic signaling centers (GSCs), and/or insulated neighborhoods (INs) related to PAH as described herein may be altered by contacting the systems containing such networks, centers and/or neighborhoods with a perturbation stimulus. Potential stimuli may include exogenous biomolecules such as small molecules, antibodies, proteins, peptides, lipids, fats, nucleic acids, and the like or environmental stimuli such as radiation, pH, temperature, ionic strength, sound, light and the like.
[0036] The present disclosure serves, not only as a discovery tool for the elucidation of better defined gene signaling networks (GSNs) and consequently a better understanding of biological systems. The present disclosure allows the ability to properly define gene signaling for PAH at the gene level in a manner which allows the prediction, a priori, of potential treatment outcomes, the identification of novel compounds or targets which may have never been implicated in the treatment of a PAH-related disease or condition, reduction or removal of one or more treatment liabilities associated with new or known drugs such as toxicity, poor half-life, poor bioavailability, lack of or loss of efficacy or pharmacokinetic or pharmacodynamic risks.
[0037] Treatment of disease by altering gene expression of canonical cellular signaling pathways has been shown to be effective. Even small changes in gene expression may have a significant impact on disease. For example, changes in signaling centers leading to signaling pathways affecting cell suicide suppression are associated with disease. The present disclosure, by elucidating a more definitive set of connectivities of the GSNs provides a fine-tuned mechanism to address disease, including genetic diseases. A method of treating a disease may include modifying a signaling center that is involved in a gene associated with that disease. Such genes may not presently be associated with the disease except as is elucidated using the methods described herein.
[0038] A perturbation stimulus may be a small molecule, a known drug, a biological, a vaccine, an herbal preparation, a hybridizing oligonucleotide (e.g., siRNA and antisense oligonucleotide), a gene or cell therapy product, or other treatment product.
[0039] In some embodiments, methods of the present disclosure include applying a perturbation stimulus to perturb GSNs, genomic signaling centers, and/or insulated neighborhoods associated with the PAH gene. Perturbation stimuli that cause changes in PAH gene expression may inform the connectivities of the associated GSNs and provide potential targets and/or treatments for phenylalanine hydroxylase deficiency such as phenylketonuria.
Downstream targets
[0040] In certain embodiments, a stimulus is administered that targets a downstream product of a gene of a gene signaling network. Alternatively, the stimulus disrupts a gene signaling network that affects downstream expression of at least one downstream target. In some embodiments, the gene is PAH.
mRNA
[0041] Perturbation of a single or multiple gene signaling network (GSN) associated with a single insulated neighborhood or across multiple insulated neighborhoods can affect the transcription of a single gene or a multiple set of genes by altering the boundaries of the insulated neighborhood due to loss of anchor sites comprising cohesins. Perturbation stimuli may result in the modification of the RNA expression and/or the sequences in the primary transcript within the mRNA, i.e. the exons or the RNA sequences between the exons that are removed by splicing, i.e. the introns. Such changes may consequently alter the members of the set of signaling molecules within the gene signaling network of a gene, thereby defining a variant of the gene signaling network.
Proteins
[0042] Perturbation of a single or multiple gene signaling networks associated with a single insulated neighborhood or across multiple insulated neighborhoods can affect the translation of a single gene or a multiple set of genes that are part of the genomic signaling center, as well as those downstream to the genomic signaling center. Perturbation might result in the inhibition of the translated protein.
Nearest neighbor gene
[0043] Perturbation stimuli may cause interactions with signaling molecules to occur in order to alter expression of the nearest primary neighborhood gene that may be located upstream or downstream of the primary neighborhood gene. Neighborhood genes may have any number of upstream or downstream genes along the chromosome. Within any insulated neighborhood, there may be one or more, e.g., one, two, three, four or more, upstream and/or downstream neighborhood genes relative to the primary neighborhood gene. A“primary neighborhood gene” is a gene which is most commonly found within a specific insulated neighborhood along a chromosome. An upstream neighborhood gene of a primary neighborhood gene may be located within the same insulated neighborhood as the primary neighborhood gene. A downstream neighborhood gene of a primary neighborhood gene may be located within the same insulated neighborhood as the primary neighborhood gene.
Canonical cell signaling pathways
[0044] It is understood that there may, and most likely will, be some overlap between the canonical pathways detailed in the art and the gene signaling networks (GSNs) defined herein.
[0045] Whereas canonical pathways permit a certain degree of promiscuity of members across pathways (cross talk), gene signaling networks (GSN) of the disclosure are defined at the gene level and characterized based on any number of stimuli or perturbation to the cell, tissue, organ or organ system expressing that gene. Hence the nature of a GSN is both structurally (e.g., the gene) and situationally (e.g., the function, e.g., expression profile) defined. And while two different gene signaling networks may share members, they are still unique in that the nature of the perturbation can distinguish them. Hence, the value of gene signaling networks in the elucidation of the function of biological systems in support of therapeutic research and development.
[0046] It should be understood that it is not intended that no connection ever be made between canonical pathways and gene signaling networks; in fact, the opposite is the case. In order to bridge the two signaling paradigms for further scientific insights, it will be instructive to compare the canonical signaling pathway paradigm with the gene signaling networks of the present disclosure.
[0047] In some embodiments, methods of the present disclosure involve altering the Janus kinases (JAK)/signal transducers and activators of transcription (STAT) pathway. The JAK/STAT pathway is the major mediator for a wide array of cytokines and growth factors. Cytokines are regulatory molecules that coordinate immune responses. JAKs are a family of intracellular, nonreceptor tyrosine kinases that are typically associated with cell surface receptors such as cytokine receptors. Mammals are known to have 4 JAKs: JAK1, JAK2, JAK3, and Tyrosine kinase 2 (TYK2). Binding of cytokines or growth factors to their respective receptors at the cell surface initiates trans-phosphorylation of JAKs, which activates downstream STATs. STATs are latent transcription factors that reside in the cytoplasm until activated. There are seven mammalian STATs: STAT1, STAT2, STAT3, STAT4, STAT5 (STAT5A and STAT5B), and STAT6. Activated STATs translocate to the nucleus where they complex with other nuclear proteins and bind to specific sequences to regulate the expression of target genes. Thus the JAK/STAT pathway provides a direct mechanism to translate an extracellular signal into a transcriptional response. Target genes regulated by JAK/STAT pathway are involved in immunity, proliferation, differentiation, apoptosis and oncogenesis. Activation of JAKs may also activate the phosphatidylinositol 3 -kinase (PI3K) and mitogen- activated protein kinase (MAPK) pathways.
[0048] In some embodiments, methods of the present disclosure involve altering the mitogen-activated protein kinase (MAPK) signaling pathway. The MAPK pathway involves a chain of signaling molecules (e.g., Ras, Raf, MEK, and ERK) in the cell that communicates a signal from a receptor at the cell membrane to the nucleus. This pathway can be activated by receptor- linked tyrosine kinases such as epidermal growth factor receptor (EGFR), Trk A/B, Fibroblast growth factor receptor (FGFR) and PDGFR. The MAPK signaling pathway is essential in regulating numerous cellular processes including cell stress response, cell differentiation, cell division, cell proliferation, inflammation, metabolism, motility and apoptosis. MAPK interacts with major pathway targets: ERK1/2, ERK5, JNK, and p38 kinase. MAPK regulates the activities of several transcription factors including C-myc, CREB and C-Fos. MAPK also interacts with other pathways such as the PI3K networks, NF-kB and JAK/STAT pathways.
[0049] In some embodiments, methods of the present disclosure involve altering the Platelet-derived Growth Factor Receptor (PDGFR)-mediated signal pathway. PDGFRs are cell surface tyrosine kinase receptors for members of the platelet-derived growth factor (PDGF) family. There are two isoforms of PDGFRs, PDGFRa and PDGFRp. The two receptor isoforms dimerize upon binding the PDGF dimer, leading to the activation of the kinase. PDGFRs mediate a number of signaling pathways that are important for regulating cell proliferation, cellular differentiation, cell growth and development. Inhibition of the PDGFR-mediated signaling pathway has been correlated with reduced expression of PDGF, angl/2, and VEGF mRNA. Since PDGF is a known stimulus for PI3-K activation, inhibiting PDGFR may lead to decreased activation of the PI3-K signaling cascade. The role of PDGFs and PDGFRs in physiology and medicine is reviewed in Andrae et ak,
Genes Dev. 2008 May 15;22(10): 1276-312, which is hereby incorporated by reference in its entirety.
[0050] Other canonical pathways which may also be altered according to the present disclosure include, but are not limited to the 2-arachidonoylglycerol biosynthesis pathway, 2-oxocarboxylic acid metabolism pathway, 5HT1 type receptor mediated signaling pathway, 5HT2 type receptor mediated signaling pathway, 5HT3 type receptor mediated signaling pathway, 5HT4 type receptor mediated signaling pathway, 5 -hydroxy tryptamine biosynthesis pathway, 5 -hydroxy tryptamine degradation pathway, abacavir transport and metabolism pathway, ABC transporters pathway, ABC-family proteins mediated transport pathway, ACE inhibitor pathway, acetate utilization pathway, acetylcholine synthesis pathway, activation of camp-dependent PKA pathway, activin beta signaling pathway, adenine and hypoxanthine salvage pathway, adherens junction pathway, adipocytokine signaling pathway, adipogenesis pathway, adrenaline and noradrenaline biosynthesis pathway, adrenergic signaling in cardiomyocytes pathway, advanced glycation end-products (age/rage) pathway, advanced glycosylation end product receptor signaling pathway, aflatoxin bl metabolism pathway, age/rage pathway, AHR pathway, AKT signaling pathway, alanine and aspartate metabolism pathway, alanine biosynthesis pathway, aldosterone synthesis and secretion pathway, aldosterone-regulated sodium reabsorption pathway, allantoin degradation pathway, allograft rejection pathway, all-trans- retinoic acid signaling pathway, alp23b signaling pathway, alpha 6 beta 4 signaling pathway, alpha adrenergic receptor signaling pathway, alpha6 beta4 integrin pathway, alpha- linolenic acid metabolism pathway, Alzheimer disease-amyloid secretase pathway, Alzheimer disease-presenilin pathway, amino acid conjugation pathway, amino sugar and nucleotide sugar metabolism pathway, aminoacyl-tRNA biosynthesis pathway, aminobutyrate degradation pathway, AMP-activated protein kinase pathway, AMPK signaling pathway, anandamide biosynthesis pathway, anandamide degradation pathway, androgen receptor signaling pathway, androgen/estrogen/progesterone biosynthesis pathway, angiogenesis pathway, angiopoietin-Tie2 signaling pathway, angiotensin II- stimulated signaling through g proteins and beta-arrestin pathway, antigen processing and presentation by MHC's pathway, apoptosis modulation and signaling pathway, apoptosis modulation by HSP70 pathway, apoptosis signaling pathway, apoptosis through death receptors pathway, apoptotic execution phase pathway, arachidonate epoxygenase/epoxide hydrolase pathway, arachidonic acid metabolism pathway, arginine and proline metabolism pathway, arginine biosynthesis pathway, aripiprazole metabolic pathway, arylamine metabolism pathway, ascorbate and aldarate metabolism pathway, ascorbate degradation pathway, asparagine and aspartate biosynthesis pathway, asparagine N-linked
glycosylation pathway, aspartate and glutamate metabolism pathway, assembly of RNA polymerase-II initiation complex pathway, ATM pathway, ATP synthesis pathway, axon guidance pathway, axon guidance mediated by netrin pathway, axon guidance mediated by semaphorins pathway, axon guidance mediated by slit/robo pathway, B cell activation pathway, B cell receptor (BCR) pathway, B cell receptor signaling pathway, bacterial invasion of epithelial cells pathway, basal transcription factors pathway, base excision repair pathway, B-cell development pathway, B-cell receptor pathway, B-cell receptor complex pathway, benzo pathway, betal adrenergic receptor signaling pathway, beta2 adrenergic receptor signaling pathway, beta3 adrenergic receptor signaling pathway, beta- alanine metabolism pathway, bile acid and bile salt metabolism pathway, bile secretion pathway, binding and uptake of ligands by scavenger receptors pathway, biogenic amine synthesis pathway, biosynthesis of amino acids pathway, biosynthesis of unsaturated fatty acids pathway, biotin biosynthesis pathway, blakely network pathway, blood clotting cascade pathway, blood coagulation pathway, bmp/activin signaling -drosophila pathway, bone morphogenic protein pathway, brain-derived neurotrophic factor (BDNF) pathway, BRCA1 pathway, bupropion degradation pathway, butanoate metabolism pathway, butirosin and neomycin biosynthesis pathway, butyrate-induced histone acetylation pathway, cadherin signaling pathway, caffeine metabolism pathway, calcium regulation in the cardiac cell pathway, calcium signaling pathway, cAMP pathway, carbohydrate digestion and absorption pathway, carbon metabolism pathway, cardiac muscle contraction pathway, cardiac progenitor differentiation pathway, carnitine metabolism pathway, caspase cascade pathway, catalytic cycle of mammalian flavin-containing monooxygenases pathway, CCKR signaling map pathway, CCR5 in macrophages pathway, CD4 and CD8 T-cell lineage pathway, CD40 signaling pathway, CDK5 pathway, cell adhesion molecules (cams) pathway, cell cycle checkpoints pathway, cell cycle pathway, cell differentiation - meta pathway, cell junction organization pathway, cell surface interactions at the vascular wall pathway, CGMP-PKG signaling pathway, chemical carcinogenesis pathway, chemokine signaling pathway, cholesterol biosynthesis pathway, cholinergic synapse pathway, chorismate biosynthesis pathway, chromatin remodeling pathway, circadian clock system pathway, circadian entrainment pathway, citrate cycle (TCA cycle) pathway, c-met pathway, cobalamin biosynthesis pathway, codeine and morphine metabolism pathway, coenzyme A biosynthesis pathway, coenzyme A linked carnitine metabolism pathway, colchicine metabolic pathway, collecting duct acid secretion pathway, complement and coagulation cascades pathway, cori cycle pathway, corticotropin-releasing hormone (CRH) pathway, costimulation by the CD28 family pathway, CREB pathway, CTL mediated apoptosis pathway, CTLA4 signaling pathway, cyanoamino acid metabolism pathway, cyclins and cell cycle regulation pathway, cysteine and methionine metabolism pathway, cysteine biosynthesis pathway, cytokine network pathway, cytokine- cytokine receptor interaction pathway, cytokines and inflammatory response pathway, cytoplasmic ribosomal proteins pathway, cytoskeletal regulation by rho GTPase pathway, cytosolic DNA-sensing pathway, de novo purine biosynthesis pathway, de novo pyrimidine deoxyribonucleotide biosynthesis pathway, de novo pyrimidine ribonucleotides biosynthesis pathway, depolarization of the presynaptic terminal triggers the opening of calcium channels pathway, diclofenac metabolic pathway, differentiation pathway, digestion resistant carbohydrate metabolism pathway, dilated cardiomyopathy pathway, dissolution of fibrin clot pathway, diurnally regulated genes with circadian orthologs pathway, div no colors pathway, div pathway, DNA damage bypass pathway, DNA damage response pathway, DNA damage reversal pathway, DNA methylation and transcriptional repression pathway, DNA repair mechanisms pathway, DNA replication pathway, dopamine metabolism pathway, dopamine receptor mediated signaling pathway, dopaminergic synapse pathway, dorso-ventral axis formation pathway, DPP signaling pathway, DPP-SCW signaling pathway, drug metabolism pathway, drug metabolism - cytochrome p450 pathway, dscam interactions pathway, E2F/MIRHG1 feedback- loop - delete pathway, EBV LMP1 signaling pathway, ECM-receptor interaction pathway, effects of nitric oxide pathway, effects of pip2 hydrolysis pathway, EGF pathway, EGF receptor signaling pathway, eicosanoid synthesis pathway, electron transport chain pathway, endochondral ossification pathway, endocrine and other factor-regulated calcium reabsorption pathway, endocytosis pathway, endoderm differentiation pathway, endogenous cannabinoid signaling pathway, endothelin pathway, endothelin signaling pathway, energy metabolism pathway, enkephalin release pathway, enos signaling pathway, ephrin-EPHR signaling pathway, epidermal growth factor receptor (EGFR) pathway, epithelial cell signaling in helicobacter pylori infection pathway, epithelial tight junctions pathway, EPO receptor signaling pathway, ERBB signaling pathway, ERK signaling pathway, erythropoietin pathway, estrogen signaling pathway, ether lipid metabolism pathway, eukaryotic transcription initiation pathway, eukaryotic translation elongation pathway, eukaryotic translation initiation pathway, eukaryotic translation termination pathway, FAK1 signaling pathway, Fas signaling pathway, fat digestion and absorption pathway, fatty acid pathway, fatty acid beta oxidation pathway, fatty acid biosynthesis pathway, fatty acid degradation pathway, fatty acid elongation pathway, fatty acid metabolism pathway, fatty acid omega oxidation pathway, FGF pathway, FGF signaling pathway, fibroblast growth factor- 1 (FGF1) pathway, flavin biosynthesis pathway, FLT3 signaling pathway, fluoropyrimidine activity pathway, focal adhesion pathway, folate biosynthesis pathway, folate metabolism pathway, follicle stimulating hormone pathway, formation of fibrin clot pathway, formyltetrahydroformate biosynthesis pathway, Foxo signaling pathway, fructose galactose metabolism pathway, G protein signaling pathway, Gl to S cell cycle control pathway, G13 signaling pathway, GABA synthesis pathway, GABA-B receptor II signaling pathway, galactose metabolism pathway, gamma-aminobutyric acid synthesis pathway, ganglio sphingolipid metabolism pathway, gap junction trafficking and regulation pathway, gastric acid secretion pathway, gastrin pathway, GBB signaling pathway, generic transcription pathway, ghrelin pathway, glial cell differentiation pathway, globo sphingolipid metabolism pathway, glucagon signaling pathway, glucocorticoid & mineralcorticoid metabolism pathway, glucocorticoid receptor signaling pathway, glucose homeostasis pathway, glucuronidation pathway, glutamatergic synapse pathway, glutamine glutamate conversion pathway, glutathione metabolism pathway, glycan degradation pathway, glycerolipid metabolism pathway,
glycerophospholipid biosynthetic pathway, glycerophospholipid metabolism pathway, glycine metabolism pathway, glycogen metabolism pathway, glycolysis/gluconeogenesis pathway, glycosaminoglycan biosynthesis -heparan sulfate / heparin pathway,
glycosaminoglycan biosynthesis-keratan sulfate pathway, glycosaminoglycan degradation pathway, glycosaminoglycan metabolism pathway, glycosphingolipid biosynthesis - ganglio series pathway, glycosphingolipid biosynthesis-globo series pathway,
glycosphingolipid biosynthesis - lacto and neolacto series pathway, glyoxylate and dicarboxylate metabolism pathway, gonadotropin-releasing hormone receptor pathway, GP1B-IX-V activation signaling pathway, GPCR pathway, GPCR downstream signaling pathway, GPCR ligand binding pathway, GPVI-mediated activation cascade pathway, granulocyte adhesion and diapedesis pathway, granzyme pathway, growth hormone signaling pathway, GSK 3 signaling pathway, hedgehog signaling pathway, hematopoiesis from pluripotent stem cells pathway, hematopoietic cell lineage pathway, hematopoietic stem cell differentiation pathway, heme biosynthesis pathway, hepatitis B pathway, hepatitis C pathway, heterotrimeric G-protein signaling-Gi alpha and Gs alpha mediated pathway, heterotrimeric g-protein signaling -rod outer segment phototransduction pathway, hexose transport pathway, HGF pathway, HIF-l signaling pathway, hippo signaling pathway, histamine hl receptor mediated signaling pathway, histamine h2 receptor mediated signaling pathway, histamine synthesis pathway, histidine biosynthesis pathway, histone modifications pathway, homologous recombination pathway, HTLV-I infection pathway, human complement system pathway, hypoxia response via hif activation pathway, ID signaling pathway, IGF1R signaling pathway, IL1 and megakaryocytes in obesity pathway, IL-l signaling pathway, IL-10 pathway, IL17 signaling pathway, IL-2 signaling pathway, IL-22 pathway, IL-3 signaling pathway, IL-4 signaling pathway, IL-5 signaling pathway, IL-6 pathway, IL-7 signaling pathway, IL-9 signaling pathway, ILK signaling pathway, inflammation mediated by chemokine and cytokine signaling pathway, inflammatory mediator regulation of Trp channels pathway, inflammatory response pathway, influenza a virus infection pathway, inos signaling pathway, inositol phosphate metabolism pathway, insulin receptor pathway, insulin resistance pathway, insulin secretion pathway, insulin/IGF-protein kinase b signaling cascade pathway, insulin-like growth factor-2 mRNA binding proteins pathway, integrin alphallb beta3 signaling pathway, integrin cell signaling pathway, integrin cell surface interactions pathway, integrin-mediated cell adhesion pathway, interferon pathway, interferon alpha/beta signaling pathway, interferon type I signaling pathway, interferon-gamma signaling pathway, interleukin signaling pathway, interleukin- 1 (IL-l) pathway, interleukin-1 processing pathway, interleukin- 11 signaling pathway, interleukin-2 (IL-2) pathway, interleukin-3 pathway, interleukin-4 (IL-4) pathway, interleukin-5 (IL-5) pathway, interleukin-6 (IL-6) pathway, interleukin-7 (IL-7) pathway, interleukin-9 (il-9) pathway, intracellular calcium signaling pathway, ionotropic glutamate receptor pathway, IP3 pathway, isoleucine biosynthesis pathway, JAK/STAT pathway, JNK pathway, kinesins pathway, KIT receptor pathway, LDL oxidation in atherogenesis pathway, leptin (lep) pathway, leptin signaling pathway, leucine biosynthesis pathway, leukocyte transendothelial migration pathway, linoleic acid metabolism pathway, lipid digestion pathway, lipoate_biosynthesis pathway, longevity regulating - mammal pathway, longevity regulating - multiple species pathway, long-term potentiation pathway, lysine biosynthesis pathway, lysine degradation pathway, lysosome pathway, mannose metabolism pathway, MAPK cascade pathway, MAPK targets/ nuclear events mediated by MAP kinases pathway, matrix metalloproteinases pathway, melatonin metabolism and effects pathway, meta biotransformation pathway, metabolism of carbohydrates pathway, metabolism of nitric oxide pathway, metabolism of nucleotides pathway, metabolism of porphyrins pathway, metabolism of water-soluble vitamins and cofactors pathway, metabolism of xenobiotics by cytochrome p450 pathway, metabotropic glutamate receptor group I pathway, metabotropic glutamate receptor group II pathway, metabotropic glutamate receptor group III pathway, methionine biosynthesis pathway, methylation pathway, methylcitrate cycle pathway, methylmalonyl pathway, mineral absorption pathway, miRNA biogenesis pathway, mismatch repair pathway, mitochondrial apoptosis pathway, mitochondrial gene expression pathway, mitochondrial lc-fatty acid beta-oxidation pathway, mitotic Gl-Gl/S phases pathway, mitotic G2-G2/M phases pathway, monoamine GPCRs pathway, monoamine transport pathway, mRNA capping pathway, mRNA editing pathway, mRNA processing pathway, mRNA splicing pathway, mRNA surveillance pathway, mTOR signaling pathway, muscarinic acetylcholine receptor 1 and 3 signaling pathway, muscarinic acetylcholine receptor 2 and 4 signaling pathway, myogenesis pathway, myometrial relaxation and contraction pathway, N-acetylglucosamine metabolism pathway, NAD biosynthesis II pathway, nanomaterial induced apoptosis pathway, nanoparticle triggered autophagic cell death pathway, nanoparticle triggered regulated necrosis pathway, natural killer cell mediated cytotoxicity pathway, ncam signaling for neurite out-growth pathway, nephrin interactions pathway, netrin- 1 signaling pathway, neural crest differentiation pathway, neuroactive ligand-receptor interaction pathway, neurotransmitter clearance in the synaptic cleft pathway, neurotransmitter release cycle pathway, neurotransmitter uptake and metabolism in glial cells pathway, neurotrophin signaling pathway, NFAT and cardiac hypertrophy pathway, NF-kappa b signaling pathway, NF-kappa b signaling pathway, NGF pathway, NGF signaling via TRKA from the plasma membrane pathway, N-glycan biosynthesis pathway, nicotinate and nicotinamide metabolism pathway, nicotine activity on chromaffin cells pathway, nicotine activity on dopaminergic neurons pathway, nicotine degradation pathway, nicotine metabolism pathway, nicotine pharmacodynamics pathway, nicotinic acetylcholine receptor signaling pathway, nifedipine activity pathway, nitrogen metabolism pathway, NLR proteins pathway, nod-like receptor signaling pathway, non-homologous end joining pathway, notch signaling pathway, Nrf2 pathway, nuclear receptors pathway, nucleosome assembly pathway, nucleotide excision repair pathway, nucleotide GPCRs pathway, nucleotide metabolism pathway, nucleotide-binding oligomerization domain pathway, o- antigen biosynthesis pathway, o-glycan biosynthesis pathway, olfactory transduction pathway, oncostatin m signaling pathway, one carbon metabolism pathway, opioid prodynorphin pathway, opioid proenkephalin pathway, opioid proopiomelanocortin pathway, ornithine degradation pathway, osteoblast signaling pathway, osteoclast signaling pathway, osteopontin signaling pathway, ovarian steroidogenesis pathway, oxidation by cytochrome p450 pathway, oxidative phosphorylation pathway, oxidative stress pathway, oxytocin receptor mediated signaling pathway, oxytocin signaling pathway, p38 MAPK signaling pathway, p53 feedback loops 1 pathway, p53 feedback loops 2 pathway, p53 mediated apoptosis pathway, p53 signaling pathway, pak pathway, pancreatic secretion pathway, pantothenate biosynthesis pathway, parkin-ubiquitin proteasomal system pathway, passive transport by aquaporins pathway, PDGF signaling pathway, pentose and glucuronate interconversions pathway, pentose phosphate pathway, peptide GPCRs pathway, peptidoglycan biosynthesis pathway, peroxisomal beta-oxidation of
tetracosanoyl-coA pathway, peroxisomal lipid metabolism pathway, pertussis pathway, phagosome pathway, phase 1 -functionalization of compounds pathway, phase I biotransformations pathway, phase II conjugation pathway, phenylacetate degradation pathway, phenylalanine biosynthesis pathway, phenylalanine metabolism pathway, phenylethylamine degradation pathway, phenylpropionate degradation pathway, phosphatidylinositol signaling system pathway, phospholipase D signaling pathway, phototransduction pathway, PI3 kinase pathway, PI3K signaling in B -lymphocytes pathway, PI3K-AKT signaling pathway, PIP3 activates AKT signaling pathway, plasminogen activating cascade pathway, platelet activation pathway, platelet adhesion to exposed collagen pathway, platelet aggregation pathway, platelet homeostasis pathway, polyol pathway, porphyrin and chlorophyll metabolism pathway, PPAR signaling pathway, primary bile acid biosynthesis pathway, primary focal segmental glomerulosclerosis FSGs pathway, processing of capped intron-containing pre-mRNA pathway, processing of capped intronless pre-mRNA pathway, progesterone-mediated oocyte maturation pathway, prolactin signaling pathway, proline biosynthesis pathway, propanoate metabolism pathway, prostaglandin synthesis and regulation pathway, proteasome pathway, proteasome degradation pathway, protein digestion and absorption pathway, protein export pathway, protein folding pathway, proximal tubule bicarbonate reclamation pathway,
PRPP biosynthesis pathway, PTEN pathway, purine metabolism pathway, pyridoxal phosphate salvage pathway, pyridoxal-5 -phosphate biosynthesis pathway, pyrimidine metabolism pathway, pyruvate metabolism pathway, racl pathway, rank signaling in osteoclast pathway, rankl/rank pathway, rapl signaling pathway, ras signaling pathway, Ras-RAF-MEK-ERK pathway, receptor activator of nuclear factor kappa-b ligand
(RANKL) pathway, regulation of actin cytoskeleton pathway, regulation of
apoptosis pathway, regulation of autophagy pathway, regulation of DNA replication pathway, regulation of lipolysis in adipocytes pathway, regulation of microtubule cytoskeleton pathway, regulation of toll-like receptor signaling pathway, remodeling of adherens junctions pathway, renin secretion pathway, renin-angiotensin system pathway, respiratory electron transport pathway, retinol metabolism pathway, retrograde
endocannabinoid signaling pathway, Rho family GTPase pathway, Rhoa pathway, ribosome biogenesis in eukaryotes pathway, RIG-I-like receptor signaling pathway, RNA degradation pathway, RNA polymerase I pathway, RNA polymerase II transcription pathway, RNA transport pathway, RNAi pathway, s-adenosylmethionine biosynthesis pathway, salivary secretion pathway, salvage pyrimidine deoxyribonucleotides pathway, salvage pyrimidine ribonucleotides pathway, SCW signaling pathway, selenium metabolism and selenoproteins pathway, selenium micronutrient network pathway, selenocompound metabolism pathway, semaphorin interactions pathway, serine and threonine metabolism pathway, serine glycine biosynthesis pathway, serotonergic synapse pathway, serotonin htrl group and fos pathway, serotonin receptor 2 and ELK-SRF/gata4 signaling pathway, serotonin receptor 2 and STAT3 signaling pathway, serotonin receptor 4/6/7 and NR3C signaling pathway, serotonin transporter activity pathway, signal amplification pathway, signal regulatory protein pathway, signal transduction of S1P receptor pathway, signaling by EGFR pathway, signaling by insulin receptor pathway, signaling by PDGF pathway, signaling by rho GTPases pathway, signaling by robo receptor pathway, signaling by VEGF pathway, signaling in gap junction pathway, signaling of hepatocyte growth factor receptor pathway, signaling regulating pluripotency of stem cells pathway, signaling in glioblastoma pathway, signaling by NGF pathway, SMAD signaling network pathway, small ligand GPCRs pathway, SNARE interactions in vesicular transport pathway, sphingolipid (SM) signaling pathway, sphingolipid metabolism pathway, spliceosome pathway, starch and sucrose metabolism pathway, stat signaling pathway, STAT3 pathway, statin pathway, steroid biosynthesis pathway, steroid hormone biosynthesis pathway, sterol regulatory element-binding proteins pathway, striated muscle contraction pathway, succinate to proprionate conversion pathway, sulfate assimilation pathway, sulfation biotransformation reaction pathway, sulfur metabolism pathway, sulfur relay system pathway, sumo pathway, synaptic vesicle pathway, synthesis and degradation of ketone bodies pathway, synthesis of DNA pathway, T cell receptor (TCR) pathway, tamoxifen metabolism pathway, tarbase pathway, target of rapamycin pathway, taste transduction pathway, taurine and hypotaurine metabolism pathway, TCA and urea cycles pathway, T-cell antigen receptor pathway, T-cell receptor and co stimulatory signaling pathway, telomere maintenance pathway, terpenoid backbone biosynthesis pathway, tetrahydrofolate biosynthesis pathway, TFS regulate miRNAs related to cardiac hypertrophy pathway, TGF-beta pathway, TGF-beta receptor signaling pathway, THC differentiation pathway, thiamin biosynthesis pathway, thiamin metabolism pathway, threonine biosynthesis pathway, thymic stromal lymphopoietin pathway, thymic stromal lymphopoietin (tslp) pathway, thyroid-stimulating hormone (tsh) pathway, thyrotropin-releasing hormone receptor signaling pathway, tie2/tek signaling pathway, tight junction pathway, TNF alpha signaling pathway, TNF related weak inducer of apoptosis pathway, TNF signaling pathway, TNF superfamily pathway, TNF-related weak inducer of apoptosis (tweak) pathway, toll receptor signaling pathway, toll-like receptors pathway, TP53 network pathway, Traf pathway, trail pathway, transcription regulation by bzip transcription factor pathway, transcriptional activation by Nrf2 pathway,
transendothelial migration of leukocytes pathway, transforming growth factor beta (TGF- beta) receptor pathway, translation factors pathway, transmission across electrical synapses pathway, transport of glucose and other sugars pathway, transport of glycerol from adipocytes to the liver by aquaporins pathway, transport of vitamins pathway, trans- sulfuration pathway, trans-sulfuration and one carbon metabolism pathway,
triacylglyceride synthesis pathway, triacylglycerol metabolism pathway, tRNA
aminoacylation pathway, tryptophan biosynthesis pathway, tryptophan metabolism pathway, tumor necrosis factor (TNF) alpha pathway, tumoricidal effects of hepatic NK cells pathway, tweak pathway, type II diabetes mellitus pathway, type II interferon signaling pathway, type III interferon signaling pathway, tyrosine and tryptophan biosynthesis pathway, tyrosine biosynthesis pathway, tyrosine metabolism pathway, ubiquinone and other terpenoid-quinone biosynthesis pathway, ubiquitin mediated proteolysis pathway, ubiquitin proteasome pathway, unfolded protein response pathway, urea cycle and metabolism of amino groups pathway, valine biosynthesis pathway, vascular smooth muscle contraction pathway, vasopressin synthesis pathway, vasopressin- regulated water reabsorption pathway, VEGF signaling pathway, vitamin a and carotenoid metabolism pathway, vitamin bl2 metabolism pathway, vitamin b6 biosynthesis pathway, vitamin b6 metabolism pathway, vitamin d metabolism pathway, vitamin digestion and absorption pathway, Wnt signaling pathway, xanthine and guanine salvage pathway, and/or the zinc homeostasis pathway.
Additional Definitions
[0051] The term“analog”, as used herein, refers to a compound that is structurally related to the reference compound and shares a common functional activity with the reference compound.
[0052] The term“biologic”, as used herein, refers to a medical product made from a variety of natural sources such as micro-organism, plant, animal, or human cells.
[0053] The term“boundary”, as used herein, refers to a point, limit, or range indicating where a feature, element, or property ends or begins.
[0054] The term“compound”, as used herein, refers to a single agent or a
pharmaceutically acceptable salt thereof, or a bioactive agent or drug.
[0055] The term“derivative”, as used herein, refers to a compound that differs in structure from the reference compound, but retains the essential properties of the reference molecule. [0056] The term“downstream neighborhood gene”, as used herein, refers to a gene downstream of primary neighborhood gene that may be located within the same insulated neighborhood as the primary neighborhood gene.
[0057] The term“gene”, as used herein, refers to a unit or segment of the genomic architecture of an organism, e.g., a chromosome. Genes may be coding or non-coding. Genes may be encoded as contiguous or non-contiguous polynucleotides. Genes may be DNA or RNA.
[0058] The term "genomic system architecture", as used herein, refers to the organization of an individual’s genome and includes chromosomes, topologically associating domains (TADs), and insulated neighborhoods.
[0059] The term“master transcription factor”, as used herein, refers to signaling molecules which alter, whether to increase or decrease, the transcription of a target gene, e.g., a neighborhood gene and establish cell-type specific enhancers. Master transcription factors recruit additional signaling proteins, such as other transcription factors to enhancers to form signaling centers.
[0060] The term“modulate”, as used herein, refers to an alteration (e.g., increase or decrease) in the expression of the target gene and/or activity of the gene product.
[0061] The term“penetrance”, as used herein, refers to the proportion of individuals carrying a particular variant of a gene (e.g., mutation, allele or generally a genotype, whether wild type or not) that also exhibits an associated trait (phenotype) of that variant gene and in some situations is measured as the proportion of individuals with the mutation who exhibit clinical symptoms thus existing on a continuum.
[0062] The term“polypeptide”, as used herein, refers to a polymer of amino acid residues (natural or unnatural) linked together most often by peptide bonds. The term, as used herein, refers to proteins, polypeptides, and peptides of any size, structure, or function. In some instances, the polypeptide encoded is smaller than about 50 amino acids and the polypeptide is then termed a peptide. If the polypeptide is a peptide, it will be at least about 2, 3, 4, or at least 5 amino acid residues long.
[0063] The term“primary neighborhood gene” as used herein, refers to a gene which is most commonly found within a specific insulated neighborhood along a chromosome.
[0064] The term“primary downstream boundary”, as used herein, refers to the insulated neighborhood boundary located downstream of a primary neighborhood gene. [0065] The term“primary upstream boundary”, as used herein, refers to the insulated neighborhood boundary located upstream of a primary neighborhood gene.
[0066] The term“promoter” as used herein, refers to a DNA sequence that defines where transcription of a gene by RNA polymerase begins and defines the direction of
transcription indicating which DNA strand will be transcribed.
[0067] The term“regulatory sequence regions”, as used herein, include but are not limited to regions, sections or zones along a chromosome whereby interactions with signaling molecules occur in order to alter expression of a neighborhood gene.
[0068] The term“repressor”, as used herein, refers to any protein that binds to DNA and therefore regulates the expression of genes by decreasing the rate of transcription.
[0069] The term“secondary downstream boundary”, as used herein, refers to the downstream boundary of a secondary loop within a primary insulated neighborhood.
[0070] The term“secondary upstream boundary”, as used herein, refers to the upstream boundary of a secondary loop within a primary insulated neighborhood.
[0071] The term“signaling molecule”, as used herein, refers to any entity, whether protein, nucleic acid (DNA or RNA), organic small molecule, lipid, sugar or other biomolecule, which interacts directly, or indirectly, with a regulatory sequence region on a chromosome.
[0072] The term“signaling transcription factor”, as used herein, refers to signaling molecules which alter, whether to increase or decrease, the transcription of a target gene, e.g., a neighborhood gene and also act as cell-cell signaling molecules.
[0073] The term“small molecule”, as used herein, refers to a low molecular weight drug, i.e. <5000 Daltons organic compound that may help regulate a biological process.
[0074] The terms“subject” and“patient” are used interchangeably herein and refer to an animal to whom treatment with the compositions according to the present disclosure is provided.
[0075] The term“therapeutic agent”, as used herein, refers to a substance that has the ability to cure a disease or ameliorate the symptoms of the disease.
[0076] The term“therapeutic or treatment outcome”, as used herein, refers to any result or effect (whether positive, negative or null) which arises as a consequence of the perturbation of a GSC or GSN. Examples of therapeutic outcomes include, but are not limited to, improvement or amelioration of the unwanted or negative conditions associated with a disease or disorder, lessening of side effects or symptoms, cure of a disease or disorder, or any improvement associated with the perturbation of a GSC or GSN.
[0077] The term“therapeutic or treatment liability”, as used herein, refers to a feature or characteristic associated with a treatment or treatment regime which is unwanted, harmful or which mitigates the therapies positive outcomes. Examples of treatment liabilities include for example toxicity, poor half-life, poor bioavailability, lack of or loss of efficacy or pharmacokinetic or pharmacodynamic risks.
[0078] The term“upstream neighborhood gene”, as used herein, refers to a gene upstream of a primary neighborhood gene that may be located within the same insulated neighborhood as the primary neighborhood gene.
II. THE PAH GENE AND PAH DEFICIENCY
[0079] In some embodiments, methods of the present disclosure involve modulating the expression of PAH gene. The PAH gene encodes phenylalanine hydroxylase which is a metabolic enzyme that converts of phenylalanine to tyrosine. PAH may also be referred to as Phenylalaninase, Phenylalanine 4-Monooxygenase, Phe-4-Monooxygenase, EC
1.14.16.1, Phenylalanine-4-Hydroxylase, or PH. The PAH gene has a cytogenetic location of l2q23.2 and is located on Chromosome 12 at position 102,836,885-102,958,410 on the reverse strand. PAH has a NCBI gene ID of 5053, Uniprot ID of P00439 and Ensembl Gene ID of ENSG00000171759.
[0080] PAH is primarily expressed in the liver, kidney, nervous system, gall bladder, and blood. More than 900 pathogenic variants have been described in PAH. Information on the pathogenic variants, associated phenotypes, gene structure, and enzyme structure can be found on the Phenylalanine Hydroxylase Locus Knowledgebase (PAHdb).
[0081] The majority of PAH mutations lead to deficiency in phenylalanine hydroxylase activity, a condition known as phenylalanine hydroxylase deficiency. As used herein, the term“phenylalanine hydroxylase deficiency” or“PAH deficiency” refers to any condition or disorder that is manifested in an elevated phenylalanine level in the blood. PAH deficiency can be detected in newborn screening using methods well known in the art (e.g., Guthrie microbial inhibition assay) based on the presence of hyperphenylalaninemia on a blood spot obtained from a heel prick.
[0082] Deficiency of phenylalanine hydroxylase results in a spectrum of disorders including mild hyperphenylalaninemia, mild phenylketonuria, and classic phenylketonuria. As used herein, the term“mild hyperphenylalaninemia” or“non-PKU hyperphenylalaninemia” is defined as the presence of plasma phenylalanine levels that exceed the limits of the upper reference range (120 pmol/L or 2 mg/dL) without treatment but that are below the level found in patients with PKU.“Mild PKU” or“moderate PKU” refers to conditions with plasma phenylalanine levels over 600 pmol/L (10 mg/dL) but lower than 1200 pmol/L (20 mg/dL) without treatment. Plasma phenylalanine levels that exceed 1200 pmol/L (20 mg/dL) are classified as“classic PKU”, which is the most severe form of PKU.“Mild PKU” or“classic PKU” are herein broadly referred to as“PKU.”
[0083] While classic PKU is caused by the absence or severe deficiency of phenylalanine hydroxylase, milder versions of PKU, or mild hyperphenylalaninemia, are often due to partial deficiency of the PAH enzyme. In some embodiments, methods and compositions of the present disclosure increase the levels of partially functional PAH protein to breakdown phenylalanine and prevent or treat PKU.
[0084] In some embodiments, methods of the present disclosure involve altering the composition and/or the structure of the insulated neighborhood containing the PAH gene. The present inventors have identified the insulated neighborhood containing the PAH gene in primary human hepatocytes. The insulated neighborhood containing the PAH gene is approximately 668 kb in length and contains 14 signaling centers. The insulated neighborhood contains PAH and 2 other genes, namely ASCL1 and C120RF42, both of which are located downstream of PAH. The chromatin marks and/or chromatin-associated proteins include H3K27Ac, BRD4, p300, and SMC1. The transcription factors include FOXA2, HNF4A, ONECUT1, ONECUT2, YY1, and HNF1A. The signaling proteins include TCF7L2, ESR1, HIF1A, FOS, NR3C1, JUN, NF-kB, RBPJ, RXR, STAT3, NR1I1, SMAD2/3, SMAD1, SMAD4, STAT1, TEAD1, and TP53. Any components of these signaling centers and/or signaling molecules, or any regions within or near the insulated neighborhood, may be targeted or altered to change the composition and/or structure of the insulated neighborhood, thereby modulating the expression of PAH.
III. PAH and GTP cyclohydrolase I (GCH1)
[0085] Shown in FIG. 9 is the BH4 syhtnesis pathway. GCH1 is the rate limiting enzyme in BH4 synthesis. This patheway is subject to allosteric feed-forward activation by L- phenylalanine and feedback inhibition by BH4. GCH1 is not liver specific, rather it is expressed in multiple cell types. An advantage of targeting GCH1 is a concomitant increase in PAH activity by increasing the BH4 levels. This will lead to increase in intracellular BH4. Modulating the BH4 pathway will result in greater treatment opportunities to rare PKU patients with mutations in BH4 pathway such as 6-pyruvoyl- tetrahydropterin synthase deficiency (PTPS), GTP cyclohydrolase I (GTPCH), and Dihydropteridine reductase deficiency (DHPR). In addition, upregulation of GCH1 could be beneficial for a wide range of cardiovascular disorders (Cunnington, Heart 96
(2010): 1872).
[0086] Methods to increase intracellular BH4 include administering Kuvan, a synthetic BH4 which stabilizes and increases the activity of some PAH mutants; enzyme substitution therapy using pegValiase which converts phenylalanine to a non-toxic metabolite; and gene therapy. (Burnett, J. R.; Sapropterin dihydrochloride/Kuvan/phenoptin, an orally active synthetic form of BH4 for the treatment of phenylketonuria; IDrugs; Nov 2010 (11): 805-13).
III. COMPOSITIONS AND METHODS
[0087] Some embodiments, provides compositions and methods for modulating the expression of the PAH gene. Any one of the compositions and methods described herein may be used to treat or prevent a PAH-related disorder such as phenylketonuria. In some embodiments, a combination of the compositions and methods described herein may be used to treat a PAH-related disorder.
[0088] The terms“subject” and“patient” are used interchangeably herein and refer to an animal to whom treatment with the compositions according to the present disclosure is provided. In some embodiments, the subject is a mammal. In some embodiments, the subject is a human being.
[0089] In some embodiments, subjects may have been diagnosed with or have symptoms for phenylalanine hydroxylase deficiency, e.g., mild hyperphenylalaninemia, mild PKU, or classic PKU. In other embodiments, subjects may be susceptible to or at risk for phenylalanine hydroxylase deficiency, e.g., mild hyperphenylalaninemia, mild PKU, or classic PKU.
[0090] In some embodiments, subjects may carry one or more mutations within or near the PAH gene. In some embodiment, subjects may carry one functional allele and one mutated allele of the PAH gene. In some embodiment, subjects may carry two mutated alleles of the PAH gene.
[0091] In some embodiments, subjects may have dysregulated expression of the PAH gene. In some embodiments, subjects may have a deficiency of the phenylalanine hydroxylase enzyme. In some embodiments, subjects may have a partially functional phenylalanine hydroxylase.
[0092] In some embodiments, compositions and methods may be used to increase the expression of the PAH gene in a cell or a subject. Changes in gene expression may be assessed at the RNA level or protein level by various techniques known in the art and described herein, such as RNA-seq, qRT-PCR, Western Blot, or enzyme-linked immunosorbent assay (ELISA). Changes in gene expression may be determined by comparing the level of target gene expression in the treated cell or subject to the level of expression in an untreated or control cell or subject. In some embodiments, the
compositions and methods cause an increase in the expression of the PAH gene by at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 100%, at least about 125%, at least about 150%, at least about 175%, at least about 200%, at least about 250%, at least about 300%, at least about 400%, at least about 500%, from about 25% to about 50%, from about 40% to about 60%, from about 50% to about 70%, from about 60% to about 80%, from about 80% to about 100%, from about 100% to about 125%, from about 100 to about 150%, from about 150% to about 200%, from about 200% to about 300%, from about 300% to about 400%, from about 400% to about 500% as compared to baseline or to administration of a control, or more than 500% as compared to baseline or to administration of a control. In some embodiments, the compositions and methods cause a fold change in the expression of the PAH gene by about 2 fold, about 3 fold, about 4 fold, about 5 fold, about 6 fold, about 7 fold, about 8 fold, about 9 fold, about 10 fold, about 12 fold, about 15 fold, about 18 fold, about 20 fold, about 25 fold, or more than 30 fold as compared to baseline or to administration of a control.
[0093] In some embodiments, the increase in the expression of the PAH gene induced by compositions and methods of the present disclosure may be sufficient to prevent or alleviate one or more signs or symptoms of phenylalanine hydroxylase deficiency in a subject. In some embodiments, administering to a subject with phenylalanine hydroxylase deficiency compositions and methods of the present disclosure may result in reduction of blood phenylalanine levels in the subject to below 120 pmol/L, below 240 pmol/L, below 360 pmol/L, below 480 pmol/L, below 600 pmol/L, below 720 pmol/L, from about 120 pmol/L to about 360 pmol/L, from about 240 pmol/L to about 480 pmol/L, from about 360 mihoI/L to about 600 mpioI/L, from about 480 pmol/L to about 720 pmol/L, from about 600 pmol/L to about 840 pmol/L, or from about 720 pmol/L to about 960 mmol/L.
[0094] In some embodiments, the compounds may be used in combination with other drugs, such as KUVAN® (sapropterin dihydrochloride), to treat phenylalanine hydroxylase deficiency (e.g., PKU).
Small molecules
[0095] In some embodiments, compounds used to modulate the expression of the PAH gene may include small molecules. As used herein, the term“small molecule” refers a low molecular weight drug, i.e. <5000 Daltons organic compound that may help regulate a biological process. In some embodiments, small molecule compounds described herein are applied to a genomic system to interfere with components (e.g., transcription factor, signaling proteins) of the gene signaling networks associated with the PAH gene, thereby modulating the expression of PAH. In some embodiments, small molecule compounds described herein are applied to a genomic system to alter the boundaries of an insulated neighborhood and/or disrupt signaling centers associated with the PAH gene, thereby modulating the expression of PAH.
[0096] A small molecule screen may be performed to identify small molecules that act through signaling centers of an insulated neighborhood to alter gene signaling networks which may modulate expression of the PAH gene. For example, known signaling agonists/antagonists may be administered. Credible hits are identified and validated by the small molecules that are known to work through a signaling center and modulate expression of the target gene.
[0097] In some embodiments, small molecule compounds capable of modulating expression of the PAH gene include compounds that modulate the JAK/STAT signaling pathway. Such compounds may be JAK inhibitors, including but not limited to Ruxolitinib, Oclacitinib, Baricitinib, Filgotinib, Gandotinib, Lestaurtinib, PF-04965842, Upadacitinib, Cucurbitacin I, CHZ868, Fedratinib, AC430, AT9283, ati-5000l and ati-50002, AZ 960, AZD1480, BMS-911543, CEP-33779, Cerdulatinib (PRT062070, PRT2070), Curcumol, Decemotinib (VX-509), Fedratinib (SAR302503, TG101348), FLLL32, FM-381,
GLPG0634 analogue, Go6976, JANEX-l (WHI-P131), Momelotinib (CYT387), NVP- BSK805, Pacritinib (SB1518), Peficitinib (ASP015K, JNJ-54781532), PF-06651600, PF- 06700841, R256 (AZD0449), Solcitinib (GSK2586184 or GLPG0778), S-Ruxolitinib (INCB018424), TG101209, Tofacitinib (CP-690550), WHI-P154, WP1066, XL019, ZM 39923 HC1, and those described herein.
[0098] In some embodiments, compounds capable of modulating the expression of the PAH gene may include Pacritinib (SB 1518), or a derivative or an analog thereof.
Pacritinib, also known as SB 1518, is a potent and selective inhibitor JAK2 and
FLT3 with IC50S of 23 and 22 nM in cell-free assays, respectively.
[0099] In some embodiments, compounds capable of modulating the expression of the PAH gene may include Momelotinib, or a derivative or an analog thereof. Momelotinib, also known as CYT387, is an ATP-competitive inhibitor of JAK1/JAK2 with IC50 of 11 hM/18 nM and approximately lO-fold selectivity versus JAK3.
[00100] In some embodiments, compounds capable of modulating the expression of the PAH gene may include Tofacitinib, or a derivative or an analog thereof. Tofacitinib, also known as CP-690550, is an inhibitor of JAK1 and JAK3. It is currently approved for the treatment of rheumatoid arthritis (RA) in the United States and other countries.
[00101] In some embodiments, compounds capable of modulating the expression of the PAH gene may include Ruxolitinib, or a derivative or an analog thereof. Ruxolitinib is an oral bioavailable JAK inhibitor with selectivity for JAK1 and JAK2. It is used in the treatment of intermediate or high risk myelofibrosis.
[00102] In some embodiments, compounds capable of modulating the expression of the PAH gene may include Cerdulatinib, or a derivative or an analog thereof. Cerdulatinib is an oral, dual-JAK and Syk inhibitor with IC50 of 12 nM/6 nM/8 nM/0.5 nM and 32 nM for JAK1/JAK2/JAK3/TYK2 and Syk, respectively.
[00103] In some embodiments, compounds capable of modulating the expression of the PAH gene may include JANEX-l (WHI-P131), or a derivative or an analog thereof.
JANEX-l is a cell-permeable JAK3 inhibitor and does not inhibit JAK1, JAK2, or Zap/S yk or Src tyrosine kinases.
[00104] In some embodiments, compounds capable of modulating the expression of the PAH gene may include Oclacitinib, or a derivative or an analog thereof. Oclacitinib is an orally bioavailable, broad spectrum JAK inhibitor with IC50S ranging from 10 to 99 nM. It is used as a veterinary medication in the control of pruritus (itching) associated with allergic dermatitis and atopic dermatitis in dogs.
[00105] In some embodiments, small molecule compounds capable of modulating expression of the PAH gene include compounds that modulate the Tyrosine Kinase/MAPK signaling pathway. Such compounds may be Tyrosine kinase inhibitors, including but not limited to Amuvatinib, Bosutinib, Cediranib, Ceritinib, CP-673451, Dasatinib, GZD824 Dimesylate, Merestinib, R788 (fostamatinib disodium hexahydrate), and those described herein.
[00106] In some embodiments, compounds capable of modulating the expression of the PAH gene may include Amuvatinib, or a derivative or an analog thereof. Amuvatinib, also known as MP-470, is a potent and multi-targeted inhibitor of c-Kit, PDGFRa and FLT3 with IC50 of 10 nM, 40 nM and 81 nM, respectively. Amuvatinib also suppresses c-Met and c-Ret.
[00107] In some embodiments, compounds capable of modulating the expression of the PAH gene may include Bosutinib, or a derivative or an analog thereof. Bosutinib, also known as SKI-606, is a novel, dual Src/Abl inhibitor with IC50 of 1.2 nM and 1 nM, respectively.
[00108] In some embodiments, compounds capable of modulating the expression of the PAH gene may include Cediranib, or a derivative or an analog thereof. Cediranib is a potent inhibitor of vascular endothelial growth factor (VEGF) receptor tyrosine kinases.
[00109] In some embodiments, compounds capable of modulating the expression of the PAH gene may include Ceritinib, or a derivative or an analog thereof. Ceritinib, also known as LDK378, is potent inhibitor against ALK with IC50 of 0.2 nM, exhibiting 40- and 35-fold selectivity against IGF-1R and InsR, respectively.
[00110] In some embodiments, compounds capable of modulating the expression of the PAH gene may include CP-673451, or a derivative or an analog thereof. CP-673451 is a selective inhibitor of PDGFRa/b with IC50 of 10 nM/l nM, exhibiting >450-fold selectivity over other angiogenic receptors. CP-673451 also has antiangiogenic and antitumor activity.
[00111] In some embodiments, compounds capable of modulating the expression of the PAH gene may include Dasatinib, or a derivative or an analog thereof. Dasatinib is a novel, potent and multi-targeted inhibitor that targets Abl, Src, and c-Kit, with IC50 of < 1 nM,
0.8 nM, and 79 nM, respectively.
[00112] In some embodiments, compounds capable of modulating the expression of the PAH gene may include GZD824 Dimesylate, or a derivative or an analog thereof. GZD824 is a novel orally bioavailable Bcr-Abl inhibitor for Bcr-Abl (wildtype) and Bcr-Abl (T315I) with IC50 of 0.34 nM and 0.68 nM, respectively. [00113] In some embodiments, compounds capable of modulating the expression of the PAH gene may include Merestinib, or a derivative or an analog thereof. Merestinib, also known as LY2801653, is a type-II ATP competitive, slow-off inhibitor of MET tyrosine kinase with a Kd of 2 nM, a pharmacodynamic residence time (Koff) of 0.00132 min-l and half life (tl/2) of 525 min.
[00114] In some embodiments, compounds capable of modulating the expression of the PAH gene may include R788 (fostamatinib disodium hexahydrate), or a derivative or an analog thereof. R788 sodium salt hydrate (fostamatinib), a prodrug of the active metabolite R406, is a potent Syk inhibitor with IC50 of 41 nM.
[00115] In some embodiments, small molecule compounds capable of modulating expression of the PAH gene include, but are not limited to, 17-AAG (Tanespimycin), Amlodipine Besylate, ATRA (all-trans retinoic acid), Chloroquine phosphate,
Deoxycorticosterone, Darapladib, Echinomycin, Enzastaurin, Epinephrine, EVP-6124 (hydrochloride) (encenicline), EW-7197, FRAX597, Ibrutinib, Perphenazine, Phenformin, PND- 1186, Rifampicin, Semagacestat, Thalidomide, WAY600, WYE-125132 (WYE-132), and Zibotentan, or derivatives or analogs thereof.
[00116] In some embodiments, compounds capable of modulating the expression of the PAH gene may include 17-AAG (Tanespimycin), or a derivative or an analog thereof. 17- AAG (Tanespimycin), also known as NSC 330507 or CP 127374, is a potent HSP90 inhibitor with half-maximal inhibitory concentration (IC50) of 5 nM, a lOO-fold higher binding affinity for HSP90 derived from tumor cells than HSP90 from normal cells.
[00117] In some embodiments, compounds capable of modulating the expression of the PAH gene may include Amlodipine Besylate, or a derivative or an analog thereof.
Amlodipine, also known as Norvasc, is a long-acting calcium channel blocker with an IC50 of 1.9 nM.
[00118] In some embodiments, compounds capable of modulating the expression of the PAH gene may include ATRA (all-trans retinoic acid), or a derivative or an analog thereof. ATRA (all-trans retinoic acid) is an active metabolite of vitamin A under the family retinoid, which exert potent effects on cell growth, differentiation and apoptosis through their cognate nuclear receptors.
[00119] In some embodiments, compounds capable of modulating the expression of the PAH gene may include Chloroquine phosphate, or a derivative or an analog thereof.
Chloroquine phosphate is an aminoquinoline antimaiarial compound. [00120] In some embodiments, compounds capable of modulating the expression of the PAH gene may include Deoxycorticosterone, or a derivative or an analog thereof.
Deoxycorticosterone acetate is a steroid hormone used for intramuscular injection for replacement therapy of the adrenocortical steroid. 1 I b-hydroxylation of
deoxycorticosterone leads to corticosterone.
[00121] In some embodiments, compounds capable of modulating the expression of the PAH gene may include Darapladib, or a derivative or an analog thereof. Darapladib is a selective and orally active inhibitor of lipoprotein-associated phospholipase A2 (Lp-PLA2) with IC50 of 270 pM. Lp-PLA2 may link lipid metabolism with inflammation, leading to the increased stability of atherosclerotic plaques present in the major arteries. Darapladib is being studied as a possible add-on treatment for atherosclerosis.
[00122] In some embodiments, compounds capable of modulating the expression of the PAH gene may include Echinomycin, or a derivative or an analog thereof. Hypoxia- inducible factor- 1 (HIF-l) is a transcription factor that controls genes involved in glycolysis, angiogenesis, migration, and invasion. Echinomycin is a cell-permeable inhibitor of HIF-l -mediated gene transcription. It acts by intercalating into DNA in a sequence- specific manner, blocking the binding of either HIF-l a or HIF-l b to the hypoxia- responsive element.
[00123] In some embodiments, compounds capable of modulating the expression of the PAH gene may include Enzastaurin, or a derivative or an analog thereof. Enzastaurin, also known as LY317615, is a potent RKCϋb selective inhibitor with IC50 of 6 nM, exhibiting 6- to 20-fold selectivity against PKCa, PKCy and PKCs.
[00124] In some embodiments, compounds capable of modulating the expression of the PAH gene may include Epinephrine, or a derivative or an analog thereof. Epinephrine HC1 is a hormone and a neurotransmitter·
[00125] In some embodiments, compounds capable of modulating the expression of the PAH gene may include EVP-6124 (hydrochloride) (encenicline), or a derivative or an analog thereof. EVP-6124 hydrochloride, also known as encenicline, is a novel partial agonist of a7 neuronal nicotinic acetylcholine receptors (nAChRs). EVP- 6124 shows selectivity for a7 nAChRs and does not activate or inhibit heteromeric a4b2 nAChRs.
[00126] In some embodiments, compounds capable of modulating the expression of the PAH gene may include EW-7197. EW-7197 is a highly potent, selective, and orally bioavailable TGF-b receptor ALK4/ALK5 inhibitor with IC50 of 13 nM and 11 nM, respectively.
[00127] In some embodiments, compounds capable of modulating the expression of the PAH gene may include FRAX597, or a derivative or an analog thereof. FRAX597 is a potent, ATP-competitive inhibitor of group I PAKs with IC50 of 8 nM, 13 nM, and 19 nM for PAK1, PAK2, and PAK3, respectively.
[00128] In some embodiments, compounds capable of modulating the expression of the PAH gene may include Ibrutinib, or a derivative or an analog thereof. Ibrutinib is a Tec family kinase inhibitor that irreversibly inhibits Bruton tyrosine kinase (BTK) and IL-2 Inducible T-cell Kinase (ITK). BTK and ITK are enzymes responsible for the
phosphorylation and activation of downstream effectors in the B-cell receptor (BCR) signaling and T cell receptor (TCR) signaling pathways, respectively.
[00129] In some embodiments, compounds capable of modulating the expression of the PAH gene may include Perphenazine, or a derivative or an analog thereof. Perphenazine is a phenothiazine derivative that binds with high affinity to a wide variety of receptors, including dopamine, serotonin (5-HT), histamine, and a-adrenergic receptors.
Perphenazine is used as an antipsychotic for the symptomatic management of psychotic disorders (e.g., schizophrenia).
[00130] In some embodiments, compounds capable of modulating the expression of the PAH gene may include Phenformin, or a derivative or an analog thereof. Phenformin hydrochloride is a hydrochloride salt of phenformin that is an anti-diabetic drug from the biguanide class.
[00131] In some embodiments, compounds capable of modulating the expression of the PAH gene may include PND-1186, or a derivative or an analog thereof. PND-1186, VS- 4718, is a reversible and selective focal adhesion kinase (FAK) inhibitor with IC50 of 1.5 nM.
[00132] In some embodiments, compounds capable of modulating the expression of the PAH gene may include Rifampicin, or a derivative or an analog thereof. Rifampicin is a member of the rifamycin class of antibiotics, as it inhibits bacterial DNA-dependent RNA synthesis (Ki= ~l nM). While this compound does not directly affect RNA synthesis in humans, its use as an antibiotic is limited by its potency toward activation of the pregnane X receptor (PXR, EC5o= ~2 mM), which results in the up-regulation of enzymes that alter drug metabolism. Access of rifampicin to the nuclear receptor PXR requires its import into the cell viaorganic anion transporters (OATs) in the OAT polypeptide (OATP) family. By acting as a transporter substrate, rifampicin inhibits OATPs with K1/IC50 values ranging from 0.58-18 mM.
[00133] In some embodiments, compounds capable of modulating the expression of the PAH gene may include Semagacestat, or a derivative or an analog thereof. Semagacestat, also known as LY-450139, is a g-secretase inhibitor for Ab42, Ab40 and Ab38 with IC50 of 10.9 nM, 12.1 nM and 12.0 nM, respectively. Semagacestat also inhibits Notch signaling with IC50 of 14.1 nM.
[00134] In some embodiments, compounds capable of modulating the expression of the PAH gene may include Thalidomide, or a derivative or an analog thereof. Thalidomide was introduced as a sedative drug, immunomodulatory agent and also is investigated for treating symptoms of many cancers. Thalidomide inhibits an E3 ubiquitin ligase, which is a CRBN-DDB l-Cul4A complex.
[00135] In some embodiments, compounds capable of modulating the expression of the PAH gene may include WAY600, or a derivative or an analog thereof. WAY600 is a potent, ATP-competitive and selective inhibitor of mTOR with IC50 of 9 nM.
[00136] In some embodiments, compounds capable of modulating the expression of the PAH gene may include WYE-125132 (WYE-132), or a derivative or an analog thereof. WYE-125132, also known as WYE-132, is a highly potent, ATP-competitive mTOR inhibitor with IC50 of 0.19 nM. It is highly selective for mTOR versus PI3Ks or PI3K- related kinases hSMGl and ATR.
[00137] In some embodiments, compounds capable of modulating the expression of the PAH gene may include Zibotentan, or a derivative or an analog thereof. Zibotentan, also known as ZD4054, is an orally administered, potent and specific endothelin A receptor (ETA) -receptor antagonist with IC50 of 21 nM.
Polypeptides
[00138] In some embodiments, compounds for altering expression of the PAH gene comprise a polypeptide. As used herein, the term“polypeptide” refers to a polymer of amino acid residues (natural or unnatural) linked together most often by peptide bonds. The term, as used herein, refers to proteins, polypeptides, and peptides of any size, structure, or function. In some instances, the polypeptide encoded is smaller than about 50 amino acids and the polypeptide is then termed a peptide. If the polypeptide is a peptide, it will be at least about 2, 3, 4, or at least 5 amino acid residues long. Thus, polypeptides include gene products, naturally occurring polypeptides, synthetic polypeptides, homologs, orthologs, paralogs, fragments and other equivalents, variants, and analogs of the foregoing. A polypeptide may be a single molecule or may be a multi-molecular complex such as a dimer, trimer or tetramer. They may also comprise single chain or multichain polypeptides and may be associated or linked. The term polypeptide may also apply to amino acid polymers in which one or more amino acid residues are an artificial chemical analog of a corresponding naturally occurring amino acid.
[00139] In some embodiments, polypeptide compounds capable of modulating expression of the PAH gene include, but are not limited to, Activin, Anti mullerian hormone, GDF10 (BMP3b), IGF-l, Nodal, PDGF, TNF-a, Wnt3a, or derivatives or analogs thereof. Any one of these compounds or a combination thereof may be administered to a subject to treat phenylalanine hydroxylase deficiency.
[00140] In some embodiments, compounds capable of modulating the expression of the PAH gene may include Activin, or a derivative or an analog thereof. Activins are homodimers or heterodimers of the different b subunit isoforms, part of the transforming growth factor-beta (TGF-B) family. Mature Activin A has two 116 amino acids residues bA subunits (bA-bA). Activin displays an extensive variety of biological activities, including mesoderm induction, neural cell differentiation, bone remodeling, hematopoiesis, and reproductive physiology. Activins takes part in the production and regulation of hormones such as FSH, LH, GnRH and ACTH. Cells that are identified to express Activin A include fibroblasts, endothelial cells, hepatocytes, vascular smooth muscle cells, macrophages, keratinocytes, osteoclasts, bone marrow monocytes, prostatic epithelium, neurons, chondrocytes, osteoblasts, Leydig cells, Sertoli cells, and ovarian granulosa cells.
[00141] In some embodiments, compounds capable of modulating the expression of the PAH gene may include anti Mullerian hormone, or a derivative or an analog thereof. Anti Mullerian hormone is a member of the TGF-B gene family which mediates male sexual differentiation. Anti Mullerian hormone causes the regression of Mullerian ducts which would otherwise differentiate into the uterus and fallopian tubes. Some mutations in the anti-Mullerian hormone result in persistent Mullerian duct syndrome.
[00142] In some embodiments, compounds capable of modulating the expression of the PAH gene may include GDF10 (BMP3b), or a derivative or an analog thereof. GDF10, also known as BMP3b, is a member of the BMP family and the TGF-B superfamily.
GDF10 is expressed in femur, brain, lung, skeletal, muscle, pancreas and testis, and has a role in head formation and possibly multiple roles in skeletal morphogenesis. In humans, GDF10 mRNA is found in the cochlea and lung of fetuses, and in testis, retina, pineal gland, and other neural tissues of adults. These proteins are characterized by a polybasic proteolytic processing site which is cleaved to produce a mature protein containing 7 conserved cysteine residues.
[00143] In some embodiments, compounds capable of modulating the expression of the PAH gene may include IGF-l, or a derivative or an analog thereof. Insulin-like growth factor I (IGF-I) also known as Somatamedin C is a hormone similar in molecular structure to insulin. Human IGF-I has two isoforms (IGF-IA and IGF-IB) which is differentially expressed by various tissues. Mature human IGF-I respectively shares 94% and 96% aa sequence identity with mouse and rat IGF-I. Both IGF-I and IGF-II (another ligand of IGF) can signal through the IGF-I receptor (IGF1R), but IGF-II can alone bind the IGF-II receptor (IGFIIR/Mannose-6-phosphate receptor). IGF-I plays an important role in childhood growth and continues to have anabolic effects in adults.
[00144] In some embodiments, compounds capable of modulating the expression of the PAH gene may include Nodal, or a derivative or an analog thereof. Nodal is a 13 kDa member of the TGF-B superfamily of molecules. In human, it is synthesized as a 347 amino acid preproprecursor that contains a 26 amino acid signal sequence, a 211 amino acid prodomain, and a 110 amino acid mature region. Consistent with its TGF-B superfamily membership, it exists as a disulfide-linked homodimer and would be expected to demonstrate a cysteine-knot motif. Mature human Nodal is 99%, 98%, 96% and 98% amino acid identical to mature canine, rat, bovine and mouse Nodal, respectively. Nodal signals through two receptor complexes, both of which contain members of the TGF-beta family of Ser/Thr kinase receptors.
[00145] In some embodiments, compounds capable of modulating the expression of the PAH gene may include PDGF, or a derivative or an analog thereof the Platelet-derived growth factor (PDGF) is a disulfide-linked dimer consisting of two peptides -chain A and chain B. PDGF has three subforms: PDGF-AA, PDGF-BB, PDGF-AB. It is involved in a number of biological processes, including hyperplasia, embryonic neuron development, chemotaxis, and respiratory tubule epithelial cell development. The function of PDGF is mediated by two receptors (PDGFRa and PDGFR ).
[00146] In some embodiments, compounds capable of modulating the expression of the PAH gene may include TNF-a, or a derivative or an analog thereof. TNF-a, the prototypical member of the TNF protein superfamily, is a homotrim eric type-II membrane protein. Membrane bound TNF-a is cleaved by the metalloprotease TACE/ADAM17 to generate a soluble homotrimer. Both membrane and soluble forms of TNF-a are biologically active. TNF-a is produced by a variety of immune cells including T cells, B cells, NK cells and macrophages. Cellular response to TNF-a is mediated through interaction with receptors TNF-R1 and TNF-R2 and results in activation of pathways that favor both cell survival and apoptosis depending on the cell type and biological context. Activation of kinase pathways (including JNK, ERK (p44/42), p38 MAPK and NF-kB) promotes the survival of cells, while TNF-a mediated activation of caspase-8 leads to programmed cell death. TNF-a plays a key regulatory role in inflammation and host defense against bacterial infection, notably Mycobacterium tuberculosis. The role of TNF- a in autoimmunity is underscored by blocking TNF-a action to treat rheumatoid arthritis and Crohn’ s disease.
[00147] In some embodiments, compounds capable of modulating the expression of the PAH gene may include Wnt3a, or a derivative or an analog thereof. The WNT gene family consists of structurally related genes which encode secreted signaling proteins. These proteins have been implicated in oncogenesis and in several developmental processes, including regulation of cell fate and patterning during embryogenesis. This gene is a member of the WNT gene family. It encodes a protein which shows 96% amino acid identity to mouse Wnt3a protein, and 84% to human WNT3 protein, another WNT gene product. This gene is clustered with WNT14 gene, another family member, in chromosome lq42 region.
Antibodies
[00148] In some embodiments, compounds for altering expression of the PAH gene comprise an antibody. In certain embodiments, antibodies described herein comprise antibodies, antibody fragments, their variants or derivatives that are specifically immunoreaetive with at least one component of the gene signaling networks associated with the PAH gene.
[00149] As used herein, the term "antibody" is used in the broadest sense and specifically covers various embodiments including, but not limited to monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g. bispecific antibodies formed from at least two intact antibodies), and antibody fragments such as diabodies so long as they exhibit a desired biological activity. Antibodies are primarily amino-acid based molecules but may also comprise one or more modifications such as with sugar moieties.
[00150] “Antibody fragments" comprise a portion of an intact antibody, preferably comprising an antigen binding region thereof. Examples of antibody fragments include Fab, Fab', F(ab')2, and Fv fragments; diabodies; linear antibodies; single-chain antibody molecules; and multispecific antibodies formed from antibody fragments. Papain digestion of antibodies produces two identical antigen-binding fragments, called "Fab" fragments, each with a single antigen-binding site. Also produced is a residual "Fc" fragment, whose name reflects its ability to crystallize readily. Pepsin treatment yields an F(ab')2 fragment that has two antigen-binding sites and is still capable of cross-linking antigen. Antibodies of the present disclosure may comprise one or more of these fragments. For the purposes herein, an "antibody" may comprise a heavy and light variable domain as well as an Fc region.
[00151] " Native antibodies" are usually heterotetrameric glycoproteins of about 150,000 Daltons, composed of two identical light (L) chains and two identical heavy (H) chains. Each light chain is linked to a heavy chain by one covalent disulfide bond, while the number of disulfide linkages varies among the heavy chains of different immunoglobulin isotypes. Each heavy and light chain also has regularly spaced intrachain disulfide bridges. Each heavy chain has at one end a variable domain (VH) followed by a number of constant domains. Each light chain has a variable domain at one end (VL) and a constant domain at its other end; the constant domain of the light chain is aligned with the first constant domain of the heavy chain, and the light chain variable domain is aligned with the variable domain of the heavy chain.
[00152] As used herein, the term "variable domain" refers to specific antibody domains that differ extensively in sequence among antibodies and are used in the binding and specificity of each particular antibody for its particular antigen. As used herein, the term “Fv” refers to antibody fragments which contain a complete antigen-recognition and antigen-binding site. This region consists of a dimer of one heavy chain and one light chain variable domain in tight, non-covalent association.
[00153] Antibody "light chains" from any vertebrate species can be assigned to one of two clearly distinct types, called kappa and lambda based on amino acid sequences of their constant domains. Depending on the amino acid sequence of the constant domain of their heavy chains, antibodies can be assigned to different classes. There are five major classes of intact antibodies: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgGl, IgG2, IgG3, IgG4, IgA, and IgA2.
[00154] "Single-chain Fv" or "scFv" as used herein, refers to a fusion protein of VH and VL antibody domains, wherein these domains are linked together into a single polypeptide chain. In some embodiments, the Fv polypeptide linker enables the scFv to form the desired structure for antigen binding.
[00155] The term "diabodies" refers to small antibody fragments with two antigen-binding sites, which fragments comprise a heavy chain variable domain VH connected to a light chain variable domain VL in the same polypeptide chain. By using a linker that is too short to allow pairing between the two domains on the same chain, the domains are forced to pair with the complementary domains of another chain and create two antigen-binding sites. Diabodies are described more fully in, for example, EP 404,097; WO 93/11161; and Hollinger et al., Proc. Natl. Acad. Sci. USA, 90:6444-6448 (1993), the contents of each of which are incorporated herein by reference in their entirety.
[00156] Antibodies of the present disclosure may be polyclonal or monoclonal or recombinant, produced by methods known in the art or as described in this application. The term "monoclonal antibody" as used herein refers to an antibody obtained from a population of substantially homogeneous cells (or clones), i.e., the individual antibodies comprising the population are identical and/or bind the same epitope, except for possible variants that may arise during production of the monoclonal antibody, such variants generally being present in minor amounts. In contrast to polyclonal antibody preparations that typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen.
[00157] The modifier "monoclonal" indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method. The monoclonal antibodies herein include "chimeric" antibodies (immunoglobulins) in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies. [00158] " Humanized" forms of non-human (e.g., murine) antibodies are chimeric antibodies that contain minimal sequence derived from non-human immunoglobulin. For the most part, humanized antibodies are human immunoglobulins (recipient antibody) in which residues from the hypervariable region from an antibody of the recipient are replaced by residues from the hypervariable region from an antibody of a non-human species (donor antibody) such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity, and capacity.
[00159] The term "hypervariable region" when used herein in reference to antibodies refers to regions within the antigen binding domain of an antibody comprising the amino acid residues that are responsible for antigen binding. The amino acids present within the hypervariable regions determine the structure of the complementarity determining region (CDR). As used herein, the“CDR” refers to the region of an antibody that comprises a structure that is complimentary to its target antigen or epitope.
[00160] In some embodiments, the compositions of the present disclosure may be antibody mimetics. The term“antibody mimetic” refers to any molecule which mimics the function or effect of an antibody and which binds specifically and with high affinity to their molecular targets. As such, antibody mimics include nanobodies and the like.
[00161] In some embodiments, antibody mimetics may be those known in the art including, but are not limited to affibody molecules, affilins, affitins, anticalins, avimers, DARPins, Fynomers and Kunitz and domain peptides. In other embodiments, antibody mimetics may include one or more non-peptide region.
[00162] As used herein, the term“antibody variant” refers to a biomolecule resembling an antibody in structure and/or function comprising some differences in their amino acid sequence, composition or structure as compared to a native antibody.
[00163] The preparation of antibodies, whether monoclonal or polyclonal, is known in the art. Techniques for the production of antibodies are well known in the art and described, e.g. in Harlow and Lane "Antibodies, A Laboratory Manual", Cold Spring Harbor Laboratory Press, 1988 and Harlow and Lane“Using Antibodies: A Laboratory Manual” Cold Spring Harbor Laboratory Press, 1999.
[00164] Antibodies of the present disclosure may be characterized by their target molecule(s), by the antigens used to generate them, by their function (whether as agonists or antagonists) and/or by the cell niche in which they function. [00165] Measures of antibody function may be made relative to a standard under normal physiologic conditions, in vitro or in vivo. Measurements may also be made relative to the presence or absence of the antibodies. Such methods of measuring include standard measurement in tissue or fluids such as serum or blood such as Western blot, enzyme- linked immunosorbent assay (ELISA), activity assays, reporter assays, luciferase assays, polymerase chain reaction (PCR) arrays, gene arrays, Real Time reverse transcriptase (RT) PCR and the like.
[00166] Antibodies exert their effects via binding (reversibly or irreversibly) to one or more target sites. While not wishing to be bound by theory, target sites which represent a binding site for an antibody, are most often formed by proteins or protein domains or regions. However, target sites may also include biomolecules such as sugars, lipids, nucleic acid molecules or any other form of binding epitope.
[00167] Alternatively, or additionally, antibodies of the present disclosure may function as ligand mimetics or nontraditional payload carriers, acting to deliver or ferry bound or conjugated drug payloads to specific target sites.
[00168] Changes elicited by antibodies may result in a neomorphic change in the cell. As used herein,“a neomorphic change” is a change or alteration that is new or different. Such changes include extracellular, intracellular and cross cellular signaling.
[00169] In some embodiments, compounds or agents act to alter or control proteolytic events. Such events may be intracellular or extracellular.
[00170] Antibodies, as well as antigens used to generate them, are primarily amino acid- based molecules. These molecules may be "peptides," "polypeptides," or "proteins.”
[00171] As used herein, the term“peptide” refers to an amino-acid based molecule having from 2 to 50 or more amino acids. Special designators apply to the smaller peptides with “dipeptide” referring to a two amino acid molecule and“tripeptide” referring to a three amino acid molecule. Amino acid based molecules having more than 50 contiguous amino acids are considered polypeptides or proteins.
[00172] The terms "amino acid" and "amino acids" refer to all naturally occurring L- alpha-amino acids as well as non-naturally occurring amino acids. Amino acids are identified by either the one-letter or three-letter designations as follows: aspartic acid (Asp:D), isoleucine (He : I) , threonine (Thr:T), leucine (Leu:L), serine (Ser:S), tyrosine (Tyr:Y), glutamic acid (Glu:E), phenylalanine (Phe:F), proline (Pro:P), histidine (His:H), glycine (Gly:G), lysine (Lys:K), alanine (Ala:A), arginine (Arg:R), cysteine (Cys:C), tryptophan (Trp:W), valine (Val:V), glutamine (Gln:Q) methionine (Met:M), asparagines (Asn:N), where the amino acid is listed first followed parenthetically by the three and one letter codes, respectively.
Hybridizing oligonucleotides
[00173] In some embodiments, oligonucleotides, including those which function via a hybridization mechanism, whether single of double stranded such as antisense molecules, RNAi constructs (including siRNA, saRNA, microRNA, etc.), aptamers and ribozymes may be used to alter or as perturbation stimuli of the gene signaling networks associated with the PAH gene.
[00174] In some embodiments, hybridizing oligonucleotides (e.g., siRNA) may be used to knock down signaling molecules involved in the pathways regulating PAH expression such that PAH expression is enhanced in the absence of the signaling molecule. For example, once a pathway is identified to negatively regulate PAH expression, a component of the pathway (e.g., a receptor, a protein kinase, a transcription factor) may be knocked down with a RNAi agent (e.g., siRNA) to enhance the expression of PAH.
[00175] In some embodiments, the pathway targeted with a hybridizing oligonucleotide (e.g., siRNA) of the present disclosure to enhance PAH expression is the JAK/STAT pathway. In one embodiment, the hybridizing oligonucleotide (e.g., siRNA) is used to knock down JAK1. In one embodiment, the hybridizing oligonucleotide (e.g., siRNA) is used to knock down JAK2.
[00176] In some embodiments, the pathway targeted with a hybridizing oligonucleotide (e.g., siRNA) of the present disclosure to enhance PAH expression is the PDGFR pathway. In one embodiment, the hybridizing oligonucleotide (e.g., siRNA) is used to knock down PDGFRA. In one embodiment, the hybridizing oligonucleotide (e.g., siRNA) is used to knock down PDGFRB.
[00177] In some embodiments, the pathway targeted with a hybridizing oligonucleotide (e.g., siRNA) of the present disclosure to enhance PAH expression is the Src/Abl pathway. In one embodiment, the hybridizing oligonucleotide (e.g., siRNA) is used to knock-down SRC. In one embodiment, the hybridizing oligonucleotide (e.g., siRNA) is used to knock down ABL.
[00178] In some embodiments, a hybridizing oligonucleotide as described above may be used together with another hybridizing oligonucleotide to target more than one components in the same pathway, or more than one components from different pathways, to enhance PAH expression. Such combination therapies may achieve additive or synergetic effects by simultaneously blocking multiple signaling molecules and/or pathways that negatively regulate PAH expression.
[00179] As such oligonucleotides may also serve as therapeutics, their therapeutic liabilities and treatment outcomes may be ameliorated or predicted, respectively by interrogating the gene signaling networks of the disclosure.
Genome editing approaches
[00180] In certain embodiments, expression of the PAH gene may be modulated by altering the chromosomal regions defining the insulated neighborhood(s) and/or genome signaling center(s) associated with the PAH gene. For example, protein production may be increased by targeting a component of the gene signaling network that functions to repress the expression of the PAH gene.
[00181] Methods of altering the gene expression attendant to an insulated neighborhood include altering the signaling center (e.g. using CRISPR/Cas to change the signaling center binding site or repair/replace if mutated). These alterations may result in a variety of results including: activation of cell death pathways prematurely/inappropriately (key to many immune disorders), production of too little/much gene product (also known as the rheostat hypothesis), production of too little/much extracellular secretion of enzymes, prevention of lineage differentiation, switch of lineage pathways, promotion of sternness, initiation or interference with auto regulatory feedback loops, initiation of errors in cell metabolism, inappropriate imprinting/gene silencing, and formation of flawed chromatin states.
Additionally, genome editing approaches including those well-known in the art may be used to create new signaling centers by altering the cohesin necklace or moving genes and enhancers.
[00182] In certain embodiments, genome editing approaches describe herein may include methods of using site-specific nucleases to introduce single-strand or double-strand DNA breaks at particular locations within the genome. Such breaks can be and regularly are repaired by endogenous cellular processes, such as homology-directed repair (HDR) and non-homologous end joining (NHEJ). HDR is essentially an error- free mechanism that repairs double-strand DNA breaks in the presence of a homologous DNA sequence. The most common form of HDR is homologous recombination. It utilizes a homologous sequence as a template for inserting or replacing a specific DNA sequence at the break point. The template for the homologous DNA sequence can be an endogenous sequence (e.g., a sister chromatid), or an exogenous or supplied sequence (e.g., plasmid or an oligonucleotide). As such, HDR may be utilized to introduce precise alterations such as replacement or insertion at desired regions. In contrast, NHEJ is an error-prone repair mechanism that directly joins the DNA ends resulting from a double-strand break with the possibility of losing, adding or mutating a few nucleotides at the cleavage site. The resulting small deletions or insertions (termed“Indels”) or mutations may disrupt or enhance gene expression. Additionally, if there are two breaks on the same DNA, NHEJ can lead to the deletion or inversion of the intervening segment. Therefore, NHEJ may be utilized to introduce insertions, deletions or mutations at the cleavage site.
CRISPR/Cas systems
[00183] In certain embodiments, a CRISPR/Cas system may be used to delete CTCF anchor sites to modulate gene expression within the insulated neighborhood associated with that anchor site. See, Hnisz et ak, Cell 167, November 17, 2016, which is hereby incorporated by reference in its entirety. Disruption of the boundaries of insulated neighborhood prevents the interactions necessary for proper function of the associated signaling centers. Changes in the expression genes that are immediately adjacent to the deleted neighborhood boundary have also been observed due to such disruptions.
[00184] In certain embodiments, a CRISPR/Cas system may be used to modify existing CTCF anchor sites. For example, existing CTCF anchor sites may be mutated or inverted by inducing NHEJ with a CRISPR/Cas nuclease and one or more guide RNAs, or masked by targeted binding with a catalytically inactive CRISPR/Cas enzyme and one or more guide RNAs. Alteration of existing CTCF anchor sites may disrupt the formation of existing insulated neighborhoods and alter the expression of genes located within these insulated neighborhoods.
[00185] In certain embodiments, a CRISPR/Cas system may be used to introduce new CTCF anchor sites. CTCF anchor sites may be introduced by inducing HDR at a selected site with a CRISPR/Cas nuclease, one or more guide RNAs and a donor template containing the sequence of a CTCF anchor site. Introduction of new CTCF anchor sites may create new insulated neighborhoods and/or alter existing insulated neighborhoods, which may affect expression of genes that are located adjacent to these insulated neighborhoods.
[00186] In certain embodiments, a CRISPR/Cas system may be used to alter signaling centers by changing signaling center binding sites. For example, if a signaling center binding site contains a mutation that affects the assembly of the signaling center with associated transcription factors, the mutated site may be repaired by inducing a double strand DNA break at or near the mutation using a CRISPR/Cas nuclease and one or more guide RNAs in the presence of a supplied corrected donor template.
[00187] In certain embodiments, a CRISPR/Cas system may be used to modulate expression of neighborhood genes by binding to a region within an insulated neighborhood (e.g., enhancer) and block transcription. Such binding may prevent recruitment of transcription factors to signaling centers and initiation of transcription. The CRISPR/Cas system may be a catalytically inactive CRISPR/Cas system that do not cleave DNA.
[00188] In certain embodiments, a CRISPR/Cas system may be used to knockdown expression of neighborhood genes via introduction of short deletions in coding regions of these genes. When repaired, such deletions would result in frame shifts and/or introduce premature stop codons in mRNA produced by the genes followed by the mRNA
degradation via nonsense-mediated decay. This may be useful for modulation of expression of activating and repressive components of signaling pathways that would result in decreased or increased expression of genes under control of these pathways including disease genes such as PAH.
[00189] In other embodiments, a CRISPR/Cas system may also be used to alter cohesion necklace or moving genes and enhancers.
CRISPR/Cas enzymes
[00190] CRISPR/Cas systems are bacterial adaptive immune systems that utilize RNA- guided endonucleases to target specific sequences and degrade target nucleic acids. They have been adapted for use in various applications in the field of genome editing and/or transcription modulation. Any of the enzymes or orthologs known in the art or disclosed herein may be utilized in the methods herein for genome editing.
[00191] In certain embodiments, the CRISPR/Cas system may be a Type II CRISPR/Cas9 system. Cas9 is an endonuclease that functions together with a trans-activating CRISPR RNA (tracrRNA) and a CRISPR RNA (crRNA) to cleave double stranded DNAs. The two RNAs can be engineered to form a single-molecule guide RNA by connecting the 3’ end of the crRNA to the 5’ end of tracrRNA with a linker loop. Jinek et ak, Science,
337(6096):8l6-82l (2012) showed that the CRISPR/Cas9 system is useful for RNA- programmable genome editing, and international patent application WO2013/176772 provides numerous examples and applications of the CRISPR/Cas endonuclease system for site-specific gene editing, which are incorporated herein by reference in their entirety. Exemplary CRISPR/Cas9 systems include those derived from Streptococcus pyogenes, Streptococcus thermophilus, Neisseria meningitidis, Treponema denticola, Streptococcus aureas, and Francisella tularensis.
[00192] In certain embodiments, the CRISPR/Cas system may be a Type V CRISPR/Cpfl system. Cpfl is a single RNA-guided endonuclease that, in contrast to Type II systems, lacks tracrRNA. Cpfl produces staggered DNA double-stranded break with a 4 or 5 nucleotide 5’ overhang. Zetsche et al. Cell. 2015 Oct 22;l63(3):759-7l provides examples of Cpfl endonuclease that can be used in genome editing applications, which is incorporated herein by reference in its entirety. Exemplary CRISPR/Cpfl systems include those derived from Francisella tularensis, Acidaminococcus sp., and Lachnospiraceae bacterium.
[00193] In certain embodiments, nickase variants of the CRISPR/Cas endonucleases that have one or the other nuclease domain inactivated may be used to increase the specificity of CRISPR-mediated genome editing. Nickases have been shown to promote HDR versus NHEJ. HDR can be directed from individual Cas nickases or using pairs of nickases that flank the target area.
[00194] In certain embodiments, catalytically inactive CRISPR/Cas systems may be used to bind to target regions (e.g., CTCF anchor sites or enhancers) and interfere with their function. Cas nucleases such as Cas9 and Cpfl encompass two nuclease domains. Mutating critical residues at the catalytic sites creates variants that only bind to target sites but do not result in cleavage. Binding to chromosomal regions (e.g., CTCF anchor sites or enhancers) may disrupt proper formation of insulated neighborhoods or signaling centers and therefore lead to altered expression of genes located adjacent to the target region.
[00195] In certain embodiments, a CRISPR/Cas system may include additional functional domain(s) fused to the CRISPR/Cas enzyme. The functional domains may be involved in processes including but not limited to transcription activation, transcription repression, DNA methylation, histone modification, and/or chromatin remodeling. Such functional domains include but are not limited to a transcriptional activation domain (e.g., VP64 or KRAB, SID or SID4X), a transcriptional repressor, a recombinase, a transposase, a histone remodeler, a DNA methyltransferase, a cryptochrome, a light inducible/controllable domain or a chemically inducible/controllable domain. [00196] In certain embodiments, a CRISPR/Cas enzyme may be administered to a cell or a patient as one or a combination of the following: one or more polypeptides, one or more mRNAs encoding the polypeptide, or one or more DNAs encoding the polypeptide.
Guide nucleic acid
[00197] In certain embodiments, guide nucleic acids may be used to direct the activities of an associated CRISPR/Cas enzymes to a specific target sequence within a target nucleic acid. Guide nucleic acids provide target specificity to the guide nucleic acid and
CRISPR/Cas complexes by virtue of their association with the CRISPR/Cas enzymes, and the guide nucleic acids thus can direct the activity of the CRISPR/Cas enzymes.
[00198] In one aspect, guide nucleic acids may be RNA molecules. In one aspect, guide RNAs may be single-molecule guide RNAs. In one aspect, guide RNAs may be chemically modified.
[00199] In certain embodiments, more than one guide RNAs may be provided to mediate multiple CRISPR/Cas-mediated activities at different sites within the genome.
[00200] In certain embodiments, guide RNAs may be administered to a cell or a patient as one or more RNA molecules or one or more DNAs encoding the RNA sequences.
Ribonucleoprotein complexes (RNPs)
[00201] In one embodiment, the CRISPR/Cas enzyme and guide nucleic acid may each be administered separately to a cell or a patient.
[00202] In another embodiment, the CRISPR/Cas enzyme may be pre-complexed with one or more guide nucleic acids. The pre-complexed material may then be administered to a cell or a patient. Such pre-complexed material is known as a ribonucleoprotein particle (RNP).
Zinc Finger Nucleases
[00203] In certain embodiments, genome editing approaches of the present disclosure involve the use of Zinc finger nucleases (ZFNs). Zinc finger nucleases (ZFNs) are modular proteins comprised of an engineered zinc finger DNA binding domain linked to a DNA- cleavage domain. A typical DNA-cleavage domain is the catalytic domain of the type II endonuclease Fokl. Because Fokl functions only as a dimer, a pair of ZFNs must are required to be engineered to bind to cognate target“half-site” sequences on opposite DNA strands and with precise spacing between them to allow the two enable the catalytically active Fokl domains to dimerize. Upon dimerization of the Fokl domain, which itself has no sequence specificity per se, a DNA double-strand break is generated between the ZFN half-sites as the initiating step in genome editing.
Transcription Activator-Like Effector Nucleases (TALENs)
[00204] In certain embodiments, genome editing approaches of the present disclosure involve the use of Transcription Activator-Like Effector Nucleases (TALENs). TALENs represent another format of modular nucleases which, similarly to ZFNs, are generated by fusing an engineered DNA binding domain to a nuclease domain, and operate in tandem to achieve targeted DNA cleavage. While the DNA binding domain in ZFN consists of Zinc finger motifs, the TALEN DNA binding domain is derived from transcription activator-like effector (TALE) proteins, which were originally described in the plant bacterial pathogen Xanthomonas sp. TALEs are comprised of tandem arrays of 33-35 amino acid repeats, with each repeat recognizing a single basepair in the target DNA sequence that is typically up to 20 bp in length, giving a total target sequence length of up to 40 bp. Nucleotide specificity of each repeat is determined by the repeat variable diresidue (RVD), which includes just two amino acids at positions 12 and 13. The bases guanine, adenine, cytosine and thymine are predominantly recognized by the four RVDs: Asn-Asn, Asn-Ile, His- Asp and Asn-Gly, respectively. This constitutes a much simpler recognition code than for zinc fingers, and thus represents an advantage over the latter for nuclease design. Nevertheless, as with ZFNs, the protein-DNA interactions of TALENs are not absolute in their specificity, and TALENs have also benefitted from the use of obligate heterodimer variants of the Fokl domain to reduce off-target activity.
IV. FORMULATIONS AND DELIVERY
Pharmaceutical Compositions
[00205] The compositions of the present disclosure may be prepared as pharmaceutical compositions. It will be understood that such compositions necessarily comprise one or more active ingredients and, most often, a pharmaceutically acceptable excipient.
[00206] Relative amounts of the active ingredient, a pharmaceutically acceptable excipient, and/or any additional ingredients in a pharmaceutical composition in accordance with the present disclosure may vary, depending upon the identity, size, and/or condition of the subject being treated and further depending upon the route by which the composition is to be administered. For example, the composition may comprise between 0.1% and 99% (w/w) of the active ingredient. By way of example, the composition may comprise between 0.1% and 100%, e.g., between .5 and 50%, between 1-30%, between 5-80%, at least 80% (w/w) active ingredient.
[00207] In some embodiments, the pharmaceutical compositions described herein may comprise at least one payload. As a non-limiting example, the pharmaceutical compositions may contain 1, 2, 3, 4 or 5 payloads.
[00208] Although the descriptions of pharmaceutical compositions provided herein are principally directed to pharmaceutical compositions which are suitable for administration to humans, it will be understood by the skilled artisan that such compositions are generally suitable for administration to any other animal, e.g., to non-human animals, e.g. non human mammals. Modification of pharmaceutical compositions suitable for administration to humans in order to render the compositions suitable for administration to various animals is well understood, and the ordinarily skilled veterinary pharmacologist can design and/or perform such modification with merely ordinary, if any, experimentation. Subjects to which administration of the pharmaceutical compositions is contemplated include, but are not limited to, humans and/or other primates; mammals, including commercially relevant mammals such as cattle, pigs, horses, sheep, cats, dogs, mice, rats, birds, including commercially relevant birds such as poultry, chickens, ducks, geese, and/or turkeys.
[00209] In some embodiments, compositions are administered to humans, human patients or subjects.
Formulations
[00210] Formulations can include, without limitation, saline, liposomes, lipid
nanoparticles, polymers, peptides, proteins, cells transfected with viral vectors (e.g., for transfer or transplantation into a subject) and combinations thereof.
[00211] Formulations of the pharmaceutical compositions described herein may be prepared by any method known or hereafter developed in the art of pharmacology. As used herein the term“pharmaceutical composition” refers to compositions comprising at least one active ingredient and optionally one or more pharmaceutically acceptable excipients.
[00212] In general, such preparatory methods include the step of associating the active ingredient with an excipient and/or one or more other accessory ingredients.
[00213] Formulations of the compositions described herein may be prepared by any method known or hereafter developed in the art of pharmacology. In general, such preparatory methods include the step of bringing the active ingredient into association with an excipient and/or one or more other accessory ingredients, and then, if necessary and/or desirable, dividing, shaping and/or packaging the product into a desired single- or multi dose unit.
[00214] A pharmaceutical composition in accordance with the present disclosure may be prepared, packaged, and/or sold in bulk, as a single unit dose, and/or as a plurality of single unit doses. As used herein, a“unit dose” refers to a discrete amount of the pharmaceutical composition comprising a predetermined amount of the active ingredient. The amount of the active ingredient is generally equal to the dosage of the active ingredient which would be administered to a subject and/or a convenient fraction of such a dosage such as, for example, one-half or one-third of such a dosage.
[00215] Relative amounts of the active ingredient, the pharmaceutically acceptable excipient, and/or any additional ingredients in a pharmaceutical composition in accordance with the present disclosure may vary, depending upon the identity, size, and/or condition of the subject being treated and further depending upon the route by which the composition is to be administered. For example, the composition may comprise between 0.1% and 99% (w/w) of the active ingredient. By way of example, the composition may comprise between 0.1% and 100%, e.g., between 0.5 and 50%, between 1-30%, between 5-80%, at least 80% (w/w) active ingredient.
Excipients and Diluents
[00216] In some embodiments, a pharmaceutically acceptable excipient may be at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% pure. In some embodiments, an excipient is approved for use for humans and for veterinary use. In some embodiments, an excipient may be approved by United States Food and Drug
Administration. In some embodiments, an excipient may be of pharmaceutical grade. In some embodiments, an excipient may meet the standards of the United States
Pharmacopoeia (USP), the European Pharmacopoeia (EP), the British Pharmacopoeia, and/or the International Pharmacopoeia.
[00217] Excipients, as used herein, include, but are not limited to, any and all solvents, dispersion media, diluents, or other liquid vehicles, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, and the like, as suited to the particular dosage form desired. Various excipients for formulating pharmaceutical compositions and techniques for preparing the composition are known in the art (see Remington: The Science and Practice of Pharmacy, 2lst Edition, A. R.
Gennaro, Lippincott, Williams & Wilkins, Baltimore, MD, 2006; incorporated herein by reference in its entirety). The use of a conventional excipient medium may be contemplated within the scope of the present disclosure, except insofar as any conventional excipient medium may be incompatible with a substance or its derivatives, such as by producing any undesirable biological effect or otherwise interacting in a deleterious manner with any other component(s) of the pharmaceutical composition.
[00218] Exemplary diluents include, but are not limited to, calcium carbonate, sodium carbonate, calcium phosphate, dicalcium phosphate, calcium sulfate, calcium hydrogen phosphate, sodium phosphate lactose, sucrose, cellulose, microcrystalline cellulose, kaolin, mannitol, sorbitol, inositol, sodium chloride, dry starch, cornstarch, powdered sugar, etc., and/or combinations thereof.
Inactive Ingredients
[00219] In some embodiments, the pharmaceutical compositions formulations may comprise at least one inactive ingredient. As used herein, the term“inactive ingredient” refers to one or more agents that do not contribute to the activity of the active ingredient of the pharmaceutical composition included in formulations. In some embodiments, all, none or some of the inactive ingredients which may be used in the formulations of the present disclosure may be approved by the US Food and Drug Administration (FDA).
[00220] In one embodiment, the pharmaceutical compositions comprise at least one inactive ingredient such as, but not limited to, l,2,6-Hexanetriol; l,2-Dimyristoyl-Sn- Glycero-3-(Phospho-S-(l-Glycerol)); l,2-Dimyristoyl-Sn-Glycero-3-Phosphocholine; 1,2- Dioleoyl-Sn-Glycero-3-Phosphocholine; l,2-Dipalmitoyl-Sn-Glycero-3-(Phospho-Rac-(l- Glycerol)); l,2-Distearoyl-Sn-Glycero-3-(Phospho-Rac-(l-Glycerol)); l,2-Distearoyl-Sn- Glycero-3-Phosphocholine; l-O-Tolylbiguanide; 2-Ethyl- l,6-Hexanediol; Acetic Acid; Acetic Acid, Glacial; Acetic Anhydride; Acetone; Acetone Sodium Bisulfite; Acetylated Lanolin Alcohols; Acetylated Monoglycerides; Acetylcysteine; Acetyltryptophan, DL-; Acrylates Copolymer; Acrylic Acid-Isooctyl Acrylate Copolymer; Acrylic Adhesive 788; Activated Charcoal; Adcote 72A103; Adhesive Tape; Adipic Acid; Aerotex Resin 3730; Alanine; Albumin Aggregated; Albumin Colloidal; Albumin Human; Alcohol; Alcohol, Dehydrated; Alcohol, Denatured; Alcohol, Diluted; Alfadex; Alginic Acid; Alkyl
Ammonium Sulfonic Acid Betaine; Alkyl Aryl Sodium Sulfonate; Allantoin; Allyl .Alpha.-Ionone; Almond Oil; Alpha-Terpineol; Alpha-Tocopherol; Alpha-Tocopherol Acetate, D1-; Alpha-Tocopherol, D1-; Aluminum Acetate; Aluminum Chlorhydroxy Allantoinate; Aluminum Hydroxide; Aluminum Hydroxide - Sucrose, Hydrated; Aluminum Hydroxide Gel; Aluminum Hydroxide Gel F 500; Aluminum Hydroxide Gel F 5000; Aluminum Monostearate; Aluminum Oxide; Aluminum Polyester; Aluminum Silicate; Aluminum Starch Octenylsuccinate; Aluminum Stearate; Aluminum Subacetate; Aluminum Sulfate Anhydrous; Amerchol C; Amerchol-Cab; Aminomethylpropanol;
Ammonia; Ammonia Solution; Ammonia Solution, Strong; Ammonium Acetate;
Ammonium Hydroxide; Ammonium Lauryl Sulfate; Ammonium Nonoxynol-4 Sulfate; Ammonium Salt Of C-12-C-15 Linear Primary Alcohol Ethoxylate; Ammonium Sulfate; Ammonyx; Amphoteric-2; Amphoteric-9; Anethole; Anhydrous Citric Acid; Anhydrous Dextrose; Anhydrous Lactose; Anhydrous Trisodium Citrate; Aniseed Oil; Anoxid Sbn; Antifoam; Antipyrine; Apaflurane; Apricot Kernel Oil Peg-6 Esters; Aquaphor; Arginine; Arlacel; Ascorbic Acid; Ascorbyl Palmitate; Aspartic Acid; Balsam Peru; Barium Sulfate; Beeswax; Beeswax, Synthetic; Beheneth-lO; Bentonite; Benzalkonium Chloride;
Benzenesulfonic Acid; Benzethonium Chloride; Benzododecinium Bromide; Benzoic Acid; Benzyl Alcohol; Benzyl Benzoate; Benzyl Chloride; Betadex; Bibapcitide; Bismuth Subgallate; Boric Acid; Brocrinat; Butane; Butyl Alcohol; Butyl Ester Of Vinyl Methyl Ether/Maleic Anhydride Copolymer (125000 Mw); Butyl Stearate; Butylated
Hydroxyanisole; Butylated Hydroxy toluene; Butylene Glycol; Butylparaben; Butyric Acid; C20-40 Pareth-24; Caffeine; Calcium; Calcium Carbonate; Calcium Chloride; Calcium Gluceptate; Calcium Hydroxide; Calcium Lactate; Calcobutrol; Caldiamide Sodium;
Caloxetate Trisodium; Calteridol Calcium; Canada Balsam; Caprylic/Capric Triglyceride; Caprylic/Capric/Stearic Triglyceride; Captan; Captisol; Caramel; Carbomer 1342;
Carbomer 1382; Carbomer 934; Carbomer 934p; Carbomer 940; Carbomer 941; Carbomer 980; Carbomer 981; Carbomer Homopolymer Type B (Allyl Pentaerythritol Crosslinked); Carbomer Homopolymer Type C (Allyl Pentaerythritol Crosslinked); Carbon Dioxide; Carboxy Vinyl Copolymer; Carboxymethylcellulose; Carboxymethylcellulose Sodium; Carboxypolymethylene; Carrageenan; Carrageenan Salt; Castor Oil; Cedar Leaf Oil;
Cellulose; Cellulose, Microcrystalline; Cerasynt-Se; Ceresin; Ceteareth-l2; Ceteareth-l5; Ceteareth-30; Cetearyl Alcohol/Ceteareth-20; Cetearyl Ethylhexanoate; Ceteth-lO; Ceteth- 2; Ceteth-20; Ceteth-23; Cetostearyl Alcohol; Cetrimonium Chloride; Cetyl Alcohol; Cetyl Esters Wax; Cetyl Palmitate; Cetylpyridinium Chloride; Chlorobutanol; Chlorobutanol Hemihydrate; Chlorobutanol, Anhydrous; Chlorocresol; Chloroxylenol; Cholesterol;
Choleth; Choleth-24; Citrate; Citric Acid; Citric Acid Monohydrate; Citric Acid, Hydrous; Cocamide Ether Sulfate; Cocamine Oxide; Coco Betaine; Coco Diethanolamide; Coco Monoethanolamide; Cocoa Butter; Coco-Glycerides; Coconut Oil; Coconut Oil,
Hydrogenated; Coconut Oil/Palm Kernel Oil Glycerides, Hydrogenated; Cocoyl
Caprylocaprate; Cola Nitida Seed Extract; Collagen; Coloring Suspension; Corn Oil;
Cottonseed Oil; Cream Base; Creatine; Creatinine; Cresol; Croscarmellose Sodium;
Crospovidone; Cupric Sulfate; Cupric Sulfate Anhydrous; Cyclomethicone;
Cyclomethicone/Dimethicone Copolyol; Cysteine; Cysteine Hydrochloride; Cysteine Hydrochloride Anhydrous; Cysteine, D1-; D&C Red No. 28; D&C Red No. 33; D&C Red No. 36; D&C Red No. 39; D&C Yellow No. 10; Dalfampridine; Daubert 1-5 Pestr (Matte) l64z; Decyl Methyl Sulfoxide; Dehydag Wax Sx; Dehydroacetic Acid; Dehymuls E; Denatonium Benzoate; Deoxycholic Acid; Dextran; Dextran 40; Dextrin; Dextrose;
Dextrose Monohydrate; Dextrose Solution; Diatrizoic Acid; Diazolidinyl Urea;
Dichlorobenzyl Alcohol; Dichlorodifluorome thane; Dichlorotetrafluoroethane;
Diethanolamine; Diethyl Pyrocarbonate; Diethyl Sebacate; Diethylene Glycol Monoethyl Ether; Diethylhexyl Phthalate; Dihydroxyaluminum Aminoacetate; Diisopropanolamine; Diisopropyl Adipate; Diisopropyl Dilinoleate; Dimethicone 350; Dimethicone Copolyol; Dimethicone Mdx4-42l0; Dimethicone Medical Fluid 360; Dimethyl Isosorbide; Dimethyl Sulfoxide; Dimethylaminoethyl Methacrylate-Butyl Methacrylate - Methyl Methacrylate Copolymer; Dimethyldioctadecylammonium Bentonite;
Dimethylsiloxane/Methylvinylsiloxane Copolymer; Dinoseb Ammonium Salt;
Dipalmitoylphosphatidylglycerol, D1-; Dipropylene Glycol; Disodium
Cocoamphodiacetate; Disodium Laureth Sulfosuccinate; Disodium Lauryl Sulfosuccinate; Disodium Sulfosalicylate; Disofenin; Divinylbenzene Styrene Copolymer; Dmdm
Hydantoin; Docosanol; Docusate Sodium; Duro-Tak 280-2516; Duro-Tak 387-2516; Duro- Tak 80-1196; Duro-Tak 87-2070; Duro-Tak 87-2194; Duro-Tak 87-2287; Duro-Tak 87- 2296; Duro-Tak 87-2888; Duro-Tak 87-2979; Edetate Calcium Disodium; Edetate Disodium; Edetate Disodium Anhydrous; Edetate Sodium; Edetic Acid; Egg
Phospholipids; Entsufon; Entsufon Sodium; Epilactose; Epitetracycline Hydrochloride; Essence Bouquet 9200; Ethanolamine Hydrochloride; Ethyl Acetate; Ethyl Oleate;
Ethylcelluloses; Ethylene Glycol; Ethylene Vinyl Acetate Copolymer; Ethylenediamine; Ethylenediamine Dihydrochloride; Ethylene-Propylene Copolymer; Ethylene- Vinyl Acetate Copolymer (28% Vinyl Acetate); Ethylene- Vinyl Acetate Copolymer (9%
Vinylacetate); Ethylhexyl Hydroxystearate; Ethylparaben; Eucalyptol; Exametazime; Fat, Edible; Fat, Hard; Fatty Acid Esters; Fatty Acid Pentaerythriol Ester; Fatty Acids; Fatty Alcohol Citrate; Fatty Alcohols; Fd&C Blue No. 1; Fd&C Green No. 3; Fd&C Red No. 4; Fd&C Red No. 40; Fd&C Yellow No. 10 (Delisted); Fd&C Yellow No. 5; Fd&C Yellow No. 6; Ferric Chloride; Ferric Oxide; Flavor 89-186; Flavor 89-259; Flavor Df-l l9; Flavor Df-l530; Flavor Enhancer; Flavor Fig 827118; Flavor Raspberry Pfc-8407; Flavor Rhodia Pharmaceutical No. Rf 451; Fluorochlorohydrocarbons; Formaldehyde; Formaldehyde Solution; Fractionated Coconut Oil; Fragrance 3949-5; Fragrance 520a; Fragrance 6.007; Fragrance 91-122; Fragrance 9128-Y; Fragrance 93498g; Fragrance Balsam Pine No.
5124; Fragrance Bouquet 10328; Fragrance Chemoderm 6401-B; Fragrance Chemoderm 6411; Fragrance Cream No. 73457; Fragrance Cs-28l97; Fragrance Felton 066m;
Fragrance Firmenich 47373; Fragrance Givaudan Ess 9090/lc; Fragrance H-6540;
Fragrance Herbal 10396; Fragrance Nj-l085; Fragrance P O F1-147; Fragrance Pa 52805; Fragrance Pera Derm D; Fragrance Rbd-98l9; Fragrance Shaw Mudge U-7776; Fragrance Tf 044078; Fragrance Ungerer Honeysuckle K 2771; Fragrance Ungerer N5195; Fructose; Gadolinium Oxide; Galactose; Gamma Cyclodextrin; Gelatin; Gelatin, Crosslinked;
Gelfoam Sponge; Gellan Gum (Low Acyl); Gelva 737; Gentisic Acid; Gentisic Acid Ethanolamide; Gluceptate Sodium; Gluceptate Sodium Dihydrate; Gluconolactone;
Glucuronic Acid; Glutamic Acid, D1-; Glutathione; Glycerin; Glycerol Ester Of
Hydrogenated Rosin; Glyceryl Citrate; Glyceryl Isostearate; Glyceryl Laurate; Glyceryl Monostearate; Glyceryl Oleate; Glyceryl Oleate/Propylene Glycol; Glyceryl Palmitate; Glyceryl Ricinoleate; Glyceryl Stearate; Glyceryl Stearate - Laureth-23; Glyceryl Stearate/Peg Stearate; Glyceryl Stearate/Peg- 100 Stearate; Glyceryl Stearate/Peg-40 Stearate; Glyceryl Stearate-Stearamidoethyl Diethylamine; Glyceryl Trioleate; Glycine; Glycine Hydrochloride; Glycol Distearate; Glycol Stearate; Guanidine Hydrochloride; Guar Gum; Hair Conditioner (l8nl95-lm); Heptane; Hetastarch; Hexylene Glycol; High Density Polyethylene; Histidine; Human Albumin Microspheres; Hyaluronate Sodium; Hydrocarbon; Hydrocarbon Gel, Plasticized; Hydrochloric Acid; Hydrochloric Acid, Diluted; Hydrocortisone; Hydrogel Polymer; Hydrogen Peroxide; Hydrogenated Castor Oil; Hydrogenated Palm Oil; Hydrogenated Palm/Palm Kernel Oil Peg-6 Esters;
Hydrogenated Polybutene 635-690; Hydroxide Ion; Hydroxyethyl Cellulose;
Hydroxyethylpiperazine Ethane Sulfonic Acid; Hydroxymethyl Cellulose;
Hydroxy octacosanyl Hydroxystearate; Hydroxypropyl Cellulose; Hydroxypropyl Methylcellulose 2906; Hydroxypropyl-Beta-cyclodextrin; Hypromellose 2208 (15000 Mpa.S); Hypromellose 2910 (15000 Mpa.S); Hypromelloses; Imidurea; Iodine; Todoxamic Acid; Tofetamine Hydrochloride; Irish Moss Extract; Isobutane; Isoceteth-20; Isoleucine; Isooctyl Acrylate; Isopropyl Alcohol; Isopropyl Isostearate; Isopropyl Myristate; Isopropyl Myristate - Myristyl Alcohol; Isopropyl Palmitate; Isopropyl Stearate; Isostearic Acid; Isostearyl Alcohol; Isotonic Sodium Chloride Solution; Jelene; Kaolin; Kathon Cg; Kathon Cg II; Lactate; Lactic Acid; Lactic Acid, D1-; Lactic Acid, L-; Lactobionic Acid; Lactose; Lactose Monohydrate; Lactose, Hydrous; Laneth; Lanolin; Lanolin Alcohol - Mineral Oil; Lanolin Alcohols; Lanolin Anhydrous; Lanolin Cholesterols; Lanolin Nonionic
Derivatives; Lanolin, Ethoxylated; Lanolin, Hydrogenated; Lauralkonium Chloride;
Lauramine Oxide; Laurdimonium Hydrolyzed Animal Collagen; Laureth Sulfate; Laureth- 2; Laureth-23; Laureth-4; Laurie Diethanolamide; Laurie Myristic Diethanolamide;
Lauroyl Sarcosine; Lauryl Lactate; Lauryl Sulfate; Lavandula Angustifolia Flowering Top; Lecithin; Lecithin Unbleached; Lecithin, Egg; Lecithin, Hydrogenated; Lecithin,
Hydrogenated Soy; Lecithin, Soybean; Lemon Oil; Leucine; Levulinic Acid; Lidofenin; Light Mineral Oil; Light Mineral Oil (85 Ssu); Limonene, (+/-)-; Lipocol Sc-l5; Lysine; Lysine Acetate; Lysine Monohydrate; Magnesium Aluminum Silicate; Magnesium Aluminum Silicate Hydrate; Magnesium Chloride; Magnesium Nitrate; Magnesium Stearate; Maleic Acid; Mannitol; Maprofix; Mebrofenin; Medical Adhesive Modified S- 15; Medical Antiform A-L Emulsion; Medronate Disodium; Medronic Acid; Meglumine; Menthol; Metacresol; Metaphosphoric Acid; Methanesulfonic Acid; Methionine; Methyl Alcohol; Methyl Gluceth-10; Methyl Gluceth-20; Methyl Gluceth-20 Sesquistearate; Methyl Glucose Sesquistearate; Methyl Laurate; Methyl Pyrrolidone; Methyl Salicylate; Methyl Stearate; Methylboronic Acid; Methylcellulose (4000 Mpa.S); Methylcelluloses; Methylchloroisothiazolinone; Methylene Blue; Methylisothiazolinone; Methylparaben; Microcrystalline Wax; Mineral Oil; Mono And Diglyceride; Monostearyl Citrate;
Monothioglycerol; Multisterol Extract; Myristyl Alcohol; Myristyl Lactate; Myristyl- . Gamma. -Picolinium Chloride; N-(Carbamoyl-Methoxy Peg-40)- 1 ,2-Distearoyl-Cephalin Sodium; N,N-Dimethylacetamide; Niacinamide; Nioxime; Nitric Acid; Nitrogen;
Nonoxynol Iodine; Nonoxynol-15; Nonoxynol-9; Norflurane; Oatmeal; Octadecene- 1/Maleic Acid Copolymer; Octanoic Acid; Octisalate; Octoxynol-1; Octoxynol-40;
Octoxynol-9; Octyldodecanol; Octylphenol Polymethylene; Oleic Acid; Oleth-lO/Oleth-5; Oleth-2; Oleth-20; Oleyl Alcohol; Oleyl Oleate; Olive Oil; Oxidronate Disodium;
Oxy quinoline; Palm Kernel Oil; Palmitamine Oxide; Parabens; Paraffin; Paraffin, White Soft; Parfum Creme 45/3; Peanut Oil; Peanut Oil, Refined; Pectin; Peg 6-32 Stearate/Glycol Stearate; Peg Vegetable Oil; Peg-lOO Stearate; Peg-l2 Glyceryl Laurate; Peg-l20 Glyceryl Stearate; Peg-l20 Methyl Glucose Dioleate; Peg-l5 Cocamine; Peg-l50 Distearate; Peg-2 Stearate; Peg-20 Sorbitan Isostearate; Peg-22 Methyl Ether/Dodecyl Glycol Copolymer; Peg-25 Propylene Glycol Stearate; Peg-4 Dilaurate; Peg-4 Laurate; Peg-40 Castor Oil; Peg-40 Sorbitan Diisostearate; Peg-45/Dodecyl Glycol Copolymer; Peg-5 Oleate; Peg-50 Stearate; Peg-54 Hydrogenated Castor Oil; Peg-6 Isostearate; Peg-60 Castor Oil; Peg-60 Hydrogenated Castor Oil; Peg-7 Methyl Ether; Peg-75 Lanolin; Peg-8 Laurate; Peg-8 Stearate; Pegoxol 7 Stearate; Pentadecalactone; Pentaerythritol Cocoate; Pentasodium Pentetate; Pentetate Calcium Trisodium; Pentetic Acid; Peppermint Oil; Perflutren; Perfume 25677; Perfume Bouquet; Perfume E-1991; Perfume Gd 5604;
Perfume Tana 90/42 Scba; Perfume W-1952-1; Petrolatum; Petrolatum, White; Petroleum Distillates; Phenol; Phenol, Liquefied; Phenonip; Phenoxy ethanol; Phenylalanine;
Phenylethyl Alcohol; Phenylmercuric Acetate; Phenylmercuric Nitrate; Phosphatidyl Glycerol, Egg; Phospholipid; Phospholipid, Egg; Phospholipon 90g; Phosphoric Acid; Pine Needle Oil (Pinus Sylvestris); Piperazine Hexahydrate; Plastibase-50w; Polacrilin;
Polidronium Chloride; Poloxamer 124; Poloxamer 181; Poloxamer 182; Poloxamer 188; Poloxamer 237; Poloxamer 407; Poly(Bis(P-Carboxyphenoxy)Propane
Anhydride) :Sebacic Acid;
Poly(Dimethylsiloxane/Methylvinylsiloxane/Methylhydrogensiloxane) Dimethylvinyl Or Dimethylhydroxy Or Trimethyl Endblocked; Poly(Dl-Lactic-Co-Glycolic Acid), (50:50; Poly(Dl-Lactic-Co-Glycolic Acid), Ethyl Ester Terminated, (50:50; Polyacrylic Acid (250000 Mw); Polybutene (1400 Mw); Polycarbophil; Polyester; Polyester Polyamine Copolymer; Polyester Rayon; Polyethylene Glycol 1000; Polyethylene Glycol 1450;
Polyethylene Glycol 1500; Polyethylene Glycol 1540; Polyethylene Glycol 200;
Polyethylene Glycol 300; Polyethylene Glycol 300-1600; Polyethylene Glycol 3350; Polyethylene Glycol 400; Polyethylene Glycol 4000; Polyethylene Glycol 540;
Polyethylene Glycol 600; Polyethylene Glycol 6000; Polyethylene Glycol 8000;
Polyethylene Glycol 900; Polyethylene High Density Containing Ferric Oxide Black (<l%); Polyethylene Low Density Containing Barium Sulfate (20-24%); Polyethylene T; Polyethylene Terephthalates; Polyglactin; Poly glyceryl-3 Oleate; Poly glyceryl-4 Oleate; Polyhydroxyethyl Methacrylate; Polyisobutylene; Polyisobutylene (1100000 Mw);
Polyisobutylene (35000 Mw); Polyisobutylene 178-236; Polyisobutylene 241-294;
Polyisobutylene 35-39; Polyisobutylene Low Molecular Weight; Polyisobutylene Medium Molecular Weight; Polyisobutylene/Polybutene Adhesive; Polylactide; Polyols;
Polyoxyethylene - Polyoxypropylene 1800; Polyoxyethylene Alcohols; Polyoxyethylene Fatty Acid Esters; Polyoxyethylene Propylene; Polyoxyl 20 Cetostearyl Ether; Polyoxyl 35 Castor Oil; Polyoxyl 40 Hydrogenated Castor Oil; Polyoxyl 40 Stearate; Polyoxyl 400 Stearate; Polyoxyl 6 And Polyoxyl 32 Palmitostearate; Polyoxyl Distearate; Polyoxyl Glyceryl Stearate; Polyoxyl Lanolin; Polyoxyl Palmitate; Polyoxyl Stearate;
Polypropylene; Polypropylene Glycol; Polyquatemium-lO; Polyquatemium-7 (70/30 Acrylamide/Dadmac; Polysiloxane; Polysorbate 20; Polysorbate 40; Polysorbate 60;
Polysorbate 65; Polysorbate 80; Polyurethane; Polyvinyl Acetate; Polyvinyl Alcohol; Polyvinyl Chloride; Polyvinyl Chloride-Polyvinyl Acetate Copolymer; Polyvinylpyridine; Poppy Seed Oil; Potash; Potassium Acetate; Potassium Alum; Potassium Bicarbonate; Potassium Bisulfite; Potassium Chloride; Potassium Citrate; Potassium Hydroxide;
Potassium Metabisulfite; Potassium Phosphate, Dibasic; Potassium Phosphate, Monobasic; Potassium Soap; Potassium Sorbate; Povidone Acrylate Copolymer; Povidone Hydrogel; Povidone K17; Povidone K25; Povidone K29/32; Povidone K30; Povidone K90; Povidone K90f; Povidone/Eicosene Copolymer; Povidones; Ppg-l2/Smdi Copolymer; Ppg-l5 Stearyl Ether; Ppg-20 Methyl Glucose Ether Distearate; Ppg-26 Oleate; Product Wat; Proline; Promulgen D; Promulgen G; Propane; Propellant A-46; Propyl Gallate; Propylene Carbonate; Propylene Glycol; Propylene Glycol Diacetate; Propylene Glycol Dicaprylate; Propylene Glycol Monolaurate; Propylene Glycol Monopalmitostearate; Propylene Glycol Palmitostearate; Propylene Glycol Ricinoleate; Propylene Glycol/Diazolidinyl
Urea/Methylparaben/Propylparben; Propylparaben; Protamine Sulfate; Protein
Hydrolysate; Pvm Ma Copolymer; Quaternium-l5; Quaternium-l5 Cis-Form; Quaternium- 52; Ra-2397; Ra-30l l; Saccharin; Saccharin Sodium; Saccharin Sodium Anhydrous; Safflower Oil; Sd Alcohol 3a; Sd Alcohol 40; Sd Alcohol 40-2; Sd Alcohol 40b; Sepineo P 600; Serine; Sesame Oil; Shea Butter; Silastic Brand Medical Grade Tubing; Silastic Medical Adhesive, Silicone Type A; Silica, Dental; Silicon; Silicon Dioxide; Silicon Dioxide, Colloidal; Silicone; Silicone Adhesive 4102; Silicone Adhesive 4502; Silicone Adhesive Bio-Psa Q7-4201; Silicone Adhesive Bio-Psa Q7-4301; Silicone Emulsion; Silicone/Polyester Film Strip; Simethicone; Simethicone Emulsion; Sipon Ls 20np; Soda Ash; Sodium Acetate; Sodium Acetate Anhydrous; Sodium Alkyl Sulfate; Sodium
Ascorbate; Sodium Benzoate; Sodium Bicarbonate; Sodium Bisulfate; Sodium Bisulfite; Sodium Borate; Sodium Borate Decahydrate; Sodium Carbonate; Sodium Carbonate Decahydrate; Sodium Carbonate Monohydrate; Sodium Cetostearyl Sulfate; Sodium Chlorate; Sodium Chloride; Sodium Chloride Injection; Sodium Chloride Injection, Bacteriostatic; Sodium Cholesteryl Sulfate; Sodium Citrate; Sodium Cocoyl Sarcosinate; Sodium Desoxycholate; Sodium Dithionite; Sodium Dodecylbenzenesulfonate; Sodium Formaldehyde Sulfoxylate; Sodium Gluconate; Sodium Hydroxide; Sodium Hypochlorite; Sodium Iodide; Sodium Lactate; Sodium Lactate, L-; Sodium Laureth-2 Sulfate; Sodium Laureth-3 Sulfate; Sodium Laureth-5 Sulfate; Sodium Lauroyl Sarcosinate; Sodium Lauryl Sulfate; Sodium Lauryl Sulfoacetate; Sodium Metabisulfite; Sodium Nitrate; Sodium Phosphate; Sodium Phosphate Dihydrate; Sodium Phosphate, Dibasic; Sodium Phosphate, Dibasic, Anhydrous; Sodium Phosphate, Dibasic, Dihydrate; Sodium Phosphate, Dibasic, Dodecahydrate; Sodium Phosphate, Dibasic, Heptahydrate; Sodium Phosphate,
Monobasic; Sodium Phosphate, Monobasic, Anhydrous; Sodium Phosphate, Monobasic, Dihydrate; Sodium Phosphate, Monobasic, Monohydrate; Sodium Polyacrylate (2500000 Mw); Sodium Pyrophosphate; Sodium Pyrrolidone Carboxylate; Sodium Starch Glycolate; Sodium Succinate Hexahydrate; Sodium Sulfate; Sodium Sulfate Anhydrous; Sodium Sulfate Decahydrate; Sodium Sulfite; Sodium Sulfosuccinated Undecyclenic
Monoalky lolamide; Sodium Tartrate; Sodium Thioglycolate; Sodium Thiomalate; Sodium Thiosulfate; Sodium Thiosulfate Anhydrous; Sodium Trimetaphosphate; Sodium
Xylenesulfonate; Somay 44; Sorbic Acid; Sorbitan; Sorbitan Isostearate; Sorbitan
Monolaurate; Sorbitan Monooleate; Sorbitan Monopalmitate; Sorbitan Monostearate; Sorbitan Sesquioleate; Sorbitan Trioleate; Sorbitan Tristearate; Sorbitol; Sorbitol Solution; Soybean Flour; Soybean Oil; Spearmint Oil; Spermaceti; Squalane; Stabilized Oxychloro Complex; Stannous 2-Ethylhexanoate; Stannous Chloride; Stannous Chloride Anhydrous; Stannous Fluoride; Stannous Tartrate; Starch; Starch 1500, Pregelatinized; Starch, Corn; Stearalkonium Chloride; Stearalkonium Hectorite/Propylene Carbonate; Stearamidoethyl Diethylamine; Steareth-10; Steareth-100; Steareth-2; Steareth-20; Steareth-21; Steareth-40; Stearic Acid; Stearic Diethanolamide; Stearoxytrimethylsilane; Steartrimonium
Hydrolyzed Animal Collagen; Stearyl Alcohol; Sterile Water For Inhalation;
Styrene/Isoprene/Styrene Block Copolymer; Succimer; Succinic Acid; Sucralose; Sucrose; Sucrose Distearate; Sucrose Polyesters; Sulfacetamide Sodium; Sulfobutylether .Beta.- Cyclodextrin; Sulfur Dioxide; Sulfuric Acid; Sulfurous Acid; Surfactol Qs; Tagatose, D-; Talc; Tall Oil; Tallow Glycerides; Tartaric Acid; Tartaric Acid, D1-; Tenox; Tenox-2; Tert- Butyl Alcohol; Tert-Butyl Hydroperoxide; Tert-Butylhydroquinone; Tetrakis(2- Methoxyisobutylisocyanide)Copper(I) Tetrafluoroborate; Tetrapropyl Orthosilicate;
Tetrofosmin; Theophylline; Thimerosal; Threonine; Thymol; Tin; Titanium Dioxide; Tocopherol; Tocophersolan; Total parenteral nutrition, lipid emulsion; Triacetin;
Tricaprylin; Trichloromonofluoromethane; Trideceth-lO; Triethanolamine Lauryl Sulfate; Trifluoroacetic Acid; Triglycerides, Medium Chain; Trihydroxystearin; Trilaneth-4 Phosphate; Trilaureth-4 Phosphate; Trisodium Citrate Dihydrate; Trisodium Hedta; Triton 720; Triton X-200; Trolamine; Tromantadine; Tromethamine (TRIS); Tryptophan;
Tyloxapol; Tyrosine; Undecylenic Acid; Union 76 Amsco-Res 6038; Urea; Valine;
Vegetable Oil; Vegetable Oil Glyceride, Hydrogenated; Vegetable Oil, Hydrogenated; Versetamide; Viscarin; Viscose/Cotton; Vitamin E; Wax, Emulsifying; Wecobee Fs; White Ceresin Wax; White Wax; Xanthan Gum; Zinc; Zinc Acetate; Zinc Carbonate; Zinc Chloride; and Zinc Oxide.
[00221] Pharmaceutical composition formulations disclosed herein may include cations or anions. In one embodiment, the formulations include metal cations such as, but not limited to, Zn2+, Ca2+, Cu2+, Mn2+, Mg+ and combinations thereof. As a non-limiting example, formulations may include polymers and complexes with a metal cation ( See e.g. , U.S. Pat. Nos. 6,265,389 and 6,555,525, each of which is herein incorporated by reference in its entirety).
[00222] Formulations may also include one or more pharmaceutically acceptable salts.
As used herein,“pharmaceutically acceptable salts” refers to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or base moiety to its salt form (e.g., by reacting the free base group with a suitable organic acid). Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. Representative acid addition salts include acetate, acetic acid, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzene sulfonic acid, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate, heptonate, hexanoate, hydrobromide, hydrochloride, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3- phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, toluenesulfonate, undecanoate, valerate salts, and the like.
Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like, as well as nontoxic ammonium, quaternary ammonium, and amine cations, including, but not limited to ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, and the like. The pharmaceutically acceptable salts of the present disclosure include the conventional non-toxic salts of the parent compound formed, for example, from non-toxic inorganic or organic acids.
[00223] Solvates may be prepared by crystallization, recrystallization, or precipitation from a solution that includes organic solvents, water, or a mixture thereof. Examples of suitable solvents are ethanol, water (for example, mono-, di-, and trl -hydrates), N- methylpyrrolidinone (NMP), dimethyl sulfoxide (DMSO), NN '-di methyl formamide (DMF), 7V,7V’-dimethylacetamide (DMAC), l,3-dimethyl-2-imidazolidinone (DMEU), 1,3- dimethyl-3,4,5,6-tetrahydro-2-(lH)-pyrimidinone (DMPU), acetonitrile (ACN), propylene glycol, ethyl acetate, benzyl alcohol, 2-pyrrolidone, benzyl benzoate, and the like. When water is the solvent, the solvate is referred to as a“hydrate.”
V. ADMINISTRATION AND DOSING
Administration
[00224] The terms "administering" and "introducing" are used interchangeable herein and refer to the delivery of the pharmaceutical composition into a cell or a subject. In the case of delivery to a subject, the pharmaceutical composition is delivered by a method or route that results in at least partial localization of the introduced cells at a desired site, such as hepatocytes, such that a desired effect(s) is produced.
[00225] In one aspect of the method, the pharmaceutical composition may be
administered via a route such as, but not limited to, enteral (into the intestine), gastroenteral, epidural (into the dura matter), oral (by way of the mouth), transdermal, peridural, intracerebral (into the cerebrum), intracerebro ventricular (into the cerebral ventricles), epicutaneous (application onto the skin), intradermal, (into the skin itself), subcutaneous (under the skin), nasal administration (through the nose), intravenous (into a vein), intravenous bolus, intravenous drip, intraarterial (into an artery), intramuscular (into a muscle), intracardiac (into the heart), intraosseous infusion (into the bone marrow), intrathecal (into the spinal canal), intraperitoneal, (infusion or injection into the peritoneum), intravesical infusion, intravitreal, (through the eye), intracavernous injection (into a pathologic cavity) intracavitary (into the base of the penis), intravaginal
administration, intrauterine, extra- amniotic administration, transdermal (diffusion through the intact skin for systemic distribution), transmucosal (diffusion through a mucous membrane), transvaginal, insufflation (snorting), sublingual, sublabial, enema, eye drops (onto the conjunctiva), in ear drops, auricular (in or by way of the ear), buccal (directed toward the cheek), conjunctival, cutaneous, dental (to a tooth or teeth), electro-osmosis, endocervical, endosinusial, endotracheal, extracorporeal, hemodialysis, infiltration, interstitial, intra-abdominal, intra-amniotic, intra-articular, intrabiliary, intrabronchial, intrabursal, intracartilaginous (within a cartilage), intracaudal (within the cauda equine), intracistemal (within the cistema magna cerebellomedularis), intracorneal (within the cornea), dental intracomal, intracoronary (within the coronary arteries), intracorporus cavernosum (within the dilatable spaces of the corporus cavernosa of the penis), intradiscal (within a disc), intraductal (within a duct of a gland), intraduodenal (within the duodenum), intradural (within or beneath the dura), intraepidermal (to the epidermis), intraesophageal (to the esophagus), intragastric (within the stomach), intragingival (within the gingivae), intraileal (within the distal portion of the small intestine), intralesional (within or introduced directly to a localized lesion), intraluminal (within a lumen of a tube), intralymphatic (within the lymph), intramedullary (within the marrow cavity of a bone), intrameningeal (within the meninges), intramyocardial (within the myocardium), intraocular (within the eye), intraovarian (within the ovary), intrapericardial (within the pericardium), intrapleural (within the pleura), intraprostatic (within the prostate gland), intrapulmonary (within the lungs or its bronchi), intrasinal (within the nasal or periorbital sinuses), intraspinal (within the vertebral column), intrasynovial (within the synovial cavity of a joint), intratendinous (within a tendon), intratesticular (within the testicle), intrathecal (within the cerebrospinal fluid at any level of the cerebrospinal axis), intrathoracic (within the thorax), intratubular (within the tubules of an organ), intratumor (within a tumor), intratympanic (within the aurus media), intravascular (within a vessel or vessels), intraventricular (within a ventricle), iontophoresis (by means of electric current where ions of soluble salts migrate into the tissues of the body), irrigation (to bathe or flush open wounds or body cavities), laryngeal (directly upon the larynx), nasogastric (through the nose and into the stomach), occlusive dressing technique (topical route administration which is then covered by a dressing which occludes the area), ophthalmic (to the external eye), oropharyngeal (directly to the mouth and pharynx), parenteral, percutaneous, periarticular, peridural, perineural, periodontal, rectal, respiratory (within the respiratory tract by inhaling orally or nasally for local or systemic effect), retrobulbar (behind the pons or behind the eyeball), intramyocardial (entering the myocardium), soft tissue,
subarachnoid, subconjunctival, submucosal, topical, transplacental (through or across the placenta), transtracheal (through the wall of the trachea), transtympanic (across or through the tympanic cavity), ureteral (to the ureter), urethral (to the urethra), vaginal, caudal block, diagnostic, nerve block, biliary perfusion, cardiac perfusion, photopheresis and spinal.
[00226] Modes of administration include injection, infusion, instillation, and/or ingestion. "Injection" includes, without limitation, intravenous, intramuscular, intra-arterial, intrathecal, intraventricular, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, sub capsular, subarachnoid, intraspinal, intracerebro spinal, and intrasternal injection and infusion. In some examples, the route is intravenous. For the delivery of cells, administration by injection or infusion can be made.
[00227] The cells can be administered systemically. The phrases "systemic
administration," "administered systemically", "peripheral administration" and
"administered peripherally" refer to the administration other than directly into a target site, tissue, or organ, such that it enters, instead, the subject's circulatory system and, thus, is subject to metabolism and other like processes.
Dosing
[00228] The term "effective amount" refers to the amount of the active ingredient needed to prevent or alleviate at least one or more signs or symptoms of a specific disease and/or condition, and relates to a sufficient amount of a composition to provide the desired effect. The term "therapeutically effective amount" therefore refers to an amount of active ingredient or a composition comprising the active ingredient that is sufficient to promote a particular effect when administered to a typical subject. An effective amount would also include an amount sufficient to prevent or delay the development of a symptom of the disease, alter the course of a symptom of the disease (for example but not limited to, slow the progression of a symptom of the disease), or reverse a symptom of the disease. It is understood that for any given case, an appropriate "effective amount" can be determined by one of ordinary skill in the art using routine experimentation.
[00229] The pharmaceutical, diagnostic, or prophylactic compositions of the present disclosure may be administered to a subject using any amount and any route of administration effective for preventing, treating, managing, or diagnosing diseases, disorders and/or conditions. The exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the disease, the particular composition, its mode of administration, its mode of activity, and the like. The subject may be a human, a mammal, or an animal. Compositions in accordance with the disclosure are typically formulated in unit dosage form for ease of administration and uniformity of dosage. It will be understood, however, that the total daily usage of the compositions of the present disclosure may be decided by the attending physician within the scope of sound medical judgment. The specific therapeutically effective,
prophylactically effective, or appropriate diagnostic dose level for any particular individual will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific payload employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, and route of administration; the duration of the treatment; drugs used in combination or coincidental with the active ingredient; and like factors well known in the medical arts.
[00230] In certain embodiments, pharmaceutical compositions in accordance with the present disclosure may be administered at dosage levels sufficient to deliver from about 0.01 mg/kg to about 100 mg/kg, from about 0.01 mg/kg to about 0.05 mg/kg, from about 0.05 mg/kg to about 0.5 mg/kg, from about 0.01 mg/kg to about 50 mg/kg, from about 0.1 mg/kg to about 40 mg/kg, from about 0.5 mg/kg to about 30 mg/kg, from about 0.01 mg/kg to about 10 mg/kg, from about 0.1 mg/kg to about 10 mg/kg, or from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic, diagnostic, or prophylactic, effect.
[00231] The desired dosage of the composition present disclosure may be delivered only once, three times a day, two times a day, once a day, every other day, every third day, every week, every two weeks, every three weeks, or every four weeks. In certain embodiments, the desired dosage may be delivered using multiple administrations (e.g. , two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, or more administrations). When multiple administrations are employed, split dosing regimens such as those described herein may be used. As used herein, a“split dose” is the division of “single unit dose” or total daily dose into two or more doses, e.g., two or more
administrations of the“single unit dose”. As used herein, a“single unit dose” is a dose of any therapeutic administered in one dose/at one time/single route/single point of contact, i.e., single administration event.
[00232] The present disclosure is further illustrated by the following non- limiting examples.
VII. EXAMPLES
Example 1. Experimental procedures
A. Human hepatocvte cell culture
[00233] Human hepatocytes were obtained from two donors from Massachusetts General Hospital, namely MGH54 and MGH63, and one donor from Lonza, namely HUM4111B. Cryopreserved hepatocytes were cultured in plating media for 16 hours, transferred to maintenance media for 4 hours. Cultured on serum-free media for 2 hours, then a compound was added. The hepatocytes were maintained on the serum-free media for 16 hours prior to gene expression analysis. Primary human hepatocytes were stored in the vapor phase of a liquid nitrogen freezer (about -l30°C).
[00234] To seed the primary human hepatocytes, vials of cells were retrieved from the LN2 freezer, thawed in a 37°C water bath, and swirled gently until only a sliver of ice remains. Using a lOml serological pipet, cells were gently pipetted out of the vial and gently pipetted down the side of 50mL conical tube containing 20mL cold thaw medium. The vial was rinsed with about lmL of thaw medium, and the rinse was added to the conical tube. Up to 2 vials were added to one tube of 20mL thaw medium.
[00235] The conical tube(s) were gently inverted 2-3 times and centrifuged at 100 g for 10 minutes at 4°C with reduced braking (e.g. 4 out of 9). The thaw medium slowly was slowly aspirated to avoid the pellet. 4 mL cold plating medium was added slowly down the side (8 mL if combined 2 vials to 1 tube), and the vial was inverted gently several times to resuspend cells.
[00236] Cells were kept on ice until 100 pl of well-mixed cells were added to 400 mΐ diluted Trypan blue and mixed by gentle inversion. They were counted using a hemocytometer (or Cellometer), and viability and viable cells/mL were noted. Cells were diluted to a desired concentration and seeded on collagen I-coated plates. Cells were pipetted slowly and gently onto plate, only 1-2 wells at a time. The remaining cells were mixed in the tubes frequently by gentle inversion. Cells were seeded at about 8.5xl06 cells per plate in 6 mL cold plating medium (lOcm). Alternatively, l.5xl06 per well for a 6-well plate (1 mL medium/well); 7xl05 per well for l2-well plate (0.5 mL/well); or 3.75xl05 per well for a 24-well plate (0.5 mL/well)
[00237] After all cells and medium were added to the plate, the plate was transferred to an incubator (37°C, 5% CC about 90% humidity) and rocked forwards and backwards, then side to side several times each to distribute cells evenly across the plate or wells. The plate(s) were rocked again every 15 minutes for the first hour post-plating. About 4 hours post-plating (or first thing the morning if cells were plated in the evening), cells were washed once with PBS and complete maintenance medium was added. The primary human hepatocytes were maintained in the maintenance medium and transferred to fresh medium daily.
B. Starvation and compound treatment of human hepatocytes
[00238] Human hepatocytes cultured as described above were plated in 24- well format, adding 375,000 cells per well in a volume of 500 ul plating medium. Four hours before treatment, cells were washed with PBS and the medium was changed to either: fresh maintenance medium (complete) or modified maintenance medium.
[00239] Compound stocks were prepared at lOOOx final concentration and added in a 2- step dilution to the medium to reduce risk of a compound precipitating out of solution when added to the cells, and to ensure reasonable pipetting volumes. One at a time, each compound was first diluted lO-fold in warm (about 37°C) modified maintenance medium (initial dilution = ID), mixed by vortexing, and the ID was diluted lOO-fold into the cell culture (e.g. 5.1 pl into 1 well of a 24- well plate containing 0.5 mL medium). The plate was mixed by carefully swirling and after all wells were treated and returned to the incubator overnight. If desired, separate plates/wells were treated with vehicle-only controls and/or positive controls. If using multi-well plates, controls were included on each plate. After about 18 hours, cells were harvested for further analysis, e.g., ChIP-seq, RNA- seq, ATAC-seq, etc.
C. Mouse heuatocvte cell culture and compound treatment
[00240] Female C57BL/6 mouse hepatocytes (F005152-cry opreserved) were purchased from BioreclamationIVT as a pool of 45 donors. Cells were plated in InvitroGRO CP Rodent Medium (Z990028) and Torpedo Rodent Antibiotic Mix (Z99027) on Collagen- coated 24-well plates for 24 hours at 200K cells/well in 0.5 mL media. Compound stocks in 10 mM DMSO, were diluted to 10 uM (with final concentration of 1% DMSO), and applied on cells in biological triplicates. Medium was removed after 20 hours and cells processed for further analysis, e.g. qRT-PCR.
D. Media composition
[00241] The thaw medium contained 6 mL isotonic percoll and 14 mL high glucose DMEM (Invitrogen #11965 or similar). The plating medium contained 100 mL Williams E medium (Invitrogen #A1217601, without phenol red) and the supplement pack #CM3000 from ThermoFisher Plating medium containing 5mL FBS, 10m1 dexamethasone, and 3.6 mL plating/maintenance cocktail. Stock trypan blue (0.4%, Invitrogen #15250) was diluted 1:5 in PBS. Normocin was added at 1:500 to both the thaw medium and the plating medium.
[00242] The ThermoFisher complete maintenance medium contained supplement pack #CM4000 (1 pl dexamethasone and 4 mL maintenance cocktail) and 100 mL Williams E (Invitrogen #A1217601, without phenol red).
[00243] The modified maintenance media had no stimulating factors (dexamethasone, insulin, etc.), and contained lOOmL Williams E (Invitrogen #A1217601, without phenol red), lmL L-Glutamine (Sigma #G75l3) to 2 mM, 1.5 mL HEPES (VWR #J848) to 15 mM, and 0.5 mL penicillin/streptomycin (Invitrogen #15140) to a final concentration of 50 U/mL each.
E. DNA purification
[00244] DNA purification was conducted as described in Ji et ah, PNAS 112(12):3841- 3846 (2015) Supporting Information, which is hereby incorporated by reference in its entirety. One milliliter of 2.5 M glycine was added to each plate of fixed cells and incubated for 5 minutes to quench the formaldehyde. The cells were washed twice with PBS. The cells were pelleted at 1,300 g for 5 minutes at 4°C. Then, 4 x 107 cells were collected in each tube. The cells were lysed gently with 1 mL of ice-cold Nonidet P-40 lysis buffer containing protease inhibitor on ice for 5 minutes (buffer recipes are provided below). The cell lysate was layered on top of 2.5 volumes of sucrose cushion made up of 24% (wt/vol) sucrose in Nonidet P-40 lysis buffer. This sample was centrifuged at 18,000 g for 10 minutes at 4°C to isolate the nuclei pellet (the supernatant represented the cytoplasmic fraction). The nuclei pellet was washed once with PBS/l mM EDTA. The nuclei pellet was resuspended gently with 0.5 mL glycerol buffer followed by incubation for 2 minutes on ice with an equal volume of nuclei lysis buffer. The sample was centrifuged at 16,000 g for 2 minutes at 4°C to isolate the chromatin pellet (the supernatant represented the nuclear soluble fraction). The chromatin pellet was washed twice with PBS/l mM EDTA. The chromatin pellet was stored at -80°C.
[00245] The Nonidet P-40 lysis buffer contained 10 mM Tris-HCl (pH 7.5), 150 mM NaCl, and 0.05% Nonidet P-40. The glycerol buffer contained 20 mM Tris-HCl (pH 7.9), 75 mM NaCl, 0.5 mM EDTA, 0.85 mM DTT, and 50% (vol/vol) glycerol. The nuclei lysis buffer contained 10 mM Hepes (pH 7.6), 1 mM DTT, 7.5 mM MgCK 0.2 mM EDTA, 0.3 M NaCl, 1 M urea, and 1% Nonidet P-40.
F. Chromatin immunoprecipitation sequencing (ChIP-seq)
[00246] ChIP-seq was performed using the following protocol for primary hepatocytes and HepG2 cells to determine the composition and confirm the location of signaling centers.
i. Cell cross-linking
[00247] 2 x 107 cells were used for each run of ChIP-seq. Two ml of fresh 11% formaldehyde (FA) solution was added to 20 ml media on 15 cm plates to reach a 1.1% final concentration. Plates were swirled briefly and incubated at room temperature (RT) for 15 minutes. At the end of incubation, the FA was quenched by adding 1 ml of 2.5 M Glycine to plates and incubating for 5 minutes at RT. The media was discarded to a 1 L beaker, and cells were washed twice with 20 ml ice-cold PBS. PBS (10 ml) was added to plates, and cells were scraped off the plate. The cells were transferred to 15 ml conical tubes, and the tubes were placed on ice. Plates were washed with an additional 4 ml of PBS and combined with cells in 15 ml tubes. Tubes were centrifuged for 5 minutes at 1,500 rpm at 4°C in a tabletop centrifuge. PBS was aspirated, and the cells were flash frozen in liquid nitrogen. Pellets were stored at -80°C until ready to use.
ii. Pre-block magnetic beads
[00248] Thirty pl Protein G beads (per reaction) were added to a l.5ml Protein LoBind Eppendorf tube. The beads were collected by magnet separation at RT for 30 seconds. Beads were washed 3 times with lml of blocking solution by incubating beads on a rotator at 4°C for 10 minutes and collecting the beads with the magnet. Five pg of an antibody was added to the 250 pl of beads in block solution. The mix was transferred to a clean tube, and rotated overnight at 4°C. On the next day, buffer containing antibodies was removed, and beads were washed 3 times with 1.1 ml blocking solution by incubating beads on a rotator at 4°C for 10 minutes and collecting the beads with the magnet. Beads were resuspended in 50 pl of block solution and kept on ice until ready to use. iii. Cell lysis, genomic fragmentation, and chromatin immunoprecipitation
[00249] COMPLETE® protease inhibitor cocktail was added to lysis buffer 1 (LB1) before use. One tablet was dissolved in 1 ml of fLO for a 50x solution. The cocktail was stored in aliquots at -20°C. Cells were resuspended in each tube in 8 ml of LB1 and incubated on a rotator at 4°C for 10 minutes. Nuclei were spun down at 1,350 g for 5 minutes at 4°C. LB1 was aspirated, and cells were resuspended in each tube in 8 ml of LB2 and incubated on a rotator at 4°C for 10 minutes.
[00250] A COVARIS® E220E VOLUTION ultrasonicator was programmed per the manufacturer’s recommendations for high cell numbers. HepG2 cells were sonicated for 12 minutes, and primary hepatocyte samples were sonicated for 10 minutes. Lysates were transferred to clean l.5ml Eppendorf tubes, and the tubes were centrifuged at 20,000 g for 10 minutes at 4°C to pellet debris. The supernatant was transferred to a 2 ml Protein LoBind Eppendorf tube containing pre-blocked Protein G beads with pre -bound antibodies. Fifty pl of the supernatant was saved as input. Input material was kept at -80°C until ready to use. Tubes were rotated with beads overnight at 4°C.
iv. Wash, elution, and cross-link reversal
[00251] All washing steps were performed by rotating tubes for 5 minutes at 4°C. The beads were transferred to clean Protein LoBind Eppendorf tubes with every washing step. Beads were collected in 1.5 ml Eppendorf tube using a magnet. Beads were washed twice with 1.1 ml of sonication buffer. The magnetic stand was used to collect magnetic beads. Beads were washed twice with 1.1 ml of wash buffer 2, and the magnetic stand was used again to collect magnetic beads. Beads were washed twice with 1.1 ml of wash buffer 3.
All residual Wash buffer 3 was removed, and beads were washed once with 1.1 ml TE + 0.2% Triton X-100 buffer. Residual TE + 0.2% Triton X-100 buffer was removed, and beads were washed twice with TE buffer for 30 seconds each time. Residual TE buffer was removed, and beads were resuspended in 300pl of ChIP elution buffer. Two hundred fifty mΐ of ChIP elution buffer was added to 50m1 of input, and the tubes were rotated with beads 1 hour at 65°C. Input sample was incubated overnight at 65°C oven without rotation. Tubes with beads were placed on a magnet, and the eluate was transferred to a fresh DNA LoBind Eppendorf tube. The eluate was incubated overnight at 65°C oven without rotation
v. Chromatin extraction and precipitation
[00252] Input and immunoprecipitant (IP) samples were transferred to fresh tubes, and 300 mΐ of TE buffer was added to IP and Input samples to dilute SDS. RNase A (20 mg/ml) was added to the tubes, and the tubes were incubated at 37°C for 30 minutes.
Following incubation, 3m1 of 1 M CaCL and 7 mΐ of 20 mg/ml Proteinase K were added, and incubated 1.5 hours at 55°C. MaXtract High Density 2 ml gel tubes (Qiagen) were prepared by centrifugation at full speed for 30 seconds at RT. Six hundred mΐ of phenol/chloroform/isoamyl alcohol was added to each proteinase K reaction and transferred in about 1.2 ml mixtures to the MaXtract tubes. Tubes were spun at 16,000 g for 5 minutes at RT. The aqueous phase was transferred to two clean DNA LoBind tubes (300 mΐ in each tube), and 1.5 mΐ glycogen, 30 mΐ of 3M sodium acetate, and 900 mΐ ethanol were added. The mixture was precipitated overnight at -20°C or for 1 hour at -80°C, and spun down at maximum speed for 20 minutes at 4°C. The ethanol was removed, and pellets were washed with 1 ml of 75% ethanol by spinning tubes down at maximum speed for 5 minutes at 4°C. Remnants of ethanol were removed, and pellets were dried for 5 min at RT. Twenty-five mΐ of ¾0 was added to each immunoprecipitant (IP) and input pellet, left standing for 5 minutes, and vortexed briefly. DNA from both tubes was combined to obtain 50 mΐ of IP and 50 mΐ of input DNA for each sample. One mΐ of this DNA was used to measure the amount of pulled down DNA using Qubit dsDNA HS assay (ThermoFisher, #Q32854). The total amount of immunoprecipitated material ranged from several ng (for TFs) to several hundred ng (for chromatin modifications). Six mΐ of DNA was analyzed using qRT-PCR to determine enrichment. The DNA was diluted if necessary. If enrichment was satisfactory, the rest was used for library preparation for DNA sequencing.
vi. Library preparation for DNA sequencing
[00253] Libraries were prepared using NEBNext Ultra II DNA library prep kit for Illumina (NEB, #E7645) using NEBNext Multiplex Oligos for Illumina (NEB, #6609S) according to manufacturer’s instructions with the following modifications. The remaining ChIP sample (about 43m1) and lpg of input samples for library preparations were brought up the volume of 50 mΐ before the End Repair portion of the protocol. End Repair reactions were run in a PCR machine with a heated lid in a 96-well semi-skirted PCR plate
(ThermoFisher, #ABl400) sealed with adhesive plate seals (ThermoFisher, #AB0558) leaving at least one empty well in-between different samples. Undiluted adapters were used for input samples, 1: 10 diluted adapters for 5-l00ng of ChIP material, and 1:25 diluted adapters for less than 5 ng of ChIP material. Ligation reactions were run in a PCR machine with the heated lid off. Adapter ligated DNA was transferred to clean DNA LoBind Eppendorf tubes, and the volume was brought to 96.5 mΐ using fLO. [00254] 200-600bp ChIP fragments were selected using SPRIselect magnetic beads (Beckman Coulter, #B233l7). Thirty mΐ of the beads were added to 96.5 mΐ of ChIP sample to bind fragments that are longer than 600 bp. The shorter fragments were transferred to a fresh DNA LoBind Eppendorf tube. Fifteen mΐ of beads were added to bind the DNA longer than 200bp, and beads were washed with DNA twice using freshly prepared 75% ethanol. DNA was eluted using 17 mΐ of 0.1X TE buffer. About 15m1 was collected.
[00255] Three mΐ of size-selected Input sample and all (15 mΐ) of the ChIP sample was used for PCR. The amount of size-selected DNA was measured using a Qubit dsDNA HS assay. PCR was ran for 7 cycles of for Input and ChIP samples with about 5-10 ng of size- selected DNA, and 12 cycles with less than 5 ng of size-selected DNA. One -half of the PCR product (25 mΐ) was purified with 22.5 mΐ of AMPure XP beads (Beckman Coulter, #A63880) according to the manufacturer’s instructions. PCR product was eluted with 17 mΐ of 0.1X TE buffer, and the amount of PCT product was measured using Qubit dsDNA HS assay. An additional 4 cycles of PCR were run for the second half of samples with less than 5 ng of PCR product, DNA was purified using 22.5 mΐ of AMPure XP beads. The concentration was measured to determine whether there was an increased yield. Both halves were combined, and the volume was brought up to 50 mΐ using fTO.
[00256] A second round of purifications of DNA was ran using 45 mΐ of AMPure XP beads in 17 mΐ of 0.1X TE, and the final yield was measured using Qubit dsDNA HS assay. This protocol produces from 20 ng to 1 mg of PCR product. The quality of the libraries was verified by diluting 1 mΐ of each sample with ¾0 if necessary using the High
Sensitivity Bio Analyzer DNA kit (Agilent, #5067-4626) based on manufacturer’s recommendations
vii. Reagents
[00257] 11% Formaldehyde Solution (50mL) contained l4.9ml of 37% formaldehyde (final cone. 11%), 1 ml of 5 M NaCl (final cone. 0.1 M), IOOmI of 0.5M EDTA (pH 8)
(final cone. 1 mM), 50 mΐ of 0.5 M EGTA (pH 8) (final cone. 0.5 mM), and 2.5 ml 1 M Hepes (pH 7.5) (final cone. 50 mM).
[00258] Block Solution contained 0.5% BSA (w/v) in PBS and 500 mg BSA in 100 ml PBS. Block solution may be prepared up to about 4 days prior to use.
[00259] Lysis buffer 1 (LB1) (500 ml) contained 25 ml of 1 M Hepes-KOH, pH 7.5; 14 ml of 5 M NaCl; 1 ml of 0.5 M EDTA, pH 8.0; 50 ml of 100% Glycerol solution; 25 ml of 10% NP-40; and 12.5 ml of 10% Triton X-100. The pH was adjusted to 7.5. The buffer was sterile-filtered, and stored at 4 °C. The pH was re-checked immediately prior to use.
[00260] Lysis buffer 2 (LB2) (lOOOml) contained 10 ml of 1 M Tris-HCL, pH 8.0; 40 ml of 5 M NaCl; 2 ml of 0.5 M EDTA, pH 8.0; and 2 ml of 0.5 M EGTA, pH 8.0. The pH was adjusted to 8.0. The buffer was sterile-filtered, and stored at 4 °C. The pH was re-checked immediately prior to use.
[00261] Sonication buffer (500 ml) contained 25 ml of 1 M Hepes-KOH, pH 7.5; 14 ml of 5 M NaCl; 1 ml of 0.5 M EDTA, pH 8.0; 50 ml of 10% Triton X-100; 10 ml of 5% Na- deoxycholate; and 5 ml of 10% SDS. The pH was adjusted to 7.5. The buffer was sterile- filtered, and stored at 4 °C. The pH was re-checked immediately prior to use.
[00262] Proteinase inhibitors were included in the LB1, LB2, and Sonication buffer.
[00263] Wash Buffer 2 (500 ml) contained 25 ml of 1 M Hepes-KOH, pH 7.5; 35 ml of 5 M NaCl; 1 ml of 0.5 M EDTA, pH 8.0; 50 ml of 10% Triton X-100; 10 ml of 5% Na- deoxycholate; and 5 ml of 10% SDS. The pH was adjusted to 7.5. The buffer was sterile- filtered, and stored at 4 °C. The pH was re-checked immediately prior to use.
[00264] Wash Buffer 3 (500ml) contained 10ml of 1 M Tris-HCL, pH 8.0; 1 ml of 0.5 M EDTA, pH 8.0; 125 ml of 1 M LiCl solution; 25 ml of 10% NP-40; and 50 ml of 5% Na- deoxycholate. The pH was adjusted to 8.0. The buffer was sterile-filtered, and stored at 4 °C. The pH was re-checked immediately prior to use.
[00265] ChIP elution Buffer (500ml) contained 25ml of 1 M Tris-HCL, pH 8.0; 10 ml of 0.5 M EDTA, pH 8.0; 50 ml of 10% SDS; and 415 ml of ddH20. The pH was adjusted to 7.5. The buffer was sterile-filtered, and stored at 4 °C. The pH was re-checked immediately prior to use.
G. Analysis of ChIP-seq results
[00266] All pass filter reads from each sample were trimmed of sequencing adapters using trim_galore 0.4.4 with default options. Trimmed reads were mapped against the human genome (assembly GRCh38/GCA_00000l405.l5“no alt” analysis set merged with hs38dl/GCA_000786075.2) using bwa version 0.7.15 (Li (2013) arXiv:l303.3997vl) with default parameters. Aligned read duplicates were assessed using picard 2.9.0
(http://broadinstitute.hithub.io/picard) and reads with a MAPQ<20 or those matching standard SAM flags 0x1804 were discarded. Standard QC were applied (read integrity, mapping statistics, library complexity, fragment bias) to remove unsatisfactory samples. Enriched ChIP-seq peaks were identified by comparing samples against whole cell extract controls using MACS2 version 2.1.0 (Zhang et al., Genome Biol. (2008) 9(9):Rl37), with significant peaks selected as those with an adjusted p-value < 0.01. Peaks overlapping known repetitive“blacklist” regions (ENCODE Project Consortium, Nature (2012) 489(7414:57-74) were discarded. ChIP-seq signals were also normalized by read depth and visualized using the UCSC browser.
H. RNA-seq
[00267] This protocol is a modified version of the following protocols: MagMAX m/rVana Total RNA Isolation Kit User Guide (Applied Biosystems #MAN00ll l3l Rev B.0), NEBNext Poly(A) mRNA Magnetic Isolation Module (E7490), and NEBNext Ultra Directional RNA Library Prep Kit for Illumina (E7420) (New England Biosystems #E7490l).
[00268] The MagMAX m/rVana kit instructions (the section titled“Isolate RNA from cells” on pages 14-17) were used for isolation of total RNA from cells in culture. Two hundred pl of Lysis Binding Mix was used per well of the multiwell plate containing adherent cells (usually a 24-well plate).
[00269] For mRNA isolation and library prep, the NEBNext Poly(A) mRNA Magnetic Isolation Module and Directional Prep kit was used. RNA isolated from cells above was quantified, and prepared in 500 pg of each sample in 50 pl of nuclease-free water. This protocol may be run in microfuge tubes or in a 96-well plate.
[00270] The 80% ethanol was prepared fresh, and all elutions are done in 0.1X TE Buffer. For steps requiring Ampure XP beads, beads were at room temperature before use. Sample volumes were measured first and beads were pipetted. Section 1.9B (not 1.9A) was used for NEBNext Multiplex Oligos for Illumina (#E6609). Before starting the PCR enrichment, cDNA was quantified using the Qubit (DNA High Sensitivity Kit, ThermoFisher
#Q32854). The PCR reaction was ran for 12 cycles.
[00271] After purification of the PCR Reaction (Step 1.10), the libraries were quantified using the Qubit DNA High Sensitivity Kit. 1 mΐ of each sample were diluted to 1-2 ng/pl to ran on the Bioanalyzer (DNA High Sensitivity Kit, Agilent # 5067-4626). If Bioanalyzer peaks were not clean (one narrow peak around 300bp), the AMPure XP bead cleanup step was repeated using a 0.9X or 1.0X beads: sample ratio. Then, the samples were quantified again with the Qubit, and ran again on the Bioanalyzer (1-2 ng/pl). [00272] Nuclear RNA from INTACT -purified nuclei or whole neocortical nuclei was converted to cDNA and amplified with the Nugen Ovation RNA-seq System V2. Libraries were sequenced using the Illumina HiSeq 2500.
I. RNA-seq data analysis
[00273] All pass filter reads from each sample were mapped against the human genome (assembly GRCh38/GCA_00000l405.l5“no alt” analysis set merged with
hs38dl/GCA_000786075.2) using two pass mapping via STAR version 2.5.3a (alignment parameters alignIntronMin=20; alignIntronMax= 1000000; outFilterMismatchNmax=999; outFilterMismatchNoverLmax=0.05 ; outFilterType=BySJout;
outFilterMultimapNmax=20; alignS JoverhangMin=8; alignS JDBoverhangMin=l ;
alignMatesGapMax= 1000000) (Dobin et al., Bioinformatics (2012) 29(1): 15-21). Genomic alignments were converted to transcriptome alignments based on reference transcript annotations from the The Human GENCODE Gene Set release 24 (Harrow et al., Genome Res. (2012) 22(9): 1760-1774). Using unique and multimapped transcriptomic alignments, gene-level abundance estimates were computed using RSEM version 1.3.0 (Li and Dewey, BMC Bioinformatics (2011) 12:323) in a strand-aware manner, and including confidence interval sampling calculations, to arrive at posterior mean estimates (PME) of abundances (counts and normalized FPKM - fragments per kilobase of exon per million mapped fragments) from the underlying Bayesian model. Standard QC were applied (read integrity, mapping statistics, library complexity, fragment bias) to remove unsatisfactory samples. Differential gene expression was computed using the negative binomial model
implemented by DESeq2 version 1.16.1 (Love et al., Genome Biol. (2014) 15(12):550). Log2 fold change and significance values were computed using PME count data (with replicates explicitly modeled versus pan-experiment controls), median ratio normalized, using maximum likelihood estimation rather than maximum a posteriori, and disabling the use of Cook’s distance cutoff when determining acceptable adjusted p-values. Significantly differential genes were assigned as those with an adjusted p-value < 0.01, a log2 fold change of >=l or <=-l, and at least one replicate with PME FPKM >=1. RNA-seq signals were also normalized by read depth and visualized using the UCSC browser.
J. ATAC-seq
[00274] Hepatocytes were seeded overnight, then the serum and other factors were removed. After 2-3 hours, the cells were treated with the compound and incubated overnight. The cells were harvested and the nuclei were prepared for the transposition reaction. 50,000 bead bound nuclei were transposed using Tn5 transposase (Illumina FC- 121-1030) as described in Mo et al., 2015, Neuron 86, 1369-1384, which is hereby incorporated by reference in its entirety. After 9-12 cycles of PCR amplification, libraries were sequenced on an Illumina HiSeq 2000. PCR was performed using barcoded primers with extension at 72°C for 5 minutes, PCR, then the final PCR product was sequenced.
[00275] All obtained reads from each sample were trimmed using trim_galore 0.4.1 requiring Phred score > 20 and read length > 30 for data analysis. The trimmed reads were mapped against the human genome (hgl9 build) using Bowtie2 (version 2.2.9) with the parameters: -t -q -N 1 -L 25 -X 2000 no-mixed no-discordant. All unmapped reads, non- uniquely mapped reads and PCR duplicates were removed. All the ATAC-seq peaks were called using MACS2 with the parameters—nolambda -nomodel -q 0.01 -SPMR. The ATAC-seq signal was visualized in the UCSC genome browser. ATAC-seq peaks that were at least 2 kb away from annotated promoters (RefSeq, Ensemble and UCSC Known Gene databases combined) were selected as distal ATAC-seq peaks.
K. qRT-PCR
[00276] qRT-PCR was performed as described in North et ak, PNAS, 107(40) 17315- 17320 (2010), which is hereby incorporated by reference in its entirety. Prior to qRT-PCR analysis, cell medium was removed and replaced with RLT Buffer for RNA extraction (Qiagen RNeasy 96 QIAcube HT Kit Cat#74l7l). Cells were processed for RNA extraction using RNeasy 96 kit (Qiagen Cat#74l82). For Taqman qPCR analysis, cDNA was synthesized using High-Capacity cDNA Reverse Transcription Kit (ThermoFisher Scientific cat:43688l3 or 4368814) according to manufacturer instructions. qRT-PCR was performed with cDNA using the iQ5 Multicolor rtPCR Detection system from BioRad with 60°C annealing. Samples were amplified using the following Taqman probes from ThermoFisher: Hs00609359_ml (human PAH), Mm005009l8_ml (mouse PAH); Hs0l026983_ml (JAK1); Hs0l078l36_ml (JAK2); 4352341E (ACTB); 4326320E (GUSB); 4326319E (B2M); and 4326317E (GAPDH).
[00277] Analysis of the fold changes in expression as measured by qRT-PCR were performed using the technique below. The control was DMSO, and the treatment was the selected compound (CPD). The internal control was GAPDH or B-Actin (or otherwise indicated), and the gene of interest is the target. First, the averages of the 4 conditions were calculated for normalization: DMSO:GAPDH, DMSO:Target, CPD: GAPDH, and CPD:Target. Next, the ACT of both control and treatment were calculated to normalize to internal control (GAPDH) using (DMSO:Target) - (DMSO:GAPDH) = ACT control and (CPD:Target) - (CPD: GAPDH) = ACT experimental. Then, the AACT was calculated by ACT experimental - ACT control. The Expression Fold Change (RQ) was calculated by 2-( AACT) (2-fold expression change was shown by RNA-Seq results provided herein).
[00278] In some examples, RQ Min and RQ Max values are also reported. RQ Min and RQ Max are the the minimum and maximum relative levels of gene expression in the test samples, respectively. They were calculated using the confidence level set in the analysis settings and the confidence level was set to one standard deviation (SD). These values were calculated using standard deviation as follows: RQ Min= 2 -(AACT-SD); and RQ Max= 2 - (AACT+SD).
L. Chromatin Interaction Analysis by Paired-End Tag Sequencing (ChlA-PET)
[00279] ChlA-PET was performed as previously described in Chepelev et al. (2012) Cell Res. 22, 490-503; Fullwood et al. (2009) Nature 462, 58-64; Goh et al. (2012) J. Vis. Exp., http://dx.doi.org/l0.379l/3770; Li et al. (2012) Cell 148, 84-98; and Dowen et al. (2014) Cell 159, 374-387, which are each hereby incorporated by reference in their entireties. Briefly, embryonic stem (ES) cells (up to lxlO8 cells) were treated with 1% formaldehyde at room temperature for 20 minutes and then neutralized using 0.2M glycine. The crosslinked chromatin was fragmented by sonication to size lengths of 300-700 bp. The anti-SMCl antibody (Bethyl, A300-055A) was used to enrich SMCl-bound chromatin fragments. A portion of ChIP DNA was eluted from antibody-coated beads for concentration quantification and for enrichment analysis using quantitative PCR. For ChlA-PET library construction, ChIP DNA fragments were end-repaired using T4 DNA polymerase (NEB). ChIP DNA fragments were divided into two aliquots and either linker A or linker B were ligated to the fragment ends. The two linkers differ by two nucleotides which were used as a nucleotide barcode (Linker A with CG; I .inker B with AT). After linker ligation, the two samples were combined and prepared for proximity ligation by diluting in a 20 ml volume to minimize ligations between different DNA-protein complexes. The proximity ligation reaction was performed with T4 DNA ligase
(Fermentas) and incubated without rocking at 22°C for 20 hours. During the proximity ligation DNA fragments with the same linker sequence were ligated within the same chromatin complex, which generated the ligation products with homodimeric linker composition. However, chimeric ligations between DNA fragments from different chromatin complexes could also occur, thus producing ligation products with heterodimeric linker composition. These heterodimeric linker products were used to assess the frequency of nonspecific ligations and were then removed.
i. DAY 1
[00280] The cells were crosslinked as described for ChIP. Frozen cell pellets are stored in the -80°C freezer until ready to use. This protocol required at least 3xl08 cells frozen in six l5ml Falcon tubes (50 million cells per tube). Six 100 pl Protein G Dynabeads (for each ChIA-RET sample) are added to six l.5ml Eppendorf tubes on ice. Beads were washed three times with 1.5 ml Block solution, and incubated end over end at 4°C for 10 minutes between each washing step to allow for efficient blocking. Protein G Dynabeads were resuspended in 250 mΐ of Block solution in each of six tubes and 10 mg of SMC1 antibody (Bethyl A300- 055A) was added to each tube. The bead-antibody mixes were incubated at 4°C end-over- end overnight.
ii. DAY 2
[00281] Beads were washed three times with l.5ml Block solution to remove unbound IgG and incubated end-over-end at 4°C for 10 minutes each time. Smcl -bound beads were resuspended in 100 mΐ of Block solution and stored at 4 °C. Final lysis buffer 1 (8 ml per sample) was prepared by adding 50x Protease inhibitor cocktail solution to Lysis buffer 1 (LB1) (1:50). Eight ml of Final lysis buffer 1 was added to each frozen cell pellet (8 ml per sample x 6). The cells were thoroughly resuspended and thawed on ice by pipetting up and down. The cell suspension was incubated again end-over-end for 10 minutes at 4 °C. The suspension was centrifuged at 1,350 g for 5 minutes at 4 °C. Concurrently, Final lysis buffer 2 (8 ml per sample) was prepared by adding 50x Protease inhibitor cocktail solution to lysis buffer 2 (LB2) (1:50)
[00282] After centrifugation, the supernatant was discarded, and the nuclei were thoroughly resuspended in 8 ml Final lysis buffer 2 by pipetting up and down. The cell suspension was incubated end-over-end for 10 minutes at 4°C. The suspension is centrifuged at 1,350 g for 5 minutes at 4°C. During incubation and centrifugation, the Final sonication buffer (l5ml per sample) was prepared by adding 50x Protease inhibitor cocktail solution to sonication buffer (1:50). The supernatant was discarded, and the nuclei were fully resuspended in 15 ml Final sonication buffer by pipetting up and down. The nuclear extract was extracted to fifteen 1 ml Covaris Evolution E220 sonication tubes on ice. An aliquot of 10 mΐ was used to check the size of unsonicated chromatin on a gel. [00283] A Covaris sonicator was programmed according to manufacturer’s instructions (12 minutes per 20 million cells = 12x15= 3 hours). The samples were sequentially sequenced as described above. The goal is to break chromatin DNA to 200-600 bp. If sonication fragments were too big, false positives became more frequent. The sonicated nuclear extract was dispensed into 1.5 ml Eppendorf tubes. l.5ml samples were centrifuged at full speed at 4°C for 10 minutes. Supernatant (SNE) was pooled into a new pre-cooled 50 ml Falcon tube, and brought to a volume of 18 ml with sonication buffer. Two tubes of 50 pl were taken as input and to check the size of fragments. 250 pl of ChIP elution buffer was added and reverse crosslinking occured at 65°C overnight in the oven. After reversal of crosslinking, the size of sonication fragments wa determined on a gel.
[00284] Three ml of sonicated extract was added to 100 mΐ Protein G beads with SMC1 antibodies in each of six clean 15 ml Falcon tubes. The tubes containing SNE-bead mix were incubated end-over-end at 4°C overnight (14 to 18 hours)
iii. DAY 3
[00285] Half the volume (l.5ml) of the SNE-bead mix was added to each of six pre chilled tubes and SNE was removed using a magnet. The tubes were sequentially washed as follows: 1) 1.5 ml of Sonication buffer is added, the beads are resuspended and rotated for 5 minutes at 4°C for binding, then the liquid was removed (step performed twice); 2) 1.5 ml of high-salt sonication buffer is added, and the beads were resuspended and rotated for 5 minutes at 4°C for binding, then the liquid was removed (step performed twice); 3) 1.5 ml of high-salt sonication buffer was added, and the beads were resuspended and rotated for 5 minutes at 4°C for binding, then the liquid was removed (step performed twice); 4) 1.5 ml of LiCl buffer was added, and the cells were resuspended and incubated end-over-end for 5 minutes for binding, then the liquid was removed (step performed twice); 5) 1.5 ml of IX TE + 0.2% Triton X-100 was used to wash the cells for 5 minutes for binding, then the liquid was removed; and 1.5 ml of ice-cold TE Buffer was used to wash the cells for 30 seconds for binding, then the liquid was removed (step performed twice). Beads from all six tubes were sequentially resuspended in beads in one l,000ul tube of IX ice-cold TE buffer.
[00286] ChIP-DNA was quantified using the following protocol. Ten percent of beads (by volume), or 100 mΐ, were transferred into a new 1.5 ml tube, using a magnet. Beads were resuspended in 300 mΐ of ChIP elution buffer and the tube was rotated with beads for 1 hour at 65°C. The tube with beads was placed on a magnet and the eluate was transferred to a fresh DNA LoBind Eppendorf tube. The eluate was incubated overnight at 65°C oven without rotating. Immuno-precipitated samples were transferred to fresh tubes, and 300 pl of TE buffer was added to the immuno-precipitants and Input samples to dilute. Five pl of RNase A (20mg/ml) was added, and the tube was incubated at 37°C for 30 minutes.
[00287] Following incubation, 3 pl of 1 M CaCE and 7 pl of 20 mg/ml Proteinase K was added to the tube and incubated 1.5 hours at 55°C. MaXtract High Density 2 ml gel tubes (Qiagen) were prepared by centrifuging them at full speed for 30 seconds at RT. 600 mΐ of phenol/chloroform/isoamyl alcohol was added to each proteinase K reaction. About 1.2 ml of the mixtures was transferred to the MaXtract tubes. Tubes are spun at 16,000 g for 5 minutes at RT. The aqueous phase was transferred to two clean DNA LoBind tubes (300 mΐ in each tube), and 1 pl glycogen, 30 pl of 3 M sodium acetate, and 900 mΐ ethanol was added. The mixture was allowed to precipitate overnight at -20°C or for 1 hour at -80°C.
[00288] The mixture was spun down at maximum speed for 20 minutes at 4°C, ethanol was removed, and the pellets were washed with 1 ml of 75% ethanol by spinning tubes down at maximum speed for 5 minutes at 4°C. All remnants of ethanol were removed, and pellets were dried for 5 minutes at RT. ¾0 was added to each tube. Each tube was allowed to stand for 5 minutes, and vortexed briefly. DNA from both tubes was combined to obtain 50 mΐ of IP and 100 mΐ of Input DNA.
[00289] The amount of DNA collected was quantitated by ChIP using Qubit (Invitrogen #Q32856). One pl intercalating dye was combined with each measure 1 pl of sample. Two standards that come with the kit were used. DNA from only 10% of the beads was being measured. About 400 ng of chromatin in 900 mΐ of bead suspension was obtained with a good enrichment at enhancers and promoters as measured by qPCR.
iv. DAY 3 or 4
[00290] End-blunting of ChIP-DNA was performed on the beads using the following protocol. The remaining chromatin/beads were split by pipetting, and 450 mΐ of bead suspension was aliquoted into 2 tubes. Beads were collected on a magnet. Supernatant was removed, and then the beads were resuspended in the following reaction mix: 70 mΐ 10X NEB buffer 2.1 (NEB, M0203L), 7 pl 10 mM dNTPs, 615.8 mΐ dH20, and 7.2 pl of 3 U/pl T4 DNA Polymerase (NEB, M0203L). The beads were incubated at 37°C with rotation for 40 minutes. Beads were collected with a magnet, then the beads were washed 3 times with 1 ml ice-cold ChIA-RET Wash Buffer (30 seconds per each wash).
[00291] On-Bead A-tailing was performed by preparing Klenow (3 To 5'exo-) master mix as stated below: 70 mΐ 10X NEB buffer 2, 7 pl 10 mM dATP, 616 mΐ dH20, and 7 pl of 3U/pl Klenow (3 "to 5"exo-) (NEB, M0212L). The mixture was incubated at 37°C with rotation for 50 minutes. Beads were collected with a magnet, then beads were washed 3 times with 1 ml of ice-cold ChIA-RET Wash Buffer (30 seconds per each wash).
[00292] Tinkers were thawed gently on ice. T nkers were mixed weii with water gentiy by pipetting, then with PEG buffer, then gentiy vortexed. Then, T394 pi of master mix and 6 pi of ligase was added per tube and mixed by inversion. Parafilm was put on the tube, and the tube was incubated at 16°C with rotation overnight (at least 16 hours). The biotinylated linker was ligated to ChIP-DNA on beads by setting up the following reaction mix and adding reagents in order: 1110 pi d!TO, 4 pi 200 ng/pl biotinylated bridge linker, 280 pi 5X T4 DNA ligase buffer with PEG (Invitrogen), and 6 pi 30 U/pl T4 DNA ligase (Fermentas).
v. DAY 5
[00293] Exonuclease lambda/Exonuclease I On-Bead digestion was performed using the following protocol. Beads were collected with a magnet and washed 3 times with 1ml of ice- cold ChIA-RET Wash Buffer (30 seconds per each wash). The Wash buffer was removed from beads, then resuspended in the following reaction mix: 70 pi 10X lambda nuclease buffer (NEB, M0262L), 618 pi nuclease-free dH20, 6 pi 5 U/pl Lambda Exonuclease (NEB, M0262L), and 6 pi Exonuclease I (NEB, M0293L). The reaction was incubated at 37°C with rotation for 1 hour. Beads were collected with a magnet, and beads are washed 3 times with lml ice-cold ChIA-RET Wash Buffer (30 seconds per each wash).
[00294] Chromatin complexes were eluted off the beads by removing all residual buffer and resuspending the beads in 300 pi of ChIP elution buffer. The tube with beads was rotated 1 hour at 65°C. The tube was placed on a magnet and the eluate was transferred to a fresh DNA LoBind Eppendorf tube. The eluate was incubated overnight at 65°C in an oven without rotating.
vi. DAY 6
[00295] The eluted sample was transferred to a fresh tube and 300 pi of TE buffer was added to dilute the SDS. Three pi of RNase A (30 mg/ml) was added to the tube, and the mixture was incubated at 37°C for 30 minutes. Following incubation, 3pl of 1M CaCE and 7 pi of 20 mg/ml Proteinase K was added, and the tube was incubated again for 1.5 hours at 55°C.
MaXtract High Density 2 ml gel tubes (Qiagen) are precipitated by centrifuging them at full speed for 30 seconds at RT. Six hundred pi of phenol/chloroform/isoamyl alcohol was added to each proteinase K reaction, and about 1.2 ml of the mixture was transferred to the MaXtract tubes. Tubes were spun at 16,000 g for 5 minutes at RT. [00296] The aqueous phase was transferred to two clean DNA LoBind tubes (300 mΐ in each tube), and 1 mΐ glycogen, 30 mΐ of 3M sodium acetate, and 900 mΐ ethanol was added. The mixture was precipitated for 1 hour at -80°C. The tubes were spun down at maximum speed for 30 minutes at 4°C, and the ethanol was removed. The pellets were washed with 1 ml of 75% ethanol by spinning tubes down at maximum speed for 5 minutes at 4°C. Remnants of ethanol were removed, and the pellets were dried for 5 minutes at RT. Thirty mΐ of fTO was added to the pellet and allowed to stand for 5 minutes. The pellet mixture was vortexed briefly, and spun down to collect the DNA.
[00297] Qubit and DNA High Sensitivity ChIP were performed to quantify and assess the quality of proximity ligated DNA products. About 120 ng of the product was obtained.
vii. DAY 7
[00298] Components for Nextera tagmentation were then prepared. One hundred ng of DNA was divided into four 25 mΐ reactions containing 12.5 mΐ 2X Tagmentation buffer (Nextera),
1 mΐ nuclease-free d¾0, 2.5 mΐ Tn5 enzyme(Nextera), and 9 mΐ DNA (25 ng). Fragments of each of the reactions were analyzed on a Bioanalyzer for quality control.
[00299] The reactions were incubated at 55°C for 5 minutes, then at l0°C for 10 minutes. Twenty-five mΐ of ¾0 was added, and tagmented DNA was purified using Zymo columns. Three hundred fifty mΐ of Binding Buffer was added to the sample, and the mixture was loaded into a column and spun at 13,000 rpm for 30 seconds. The flow through was re-applied and the columns were spun again. The columns were washed twice with 200 mΐ of wash buffer and spun for 1 minute to dry the membrane. The column was transferred to a clean Eppendorf tube and 25 mΐ of Elution buffer was added. The tube was spun down for 1 minute. This step was repeated with another 25 mΐ of elution buffer. All tagmented DNA was combined into one tube.
[00300] ChlA-PETs were immobilized on Streptavidin beads using the following steps. 2X B&W Buffer (40 ml) was prepared as follows for coupling of nucleic acids: 400 mΐ 1M Tris- HC1 pH 8.0 (10 mM final), 80 mΐ 1M EDTA (1 mM final), 16 ml 5 M NaCl (2 M final), and 23.52 ml dH20. IX B&W Buffer (40 ml total) was prepared by adding 20 ml dfTO to 20 ml of the 2X B&W Buffer.
[00301] MyOne Streptavidin Dynabeads M-280 were allowed to come to room temperature for 30 minutes, and 30 mΐ of beads were transferred to a new 1.5 ml tube. Beads were washed with 150 mΐ of 2X B&W Buffer twice. Beads were resuspended in 100 mΐ of iBlock buffer (Applied Biosystems), and mixed. The mixture was incubated at RT for 45 minutes on a rotator.
[00302] I-BLOCK Reagent was prepared to contain: 0.2% I-Block reagent (0.2 g), IX PBS or IX TBS (10 ml 10X PBS or 10X TBS), 0.05% Tween-20 (50 pl), and H20 to 100 ml. 10X PBS and I-BLOCK reagent was added to H20, and the mixture was microwaved for 40 seconds (not allowed to boil), then stirred. Tween-20 was added after the solution was cooled. The solution remained opaque, but particles were dissolved. The solution was cooled to RT for use.
[00303] During incubation of beads, 500 ng of sheared genomic DNA was added to 50 mΐ of H20 and 50 mΐ of 2X B&W Buffer. When the beads finish incubating with the iBLOCK buffer, they were washed twice with 200 mΐ of IX B&W buffer. The wash buffer was discarded, and 100 mΐ of the sheared genomic DNA was added. The mixture was incubated with rotation for 30 minutes at RT. The beads were washed twice with 200 mΐ of IX B&W buffer. Tagmented DNA was added to the beads with an equal volume of 2X B&W buffer and incubated for 45 minutes at RT with rotation. The beads were washed 5 times with 500 mΐ of 2xSSC/0.5% SDS buffer (30 seconds each time) followed by 2 washes with 500 ml of IX B&W Buffer and incubating each after wash for 5 minutes at RT with rotation. The beads were washed once with 100 mΐ elution buffer (EB) from a Qiagen Kit by resuspending beads gently and putting the tube on a magnet. The supernatant was removed from the beads, and they were resuspended in 30 mΐ of EB.
[00304] A paired end sequencing library was constructed on beads using the following protocol. Ten mΐ of beads are tested by PCR with 10 cycles of amplification. The 50 mΐ of the PCR mixture contained: 10 mΐ of bead DNA, 15 mΐ NPM mix (from Illumina Nextera kit), 5 mΐ of PPC PCR primer, 5 mΐ of Index Primer 1 (i7), 5 mΐ of Index Primer 2 (i5), and 10 mΐ of H20. PCR was performed using the following cycle conditions: denaturing the DNA at 72°C for 3 minutes, then 10-12 cycles of 98°C for 10 seconds, 63°C for 30 seconds, and 72°C for 50 seconds, and a final extension of 72°C for 5 minutes. The number of cycles was adjusted to obtain about 300 ng of DNA total with four 25 mΐ reactions. The PCR product was held at 4°C for an indefinite amount of time.
[00305] The PCR product was cleaned-up using AMPure beads. Beads were allowed to come to RT for 30 minutes before using. Fifty mΐ of the PCR reaction was transferred to a new Low- Bind Tube and (l.8x volume) 90 mΐ of AMPure beads was added. The mixture was pipetted well and incubated at RT for 5 minutes. A magnet was used for 3 minutes to collect beads and remove the supernatant. Three hundred mΐ of freshly prepared 80% ethanol was added to the beads on the magnet, and the ethanol was carefully dicarded. The wash was repeated, and then all ethanol was removed. The beads were dried on the magnet rack for 10 minutes. Ten mΐ EB was added to the beads, mixed well, and incubated for 5 minutes at RT. The eluate was collected, and 1 mΐ of eluate was used for Qubit and Bioanalyzer.
[00306] The library was cloned to verify complexity using the following protocol. One mΐ of the library was diluted at 1:10. A PCR reaction was performed as described below. Primers that anneal to Illumina adapters were chosen (Tm=52.2°C). The PCR reaction mixture (total volume: 50 mΐ) contained the following: 10 mΐ of 5X GoTaq buffer, 1 mΐ of 10 mM dNTP, 5 mΐ of 10 mM primer mix, 0.25 mΐ of GoTaq polymerase, 1 mΐ of diluted template DNA, and 32.75 mΐ of H20. PCR was performed using the following cycle conditions: denaturing the DNA at 95°C for 2 minutes and 20 cycles at the following conditions: 95°C for 60 seconds, 50°C for 60 seconds, and 72°C for 30 seconds with a final extension at 72°C for 5 minutes. The PCR product was held at 4°C for an indefinite amount of time.
[00307] The PCR product was ligated with the pGEM® T-Easy vector (Promega) protocol. Five mΐ of 2X T4 Quick ligase buffer, Imΐ of pGEM® T-Easy vector, 1 mΐ of T4 ligase, 1 mΐ of PCR product, and 2 mΐ of H20 were combined to a total volume of 10 mΐ. The product was incubated for 1 hour at RT and 2 mΐ was used to transform Stellar competent cells. Two hundred mΐ of 500 mΐ of cells were plated in SOC media. The next day, 20 colonies were selected for Sanger sequencing using a T7 promoter primer. 60% clones had a full adapter, and 15% had a partial adapter
viii. Reagents
[00308] Protein G Dynabeads for 10 samples were from Invitrogen Dynal, Cat# 10003D. Block solution (50 ml) contained 0.25g BSA dissolved in 50 ml of ddH20 (0.5% BSA, w/v), and was stored at 4°C for 2 days before use.
[00309] Lysis buffer 1 (LB1) (500 ml) contained 25 ml of 1 M Hepes-KOH, pH 7.5; 14 ml of 5 M NaCl; 1 ml of 0.5 M EDTA, pH 8.0; 50 ml of 100% Glycerol solution; 25 ml of 10% NP- 40; and 12.5 ml of 10% Triton X-100. The pH was adjusted to 7.5. The buffer was sterile - filtered, and stored at 4°C. The pH was re-checked immediately prior to use. Lysis buffer 2 (LB2) (1000 ml) contained 10 ml of 1 M Tis-HCL, pH 8.0; 40 ml of 5 M NaCl; 2 ml of 0.5 M EDTA, pH 8.0; and 2 ml of 0.5 M EGTA, pH 8.0. The pH was adjusted to 8.0. The buffer was sterile-filtered, and stored at 4 °C. The pH was re-checked immediately prior to use. [00310] Sonication buffer (500 ml) contained 25 ml of 1 M Hepes-KOH, pH 7.5; 14 ml of 5M NaCl; 1 ml of 0.5 M EDTA, pH 8.0; 50 ml of 10% Triton X-100; 10 ml of 5% Na-deoxycholate; and 5 ml of 10% SDS. The buffer was sterile-filtered, and stored at 4 °C. The pH was re-checked immediately prior to use. High-salt sonication buffer (500ml) contained 25 ml of 1 M Hepes- KOH, pH 7.5; 35 ml of 5M NaCl; 1 ml of 0.5 M EDTA, pH 8.0; 50 ml of 10% Triton X-100; 10 ml of 5% Na-deoxycholate; and 5 ml of 10% SDS. The buffer was sterile-filtered, and stored at 4 °C. The pH was re-checked immediately prior to use.
[00311] LiCl wash buffer (500 ml) contained 10 ml of 1 M Tris-HCL, pH 8.0; 1 ml of 0.5M EDTA, pH 8.0; 125 ml of 1M LiCl solution; 25 ml of 10% NP-40; and 50 ml of 5% Na- deoxycholate. The pH was adjusted to 8.0. The buffer was sterile-filtered, and stored at 4 °C. The pH was re-checked immediately prior to use.
[00312] Elution buffer (500ml) used to quantify the amount of ChIP DNA contained 25 ml of 1M Tris-HCL, pH 8.0; 10 ml of 0.5M EDTA, pH 8.0; 50 ml of 10% SDS; and 415 ml of ddH20. The pH was adjusted to 8.0. The buffer was sterile-filtered, and stored at 4 °C. The pH was re checked immediately prior to use.
[00313] ChIA-RET Wash Buffer (50 ml) contained 500 pl of 1 M Tris-HCl, pH 8.0 (final 10 mM); 100 pl of 0.5 M EDTA, pH 8.0 (final 1 mM); 5 ml of 5 M NaCl (final 500 mM); and 44.4 ml of dhkO.
M. HiChIP
[00314] Alternatively to ChIA-RET, HiChIP was used to analyze chromatin interactions and conformation. HiChIP requires fewer cells than ChlA-PET.
i. Cell crosslinking
[00315] Cells were cross-linked as described in the ChIP protocol above. Crosslinked cells were either stored as pellets at -80°C or used for HiChIP immediately after flash-freezing the cells.
ii. Lysis and restriction
[00316] Fifteen million cross-linked cells were resuspended in 500 pL of ice-cold Hi-C Lysis Buffer and rotated at 4°C for 30 minutes. For cell amounts greater than 15 million, the pellet was split in half for contact generation and then recombined for sonication. Cells were spun down at 2500g for 5 minutes, and the supernatant was discarded. The pelleted nuclei were washed once with 500 pL of ice-cold Hi-C Lysis Buffer. The supernatant was removed, and the pellet was resuspended in 100 pL of 0.5% SDS. The resuspension was incubated at 62°C for 10 minutes, and then 285 pL of H20 and 50 pL of 10% Triton X-100 were added to quench the SDS. The resuspension was mixed well, and incubated at 37°C for 15 minutes. Fifty pL of 10X NEB Buffer 2 and 375 U of Mbol restriction enzyme (NEB, R0147) was added to the mixture to digest chromatin for 2 hours at 37°C with rotation. For lower starting material, less restriction enzyme is used: 15 pL was used for 10-15 million cells, 8 pL for 5 million cells, and 4 pL for 1 million cells. Heat (62°C for 20 minutes) was used to inactivate Mbol.
iii. Biotin Incorporation and Proximity Ligation
[00317] To fill in the restriction fragment overhangs and mark the DNA ends with biotin, 52 pL of fill-in master mix was reacted by combining 37.5 pL of 0.4 mM biotin-dATP (Thermo 19524016); 1.5 pL of 10 mM dCTP, dGTP, and dTTP; and 10 pL of 5 U/pL DNA Polymerase I, Large (Klenow) Fragment (NEB, M0210). The mixture was incubated at 37°C for 1 hour with rotation.
[00318] 948 pL of ligation master mix was added. Ligation Master Mix contains 150 pL of
10X NEB T4 DNA ligase buffer with lOmM ATP (NEB, B0202); 125 pL of 10% Triton X-100; 3 pL of 50 mg/mL BSA; 10 pL of 400 U/pL T4 DNA Ligase (NEB, M0202); and 660 pL of water. The mixture was incubated at room temperature for 4 hours with rotation. The nuclei were pelleted at 2500g for 5 minutes, and the supernatant was removed.
iv. Sonic ation
[00319] For sonication, the pellet was brought up to 1000 pL in Nuclear Lysis Buffer. The sample was transferred to a Covaris millitube, and the DNA was sheared using a Covaris® E220Evolution with the manufacturer recommended parameters. Each tube (15 million cells) was sonicated for 4 minutes under the following conditions: Fill Level 5; Duty Cycle 5%; PIP 140; and Cycles/Burst 200.
v. Preclearing. Tmmunoprecipitation. IP Bead Capture, and Washes
[00320] The sample was clarified for 15 minutes at l6,l00g at 4°C. The sample is split into 2 tubes of about 400 pL each and 750 pL of ChIP Dilution Buffer is added. For the Smcla antibody (Bethyl A300-055A), the sample is diluted 1:2 in ChIP Dilution Buffer to achieve an SDS concentration of 0.33%. 60 pL of Protein G beads were washed for every 10 million cells in ChIP Dilution Buffer. Amounts of beads (for preclearing and capture) and antibodies were adjusted linearly for different amounts of cell starting material. Protein G beads were resuspended in 50 pL of Dilution Buffer per tube (100 pL per HiChIP). The sample was rotated at 4°C for 1 hour. The samples were put on a magnet, and the supernatant was transferred into new tubes. 7.5 pg of antibody was added for every 10 million cells, and the mixture was incubated at 4°C overnight with rotation. Another 60 pL of Protein G beads for every 10 million cells in ChIP Dilution Buffer was added. Protein G beads were resuspended in 50 pL of Dilution Buffer (100 pL per HiChIP), added to the sample, and rotated at 4°C for 2 hours. The beads were washed three times each with Low Salt Wash Buffer, High Salt Wash Buffer, and LiCl Wash Buffer. Washing was performed at room temperature on a magnet by adding 500 pL of a wash buffer, swishing the beads back and forth twice by moving the sample relative to the magnet, and then removing the supernatant
vi. ChTP DNA Elution
[00321] ChIP sample beads were resuspended in 100 pL of fresh DNA Elution Buffer. The sample beads were incubated at RT for 10 minutes with rotation, followed by 3 minutes at 37°C with shaking. ChIP samples were placed on a magnet, and the supernatant was removed to a fresh tube. Another 100 pL of DNA Elution Buffer was added to ChIP samples and incubations were repeated. ChIP sample supernatants were removed again and transferred to a new tube. There was about 200 pL of ChIP sample. Ten pL of Proteinase K (20 mg/ml) was added to each sample and incubated at 55°C for 45 minutes with shaking. The temperature was increased to 67°C, and the samples were incubated for at least 1.5 hours with shaking. The DNA was Zymo- purified (Zymo Research, #D40l4) and eluted into 10 pL of water. Post-ChIP DNA was quantified to estimate the amount of Tn5 needed to generate libraries at the correct size distribution. This assumed that contact libraries were generated properly, samples were not over sonicated, and that material was robustly captured on streptavidin beads. SMC1 HiChIP with 10 million cells had an expected yield of post-ChIP DNA from 15 ng-50 ng. For libraries with greater than 150 ng of post-ChIP DNA, materials were set aside and a maximum of l50ng was taken into the biotin capture step
vii. Biotin Pull-Down and Preparation for Tllumina Sequencing
[00322] To prepare for biotin pull-down, 5 pL of Streptavidin C-l beads were washed with Tween Wash Buffer. The beads were resuspended in 10 pL of 2X Biotin Binding Buffer and added to the samples. The beads were incubated at RT for 15 minutes with rotation. The beads were separated on a magnet, and the supernatant was discarded. The beads were washed twice by adding 500 pL of Tween Wash Buffer and incubated at 55°C for 2 minutes while shaking.
The beads were washed in 100 pL of IX (diluted from 2X) TD Buffer. The beads were resuspended in 25 pL of 2X TD Buffer, 2.5 pL of Tn5 for each 50 ng of post-ChIP DNA, and water to a volume of 50 pL.
[00323] The Tn5 had a maximum amount of 4 pL. For example, for 25 ng of DNA transpose, 1.25 pL of Tn5 was added, while for 125 ng of DNA transpose, 4 pL of Tn5 was used. Using the correct amount of Tn5 resulted in proper size distribution. An over-transposed sample had shorter fragments and exhibited lower alignment rates (when the junction was close to a fragment end). An undertransposed sample has fragments that are too large to cluster properly on an Illumina sequencer. The library was amplified in 5 cycles and had enough complexity to be sequenced deeply and achieve proper size distribution regardless of the level of transposition of the library.
[00324] The beads were incubated at 55°C with interval shaking for 10 minutes. Samples were placed on a magnet, and the supernatant was removed. Fifty mM EDTA was added to samples and incubated at 50°C for 30 minutes. The samples were then quickly placed on a magnet, and the supernatant was removed. The samples were washed twice with 50 mM EDTA at 50°C for 3 minutes, then were removed quickly from the magnet. Samples were washed twice in Tween Wash Buffer for 2 minutes at 55°C, then were removed quickly from the magnet. The samples were washed with 10 mM Tris-HCl, pH8.0.
viii. PCR and Post-PCR Size Selection
[00325] The beads were resuspended in 50 pL of PCR master mix (use Nextera XT DNA library preparation kit from Illumina, #15028212 with dual-index adapters # 15055289). PCR was performed using the following program. The cycle number was estimated using one of two methods: (1) A first run of 5 cycles (72°C for 5 minutes, 98°C for 1 minute, 98°C for 15 seconds, 63°C for 30 seconds, 72°C for 1 minute) was performed on a regular PCR and then the product was removed from the beads. Then, 0.25X SYBR green was added, and the sample was ran on a qPCR. Samples were pulled out at the beginning of exponential amplification; or (2) Reactions were run on a PCR and the cycle number was estimated based on the amount of material from the post-ChIP Qubit (greater than 50ng is run in 5 cycles, while approximately 50 ng was run in 6 cycles, 25ng is ran in 7 cycles, l2.5ng is ran in 8 cycles, etc.).
[00326] Libraries were placed on a magnet and eluted into new tubes. The libraries were purified using a kit form Zymo Research and eluted into 10 pL of water. A two-sided size selection was performed with AMPure XP beads. After PCR, the libraries were placed on a magnet and eluted into new tubes. Then, 25 pL of AMPure XP beads were added, and the supernatant was kept to capture fragments less than 700 bp. The supernatant was transferred to a new tube, and 15 pL of fresh beads were added to capture fragments greater than 300 bp. A final elution was performed from the Ampure XP beads into 10 pL of water. The library quality was verified using a Bioanalyzer.
ix. Buffers [00327] Hi-C Lysis Buffer (10 mL) contains 100 pL of 1 M Tris-HCl pH 8.0; 20 pL of 5 M NaCl; 200 pL of 10% NP-40; 200 pL of 50X protease inhibitors; and 9.68 mL of water. Nuclear Lysis Buffer (10 mL) contains 500 pL of 1 M Tris-HCl pH 7.5; 200 pL of 0.5 M EDTA; 1 mL of 10% SDS; 200 pL of 50X Protease Inhibitor; and 8.3mL of water. ChIP Dilution Buffer (10 mL) contains 10 pL of 10% SDS; 1.1 mL of 10% Triton X-100; 24 pL of 500 mM EDTA; 167 pL of 1 M Tris pH 7.5; 334 pL of 5 M NaCl; and 8.36 5mL of water. Low Salt Wash Buffer (lOmL) contains 100 pL of 10% SDS; 1 mL of 10% Triton X-100; 40 pL of 0.5 M EDTA; 200 pL of 1 M Tris-HCl pH 7.5; 300 pL of 5 M NaCl; and 8.36 mL of water. High Salt Wash Buffer (10 mL) contains 100 pL of 10% SDS; 1 mL of 10% Triton X-100; 40 pL of 0.5 M EDTA; 200 pL of 1 M Tris-HCl pH 7.5; 1 mL of 5 M NaCl; and 7.66 mL of water. LiCl Wash Buffer (10 mL) contains 100 pL of 1 M Tris pH 7.5; 500 pL of 5 M LiCl; 1 mL of 10% NP-40; 1 mL of 10% Na-deoxycholate; 20 pL of 0.5 M EDTA; and 7.38 mL of water.
[00328] DNA Elution Buffer (5 mL) contains 250 pL of fresh 1 M NaHC03; 500 pL of 10% SDS; and 4.25 mL of water. Tween Wash Buffer (50 mL) contains 250 pL of 1 M Tris-HCl pH 7.5; 50 pL of 0.5 M EDTA; 10 mL of 5 M NaCl; 250 pL of 10% Tween-20; and 39.45 mL of water. 2X Biotin Binding Buffer (10 mL) contains 100 pL 1 M Tris-HCl pH 7.5; 20 pL of 0.5 M; 4 mL of 5 M NaCl; and 5.88 mL of water. 2X TD Buffer (lmL) contains 20 pL of 1 M Tris- HCl pH 7.5; 10 pL of 1 M MgCL; 200 pL of 100% Dimethylformamide; and 770 pL of water.
N. Drug dilutions for administration to hepatocvtes
[00329] Prior to compound treatment of hepatocytes, 100 mM stock drugs in DMSO were diluted to 10 mM by mixing 0.1 mM of the stock drug in DMSO with 0.9 ml of DMSO to a final volume of 1.0 ml. Five pl of the diluted drug was added to each well, and 0.5 ml of media was added per well of drug. Each drug was analyzed in triplicate. Dilution to lOOOx was performed by adding 5 pl of drug into 45 pl of media, and the 50 pl being added to 450 pl of media on cells.
[00330] Bioactive compounds were also administered to hepatocytes. To obtain lOOOx stock of the bioactive compounds in 1 ml DMSO, 0.1 ml of IO,OOOC stock was combined with 0.9 ml DMSO.
O. siRNA knockdown
[00331] Primary human hepatocytes were reverse transfected with siRNA with 6 pmol siRNA using RNAiMAX Reagent (ThermoFisher Cat#l3778030) in 24 well format, 1 pl per well. The following morning, the medium was removed and replaced with modified maintenance medium for an additional 24 hours. The entire treatment lasted 48 hours, at which point the medium was removed and replaced with RLT Buffer for RNA extraction (Qiagen RNeasy 96 QIAcube HT Kit Cat#74l7l). Cells were processed for qRT-PCR analysis and then levels of target mRNA were measured.
[00332] siRNAs were obtained from Dharmacon and are a pool of four siRNA duplex all designed to target distinct sites whitin the specific gene of interest (“SMARTpool”). The following siRNAs were used for each target: D-001206-13-05 (non- targeting); M-003145-02- 0005 (JAK1); and M-003146-02-0005 (JAK2).
P. Mice studies
[00333] A group of 6 mice (C57BL/6J strain), 3 male and 3 female, were administered with a candidate compound once daily via oral gavage for four consecutive days. Mice were sacrificed 4 hours post-last dose on the fourth day. Organs including liver, spleen, kidney, adipose, plasma were collected. Mouse liver tissues were pulverized in liquid nitrogen and aliquoted into small microtubes. TRIzol (Invitrogen Cat# 15596026) was added to the tubes to facilitate cell lysis from tissue samples. The TRIzol solution containing the disrupted tissue was then centrifuged and the supernatant phase was collected. Total RNA was extracted from the supernatant using Qiagen RNA Extraction Kit (Qiagen Cat#74l82) and the target mRNA levels were analyzed using qRT-PCR.
Example 2. RNA-seq study for stimulated hepatocytes
[00334] To identify small molecules that modulate PAH expression, primary human hepatocytes were prepared as a monoculture, and at least one small molecule compound was applied to the cells.
[00335] RNA-seq was performed to determine the effects of the compounds on the expression of PAH in hepatocytes. Fold change was calculated by dividing the level of expression in the cell system that had been perturbed by the level of expression in an unperturbed system. Changes in expression having a p-value < 0.05 were considered significant.
[00336] Compounds used to perturb the signaling centers of hepatocytes include at least one compound listed in Table 1. In the table, compounds are listed with their ID, target, pathway, and pharmaceutical action. Most compounds chosen as perturbation signals are known in the art to modulate at least one canonical cellular pathway. Some compounds were selected from compounds that failed in Phase III clinical evaluation due to lack of efficacy.
Table 1. Compounds used in RNA-seq
Figure imgf000107_0001
Figure imgf000108_0001
Figure imgf000109_0001
Figure imgf000110_0001
Figure imgf000111_0001
Figure imgf000112_0001
Figure imgf000113_0001
Figure imgf000114_0001
Figure imgf000115_0001
Figure imgf000116_0001
Example 3. Identification of compounds that modulate expression of PAH
[00337] Analysis of RNA-seq data revealed a number of compounds that caused significant changes in the expression of PAH. Significance was defined as an FPKM > 1, a log2 (fold change) > 0.5, and a q- value of < 0.05 for selected gene target. RNA-seq results for compounds that significantly increase expression of PAH are shown in Table 2.
Table 2. RNA-seq results for PAH
Figure imgf000116_0002
Figure imgf000117_0001
[00338] Interestingly, a number of tyrosine kinase inhibitors were among the identified compounds that upregulate PAH expression (including R788, Dasatinib, Bosutinib, CP-673451, Merestinib, Pacritinib, Cediranib, GZD824, Amuvatinib, and Ceritinib), suggesting that PAH expression may be strongly regulated via one or more tyrosine kinase-mediated signaling pathways. R788 (fostamatinib disodium hexahydrate) is a known Syk inhibitor. Dasatinib inhibits BCR/ABL and the Src kinase family. Bosutinib is a dual Src/Abl inhibitor. CP-673451 is a selective inhibitor of PDGFRa/b. Merestinib selectively inhibits c-MET and several other receptor tyrosine kinases such as MST1R, FLT3, AXL, MERTK, TEK, ROS1, NTRK1/2/3, and DDR1/2. Pacritinib selectively inhibits JAK2 and FLT3. PND- 1186 is a reversible and selective FAK inhibitor. Cediranib is a potent inhibitor of vascular endothelial growth factor (VEGF) receptor tyrosine kinases. GZD824 is known to inhibit a broad spectrum of Bcr/Abl tyrosine kinase mutants. Amuvatinib is a multi-targeted tyrosine kinase inhibitor with potent activity against mutant c-Met, c-Kit, PDGFRa, Flt3, and c-Ret. Ceritinib is a potent inhibitor against ALK. These tyrosine kinases are associated with the JAK/STAT pathway and MAPK pathway. Pathways associated with these tyrosine kinases may be manipulated to increase PAH expression.
[00339] The results also suggest that expression of PAH expression may be associated with other signaling pathways, such as the TGF-beta/SMAD pathway, Hypoxia activated pathway, Mineralocorticoid receptor signaling pathway, WNT pathway, adrenergic receptor pathway, NF- kB pathway, neuronal signaling pathway, AMPK pathway, Calcium channel pathway, PXR pathway, IGF-lR/InsR signaling pathway, P53 signaling, GPCR/G protein signaling pathway, and/or Notch signaling pathway. Modulating one of these pathways also leads to the up- regulation of PAH. Example 4. Determining genomic position and composition of signaling centers
[00340] A multilayered approach was used herein to identify locations or the“footprint” of signaling centers. The linear proximity of genes and enhancers is not always instructive to determine the 3D conformation of the signaling centers.
[00341] ChIP-seq was used to determine the genomic position and composition of signaling centers. Antibodies specific to 67 targets, including transcription factors, signaling proteins, and chromatin modifications or chromatin-associated proteins, were used in ChIP-seq studies. These antibody targets are shown in Table 3. In the signaling proteins column, the associated canonical pathway is included after the
Figure imgf000118_0001
Table 3. ChIP-seq targets for primary human hepatocytes
Figure imgf000118_0002
Figure imgf000119_0001
[00342] In primary human hepatocytes, the insulated neighborhood that contains the PAH gene was identified to be on chromosome 12 with a size of approximately 668 kb. 14 signaling centers were found within the insulated neighborhood. The chromatin marks or chromatin- associated proteins, transcription factors, and signaling proteins/or factors that were found in the insulated neighborhood are presented in Table 4.
Table 4. ChIP-seq results for PAH
Figure imgf000119_0002
[00343] The ChIP-seq profile indicates that the insulated neighborhood containing PAH may be regulated by JAK/STAT signaling, TGF signaling, WNT signaling, nuclear receptor signaling, BMP signaling, NF-kB signaling, MAPK signaling, and/or Hippo signaling pathways. STAT1 and STAT3, both associated with the JAK/STAT pathway, were observed to bind to the signaling centers within the insulated neighborhood. Moreover, the insulated neighborhood is also enriched with NF-kB and AP- 1 transcription factors, which are associated with tyrosine kinase/MAPK pathway. Targeting these pathways can upregulate PAH expression.
Example 5. Determining genome architecture in hepatocytes
[00344] Hl-ChIP was performed as described in Example 1 to decipher genome architecture. In some cases, ChIA-RET for SMC1 structural protein was used for the same purpose. These techniques identify portions of the chromatin that interact to form 3D structures, such as insulated neighborhood and gene loops.
[00345] The insulated neighborhood containing the PAH gene was identified to be on chromosome 12 at position 102,882,556 to 103,550,727 (human CRCH38/hg38 genome assembly) with a size of approximately 668 kb. The insulated neighborhood contains PAH and 2 other genes, which are positioned in the following order with respect to PAH: (PAH), ASCL1 and C120RF42.
Example 6. Validating compounds and pathways in human hepatocvtes
[00346] qRT-PCR was performed on samples of primary human hepatocytes from two donors to validate the identified compounds for disease associated targets such as PAH. Cryopreserved hepatocytes were thawed and cultured in plating media for 16 hours, and transferred to complete maintenance media for 2 hours. Cells were then either maintained in the complete maintenance media or transferred to modified maintenance media for another 2 hours, and a compound was added. Compounds were added at concentrations ranging from 0.01 mM to 50 mM. Cells were maintained in respective media for 16 hours prior to qRT-PCR analysis. qRT-PCR was performed as described in Example 1. Gene expression level was determined as the relative mRNA level of the gene to an internal control, GAPDH.
[00347] Three compounds that were observed to increase PAH expression in RNA-seq study were selected for validation. Echinomycin is an inhibitor of hypoxia- inducible factor- 1 DNA- binding activity. Dasatinib and CP-673451 are both tyrosine kinase inhibitors. Relative PAH mRNA expression in human hepatocytes treated with Echinomycin, Dasatinib, and CP-673451 are presented in Table 5. The data demonstrated that the compounds caused a dose-dependent increase in PAH levels. The effect of the compounds on PAH expression was reproducible in multiple human donor and culture conditions. This experiment confirms that Dasatinib, CP- 67345! and Echinomycin are capable of increasing expression of PAH in primary human hepatocytes.
Table 5. Echinomycin, Dasatinib and CP-673451 in human hepatocytes
Figure imgf000120_0001
[00348] As both RNA-seq and ChIP analysis suggest the role of JAK/STAT pathway in PAH regulation, a number of JAK pathway inhibitors were tested in primary human hepatocytes. Effects of selected JAK inhibitors (10 mM) on PAH expression are presented in Tables 6 and 7. In Table 7, PAH mRNA levels were normalized to the geometric mean of two internal controls, GAPDH and B2M. The data showed substantial upregulation of PAH by the predicted JAK pathway inhibitors. This was observed across multiple donors and culture conditions.
Table 6. JAK pathway inhitors in human hepatocytes
Figure imgf000121_0001
Table 7. Additional JAK pathway inhitors in human hepatocytes
Figure imgf000121_0002
[00349] Momelotinib was observed to increase PAH expression in a dose-dependent manner (see FIG. 6).
Example 7. Interrogating pathways of interest via siRNA
[00350] The aim of this example was to confirm the role of the identified signaling pathways (e.g., JAK/STAT) that are controlling PAH expression. The end component of the JAK/STAT pathway was targeted via siRNA-mediated knock-down. Primary human hepatocytes were reverse transfected with 10 nM siRNA targeting JAK1, JAK2 or both. Levels of the target mRNA were measured via qRT-PCR and normalized to a non-targeting siRNA control to evaluate the known-down efficiency. PAH mRNA levels were then assayed via qRT-PCR to assess the effect of JAK1 or JAK2 knock-down on PAH expression.
[00351] Results of JAK1/JAK2 knock-down and the corresponding effect on PAH mRNA are shown in FIG. 7. Plotted values are the relative fold changes of target mRNA level with repect to the non-targeting control. As shown in FIG. 7, JAK1 and JAK2 were specifically knocked-down by siRNA at an efficiency of ~ 95% and 75%, respectively. While knocking down of only JAK1 or JAK2 led to a slight increase in PAH expression, JAK1 + JAK2 knock-down resulted in a notable (-40%) increase in PAH mRNA, confirming the role of JAK/STAT pathway in PAH expression regulation.
Example 8. Compound testing in mouse hepatocvtes
[00352] Selected compounds were tested in mouse hepatocytes to evaluate their ability to upregulate PAH. qRT-PCR was performed on samples of mouse hepatocytes treated with the candidate compounds. Compounds were tested at concentrations ranging from 0.01 mM to 50 mM. Fold change in PAH expression observed via qRT-PCR was analyzed as described in Example 1. Compounds that caused robust increase in PAH expression are shown in Table 8. In Table 8,“N.D.” indicates data not determined.
Table 8. Compound treatment in mouse hepatocytes
Figure imgf000122_0001
[00353] As shown above, Dasatinib, Momelotinib and 17-AAG showed consistent upregulation of PAH in mouse hepatocytes. Additional compounds were identified that upregulated PAH, including ATRA, WYE-132, Ibrutinib, and WAY600. ATRA (all-trans retinoic acid) is an active metabolite of vitamin A under the family retinoid, which exert potent effects on cell growth, differentiation and apoptosis through their cognate nuclear receptors. WYE- 125132 (WYE- 132) and WAY600 are highly potent mTOR inhibitors. Ibrutinib is a Syk pathway inhibitor that inhibits Tec family kinases (e.g., BTK). These compounds were chosed for further characterization in the in vivo experiments. Example 9. In vivo compound testing in mice
[00354] Compounds that showed effective upregulation in ex vivo validation studies were chosen for in vivo testing in mice. Candidate compounds were administered at an appropriate dose once daily to a group of wild-type mice consisting of 3 male and 3 female mice. Table 9 lists the doses used for each compound in the study. Mice were sacrificed on the fourth day and liver tissue was collected and analyzed for PAH expression by qRT-PCR.
Table 9. Compound dose
Figure imgf000123_0001
[00355] Relative PAH mRNA levels in each treatment group are presented in FIGs. 8A and 8B. As predicted, all tested compounds demonstrated modest upregulation of PAH in mice liver. The fold change induced by the two tyrosine kinase inhibitors, Bosutinib and Dasatinib, are statistically significant with p-values of 0.0009 and 6.6e-007.
Example 10. Compound testing in other cell lines
[00356] In one embodiment, candidate compounds are evaluated in kidney cells to confirm their efficacy. Changes in target gene expression in kidney cells are analyzed with qRT-PCR. Results are compared with that from the primary hepatocytes. Compounds that show consistent induction of PAH expression are selected for further analysis.
Example 11. Compound testing in patient cells
[00357] Candidate compounds are evaluated in patient derived induced pluripotent stem (iPS)-hepatoblast cells to confirm their efficacy. Selected patients have a deficiency in PAH. Changes in target gene expression in iPS-hepatoblast cells are analyzed with qRT-PCR. Results are used to confirm if the pathway is similarly functional in patient cells and if the compound has the same impact.
Example 12. Compound testing in a mouse model
[00358] Candidate compounds are evaluated in a mouse model of phenylketonuria for in vivo activity and safety. Example 13. Phenylalanine hydroxylase (PAH) enzyme assay
[00359] A biochemical reaction assay in a cell free system was developed to monitor the rate and amount of tyrosine product made by PAH as a function of enzyme concentration. PAH enzyme was collected from hepatocyte lysate and mixed with a phenylalanine substrate, tetrahydrobiopterin (BH4) coenzyme, catalase, and a ferrous ammonium sulfate non-heme Fe(II) cofactor. The reaction is shown in FIG. 10A, 10B, and 10C; and a schematic of the assay is shown in FIG. 11. The samples were processed via Thin Layer Chromatography to resolve the substrate phenylalanine from the tyrosine product. The chromatography assay used Silica Gel 60 plates. The mobile phase was Isopropanol: Acetone: Ammonium Hydroxide: in a 25:25:13 ratio; and detection was via Ninhydrin Spray. This allowed separation of the phenylalanine from the tyrosine. The tyrosine product was detected via fluorometric assays using a 274 nm wavelength light source.
[00360] As shown in FIG. 12, the reaction rates and the amount of tyrosine generated increase linearly with the amount of lysate (amount of PAH enzyme) used in the reaction.
Example 14. Phenylalanine hydroxylase (PAH) enzyme assay from in vivo samples
[00361] The PAH enzyme levels in the mouse liver samples from Example 9 were next assessed using the PAH enzyme assay. As shown in FIG. 13, the treatment induced increase in the liver PAH mRNA upregulation correlates with an increase in the PAH enzyme activity.
[00362] The PAH activity for each compound is shown in FIG. 14 and the mRNA levels are quantified in Table 10 below.
Table 10
Figure imgf000124_0001
Example 15. PAH and GCH1 in vitro and in vivo compound testing [00363] Next, additional compounds were tested in vitro in two different primary human hepatocyte cell lines and one primary mouse hepatocyte cell line for PAH and GCH1 mRNA expression upregulation. Table 11 lists the compounds used in the study.
Table 11
Figure imgf000125_0001
[00364] Table 12 shows the PAH and GCH1 mRNA results for the HH4178 human primary hepatocytes
Table 12
Figure imgf000125_0002
Figure imgf000126_0001
Table 13
Figure imgf000126_0002
[00366] Table 14 shows the PAH and GCH1 mRNA results for the mouse primary hepatocytes.
Table 14
Figure imgf000126_0003
Figure imgf000127_0001
[00367] Compounds were also assessed in vivo as previously described in Example 9. Candidate compounds were administered at an appropriate dose once daily to a group of wild- type mice consisting of 6 male mice. Mice were sacrificed 12 hours post dose on the fourth day and liver tissue was collected and analyzed for PAH enzyme activity and mRNA levels.
[00368] Table 15 shows the PAH and GCH1 mRNA results for the in vivo samples.
Table 15
Figure imgf000127_0002
0.080874
Figure imgf000128_0001
Example 16: Perturbogen upregulation of PAH and GCH1 in primary human hepatocvtes
[00369] Additional compounds were tested in vitro in three different primary human hepatocyte cell lines and one primary mouse hepatocyte cell line for PAH and GCH1 mRNA expression upregulation. Table 16 lists the compounds used in the study. Figure 15 is a diagram showing the average fold increase in PAH and GCHI mRNA upon perturbation of the indicated gene regulatory pathways.
Table 16
HH105 Human Primary Hepatocytes
Figure imgf000128_0002
values represent fold change relative to vehicle
HH4182 Human Primary Hepatocytes
Figure imgf000129_0001
values represent fold change relative to vehicle
HH4178 Human Primary Hepatocytes
Figure imgf000129_0002
Figure imgf000130_0001
values represent fold change relative to vehicle
Mouse Primary Hepatocytes
Figure imgf000130_0002
values represent fold change relative to vehicle
Example 17 : Compounds tested in mice in vivo for upregulation of PAH and GCH1 expression.
Additional compounds were tested in mice for upregulation of PAH and GCH1 expression. Six mice were were tested per group. Select compounds (Table 17) were administered by gavage daily for 4 consequtive days. Animals were sacraficed 6 h after admini station of final dose. Table 17 shows compund names, dosing and signaling pathway that is the target of each compound tested. FIG. 16A shows the fold change in PAH mRNA in treated mice relative to mice administered vehicle alone for select compounds. FIG. 16B shows the fold change in GCH1 mRNA in treated mice relative to mice administered vehicle alone for select compounds. FIG 17 shows the relative increase in PAH protein in livers of treated mice. Table 18 summarizes the results and shows fold change in mRNA for PAH and GCH1 and PAH protein. Mouse liver lysates were prepared for western blotting using a lysis buffer comprising M- PER/HALT. Lysis buffer was prepared by mixing 10 ml of M-PER and 100 uL HALT
Protease/phosphatase cocktail HALT cocktail. 100 uL M-PER/HALT mix was added to each tissue powder sample. The samples were mixed well and then placed at 4°C for 30 min. Tubes were spun at 4°C for 10 min at 5 K rpm. Supernatant was removed ( ~l00uL) and transfered to ice. Protein was quantified, mixed appropriate amount with Lamelli’s buffer, heated at 95°C for 10 min, and loaded on a polyacrylamide gel.
Table 17
Figure imgf000131_0001
Figure imgf000131_0002
Figure imgf000132_0001
Example 18: PAH activity in vivo
[00370] Mice were treated with Dasatinib by oral gavage for 4 days and mice were sacraficed on the fouth day of treatment (FIG. 18 A). Liver lysates were prepared for in vitro analysis of PAH activity as described above. The relative increase in tyrosine in the livers from mice treated with Dasatinib versus livers from mice treated with vehicle alone is shown in FIG. 18B. Three additional compounds, XL228, SIS3 and PF-562271 were tested in mice following the dosing schedule as shown in FIG. 19. PAH activity (FIG. 20) and PAH specific activity (FIG. 21) was increased in mice treated with PF-562271 and XL228. Example 19: Increased amount of PAH protein and GCH1 protein increases activity in vitro for select PAH mutants.
[00371] Mutants of PAH were cloned into plasmids in frame with a Renilla luciferase reporter gene and transfected into 293XT cells that do not express endogenous PAH. The human and mouse PAH constructs are l356bp and l359bp in length, respectively and are based on the Consensus Coding Sequences (CCDS) attained from NCBI. All constructs include a minimal Kozak sequence for optimal expression (GCCGCCACC) located directly upstream of the start codon (ATG) of PAH. Additionally, all PAH sequences include a C-terminal FLAG tag
(GACTACAAAGACGATGACGACAAG) for quantification and detection of the over expression constructs by western blot or immunofluorescence. Constructs were synthesized and cloned into a modified version of the pCDNA3.l using restriction enzymes Agel and Mlul. Briefly, pCDNA3.l was modified by incorporating a viral self-cleaving peptide P2A cleavage sequence(GCAACAAACTTCTCTCTGCTGAAACAAGCCGGAGATGTCGAAGAGAATCC TGGACCG), which can induce the cleaving of the recombinant protein in cells followed by Renilla Luciferase for quantifying the transfection efficiency of the constructs. P2A and Renilla Luciferase are located downstream of the Mlul restriction site in the pCDN3.l vector. All PAH constructs were cloned upstream of the P2A-Renilla Luciferase. The bicistronic expression of both PAH and Renilla luciferase was achieved using a minimal CMV promoter in the pCDNA3.l vector.
[00372] Transfection in 293XT cells was performed as follows. All PAH constructs were transfected into the Lenti-X 293T cell line (293XT) from Takara (Clontech). The transfection reagent used was Fugene6 (Promega) as recommended by the manufacturer. Briefly, various Fugene:DNA ratios were tested in 293XT cells and the optimal plasmid to DNA ratio was determined to be 4:1. 293XT ells were plated for l6h prior to transfection. Transfection was allowed to proceed for a period of 48h. Renilla Luciferase was assessed at 48h and cells were collected, washed in PBS and stored at -80C prior to PAH enzyme activity assays or western blot analysis.
[00373] Cells were lysed and PAH activity was measured in vitro as described above.
Increasing amounts of cell lysate was added to the reaction mixture. Table 19 shows the PAH activity as percent WT activity measured for different PAH constructs harboring different mutations of PAH. FIG. 22 shows the relative protein expression of PAH in lystaes transfected with the different constructs harboroing PAH mutations. FIG. 23 shows increased amonts of cell lysate from cells transfected with a construct harboring PAH 198R243Q lead to increases in PAH activity.
[00374] GCH1 is also cloned into plasmids and transfected into cells. PAH activity in vitro is increased in cells overexpressing GCH1 or in cells where GCH1 expression is increased by treatment of cells with select compounds as described above.
Table 19
Figure imgf000134_0001
Figure imgf000135_0001
Jill · ¾
- Tested in-house using the cell-fee biochemical assay; - splice-site valiant; null in vivo. Should have no effect in the in vitro over-expression system.
Equivalents and Scope
[00375] Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments in accordance with the disclosure described herein. The scope of the present disclosure is not intended to be limited to the above Description, but rather is as set forth in the appended claims.In the claims, articles such as“a,”“an,” and“the” may mean one or more than one unless indicated to the contrary or otherwise evident from the context. Claims or descriptions that include“or” between one or more members of a group are considered satisfied if one, more than one, or all of the group members are present in, employed in, or otherwise relevant to a given product or process unless indicated to the contrary or otherwise evident from the context. The disclosure includes embodiments in which exactly one member of the group is present in, employed in, or otherwise relevant to a given product or process. The disclosure includes embodiments in which more than one, or the entire group members are present in, employed in, or otherwise relevant to a given product or process. It is also noted that the term“comprising” is intended to be open and permits but does not require the inclusion of additional elements or steps. When the term“comprising” is used herein, the term“consisting of’ is thus also encompassed and disclosed. Where ranges are given, endpoints are included. Furthermore, it is to be understood that unless otherwise indicated or otherwise evident from the context and understanding of one of ordinary skill in the art, values that are expressed as ranges can assume any specific value or subrange within the stated ranges in different embodiments of the disclosure, to the tenth of the unit of the lower limit of the range, unless the context clearly dictates otherwise.In addition, it is to be understood that any particular embodiment of the present disclosure that falls within the prior art may be explicitly excluded from any one or more of the claims. Since such embodiments are deemed to be known to one of ordinary skill in the art, they may be excluded even if the exclusion is not set forth explicitly herein. Any particular embodiment of the compositions of the disclosure (e.g., any antibiotic, therapeutic or active ingredient; any method of production; any method of use; etc.) can be excluded from any one or more claims, for any reason, whether or not related to the existence of prior art. It is to be understood that the words which have been used are words of description rather than limitation, and that changes may be made within the purview of the appended claims without departing from the true scope and spirit of the disclosure in its broader aspects.
While the present invention has been described at some length and with some particularity with respect to the several described embodiments, it is not intended that it should be limited to any such particulars or embodiments or any particular embodiment, but it is to be construed with references to the appended claims so as to provide the broadest possible interpretation of such claims in view of the prior art and, therefore, to effectively encompass the intended scope of the invention.

Claims

1. A method of treating a subject with phenylalanine hydroxylase deficiency, comprising administering to the subject an effective amount of a compound capable of modulating the expression of the PAH gene.
2. The method of claim 1, wherein the compound is at least one selected from the group consisting of: a small molecule, a polypeptide, an antibody, a hybridizing
oligonucleotide, and a genome editing agent.
3. The method of claim 2, wherein the compound comprises an inhibitor of the JAK/STAT pathway.
4. The method of claim 3, wherein the compound comprises at least one selected from the group consisting of: Ruxolitinib, Oclacitinib, Baricitinib, Filgotinib, Gandotinib, Lestaurtinib, PF-04965842, Upadacitinib, Cucurbitacin I, CHZ868, Fedratinib, AC430, AT9283, ati-5000l and ati-50002, AZ 960, AZD1480, BMS-911543, CEP-33779, Cerdulatinib (PRT062070, PRT2070), Curcumol, Decernotinib (VX-509), Fedratinib (SAR302503, TG101348), FLLL32, FM-381, GLPG0634 analogue, Go6976, JANEX-l (WHI-P131), Momelotinib (CYT387), NVP-BSK805, Pacritinib (SB1518), Peficitinib (ASP015K, JNJ-54781532), PF-06651600, PF-06700841, R256 (AZD0449), Solcitinib (GSK2586184 or GLPG0778), S-Ruxolitinib (INCB018424), TG101209, Tofacitinib (CP-690550), WHI-P154, WP1066, XL019, ZM 39923 HC1, or a derivative or an analog thereof.
5. The method of claim 2, wherein the compound comprises an inhibitor of the Tyrosine kinase/MAPK pathway.
6. The method of claim 5, wherein the compound comprises at least one selected from the group consisting of: Amuvatinib, Bosutinib, Cediranib, Ceritinib, CP-673451, Dasatinib, GZD824 Dimesylate, Merestinib, R788 (fostamatinib disodium hexahydrate), or a derivative or an analog thereof.
7. The method of claim 2, wherein the compound comprises at least one selected from the group consisting of: 17-AAG (Tanespimycin), Amlodipine Besylate, ATRA (all-trans retinoic acid), Chloroquine phosphate, Deoxycorticosterone, Darapladib, Echinomycin, Enzastaurin, Epinephrine, EVP-6124 (hydrochloride) (encenicline), EW-7197,
FRAX597, Ibrutinib, Perphenazine, Phenformin, PND- 1186, Rifampicin, Semagacestat, Thalidomide, WAY600, WYE-125132 (WYE-132), Zibotentan, Activin, Anti mullerian hormone, GDF10 (BMP3b), IGF-l, Nodal, PDGF, TNF-a, Wnt3a, or a derivative or an analog thereof.
8. The method of claim 2, wherein the compound comprises one or more small interfering RNA (siRNA) targeting one or more genes selected from the group consisting of JAK1 , JAK2, PDGFRA, PDGFRB, SRC and ABL.
9. The method of any one of claims 1-8, wherein the compound increases the expression of the PAH gene in the subject.
10. The method of claim 9, wherein the expression of the PAH gene is increased by at least about 40% over baseline or over levels measured following administration of a control.
11. The method of claim 9, wherein the expression of the PAH gene is increased in the liver of the subject.
12. The method of any one of claims 1-11, wherein the subject has at least one mutated allele of the PAH gene.
13. The method of any one of claims 12, wherein mutation occurs within or near the PAH gene.
14. The method of claim 13, wherein the mutation decreases the activity of phenylalanine hydroxylases or reduces the expression of PAH in the subject as compared to activity or expression associated with a canonical wild-type sequence.
15. The method of any one of claims 1-14, wherein the phenylalanine hydroxylase deficiency is mild hyperphenylalaninemia.
16. The method of any one of claims 1-14, wherein the phenylalanine hydroxylase deficiency is mild phenylketonuria (PKU).
17. The method of any one of claims 1-14, wherein the phenylalanine hydroxylase deficiency is classic phenylketonuria (PKU).
18. A method of modulating the expression of a PAH gene in a cell, comprising introducing to the cell an effective amount of a compound capable of altering one or more signaling molecules associated with the regulatory sequence regions (RSRs) or portion thereof of the PAH gene.
19. The method of claim 18, wherein the compound is selected from the group consisting of: a small molecule, an antibody, a hybridizing oligonucleotide, and a genome editing agent.
20. The method of claim 19, wherein the compound comprises an inhibitor of the JAK/STAT pathway.
21. The method of claim 20, wherein the compound comprises at least one selected from the group consisting of: Ruxolitinib, Oclacitinib, Baricitinib, Filgotinib, Gandotinib, Lestaurtinib, PF-04965842, Upadacitinib, Cucurbitacin I, CHZ868, Fedratinib, AC430, AT9283, ati-5000l and ati-50002, AZ 960, AZD1480, BMS-911543, CEP-33779, Cerdulatinib (PRT062070, PRT2070), Curcumol, Decernotinib (VX-509), Fedratinib (SAR302503, TG101348), FLLL32, FM-381, GLPG0634 analogue, Go6976, JANEX-l (WHI-P131), Momelotinib (CYT387), NVP-BSK805, Pacritinib (SB1518), Peficitinib (ASP015K, JNJ-54781532), PF-06651600, PF-06700841, R256 (AZD0449), Solcitinib (GSK2586184 or GLPG0778), S-Ruxolitinib (INCB018424), TG101209, Tofacitinib (CP-690550), WHI-P154, WP1066, XL019, ZM 39923 HC1, or a derivative or an analog thereof.
22. The method of claim 19, wherein the compound comprises an inhibitor of the Tyrosine kinase/MAPK pathway.
23. The method of claim 22, wherein the compound comprises at least one selected from the group consisting of: Amuvatinib, Bosutinib, Cediranib, Ceritinib, CP-673451, Dasatinib, GZD824 Dimesylate, Merestinib, R788 (fostamatinib disodium hexahydrate), or a derivative or an analog thereof.
24. The method of claim 19, wherein the compound comprises at least one selected from the group consisting of: 17-AAG (Tanespimycin), Amlodipine Besylate, ATRA (all-trans retinoic acid), Chloroquine phosphate, Deoxycorticosterone, Darapladib, Echinomycin, Enzastaurin, Epinephrine, EVP-6124 (hydrochloride) (encenicline), EW-7197,
FRAX597, Ibrutinib, Perphenazine, Phenformin, PND- 1186, Rifampicin, Semagacestat, Thalidomide, WAY600, WYE-125132 (WYE-132), Zibotentan, Activin, Anti mullerian hormone, GDF10 (BMP3b), IGF-l, Nodal, PDGF, TNF-a, Wnt3a, or a derivative or an analog thereof.
25. The method of claim 19, wherein the compound comprises one or more small interfering RNA (siRNA) targeting one or more genes selected from the group consisting of JAK1 , JAK2, PDGFRA, PDGFRB, SRC and ABL.
26. The method of any one of claims 18-25, wherein the compound increases the expression of the PAH gene.
27. The method of claim 26, wherein the expression of the PAH gene is increased by at least about 40% over baseline or over levels measured following administration of a control.
28. The method of any one of claims 18-27, wherein the cell has at least one mutation within or near the PAH gene.
29. The method of claim 28, wherein the at least one mutation decreases the activity of phenylalanine hydroxylases or reduces the expression of PAH in the cell as compared to activity or expression associated with a canonical wild-type sequence.
30. The method of any one of claims 18-29, wherein the cell is a mammalian cell.
31. The method of claim 30, wherein the cell is a human cell.
32. The method of claim 30, wherein the cell is a mouse cell.
33. The method of any one of claims 18-32, wherein the cell is a hepatocyte.
34. A method of modulating the expression of a PAH gene in a cell, comprising introducing to the cell one or more compounds that alter one or more of the upstream or downstream neighborhood genes or its RSRs of the insulated neighborhood comprising the PAH gene.
35. The method of claim 34, wherein the insulated neighborhood comprises the region on chromosome 12 at position 102,882,556 to 103,550,727 (human CRCH38/hg38 genome assembly).
36. The method of claim 34, wherein the one or more downstream neighborhood gene comprises at least one selected from the group consisting of ASCL1 and C120RF42.
37. The method of claim 34, wherein the cell is a hepatocyte.
PCT/US2019/026399 2018-04-06 2019-04-08 Compositions and methods for treating phenylketonuria WO2019195854A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201862653748P 2018-04-06 2018-04-06
US62/653,748 2018-04-06
US201962789477P 2019-01-07 2019-01-07
US62/789,477 2019-01-07

Publications (1)

Publication Number Publication Date
WO2019195854A1 true WO2019195854A1 (en) 2019-10-10

Family

ID=68101255

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2019/026399 WO2019195854A1 (en) 2018-04-06 2019-04-08 Compositions and methods for treating phenylketonuria

Country Status (1)

Country Link
WO (1) WO2019195854A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021114020A1 (en) * 2019-12-09 2021-06-17 苏州亚盛药业有限公司 Pharmaceutical composition of alkynyl-containing compound, preparation method therefor and application thereof
CN113077841A (en) * 2021-03-01 2021-07-06 华中科技大学 Method for predicting functional gene for regulating and controlling autophagy of yeast

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016207647A1 (en) * 2015-06-24 2016-12-29 Oxford Biodynamics Limited Epigenetic chromosome interactions
WO2017031370A1 (en) * 2015-08-18 2017-02-23 The Broad Institute, Inc. Methods and compositions for altering function and structure of chromatin loops and/or domains
WO2017158358A1 (en) * 2016-03-15 2017-09-21 Almac Diagnostics Limited Gene signatures for cancer detection and treatment

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016207647A1 (en) * 2015-06-24 2016-12-29 Oxford Biodynamics Limited Epigenetic chromosome interactions
WO2017031370A1 (en) * 2015-08-18 2017-02-23 The Broad Institute, Inc. Methods and compositions for altering function and structure of chromatin loops and/or domains
WO2017158358A1 (en) * 2016-03-15 2017-09-21 Almac Diagnostics Limited Gene signatures for cancer detection and treatment

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
DATABASE Genome [online] 21 December 2017 (2017-12-21), "NC_000012.12: Homo sapiens chromosome 12, GRCh38.p13 Primary Assembly", retrieved from NCBI Database accession no. GCA_000001405.27 *
SCHERER ET AL.: "The finished DNA sequence of human chromosome 12", NATURE, vol. 440, no. 7082, 16 March 2006 (2006-03-16), pages 346 - 351, XP055642616 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021114020A1 (en) * 2019-12-09 2021-06-17 苏州亚盛药业有限公司 Pharmaceutical composition of alkynyl-containing compound, preparation method therefor and application thereof
CN113077841A (en) * 2021-03-01 2021-07-06 华中科技大学 Method for predicting functional gene for regulating and controlling autophagy of yeast
CN113077841B (en) * 2021-03-01 2022-05-24 华中科技大学 Method for predicting functional gene for regulating and controlling autophagy of yeast

Similar Documents

Publication Publication Date Title
US20200208128A1 (en) Methods of treating liver diseases
US11624065B2 (en) Methods and compositions for modulating gene expression
US20210161997A1 (en) Treating diseases via targeted modulation of gene signaling networks
Denk et al. HDAC inhibitors attenuate the development of hypersensitivity in models of neuropathic pain
US20220107328A1 (en) Methods of treating liver diseases
Lim et al. Enhancing inhibitory synaptic function reverses spatial memory deficits in Shank2 mutant mice
Machado et al. Indoxyl sulfate upregulates liver P-glycoprotein expression and activity through aryl hydrocarbon receptor signaling
US20100267626A1 (en) Methods and compositions for measuring wnt activation and for treating wnt-related cancers
Sachewsky et al. Cyclosporin A enhances neural precursor cell survival in mice through a calcineurin-independent pathway
CA3078247A1 (en) Methods and compositions for treating urea cycle disorders, in particular otc deficiency
WO2019195854A1 (en) Compositions and methods for treating phenylketonuria
JP2023181162A (en) Compositions and methods for inhibiting acss2
Wei et al. Microinjection of histone deacetylase inhibitor into the ventrolateral orbital cortex potentiates morphine induced behavioral sensitization
TW201420105A (en) Mucosal healing promoter
Focchi et al. ATM rules neurodevelopment and glutamatergic transmission in the hippocampus but not in the cortex
US20220168316A1 (en) Methods and compositions for treating urea cycle disorders
US20220288004A1 (en) Treatment and prevention of aging related-disease and/or aging by the inhibition of sphingolipids
Wei et al. Neurofibromin 1 controls metabolic balance and Notch-dependent quiescence of juvenile myogenic progenitors
Zhao et al. Triazole/thiadiazole substituted 4′-demethylepipodophyllotoxin derivatives induced apoptosis in HeLa cells by up-regulating TMEM133
Villamor Payà The Tousled-like kinases and their implications in cancer and neurodevelopmental disorders
WO2019188950A1 (en) Medicinal composition
Chatterjee et al. DNA-PK sustains autophagy and pancreatic cancer cell growth
Sokolov et al. NMNAT1 facilitates energy metabolism and gene regulation in developing retinal neurons
Kurmis Activity of Py-Im polyamides in anti-androgen resistant prostate cancer models
Castro Neurogenetic Function of Host Cell Factor-1

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: 19781469

Country of ref document: EP

Kind code of ref document: A1

DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)
NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 19781469

Country of ref document: EP

Kind code of ref document: A1