+

WO2018129421A1 - Candidat-médicament prometteur pour la maladie de parkinson - Google Patents

Candidat-médicament prometteur pour la maladie de parkinson Download PDF

Info

Publication number
WO2018129421A1
WO2018129421A1 PCT/US2018/012716 US2018012716W WO2018129421A1 WO 2018129421 A1 WO2018129421 A1 WO 2018129421A1 US 2018012716 W US2018012716 W US 2018012716W WO 2018129421 A1 WO2018129421 A1 WO 2018129421A1
Authority
WO
WIPO (PCT)
Prior art keywords
disease
parkinson
genes
expression
regulates
Prior art date
Application number
PCT/US2018/012716
Other languages
English (en)
Inventor
Pui-Yuk Dorothy Ko
William Swindell
Original Assignee
Genervon Biopharmaceuticals, LLC
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 Genervon Biopharmaceuticals, LLC filed Critical Genervon Biopharmaceuticals, LLC
Publication of WO2018129421A1 publication Critical patent/WO2018129421A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/08Peptides having 5 to 11 amino acids
    • A61K38/095Oxytocins; Vasopressins; Related peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/08Peptides having 5 to 11 amino acids

Definitions

  • the field includes using the MNTF Factor known as GM6 for the diagnosing, monitoring, prognosing Parkinson's disease, and preventing, delaying the onset, or treating Parkinson's disease through regulation of expression of multiple genes by GM6 to regulate the pathology of Parkinson's disease.
  • MNTF Factor known as GM6
  • MNTF1 and MNTF2 motoneuronotrophic factors
  • MNTF1 polypeptides were shown to selectively enhance the survival in vitro of anterior horn motor neurons isolated from rat lumbar spinal cord explants. Photomicrographs of treated cultures exhibited neurite outgrowth of myelinated nerve fibers and a marked reduction in the growth of non-neuronal cells, e.g. glial cells and fibroblasts. Similarly, in vivo administration of MNTF1 to surgically axotomized rat peripheral nerves resulted in a markedly higher percentage of surviving motor neurons than untreated controls, which could be blocked by co-administration of anti-MNTFl monoclonal antibody.
  • MNTF1 myelinated nerve fiber formation.
  • spinal cord hemi-section repaired by a peripheral nerve autograft and implanted with MNTF1- containing gel sections in close proximity to the nerve graft junctions with spinal cord.
  • MNTF1 treated animals exhibited greater numbers of surviving motor neurons, improved recovery of motor and sensory function, reduced inflammatory response (fewer infiltrating macrophages and lymphocytes) and reduced collagen-containing scar tissue formation at the site of the graft, normal Schwann cell morphology and normal myelinated and non-myelinated nerve fiber formation.
  • Each of these domains are sufficient to stimulate the proliferation of motor neuron derived cell lines in a manner similar to the MNTF1-F6 33-mer.
  • the "FSRYAR” domain is sufficient to direct selective reinnervation of muscle targets by motor neurons in vivo in a manner similar to the MNTF1-F6 33-mer.
  • the "FSRYAR” domain provides an antigenic epitope sufficient to raise antibody that recognizes any MNTF peptide containing the "FSRYAR” sequence, including the MNTF1-F6 33-mer.
  • Novel peptides and composition from active fragments of MNTF that are capable of modulating viability and growth in neuronal cells, and to methods of modulating neuronal cell viability and growth employing the novel peptides and compositions, containing either a "WMLSAFS domain” or "FSRYAR domain", which is sufficient for neurotrophic or neurotropic function is described in US Patent 7,183,373.
  • the polypeptide domain demonstrated therein were sufficient for the selective maintenance and axonal regeneration of neuronal cells, and to peptides and/or molecules capable of mimicking their structure and/or function.
  • Preferred embodiments of that invention comprise a peptide having the amino acid sequence: FSRYAR [SEQ ID NO:2], the sequence of GM6, also known as GM604 for ALS indication, as well as analogues thereof.
  • FSRYAR amino acid sequence
  • GM6 also known as GM604 for ALS indication
  • analogues thereof are functional equivalents of the GM6 [SEQ ID NO:2].
  • GM6 encompasses the active domain of MNTF, an endogenous master neural growth regulator present during the fetal development phase when neurons are being created and reaching terminal synaptic targets.
  • the fetal phase is the most intense and rapid period of human growth and development, especially within the CNS.
  • treatment of GM604 in rodents demonstrated that it promotes neural regeneration and exhibits both trophic and tropic effects (Chau RMW et al. 1990 Neuronotrophic Factor. Chin J. Neuroanat. 6:129-138; Chau RMW et al. 1992. Muscle neurotrophic factors specific for anterior horn motoneurons of rat spinal cord. Recent Adv. Cell Mol Biol. 5:89-94; Yu J. et al. 2008.
  • Motoneuronotrophic Factor analog GM604 reduces infarct volume and behavioral deficits following transient ischemia in the mouse. Brain Res. 1238:143-153; US 7,183,373).
  • Parkinson's disease or paralysis agitans is a basal ganglia disorder characterized by the loss of dopaminergic neurons within the midbrain substantia nigra pars compacta and ventral tegmental area. It is a relatively common degenerative disease of aging affecting approximately 1% of the population 50 years of age and older. The disease is usually diagnosed based upon clinical symptoms although radioactive positron emission tomography can be used as a diagnostic aide.
  • Clinical symptoms of PD include a "pill rolling" tremor at rest, cogwheel or lead-pipe rigidity, masked (expressionless) facies, restlessness (akinesia), bradykinesia, postural instability, and shuffling gait with festination and loss of arm swing. Patients may additionally present with sialorrhea (excess of saliva) and display micrographia (small handwriting). In contrast to other forms of tremor, the PD resting tremor is most noticeable in the hands and distal appendages and is alleviated by intentional movement.
  • PD neuromelanin pigment granules and the presence of intracellular eosinophilic alpha- synuclein inclusions known as Lewy bodies. It is likely that both genetic and environmental factors contribute to PD.
  • the genetic component is polygenic and thus far more than 41 genetic loci have been linked to PD based upon genome- wide association studies (PMID: 28892059).
  • PD is driven by decreased activity of the nigrostriatal dopamine pathway secondary to the loss of dopaminergic neurons leading to decreased dopamine abundance in the caudate nucleus and putamen.
  • disturbance of other neurotransmitters is also observed, with decreased abundance of serotonin and elevations of acetylcholine, leading to altered neurotransmitter balance that may worsen or contribute to the disease process (PMID: 20590830).
  • GABA A inhibitory neurotransmitter gamma- Aminobutyric acid
  • L-DOPA/carbidopa agents increasing L-DOPA availability and preventing its degradation
  • agents that prevent dopamine degradation agents that prevent dopamine degradation
  • dopamine agonists agents that prevent dopamine degradation
  • bromocriptine, pramipexole, ropinirole dopamine agonists
  • drugs increasing dopamine availability amantadine
  • anti-cholinergic drugs benztropine, trihexyphenidyl
  • the mainstay of PD therapy has been the synergistic combination of Levodopa with carbidopa (Sinemet), which serves to increase L-DOPA while the addition of DOPA decarboxylase inhibitor (carbidopa) increases L-DOPA bioavailability to facilitate its transport across the blood-brain barrier.
  • Sinemet carbidopa
  • DOPA decarboxylase inhibitor carbidopa
  • the long-term use of levodopa/carbidopa is limited by adverse effects, which include development of arrhythmias along with dyskinesia.
  • All current PD treatments are supportive and intended to address symptoms and delay disease progression, but no existing treatment has been demonstrated to restore loss of dopamine production in the midbrain of advanced PD patients. Given that defects in PD neurotransmission may thus be broader than currently recognized and not limited to dopamine, development of new multi-target may will be needed to fully prevent functional decline in PD patients (PMID: 28328536).
  • GM6 is an oligopeptide containing 6 amino acids, corresponding to the active site of an embryonic-stage protein functioning within the developing nervous system. GM6 has been shown to cross the blood-brain barrier (Yu et al. 2008, Brain Res. 1238: 143-153). The safety and tolerability of GM6 has been demonstrated in phase 1 and phase 2 clinical trials in Parkinson's Disease (PD), Amyotrophic Lateral Sclerosis (ALS) and Ischemic stroke. It has been proposed that GM6 triggers activation of pro-survival pathways mediating neural development, which may in turn have beneficial effects for treatment of multiple neurodegenerative diseases.
  • PD Parkinson's Disease
  • ALS Amyotrophic Lateral Sclerosis
  • Ischemic stroke Ischemic stroke. It has been proposed that GM6 triggers activation of pro-survival pathways mediating neural development, which may in turn have beneficial effects for treatment of multiple neurodegenerative diseases.
  • one embodiment is directed to a method of treating or preventing Parkinson's disease, the method comprising administering a MNTF peptide with the peptide sequence FSRYAR (GM6) to a subject to regulate the pathology of Parkinson's disease, wherein GM6 regulates more than one downstream target to regulate the pathology of the disease.
  • GM6 peptide sequence FSRYAR
  • GM6 acts as a master regulator of multiple targets to regulate the pathology of Parkinson' s disease to treat or prevent Parkinson's disease, where GM6 functions both as an agonist and an antagonist for different targets that regulate the pathology of Parkinson's disease, and where GM6 functions both as an agonist and an antagonist for different targets that regulate the pathology of Parkinson's disease, wherein GM6 regulates the pathology of Parkinson's disease by up regulating some targets and downregulating other targets to achieve a homeostasis.
  • Another embodiment is a method of treating or preventing Parkinson's disease, the method comprising the administering a MNTF peptide consisting of the amino acids FSRYAR (GM6) to a subject to regulate the pathology of Parkinson's disease, wherein GM6 regulates more than one downstream target to inhibit mitochondria-mediated neuronal cell death associated with Parkinson' s disease.
  • GM6 a MNTF peptide consisting of the amino acids FSRYAR
  • Another embodiment is a method of treating or preventing Parkinson's disease, the method comprising administering a MNTF peptide with the peptide sequence FSRYAR (GM6) to a subject, wherein GM6 regulates more than one of the following downstream targets to regulate the pathology of the disease: GNAI1, CASP3, PRKACB, UBE2L3, CASP9, UBE2J1, SNCAIP, ATP5B, GPR37, GNAI3, SLC18A1, UBE2G1, APAF1, UQCRC2, NDUFS1, NDUFV3, PINK1, PARK2, SNCA, NDUFA4L2, NDUFV1, ATP5E, UBE2J2, NDUFS2, UQCRC1, HTRA2, ADORA2A, COX6A2, COX7A1, LRRK2, PPIF, SLC18A2, VDAC3, SDHD, SLC25A6, COX4I2, NDUFC2, SLC25A31, GNAL, PRKACG, COX7B2,
  • Another embodiment is a method of treating or preventing Parkinson's disease, the method comprising administering a MNTF peptide with the peptide sequence FSRYAR (GM6) to a subject, wherein GM6 down-regulates at least one of the following downstream targets to regulate the pathology of the disease: NDUFBl, NDUFA12, PARK7, COX5A, ATP50, COX7B, COX7A2, NDUFB7, NDUFB2, NDUFAB1, COX6C, NDUFC1, NDUFB6, NDUFB4, COX7C, UQCRH, NDUFA2, NDUFA8, NDUFS6, NDUFA7, NDUFBl 1, NDUFB10, NDUFS5, NDUFB9, NDUFA13, ATP5D, NDUFS8, NDUFA6, COX5B, NDUFS4, SLC25A4, NDUFA1, COX6B1, NDUFS3, UQCRQ, PRKACA, NDUFA9, COX8A, ATP5G3,
  • Another embodiment is a method of treating or preventing Parkinson's disease, the method comprising administering a MNTF peptide consisting of the amino acids FSRYAR (GM6) to a subject, wherein GM6 down-regulates at least one of the following downstream targets to regulate the pathology of the disease: NDUFBl, NDUFA12, PARK7, COX5A, ATP50, COX7B, COX7A2, NDUFB7, NDUFB2, NDUFAB1, COX6C, NDUFC1, NDUFB6, NDUFB4, COX7C, UQCRH, NDUFA2, NDUFA8, NDUFS6, NDUFA7, NDUFBl 1, NDUFB10, NDUFS5, NDUFB9, NDUFA13, ATP5D, NDUFS8, NDUFA6, COX5B, NDUFS4, SLC25A4, NDUFA1, COX6B1, NDUFS3, UQCRQ, PRKACA, NDUFA9, COX8A, ATP5G3,
  • Another embodiment is a method of treating or preventing Parkinson's disease, the method comprising administering a MNTF peptide consisting of the amino acids FSRYAR (GM6) to a subject, wherein GM6 up- regulates at least one of the following downstream targets to regulate the pathology of the disease: GNAI1, CASP3, PRKACB, UBE2L3, CASP9, UBE2J1, SNCAIP.
  • GM6 up- regulates at least one of the following downstream targets to regulate the pathology of the disease: GNAI1, CASP3, PRKACB, UBE2L3, CASP9, UBE2J1, SNCAIP.
  • Another embodiment is a method of treating or preventing Parkinson's disease, the method comprising administering a MNTF peptide consisting of the amino acids FSRYAR (GM6) to a subject, wherein GM6 up- regulates at least one of the following downstream targets to regulate the pathology of the disease: GNAI1, CASP3, PRKACB.
  • GM6 up- regulates at least one of the following downstream targets to regulate the pathology of the disease: GNAI1, CASP3, PRKACB.
  • Another embodiment is a method of treating or preventing Parkinson's disease, the method comprising administering a MNTF peptide with the peptide sequence FSRYAR (GM6) to a subject, wherein GM6regulate the genes in pathways selected from the following: mitochondrial protein complex, respiratory chain complex, mitochondrial respiratory chain complex, NADH dehydrogenase complex, mitochondrial envelope, envelope, organelle membrane, catalytic complex, intracellular organelle part, mitochondrion, macromolecular complex, mitochondrial inner membrane, mitochondrial membrane part, mitochondrial respiratory chain, mitochondrial intermembrane space.
  • GM6 peptide sequence FSRYAR
  • Another embodiment is a method of treating or preventing Parkinson's disease, the method comprising the steps of: i) administering a MNTF peptide consisting of the amino acids FSRYAR [SEQ ID NO:2] (GM6) to a subject to inhibit or prevent Parkinson's disease associated neuron loss or dysfunction by (up) -regulating the expression of one or more genes selected from the group consisting of: MAPT; RAB29; PERI; HSPB1; SLC45A3; HIP1R; RET; CAPN1; ACHE; LINGOl; DNM1; NPTX2; HGF; BCKDK; SLC41A1; SNCAIP; NEDD9; GPR37; CNKSR3; HMOX1; MAOA; DRD2; SREBF1; GAD1; GFRA1; SEPT4; APOE; GST02; SNCB; WNT3; VEGFA; SYT4; RTN3; KCNN3; PRSS53; UBE2L6
  • Another embodiment is a method of treating or preventing Parkinson's disease, the method comprising the steps of: i) administering a MNTF peptide consisting of the amino acids FSRYAR [SEQ ID NO:2] (GM6) to a subject to inhibit or prevent Parkinson's disease associated neuron loss or dysfunction by (down) -regulating the expression of one or more genes selected from the group consisting of: NDUFS2; LMNB1; PRDM15; CD200; MRPL3; NSF; BRINP1; WNT3; ASCL1; SEMA5A; SFXN2; GRK5; CXCR4; DDIT4; GSTA4; DDC; VEGFA; NEDD9 and COX4I2; ATXN3; MCCC1; NDUFB2; HSPA8; SIPA1L2; CAT; TP53; CNNM2; SLC25A4; PRDX3; BORCS7; ZNF646; TRAF6; SIAH1; HTT; DD
  • Another embodiment is a method of treating or preventing Parkinson's disease, the method comprising the steps of: i) administering a MNTF peptide consisting of the amino acids FSRYAR [SEQ ID NO:2] (GM6) to a subject to inhibit or prevent Parkinson's disease associated neuron loss or dysfunction by (up) -regulating the expression of one or more genes selected from the group consisting of: MAPT; SLC41A1; RAB29; PERI; SNCAIP; HSPB1; SLC45A3; HIP1R; NEDD9; RET; GPR37; CAPN1; ACHE; CNKSR3; LINGOl; DNM1; NPTX2; HMOX1; MAOA; DRD2; SREBF1; HGF; GAD1; GFRA1; SEPT4; BCKDK and APOE.
  • a MNTF peptide consisting of the amino acids FSRYAR [SEQ ID NO:2] (GM6)
  • Another embodiment is a method of treating or preventing Parkinson's disease, the method comprising the steps of: i) administering a MNTF peptide consisting of the amino acids FSRYAR [SEQ ID NO:2] (GM6) to a subject to inhibit or prevent Parkinson's disease associated neuron loss or dysfunction by (down) -regulating the expression of one or more genes selected from the group consisting of: NSF; BRINP1; NDUFS2; WNT3; ASCL1; SEMA5A; LMNB1; PRDM15; SFXN2; CD200; GRK5; MRPL3; CXCR4; DDIT4; GSTA4; DDC; VEGFA; NEDD9 and COX4I2.
  • Another embodiment is a method of treating or preventing Parkinson's disease, the method comprising the steps of: i) administering a MNTF peptide consisting of the amino acids FSRYAR [SEQ ID NO:2] (GM6) to a subject to inhibit or prevent Parkinson's disease associated neuron loss or dysfunction by (up) -regulating the expression of one or more genes selected from the group consisting of: CAPN1; HGF; GST02; SNCB; ACHE; BCKDK; HSPB1; DNM1; WNT3; VEGFA; RAB29; HIP1R; SYT4; RTN3; KCNN3; PRSS53; UBE2L6; SLC45A3; KRTCAP2; RET; SLC50A1; LINGOl; C8orf4; PERI; MAPT; CRHR1-IT1; VDAC1; NOS1; NPTX2; BST1; CRYAB; SOD2; HIVEP3; ARNTL; S100B;
  • Another embodiment is a method of treating or preventing Parkinson's disease, the method comprising the steps of: i) administering a MNTF peptide consisting of the amino acids FSRYAR [SEQ ID NO:2] (GM6) to a subject to inhibit or prevent Parkinson's disease associated neuron loss or dysfunction by (down) -regulating the expression of one or more genes selected from the group consisting of: ATXN3; MCCC1; NDUFB2; HSPA8; LMNB1; MRPL3; SIPA1L2; CAT; TP53; PRDM15; CNNM2; SLC25A4; NDUFS2; PRDX3; BORCS7; ZNF646; TRAF6; SIAH1; HTT; DDRGK1; UNC13B; GIGYF2; P2RX7; CD200; NDUFS4 and LRRK2.
  • Another embodiment is a method of diagnosing Parkinson's disease in a patient, comprising: detecting the level of expression of one or more genes selected from the group consisting of: MAPT; RAB29; PERI; HSPB1; SLC45A3; HIP1R; RET; CAPN1; ACHE; LINGOl; DNM1; NPTX2; HGF; BCKDK; SLC41A1; SNCAIP; NEDD9; GPR37; CNKSR3; HMOX1; MAOA; DRD2; SREBF1; GAD1; GFRA1; SEPT4; APOE; GST02; SNCB; WNT3; VEGFA; SYT4; RTN3; KCNN3; PRSS53; UBE2L6; KRTCAP2; SLC50A1; C8orf4; CRHR1- IT1 VDAC1; NOS1; BST1; CRYAB; SOD2; HIVEP3; ARNTL; S100B;
  • Another embodiment is a method of diagnosing Parkinson's disease in a patient, comprising: detecting the level of expression of one or more genes selected from the group consisting of: NDUFS2; LMNB1; PRDM15; CD200; MRPL3; NSF; BRINP1; WNT3; ASCL1; SEMA5A; SFXN2; GRK5; CXCR4; DDIT4; GSTA4; DDC; VEGFA; NEDD9 and COX4I2; ATXN3; MCCC1; NDUFB2; HSPA8; SIPA1L2; CAT; TP53; CNNM2; SLC25A4; PRDX3; BORCS7; ZNF646; TRAF6; SIAH1; HTT; DDRGK1; UNC13B; GIGYF2; P2RX7; NDUFS4; and LRRK2 in a biological sample from said patient, wherein differential expression of said one or more gene variants in the sample as compared
  • Another embodiment is a method of diagnosing Parkinson's disease in a patient, comprising: detecting the level of expression of one or more genes selected from the group consisting of: MAPT; SLC41A1; RAB29; PERI; SNCAIP; HSPB1; SLC45A3; HIP1R; NEDD9; RET; GPR37; CAPN1; ACHE; CNKSR3; LINGOl; DNM1; NPTX2; HMOX1; MAOA; DRD2; SREBFl; HGF; GADl; GFRAl; SEPT4; BCKDK and APOE in a biological sample from said patient, wherein differential expression of said one or more gene variants in the sample as compared to control levels of expression of said one or more genes or gene variants is indicative of Parkinson' s disease.
  • one or more genes selected from the group consisting of: MAPT; SLC41A1; RAB29; PERI; SNCAIP; HSPB1; SLC45A3
  • Another embodiment is a method of diagnosing Parkinson's disease in a patient, comprising: detecting the level of expression of one or more genes selected from the group consisting of: NSF; BRINP1; NDUFS2; WNT3; ASCL1; SEMA5A; LMNB1; PRDM15; SFXN2; CD200; GRK5; MRPL3; CXCR4; DDIT4; GSTA4; DDC; VEGFA; NEDD9 and COX4I2 in a biological sample from said patient, wherein differential expression of said one or more gene variants in the sample as compared to control levels of expression of said one or more genes or gene variants is indicative of Parkinson' s disease.
  • one or more genes selected from the group consisting of: NSF; BRINP1; NDUFS2; WNT3; ASCL1; SEMA5A; LMNB1; PRDM15; SFXN2; CD200; GRK5; MRPL3; CXCR4; DDIT4;
  • Another embodiment is a method of diagnosing Parkinson's disease in a patient, comprising: detecting the level of expression of one or more genes selected from the group consisting of: CAPN1; HGF; GST02; SNCB; ACHE; BCKDK; HSPB1; DNM1; WNT3; VEGFA; RAB29; HIP1R; SYT4; RTN3; KCNN3; PRSS53; UBE2L6; SLC45A3; KRTCAP2; RET; SLC50A1; LINGOl; C8orf4; PERI; MAPT; CRHR1-IT1; VDAC1; NOS1; NPTX2; BST1; CRYAB; SOD2; HIVEP3; ARNTL; S100B; LY6E; CTSB; IGF2; DDIT4; NQOl; TMEM229B; TGM2 and HSPA1A in a biological sample from said patient, wherein differential expression of said one or more gene variants
  • Another embodiment is a method of diagnosing Parkinson's disease in a patient, comprising: detecting the level of expression of one or more genes selected from the group consisting of: ATXN3; MCCC1; NDUFB2; HSPA8; LMNB1; MRPL3; SIPA1L2; CAT; TP53; PRDM15; CNNM2; SLC25A4; NDUFS2; PRDX3; BORCS7; ZNF646; TRAF6; SIAH1; HTT; DDRGK1; UNC13B; GIGYF2; P2RX7; CD200; NDUFS4 and LRRK2 in a biological sample from said patient, wherein differential expression of said one or more gene variants in the sample as compared to control levels of expression of said one or more genes or gene variants is indicative of Parkinson' s disease.
  • one or more genes selected from the group consisting of: ATXN3; MCCC1; NDUFB2; HSPA8; LMNB1; MRPL
  • GM6 is used in combination with another drug or therapy, where GM6 is administered intravenously, where GM6 is administered subcutaneously, where GM6 is administered orally, and those where GM6 is administered by combination of intravenously and subcutaneously.
  • Fig. 1 shows that GM6 prevents cell death following treatment with salsolinol or post-mortem CSF from PD patients.
  • thiazolyl blue (MTT) assays were used to assess cell viability. Groups without the same letter differ significantly (P ⁇ 0.05, Tukey HSD test).
  • Fig. 2 shows PD-associated genes most strongly altered by GM6 (Microarray).
  • the figure shows PD-associated genes most strongly altered in SH-5YSY cells treated with GM6 for 48 hours. PD-associated genes with lowest p-values are shown.
  • An asterisk (*) is used to denote genes significantly altered with FDR ⁇ 0.10 (left margin).
  • Black bars denote genes additionally altered by GM6 based upon a FC threshold (FC > 1.50 or FC ⁇ 0.67). All genes were significantly altered by GM6 (FDR ⁇ 0.10) with FC > 1.50 or FC ⁇ 0.67. Genes were linked to PD based upon 2 or more of 9 possible database sources.
  • Fig. 3 illustrates PD-associated genes are predominantly repressed by GM6 in SH-SY5Y cells and associated with mitochondria.
  • A Simulation analyses. Sets of 104 SH- SY5Y-expressed genes were sampled at random. The histogram shows average FC among sets of 104 randomly sampled genes (arrow: observed average FC among 104 PD-associated genes).
  • C GO CC terms enriched among PD- associated GM6-decreased genes (conditional hypergeometric test; left margin parentheses: no. of GM6-decreased genes associated with each term; right margin: example GM6-decreased genes associated with each term).
  • Fig. 4 illustrates the KEGG PD pathway (hsa05012). Dark grey elements are associated with genes up-regulated in GM6-treated cells compared to CTL cells, and light grey elements are associated with genes down-regulated in GM6-treated cells compared to CTL cells (see color scale, lower right). White elements have not been assigned to specific genes.
  • An interactive version of this diagram is available online:
  • Fig. 5 illustrates that GM6 improves behavioral, biochemical, and histological features in the 6-OHDA mouse model.
  • Male C57BL/6 mice received intracerebral injection of 6- OHDA or solvent (CTL).
  • CTL solvent
  • Treated mice received GM6 twice daily (i.v.) over 5 days after 6- OHDA treatment (0, 1, 5, 10 or 20 mg/kg in saline).
  • CTL rotarod performance
  • C brain dopamine
  • D brain DOPAC
  • E brain HVA
  • F number of tyrosine hydroxylase positive neurons in the substantia nigra pars compacta
  • Groups without the same letter differ significantly (P ⁇ 0.05, Tukey HSD test).
  • Fig. 6 shows how GM6 improves behavioral, biochemical, and histological features in the MPTP mouse model.
  • C57BL/6 mice received intracerebral injection of MPTP or solvent (CTL).
  • Treated mice received GM6 twice daily (i.v.) over 4 days after MPTP treatment (0, 1, 5, 10 or 20 mg/kg in saline).
  • A spontaneous activity
  • B rotarod performance
  • C brain dopamine
  • D brain DOPAC
  • E brain HVA
  • F number of tyrosine hydroxylase positive neurons in the substantia nigra pars compacta
  • Groups without the same letter differ significantly (P ⁇ 0.05, Tukey HSD test).
  • Fig. 7 shows short-term GM6 treatment improves BDNF blood levels in PD patients (Phase 2A trial).
  • BDNF was evaluated at baseline (visit 1), after 4 doses (visit 4), after 6 doses (visit 6) and 10 weeks post-treatment (visit 8).
  • visit 4 after 4 doses
  • visit 6 after 6 doses
  • visit 8 10 weeks post-treatment
  • Fig. 8 shows genes associated with PD and their response to GM6 treatment (3+ database sources; directional test).
  • PD-associated genes were identified and their average fold- change (GM6/CTL) was compared to randomly sampled gene sets.
  • the arrow indicates the average fold-change (GM6/CTL) among PD-associated genes.
  • the grey histogram represents the distribution of average fold-change estimates in randomly sampled gene sets (10,000 random samples for each analysis).
  • PD-associated genes used for this analysis include only those linked to PD based upon at least 3 database sources.
  • Fig. 9 shows genes associated with PD and their response to GM6 treatment (3+ database sources; non-directional test).
  • PD-associated genes were identified and their average absolute fold-change [2 A abs(log2(GM6/CTL))] was compared to randomly sampled gene sets.
  • the arrow indicates the average absolute fold-change among PD-associated genes.
  • the grey histogram represents the distribution of average absolute fold-change estimates in randomly sampled gene sets (10,000 random samples for each analysis).
  • PD-associated genes used for this analysis include only those linked to PD based upon at least 3 database sources.
  • Fig. 10 shows PD-associated genes most strongly altered by GM6 (RNA-seq).
  • the figure shows PD-associated genes most strongly altered in SH-5YSY cells (A) treated with GM6 for 6 hours (UM, RNA-seq), (B) treated with GM6 for 24 hours (UM, RNA-seq), (C) treated with GM6 for 48 hours (UM, RNA-seq), (D) treated with GM6 for 6 hours (SBH, 6 hours), and (E) treated with GM6 for 24 hours (SBH, 24 hours).
  • PD-associated genes with lowest p-values are shown in each case. Genes with black bars were altered by GM6 with FC > 1.50 or FC ⁇ 0.67. An asterisk (*) is used to denote genes significantly altered with FDR ⁇ 0.10 (left margin). Genes were linked to PD based upon 2 or more of 9 possible database sources.
  • Fig. 11 shows Gene ontology biological process (BP) terms associated with PD- associated genes altered by GM6 (UM, RNA-seq).
  • BP Gene ontology biological process
  • A Gene ontology BP terms enriched with respect to GM6-increased PD-associated genes (FDR ⁇ 0.10; pooled from the 3 UM RNA-seq experiments). Values in parentheses (left margin) indicate the number of GM6-increased genes in each category (right margin: exemplar genes most strongly induced by GM6).
  • B Gene ontology BP terms enriched with respect to GM6-decreased PD-associated genes (FDR ⁇ 0.10; pooled from the 3 UM RNA-seq experiments). Values in parentheses (left margin) indicate the number of GM6-decreased genes in each category (right margin: exemplar genes most strongly repressed by GM6).
  • Fig. 12 shows Gene ontology biological process (BP) terms associated with PD- associated genes altered by GM6 (SBH, RNA-seq).
  • A Gene ontology BP terms enriched with respect to GM6-increased PD-associated genes (FDR ⁇ 0.10; pooled from the 2 SBH RNA-seq experiments). Values in parentheses (left margin) indicate the number of GM6-increased genes in each category (right margin: exemplar genes most strongly induced by GM6).
  • B Gene ontology BP terms enriched with respect to GM6-decreased PD-associated genes (FDR ⁇ 0.10; pooled from the 2 SBH RNA-seq experiments). Values in parentheses (left margin) indicate the number of GM6-decreased genes in each category (right margin: exemplar genes most strongly repressed by GM6)
  • Fig. 13 shows KEGG pathways in PD diagram (hsa05012; SH-5YSY cells, UM RNA-seq, 6 hours GM6 treatment). Dark grey elements are associated with genes up-regulated in GM6-treated cells compared to CTL cells, and light grey elements are associated with genes down-regulated in GM6-treated cells compared to CTL cells (see color scale, lower right). White elements have not been assigned to specific genes..
  • An interactive version of this diagram is available online:
  • Figure 14 shows KEGG pathways in PD diagram (hsa05012; SH-5YSY cells, UM RNA-seq, 24 hours GM6 treatment). Dark grey elements are associated with genes up- regulated in GM6-treated cells compared to CTL cells, and light grey elements are associated with genes down-regulated in GM6-treated cells compared to CTL cells (see color scale, lower right). White elements have not been assigned to specific genes.
  • An interactive version of this diagram is available online:
  • Fig. 15 shows KEGG pathways in PD diagram (hsa05012; SH-5YSY cells, UM RNA-seq, 48 hours GM6 treatment). Dark grey elements are associated with genes up-regulated in GM6-treated cells compared to CTL cells, and light grey elements are associated with genes down-regulated in GM6-treated cells compared to CTL cells (see color scale, lower right). White elements have not been assigned to specific genes.
  • An interactive version of this diagram is available online:
  • Fig. 16 shows KEGG pathways in PD diagram (hsa05012; SH-5YSY cells, SBH
  • RNA-seq 6 hours GM6 treatment.
  • Dark grey elements are associated with genes up-regulated in GM6-treated cells compared to CTL cells, and light grey elements are associated with genes down-regulated in GM6-treated cells compared to CTL cells (see color scale, lower right).
  • White elements have not been assigned to specific genes.
  • Fig. 17 shows KEGG pathways in PD diagram (hsa05012; SH-5YSY cells, SBH
  • RNA-seq 24 hours GM6 treatment. Dark grey elements are associated with genes up-regulated in GM6-treated cells compared to CTL cells, and light grey elements are associated with genes down-regulated in GM6-treated cells compared to CTL cells (see color scale, lower right). White elements have not been assigned to specific genes.An interactive version of this diagram is available online:
  • Parkinson's is a debilitating disease for which existing drugs primarily control symptoms without preventing underlying neurodegeneration.
  • the purpose of this particular study was to evaluate GM6 as a PD drug candidate, utilizing both in vitro and in vivo approaches to model PD pathogenesis. Additionally, to gain insight into potential mechanisms of action, we used gene expression profiling (DNA microarrays and RN A- sequencing) to identify and characterize PD-associated genes altered by GM6 treatment.
  • Salsolinol is a neurotoxin byproduct of dopamine metabolism elevated in CSF from PD patients.
  • GM6 GM6 up-regulated 3 genes 2 fold (FDR ⁇ 0.10 and FC >2.0) and 7 genes 1.5 fold (FDR ⁇ 0.10 and FC > 1.50).
  • GM6 blood-derived neurotrophic factor, BDNF; Figure 7). Long-term data based upon larger patient cohorts appear justified and are needed to fully address clinical efficacy of GM6 in this context.
  • GM6 is not a cocktail of different molecules. It is an endogenous embryonic stage tyrosine kinase motoronotropic factor regulator.
  • GM6 binds to insulin receptor, IGF1 and IGF2 receptors of the human nervous system.
  • Our data suggest that the mechanism of action for GM6 appears to involve a number of different pathways and that, collectively, the combined action serves to promote homeostasis and correction of multiple disease states.
  • Our data also suggest that GM6 does not act solely as an agonist or antagonist.
  • Our findings indicate that GM6 can up and down regulate the same genes depending on whether the disease gene is below or above the normal range respectively. The findings also suggest that GM6 regulates through multiple signaling pathways in response to distress signals.
  • GM6 regulates 1,259 (2 fold) and 2,207 (1.5 fold) genes in the DNA microarray, within which GM6 regulates 89 (2 fold) to 238 (1.5 fold) ALS associated genes, 48 (2 fold) to 68 (1.5 fold) AD associated genes, 46 (2 fold) and to 59 (1.5 fold) PD associated genes.
  • GM6 may involve attenuation of mitochondrial dysfunction, e.g., decreased ROS production and/or dampening of intrinsic apoptosis cascades.
  • GM6 has the utility of therapeutic effect to treat or prevent PD disease. By preventing death of dopaminergic neurons, GM6 may improve symptoms but also address the underlying disease etiology.
  • the pharmaceutical formulations provided herein may further include, as optional ingredients, pharmaceutically acceptable carriers, diluents, solubilizing or emulsifying agents, and salts of the type that are available in the art.
  • pharmaceutically acceptable carriers such as physiologically buffered saline solutions and water.
  • diluents such as phosphate buffered saline solutions pH 7.0-8.0.
  • Suitable pharmaceutical carriers include, but are not limited to sterile water, salt solutions (such as Ringer's solution), alcohols, polyethylene glycols, gelatin, carbohydrates such as lactose, amylose or starch, magnesium stearate, talc, silicic acid, viscous paraffin, fatty acid esters, hydroxymethylcellulose, polyvinyl pyrolidone, etc.
  • the pharmaceutical preparations can be sterilized and desired, mixed with auxiliary agents, e.g., lubricants, preservatives, stabilizers, wetting agents, emulsiers, salts for influencing osmotic pressure, buffers, coloring, and/or aromatic substances and the like which do not deleteriously react with the active compounds. They can also be combined where desired with other active substances, e.g., enzyme inhibitors, to reduce metabolic degradation.
  • auxiliary agents e.g., lubricants, preservatives, stabilizers, wetting agents, emulsiers, salts for influencing
  • Compounds provided herein may be formulated in a pharmaceutical composition, which may include pharmaceutically acceptable carriers, thickeners, diluents, buffers, preservatives, surface active agents, neutral or cationic lipids, lipid complexes, liposomes, penetration enhancers, carrier compounds and other pharmaceutically acceptable carriers or excipients and the like in addition to the compound.
  • a pharmaceutical composition may include pharmaceutically acceptable carriers, thickeners, diluents, buffers, preservatives, surface active agents, neutral or cationic lipids, lipid complexes, liposomes, penetration enhancers, carrier compounds and other pharmaceutically acceptable carriers or excipients and the like in addition to the compound.
  • compositions may also include one or more active ingredients such as interferons, antimicrobial agents, anti-inflammatory agents, anesthetics, and the like.
  • Formulations for parenteral administration may include sterile aqueous solutions which may also contain buffers, liposomes, diluents and other suitable additives.
  • Pharmaceutical compositions comprising the compounds provided herein may include penetration enhancers in order to enhance the alimentary delivery of the compounds.
  • Penetration enhancers may be classified as belonging to one of five broad categories, i.e., fatty acids, bile salts, chelating agents, surfactants and non-surfactants (Lee et al, Critical Reviews in Therapeutic Drug Carrier Systems 8, 91-192 (1991); Muranishi, Critical Reviews in Therapeutic Drug Carrier Systems 7, 1-33 (1990)).
  • One or more penetration enhancers from one or more of these broad categories may be included.
  • the pharmaceutical composition of the invention can be administered locally, nasally, orally, gastrointestinally, intrabronchially, intravesically, intravaginally, into the uterus, sub-cutaneously, intramuscularly, periarticularly, intraarticularly, into the cerebrospinal fluid (ICSF), into the brain tissue (e.g. intracranial administration), into the spinal medulla, into wounds, intraperitoneally or intrapleurally, or systemically, e.g. intravenously, intraarterially, intraportally or into the organ directly, such as the heart.
  • ISF cerebrospinal fluid
  • the compounds provided herein may be administered parentally. It is sometimes preferred that certain compounds are combined with a pharmaceutically acceptable carrier or diluent to produce a pharmaceutical composition.
  • Suitable carriers and diluents include isotonic saline solutions, for example phosphate-buffered saline.
  • the composition may be formulated for parenteral, intramuscular, intracerebral, intravenous, subcutaneous, or transdermal administration. Uptake of nucleic acids by mammalian cells is enhanced by several known transfection techniques, for example, those that use transfection agents.
  • the formulation which is administered may contain such agents. Example of these agents include cationic agents (for example calcium phosphate and DEAE-dextran) and lipofectants (for example lipofectamTM and transfectam TM).
  • Formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders.
  • Conventional pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable.
  • Coated gloves, condoms, and the like may also be useful.
  • Compositions for oral administration include powders or granules, suspensions or solutions in water or non-aqueous media, capsules, sachets or tablets. Thickeners, flavoring agents, diluents, emulsifiers, dispersing aids or binders may be desirable.
  • compositions for parenteral administration may include sterile aqueous solutions which may also contain buffers, diluents and other suitable additives. In some cases it may be more effective to treat a patient with an compound in conjunction with other traditional therapeutic modalities in order to increase the efficacy of a treatment regimen.
  • treatment regimen is meant to encompass therapeutic, palliative and prophylactic modalities.
  • Dosing can be dependent on a number of factors, including severity and responsiveness of the disease state to be treated, and with the course of treatment lasting from several days to several months, or until a cure is effected or a diminution of the disease state is achieved.
  • Toxicity and therapeutic efficacy of compounds provided herein can be determined by standard pharmaceutical procedures in cell cultures or experimental animals. For example, for determining The LD5 0 (the dose lethal to 50% of the population) and the ED5 0 (the dose therapeutically effective in 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD5 0 /ED5 0 .
  • Compounds which exhibit large therapeutic indices are preferred.
  • While compounds that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets such compounds to the site of affected tissues in order to minimize potential damage to uninfected cells and, thereby, reduce side effects.
  • the data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage for use in humans.
  • the dosage of such compounds lies preferably within a range of circulating concentrations that include the ED5 0 with little or no toxicity.
  • the dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
  • the therapeutically effective dose can be estimated initially from cell culture assays.
  • a dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC 50 (i.e., the concentration of the test compound which achieves a half-maximal inhibition of symptoms) as determined in cell culture. Such information can be used to more accurately determine useful doses in humans. Levels in plasma may be measured, for example, by high performance liquid chromatography. Dosing schedules can be calculated from measurements of drug accumulation in the body of the patient.
  • Suitable dosage amounts may, for example, vary from about 0.1 ug up to a total dose of about 1 gram, depending upon the route of administration.
  • Guidance as to particular dosages and methods of delivery is provided in the literature and generally available to practitioners in the art. Those skilled in the art will employ different formulations for nucleotides than for proteins or their inhibitors. Similarly, delivery of compounds provided herein will be specific to particular cells, conditions, and locations. In general, dosage is from 0.01 mg/kg to 100 mg per kg of body weight, and may be given once or more daily, weekly, monthly or yearly, or even less frequently. In the treatment or prevention of certain conditions, an appropriate dosage level will generally be about 0.001 to 100 mg per kg patient body weight per day which can be administered in single or multiple doses.
  • compositions may, for example, comprise one or more recombinant expression constructs, and/or expression products of such constructs, in combination with a pharmaceutically acceptable carrier, excipient or diluent.
  • a pharmaceutically acceptable carrier excipient or diluent.
  • Such carriers will be nontoxic to recipients at the dosages and concentrations employed.
  • a suitable dosage may be from about 0.01 g/kg to about 1 g/kg body weight, typically by the intradermal, subcutaneous, intramuscular or intravenous route, or by other routes.
  • a more typical dosage is about 1 g/kg to about 500 mg/kg, with about 10 ⁇ g kg, 100 ⁇ g/kg, 1 mg/kg, 10 mg/kg, 20 mg/kg, 50 mg/kg, 100 mg/kg, 200 mg/kg, 300 mg/kg, 400 mg/kg, and various ranges within these amount being still more typical for administration. It will be evident to those skilled in the art that the number and frequency of administration will be dependent upon the response of the host. "Pharmaceutically acceptable carriers" for therapeutic use are well known in the pharmaceutical art, and are described, for example, in Remingtons Pharmaceutical Sciences, Mack Publishing Co. (A.R. Gennaro edit. 1985).
  • sterile saline and phosphate-buffered saline at physiological pH may be used.
  • Preservatives, stabilizers, dyes and even flavoring agents may be provided in the pharmaceutical composition.
  • sodium benzoate, sorbic acid and esters of p-hydroxybenzoic acid may be added as preservatives. Id. at 1449.
  • antioxidants and suspending agents may be used. Id.
  • “Pharmaceutically acceptable salt” refers to salts of the compounds of the present invention derived from the combination of such compounds and an organic or inorganic acid (acid addition salts) or an organic or inorganic base (base addition salts).
  • the compounds of the present invention may be used in either the free base or salt forms, with both forms being considered as being within the scope of the present invention.
  • compositions provided herein may be in any form which allows for the composition to be administered to a patient.
  • the composition may be in the form of a solid, liquid or gas (aerosol).
  • routes of administration include, without limitation, oral, topical, parenteral (e.g., sublingually or buccally), sublingual, rectal, vaginal, and intranasal.
  • parenteral e.g., sublingually or buccally
  • parenteral includes subcutaneous injections, intravenous, intramuscular, intrasternal, intracavernous, intrathecal, intrameatal, intraurethral injection or infusion techniques.
  • the pharmaceutical composition is formulated so as to allow the active ingredients contained therein to be bioavailable upon administration of the composition to a patient.
  • Compositions that will be administered to a patient take the form of one or more dosage units, where for example, a tablet may be a single dosage unit, and a container of one or more compounds of the invention in aerosol form may hold a plurality of
  • an excipient and/or binder may be present.
  • examples are sucrose, kaolin, glycerin, starch dextrins, sodium alginate, carboxymethylcellulose and ethyl cellulose.
  • Coloring and/or flavoring agents may be present.
  • a coating shell may be employed.
  • the composition may be in the form of a liquid, e.g., an elixir, syrup, solution, emulsion or suspension.
  • the liquid may be for oral administration or for delivery by injection, as two examples.
  • preferred compositions contain, in addition to one or more binding domain-immunoglobulin fusion construct or expressed product, one or more of a sweetening agent, preservatives, dye/colorant and flavor enhancer.
  • a surfactant, preservative, wetting agent, dispersing agent, suspending agent, buffer, stabilizer and isotonic agent may be included.
  • a liquid pharmaceutical composition as used herein, whether in the form of a solution, suspension or other like form, may include one or more of the following adjuvants: sterile diluents such as water for injection, saline solution, preferably physiological saline, Ringer's solution, isotonic sodium chloride, fixed oils such as synthetic mono or digylcerides which may serve as the solvent or suspending medium, polyethylene glycols, glycerin, propylene glycol or other solvents; antibacterial agents such as benzyl alcohol or methyl paraben; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose.
  • the parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
  • Compounds described herein can be used in diagnostics, therapeutics, prophylaxis, and as research reagents and in kits. Provision of means for detecting compounds of the invention can routinely be accomplished. Such provision may include enzyme conjugation, radiolabelling or any other suitable detection systems. Kits for detecting the presence or absence of compounds of the invention may also be prepared.
  • the compounds of the invention may also be used for research purposes.
  • the specific activities or modalities exhibited by the compounds may be used for assays, purifications, cellular product preparations and in other methodologies which may be appreciated by persons of ordinary skill in the art.
  • Example 1 DNA microarray and RNA-Seq Analysis of GM6 regulated PD- associated genes
  • the purpose of this study was to provide a bioinformatic analysis to assess whether GM6 alters the expression of genes associated with PD.
  • Gene expression analyses are performed using several gene expression profiling datasets generated using DNA microarray or RNA-seq technology. These datasets evaluated effects of GM6 on gene expression in SH-SY5Y neuroblastoma cells. Effects of GM6 in SH-SY5Y neuroblastoma cells were of main interest as an in vitro dopaminergic neuron model (PMID: 20497720). Goals were to determine whether PD-associated genes exhibit unique responses to GM6 treatment, which would suggest that GM6 is impacting signaling pathways linked to core processes that underlie PD onset and/or progression.
  • Dataset (1) was generated using DNA microarray technology and 2 technical replicates. For these data, differential expression analyses are performed based upon estimated fold-change (FC) values and FDR estimates. Since technical and not biological replicates were used in these experiments, FDR estimates are used for heuristic purposes but are not true FDR estimates (which would require biological replication).
  • FC estimated fold-change
  • Datasets (2) - (6) were generated using RNA-seq technology with experiments carried out by laboratories at Sunny Biodiscovery (Santa Paula, CA) or SBH Sciences (Natick,
  • RNA-seq data were generated by core facilities at the University of Michigan (Ann Arbor, MI), while for datasets (5) and (6) RNA-seq was carried out by Phalanx
  • Genes were linked to PD based upon an association from one or more of 9 database sources (Table 2).
  • the databases used vary in terms of their criteria and stringency for identifying a gene as PD-associated. All analyses included in this report were performed using genes linked to PD based upon at least 2 of the 9 database sources, with many highlighted genes linked to PD based upon several sources (Table 2). Table 2.
  • MeSH Medical Subject Headings
  • cDisease Ontology is a disease-centered database with genes organized according to disease etiology (http://www.disease-ontology.org). PD-associated genes were identified based upon the DO identifier 10652.
  • dDisGeNET provides a comprehensive catalogue of genes and variants associated to human diseases (http://www.disgenet.org).
  • eKyoto Encyclopedia of Genes and Genomes eKyoto Encyclopedia of Genes and Genomes
  • PD-associated genes were identified based upon the KEGG pathway identifier hsa05010 (http://www.kegg.jp/).
  • eDGAR Database of Disease-Gene Associations
  • the eDGAR database integrates gene-disease associations based upon the OMIM, HUMSAVAR and CLINVAR databases.
  • hGenes were obtained from Table S6 from Mariani et al. 2016, PLoS ONE l l:e0161567.
  • the table lists genes identified as differentially expressed between PD patient substantia nigra samples and those from healthy control subjects (whole tissue or laser microdissected samples).
  • LRRK2 leucine rich repeat kinase 2
  • SNCA synuclein alpha
  • Table 3 Gene set analysis p-value summary. The table lists p-values from simulation analyses evaluating whether average fold-change estimates (GM6/CTL) of PD- associated genes differ significantly from randomly sampled gene sets of the same size ( Figures 8 -9)
  • aDirectional test Test evaluates whether PD-associated genes are more strongly increased or decreased in comparison to randomly sampled gene sets. The test statistic is average fold-change (GM6/CTL).
  • bNon-directional test Test evaluates whether PD-associated genes are more strongly altered (either direction) in comparison to randomly sampled gene sets. The test statistic is average absolute fold-change [abs(log2(GM6/CTL))]. It is instructive to examine trends among individual genes for which links to PD are most robustly supported by strong evidence. Overall, 3358 genes were identified as linked to PD based upon at least one database source, but only a small fraction of these were linked to PD based upon multiple sources (Data not shown). No genes were linked to PD based upon all 9 database sources, although two genes were linked to PD based upon 8 of the 9 sources (leucine rich repeat kinase 2, LRRK2; synuclein alpha, SNCA).
  • SNCA expression was not significantly altered by GM6 in any of the 6 experimental studies (P > 0.075).
  • expression of LRRK2 was decreased by GM6 in all 6 experimental studies (FC ⁇ 1.00), with a significant decrease observed in 3 of 6 experiments (P ⁇ 0.05; Data not shown).
  • GM6 increased expression of synuclein alpha interacting protein (SNCAIP) with respect to 3 of 6 experiments, including both microarray and RNA-seq studies (Data not shown).
  • RNA-seq analyses demonstrated strongly increased expression of SNCAIP, ACHE, GAD1 and APOE, and strongly decreased expression of SEMA5A, NEDD9 and COX4I2 (Figure 10).
  • UM, GM6-increased (27 genes) are:
  • GPR37 CAPN1; ACHE; CNKSR3; LINGOl; DNM1; NPTX2; HMOX1; MAOA; DRD2; SREBF1; HGF; GAD1; GFRA1; SEPT4; BCKDK; APOE.
  • UM, GM6-decreased (19 genes) are:
  • NSF NSF
  • BRINP1 NDUFS2
  • WNT3 WNT3
  • ASCL1 SEMA5A
  • LMNB1 PRDM15
  • SFXN2 CD200
  • GRK5 MRPL3
  • CXCR4 DDIT4
  • GSTA4 DDC
  • VEGFA VEGFA
  • NEDD9 COX4I2
  • the 27 GM6-increased genes were significantly associated with neurotransmitter biosynthesis, negative regulation of cellular response to oxidative stress, secretion, and positive regulation of angiogenesis (Figure 11A), while the 19 GM6-decreased PD-associated genes were significantly associated with regulation of axon guidance, neuron projection extension and positive regulation of nervous system development (Figure 1 IB).
  • the 43 GM6-increased PD-associated genes were associated with regulation of neurogenesis, signaling, apoptotic process and reactive oxygen species metabolism (Figure 12A), and the 26 GM6-decreased genes were associated with the metabolism of ribonucleoside triphosphate, purine nucleoside triphosphate, glycosyl metabolism, purine nucleoside monophosphate and reactive oxygen species ( Figure 12B).
  • RNA-seq There was a total of 40 PD-associated gene decreased by GM6 in RNA-seq: NDUFS2, LMNB1, PRDM15, CD200, MRPL3, NSF, BRINP1, WNT3, ASCL1, SEMA5A, SFXN2, GRK5, CXCR4, DDIT4, GSTA4, DDC, VEGFA, NEDD9, COX4I2,
  • GM6-decreased PD-associated genes from RNA-seq with FC ⁇ 0.67 are PRDM15, SEMA5A, VEGFA, NEDD9, and COX4I2
  • Patterns of GM6 gene regulation were visualized using color-coded KEGG PD pathway diagrams (hsa05012). Dark grey elements are associated with genes up-regulated in GM6-treated cells compared to CTL cells, and light grey elements are associated with genes down-regulated in GM6-treated cells compared to CTL cells (see color scale, lower right). White elements have not been assigned to specific genes.
  • This diagram provides a gene mapping to pathways leading to cell death and the production of Lewy bodies and disease-related peptides in dopaminergic neurons, including key processes related to disease-mediating ubiquitin and mitochondrial pathways ( Figure 4, Figures 13 - 17).
  • Pathway maps showed that GM6 induced down-regulation of ubiquitin and mitochondrial pathway components in the microarray dataset, including genes associated with protein kinase A (PKA) associated with motor impairment, as well as genes linked to ubiquitin B, tyrosine 3-monooxygenase and mitochondrial enzymes (Figure 13). Some of these patterns were attenuated with respect to RNA-seq comparisons ( Figures 11 - 17).
  • PKA protein kinase A
  • GM6 decreased expression of genes encoding tyrosine 3-monooxygenase as well as mitochondrial enzyme components, such as NADH dehydrogenase (ubiquinone) 1 subunit C2, ubiquinol-cytochrome c reductase subunit 10, cytochrome c oxidase subunit 6b, and F-type H+-transporting ATPase subunit d (UM-RNA seq experiments; 24 - 48 hours GM6 treatment; Figures 14 and 15).
  • NADH dehydrogenase ubiquinone
  • ubiquinol-cytochrome c reductase subunit 10 ubiquinol-cytochrome c reductase subunit 10
  • F-type H+-transporting ATPase subunit d UM-RNA seq experiments; 24 - 48 hours GM6 treatment; Figures 14 and 15.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Zoology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

L'objet des présents étude et rapport était d'évaluer le GM6 à titre de candidat-médicament dans le traitement de la maladie de Parkinson (MP) et ses éventuels mécanismes d'action et voies. Il a été démontré que le GM6 prolonge la survie in vitro des cellules SH-SY5Y traitées par salsolinol et des neurones corticaux de rats traités par le CSF post-mortem provenant de patients atteints de la maladie de Parkinson (MP). Un traitement à court terme par GM6 améliore les taux sanguins du BDNF chez les patients atteints de MP. De plus, le GM6 supprime tout ou partie d'une dysfonction dans 2 modèles murins de MP in vivo (6-OHDA, MPTP), avec pour effets une amélioration de la performance motrice et de la survie des neurones dopaminergiques dans les deux modèles. Des gènes associés à la MP régulés par le GM6 ont en outre été identifiés. Ces gènes ont été catégorisés en fonction de 6 mécanismes d'action hypothétiques, dont (1) l'atténuation d'une dysfonction kinasique par régulation négative de LRKK2, (2) leurs actions cholinergiques, (3) leurs effets pro-GABAergiques, (4) l'amélioration de l'activité GDNF par activation d'un axe GRFA1-RET, (5) l'amélioration des défenses contre le stress oxydatif et (6) l'émoussement de la génération des radicaux libres par réduction de l'activité ou de l'abondance mitochondriale.
PCT/US2018/012716 2017-01-09 2018-01-06 Candidat-médicament prometteur pour la maladie de parkinson WO2018129421A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201762444032P 2017-01-09 2017-01-09
US62/444,032 2017-01-09

Publications (1)

Publication Number Publication Date
WO2018129421A1 true WO2018129421A1 (fr) 2018-07-12

Family

ID=62791309

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2018/012716 WO2018129421A1 (fr) 2017-01-09 2018-01-06 Candidat-médicament prometteur pour la maladie de parkinson

Country Status (1)

Country Link
WO (1) WO2018129421A1 (fr)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019191148A1 (fr) * 2018-03-26 2019-10-03 Whitehead Institute For Biomedical Research Sidéroflexines en tant que transporteurs mitochondriaux de la sérine
CN110824156A (zh) * 2018-08-14 2020-02-21 杭州欧蒙医学检验所有限公司 神经自身免疫疾病的诊断
CN111733154A (zh) * 2020-04-30 2020-10-02 和也健康科技有限公司 磁场处理的免疫细胞及其用途
KR20230000180A (ko) * 2021-06-24 2023-01-02 연세대학교 산학협력단 도파민 미모톱 기능 합성 펩티드 및 이를 포함하는 mao-b 억제제
WO2023144742A1 (fr) 2022-01-26 2023-08-03 Universidade De Aveiro Formulations à base de liquide ionique pour la prévention ou le traitement de maladies neurologiques

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020045206A1 (en) * 1997-07-24 2002-04-18 Ningning Miao Vertebrate embryonic patterning-inducing proteins, compositions and uses related therto
WO2007124096A2 (fr) * 2006-04-21 2007-11-01 The Trustees Of Columbia University In The City Of New York Régulation de la morphologie des processus neuronaux par lrrk2
WO2008057609A2 (fr) * 2006-11-10 2008-05-15 Genervon Biopharmaceuticals Llc Procédés de traitement de troubles neuronaux à l'aide de peptides mntf et de leurs analogues

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020045206A1 (en) * 1997-07-24 2002-04-18 Ningning Miao Vertebrate embryonic patterning-inducing proteins, compositions and uses related therto
WO2007124096A2 (fr) * 2006-04-21 2007-11-01 The Trustees Of Columbia University In The City Of New York Régulation de la morphologie des processus neuronaux par lrrk2
WO2008057609A2 (fr) * 2006-11-10 2008-05-15 Genervon Biopharmaceuticals Llc Procédés de traitement de troubles neuronaux à l'aide de peptides mntf et de leurs analogues

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
TASSIGNY ET AL.: "GDNF-based therapies, GDNF-producing interneurons, and trophic support of the dopaminergic nigrostriatal pathway", IMPLICATIONS FOR PARKINSON'S DISEASE. FRONTIERS IN NEUROANATOMY, vol. 9, 13 February 2015 (2015-02-13), XP055510196 *

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019191148A1 (fr) * 2018-03-26 2019-10-03 Whitehead Institute For Biomedical Research Sidéroflexines en tant que transporteurs mitochondriaux de la sérine
US11571441B2 (en) 2018-03-26 2023-02-07 Whitehead Institute For Biomedical Research Sideroflexins as mitochondrial serine transporters
CN110824156A (zh) * 2018-08-14 2020-02-21 杭州欧蒙医学检验所有限公司 神经自身免疫疾病的诊断
CN110824156B (zh) * 2018-08-14 2023-08-11 欧蒙医学诊断(中国)有限公司 神经自身免疫疾病的诊断
CN111733154A (zh) * 2020-04-30 2020-10-02 和也健康科技有限公司 磁场处理的免疫细胞及其用途
KR20230000180A (ko) * 2021-06-24 2023-01-02 연세대학교 산학협력단 도파민 미모톱 기능 합성 펩티드 및 이를 포함하는 mao-b 억제제
KR102790040B1 (ko) 2021-06-24 2025-04-01 연세대학교 산학협력단 도파민 미모톱 기능 합성 펩티드 및 이를 포함하는 mao-b 억제제
WO2023144742A1 (fr) 2022-01-26 2023-08-03 Universidade De Aveiro Formulations à base de liquide ionique pour la prévention ou le traitement de maladies neurologiques

Similar Documents

Publication Publication Date Title
WO2018129421A1 (fr) Candidat-médicament prometteur pour la maladie de parkinson
KR102607599B1 (ko) 인플루엔자의 치료 방법
Harding et al. Proteostasis in Huntington's disease: disease mechanisms and therapeutic opportunities
Decressac et al. TFEB-mediated autophagy rescues midbrain dopamine neurons from α-synuclein toxicity
Evonuk et al. Inhibition of system xc− transporter attenuates autoimmune inflammatory demyelination
US20200095280A1 (en) Methods of using GM6 in diagnosing and treating Alzheimers disease
DE69533463T2 (de) Peptid-Inhibitoren von p33cdk2 und p34cdc2 Zellzyklus regulierenden Kinasen und humanem Papillomavirus E7 Onkoprotein
Shao et al. Therapeutic potential of the target on NLRP3 inflammasome in multiple sclerosis
US10729636B2 (en) Compositions comprising peptide WKDEAGKPLVK
Van der Walt et al. Neuroprotection in multiple sclerosis: a therapeutic challenge for the next decade
Chen et al. Effect of hypoxia‑inducible factor‑1/vascular endothelial growth factor signaling pathway on spinal cord injury in rats
EP2841068A2 (fr) Antagonistes de crhr1 pour une utilisation dans le traitement de patients présentant une hyperactivité de la crh
Ahuja et al. Harnessing the therapeutic potential of the nrf2/bach1 signaling pathway in Parkinson’s disease
US20240024304A1 (en) Methods of managing conditioned fear with neurokinin receptor antagonists
Deng et al. Role of matrix metalloproteinases in myelin abnormalities and mechanical allodynia in rodents with diabetic neuropathy
JP2024026109A (ja) 疼痛を治療し、疼痛感受性を増大させるためのペプチド及び他の薬剤
Ke et al. The mitochondrial biogenesis signaling pathway is a potential therapeutic target for myasthenia gravis via energy metabolism
Li et al. Role of catalpol in ameliorating the pathogenesis of experimental autoimmune encephalomyelitis by increasing the level of noradrenaline in the locus coeruleus
Hathazi et al. INPP5K and SIL1 associated pathologies with overlapping clinical phenotypes converge through dysregulation of PHGDH
EP1278537B1 (fr) Compositions de stimulation de la regeneration et de la reparation du systeme nerveux par regulation de l'activite de l'arginase 1 et de la synthese des polyamines
RU2620070C2 (ru) Модифицированные пептиды и их применение для лечения аутоиммунных заболеваний
CN102481265B (zh) 保护和治疗神经损伤的方法和组合物
CN112203675A (zh) 用于改善虚弱和衰老的方法
KR20230155941A (ko) 신규한 e3 리가아제 리간드 및 이의 용도
CN113209301B (zh) γCamkⅡ表达促进剂或稳定剂在制备减缓衰老和抗神经退行性药物中的应用

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: 18736739

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18736739

Country of ref document: EP

Kind code of ref document: A1

点击 这是indexloc提供的php浏览器服务,不要输入任何密码和下载