WO2017122789A1 - てんかん治療のためのアデノ随伴ウイルスビリオン - Google Patents
てんかん治療のためのアデノ随伴ウイルスビリオン Download PDFInfo
- Publication number
- WO2017122789A1 WO2017122789A1 PCT/JP2017/001048 JP2017001048W WO2017122789A1 WO 2017122789 A1 WO2017122789 A1 WO 2017122789A1 JP 2017001048 W JP2017001048 W JP 2017001048W WO 2017122789 A1 WO2017122789 A1 WO 2017122789A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- protein
- amino acid
- sequence
- promoter
- associated virus
- Prior art date
Links
Images
Classifications
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
- C12N15/79—Vectors or expression systems specially adapted for eukaryotic hosts
- C12N15/85—Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
- C12N15/86—Viral vectors
- C12N15/864—Parvoviral vectors, e.g. parvovirus, densovirus
- C12N15/8645—Adeno-associated virus
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K35/00—Medicinal preparations containing materials or reaction products thereof with undetermined constitution
- A61K35/66—Microorganisms or materials therefrom
- A61K35/76—Viruses; Subviral particles; Bacteriophages
- A61K35/761—Adenovirus
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/177—Receptors; Cell surface antigens; Cell surface determinants
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K45/00—Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
- A61K45/06—Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K48/00—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
- A61K48/005—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K48/00—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
- A61K48/0075—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the delivery route, e.g. oral, subcutaneous
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
- A61P25/08—Antiepileptics; Anticonvulsants
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
- A61P25/28—Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/705—Receptors; Cell surface antigens; Cell surface determinants
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K48/00—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
- A61K48/005—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
- A61K48/0058—Nucleic acids adapted for tissue specific expression, e.g. having tissue specific promoters as part of a contruct
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2750/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
- C12N2750/00011—Details
- C12N2750/14011—Parvoviridae
- C12N2750/14111—Dependovirus, e.g. adenoassociated viruses
- C12N2750/14122—New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2750/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
- C12N2750/00011—Details
- C12N2750/14011—Parvoviridae
- C12N2750/14111—Dependovirus, e.g. adenoassociated viruses
- C12N2750/14141—Use of virus, viral particle or viral elements as a vector
- C12N2750/14143—Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
Definitions
- the present invention relates to recombinant adeno-associated virus (rAAV) virions for neurological diseases. More specifically, the present invention relates to epilepsy, schizophrenia, autism spectrum disorder, mental retardation, anxiety, depression, migraine headache, fear compulsive symptoms, drug dependence, Angelman's syndrome, dyskinesia, dystonia, Alzheimer's disease
- the present invention relates to rAAV for the treatment of neuropsychiatric diseases such as diseases, developmental disorders (attention deficit hyperactivity disorder, Asperger syndrome).
- Epilepsy is a partial seizure that occurs in a part of the body or a generalized seizure that occurs as a whole body convulsions due to the abnormal excitation spreading to the whole cortex as a result of the occurrence of aberrant excitation of brain neurons in a relatively limited area. Such as clonic seizures).
- General seizures cause loss of consciousness. It is a disease that does not cause cramps, and may cause unconsciousness only or abnormal mental symptoms (such as psychomotor seizures).
- the current general therapeutic means mainly consists of drug treatment with antiepileptic drugs (phenytoin, carbamazepine, valproic acid etc.), surgical treatment is performed for intractable ones.
- epilepsy cases are intractable diseases whose seizures are not suppressed even by drug treatment.
- surgical treatment such as temporal lobe resection may be effective, but excision of the hippocampus on both sides causes memory loss, so if the seizure focal point is in both hippocampus, surgery It does not become adaptation.
- about 100,000 intractable epilepsy patients of unknown seizure focus such as childhood epilepsy encephalopathy (West syndrome etc.) are expected to exist in Japan, and there is no curative treatment for such patients.
- rAAV genetically modified adeno-associated virus
- Non-patent Document 1 directly administers overexpression of NLGN2 by directly administering to the hippocampus a rAAV vector that expresses neuroligin 2 (NLGN2), which is a synaptic localization protein of neurons.
- NLGN2 neuroligin 2
- the results show that it has produced changes in social behavior and inhibitory synaptic transmission, but has not been described with respect to the treatment of specific diseases.
- Moe 'et al. Non-patent Document 2
- epilepsy treatment is performed using a rAAV vector expressing neuropeptide Y to alleviate the epileptic condition.
- Non-patent document 3 epilepsy symptoms are alleviated as a result of directly administering to the hippocampus a rAAV vector that expresses antisense of neuroligin 1 (NLGN1), which is a synaptic localization protein of neurons. It is shown that it was done.
- NLGN1 neuroligin 1
- the present inventors prepared a recombinant adeno-associated virus vector containing a polynucleotide encoding neuroligin 2, which is a protein that improves the ability of inhibitory synaps to suppress the stimulation.
- the present invention was completed by finding that administration of this vector to a living body ameliorates the symptoms of epilepsy.
- the present application provides a recombinant adeno-associated virus (rAAV) vector shown below, a pharmaceutical composition containing the same, etc. for treatment of a disease related to a nerve cell such as epilepsy described below.
- rAAV adeno-associated virus
- a pharmaceutical composition containing the same, etc. for treatment of a disease related to a nerve cell such as epilepsy described below.
- rAAV adeno-associated virus
- the above polynucleotide encodes a neuroligin 2 protein comprising the amino acid sequence of SEQ ID NO: 2, 4 or 6 or an amino acid sequence having about 90% or more identity to these sequences and binding to neurexin
- the recombinant adeno-associated virus vector is a protein having a mutated amino acid sequence in which tyrosine at position 445 in the amino acid sequence of wild type AAV1 capsid protein is substituted with phenylalanine, 445 in the amino acid sequence of wild type AAV2 capsid protein Including a protein having a mutated amino acid sequence in which tyrosine at the position is replaced with phenylalanine, or a protein having a mutated amino acid sequence in which tyrosine at position 446 in the amino acid sequence of wild type AAV9 capsid protein is replaced by phenylalanine
- the adeno-associated virus recombinant vector according to any one of [1] to [3].
- the polynucleotide is synapsin I promoter sequence, myelin basic protein promoter sequence, neuron specific enolase promoter sequence, calcium / calmodulin dependent protein kinase II (CMKII) promoter sequence, tubulin ⁇ I promoter sequence, platelet derived growth Factor ⁇ chain promoter sequence, glial fibrillary acidic protein (GFAP) promoter sequence, L7 promoter sequence (cerebellar Purkinje cell specific promoter), glial fibrillary acidic protein (hGfa2) promoter sequence, and glutamate receptor delta 2 promoter (cerebellar Purkinje cell)
- Associated viral vector [6] The polynucleotide according to any one of [1] to [5], wherein the polynucleotide comprises an inverted terminal repeat (ITR) selected from the group consisting of AAV1, AAV2, AAV3, AAV4, AAV8, and AAV9. Recombinant Adeno-Associated Virus Vector. [7] The recombinant adeno-associated virus vector according to any one of [1] to [6], wherein the polynucleotide further comprises a polynucleotide for suppressing excitatory synaptic excitation. [8] A pharmaceutical composition comprising the recombinant adeno-associated virus recombinant vector according to any one of [1] to [7].
- ITR inverted terminal repeat
- Gene therapy for enhancing the function of the synaptic suppression system according to the present invention is useful as a treatment for epilepsy.
- the composition of the present invention can be expected to be effective also for intractable epilepsy patients whose seizure focus is unknown such as childhood epilepsy encephalopathy (West syndrome etc.).
- FIG. 1 a shows the results of confirmation of expression of recombinant neuroligin 2 protein in mice administered vascularized rAAV by FLAG antibody staining of tissue sections.
- FIG. 1 b shows the results of detection of expression of recombinant neuroligin 2 in mice receiving control.
- the horizontal axis represents epileptic seizure frequency in mice treated with 3 types of intracardiac administration (vascular administration type rAAV expressing NLGN2, vascular administration type rAAV expressing GFP protein, and intracardiac administration of saline). The results measured at the indicated age are shown. The result of the seizure time of the mouse
- mouth which administered said 3 types of intracardiac administration is shown. It shows the tabulated result of seizure time ⁇ seizure intensity shown in FIGS. 3 and 4.
- mouth which administered said 3 types of said administration in each week is shown.
- Fig. 7 shows the results of measuring the seizure frequency of epilepsy in a mouse (Fig.
- epilepsy schizophrenia, autism spectrum disorder, mental retardation, anxiety, depression, migraine headache, fear compulsory, including a polynucleotide encoding a protein that improves the inhibitory synaptic excitatory function in vivo
- a recombinant adeno-associated virus vector for the treatment of a disease selected from the group consisting of symptoms, drug addiction, Angelman's syndrome, dyskinesia, dystonia, Alzheimer's disease, developmental disorders (attention deficit hyperactivity disorder, Asperger syndrome) Provided.
- a synapse refers to a junction between a synaptic small head in which an axon terminal or the like of a nerve cell is swollen and a target neuron or muscle cell.
- synapses include excitatory synapses that transmit excitation and inhibitory synapses that suppress transmission of excitation.
- most synapses are chemical synapses (slow signaling) mediated by chemical transmission.
- Another type is the electrical synapse, which shows a quick response in time, but is less likely to be found in the central nervous system of mature mammals.
- amino acids such as glutamate, aspartate, cysteic acid, homocystenone etc. function as transmitters, and excitatory postsynaptic potential (EPSP) is generated, and the potential exceeds the threshold. And transmission of excitement (impulse) is performed.
- amino acids such as ⁇ -aminobutyric acid (GABA), glycine, taurine, alanine, cystathionine, serine, etc. function as transmitters to generate inhibitory postsynaptic potential (IPSP). It is believed that it suppresses or makes it difficult to generate postsynaptic neuron impulses.
- GABA ⁇ -aminobutyric acid
- IPSP inhibitory postsynaptic potential
- fast EPSP In EPSP and IPSP, one showing fast time lapse (within 100 milliseconds of full lapse) (fast EPSP or fast IPSP) and one showing slow slow time lapse lasting several tens of seconds to several tens of minutes (slow EPSP or slow IPSP) And exist.
- fast IPSP GABA related to the GABA A receptor (Cl ⁇ ) channel and glycine related to the glycine receptor (Cl ⁇ ) channel act, and as a transmitter of slow IPSP, GABA B through GABA B receptor or GABA B It is known that acetylcholine and catecholamines act.
- Proteins that enhance excitatory inhibitory function of inhibitory synapses include neuroligin 2 and neurexin involved in stabilization of synapse, GABA receptor, glutamine decarboxylase (GAD) involved in GABA biosynthesis, and glycine transport These include Na + channel protein and Cl 2 - channel protein, neuropeptide Y, the scaffold protein gephyrin, StranstK3 which is a transmembrane protein involved in inhibitory synapse formation, and PTPRD as a receptor-type tyrosine phosphatase which binds to SLITRK3.
- the polynucleotide contained in the vector of the present invention preferably contains a nucleotide sequence (SEQ ID NO: 2, 4 or 6) encoding a neuroligin 2 protein as a protein for improving the excitatory inhibitory function of inhibitory synapse.
- Neurorigin refers to a family of membrane proteins present in the postsynaptic membrane, generally divided into neuroligins 1-4. Each of these neuroligins specifically binds to the cell adhesion factor neurexin (Nurexin: NRXN) protein of the presynaptic membrane to connect the presynaptic terminal and the postsynaptic terminal.
- Neurorigin 1 is localized to excitatory synapses and is thought to mediate excitatory synaptic transmission.
- neuroligin 2 is localized to inhibitory synapses and is thought to mediate inhibitory synaptic transmission.
- neuroligin 3 is expressed in both the excitatory synapse and inhibitory synapse, in the heart, the pancreas and the like, and neuroligin 4 is expressed in the heart, the liver and the like.
- Neurexin proteins which are binding partners of neuroligin, are generally divided into neurexin 1 ⁇ -3 ⁇ and 1 ⁇ -3 ⁇ .
- the ⁇ form and the ⁇ form are long protein and short protein, respectively, which are produced by the action of different promoters from the same gene.
- Neuroligin 2 used in the present invention functionally binds to neurexin 1 ⁇ .
- amino acid sequence of neuroligin 2 protein used in the present invention known ones can be used. Such amino acid sequences include, for example, Genbank Accession Nos. AAM 46111 (human), EDL 12455 (mouse), EDMO 4903 (rat). Other available animal species include, for example, those derived from mammals such as monkeys, dogs, pigs, cows and horses. The amino acid sequences of human, mouse and rat neuroligin 2 proteins are described in SEQ ID NOs: 2, 4 and 6, respectively.
- the neuroligin 2 protein used in the present invention is about 90% or more, 91% or more, 92% or more, 93% or more, 94% or more, 95% or more, with respect to the amino acid sequence of SEQ ID NO: 2, 4 or 6 , 96% or more, 97% or more, 98% or more, 99% or more, 99.1% or more, 99.2% or more, 99.3% or more, 99.4% or more, 99.5% or more, 99.6% or more, 99.7% or more, 99.8% or more, 99.9 Included are proteins that have an amino acid sequence with% or more identity and are capable of binding to neurexin 1 ⁇ protein under physiological conditions. Generally, the larger the value, the better.
- the amino acid sequences of human and mouse neuroligin 2 are at least 98% identical, and the amino acid sequences of human and rat neuroligin 2 are at least 91% identical.
- that the mutant protein and the original protein function to the same degree means, for example, a specific activity of about 0.01 to 100, preferably about 0.5 to 20, more preferably Means that it is within the range of about 0.5 to 2, but is not limited thereto.
- the neuroligin 2 protein used in the present invention has one or more amino acids deleted, substituted, inserted and / or added in the amino acid sequence of SEQ ID NO: 2, 4 or 6 or the amino acid sequence having the above identity. Included are proteins which contain an amino acid sequence and which are capable of binding to the neurexin 1 ⁇ protein under physiological conditions. Two or more of the above amino acid deletions, substitutions, insertions and additions may occur simultaneously. Examples of such a protein include, for example, 1 to 50, 1 to 40, 1 to 39, 1 to 38, 1 to 37, and 1 to the amino acid sequence of SEQ ID NO: 2, 4 or 6.
- Examples of mutually replaceable amino acid residues in the protein (polypeptide) of the present invention are shown below.
- Amino acid residues included in the same group can be mutually substituted.
- Group A leucine, isoleucine, norleucine, valine, norvaline, alanine, 2-aminobutanoic acid, methionine, o-methylserine, t-butylglycine, t-butylalanine, cyclohexylalanine;
- Group B aspartic acid, glutamic acid, isoaspartic acid , Isoglutamic acid, 2-aminoadipic acid, 2-aminosuberic acid;
- group C asparagine, glutamine;
- group D lysine, arginine, ornithine, 2,4-diaminobutanoic acid, 2,3-diaminopropionic acid;
- group E Proline, 3-hydroxyproline, 4-hydroxyproline; F
- Neuroligin proteins in which amino acid residues are substituted can be produced according to methods known to those skilled in the art, such as ordinary genetic engineering techniques. For such gene manipulation procedures, see, for example, Molecular Cloning 3rd Edition, J. Sambrook et al., Cold Spring Harbor Lab. Press. 2001, Current Protocols in Molecular Biology, John Wiley & Sons 1987-1997, etc. Can.
- preferred polynucleotides to be used in the present invention are, for example, one or more (for example, 1 to 50, 1 to 40, 1 to 30, 1) or the like in the polynucleotide sequence of SEQ ID NO: 1, 3 or 5. -25 pieces, 1-20 pieces, 1-15 pieces, 1-10 pieces, 1-9 pieces (1-several pieces), 1-8 pieces, 1-7 pieces, 1-6 pieces, 1-5 pieces, A polynucleotide having deletion, substitution, insertion and / or addition of 1 to 4, 1 to 3, 1 to 2, 1 etc.
- nucleotides wherein the amino acid sequence of SEQ ID NO: 2, 4 or 6 Or a protein which is capable of binding to neurexin 1 ⁇ and containing an amino acid sequence in which one or more of the above amino acids are deleted, substituted, inserted and / or added in the amino acid sequence of SEQ ID NO: 2, 4 or 6 Containing polynucleotides. Combinations of two or more of these deletions, substitutions, insertions and additions may be included simultaneously. Generally, the smaller the number of deletions, substitutions, insertions and / or additions of the above nucleotides, the better.
- a preferred polynucleotide in the present invention is, for example, a polynucleotide hybridizable under stringent hybridization conditions to SEQ ID NO: 7, 9 or 11 or a complementary sequence thereof, which comprises SEQ ID NO: 2, 4 or 6 Or a polynucleotide encoding the amino acid sequence of SEQ ID NO: 2, or an amino acid sequence in which one or more of the above amino acids are deleted, substituted, inserted and / or added in the amino acid sequence of SEQ ID NO: 2, 4 or 6 It contains a polynucleotide encoding a certain protein.
- Hybridization can be performed according to a known method or a method according thereto, such as a method described in Molecular Cloning (Molecular Cloning 3rd Edition, J. Sambrook et al., Cold Spring Harbor Lab. Press. 2001). Moreover, when using a commercially available library, it can carry out according to the method as described in the instruction manual etc. which are provided by the manufacturer.
- stringent conditions may be any of low stringency conditions, moderate stringency conditions and high stringency conditions.
- “Low stringency conditions” are, for example, 5x SSC, 5x Denhardt's solution, 0.5% SDS, 50% formamide, 32 ° C.
- “moderately stringent conditions” are, for example, 5x SSC, 5x Denhardt's solution, 0.5% SDS, 50% formamide, 42 ° C.
- “Highly stringent conditions” are, for example, 5 ⁇ SSC, 5 ⁇ Denhardt's solution, 0.5% SDS, 50% formamide, 50 ° C. Under these conditions, it can be expected that DNAs having high homology can be efficiently obtained as the temperature is raised. However, multiple factors such as temperature, probe concentration, probe length, ionic strength, time, and salt concentration can be considered as factors affecting the stringency of hybridization, and those skilled in the art should appropriately select these factors. Similar stringency can be achieved with
- the hybridizable polynucleotide may be, for example, 70% or more of the nucleotide sequence of SEQ ID NO: 7, 9 or 11, as calculated using homology search software such as FASTA, BLAST, etc. using default parameters. 80% or more, 90% or more, 91% or more, 92% or more, 93% or more, 95% or more, 95% or more, 96% or more, 97% or more, 98% or more, 99% or more, 99.1% or more, 99.2%
- the polynucleotides having the same identity of 99.3% or more, 99.4% or more, 99.5% or more, 99.6% or more, 99.7% or more, 99.8% or more, 99.9% or more can be mentioned. Generally, the larger the value of the homology, the better.
- the target diseases of the present invention relates to epilepsy, schizophrenia, autism spectrum disorder, mental retardation, anxiety, depression, migraine headache, fear compulsive symptoms, drug dependence, Angelman's syndrome, dyskinesia, dystonia, Alzheimer's disease
- the present invention provides rAAV vectors that are useful for the treatment of diseases selected from the group consisting of diseases, developmental disorders (attention deficit hyperactivity disorder, Asperger syndrome), especially epilepsy.
- Epilepsy refers to the occurrence of excessive synchronous firing activity of cerebral neurons, and causes the same type of clinical seizure (systemic tonic-clonic seizure, absence seizure, auditory seizure, part of the limb) in the same individual. It refers to a pathological condition in which tonic seizures repeat).
- the classification of clinical seizures is partial seizure (simple partial seizure, complex partial seizure), general seizure (absent seizure, myoclonic seizure, tonic-clonic seizure, weakness) Attacks), classified into unclassifiable attacks.
- classification of epilepsy, epilepsy syndrome and related seizure disorders includes localization relatedness (age related, symptomatic, latent classification), generality (spontaneous, latent) It is divided into causal or symptomatic, symptomatic subclasses), those in which focal or generality can not be determined, and special syndromes (such as thermal cramps).
- West syndrome mainly characterized by short-time proflex seizures beginning in infancy around 1 year old
- West syndrome or sputum spasm
- the disease forms a series in which momentary tonic seizures occur, which fork and bow over the upper body and head.
- the etiology of the West syndrome is diverse, and congenital malformations such as congenital brain malformations, tuberous sclerosis, and congenital metabolic abnormalities such as vitamin B6 deficiency are known.
- the rAAV vectors of the invention may have a therapeutic effect on West syndrome.
- Recombinant Adeno-Associated Virus (rAAV) Vector of the Present Invention As a vector for delivering a gene used for synaptic function control to neural cells, it is also possible by peripheral administration described in WO 2012/057363.
- a recombinant adeno-associated virus vector (also referred to herein as a vascular administration vector) capable of efficient gene delivery to neural cells, or those described in WO 2008/124724 and the like can be used.
- the rAAV vector of the present invention can pass through the blood-brain barrier of a living body and is therefore delivered to the brain via the blood-brain barrier, such as by peripheral administration to a patient, nerves such as the patient's brain and spinal cord It is possible to introduce a target therapeutic gene into a system cell. It can also be administered intrathecally or directly to the target site in the brain.
- the rAAV vector of the present invention is preferably derived from natural adeno-associated virus type 1 (AAV1), type 2 (AAV2), type 3 (AAV3), type 4 (AAV4), type 5 (AAV5), type 6 (AAV6) And 7 (AAV7, 8 (AAV8), 9 (AAV9), etc.
- AAV1 natural adeno-associated virus type 1
- AAV2 type 2
- AAV3 AAV3
- AAV4 type 5
- AAV6 AAV6
- AAV9 AAV9
- AAV9 AAAV 9
- types 2, 3, 5 and 9 are of human origin, in the context of the present invention, particularly capsid proteins (VP1 derived from AAV1, AAV2 or AAV 9).
- AAV1 and AAV9 are preferably human-derived AAVs. Infection efficiency of the nerve cells have been reported to be relatively high (Taymans, et al, Hum Gene Ther 18:. 195-206, such as 2007).
- the capsid protein contained in the rAAV vector used in the present invention preferably has at least one tyrosine such as phenylalanine or the like as compared to the wild-type amino acid sequence as described in WO 2012/057363 or WO 2008/124724 etc.
- a mutant protein having an amino acid sequence substituted for the amino acid of For example, an amino acid sequence in which the tyrosine at position 445 in the amino acid sequence of wild type AAV1 capsid protein is substituted with phenylalanine (SEQ ID NO: 9), and a tyrosine at position 444 in the amino acid sequence of wild type AAV2 capsid protein is substituted with phenylalanine Amino acid sequence (SEQ ID NO: 10) or a mutant protein having an amino acid sequence (SEQ ID NO: 11) in which the tyrosine residue at position 446 in the amino acid sequence of wild type AAV9 capsid protein is substituted with phenylalanine residue (SEQ ID NO: 11) WO 2012/057363, WO 2008/124724).
- capsid proteins have the function of forming capsomers either alone or together with other capsid protein members (eg, VP2, VP3 etc).
- capsid protein members eg, VP2, VP3 etc.
- a polynucleotide containing a therapeutic gene or the like to be delivered to neural cells is packaged in the capsomer.
- neural cells as gene transfer targets include at least neurons contained in the central nervous system such as the brain and spinal cord, and further include glial cells, microglia, astrocytes, oligodendrocytes, It may also include ventricular ependymal cells, cerebral vascular endothelial cells and the like.
- the percentage of neurons among the neural cells to be transfected is preferably 70% or more, 80% or more, 85% or more, 90% or more, 91% or more, 92% or more, 93% or more, 94% or more, 95% or more, 96% or more, 97% or more, 98% or more, 99% or more, 99.1% or more, 99.2% or more, 99.3% or more, 99.4% or more, 99.5% or more, 99.6% or more, 99.7% or more, 99.8% The above is 99.9% or more, or 100%.
- the Rep protein used in the present invention has a function of recognizing an ITR sequence and performing genome replication depending on the sequence, a function of recruiting and packaging a wild-type AAV genome (or rAAV genome) into a viral vector, It may have the same number of amino acid sequence identity as described above as long as it has the same function as that of the present invention such as the function to form the rAAV vector, and deletion of the same number of amino acid residues as described above , Substitution, insertion and / or addition. Functionally similar ranges include those described in the above description of specific activity. In the present invention, preferably, known AAV3-derived Rep proteins are used.
- the polynucleotide encoding the Rep protein used in the present invention has a function of recognizing an ITR sequence and performing genome replication depending on that sequence, and recruiting a wild-type AAV genome (or rAAV genome) into a viral vector It may have the same number of identities as above, as long as it encodes a Rep protein having the same degree of known functions, such as the function to package and the function to form the rAAV vector of the present invention, It may include deletions, substitutions, insertions and / or additions of as many nucleotides. Functionally similar ranges include those described in the above description of specific activity.
- a rep gene derived from AAV3 or AAV2 is used.
- the capsid protein VP1 or the like (VP1, VP2 and / or VP3) encoded in the internal region of the wild-type AAV genome as described above, and the Rep protein It is incorporated in and used.
- the capsid proteins (VP1, VP2 and / or VP3) used in the present invention, and the Rep protein may be incorporated into one, two, three or more plasmids, as required.
- one or more of these capsid and Rep proteins may be included in the AAV genome.
- capsid proteins (VP1, VP2 and / or VP3) and Rep proteins are all encoded by one polynucleotide and provided as an AAV helper plasmid.
- the polynucleotide (ie, polynucleotide) packaged in the rAAV vector of the present invention is an internal region (ie, rep gene and cap) located between the ITRs located 5 'and 3' of the wild type genome.
- the polynucleotide of one or both of the genes can be prepared by replacing it with a gene cassette including a polynucleotide encoding a protein of interest (therapeutic gene), a promoter sequence for transcribing the polynucleotide, and the like.
- the ITRs located 5 'and 3' are located at the 5 'and 3' ends of the AAV genome, respectively.
- the rAAV genome of the present invention has ITRs located at the 5 'end and 3' end including 5 'ITRs and 3' ITRs contained in the genome of AAV1, AAV2, AAV3, AAV4, AAV8 or AAV9. .
- the ITR contained in the rAAV genome of the present invention may be reversed in the 5 'and 3' direction, since the ITR portion has a flip-flop structure in which complementary sequences are easily exchanged.
- the length of the polynucleotide replaced with the internal region ie, the therapeutic gene
- the rAAV genome of the present invention is preferably about the same as 5 kb in which the full length is wild type, for example, about 2 to 6 kb, preferably about 4 to 6 kb.
- the length of the therapeutic gene to be incorporated into the rAAV genome of the present invention is preferably the length of the transcriptional regulatory region including the promoter, polyadenylation, etc. (for example, assuming about 1 to 1.5 kb).
- the length is about 0.01 to 3.7 kb, more preferably about 0.01 to 2.5 kb, still more preferably about 0.01 to 2 kb, but not limited thereto.
- a polynucleotide packaged in a recombinant adeno-associated virus vector may take several days to express a therapeutic protein of interest if it is single stranded.
- the therapeutic gene to be introduced may be designed to be a self-complementary sc (self-complementary) type to show an effect in a shorter period of time. Specific details are described in, for example, Foust KD, et al. (Nat Biotechnol. 2009 Jan; 27 (1): 59-65).
- the polynucleotide packaged in the rAAV vector of the present invention may be non-sc or sc.
- the rAAV vector of the invention preferably comprises a polynucleotide comprising a neural cell specific promoter sequence and a therapeutic gene operably linked to the promoter sequence (ie such a poly Nucleotides are packaged).
- a promoter sequence used in the present invention a promoter sequence specific to neural cells is derived from, for example, neural cells, glial cells, oligodendrocytes, cerebral vascular endothelial cells, microglia, ventricular epithelial cells, etc. Although it is not limited to these.
- promoter sequences include synapsin I promoter sequence, myelin basic protein promoter sequence, neuron specific enolase promoter sequence, glial fibrillary acidic protein promoter sequence, L7 promoter sequence (cerebrate Purkinje cell specific promoter And glutamic acid receptor delta 2 promoter (cerebellar Purkinje cell specific promoter) sequences, glial fibrillary acidic protein (hGfa2) promoter sequences, and glutamate decarboxylase (GAD65 / GAD67) promoter sequences, but not limited thereto.
- promoter sequences such as calcium / calmodulin-dependent protein kinase II (CMKII) promoter sequence, tubulin ⁇ I promoter sequence, and platelet-derived growth factor ⁇ chain promoter sequence can also be used.
- CMKII calcium / calmodulin-dependent protein kinase II
- tubulin ⁇ I promoter sequence tubulin ⁇ I promoter sequence
- platelet-derived growth factor ⁇ chain promoter sequence can also be used.
- the above promoter sequences may be used alone or in combination of any two or more. It may also be a commonly used strong promoter sequence such as CMV promoter or CAG promoter.
- promoter sequences in the present invention include synapsin I promoter sequence, myelin basic protein promoter sequence, L7 promoter sequence (cerebellar Purkinje cell specific promoter), glutamate receptor delta 2 promoter (cerebellar Purkinje cell specific promoter) Be Furthermore, it may contain known sequences such as enhancer sequences for assisting in transcription of mRNA, translation into protein, etc., Kozak sequences, appropriate polyadenylation signal sequences and the like.
- the therapeutic gene of interest to be integrated into the rAAV genome of the present invention is delivered to neural cells with high efficiency and integrated into the cell's genome.
- the number of nerves is about 10 times or more, about 20 times or more, about 30 times or more, about 40 times or more or about 50 times or more than that when using conventional rAAV vectors It is possible to introduce a gene into cells.
- the number of gene-introduced neural cells can be determined, for example, by preparing an rAAV vector packaging an rAAV vector genome incorporating any marker gene, and then administering this rAAV vector to a test animal to prepare the rAAV vector genome It can be measured by counting the number of neural cells expressing the integrated marker gene (or marker protein).
- Known marker genes can be selected. Examples of such marker genes include LacZ gene, green fluorescent protein (GFP) gene, luminescent protein gene (such as firefly luciferase) and the like.
- Other means or additional means to improve the inhibitory synaptic excitatory function of other therapeutic genes eg, means to enhance expression of neurexin 1 alpha, a binding partner of neuroligin 2, cells of neuroligin 2 Means to improve internal signaling can be expected.
- means for reducing the function of excitatory synapse for example, suppressing the expression of a protein related to the action of excitatory synapse, specifically by antisense of neuroligin 1 Measures to reduce neuroligin 1 (Fang et al., Mol. Neurobiol. 2014 Nov.) and the like may also be useful.
- the rAAV vector of the present invention may express different proteins for synaptic function control, and such different proteins include, for example, neutralizing antibodies (antigens to proteins and receptors present on synaptic membranes). And binding sites, Fab, Fab2, single chain antibodies (scFv) and the like. Examples of these antibody classes include IgG, IgM, IgA, IgD, or IgE.
- therapeutic genes incorporated into the rAAV genome of the present invention for functional suppression of excitatory synapses may be targeted internalizing agents such as antisense molecules, ribozymes, interfering RNAs (iRNAs), microRNAs (miRNAs) It may be a polynucleotide for changing (eg, disrupting, reducing) the function of a sex gene, or a polynucleotide for changing (eg, reducing) the expression level of an endogenous protein.
- iRNAs interfering RNAs
- miRNAs microRNAs
- the length of the antisense nucleic acid is 10 bases or more, 15 bases or more, 20 bases or more, and 100 bases or more And more preferably 500 bases or more.
- the length of the antisense nucleic acid used is less than 5 kb, preferably less than 2.5 kb.
- a ribozyme By using a ribozyme, the mRNA of a target protein can be specifically cleaved to suppress the expression of that protein.
- various known documents can be referred to (for example, FEBS Lett. 228: 228, 1988; FEBS Lett. 239: 285, 1988; Nucl. Acids. Res. 17: 7059, 1989; Nature 323: 349, 1986, etc.).
- RNAi is a phenomenon in which the expression of both the introduced foreign gene and the target endogenous gene is suppressed when a double stranded RNA having a sequence identical or similar to the target gene sequence is introduced into the cell.
- Point to Examples of RNA used herein include double-stranded RNA that causes RNA interference of 21 to 25 bases in length, such as dsRNA (double strand RNA), siRNA (small interfering RNA), shRNA (short hairpin RNA), or miRNA. (microRNA).
- RNA can also be locally delivered to a desired site by a delivery system such as liposome, or can be locally expressed using a vector that produces the double stranded RNA.
- dsRNA, siRNA, shRNA or miRNA double-stranded RNA
- methods for using, etc. are known from many documents (Japanese Patent Application 2002-516062; US Publication No. 2002 / 086356A; Nature Genetics, 24 (2), 180-183, 2000 Feb. et al.).
- the polynucleotide contained in the vector of the present invention can be mediated by known internal ribosome entry site (IRES) sequences.
- ITR internal ribosome entry site
- the rAAV genome of the present invention is non-sc, it is possible to select a wider range of promoters and target genes, and it is also possible to use multiple target genes.
- the total length of the polynucleotide packaged in the rAAV vector of the present invention is preferably about 5 kb or less (about 4.7 kb or less excluding the ITR region).
- rAAV vector of the present invention for example, (a) first polynucleotide encoding a capsid protein (generally, AAV helper plasmid And (b) transfecting the cultured cells with a second polynucleotide (containing a therapeutic gene of interest) packaged within the rAAV vector of the present invention.
- the preparation method of the present invention further comprises (c) transfecting a cultured cell with a plasmid encoding an adenovirus-derived factor called an adenovirus (AdV) helper plasmid, or infecting the cultured cell with adenovirus.
- AdV adenovirus
- the method may further comprise the steps of culturing the above-mentioned transfected cultured cells, and collecting the recombinant adeno-associated virus vector from the culture supernatant.
- baculoviruses respectively containing the polynucleotides of the above (a) and (b) are prepared, and infected with insect cells such as Sf9 Also includes methods of production. Such methods are already known and are also utilized in the examples herein.
- the nucleotide encoding the capsid protein of the present invention in the first polynucleotide (a) is preferably operably linked to a known promoter sequence operable in cultured cells.
- a promoter sequence for example, cytomegalovirus (CMV) promoter, EF-1 ⁇ promoter, SV40 promoter and the like can be used appropriately.
- CMV cytomegalovirus
- EF-1 ⁇ promoter EF-1 ⁇ promoter
- SV40 promoter SV40 promoter
- known enhancer sequences, Kozak sequences, poly A addition signal sequences and the like may be suitably included.
- the second polynucleotide (b) contains a therapeutic gene at a position operable with a neural cell specific promoter. Furthermore, known enhancer sequences, Kozak sequences, poly A addition signal sequences and the like may be suitably included.
- This first polynucleotide may further comprise, downstream of the neural cell specific promoter sequence, a cloning site cleavable by various known restriction enzymes. More preferred is a multicloning site comprising multiple restriction enzyme recognition sites.
- Those skilled in the art can incorporate the therapeutic gene of interest downstream of the neural cell specific promoter according to known genetic engineering procedures. For such gene manipulation procedures, see, for example, Molecular Cloning (Molecular Cloning 3rd Edition, J. Sambrook et al., Cold Spring Harbor Lab. Press. 2001).
- a helper virus plasmid eg, adenovirus, herpes virus or vaccinia
- the preparation method of the present invention further comprises the step of introducing an adenovirus (AdV) helper plasmid.
- AdV adenovirus
- the AdV helper is derived from the same species of virus as the cultured cells.
- human AdV-derived helper virus vectors can be used.
- AdV helper vectors commercially available ones (for example, AAV Helper-Free System of Agilent Technologies (Catalog No. 240071)) can be used.
- rAAV vector of the present invention methods for transfecting the above-mentioned one or more types of plasmids into cultured cells can be carried out using various known methods such as calcium phosphate method, lipofection method, electroporation method, etc. it can. Such methods are described, for example, in Molecular Cloning 3rd Ed., Current Protocols in Molecular Biology, John Wiley & Sons 1987-1997.
- compositions Comprising the rAAV Vectors of the Present Invention
- the rAAV vectors of the present invention are useful for the treatment of neurological diseases, in particular, diseases associated with protein dysfunction at synapses (eg, schizophrenia, autism spectrum disorders, etc.) It can contain a gene.
- An rAAV vector containing these genes can be administered, for example, in a blood vessel, passed through the blood-brain barrier, and integrated into nerve cells of the brain, spinal cord, and retina.
- RAAV vectors containing such therapeutic genes are included in the pharmaceutical composition of the present invention.
- a therapeutic gene for example, a polynucleotide encoding an antibody as described above, neurotrophic factor (NGF), growth factor (HGF), acidic fiber growth factor (aFGF), miRNA or the like can be selected.
- NGF neurotrophic factor
- HGF growth factor
- aFGF acidic fiber growth factor
- miRNA miRNA
- the active ingredients of the pharmaceutical composition of the present invention may be formulated singly or in combination, but may be formulated with a pharmaceutically acceptable carrier or a pharmaceutical additive to be provided as a formulation. it can.
- the active ingredient of the present invention can be contained, for example, in an amount of 0.1 to 99.9% by weight in the preparation.
- Pharmaceutically acceptable carriers or additives include, for example, excipients, disintegrants, disintegrants, binders, lubricants, coatings, dyes, diluents, solubilizers, solubilizers, etc. Agents, pH adjusters, stabilizers and the like can be used.
- excipients such as microcrystalline cellulose, sodium citrate and calcium carbonate, starch, disintegrating agents such as alginic acid, granulating binders such as polyvinyl pyrrolidone, lubricants, etc. are commonly used in the relevant field. It can be used with an excipient.
- the active ingredient is used in combination with various sweeteners or flavoring agents, coloring agents or dyes, and, if necessary, also with emulsifying and / or suspending agents.
- various sweeteners or flavoring agents, coloring agents or dyes and, if necessary, also with emulsifying and / or suspending agents.
- preparations suitable for oral administration include, for example, powders, tablets, capsules, fine granules, granules, solutions or syrups and the like.
- preparations suitable for parenteral administration include injections, intrathecal fluid, suppositories and the like.
- parenteral administration a solution of the active ingredient of the present invention in either sesame oil or peanut oil or in an aqueous propylene glycol solution can be used.
- the aqueous solution should be suitably buffered (preferably at a pH of 8 or more) as necessary, and the liquid diluent first rendered isotonic.
- physiological saline can be used.
- the prepared aqueous solutions are suitable for intravenous injection, while the oily solutions are suitable for intraarticular, intramuscular and subcutaneous injection.
- the sterile preparation of all these solutions can be readily accomplished with standard pharmaceutical techniques well known to those skilled in the art.
- the active ingredient of the present invention can also be administered topically, such as skin. In this case, it is desirable to administer topically in the form of a cream, jelly, paste, ointment according to standard pharmaceutical practice.
- the dosage of the pharmaceutical composition of the present invention is not particularly limited, and an appropriate dosage is selected according to various conditions such as the type of disease, patient's age and symptoms, administration route, purpose of treatment, presence or absence of concomitant drug. It is possible.
- the dosage of the pharmaceutical composition of the present invention is, for example but not limited to, 1 to 5000 mg, preferably 10 to 1000 mg, per day for an adult (for example, 60 kg of body weight). These daily doses may be administered in two to four divided doses.
- vg vector genome
- the rAAVs of the invention are capable of crossing the blood-brain barrier of a living body (including the uncompleted fetal and neonatal blood-brain barriers, and the established adult blood-brain barrier). Peripheral administration to an adult and a fetus or a neonate allows gene delivery of the rAAV of the present invention to neural cells such as the brain and spinal cord. Furthermore, rAAV vectors used in the present invention can target neural cells contained in adult brain, spinal cord and the like by peripheral administration.
- peripheral administration includes intravenous administration, intraarterial administration, intraperitoneal administration, intracardiac administration, intramuscular administration, umbilical cord intravascular administration (for example, in the case of fetal administration), etc.
- administration methods fluidly connected to the brain such as intrathecal administration
- the rAAV vectors of the invention can be locally administered to a target site in the brain such as the hippocampus.
- a simpler means of administration can be provided as compared to intracerebral administration.
- kits for producing the rAAV of the present invention can include, for example, (a) a first polynucleotide for expressing a capsid protein VP1 or the like, and (b) a second polynucleotide packaged in a rAAV vector.
- the first polynucleotide comprises a polynucleotide encoding an amino acid of SEQ ID NO :.
- the second polynucleotide may or may not include the therapeutic gene of interest, but preferably may include various restriction enzyme cleavage sites for incorporating such therapeutic gene of interest. .
- kits for producing a rAAV vector of the invention can further comprise any of the configurations described herein (eg, AdV helpers, etc.).
- the kits of the invention may also further comprise instructions describing a protocol for making rAAV vectors using the kits of the invention.
- the rAAV vector according to the present invention can also be used in combination with existing chemotherapeutic agents.
- chemotherapeutic agents include phenytoin, carbamazepine, valproic acid, topiramate, lamotrigine, lufinamide, phenobarbital, diazepam, clonazepam, etosuccimide, zonisamide, gabapentin, levetiracetam, midazolam, clobazam, propofol.
- chemotherapeutic agents include phenytoin, carbamazepine, valproic acid, topiramate, lamotrigine, lufinamide, phenobarbital, diazepam, clonazepam, etosuccimide, zonisamide, gabapentin, levetiracetam, midazolam, clobazam, propofol.
- it can be expected to greatly reduce the dose of the
- the therapeutic effects of the rAAV vectors of the invention for the measurement of therapeutic effects are determined using known means to determine that arousal is suppressed.
- known means for example, analysis of behavioral level, analysis of pharmacokinetics of a labeled transmitter (such as GABA), measurement of excitatory postsynaptic potential or inhibitory postsynaptic potential, epilepsy induction by drugs or electrical stimulation Examples include, but are not limited to, measurement of threshold change, electroencephalogram, optical topography, positron emission tomography (PET), and the like. . 11.
- the meanings of the terms used in the present specification for the terms in the present specification are as follows. In the present specification, terms that are not particularly described are intended to indicate the range to which the term that is ordinarily understood by those skilled in the art means.
- viral vector As used herein, unless stated otherwise, the terms “viral vector”, “viral virion”, “viral particle” are used interchangeably.
- neural system refers to an organ system comprised of neural tissue.
- neural cells include at least nerve cells contained in the central nervous system such as the brain and spinal cord, and further include glial cells, microglia, astrocytes, It may also contain oligodendrocytes, ependymal cells, cerebrovascular endothelial cells and the like.
- nucleic acid As used herein, the term “polynucleotide” is used interchangeably with “nucleic acid”, “gene” or “nucleic acid molecule” and is intended to be a polymer of nucleotides.
- nucleotide sequence is used interchangeably with “nucleic acid sequence” or “base sequence” and is the sequence of deoxyribonucleotides (abbreviated A, G, C and T) It is shown as.
- a polynucleotide or a fragment thereof comprising the nucleotide sequence of SEQ ID NO: 1 means a polynucleotide or a fragment thereof comprising a sequence represented by each deoxynucleotide A, G, C and / or T of SEQ ID NO: 1 Be done.
- the "virus genome” and the “polynucleotide” according to the invention may each be present in the form of DNA (eg cDNA or genomic DNA) but may optionally be in the form of RNA (eg mRNA).
- the viral genomes and polynucleotides used herein may each be double stranded or single stranded DNA. In the case of single stranded DNA or RNA, it may be the coding strand (also known as the sense strand) or the non-coding strand (also known as the antisense strand).
- polypeptide As used herein, “protein” and “polypeptide” are used interchangeably and are intended to be polymers of amino acids.
- the polypeptide used in the present specification has the N-terminus (amino terminus) at the left end and the C-terminus (carboxy terminus) at the right end according to the peptide label convention.
- the partial peptide of the polypeptide of the present invention (hereinafter sometimes abbreviated as the partial peptide of the present invention) is a partial peptide of the above-mentioned polypeptide of the present invention, preferably, the above-mentioned poly of the present invention It has the same properties as the peptide.
- the term "plasmid” refers to various known genetic elements such as, for example, plasmids, phages, transposons, cosmids, chromosomes and the like. Plasmids can replicate in a particular host and transfer gene sequences between cells.
- the plasmid contains various known nucleotides (DNA, RNA, PNA and a mixture thereof), and may be single-stranded or double-stranded, but preferably double-stranded.
- the term "rAAV vector plasmid” is intended to include the duplex formed by the rAAV vector genome and its complement unless otherwise stated.
- the plasmids used in the present invention may be linear or circular.
- the term "packaging” refers to events including preparation of single-stranded viral genome, assembly of coat protein (capsid), and encapsidation of viral genome and the like.
- the appropriate plasmid vector usually multiple plasmids
- the recombinant viral particles ie, viral virions, viral vectors
- rAAV intravascular-administered adeno-associated virus
- rAAV Recombinant Adeno-Associated Virus
- the vector used in this example introduces the tyrosine mutation (Y446 ⁇ F) into the capsid of AAV9 released previously (AAV9 capsid, AVR3 ITR) ) Has the Synapsin I promoter (WO 2012/057363).
- rAAV vector expressing Neuroligin 2 with FLAG tag (DDDDK) sequence linked to N-terminus was prepared (AAV9 / 3-Syn1-FLAG (DDDDK) -NL2) , Was administered to the subject animals.
- mice (6 weeks old, male, 22-32 g body weight) were used.
- Seizure intensity 1 point no seizure 2 points: raise tail or shake body only 3 points: clear seizure but keep posture without falling 4 points: violent seizure, can not keep posture, falls sideways It scored as.
- Duration of seizure 1 point no seizure 2 points: 1-10 seconds 3 points: 11-20 seconds 4 points: 21-30 seconds 5 points: 31-60 seconds 6 points: 61 seconds- It scored as.
- the incidence rate, mean seizure time, mean seizure intensity, mean seizure time ⁇ intensity of each group were evaluated weekly
- Electrodes were attached to both ears at 5 (before vector administration), 12, 18 and 22 weeks of age, and the following parameters were used for electrical stimulation (Neuron SQ Neuropack S1: duration: 1 ms , interval: 50 ms, 10 train, strength: up to 50 mA every 5 mA to 5 mA) to induce epileptic seizure and measure the seizure threshold. If a seizure was not induced at 50 mA, the seizure threshold was evaluated as 60 mA. The topical administration group was performed at 5, 12 and 22 weeks of age only.
- ⁇ rAAV9-GFP expression and cell identification A 40 ⁇ m sagittal section was prepared with a freezing microtome, and GFP expression in each part of the brain was confirmed with a fluorescence microscope. For identification of cells expressing GFP, double staining with the following markers was performed. Neuronal cells; NeuN or MAP2, glial cells; GFAP, inhibitory interneurons; Parvalbumin
- ⁇ rAAV9-NL2 expression and cell identification As in the case of GFP confirmation, a 40 ⁇ m sagittal section was prepared, and the transgene NL2 expression was confirmed by FLAG (DDDDK) antibody staining.
- the FLAG antibody was purchased from Abcam Co., Ltd., and expression was confirmed by fluorescence microscopy by image processing using a secondary antibody with Alexafluor 488.
- Hippocampal slice preparations were used to measure changes in intracellular Ca 2+ influx at ischemic load by fluorescence change of rhod 2-AM (DOJINDO catalog number: R002). Intracellular Ca 2+ influx that occurs in the hippocampus due to ischemic load was significantly elevated in the CA3 region compared to DDY mice in the EL mice, suggesting vulnerability of the inhibitory system in the CA3 region.
- the expression in the EL hippocampus of the intercalated cells which are the excitation inhibitory system was examined histologically by immunostaining with a parvalbumin antibody (catalog number: LS-C39101).
- rAAV vector expressing a gene of a suppression system synapse related molecule is prepared, the vector is administered to EL by stereotactic hippocampal injection and intravascular injection, and histologically by staining using a FLAG antibody.
- the distribution in the brain was observed (Fig. 1a, 1b).
- FLAG-tagged NLGN2 was widely expressed in neurons of hippocampus and cerebral cortex by intravascular administration of the vector of the present invention, it could be confirmed that gene delivery by rAAV was good.
- the expression of the target gene was observed in hippocampal neurons in the hippocampus injection group and in all brain neurons including the hippocampus in the intravascular administration group (results not shown).
- the NLGN 2 administration group sometimes showed significant differences in seizure frequency, but there was no significant difference in the overall effects (FIGS. 7 to 11).
- significant differences were found in the portions marked with “*” and “**” in the figure.
- the intracardiac administration group tended to be more effective in suppressing epilepsy in all of the seizure frequency, seizure time and seizure intensity.
- the target molecule was delivered to neurons throughout the brain by the intravascular administration type AAV vector.
- the potential of non-invasive epilepsy gene therapy which is more useful as a therapeutic method, does not change the threshold of electrical stimulation while suppressing the epileptic seizure with the excitatory action of this molecule.
- rAAV vector of the present invention By using the rAAV vector of the present invention, it can be expected to treat (reduce, ameliorate, repair, etc.) genetic defects (including innate and acquired ones) in neural cells.
- SEQ ID NO: 1 human neuroligin 2 nucleotide sequence
- SEQ ID NO: 2 human neuroligin 2 amino acid sequence
- SEQ ID NO: 3 mouse neuroligin 2 nucleotide sequence
- SEQ ID NO: 4 mouse neuroligin 2 amino acid sequence
- SEQ ID NO 5 rat neuroligin 2 nucleotide sequence
- SEQ ID NO: 6 rat neuroligin 2 amino acid sequence
- SEQ ID NO: 7 nucleotide sequence of Flag tagged mouse neuroligin 2
- SEQ ID NO: 8 amino acid sequence of Flag tagged mouse neuroligin 2
- SEQ ID NO: 9 amino acid of AAV1 capsid protein Y445F variant
- SEQ ID NO: 10 Amino acid sequence of AAV2 capsid protein Y444F variant
- SEQ ID NO: 11 Amino acid sequence of AAV 9 capsid protein Y446F variant
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Engineering & Computer Science (AREA)
- Veterinary Medicine (AREA)
- Pharmacology & Pharmacy (AREA)
- Public Health (AREA)
- Animal Behavior & Ethology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Organic Chemistry (AREA)
- Epidemiology (AREA)
- Genetics & Genomics (AREA)
- Biomedical Technology (AREA)
- Molecular Biology (AREA)
- Zoology (AREA)
- Neurosurgery (AREA)
- Neurology (AREA)
- Biotechnology (AREA)
- Virology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Immunology (AREA)
- General Chemical & Material Sciences (AREA)
- Gastroenterology & Hepatology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Cell Biology (AREA)
- Microbiology (AREA)
- Biochemistry (AREA)
- Biophysics (AREA)
- General Engineering & Computer Science (AREA)
- Mycology (AREA)
- Wood Science & Technology (AREA)
- Pain & Pain Management (AREA)
- Toxicology (AREA)
- Hospice & Palliative Care (AREA)
- Psychiatry (AREA)
- Plant Pathology (AREA)
- Physics & Mathematics (AREA)
Abstract
Description
[1] 生体における抑制性シナプスの興奮抑制機能を向上させるタンパク質をコードするポリヌクレオチドを含む、てんかん、統合失調症、自閉症スペクトラム障害、精神遅滞、不安、躁鬱病、片頭痛、恐怖強迫症状、薬物依存症、アンジェルマン症候群、ジスキネジア、ジストニア、アルツハイマー病、発達障害(注意欠陥多動性障害、アスペルガー症候群)からなる群より選択される疾患の治療のための組換えアデノ随伴ウイルスベクター。
[2] 前記ポリヌクレオチドが、配列番号2、4もしくは6のアミノ酸配列、またはこれら配列と約90%以上の同一性を有してニューレキシンと結合するアミノ酸配列を含むニューロリギン2タンパク質をコードするヌクレオチド配列を含む、[1]に記載の組換えアデノ随伴ウイルスベクター。
[3] 前記疾患がてんかんである、[1]又は[2]に記載の組換えアデノ随伴ウイルスベクター。
[4] 前記組換えアデノ随伴ウイルスベクターが、野生型AAV1カプシドタンパク質のアミノ酸配列中445位のチロシンがフェニルアラニンに置換されている変異アミノ酸配列を有するタンパク質、野生型のAAV2カプシドタンパク質のアミノ酸配列中445位のチロシンがフェニルアラニンに置換されている変異アミノ酸配列を有するタンパク質、または野生型のAAV9カプシドタンパク質のアミノ酸配列中446位のチロシンがフェニルアラニンに置換されている変異アミノ酸配列を有するタンパク質を含む、[1]~[3]のいずれかに記載のアデノ随伴ウイルス組換えベクター。
[5] 前記ポリヌクレオチドが、シナプシンIプロモーター配列、ミエリン塩基性タンパク質プロモーター配列、ニューロン特異的エノラーゼプロモーター配列、カルシウム/カルモジュリンー依存性蛋白キナーゼII(CMKII)プロモーター配列、チュブリンαIプロモーター配列、血小板由来成長因子β鎖プロモーター配列、グリア線維性酸性タンパク質(GFAP)プロモーター配列、L7プロモーター配列(小脳プルキンエ細胞特異的プロモーター)、グリア線維酸性タンパク質(hGfa2)プロモーター配列、およびグルタミン酸受容体デルタ2プロモーター(小脳プルキンエ細胞特異的プロモーター)配列、グルタミン酸脱炭酸酵素(GAD65/GAD67)プロモーター配列からなる群より選択されるプロモーター配列を含む、[1]~[4]のいずれかに記載の組換えアデノ随伴ウイルスベクター。
[6] 前記ポリヌクレオチドが、AAV1、AAV2、AAV3、AAV4、AAV8、およびAAV9からなる群より選択されるインバーテッドターミナルリピート(ITR)を含む、[1]~[5]のいずれかに記載の組換えアデノ随伴ウイルスベクター。
[7] 前記ポリヌクレオチドが、興奮性シナプスの興奮を抑制するためのポリヌクレオチドをさらに含む、[1]~[6]のいずれかに記載の組換えアデノ随伴ウイルスベクター。
[8] [1]~[7]のいずれかに記載の組換えアデノ随伴ウイルス組換えベクターを含む、医薬組成物。
[9] 脳内投与されるための、[8]に記載の医薬組成物。
[10] 髄腔内投与されるための、[8]に記載の医薬組成物。
[11] 末梢投与されるための、[8]に記載の医薬組成物。
[12] 神経精神疾患に係る化学療法剤と併用するための、[8]~[11]のいずれかに記載の医薬組成物。
[13] 生体における抑制性シナプスの興奮抑制機能を向上させるタンパク質をコードするヌクレオチド配列を有するポリヌクレオチドを含む組換えアデノ随伴ウイルスベクターを生体に投与することを含む、てんかん、統合失調症、自閉症スペクトラム障害、精神遅滞、不安、躁鬱病、片頭痛、恐怖強迫症状、薬物依存症、アンジェルマン症候群、ジスキネジア、ジストニア、アルツハイマー病、発達障害(注意欠陥多動性障害、アスペルガー症候群)からなる群より選択される疾患の治療方法。
[14]化学療法と併用される、[13]に記載の治療方法
本出願において、シナプスとは、神経細胞の軸索終末等が膨らんだシナプス小頭部と、その標的のニューロンまたは筋細胞との接合部を指す。生体において、シナプスには、興奮を伝達する興奮性シナプスと、興奮の伝達を抑制する抑制性シナプスとが存在する。また、大部分のシナプスは、化学物質の伝達によって媒介される化学シナプス(信号伝達が遅い)である。もう1つの型は、時間的に速い応答を示す電気シナプスであるが、成熟哺乳類の中枢神経では余り見られない。
本願発明は、てんかん、統合失調症、自閉症スペクトラム障害、精神遅滞、不安、躁鬱病、片頭痛、恐怖強迫症状、薬物依存症、アンジェルマン症候群、ジスキネジア、ジストニア、アルツハイマー病、発達障害(注意欠陥多動性障害、アスペルガー症候群)からなる群より選択される疾患、とりわけてんかんの治療に有用であるrAAVベクターを提供するものである。
本願発明において、シナプスの機能制御に用いられる遺伝子を神経系細胞に送達するためのベクターとしては、WO 2012/057363に記載される、末梢投与によっても神経細胞に効率的に遺伝子送達できる組換えアデノ随伴ウイルスベクター(本明細書中、血管投与型ベクターともいう)、あるいはWO 2008/124724などに記載されるものを用いることができる。本発明のrAAVベクターは、生体の血液脳関門を通過可能であり、したがって血液脳関門を介して脳に送達される投与手段、例えば患者への末梢投与などによって、患者の脳、脊髄などの神経系細胞に目的の治療用遺伝子を導入可能である。また、髄腔内投与や脳内の標的部位に直接投与することも可能である。
抑制性シナプスの興奮抑制機能を向上させる他の手段または追加の手段として、例えば、ニューロリギン2の結合パートナーであるニューレキシン1αの発現を増強させる手段、ニューロリギン2の細胞内シグナル伝達を向上させる手段などが期待できる。あるいは、そのような他の手段または追加の手段として、興奮性シナプスの機能を低下させる手段、例えば興奮性シナプスの作動に関するタンパク質の発現を抑制すること、具体的にはニューロリギン1のアンチセンスによってニューロリギン1を低減させる手段(非特許文献4:Fangら、Mol. Neurobiol. 2014 Nov.)等もまた有用となり得る。
本発明のrAAVベクターを調製する方法としては一般的なものを利用することができ、例えば、(a)カプシドタンパク質をコードする第1のポリヌクレオチド(一般に、AAVヘルパープラスミドと称される)、および(b)本発明のrAAVベクター内にパッケージングされる第2のポリヌクレオチド(目的の治療用遺伝子を含む)を、培養細胞にトランスフェクトする工程を含むことができる。本発明における調製方法はさらに、(c)アデノウイルス(AdV)ヘルパープラスミドと称されるアデノウイルス由来因子をコードするプラスミドを培養細胞にトランフェクトする工程、またはアデノウイルスを培養細胞に感染させる工程も含むことができる。さらに、上記のトランスフェクトされた培養細胞を培養する工程、および培養上清より組換えアデノ随伴ウイルスベクターを収集する工程を含むこともできる。(d)さらに、本発明のrAAVベクターの調製方法は、上記(a)(b)のポリヌクレオチドをそれぞれ含むバキュロウイルスを作製し、昆虫細胞であるSf9などに感染させることにより、rAAVを大量に産生する方法も含む。このような方法は既に公知であり、本明細書の実施例においても利用される。
本発明のrAAVベクターは、神経疾患、とりわけシナプスにおけるタンパク質機能異常に係る疾患(例、統合失調症、自閉症スペクトラム障害など)などに対する治療に有用である遺伝子を含むことができる。それら遺伝子を含むrAAVベクターを、例えば血管内に投与し、血液脳関門を通過して脳、脊髄、網膜の神経細胞中に組込むことができる。このような治療用遺伝子を含むrAAVベクターは、本発明の医薬組成物に含められる。このような治療用遺伝子としては、例えば上記のような抗体、神経栄養因子(NGF)、成長因子(HGF)、酸性線維細胞増殖因子(aFGF)、miRNAなどをコードするポリヌクレオチドを選択できる。このようなrAAVベクターを被検体に末梢投与することによって、などの神経疾患を治療することが期待できる。
本発明のrAAVは、生体の血液脳関門(未完成な胎児及び新生児の血液脳関門、および確立した成体の血液脳関門を含む)を通過可能であるので、生体(成体および胎児または新生児を含む)に末梢投与することによって、本発明のrAAVを脳、脊髄などの神経系細胞に遺伝子の送達が可能である。さらに、本発明に用いるrAAVベクターは、末梢投与によって成体の脳、脊髄などに含まれる神経細胞を標的とすることができる。本明細書において末梢投与とは、静脈内投与、動脈内投与、腹腔内投与、心腔内投与、筋肉内投与、臍帯血管内投与(例えば、胎児を対象とする場合)など、当業者に末梢投与として通常理解される投与経路をいう。また、血液以外に脳と流体接続される投与方法、例えば髄腔内投与も、本発明のrAAVベクターに用いることができる。別の実施形態において、本発明のrAAVベクターを、海馬などの脳内の標的部位に局所投与することも可能である。例えば、本発明のrAAVを、髄腔内投与にて髄液内に、または末梢投与によって血中内に投与する場合、脳実質内投与と比較して、より平易な投与手段を提供できる。
本発明は別の実施形態において、本発明のrAAVを作製するためのキットを提供する。このようなキットは、例えば、(a)カプシドタンパク質VP1等を発現するための第1のポリヌクレオチド、および(b)rAAVベクター内にパッケージングされる第2のポリヌクレオチドを含むことができる。例えば、第1のポリヌクレオチドは、配列番号:のアミノ酸をコードするポリヌクレオチドを含む。例えば、第2のポリヌクレオチドは、目的の治療用遺伝子を含んでも含まなくてもよいが、好ましくは、そのような目的の治療用遺伝子を組込むための種々の制限酵素切断部位を含むことができる。
本願発明に係るrAAVベクターは、既存の化学療法剤と併用することもできる。そのような化学療法剤の例としては、フェニトイン、カルバマゼピン、バルプロ酸、トピラマート、ラモトリギン、ルフィナミド、フェノバルビタール、ジアゼパム、クロナゼパム、エトスクシミド、ゾニサミド、ガバペンチン、レベチラセタム、ミダゾラム、クロバザム、プロポフォールが挙げられる。例えば、本願発明のrAAVの投与後に、上記化学治療剤の投与量を大きく低減することが期待できる。
本発明のrAAVベクターの治療効果は、興奮が抑えられることを測定するための公知手段を用いて判断される。そのような公知手段としては、例えば、行動レベルの解析、標識した伝達物質(GABAなど)の薬理動態の解析、興奮性シナプス後電位や抑制性シナプス後電位の測定、薬剤または電気刺激よるてんかん誘発閾値変化の測定、脳波、光トポグラフィー、陽電子放出断層撮影(PET)などが挙げられるが、これらに限定されない。。
11.本明細書中の用語について
本明細書中に用いられる各用語が示す意味は以下のとおりである。本明細書中、特には説明されない用語については、当業者が通常理解する用語が意味する範囲を指すことが意図される。
生後6週時にrAAV-Neuroligin2(rAAV-NL2)を心腔内投与
・発作頻度・持続時間・強さ・持続時間×強さ・電気刺激に対する閾値の変化
ニューロリギン2搭載血管内投与型rAAVの脳内発現
○ 組換えアデノ随伴ウイルス(rAAV)ベクター
本実施例において用いるベクターは、先に公開されたAAV9/3 (AAV9のカプシドにチロシン変異(Y446→F)を導入し、AAV3のITRを持つ)にSynapsin I promoterを搭載しているものである(WO 2012/057363)。内因性のNeuroligin 2 (NL2)と区別するため、FLAG tag (DDDDK)配列がN末端に結合したニューロリギン2を発現するrAAVベクターを調製し(AAV9/3-Syn1-FLAG (DDDDK)-NL2)、これを対象動物に投与した。
ELマウス(6週齢、雄、体重22 - 32g)を使用した。
2-4%セボフルレン麻酔下にAAV9/3-Syn1-FLAG (DDDDK)-NL2を4.1×1013 vector genome/ml × 0.1ml /mouse心注した (NL2心注群n = 10)。AAV9/3-Syn1-AcGFP-WPREを2.3×1013 vg/ml × 0.1ml /mouse心注した群(n=17)と生理食塩水のみを0.1ml/mouse心注した群(n=14)を対照とした。また、局所投与と比較するため、両側海馬CA3領域(Bregmaより 0.5 mm後方、3.0 mm側方、脳表より2.0 mm)にAAV9/3-Syn1-FLAG (DDDDK)-NL 2を4.1×1013 vg/ml × 0.005 ml注入したグループを局所投与群(n = 3)とした。
ベクター等投与後、22週齢に至るまで1週間毎に、尻尾を持ってマウスを一定数回転させ(8回転)、行動をビデオ記録した。後にビデオ上で発作の有無、発作を起こした場合の持続時間、強度を観察した。
発作強度を
1点: 発作なし
2点: 尻尾を挙げる、または体を震わせるのみ
3点: 明らかな発作だが、倒れずに姿勢を保つ
4点: 激しい発作で、姿勢を保てず、横に倒れる
として得点化した。
発作の持続時間を
1点: 発作なし
2点: 1-10秒
3点: 11-20秒
4点: 21-30秒
5点: 31-60秒
6点: 61秒-
として得点化した。
各グループの発作出現率、平均発作時間、平均発作強度、平均発作時間×強度を週毎に評価した
5(ベクター投与前), 12, 18, 22週齢において、両耳に電極を装着し、電気刺激(NIHON KOHDEN社製のNeuropack S1において以下のパラメーターを用いた:duration:1ms, interval:50ms, 10train, strength:0mA~5mA毎にmax 50mAまで)を与え、てんかん発作を誘発し、発作閾値を測定した。50mAで発作が誘発されなかった場合は発作閾値を60mAとして評価した。局所投与群のみ5, 12, 22週齢で行った。
発作出現率:Fisher正確検定
その他:Welchのt検定
により評価した。
脳標本作成:ペントバルビタールで深麻酔し、左心室からの4%パラフォルムアルデヒドを含む0.1Mリン酸バッファー(pH7.4)注入し灌流固定した。固定後、脳を取り出して灌流固定液での半日の浸積固定後15%ショ糖を含む0.1Mリン酸バッファー(pH7.4)に移して冷蔵庫で組織化学実験まで保存した。
凍結ミクロトームで40μmの矢状断切片を作成し蛍光顕微鏡で脳各部位のGFP発現を確認した。GFPを発現している細胞の同定は以下のマーカーとの2重染色を行った。神経細胞;NeuNまたはMAP2、グリア細胞;GFAP、抑制性介在ニューロン;Parvalbumin
GFP確認と同様、40μmの矢状断切片を作成してFLAG(DDDDK)抗体染色により導入遺伝子NL2発現を確認した。FLAG抗体はアブカム株式会社より購入し、Alexafluor 488付き2次抗体を用いた画像処理により蛍光顕微鏡で発現を確認した。
海馬スライス標本を用いて虚血負荷時の細胞内Ca2+流入変化をrhod 2-AM(DOJINDOカタログ番号:R002 )の蛍光変化により測定した。虚血負荷により海馬で起きる細胞内Ca2+流入はELマウスがDDYマウスに比べてCA3 領域で有意な上昇を示し、CA3 領域における抑制系の脆弱性が示唆された。次に興奮抑制系である介在細胞のEL海馬における発現をparvalbumin抗体(カタログ番号:LS-C39101 )による免疫染色で組織学的に検討した。海馬各領域のparvalbumin陽性細胞数においてはELマウスとDDYマウスで有意差は認められず、シナプスレベルでの変化の可能性が示唆された。以上の結果に基づき、抑制系シナプス関連分子の遺伝子を発現するrAAVベクターを調製し、定位的海馬注入および血管内注入により同ベクターをELに投与し、FLAG抗体を用いる染色によって、組織学的に脳内分布を観察した(図1a、1b)。図1aに示すように、本発明のベクターの血管内投与によって海馬及び大脳皮質の神経細胞にFLAGタグ付NLGN2が広く発現していたため、rAAVによる遺伝子送達が良好であったことを確認できた。
配列番号2:ヒトニューロリギン2アミノ酸配列
配列番号3:マウスニューロリギン2ヌクレオチド配列
配列番号4:マウスニューロリギン2アミノ酸配列
配列番号5:ラットニューロリギン2ヌクレオチド配列
配列番号6:ラットニューロリギン2アミノ酸配列
配列番号7:Flagタグ付マウスニューロリギン2のヌクレオチド配列
配列番号8:Flagタグ付マウスニューロリギン2のアミノ酸配列
配列番号9:AAV1カプシドタンパク質Y445F変異体のアミノ酸配列
配列番号10:AAV2カプシドタンパク質Y444F変異体のアミノ酸配列
配列番号11:AAV9カプシドタンパク質Y446F変異体のアミノ酸配列
Claims (12)
- 生体における抑制性シナプスの興奮抑制機能を向上させるタンパク質をコードするポリヌクレオチドを含む、てんかん、統合失調症、自閉症スペクトラム障害、精神遅滞、不安、躁鬱病、片頭痛、恐怖強迫症状、薬物依存症、アンジェルマン症候群、ジスキネジア、ジストニア、アルツハイマー病、発達障害(注意欠陥多動性障害、アスペルガー症候群)からなる群より選択される疾患の治療のための組換えアデノ随伴ウイルスベクター。
- 前記ポリヌクレオチドが、配列番号2、4もしくは6のアミノ酸配列、またはこれら配列と約90%以上の同一性を有してニューレキシンと結合するアミノ酸配列を含むニューロリギン2タンパク質をコードするヌクレオチド配列を含む、請求項1に記載の組換えアデノ随伴ウイルスベクター。
- 前記疾患がてんかんである、請求項1又は2に記載の組換えアデノ随伴ウイルスベクター。
- 前記組換えアデノ随伴ウイルスベクターが、野生型AAV1カプシドタンパク質のアミノ酸配列中445位のチロシンがフェニルアラニンに置換されている変異アミノ酸配列を有するタンパク質、野生型のAAV2カプシドタンパク質のアミノ酸配列中445位のチロシンがフェニルアラニンに置換されている変異アミノ酸配列を有するタンパク質、または野生型のAAV9カプシドタンパク質のアミノ酸配列中446位のチロシンがフェニルアラニンに置換されている変異アミノ酸配列を有するタンパク質を含む、請求項1~3のいずれかに記載のアデノ随伴ウイルス組換えベクター。
- 前記ポリヌクレオチドが、シナプシンIプロモーター配列、ミエリン塩基性タンパク質プロモーター配列、ニューロン特異的エノラーゼプロモーター配列、カルシウム/カルモジュリンー依存性蛋白キナーゼII(CMKII)プロモーター配列、チュブリンαIプロモーター配列、血小板由来成長因子β鎖プロモーター配列、グリア線維性酸性タンパク質(GFAP)プロモーター配列、L7プロモーター配列(小脳プルキンエ細胞特異的プロモーター)、グリア線維酸性タンパク質(hGfa2)プロモーター配列、およびグルタミン酸受容体デルタ2プロモーター(小脳プルキンエ細胞特異的プロモーター)配列、グルタミン酸脱炭酸酵素(GAD65/GAD67)プロモーター配列からなる群より選択されるプロモーター配列を含む、請求項1~4のいずれかに記載の組換えアデノ随伴ウイルスベクター。
- 前記ポリヌクレオチドが、AAV1、AAV2、AAV3、AAV4、AAV8、およびAAV9からなる群より選択されるインバーテッドターミナルリピート(ITR)を含む、請求項1~5のいずれかに記載の組換えアデノ随伴ウイルスベクター。
- 前記ポリヌクレオチドが、興奮性シナプスの興奮を抑制するためのポリヌクレオチドをさらに含む、請求項1~6のいずれかに記載の組換えアデノ随伴ウイルスベクター。
- 請求項1~7のいずれかに記載の組換えアデノ随伴ウイルス組換えベクターを含む、医薬組成物。
- 脳内投与されるための、請求項8に記載の医薬組成物。
- 髄腔内投与されるための、請求項8に記載の医薬組成物。
- 末梢投与されるための、請求項8に記載の医薬組成物。
- 神経精神疾患に係る化学療法剤と併用するための、請求項8~11のいずれかに記載の医薬組成物。
Priority Applications (5)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
ES17738541T ES2871527T3 (es) | 2016-01-15 | 2017-01-13 | Virión de virus adenoasociado para usar en el tratamiento de la epilepsia |
EP17738541.6A EP3403675B1 (en) | 2016-01-15 | 2017-01-13 | Adeno-associated virus virion for use in treatment of epilepsy |
US16/069,370 US20190022251A1 (en) | 2016-01-15 | 2017-01-13 | Adeno-Associated Virus Virions for Treatment of Epilepsy |
JP2017561186A JP6924441B2 (ja) | 2016-01-15 | 2017-01-13 | てんかん治療のためのアデノ随伴ウイルスビリオン |
US17/732,901 US20220275399A1 (en) | 2016-01-15 | 2022-04-29 | Adeno-Associated Virus Virions for Treatment of Epilepsy |
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
JP2016-006191 | 2016-01-15 | ||
JP2016006191 | 2016-01-15 |
Related Child Applications (2)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US16/069,370 A-371-Of-International US20190022251A1 (en) | 2016-01-15 | 2017-01-13 | Adeno-Associated Virus Virions for Treatment of Epilepsy |
US17/732,901 Division US20220275399A1 (en) | 2016-01-15 | 2022-04-29 | Adeno-Associated Virus Virions for Treatment of Epilepsy |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2017122789A1 true WO2017122789A1 (ja) | 2017-07-20 |
Family
ID=59311265
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/JP2017/001048 WO2017122789A1 (ja) | 2016-01-15 | 2017-01-13 | てんかん治療のためのアデノ随伴ウイルスビリオン |
Country Status (5)
Country | Link |
---|---|
US (2) | US20190022251A1 (ja) |
EP (1) | EP3403675B1 (ja) |
JP (1) | JP6924441B2 (ja) |
ES (1) | ES2871527T3 (ja) |
WO (1) | WO2017122789A1 (ja) |
Cited By (7)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US10335466B2 (en) | 2014-11-05 | 2019-07-02 | Voyager Therapeutics, Inc. | AADC polynucleotides for the treatment of parkinson's disease |
US10570395B2 (en) | 2014-11-14 | 2020-02-25 | Voyager Therapeutics, Inc. | Modulatory polynucleotides |
US10584337B2 (en) | 2016-05-18 | 2020-03-10 | Voyager Therapeutics, Inc. | Modulatory polynucleotides |
US10597660B2 (en) | 2014-11-14 | 2020-03-24 | Voyager Therapeutics, Inc. | Compositions and methods of treating amyotrophic lateral sclerosis (ALS) |
US11603542B2 (en) | 2017-05-05 | 2023-03-14 | Voyager Therapeutics, Inc. | Compositions and methods of treating amyotrophic lateral sclerosis (ALS) |
US11697825B2 (en) | 2014-12-12 | 2023-07-11 | Voyager Therapeutics, Inc. | Compositions and methods for the production of scAAV |
US11759506B2 (en) | 2017-06-15 | 2023-09-19 | Voyager Therapeutics, Inc. | AADC polynucleotides for the treatment of Parkinson's disease |
Families Citing this family (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
KR102546560B1 (ko) * | 2020-08-12 | 2023-06-21 | 재단법인대구경북과학기술원 | Slitrk3 및 Nlgn2 억제제를 포함하는 불안장애 예방 또는 치료용 약학적 조성물 |
WO2022232694A1 (en) * | 2021-04-30 | 2022-11-03 | The United States Government As Represented By The Department Of Veterans Affairs | Ptprd inhibitors and uses thereof |
Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2008016629A2 (en) * | 2006-08-01 | 2008-02-07 | Neurologix, Inc. | Novel glutamic acid decarboxylase (gad) proteins and methods of use |
WO2010037143A1 (en) * | 2008-09-29 | 2010-04-01 | The University Of Montana | Vectors and methods of treating brain seizures |
WO2012057363A1 (ja) * | 2010-10-27 | 2012-05-03 | 学校法人自治医科大学 | 神経系細胞への遺伝子導入のためのアデノ随伴ウイルスビリオン |
Family Cites Families (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
AU2014244167A1 (en) * | 2013-03-13 | 2015-10-08 | The Children's Hospital Of Philadelphia | Adeno-associated virus vectors and methods of use thereof |
-
2017
- 2017-01-13 JP JP2017561186A patent/JP6924441B2/ja active Active
- 2017-01-13 EP EP17738541.6A patent/EP3403675B1/en active Active
- 2017-01-13 ES ES17738541T patent/ES2871527T3/es active Active
- 2017-01-13 US US16/069,370 patent/US20190022251A1/en not_active Abandoned
- 2017-01-13 WO PCT/JP2017/001048 patent/WO2017122789A1/ja active Application Filing
-
2022
- 2022-04-29 US US17/732,901 patent/US20220275399A1/en active Pending
Patent Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2008016629A2 (en) * | 2006-08-01 | 2008-02-07 | Neurologix, Inc. | Novel glutamic acid decarboxylase (gad) proteins and methods of use |
WO2010037143A1 (en) * | 2008-09-29 | 2010-04-01 | The University Of Montana | Vectors and methods of treating brain seizures |
WO2012057363A1 (ja) * | 2010-10-27 | 2012-05-03 | 学校法人自治医科大学 | 神経系細胞への遺伝子導入のためのアデノ随伴ウイルスビリオン |
Non-Patent Citations (8)
Title |
---|
FANG, M. ET AL.: "Neuroligin-1 Knockdown Suppresses Seizure Activity by Regulating Neuronal Hyperexcitability", MOL. NEUROBIOL., vol. 53, no. 1, 2014, pages 270 - 284, XP036233661 * |
KEIJI OGURO ET AL.: "EL Mouse ni Okeru Kekkannai Toyogata Adeno Zuihan Virus (AAV) Vector ni yoru Tenkan no Idenshi Chiryo", TENKAN KENKYU, vol. 33, no. 2, 2015, pages 521 (297), XP009512132, ISSN: 0912-0890 * |
KOHL, C. ET AL.: "Hippocampal Neuroligin-2 Overexpression Leads to Reduced Aggression and Inhibited Novelty Reactivity in Rats", PLOS ONE, vol. 8, no. 2, February 2013 (2013-02-01), pages 1 - 10, XP055400186 * |
KULLMANN, D. M. ET AL.: "Gene therapy in epilepsy--is it time for clinical trials?", NATURE REVIEWS NEUROLOGY, vol. 10, no. 5, May 2014 (2014-05-01), pages 300 - 304, XP055400192 * |
NOE, F. M. ET AL.: "Gene therapy of focal onset epilepsy using adeno-asociated virus vector- mediated overexpression of neuropeptide Y", JASPER'S BASIC MECHANISMS OF THE EPILEPSIES, 2012, Bethesda, pages 1 - 17, XP055535180, Retrieved from the Internet <URL:https://www.ncbi.nlm.nih.gov/ books/NBK98184/pdf/BookshelfNBN98184.pdf> [retrieved on 20170210] * |
OGURO, K. ET AL.: "EL Mouse ni Okeru Kekkannai Toyogata Virus Vector ni yoru Tenkan no Idenshi Chiryo", TENKAN KENKYU = JOURNAL OF THE JAPAN EPILEPSY SOCIETY, vol. 34, no. 2, September 2016 (2016-09-01), pages 442 (324), XP009512133, ISSN: 0912-0890 * |
RAOL, Y. H. ET AL.: "Enhancing GABA(A) Receptor alpha 1 Subunit Levels in Hippocampal Dentate Gyrus Inhibits Epilepsy Development in an Animal Model of Temporal Lobe Epilepsy", J. NEUROSCI., vol. 26, no. 44, 2006, pages 11342 - 11346, XP055400188 * |
See also references of EP3403675A4 * |
Cited By (16)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US10335466B2 (en) | 2014-11-05 | 2019-07-02 | Voyager Therapeutics, Inc. | AADC polynucleotides for the treatment of parkinson's disease |
US11975056B2 (en) | 2014-11-05 | 2024-05-07 | Voyager Therapeutics, Inc. | AADC polynucleotides for the treatment of Parkinson's disease |
US11027000B2 (en) | 2014-11-05 | 2021-06-08 | Voyager Therapeutics, Inc. | AADC polynucleotides for the treatment of Parkinson's disease |
US10920227B2 (en) | 2014-11-14 | 2021-02-16 | Voyager Therapeutics, Inc. | Compositions and methods of treating amyotrophic lateral sclerosis (ALS) |
US10597660B2 (en) | 2014-11-14 | 2020-03-24 | Voyager Therapeutics, Inc. | Compositions and methods of treating amyotrophic lateral sclerosis (ALS) |
US11198873B2 (en) | 2014-11-14 | 2021-12-14 | Voyager Therapeutics, Inc. | Modulatory polynucleotides |
US11542506B2 (en) | 2014-11-14 | 2023-01-03 | Voyager Therapeutics, Inc. | Compositions and methods of treating amyotrophic lateral sclerosis (ALS) |
US10570395B2 (en) | 2014-11-14 | 2020-02-25 | Voyager Therapeutics, Inc. | Modulatory polynucleotides |
US12071625B2 (en) | 2014-11-14 | 2024-08-27 | Voyager Therapeutics, Inc. | Modulatory polynucleotides |
US12123002B2 (en) | 2014-11-14 | 2024-10-22 | Voyager Therapeutics, Inc. | Compositions and methods of treating amyotrophic lateral sclerosis (ALS) |
US11697825B2 (en) | 2014-12-12 | 2023-07-11 | Voyager Therapeutics, Inc. | Compositions and methods for the production of scAAV |
US10584337B2 (en) | 2016-05-18 | 2020-03-10 | Voyager Therapeutics, Inc. | Modulatory polynucleotides |
US11193129B2 (en) | 2016-05-18 | 2021-12-07 | Voyager Therapeutics, Inc. | Modulatory polynucleotides |
US12084659B2 (en) | 2016-05-18 | 2024-09-10 | Voyager Therapeutics, Inc. | Modulatory polynucleotides |
US11603542B2 (en) | 2017-05-05 | 2023-03-14 | Voyager Therapeutics, Inc. | Compositions and methods of treating amyotrophic lateral sclerosis (ALS) |
US11759506B2 (en) | 2017-06-15 | 2023-09-19 | Voyager Therapeutics, Inc. | AADC polynucleotides for the treatment of Parkinson's disease |
Also Published As
Publication number | Publication date |
---|---|
JPWO2017122789A1 (ja) | 2018-11-29 |
US20190022251A1 (en) | 2019-01-24 |
EP3403675A4 (en) | 2019-09-18 |
EP3403675A1 (en) | 2018-11-21 |
ES2871527T3 (es) | 2021-10-29 |
US20220275399A1 (en) | 2022-09-01 |
EP3403675B1 (en) | 2021-04-21 |
JP6924441B2 (ja) | 2021-08-25 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
WO2017122789A1 (ja) | てんかん治療のためのアデノ随伴ウイルスビリオン | |
US11674156B2 (en) | Adeno-associated virus virion for gene transfer to nervous system cells | |
CN108699565B (zh) | 用于定向腺相关病毒(aav)的靶向肽 | |
AU2018261003A1 (en) | Compositions and methods of treating Huntington's Disease | |
JP2022088656A (ja) | 眼疾患のための遺伝子療法 | |
US20220195457A1 (en) | Interneuron-specific therapeutics for normalizing neuronal cell excitability and treating dravet syndrome | |
US11680276B2 (en) | Compositions and methods for treating retinal disorders | |
JP7334996B2 (ja) | グルコーストランスポーター1発現用アデノ随伴ウイルスベクター | |
KR20230123925A (ko) | Neurod1 및 dlx2 벡터 | |
WO2019146745A1 (ja) | テイ-サックス病及びザンドホッフ病治療用の新規アデノ随伴ウイルスビリオン | |
JP2020533313A (ja) | 神経障害性疼痛を処置するための方法および組成物 | |
US20240067989A1 (en) | Compositions and Methods for Treating Retinal Disorders | |
KR20240128717A (ko) | 유리체내 투여용 기능성 aav 캡시드 | |
EA046019B1 (ru) | Композиции и способы лечения нарушений сетчатки |
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: 17738541 Country of ref document: EP Kind code of ref document: A1 |
|
ENP | Entry into the national phase |
Ref document number: 2017561186 Country of ref document: JP Kind code of ref document: A |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2017738541 Country of ref document: EP |
|
ENP | Entry into the national phase |
Ref document number: 2017738541 Country of ref document: EP Effective date: 20180816 |