WO2023004365A1 - Vector constructs for delivery of nucleic acids encoding therapeutic proteasome activator complex subunits and methods of using the same - Google Patents
Vector constructs for delivery of nucleic acids encoding therapeutic proteasome activator complex subunits and methods of using the same Download PDFInfo
- Publication number
- WO2023004365A1 WO2023004365A1 PCT/US2022/073965 US2022073965W WO2023004365A1 WO 2023004365 A1 WO2023004365 A1 WO 2023004365A1 US 2022073965 W US2022073965 W US 2022073965W WO 2023004365 A1 WO2023004365 A1 WO 2023004365A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- promoter
- construct
- aspects
- vector
- nucleic acid
- Prior art date
Links
- 150000007523 nucleic acids Chemical class 0.000 title claims abstract description 252
- 102000039446 nucleic acids Human genes 0.000 title claims abstract description 203
- 108020004707 nucleic acids Proteins 0.000 title claims abstract description 203
- 239000013598 vector Substances 0.000 title claims abstract description 138
- 238000000034 method Methods 0.000 title claims abstract description 72
- 108090000708 Proteasome Endopeptidase Complex Proteins 0.000 title abstract description 30
- 102000004245 Proteasome Endopeptidase Complex Human genes 0.000 title abstract description 30
- 230000001225 therapeutic effect Effects 0.000 title description 13
- 239000012190 activator Substances 0.000 title description 2
- 238000001415 gene therapy Methods 0.000 claims abstract description 100
- 239000013603 viral vector Substances 0.000 claims abstract description 53
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims abstract description 52
- 230000000694 effects Effects 0.000 claims abstract description 42
- 201000010099 disease Diseases 0.000 claims abstract description 33
- 230000014509 gene expression Effects 0.000 claims description 364
- 101710103872 Proteasome activator complex subunit 1 Proteins 0.000 claims description 280
- 102100031300 Proteasome activator complex subunit 1 Human genes 0.000 claims description 276
- 101710103684 Proteasome activator complex subunit 2 Proteins 0.000 claims description 228
- 102100031299 Proteasome activator complex subunit 2 Human genes 0.000 claims description 226
- 210000004027 cell Anatomy 0.000 claims description 119
- 239000002245 particle Substances 0.000 claims description 113
- 102000040430 polynucleotide Human genes 0.000 claims description 97
- 108091033319 polynucleotide Proteins 0.000 claims description 97
- 239000002157 polynucleotide Substances 0.000 claims description 97
- 239000013612 plasmid Substances 0.000 claims description 74
- 239000013607 AAV vector Substances 0.000 claims description 65
- 230000003612 virological effect Effects 0.000 claims description 65
- 230000008488 polyadenylation Effects 0.000 claims description 58
- 108091028043 Nucleic acid sequence Proteins 0.000 claims description 57
- 210000001519 tissue Anatomy 0.000 claims description 45
- 230000001105 regulatory effect Effects 0.000 claims description 39
- 239000008194 pharmaceutical composition Substances 0.000 claims description 34
- 101000575685 Homo sapiens Synembryn-B Proteins 0.000 claims description 33
- 102100026014 Synembryn-B Human genes 0.000 claims description 33
- 108090000765 processed proteins & peptides Proteins 0.000 claims description 33
- 108091023045 Untranslated Region Proteins 0.000 claims description 32
- 230000001124 posttranscriptional effect Effects 0.000 claims description 30
- 241001492404 Woodchuck hepatitis virus Species 0.000 claims description 28
- 241000282414 Homo sapiens Species 0.000 claims description 27
- 230000027455 binding Effects 0.000 claims description 24
- 150000001413 amino acids Chemical group 0.000 claims description 20
- 102100021519 Hemoglobin subunit beta Human genes 0.000 claims description 19
- 108091005904 Hemoglobin subunit beta Proteins 0.000 claims description 19
- 102000001435 Synapsin Human genes 0.000 claims description 19
- 108050009621 Synapsin Proteins 0.000 claims description 19
- 108010006025 bovine growth hormone Proteins 0.000 claims description 19
- 108700011259 MicroRNAs Proteins 0.000 claims description 18
- 241000702623 Minute virus of mice Species 0.000 claims description 18
- 108091036066 Three prime untranslated region Proteins 0.000 claims description 18
- 108091000117 Tyrosine 3-Monooxygenase Proteins 0.000 claims description 18
- 102000048218 Tyrosine 3-monooxygenases Human genes 0.000 claims description 18
- 239000002679 microRNA Substances 0.000 claims description 18
- 101000611338 Homo sapiens Rhodopsin Proteins 0.000 claims description 16
- 241000702423 Adeno-associated virus - 2 Species 0.000 claims description 14
- 241001164825 Adeno-associated virus - 8 Species 0.000 claims description 13
- 208000023105 Huntington disease Diseases 0.000 claims description 13
- 208000018737 Parkinson disease Diseases 0.000 claims description 13
- 208000009415 Spinocerebellar Ataxias Diseases 0.000 claims description 12
- 102000044159 Ubiquitin Human genes 0.000 claims description 12
- 108090000848 Ubiquitin Proteins 0.000 claims description 12
- 210000000234 capsid Anatomy 0.000 claims description 12
- 230000002457 bidirectional effect Effects 0.000 claims description 11
- 230000001965 increasing effect Effects 0.000 claims description 11
- 108010000521 Human Growth Hormone Proteins 0.000 claims description 10
- 102000002265 Human Growth Hormone Human genes 0.000 claims description 10
- 239000000854 Human Growth Hormone Substances 0.000 claims description 10
- 101100412856 Mus musculus Rhod gene Proteins 0.000 claims description 10
- 108010029485 Protein Isoforms Proteins 0.000 claims description 10
- 102000001708 Protein Isoforms Human genes 0.000 claims description 10
- 208000007014 Retinitis pigmentosa Diseases 0.000 claims description 10
- 102100023742 Rhodopsin kinase GRK1 Human genes 0.000 claims description 10
- 101100242191 Tetraodon nigroviridis rho gene Proteins 0.000 claims description 10
- 108050003620 Arrestin-C Proteins 0.000 claims description 9
- 102100026440 Arrestin-C Human genes 0.000 claims description 9
- 102100026031 Beta-glucuronidase Human genes 0.000 claims description 9
- 108010044266 Dopamine Plasma Membrane Transport Proteins Proteins 0.000 claims description 9
- 108090001126 Furin Proteins 0.000 claims description 9
- 101000933465 Homo sapiens Beta-glucuronidase Proteins 0.000 claims description 9
- 101000629029 Homo sapiens Myosin regulatory light chain 2, ventricular/cardiac muscle isoform Proteins 0.000 claims description 9
- 101000829506 Homo sapiens Rhodopsin kinase GRK1 Proteins 0.000 claims description 9
- 102100026925 Myosin regulatory light chain 2, ventricular/cardiac muscle isoform Human genes 0.000 claims description 9
- 102100033928 Sodium-dependent dopamine transporter Human genes 0.000 claims description 9
- 208000024827 Alzheimer disease Diseases 0.000 claims description 7
- 206010002026 amyotrophic lateral sclerosis Diseases 0.000 claims description 7
- NCYCYZXNIZJOKI-IOUUIBBYSA-N 11-cis-retinal Chemical compound O=C/C=C(\C)/C=C\C=C(/C)\C=C\C1=C(C)CCCC1(C)C NCYCYZXNIZJOKI-IOUUIBBYSA-N 0.000 claims description 6
- 241001655883 Adeno-associated virus - 1 Species 0.000 claims description 6
- 241000202702 Adeno-associated virus - 3 Species 0.000 claims description 6
- 241000580270 Adeno-associated virus - 4 Species 0.000 claims description 6
- 241001634120 Adeno-associated virus - 5 Species 0.000 claims description 6
- 241000972680 Adeno-associated virus - 6 Species 0.000 claims description 6
- 241001164823 Adeno-associated virus - 7 Species 0.000 claims description 6
- 241000649045 Adeno-associated virus 10 Species 0.000 claims description 6
- 208000005145 Cerebral amyloid angiopathy Diseases 0.000 claims description 6
- 206010067889 Dementia with Lewy bodies Diseases 0.000 claims description 6
- 201000011240 Frontotemporal dementia Diseases 0.000 claims description 6
- 208000027747 Kennedy disease Diseases 0.000 claims description 6
- 102000004330 Rhodopsin Human genes 0.000 claims description 6
- 108090000820 Rhodopsin Proteins 0.000 claims description 6
- 208000006269 X-Linked Bulbo-Spinal Atrophy Diseases 0.000 claims description 6
- FWMNVWWHGCHHJJ-SKKKGAJSSA-N 4-amino-1-[(2r)-6-amino-2-[[(2r)-2-[[(2r)-2-[[(2r)-2-amino-3-phenylpropanoyl]amino]-3-phenylpropanoyl]amino]-4-methylpentanoyl]amino]hexanoyl]piperidine-4-carboxylic acid Chemical compound C([C@H](C(=O)N[C@H](CC(C)C)C(=O)N[C@H](CCCCN)C(=O)N1CCC(N)(CC1)C(O)=O)NC(=O)[C@H](N)CC=1C=CC=CC=1)C1=CC=CC=C1 FWMNVWWHGCHHJJ-SKKKGAJSSA-N 0.000 claims description 4
- 208000001089 Multiple system atrophy Diseases 0.000 claims description 4
- 208000024777 Prion disease Diseases 0.000 claims description 4
- 208000018282 ACys amyloidosis Diseases 0.000 claims description 3
- 208000011403 Alexander disease Diseases 0.000 claims description 3
- 102000007371 Ataxin-3 Human genes 0.000 claims description 3
- 108010032947 Ataxin-3 Proteins 0.000 claims description 3
- 102000007368 Ataxin-7 Human genes 0.000 claims description 3
- 108010032953 Ataxin-7 Proteins 0.000 claims description 3
- 206010068597 Bulbospinal muscular atrophy congenital Diseases 0.000 claims description 3
- 208000010200 Cockayne syndrome Diseases 0.000 claims description 3
- 208000011990 Corticobasal Degeneration Diseases 0.000 claims description 3
- 208000010859 Creutzfeldt-Jakob disease Diseases 0.000 claims description 3
- 201000008163 Dentatorubral pallidoluysian atrophy Diseases 0.000 claims description 3
- 208000007487 Familial Cerebral Amyloid Angiopathy Diseases 0.000 claims description 3
- 208000002339 Frontotemporal Lobar Degeneration Diseases 0.000 claims description 3
- 208000032849 Hereditary cerebral hemorrhage with amyloidosis Diseases 0.000 claims description 3
- 101000891654 Homo sapiens TATA-box-binding protein Proteins 0.000 claims description 3
- 208000009829 Lewy Body Disease Diseases 0.000 claims description 3
- 201000002832 Lewy body dementia Diseases 0.000 claims description 3
- 208000002569 Machado-Joseph Disease Diseases 0.000 claims description 3
- 208000000609 Pick Disease of the Brain Diseases 0.000 claims description 3
- 208000033063 Progressive myoclonic epilepsy Diseases 0.000 claims description 3
- 208000013738 Sleep Initiation and Maintenance disease Diseases 0.000 claims description 3
- 208000032859 Synucleinopathies Diseases 0.000 claims description 3
- 102100040296 TATA-box-binding protein Human genes 0.000 claims description 3
- 208000034799 Tauopathies Diseases 0.000 claims description 3
- 208000018756 Variant Creutzfeldt-Jakob disease Diseases 0.000 claims description 3
- 208000005881 bovine spongiform encephalopathy Diseases 0.000 claims description 3
- 208000017580 chronic wasting disease Diseases 0.000 claims description 3
- 206010022437 insomnia Diseases 0.000 claims description 3
- 206010023497 kuru Diseases 0.000 claims description 3
- 210000004558 lewy body Anatomy 0.000 claims description 3
- 201000007601 neurodegeneration with brain iron accumulation Diseases 0.000 claims description 3
- 208000021629 neuronal intranuclear inclusion disease Diseases 0.000 claims description 3
- 201000002212 progressive supranuclear palsy Diseases 0.000 claims description 3
- 230000001177 retroviral effect Effects 0.000 claims description 3
- 208000008864 scrapie Diseases 0.000 claims description 3
- 208000002320 spinal muscular atrophy Diseases 0.000 claims description 3
- 201000003570 spinocerebellar ataxia type 17 Diseases 0.000 claims description 3
- 208000011580 syndromic disease Diseases 0.000 claims description 3
- 102100035233 Furin Human genes 0.000 claims 2
- 210000004958 brain cell Anatomy 0.000 claims 1
- 210000004413 cardiac myocyte Anatomy 0.000 claims 1
- 239000000203 mixture Substances 0.000 abstract description 36
- 108090000623 proteins and genes Proteins 0.000 description 83
- 102000004169 proteins and genes Human genes 0.000 description 53
- 241000701022 Cytomegalovirus Species 0.000 description 36
- 238000003776 cleavage reaction Methods 0.000 description 34
- 241000699670 Mus sp. Species 0.000 description 30
- 230000007017 scission Effects 0.000 description 28
- 238000004519 manufacturing process Methods 0.000 description 26
- 230000009089 cytolysis Effects 0.000 description 25
- 210000001508 eye Anatomy 0.000 description 25
- 230000006870 function Effects 0.000 description 25
- 108700026220 vif Genes Proteins 0.000 description 25
- 108020004414 DNA Proteins 0.000 description 22
- 239000013604 expression vector Substances 0.000 description 22
- 230000006337 proteolytic cleavage Effects 0.000 description 21
- 208000035475 disorder Diseases 0.000 description 19
- 101800001494 Protease 2A Proteins 0.000 description 18
- 101800001066 Protein 2A Proteins 0.000 description 18
- 238000001727 in vivo Methods 0.000 description 18
- 230000010076 replication Effects 0.000 description 18
- 239000003795 chemical substances by application Substances 0.000 description 17
- 239000003623 enhancer Substances 0.000 description 17
- 108091026890 Coding region Proteins 0.000 description 16
- 238000002347 injection Methods 0.000 description 16
- 239000007924 injection Substances 0.000 description 16
- 238000011282 treatment Methods 0.000 description 16
- 108090000565 Capsid Proteins Proteins 0.000 description 15
- 102100023321 Ceruloplasmin Human genes 0.000 description 15
- 210000004556 brain Anatomy 0.000 description 15
- 230000029812 viral genome replication Effects 0.000 description 15
- 241000700605 Viruses Species 0.000 description 14
- 230000004807 localization Effects 0.000 description 14
- 102000004196 processed proteins & peptides Human genes 0.000 description 13
- 230000022532 regulation of transcription, DNA-dependent Effects 0.000 description 13
- 108700019146 Transgenes Proteins 0.000 description 12
- 229920001184 polypeptide Polymers 0.000 description 12
- 238000001262 western blot Methods 0.000 description 11
- 239000002773 nucleotide Substances 0.000 description 10
- 210000003523 substantia nigra Anatomy 0.000 description 10
- 238000013518 transcription Methods 0.000 description 10
- 230000035897 transcription Effects 0.000 description 10
- 241000701447 unidentified baculovirus Species 0.000 description 10
- 241000702421 Dependoparvovirus Species 0.000 description 9
- 239000006166 lysate Substances 0.000 description 9
- 125000003729 nucleotide group Chemical group 0.000 description 9
- 239000000047 product Substances 0.000 description 9
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 8
- 230000006037 cell lysis Effects 0.000 description 8
- 230000002068 genetic effect Effects 0.000 description 8
- 230000001939 inductive effect Effects 0.000 description 8
- 208000015181 infectious disease Diseases 0.000 description 8
- 210000004962 mammalian cell Anatomy 0.000 description 8
- 108020004999 messenger RNA Proteins 0.000 description 8
- 210000004165 myocardium Anatomy 0.000 description 8
- 238000012360 testing method Methods 0.000 description 8
- 102000004961 Furin Human genes 0.000 description 7
- 101000705759 Homo sapiens Proteasome activator complex subunit 2 Proteins 0.000 description 7
- 101000821100 Homo sapiens Synapsin-1 Proteins 0.000 description 7
- 108091000080 Phosphotransferase Proteins 0.000 description 7
- 102100021905 Synapsin-1 Human genes 0.000 description 7
- 238000010172 mouse model Methods 0.000 description 7
- 102000020233 phosphotransferase Human genes 0.000 description 7
- 239000000126 substance Substances 0.000 description 7
- 241000238631 Hexapoda Species 0.000 description 6
- 208000022873 Ocular disease Diseases 0.000 description 6
- 108010076504 Protein Sorting Signals Proteins 0.000 description 6
- 230000015572 biosynthetic process Effects 0.000 description 6
- 238000004113 cell culture Methods 0.000 description 6
- 239000000243 solution Substances 0.000 description 6
- 108020005345 3' Untranslated Regions Proteins 0.000 description 5
- 108020003589 5' Untranslated Regions Proteins 0.000 description 5
- 241001465754 Metazoa Species 0.000 description 5
- 241000699666 Mus <mouse, genus> Species 0.000 description 5
- 230000009286 beneficial effect Effects 0.000 description 5
- 210000005013 brain tissue Anatomy 0.000 description 5
- 239000013592 cell lysate Substances 0.000 description 5
- 210000000349 chromosome Anatomy 0.000 description 5
- 210000005064 dopaminergic neuron Anatomy 0.000 description 5
- 239000003814 drug Substances 0.000 description 5
- 208000019622 heart disease Diseases 0.000 description 5
- 238000000338 in vitro Methods 0.000 description 5
- 239000003550 marker Substances 0.000 description 5
- 238000002703 mutagenesis Methods 0.000 description 5
- 231100000350 mutagenesis Toxicity 0.000 description 5
- 230000004770 neurodegeneration Effects 0.000 description 5
- 210000002569 neuron Anatomy 0.000 description 5
- 108091008695 photoreceptors Proteins 0.000 description 5
- 239000000758 substrate Substances 0.000 description 5
- 208000024891 symptom Diseases 0.000 description 5
- 238000001890 transfection Methods 0.000 description 5
- 210000002845 virion Anatomy 0.000 description 5
- 108020005176 AU Rich Elements Proteins 0.000 description 4
- 208000020446 Cardiac disease Diseases 0.000 description 4
- 102000055765 ELAV-Like Protein 1 Human genes 0.000 description 4
- 101000705756 Homo sapiens Proteasome activator complex subunit 1 Proteins 0.000 description 4
- 206010061323 Optic neuropathy Diseases 0.000 description 4
- 108700029229 Transcriptional Regulatory Elements Proteins 0.000 description 4
- 238000003556 assay Methods 0.000 description 4
- 230000003197 catalytic effect Effects 0.000 description 4
- 238000005352 clarification Methods 0.000 description 4
- 230000000295 complement effect Effects 0.000 description 4
- 230000006378 damage Effects 0.000 description 4
- 230000002950 deficient Effects 0.000 description 4
- 239000003599 detergent Substances 0.000 description 4
- 230000003291 dopaminomimetic effect Effects 0.000 description 4
- 238000003780 insertion Methods 0.000 description 4
- 230000037431 insertion Effects 0.000 description 4
- 230000004048 modification Effects 0.000 description 4
- 238000012986 modification Methods 0.000 description 4
- 208000020911 optic nerve disease Diseases 0.000 description 4
- 230000008569 process Effects 0.000 description 4
- 210000001525 retina Anatomy 0.000 description 4
- UVFAEQZFLBGVRM-MSMWPWNWSA-N succinyl-Leu-Leu-Val-Tyr-7-amino-4-methylcoumarin Chemical compound C([C@H](NC(=O)[C@H](C(C)C)NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](NC(=O)CCC(O)=O)CC(C)C)C(=O)NC=1C=C2OC(=O)C=C(C)C2=CC=1)C1=CC=C(O)C=C1 UVFAEQZFLBGVRM-MSMWPWNWSA-N 0.000 description 4
- 238000003786 synthesis reaction Methods 0.000 description 4
- 230000008685 targeting Effects 0.000 description 4
- 229940124597 therapeutic agent Drugs 0.000 description 4
- 238000013519 translation Methods 0.000 description 4
- 102000053602 DNA Human genes 0.000 description 3
- 241000196324 Embryophyta Species 0.000 description 3
- 241000282412 Homo Species 0.000 description 3
- 241000713666 Lentivirus Species 0.000 description 3
- 241000124008 Mammalia Species 0.000 description 3
- 101150056038 PSME2 gene Proteins 0.000 description 3
- 206010063837 Reperfusion injury Diseases 0.000 description 3
- 108091027981 Response element Proteins 0.000 description 3
- 108091081024 Start codon Proteins 0.000 description 3
- 241000700618 Vaccinia virus Species 0.000 description 3
- 208000027418 Wounds and injury Diseases 0.000 description 3
- 230000004913 activation Effects 0.000 description 3
- 230000001580 bacterial effect Effects 0.000 description 3
- 230000000747 cardiac effect Effects 0.000 description 3
- 238000004587 chromatography analysis Methods 0.000 description 3
- 230000003247 decreasing effect Effects 0.000 description 3
- 230000001419 dependent effect Effects 0.000 description 3
- 238000003306 harvesting Methods 0.000 description 3
- 238000003364 immunohistochemistry Methods 0.000 description 3
- 208000014674 injury Diseases 0.000 description 3
- 238000001990 intravenous administration Methods 0.000 description 3
- 208000028867 ischemia Diseases 0.000 description 3
- 239000006193 liquid solution Substances 0.000 description 3
- 230000035772 mutation Effects 0.000 description 3
- 208000015122 neurodegenerative disease Diseases 0.000 description 3
- 239000002777 nucleoside Substances 0.000 description 3
- 125000003835 nucleoside group Chemical group 0.000 description 3
- 238000004806 packaging method and process Methods 0.000 description 3
- 239000003755 preservative agent Substances 0.000 description 3
- 230000004845 protein aggregation Effects 0.000 description 3
- 238000000746 purification Methods 0.000 description 3
- 241000894007 species Species 0.000 description 3
- 239000006228 supernatant Substances 0.000 description 3
- 230000004083 survival effect Effects 0.000 description 3
- 238000003151 transfection method Methods 0.000 description 3
- 241000701161 unidentified adenovirus Species 0.000 description 3
- 239000003981 vehicle Substances 0.000 description 3
- 108091006112 ATPases Proteins 0.000 description 2
- 102000057290 Adenosine Triphosphatases Human genes 0.000 description 2
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 2
- 101150063057 CRYAB gene Proteins 0.000 description 2
- 101150044789 Cap gene Proteins 0.000 description 2
- 208000031229 Cardiomyopathies Diseases 0.000 description 2
- 101000709520 Chlamydia trachomatis serovar L2 (strain 434/Bu / ATCC VR-902B) Atypical response regulator protein ChxR Proteins 0.000 description 2
- 108020004705 Codon Proteins 0.000 description 2
- 108091035707 Consensus sequence Proteins 0.000 description 2
- 102000004163 DNA-directed RNA polymerases Human genes 0.000 description 2
- 108090000626 DNA-directed RNA polymerases Proteins 0.000 description 2
- 102100036912 Desmin Human genes 0.000 description 2
- 108010044052 Desmin Proteins 0.000 description 2
- 108091029865 Exogenous DNA Proteins 0.000 description 2
- 108010010803 Gelatin Proteins 0.000 description 2
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 2
- 108010036652 HSC70 Heat-Shock Proteins Proteins 0.000 description 2
- 102100027421 Heat shock cognate 71 kDa protein Human genes 0.000 description 2
- 208000012902 Nervous system disease Diseases 0.000 description 2
- 108091034117 Oligonucleotide Proteins 0.000 description 2
- 241000283973 Oryctolagus cuniculus Species 0.000 description 2
- 108010038255 PAN enzyme Proteins 0.000 description 2
- 101150118259 PSME1 gene Proteins 0.000 description 2
- ISWSIDIOOBJBQZ-UHFFFAOYSA-N Phenol Chemical compound OC1=CC=CC=C1 ISWSIDIOOBJBQZ-UHFFFAOYSA-N 0.000 description 2
- ZTHYODDOHIVTJV-UHFFFAOYSA-N Propyl gallate Chemical compound CCCOC(=O)C1=CC(O)=C(O)C(O)=C1 ZTHYODDOHIVTJV-UHFFFAOYSA-N 0.000 description 2
- 241000700159 Rattus Species 0.000 description 2
- 241000700584 Simplexvirus Species 0.000 description 2
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 2
- RAHZWNYVWXNFOC-UHFFFAOYSA-N Sulphur dioxide Chemical compound O=S=O RAHZWNYVWXNFOC-UHFFFAOYSA-N 0.000 description 2
- 108091023040 Transcription factor Proteins 0.000 description 2
- 108010005705 Ubiquitinated Proteins Proteins 0.000 description 2
- 230000001594 aberrant effect Effects 0.000 description 2
- GFFGJBXGBJISGV-UHFFFAOYSA-N adenyl group Chemical group N1=CN=C2N=CNC2=C1N GFFGJBXGBJISGV-UHFFFAOYSA-N 0.000 description 2
- 102000003802 alpha-Synuclein Human genes 0.000 description 2
- 108090000185 alpha-Synuclein Proteins 0.000 description 2
- 230000015556 catabolic process Effects 0.000 description 2
- 210000000170 cell membrane Anatomy 0.000 description 2
- 230000001413 cellular effect Effects 0.000 description 2
- OSASVXMJTNOKOY-UHFFFAOYSA-N chlorobutanol Chemical compound CC(C)(O)C(Cl)(Cl)Cl OSASVXMJTNOKOY-UHFFFAOYSA-N 0.000 description 2
- 239000007771 core particle Substances 0.000 description 2
- 238000012258 culturing Methods 0.000 description 2
- 238000006731 degradation reaction Methods 0.000 description 2
- 238000012217 deletion Methods 0.000 description 2
- 230000037430 deletion Effects 0.000 description 2
- 238000013461 design Methods 0.000 description 2
- 210000005045 desmin Anatomy 0.000 description 2
- 239000003937 drug carrier Substances 0.000 description 2
- 230000002255 enzymatic effect Effects 0.000 description 2
- CBOQJANXLMLOSS-UHFFFAOYSA-N ethyl vanillin Chemical compound CCOC1=CC(C=O)=CC=C1O CBOQJANXLMLOSS-UHFFFAOYSA-N 0.000 description 2
- 210000002950 fibroblast Anatomy 0.000 description 2
- 239000012634 fragment Substances 0.000 description 2
- 239000008273 gelatin Substances 0.000 description 2
- 229920000159 gelatin Polymers 0.000 description 2
- 235000019322 gelatine Nutrition 0.000 description 2
- 235000011852 gelatine desserts Nutrition 0.000 description 2
- UYTPUPDQBNUYGX-UHFFFAOYSA-N guanine Chemical compound O=C1NC(N)=NC2=C1N=CN2 UYTPUPDQBNUYGX-UHFFFAOYSA-N 0.000 description 2
- 210000005003 heart tissue Anatomy 0.000 description 2
- 210000003494 hepatocyte Anatomy 0.000 description 2
- 230000001900 immune effect Effects 0.000 description 2
- 230000002458 infectious effect Effects 0.000 description 2
- 238000001802 infusion Methods 0.000 description 2
- 239000004615 ingredient Substances 0.000 description 2
- 238000007918 intramuscular administration Methods 0.000 description 2
- 239000006194 liquid suspension Substances 0.000 description 2
- 238000001823 molecular biology technique Methods 0.000 description 2
- 238000010369 molecular cloning Methods 0.000 description 2
- 230000002107 myocardial effect Effects 0.000 description 2
- 210000001577 neostriatum Anatomy 0.000 description 2
- 230000003287 optical effect Effects 0.000 description 2
- 239000000546 pharmaceutical excipient Substances 0.000 description 2
- 230000000069 prophylactic effect Effects 0.000 description 2
- 230000002797 proteolythic effect Effects 0.000 description 2
- 239000013608 rAAV vector Substances 0.000 description 2
- 230000002207 retinal effect Effects 0.000 description 2
- 239000011780 sodium chloride Substances 0.000 description 2
- 239000003381 stabilizer Substances 0.000 description 2
- 238000010361 transduction Methods 0.000 description 2
- 230000026683 transduction Effects 0.000 description 2
- 238000012546 transfer Methods 0.000 description 2
- 241001430294 unidentified retrovirus Species 0.000 description 2
- CHHHXKFHOYLYRE-UHFFFAOYSA-M 2,4-Hexadienoic acid, potassium salt (1:1), (2E,4E)- Chemical compound [K+].CC=CC=CC([O-])=O CHHHXKFHOYLYRE-UHFFFAOYSA-M 0.000 description 1
- 229940126695 20S proteasome inhibitor Drugs 0.000 description 1
- FWBHETKCLVMNFS-UHFFFAOYSA-N 4',6-Diamino-2-phenylindol Chemical compound C1=CC(C(=N)N)=CC=C1C1=CC2=CC=C(C(N)=N)C=C2N1 FWBHETKCLVMNFS-UHFFFAOYSA-N 0.000 description 1
- WXNZTHHGJRFXKQ-UHFFFAOYSA-N 4-chlorophenol Chemical compound OC1=CC=C(Cl)C=C1 WXNZTHHGJRFXKQ-UHFFFAOYSA-N 0.000 description 1
- 108010022579 ATP dependent 26S protease Proteins 0.000 description 1
- 108010085238 Actins Proteins 0.000 description 1
- 102000007469 Actins Human genes 0.000 description 1
- 229930024421 Adenine Natural products 0.000 description 1
- 241000649044 Adeno-associated virus 9 Species 0.000 description 1
- 229920001817 Agar Polymers 0.000 description 1
- 108010088751 Albumins Proteins 0.000 description 1
- 102000009027 Albumins Human genes 0.000 description 1
- GUBGYTABKSRVRQ-XLOQQCSPSA-N Alpha-Lactose Chemical compound O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@H]1O[C@@H]1[C@@H](CO)O[C@H](O)[C@H](O)[C@H]1O GUBGYTABKSRVRQ-XLOQQCSPSA-N 0.000 description 1
- 241000203069 Archaea Species 0.000 description 1
- 101100043731 Caenorhabditis elegans syx-3 gene Proteins 0.000 description 1
- 208000024172 Cardiovascular disease Diseases 0.000 description 1
- 241000282693 Cercopithecidae Species 0.000 description 1
- 108091062157 Cis-regulatory element Proteins 0.000 description 1
- 101710094648 Coat protein Proteins 0.000 description 1
- 208000027205 Congenital disease Diseases 0.000 description 1
- 241000699800 Cricetinae Species 0.000 description 1
- 230000004543 DNA replication Effects 0.000 description 1
- 229920002307 Dextran Polymers 0.000 description 1
- 206010061818 Disease progression Diseases 0.000 description 1
- 101100535673 Drosophila melanogaster Syn gene Proteins 0.000 description 1
- 102000016662 ELAV Proteins Human genes 0.000 description 1
- 108010053101 ELAV Proteins Proteins 0.000 description 1
- 102100031780 Endonuclease Human genes 0.000 description 1
- 108010042407 Endonucleases Proteins 0.000 description 1
- 241000991587 Enterovirus C Species 0.000 description 1
- 102000004190 Enzymes Human genes 0.000 description 1
- 108090000790 Enzymes Proteins 0.000 description 1
- 241000206602 Eukaryota Species 0.000 description 1
- 208000024412 Friedreich ataxia Diseases 0.000 description 1
- 108700039691 Genetic Promoter Regions Proteins 0.000 description 1
- 102000003886 Glycoproteins Human genes 0.000 description 1
- 108090000288 Glycoproteins Proteins 0.000 description 1
- 241000251728 Gnathostomata <vertebrate> Species 0.000 description 1
- HTTJABKRGRZYRN-UHFFFAOYSA-N Heparin Chemical compound OC1C(NC(=O)C)C(O)OC(COS(O)(=O)=O)C1OC1C(OS(O)(=O)=O)C(O)C(OC2C(C(OS(O)(=O)=O)C(OC3C(C(O)C(O)C(O3)C(O)=O)OS(O)(=O)=O)C(CO)O2)NS(O)(=O)=O)C(C(O)=O)O1 HTTJABKRGRZYRN-UHFFFAOYSA-N 0.000 description 1
- 101000878605 Homo sapiens Low affinity immunoglobulin epsilon Fc receptor Proteins 0.000 description 1
- 241000700588 Human alphaherpesvirus 1 Species 0.000 description 1
- 102000016252 Huntingtin Human genes 0.000 description 1
- 108050004784 Huntingtin Proteins 0.000 description 1
- 208000032578 Inherited retinal disease Diseases 0.000 description 1
- 108010074328 Interferon-gamma Proteins 0.000 description 1
- 102000008070 Interferon-gamma Human genes 0.000 description 1
- QIVBCDIJIAJPQS-VIFPVBQESA-N L-tryptophane Chemical compound C1=CC=C2C(C[C@H](N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-VIFPVBQESA-N 0.000 description 1
- OUYCCCASQSFEME-QMMMGPOBSA-N L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-QMMMGPOBSA-N 0.000 description 1
- GUBGYTABKSRVRQ-QKKXKWKRSA-N Lactose Natural products OC[C@H]1O[C@@H](O[C@H]2[C@H](O)[C@@H](O)C(O)O[C@@H]2CO)[C@H](O)[C@@H](O)[C@H]1O GUBGYTABKSRVRQ-QKKXKWKRSA-N 0.000 description 1
- 108091026898 Leader sequence (mRNA) Proteins 0.000 description 1
- 201000003533 Leber congenital amaurosis Diseases 0.000 description 1
- 108090001030 Lipoproteins Proteins 0.000 description 1
- 102000004895 Lipoproteins Human genes 0.000 description 1
- 102100038007 Low affinity immunoglobulin epsilon Fc receptor Human genes 0.000 description 1
- 241000283923 Marmota monax Species 0.000 description 1
- 101100368134 Mus musculus Syn1 gene Proteins 0.000 description 1
- 208000025966 Neurological disease Diseases 0.000 description 1
- 108010077850 Nuclear Localization Signals Proteins 0.000 description 1
- 102000007999 Nuclear Proteins Human genes 0.000 description 1
- 108010089610 Nuclear Proteins Proteins 0.000 description 1
- 101710163270 Nuclease Proteins 0.000 description 1
- 108700026244 Open Reading Frames Proteins 0.000 description 1
- 235000019483 Peanut oil Nutrition 0.000 description 1
- KDCGOANMDULRCW-UHFFFAOYSA-N Purine Natural products N1=CNC2=NC=NC2=C1 KDCGOANMDULRCW-UHFFFAOYSA-N 0.000 description 1
- 108010020942 REG proteasome activator subunit beta Proteins 0.000 description 1
- 108091034057 RNA (poly(A)) Proteins 0.000 description 1
- 108020004511 Recombinant DNA Proteins 0.000 description 1
- 241001068263 Replication competent viruses Species 0.000 description 1
- 201000007737 Retinal degeneration Diseases 0.000 description 1
- 108091081021 Sense strand Proteins 0.000 description 1
- 108020004682 Single-Stranded DNA Proteins 0.000 description 1
- 241000256251 Spodoptera frugiperda Species 0.000 description 1
- 101710172711 Structural protein Proteins 0.000 description 1
- 229930006000 Sucrose Natural products 0.000 description 1
- CZMRCDWAGMRECN-UGDNZRGBSA-N Sucrose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-UGDNZRGBSA-N 0.000 description 1
- 108700009124 Transcription Initiation Site Proteins 0.000 description 1
- 102000040945 Transcription factor Human genes 0.000 description 1
- 101710195626 Transcriptional activator protein Proteins 0.000 description 1
- QIVBCDIJIAJPQS-UHFFFAOYSA-N Tryptophan Natural products C1=CC=C2C(CC(N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-UHFFFAOYSA-N 0.000 description 1
- 206010046865 Vaccinia virus infection Diseases 0.000 description 1
- 230000002159 abnormal effect Effects 0.000 description 1
- 238000009825 accumulation Methods 0.000 description 1
- 230000021736 acetylation Effects 0.000 description 1
- 238000006640 acetylation reaction Methods 0.000 description 1
- 208000037919 acquired disease Diseases 0.000 description 1
- 230000003213 activating effect Effects 0.000 description 1
- 229960000643 adenine Drugs 0.000 description 1
- 150000003838 adenosines Chemical class 0.000 description 1
- 239000008272 agar Substances 0.000 description 1
- 235000010419 agar Nutrition 0.000 description 1
- 230000009435 amidation Effects 0.000 description 1
- 238000007112 amidation reaction Methods 0.000 description 1
- AVKUERGKIZMTKX-NJBDSQKTSA-N ampicillin Chemical compound C1([C@@H](N)C(=O)N[C@H]2[C@H]3SC([C@@H](N3C2=O)C(O)=O)(C)C)=CC=CC=C1 AVKUERGKIZMTKX-NJBDSQKTSA-N 0.000 description 1
- 229960000723 ampicillin Drugs 0.000 description 1
- 238000005571 anion exchange chromatography Methods 0.000 description 1
- 239000003242 anti bacterial agent Substances 0.000 description 1
- 239000000427 antigen Substances 0.000 description 1
- 108091007433 antigens Proteins 0.000 description 1
- 102000036639 antigens Human genes 0.000 description 1
- 210000004507 artificial chromosome Anatomy 0.000 description 1
- 230000002238 attenuated effect Effects 0.000 description 1
- 230000006399 behavior Effects 0.000 description 1
- 230000003542 behavioural effect Effects 0.000 description 1
- 230000008901 benefit Effects 0.000 description 1
- 230000003115 biocidal effect Effects 0.000 description 1
- 230000000903 blocking effect Effects 0.000 description 1
- 239000006172 buffering agent Substances 0.000 description 1
- 230000003139 buffering effect Effects 0.000 description 1
- 239000001506 calcium phosphate Substances 0.000 description 1
- 229910000389 calcium phosphate Inorganic materials 0.000 description 1
- 235000011010 calcium phosphates Nutrition 0.000 description 1
- 150000001720 carbohydrates Chemical class 0.000 description 1
- 235000014633 carbohydrates Nutrition 0.000 description 1
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 description 1
- 239000000969 carrier Substances 0.000 description 1
- 238000005277 cation exchange chromatography Methods 0.000 description 1
- 125000002091 cationic group Chemical group 0.000 description 1
- 230000003915 cell function Effects 0.000 description 1
- 210000002421 cell wall Anatomy 0.000 description 1
- 210000003169 central nervous system Anatomy 0.000 description 1
- 238000005119 centrifugation Methods 0.000 description 1
- 230000008859 change Effects 0.000 description 1
- 239000002738 chelating agent Substances 0.000 description 1
- 238000006243 chemical reaction Methods 0.000 description 1
- 229960004926 chlorobutanol Drugs 0.000 description 1
- 238000010367 cloning Methods 0.000 description 1
- 239000002299 complementary DNA Substances 0.000 description 1
- 150000001875 compounds Chemical class 0.000 description 1
- 238000004590 computer program Methods 0.000 description 1
- 230000021615 conjugation Effects 0.000 description 1
- 238000007796 conventional method Methods 0.000 description 1
- 239000002577 cryoprotective agent Substances 0.000 description 1
- 238000001212 derivatisation Methods 0.000 description 1
- 238000011161 development Methods 0.000 description 1
- LOKCTEFSRHRXRJ-UHFFFAOYSA-I dipotassium trisodium dihydrogen phosphate hydrogen phosphate dichloride Chemical compound P(=O)(O)(O)[O-].[K+].P(=O)(O)([O-])[O-].[Na+].[Na+].[Cl-].[K+].[Cl-].[Na+] LOKCTEFSRHRXRJ-UHFFFAOYSA-I 0.000 description 1
- 230000005750 disease progression Effects 0.000 description 1
- 230000004064 dysfunction Effects 0.000 description 1
- 239000002532 enzyme inhibitor Substances 0.000 description 1
- 210000000981 epithelium Anatomy 0.000 description 1
- 108700002672 epoxomicin Proteins 0.000 description 1
- DOGIDQKFVLKMLQ-JTHVHQAWSA-N epoxomicin Chemical compound CC[C@H](C)[C@H](N(C)C(C)=O)C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CC(C)C)C(=O)[C@@]1(C)CO1 DOGIDQKFVLKMLQ-JTHVHQAWSA-N 0.000 description 1
- BEFDCLMNVWHSGT-UHFFFAOYSA-N ethenylcyclopentane Chemical compound C=CC1CCCC1 BEFDCLMNVWHSGT-UHFFFAOYSA-N 0.000 description 1
- 229940073505 ethyl vanillin Drugs 0.000 description 1
- 210000003527 eukaryotic cell Anatomy 0.000 description 1
- 238000011156 evaluation Methods 0.000 description 1
- 210000001808 exosome Anatomy 0.000 description 1
- 239000013613 expression plasmid Substances 0.000 description 1
- 230000004438 eyesight Effects 0.000 description 1
- 238000001914 filtration Methods 0.000 description 1
- 239000012530 fluid Substances 0.000 description 1
- 238000010353 genetic engineering Methods 0.000 description 1
- 235000011187 glycerol Nutrition 0.000 description 1
- 229960005150 glycerol Drugs 0.000 description 1
- 230000013595 glycosylation Effects 0.000 description 1
- 238000006206 glycosylation reaction Methods 0.000 description 1
- 229960002897 heparin Drugs 0.000 description 1
- 229920000669 heparin Polymers 0.000 description 1
- 208000006454 hepatitis Diseases 0.000 description 1
- 231100000283 hepatitis Toxicity 0.000 description 1
- 125000005842 heteroatom Chemical group 0.000 description 1
- 238000002744 homologous recombination Methods 0.000 description 1
- 230000006801 homologous recombination Effects 0.000 description 1
- 210000005260 human cell Anatomy 0.000 description 1
- 230000001771 impaired effect Effects 0.000 description 1
- 230000006872 improvement Effects 0.000 description 1
- 238000000099 in vitro assay Methods 0.000 description 1
- 230000001524 infective effect Effects 0.000 description 1
- 230000002757 inflammatory effect Effects 0.000 description 1
- 230000000977 initiatory effect Effects 0.000 description 1
- 230000010354 integration Effects 0.000 description 1
- 229960003130 interferon gamma Drugs 0.000 description 1
- 230000003834 intracellular effect Effects 0.000 description 1
- 238000007917 intracranial administration Methods 0.000 description 1
- 238000007913 intrathecal administration Methods 0.000 description 1
- 238000004255 ion exchange chromatography Methods 0.000 description 1
- 239000008101 lactose Substances 0.000 description 1
- 150000002632 lipids Chemical class 0.000 description 1
- 239000002502 liposome Substances 0.000 description 1
- 239000007788 liquid Substances 0.000 description 1
- 239000012139 lysis buffer Substances 0.000 description 1
- 239000000693 micelle Substances 0.000 description 1
- 201000006417 multiple sclerosis Diseases 0.000 description 1
- 210000003098 myoblast Anatomy 0.000 description 1
- 230000000926 neurological effect Effects 0.000 description 1
- 229910052757 nitrogen Inorganic materials 0.000 description 1
- 239000002736 nonionic surfactant Substances 0.000 description 1
- 231100000252 nontoxic Toxicity 0.000 description 1
- 230000003000 nontoxic effect Effects 0.000 description 1
- 210000004940 nucleus Anatomy 0.000 description 1
- 230000003204 osmotic effect Effects 0.000 description 1
- 125000001312 palmitoyl group Chemical group O=C([*])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])[H] 0.000 description 1
- 229940090668 parachlorophenol Drugs 0.000 description 1
- 239000000312 peanut oil Substances 0.000 description 1
- 239000001814 pectin Substances 0.000 description 1
- 235000010987 pectin Nutrition 0.000 description 1
- 229920001277 pectin Polymers 0.000 description 1
- 229960003742 phenol Drugs 0.000 description 1
- 239000002953 phosphate buffered saline Substances 0.000 description 1
- 230000026731 phosphorylation Effects 0.000 description 1
- 238000006366 phosphorylation reaction Methods 0.000 description 1
- 230000037081 physical activity Effects 0.000 description 1
- 230000004962 physiological condition Effects 0.000 description 1
- 239000000049 pigment Substances 0.000 description 1
- 229920001983 poloxamer Polymers 0.000 description 1
- 229920001993 poloxamer 188 Polymers 0.000 description 1
- 230000001323 posttranslational effect Effects 0.000 description 1
- 235000010241 potassium sorbate Nutrition 0.000 description 1
- 239000004302 potassium sorbate Substances 0.000 description 1
- 229940069338 potassium sorbate Drugs 0.000 description 1
- 230000002265 prevention Effects 0.000 description 1
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 1
- 239000000473 propyl gallate Substances 0.000 description 1
- 229940075579 propyl gallate Drugs 0.000 description 1
- 235000010388 propyl gallate Nutrition 0.000 description 1
- 230000017854 proteolysis Effects 0.000 description 1
- 125000000561 purinyl group Chemical group N1=C(N=C2N=CNC2=C1)* 0.000 description 1
- 238000002708 random mutagenesis Methods 0.000 description 1
- 230000008707 rearrangement Effects 0.000 description 1
- 238000010188 recombinant method Methods 0.000 description 1
- 239000013074 reference sample Substances 0.000 description 1
- 230000008844 regulatory mechanism Effects 0.000 description 1
- 101150066583 rep gene Proteins 0.000 description 1
- 230000003362 replicative effect Effects 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 230000004044 response Effects 0.000 description 1
- 230000004258 retinal degeneration Effects 0.000 description 1
- 150000003839 salts Chemical class 0.000 description 1
- 239000000523 sample Substances 0.000 description 1
- 238000012216 screening Methods 0.000 description 1
- 239000008159 sesame oil Substances 0.000 description 1
- 235000011803 sesame oil Nutrition 0.000 description 1
- 238000004904 shortening Methods 0.000 description 1
- 230000003584 silencer Effects 0.000 description 1
- 238000001542 size-exclusion chromatography Methods 0.000 description 1
- 239000007787 solid Substances 0.000 description 1
- 239000007790 solid phase Substances 0.000 description 1
- 239000002904 solvent Substances 0.000 description 1
- 235000010199 sorbic acid Nutrition 0.000 description 1
- 229940075582 sorbic acid Drugs 0.000 description 1
- 239000004334 sorbic acid Substances 0.000 description 1
- 230000009870 specific binding Effects 0.000 description 1
- 230000006641 stabilisation Effects 0.000 description 1
- 238000011105 stabilization Methods 0.000 description 1
- 238000010186 staining Methods 0.000 description 1
- 239000005720 sucrose Substances 0.000 description 1
- 229940044609 sulfur dioxide Drugs 0.000 description 1
- 235000010269 sulphur dioxide Nutrition 0.000 description 1
- 239000004094 surface-active agent Substances 0.000 description 1
- 230000002194 synthesizing effect Effects 0.000 description 1
- 238000002560 therapeutic procedure Methods 0.000 description 1
- 238000011285 therapeutic regimen Methods 0.000 description 1
- 230000005030 transcription termination Effects 0.000 description 1
- 230000002103 transcriptional effect Effects 0.000 description 1
- 230000009466 transformation Effects 0.000 description 1
- 230000009261 transgenic effect Effects 0.000 description 1
- 230000014621 translational initiation Effects 0.000 description 1
- 230000017105 transposition Effects 0.000 description 1
- QORWJWZARLRLPR-UHFFFAOYSA-H tricalcium bis(phosphate) Chemical compound [Ca+2].[Ca+2].[Ca+2].[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O QORWJWZARLRLPR-UHFFFAOYSA-H 0.000 description 1
- 230000010415 tropism Effects 0.000 description 1
- 210000004881 tumor cell Anatomy 0.000 description 1
- 230000007306 turnover Effects 0.000 description 1
- OUYCCCASQSFEME-UHFFFAOYSA-N tyrosine Natural products OC(=O)C(N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-UHFFFAOYSA-N 0.000 description 1
- 230000034512 ubiquitination Effects 0.000 description 1
- 238000010798 ubiquitination Methods 0.000 description 1
- 241001529453 unidentified herpesvirus Species 0.000 description 1
- 238000011144 upstream manufacturing Methods 0.000 description 1
- 208000007089 vaccinia Diseases 0.000 description 1
- 230000002861 ventricular Effects 0.000 description 1
- 230000035899 viability Effects 0.000 description 1
- 230000004304 visual acuity Effects 0.000 description 1
- 230000000007 visual effect Effects 0.000 description 1
- 230000004382 visual function Effects 0.000 description 1
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 1
- 210000005253 yeast cell Anatomy 0.000 description 1
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
-
- 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
-
- 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
- C07K14/46—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
- C07K14/47—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
- C07K14/4701—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
- C07K14/4702—Regulators; Modulating activity
- C07K14/4705—Regulators; Modulating activity stimulating, promoting or activating activity
-
- 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
-
- A—HUMAN NECESSITIES
- A01—AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
- A01K—ANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
- A01K2207/00—Modified animals
- A01K2207/15—Humanized animals
-
- A—HUMAN NECESSITIES
- A01—AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
- A01K—ANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
- A01K2217/00—Genetically modified animals
- A01K2217/05—Animals comprising random inserted nucleic acids (transgenic)
- A01K2217/052—Animals comprising random inserted nucleic acids (transgenic) inducing gain of function
-
- A—HUMAN NECESSITIES
- A01—AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
- A01K—ANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
- A01K2227/00—Animals characterised by species
- A01K2227/10—Mammal
- A01K2227/105—Murine
-
- A—HUMAN NECESSITIES
- A01—AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
- A01K—ANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
- A01K2267/00—Animals characterised by purpose
- A01K2267/03—Animal model, e.g. for test or diseases
- A01K2267/0306—Animal model for genetic diseases
-
- A—HUMAN NECESSITIES
- A01—AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
- A01K—ANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
- A01K2267/00—Animals characterised by purpose
- A01K2267/03—Animal model, e.g. for test or diseases
- A01K2267/0306—Animal model for genetic diseases
- A01K2267/0318—Animal model for neurodegenerative disease, e.g. non- Alzheimer's
-
- A—HUMAN NECESSITIES
- A01—AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
- A01K—ANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
- A01K2267/00—Animals characterised by purpose
- A01K2267/03—Animal model, e.g. for test or diseases
- A01K2267/035—Animal model for multifactorial diseases
- A01K2267/0375—Animal model for cardiovascular diseases
-
- 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
- C12N2750/14141—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
-
- 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
- C12N2830/00—Vector systems having a special element relevant for transcription
- C12N2830/008—Vector systems having a special element relevant for transcription cell type or tissue specific enhancer/promoter combination
-
- 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
- C12N2830/00—Vector systems having a special element relevant for transcription
- C12N2830/48—Vector systems having a special element relevant for transcription regulating transport or export of RNA, e.g. RRE, PRE, WPRE, CTE
-
- 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
- C12N2840/00—Vectors comprising a special translation-regulating system
- C12N2840/20—Vectors comprising a special translation-regulating system translation of more than one cistron
Definitions
- the present disclosure pertains to the medical field, including gene therapy constructs, vectors (e.g., viral vectors), and compositions suitable for delivery of nucleic acids encoding therapeutic proteins, and methods of using the same.
- vectors e.g., viral vectors
- compositions suitable for delivery of nucleic acids encoding therapeutic proteins and methods of using the same.
- the ubiquitin-proteasome system mediates the targeted degradation of abnormal as well as most normal intracellular proteins and generally includes two main steps: ubiquitination of a specific protein and subsequent degradation of the ubiquitinated protein by the proteasome.
- Eukaryotic proteasomes are large multisubunit complexes that degrade the majority of proteins in the cell under normal conditions.
- the proteasome can be divided into two subcomplexes: a regulatory cap and the 20S catalytic core particle.
- the catalytic core (20S) is barrel shaped in structure and contains the enzymatic activity to degrade proteins.
- the activity of the catalytic core is regulated by 2 main types of regulatory caps: 19S and PA28. When combined with the 20S catalytic core, these caps, respectively, form the 26S proteasome (20S + 19S) that degrades ubiquitin-tagged proteins or the 1 IS proteasome (20S + PA28) that degrades damaged or oxidized proteins independent of ubiquitin.
- the 19S particle in eukaryotes consists of 19 individual proteins and is divisible into two subassemblies, a 9-subunit base that binds directly to the a ring of the 20S core particle, and a 10-subunit lid.
- Six of the nine base proteins are ATPase subunits from the AAA Family, and an evolutionary homolog of these ATPases exists in archaea, called PAN (Proteasome- Activating Nucleotidase).
- PA28 also referred to as 1 IS, REG or PSME
- PA28a also known as Proteasome activator complex subunit 1, PSME1, PA28A, and REGalpha
- RA28b also known as Proteasome activator complex subunit 2, PSME2, PA28B, and REGbeta
- RA28g which assemble as heptameric hetero (RA28ab) or homo (RA28g) rings on one or both extremities of the 20S proteasome cylindrical structure.
- PSME1 (or PA28a) and PSME2 (or RA28b) are encoded by the PSME1 gene and the PSME2 gene, respectively.
- Proteinopathies are diseases caused by protein misfolding and are characterized by aberrant protein aggregation. Terminally misfolded proteins are degraded mainly by the UPS. Ubiquitinated proteins usually accumulate in the cell during proteasome functional insufficiency (PFI). Consequently, PFI and protein aggregation can result in a cycle that can result in proteinopathy and other disorders. Enhancement of proteasomal function has been shown to protect against cardiac proteinopathy and ischemia/reperfusion injury in mice. Li et ak, J Clin Invest. 2011 Sep; 121(9):3689-700. Furthermore, increased proteasomal activity has been shown to support photoreceptor survival in inherited retinal degeneration. Lobanova et ak, Nat Commun. 2018 Apr 30; 9(1): 1738.
- Certain aspects of the present disclosure are directed to constructs (e.g., gene therapy constructs) for use in packaging a PA28 cap (an alpha subunit alone, or alpha plus beta subunits) in a vector (e.g., viral vector such as AAV or lentivirus) for delivery to discrete parts of the body, e.g., the eye, brain, or heart, to increase the capacity of proteasomes to process misfolded or damaged proteins that cause disease.
- a vector e.g., viral vector such as AAV or lentivirus
- the present disclosure provides a construct (e.g., a gene therapy construct) comprising a polynucleotide comprising a promoter and a nucleic acid sequence encoding a Proteasome activator complex subunit 1 (PA28A).
- a construct e.g., a gene therapy construct
- PA28A Proteasome activator complex subunit 1
- the present disclosure provides a construct (e.g., a gene therapy construct) comprising a polynucleotide comprising a promoter, a nucleic acid sequence encoding a Proteasome activator complex subunit 1 (PA28A), and a nucleic acid sequence encoding Proteasome activator complex subunit 2 (PA28B).
- a construct e.g., a gene therapy construct
- PA28A a nucleic acid sequence encoding a Proteasome activator complex subunit 1
- PA28B nucleic acid sequence encoding Proteasome activator complex subunit 2
- the construct is a bicistronic construct.
- the present disclosure provides a gene therapy constructs (e.g., a bicistronic construct) comprising a polynucleotide comprising (a) a first promoter sequence and a nucleic acid sequence encoding a Proteasome activator complex subunit 1 (PA28A), and (b) a second promoter sequence and a nucleic acid sequence encoding Proteasome activator complex subunit 2 (PA28B).
- the first and second promoter are the same. In some aspects, the first and second promoter are different.
- the present disclosure provides a plurality of gene therapy constructs comprising (a) a polynucleotide comprising a promoter sequence and a nucleic acid sequence encoding a Proteasome activator complex subunit 1 (PA28A), and (b) a polynucleotide comprising a promoter sequence and a nucleic acid sequence encoding a promoter sequence and Proteasome activator complex subunit 2 (PA28B).
- the promoters are the same. In some aspects, the promoters are different.
- the present disclosure provides an expression cassette comprising a polynucleotide comprising a promoter sequence and a nucleic acid sequence encoding a Proteasome activator complex subunit 1 (PA28A).
- PA28A Proteasome activator complex subunit 1
- the present disclosure provides an expression cassette comprising a polynucleotide comprising a promoter, a nucleic acid sequence encoding a Proteasome activator complex subunit 1 (PA28A), and a nucleic acid sequence encoding Proteasome activator complex subunit 2 (PA28B).
- a polynucleotide comprising a promoter, a nucleic acid sequence encoding a Proteasome activator complex subunit 1 (PA28A), and a nucleic acid sequence encoding Proteasome activator complex subunit 2 (PA28B).
- the present disclosure provides a first expression cassette comprising a polynucleotide comprising a promoter, a nucleic acid sequence encoding a Proteasome activator complex subunit 1 (PA28A), and a second expression cassette comprising a polynucleotide comprising a promoter and Proteasome activator complex subunit 2 (PA28B).
- the promoter of the first and second expression cassette are the same or different.
- the promoter is a heterologous promoter. In some aspects, the promoter is tissue or cell-type specific promoter. In some aspects, the promoter is a bi directional promoter. In some aspects, the promoter is selected from a MecP2 promoter, a GUSB promoter, a CBh promoter, a PGK promoter, a human synapsin (Syn) promoter, a human rhodopsin (Rhod) kinase promoter, a cone arrestin promoter, a tyrosine hydroxylase promoter, a dopamine transporter promoter, a GRK1 promoter, a cardiac myosin light chain 2 promoter, a CAMKII promoter, a hDlx promoter, a CMV promoter, a CAG promoter, a bidirectional CAG promoter, a CBA promoter, an EFla promoter, or a human ubiquitin C promote
- the gene therapy construct or expression construct further comprises a polyadenylation site, a woodchuck hepatitis virus post-transcriptional regulatory element (WPRE), an optimized woodchuck hepatitis virus post-transcriptional regulatory element (oPRE), an intron, a microRNA binding motif, or any combination thereof.
- WPRE woodchuck hepatitis virus post-transcriptional regulatory element
- oPRE optimized woodchuck hepatitis virus post-transcriptional regulatory element
- intron a microRNA binding motif, or any combination thereof.
- the gene therapy construct or expression construct comprises a chimeric intron. In some aspects, the gene therapy construct or expression construct comprises a beta-globin intron. In some aspects, the gene therapy construct or expression construct comprises minute virus of mice (MVM) intron.
- VMM minute virus of mice
- the gene therapy construct or expression construct comprises a polyA sequence selected from the group consisting of a beta-globin poly A, a bovine growth hormone polyA (bGH polyA), a human growth hormone polyA (hGH polyA), and a SV40 immediate early poly A (SV40 polyA).
- bGH polyA bovine growth hormone polyA
- hGH polyA human growth hormone polyA
- SV40 polyA SV40 immediate early poly A
- the gene therapy construct or expression construct further comprises a WPRE. In some aspects, the gene therapy construct or expression construct further comprises an optimized woodchuck hepatitis virus post-transcriptional regulatory element (oPRE).
- oPRE woodchuck hepatitis virus post-transcriptional regulatory element
- the present disclosure provides a vector comprising gene therapy construct or expression construct disclosed herein.
- the vector is a viral vector.
- the viral vector is an adeno-associated virus (AAV) vector.
- the AAV vector comprises an inverted terminal repeat (ITR), e.g., a pair of ITRs flanking the gene therapy or expression construct.
- ITR inverted terminal repeat
- the gene therapy construct, expression construct, or viral vector disclosed herein is packaged in an AAV capsid (e.g., a recombinant AAV (rAAV)).
- the AAV serotype of the capsid is selected from AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAVrh8, AAV9, AAV10, AAVrhlO, or AAV.PHP.B.
- the disease or condition is Alzheimer’s disease, Dutch hereditary cerebral hemorrhage with amyloidosis, Congophilic angiopathy, Tauopathies, Corticobasal degeneration, Pick’s disease, Progressive supranuclear palsy, Prion diseases, Creutzfeld-Jacob disease, Gerstmann-Straussler-Schneiker syndrome, fatal familia insomnia, Kuru, Bovine spongiform encephalopathy, Scrapie, Chronic wasting disease, Synucleinopathies, Parkinson’s disease, Lewy body variant of Alzheimer’s disease, Diffuse Lewy body disease, Dementia with Lewy bodies, Multiple system atrophy, Neurodegeneration with brain iron accumulation type I, Amyotrophic lateral sclerosis and frontotemporal lobar degeneration, Amyotrophic lateral sclerosis, Huntington’s disease, Hereditary dentatorubral-pallidoluysian atrophy, Kennedy’s disease or X-linked spinal and bulbar muscular atrophy, Spin
- FIGs. 1A-1C depict exemplary plasmids for expression of PA28alpha
- FIG. 1A depicts an exemplary plasmid for expression of PA28alpha comprising a CMV enhancer, a minimal CMV promoter, PA28alpha gene, and SV40 polyadenylation signal.
- FIG. IB depicts an exemplary plasmid for expression of PA28alpha and PA28beta comprising a minimal CMV promoter, a PA28beta gene, a SV40 polyadenylation signal, a CMV enhancer, a second minimal CMV promoter, a PA28alpha gene, and a second SV40 polyadenylation signal.
- FIG. 1C depicts an exemplary empty vector plasmid which does not express PA28alpha or PA28beta.
- FIGs. 2A-2E show proteasome activity in HEK-293T cells transfected with plasmids expressing PA28alpha-only (FIG. 1A), PA28alpha/beta 11 S caps (FIG. IB), or an empty vector control (FIG. 1C).
- FIG. 2A depicts the proteasome assay, in which proteasome activity cleaves the artificial substrate Suc-LLVY-AMC (AMC) and generates free AMC which is fluorescent. Proteasome activity was analyzed with either 5 pL (FIG. 2B), 10 pL (FIG. 2C), 25 pL (FIG. 2D), or 50 pL (FIG. 2E) cell lysate.
- FIGs. 3A-3C are western blots showing protein expression of PA28alpha (FIG.
- PA28beta (FIG. 3B), or PA28alpha and PA28beta (FIG. 3C) in HEK-293T cells transfected with plasmids expressing PA28alpha-only (FIG. 1A), PA28alpha/beta 1 IS caps (FIG. IB), or an empty vector control (FIG. 1C).
- FIGs. 4A-4M depict exemplary plasmids for expression of PA28alpha
- FIG. 4A depicts an exemplary plasmid for expression of PA28beta comprising a CMV promoter, PA28beta gene, and SV40 polyadenylation signal.
- FIG. 4B depicts an exemplary plasmid for tandem expression of PA28alpha and PA28beta comprising a promoter, PA28alpha gene, an optimized WPRE (oPRE), a polyA tail, a second promoter, a PA28beta gene, a second optimized WPRE (oPRE), and a second polyA tail.
- oPRE optimized WPRE
- FIG. 4C depicts an exemplary bidirectional plasmid for expression of PA28alpha and PA28beta comprising a poly A tail, a PA28alpha gene, a promoter, an enhancer, a second promoter, a PA28beta gene, and a second polyA tail.
- FIG. 4D depicts exemplary 1:1 coexpression of PA28alpha and PA28beta comprising a first plasmid comprising a promoter, a PA28alpha gene, an optimized WPRE (oPRE), and a poly A tail and a second plasmid comprising a promoter, a PA28beta gene, a second optimized WPRE (oPRE), and a polyA tail.
- FIG. 4D depicts exemplary 1:1 coexpression of PA28alpha and PA28beta comprising a first plasmid comprising a promoter, a PA28alpha gene, an optimized WPRE (oPRE), and a poly A tail and a second plasmi
- FIG. 4E depicts an exemplary P2A self-cleavage plasmid for expression of PA28alpha and PA28beta comprising a promoter, a PA28alpha gene, a P2A site, a PA28beta gene, and a polyA tail.
- FIG. 4F depicts an exemplary P2A self-cleavage plasmid for expression of PA28alpha and PA28beta comprising a CMV promoter, a PA28alpha gene, a P2A site, a PA28beta gene, and a SV40 polyadenylation signal.
- FIG. 4E depicts an exemplary P2A self-cleavage plasmid for expression of PA28alpha and PA28beta comprising a promoter, a PA28alpha gene, a P2A site, a PA28beta gene, and a SV40 polyadenylation signal.
- FIG. 4G depicts an exemplary IRES self-cleavage plasmid for expression of PA28alpha and PA28beta comprising a promoter, a PA28alpha gene, a IRES site, a PA28beta gene, and a polyA tail.
- FIG. 4H depicts an exemplary IRES self-cleavage plasmid for expression of PA28alpha and PA28beta comprising a CMV promoter, a PA28alpha gene, an IRES site, a PA28beta gene, and a SV40 polyadenylation signal.
- FIG. 4G depicts an exemplary IRES self-cleavage plasmid for expression of PA28alpha and PA28beta comprising a promoter, a PA28alpha gene, a IRES site, a PA28beta gene, and a SV40 polyadenylation signal.
- FIG. 41 depicts an exemplary flexible linker plasmid for expression of PA28alpha and PA28beta comprising a promoter, a PA28alpha gene, a linker, a PA28beta gene, and a polyA tail.
- FIG. 4J depicts an exemplary flexible linker plasmid for expression of PA28alpha and PA28beta comprising a CMV promoter, a PA28alpha gene, a 3xGGGGS flexible linker, a PA28beta gene, and a SV40 polyadenylation signal.
- FIGs. 5A-5C are western blots showing protein expression of PA28alpha (FIG.
- PA28beta (FIG. 5B), or PA28alpha and PA28beta (FIG. 5C) in HEK-293T cells transfected with either a PA28alpha and PA28beta bicistronic plasmid (FIG. 4C), a 1 : 1 ratio of a PA28alpha and PA28beta plasmid (FIG. 4D), a PA28alpha only plasmid, a PA28beta only plasmid, a PA28alpha and PA28beta IRES bicistronic plasmid (FIGs. 4G and 4H), a PA28alpha and PA28beta P2A bicistronic plasmid (FIG.
- FIGs. 6A and 6B are graphs showing the proteasome activity of HEK-293T cells transfected with either a PA28alpha and PA28beta bicistronic plasmid (FIG. 4C), a 1 : 1 ratio of a PA28alpha and PA28beta plasmid (FIG. 4D), a PA28alpha only plasmid, a PA28beta only plasmid, a PA28alpha and PA28beta IRES bicistronic plasmid (FIGs.
- FIGs. 7A-7J depict exemplary AAV expression vectors for expression of
- FIG. 7A depicts an exemplary AAV expression vector for tandem expression of PA28alpha and PA28beta comprising a 5' ITR, CBh promoter, PA28alpha gene, an optimized WPRE (oPRE), bGH polyadenylation signal, a second CBh promoter, a PA28beta gene, a second optimized WPRE (oPRE), a second bGH polyadenylation signal, and a 3' ITR.
- FIG. 7B depicts an exemplary AAV expression vector for tandem expression of PA28alpha and PA28beta comprising a 5' ITR, CBh promoter, a PA28alpha gene, a bGH polyadenylation signal, a second CBh promoter, a PA28beta gene, a second bGH polyadenylation signal, and a 3' ITR.
- FIG. 7B depicts an exemplary AAV expression vector for tandem expression of PA28alpha and PA28beta comprising a 5' ITR, CBh promoter, a PA28alpha gene, a bGH polyadenylation signal, a second CBh promoter, a PA28beta gene, a second bGH polyadenylation signal, and a 3' ITR.
- FIG. 7C depicts an exemplary AAV expression vector for tandem expression of PA28alpha and PA28beta comprising a 5' ITR, hRHO promoter, a PA28alpha gene, a bGH polyadenylation signal, a second hRHO promoter, a PA28beta gene, a second bGH polyadenylation signal, and a 3' ITR.
- FIG. 7C depicts an exemplary AAV expression vector for tandem expression of PA28alpha and PA28beta comprising a 5' ITR, hRHO promoter, a PA28alpha gene, a bGH polyadenylation signal, a second hRHO promoter, a PA28beta gene, a second bGH polyadenylation signal, and a 3' ITR.
- FIG. 7D depicts an exemplary AAV expression vector for tandem expression of PA28alpha and PA28beta comprising a 5' ITR, a hRHO promoter, a PA28alpha gene, an optimized WPRE (oPRE), a bGH polyadenylation signal, a second hRHO promoter, a PA28beta gene, a second optimized WPRE (oPRE), a second bGH polyadenylation signal, and a 3' ITR.
- oPRE optimized WPRE
- oPRE second optimized WPRE
- a second bGH polyadenylation signal a 3' ITR.
- FIG. 7E depicts an exemplary AAV expression vector for tandem expression of PA28alpha and PA28beta comprising a 5' ITR, a SYN1 promoter, a PA28alpha gene, a bGH polyadenylation signal, a second SYN 1 promoter, a PA28beta gene, a second bGH polyadenylation signal, and a 3' ITR.
- FIG. 7E depicts an exemplary AAV expression vector for tandem expression of PA28alpha and PA28beta comprising a 5' ITR, a SYN1 promoter, a PA28alpha gene, a bGH polyadenylation signal, a second SYN 1 promoter, a PA28beta gene, a second bGH polyadenylation signal, and a 3' ITR.
- FIG. 7G depicts an exemplary AAV expression vector for tandem expression of PA28alpha and PA28beta comprising a 5' ITR, a EFla promoter, a PA28alpha gene, a bGH polyadenylation signal, a second EFla promoter, a PA28beta gene, a second bGH polyadenylation signal and a 3' ITR.
- FIG. 7G depicts an exemplary AAV expression vector for tandem expression of PA28alpha and PA28beta comprising a 5' ITR, a EFla promoter, a PA28alpha gene, a bGH polyadenylation signal, a second EFla promoter, a PA28beta gene, a second bGH polyadenylation signal and a 3' ITR.
- FIG. 7H depicts an exemplary AAV expression vector for bicistronic expression of PA28alpha and PA28beta comprising an AAV2 ITR, an SV40 polyadenylation signal, a PA28beta gene, an hRHO promoter, a CMV enhancer, a second hRHO promoter, a PA28alpha gene, a second SV40 polyadenylation signal, and a second AAV2 ITR.
- FIG. 1 depicts an exemplary AAV expression vector for bicistronic expression of PA28alpha and PA28beta comprising an AAV2 ITR, an SV40 polyadenylation signal, a PA28beta gene, an hRHO promoter, a CMV enhancer, a second hRHO promoter, a PA28alpha gene, a second SV40 polyadenylation signal, and a second AAV2 ITR.
- FIG. 71 depicts an exemplary AAV expression vector for bicistronic expression of PA28alpha and PA28beta comprising an AAV2 ITR, an SV40 polyadenylation signal, an optimized WPRE (oPRE), a PA28beta gene, a SYN1 promoter, a CMV enhancer, a second SYN1 promoter, a PA28alpha gene, a second optimized WPRE (oPRE), a second SV40 polyadenylation signal, and a AAV2 ITR.
- FIG. 7J depicts an exemplary AAV expression vector for bicistronic expression of PA28alpha and PA28beta comprising an AAV2 ITR, a SV40 polyadenylation signal, a PA28beta gene, a SYN1 promoter, a CMV enhancer, a second SYN1 promoter, a PA28beta gene, a second SV40 polyadenylation signal, and a second AAV2 ITR.
- PA28beta (FIG. 8B), or PA28alpha and PA28beta (FIG. 8C) in HEK-293T cells transfected with either a wild-type pBI-CMV vector (ZsGreen) (FIG. 1C), a PA28alpha and PA28beta bicistronic plasmid (FIG. IB), a PA28alpha only plasmid, a PA28beta only plasmid, a 1 : 1 ratio of a PA28alpha and PA28beta plasmid (FIG. 4D), a PA28alpha and PA28beta tandem plasmid under the transcriptional control of a EFla promoter (FIG.
- FIG. 7G a PA28alpha and PA28beta tandem plasmid under the transcriptional control of a hSynapsin promoter
- FIG. 7J a PA28alpha and PA28beta tandem plasmid under the transcriptional control of a hSynapsin promoter with an optimized WPRE (oPRE) sequence
- oPRE WPRE
- FIG. 7B a PA28alpha and PA28beta tandem plasmid under the transcriptional control of a CBh promoter
- FIGs. 9A-9B are graphs showing the proteasome activity of HEK-293T cells transfected with either a wild-type pBI-CMV vector (ZsGreen) (FIG. 1C), a PA28alpha and PA28beta bicistronic plasmid (pBi-PSMEl-PSME2) (FIG. IB), a PA28alpha only plasmid (PSMEl only), a PA28beta only plasmid (PSME2 only), a 1:1 ratio of a PA28alpha and PA28beta plasmid (1 : 1 PSMEl :PSME2) (FIG.
- ZsGreen wild-type pBI-CMV vector
- pBi-PSMEl-PSME2 PA28alpha and PA28beta bicistronic plasmid
- PSMEl PA28alpha only plasmid
- PSME2 PA28beta only plasmid
- a PA28alpha and PA28beta tandem plasmid under the transcriptional control of a EFla promoter (EFla tandem) (FIG. 7G)
- a PA28alpha and PA28beta tandem plasmid under the transcriptional control of a hSynapsin promoter (hSyn-tandem) (FIG. IT)
- a PA28alpha and PA28beta tandem plasmid under the transcriptional control of a hSynapsin promoter with an optimized WPRE (oPRE) sequence hSyn-OPRE tandem
- a PA28alpha and PA28beta tandem plasmid under the transcriptional control of a CBh promoter CBh- tandem
- FIGs. 10A-10B are western blots showing in vivo expression of PA28alpha and
- FIG. 10A shows expression of HSC70, beta-actin, PA28alpha, PA28beta, and GFP from mice injected in the left eye with an scAAV8 viral mixture comprising a 4:4: 1 (by volume) mixture of scAAV8-hRho-mPMSEl (PA28alpha) (titer: 5.38xl0 13 gc/mL), scAAV8-hRho-mPSME2 (PA28beta) (titer: 3.99xl0 13 gc/mL), and scAAV8-hRho-eGFP (titer: 4.49xl0 13 gc/mL).
- PA28alpha scAAV8-hRho-mPMSEl
- PA28beta scAAV8-hRho-mPSME2
- scAAV8-hRho-eGFP titer: 4.49xl0 13 gc/
- FIG. 10B shows expression of HSC70, PA28alpha, or PA28beta from mice injected in the left eye with lpl of the scAAV8 vectors, AAV8-hRHO-oPRE tandem mPA28alpha/beta or AAV8- CBh tandem mA28alpha/beta via sub-retinal injection. Uninjected lysate and recombinant PA28alpha and PA28beta were used as controls.
- FIGs. 11 A-l 1C are immunohistochemistry images showing localization of
- FIG. 1 IB shows PA28alpha localization in the retina of mice administered lpl of AAV8 expressing both PA28alpha and PA28beta in the tandem configuration under control of human rhodopsin promoter (hRho) with oPRE elements after the transgenes (FIG. 7D) via sub-retinal injection.
- DAPI was used as a marker of the nucleus.
- FIG. llC shows localization of PA28alpha in mice administered 1 pL of an AAV8 expressing both PA28oc and RA28b in the tandem configuration under control of CBh promoter (FIG. 7B).
- FIGs. 12A-12E are immunohistochemistry images showing localization of
- FIG. 12A shows localization of PA28alpha in the brain of mice administered a 1:1 mixture of scAAV9-hSyn-mPA28oc and scAAV9-hSyn-mPA28 .
- FIGs. 12B and 12C show localization of PA28alpha (FIG. 12B) and PA28beta (FIG.
- FIGs. 12D and 12E show localization of PA28alpha (FIG. 12D) and PA28beta (FIG. 12E) in the brain of mice administered AAV9 expressing both PA28oc and RA28b in the tandem configuration under control of CBh promoter (FIG. 7B) (Left side) or EFloc promoter (FIG. 7G).
- Dopaminergic neurons are indicated by staining of tyrosine hydroxylase (TH).
- a or “an” entity refers to one or more of that entity; for example, “a nucleic acid sequence,” is understood to represent one or more nucleic acid sequences, unless stated otherwise.
- the terms “a” (or “an”), “one or more,” and “at least one” can be used interchangeably herein.
- the term "at least" prior to a number or series of numbers is understood to include the number adjacent to the term “at least,” and all subsequent numbers or integers that could logically be included, as clear from context.
- the number of nucleotides in a nucleic acid molecule must be an integer.
- "at least 18 nucleotides of a 21 -nucleotide nucleic acid molecule” means that 18, 19, 20, or 21 nucleotides have the indicated property.
- At least is present before a series of numbers or a range, it is understood that “at least” can modify each of the numbers in the series or range.
- “At least” is also not limited to integers (e.g., "at least 5%” includes 5.0%, 5.1%, 5.18% without consideration of the number of significant figures).
- nucleic acid refers only to the primary structure of the molecule. Thus, these terms include double- and single-stranded DNA, as well as double- and single-stranded RNA.
- nucleic acid refers only to the primary structure of the molecule. Thus, these terms include double- and single-stranded DNA, as well as double- and single-stranded RNA.
- nucleic acid refers only to the primary structure of the molecule. Thus, these terms include double- and single-stranded DNA, as well as double- and single-stranded RNA.
- nucleic acid refers only to the primary structure of the molecule. Thus, these terms include double- and single-stranded DNA, as well as double- and single-stranded RNA.
- nucleic acid refers only to the primary structure of the molecule. Thus, these terms include double- and single-stranded DNA, as well as double- and single-stranded RNA.
- nucleic acid refers only to the primary structure of the molecule. Thus, these
- Polynucleotides can be made recombinantly, enzymatically, or synthetically, e.g., by solid-phase chemical synthesis followed by purification.
- sequence of the polynucleotide or nucleic acid reference is made to the sequence or order of nucleobase moieties, or modifications thereof, of the covalently linked nucleotides or nucleosides.
- mRNA refers to a single stranded RNA that encodes the amino acid sequence of one or more polypeptide chains.
- polypeptide is intended to encompass a singular
- polypeptide as well as plural “polypeptides,” and comprises any chain or chains of two or more amino acids.
- a “peptide,” a “peptide subunit,” a “protein,” an “amino acid chain,” an “amino acid sequence,” or any other term used to refer to a chain or chains of two or more amino acids are included in the definition of a “polypeptide,” even though each of these terms can have a more specific meaning.
- the term “polypeptide” can be used instead of, or interchangeably with any of these terms.
- polypeptides which have undergone post-translational or post synthesis modifications, for example, conjugation of a palmitoyl group, glycosylation, acetylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, or modification by non-naturally occurring amino acids.
- peptide encompasses full length peptides and fragments, variants or derivatives thereof.
- Percent (%) sequence identity with respect to a reference polynucleotide or polypeptide sequence is defined as the percentage of nucleic acids or amino acids in a candidate sequence that are identical to the nucleic acids or amino acids in the reference polynucleotide or polypeptide sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity. Alignment for purposes of determining percent nucleic acid or amino acid sequence identity can be achieved in various ways that are within the capabilities of one of skill in the art, for example, using publicly available computer software such as BLAST, BLAST-2, or Megalign software. Those skilled in the art can determine appropriate parameters for aligning sequences, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared. For example, percent sequence identity values can be generated using the sequence comparison computer program BLAST.
- a "coding sequence” or a sequence "encoding" a particular molecule is a nucleic acid that is transcribed (in the case of DNA) or translated (in the case of mRNA) into polypeptide, in vitro or in vivo , when operably linked to an appropriate regulatory sequence, such as a promoter.
- the boundaries of the coding sequence are determined by a start codon at the 5' (amino) terminus and a translation stop codon at the 3' (carboxy) terminus.
- a coding sequence can include, but is not limited to, cDNA from prokaryotic or eukaryotic mRNA, genomic DNA sequences from prokaryotic or eukaryotic DNA, and synthetic DNA sequences.
- a transcription termination sequence will usually be located 3' to the coding sequence.
- promoter refers to a DNA sequence recognized by the machinery of a cell, or introduced synthetic machinery, required to initiate the specific transcription of a coding sequence (e.g., a gene).
- the term “promoter” is also meant to encompass those nucleic acid elements sufficient for promoter-dependent gene expression controllable for cell-type specific, tissue-specific or inducible by external signals or agents; such elements can be located in the 5' or 3' regions of the native gene.
- the promoter is a constitutively active promoter, a cell-type specific promoter, or an inducible promoter.
- Enhancers are a cis-acting element that stimulates or inhibits transcription of adjacent genes.
- An enhancer that inhibits transcription is also referred to as a “silencer.”
- Enhancers can function (e.g., can be associated with a coding sequence) in either orientation, over distances of up to several kilobase pairs (kb) from the coding sequence and from a position downstream of a transcribed region.
- the term "regulatable promoter” is any promoter whose activity is affected by a cis or trans acting factor (e.g., an inducible promoter, such as an external signal or agent).
- the term "constitutive promoter” is any promoter that directs RNA production in many or all tissue/cell types at most times, e.g., the human CMV immediate early enhancer/promoter region that promotes constitutive expression of cloned DNA inserts in mammalian cells.
- transcriptional regulatory protein refers to a nuclear protein that binds a DNA response element and thereby transcriptionally regulates the expression of an associated gene or genes.
- Transcriptional regulatory proteins generally bind directly to a DNA response element, however in some cases binding to DNA can be indirect by way of binding to another protein that in turn binds to, or is bound to a DNA response element.
- termination signal sequence can be any genetic element that causes RNA polymerase to terminate transcription, such as for example a polyadenylation signal sequence.
- a polyadenylation signal sequence is a recognition region necessary for endonuclease cleavage of an RNA transcript that is followed by the polyadenylation consensus sequence AATAAA.
- a polyadenylation signal sequence provides a "polyA site,” i.e., a site on a RNA transcript to which adenine residues will be added by post-transcriptional polyadenylation.
- under control or "under transcriptional control” means that the promoter is in the correct location and orientation in relation to the nucleic acid to control RNA polymerase initiation and expression of the gene.
- operably linked means that a DNA sequence and a regulatory sequence(s) are connected in such a way as to permit gene expression when the appropriate molecules (e.g., transcriptional activator proteins) are bound to the regulatory sequence(s).
- operably inserted means that the DNA of interest introduced into the cell is positioned adjacent a DNA sequence which directs transcription and translation of the introduced DNA (i.e., facilitates the production of, e.g., a polypeptide encoded by a DNA of interest).
- expression cassette or “expression construct” means any type of genetic construct containing a nucleic acid in which part or all of the nucleic acid encoding sequence is capable of being transcribed.
- cistron refers to a nucleic acid cassette sufficient for expression of a gene product.
- a cistron is an expression cassette.
- some aspects of this invention provide nucleic acid constructs comprising two or more cistrons (e.g., multi cistronic, e.g., bicistronic), for example, two or more expression cassettes.
- the term "delivery vector” or “vector” refers to any vehicle for the transfer of a nucleic acid (e.g., for cloning or transfer into a host cell).
- the vector can be a plasmid, phage, transposon, cosmid, chromosome, artificial chromosome, virus, virion, etc.
- a vector can be a replicon to which another nucleic acid segment can be attached so as to bring about the replication of the attached segment.
- a “replicon” refers to any genetic element (e.g., plasmid, phage, cosmid, chromosome, virus) that functions as an autonomous unit of replication in vivo , i.e., capable of replication under its own control.
- delivery vector or “vector” includes both viral and nonviral vehicles for introducing the nucleic acid into a cell in vitro, ex vivo or in vivo.
- insertion of a polynucleotide into a suitable vector can be accomplished by ligating the appropriate polynucleotide fragments into a chosen vector that has complementary cohesive termini.
- Vectors can be engineered to encode selectable markers or reporters that provide for the selection or identification of cells that have incorporated the vector.
- the delivery vector is a viral vector (e.g., an AAV vector or lentivirus vector).
- the disclosure are directed to delivery vectors for gene therapy, which can include, e.g., viruses, particularly attenuated and/or replication-deficient viruses.
- the delivery vector of the disclosure is a viral vector selected from the group consisting of an adeno-associated viral (AAV) vector, an adenoviral vector, a lentiviral vector, or a retroviral vector.
- the deliver vector is a non-viral vector, a plasmid, a lipid-DNA micelle or liposome, or an exosome.
- AAV vector refers to any vector that comprises or derives from components of an adeno-associated vector and is suitable to infect mammalian cells, preferably human cells.
- AAV vector typically designates an AAV-type viral particle or virion comprising a payload.
- the AAV vector can be derived from various serotypes, including combinations of serotypes (i.e., "pseudotyped” AAV) or from various genomes (e.g., single stranded or self- complementary).
- the AAV vector can be replication defective and/or targeted.
- AAV adeno-associated virus
- AAV includes but is not limited to, AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAVrh8, AAV9, AAV10, AAVrhlO, and AAV.PHP.B.
- a "recombinant AAV particle” or a “rAAV particle” is an AAV virus capsid that comprises an AAV vector having at least one payload region (e.g., a polynucleotide encoding a therapeutic protein or peptide) and at least one inverted terminal repeat (ITR) region.
- payload region e.g., a polynucleotide encoding a therapeutic protein or peptide
- ITR inverted terminal repeat
- derived from refers to a component that is isolated from or made using a specified molecule or organism, or information (e.g., amino acid or nucleic acid sequence) from the specified molecule or organism.
- the derived species can be obtained by, for example, naturally occurring mutagenesis, artificial directed mutagenesis or artificial random mutagenesis.
- the mutagenesis used to derive polynucleotides can be intentionally directed or intentionally random, or a mixture of each.
- the mutagenesis of a polynucleotide to create a different polynucleotide derived from the first can be a random event (e.g., caused by polymerase infidelity) and the identification of the derived polynucleotide can be made by appropriate screening methods.
- modified refers to a changed state or structure of a molecule of the disclosure. Molecules can be modified in many ways including chemically, structurally, and functionally.
- mutation refers to any changing of the structure of a gene, resulting in a variant (also called “mutant") form that can be transmitted to subsequent generations. Mutations in a gene can be caused by the alternation of single base in DNA, or the deletion, insertion, or rearrangement of larger sections of genes or chromosomes.
- synthetic means produced, prepared, and/or manufactured by the hand of man. Synthesis of polynucleotides or polypeptides or other molecules of the present disclosure can be chemical or enzymatic.
- administration refers to the administration of a composition of the present disclosure (e.g., a gene therapy construct, vector, or composition disclosed herein) to a subject or system.
- Administration to an animal subject e.g., to a human
- contacting a cell includes contacting a cell directly or indirectly.
- contacting a cell includes contacting a cell in vitro or in vivo.
- the gene therapy construct, vector, or composition can be put into physical contact with the cell by the individual performing the method, or alternatively, the gene therapy construct, vector, or composition can be put into a situation that will permit or cause it to subsequently come into contact with the cell.
- a therapeutically effective amount of an agent refers to a quantity sufficient to, when administered to the subject, including a human, effect beneficial or desired results, including clinical results, and, as such, an "effective amount” or synonym thereto depends on the context in which it is being applied.
- a therapeutically effective amount of an agent is an amount that results in a beneficial or desired result in a subject as compared to a control.
- the amount of a given agent (e.g., a gene therapy construct, vector, or composition disclosed herein) will correspond to such an amount will vary depending upon various factors, such as the given agent, the pharmaceutical formulation, the route of administration, the type of disease or disorder, the identity of the subject (e.g., age, sex, and/or weight) or host being treated, and the like.
- gene therapy is the insertion of nucleic acid sequences
- Gene therapy also includes insertion of transgene that are activating in nature, i.e., that activate, increase or cause expression, activity or function of an endogenous gene or protein, such as a desirable or gene or protein.
- transgenes can be exogenous.
- An exogenous molecule or sequence is understood to be molecule or sequence not normally occurring in the cell, tissue and/or individual to be treated. Both acquired and congenital diseases are amenable to gene therapy.
- prophylactically effective amount includes the amount of an agent, (e.g., a gene therapy construct, vector or composition disclosed herein) that, when administered to a subject having or predisposed to have a disease or disorder (e.g., a neurological disease or disorder (e.g., Parkinson’s Disease (PD) or Huntington’s Disease (HD) or ocular diseases or disorders (e.g., retinitis pigmentosa)) is sufficient to prevent, reduce the symptoms of, or ameliorate the disease or disorder or one or more symptoms of the disease or disorder.
- a disease or disorder e.g., a neurological disease or disorder (e.g., Parkinson’s Disease (PD) or Huntington’s Disease (HD) or ocular diseases or disorders (e.g., retinitis pigmentosa)
- Ameliorating the disease or disorder includes slowing the course of the disease or disorder or reducing the severity of later-developing disease or disorder.
- the “prophylactically effective amount” can vary depending on the characteristics of the agent, how the agent is administered, the degree of risk of disease, and the history, age, weight, family history, genetic makeup, the types of preceding or concomitant treatments, if any, and other individual characteristics of the patient to be treated.
- in vitro refers to events that occur in an artificial environment, e.g., in a test tube or reaction vessel, in cell culture, in a Petri dish, etc., rather than within an organism (e.g., animal, plant, or microbe).
- in vivo refers to events that occur within an organism
- transfection refers to methods to introduce exogenous nucleic acids into a cell. Methods of transfection include, but are not limited to, chemical methods, physical treatments and cationic lipids or mixtures.
- level is meant a level or activity of a protein, or mRNA encoding the protein, optionally as compared to a reference.
- the reference can be any useful reference, as defined herein.
- a “decreased level” or an “increased level” of a protein or expression is meant a decrease or increase in protein or expression level, as compared to a reference.
- a “reference” is meant any useful reference used to compare protein or mRNA levels or activity.
- the reference can be any sample, standard, standard curve, or level that is used for comparison purposes.
- the reference can be a normal reference sample or a reference standard or level.
- composition represents a composition comprising a therapeutic agent disclosed herein, e.g., a gene therapy construct, vector or composition disclosed herein, formulated with a pharmaceutically acceptable excipient, and can be manufactured or sold with the approval of a governmental regulatory agency as part of a therapeutic regimen for the treatment of disease in a mammal.
- a "pharmaceutically acceptable excipient,” as used herein, refers to any ingredient other than the therapeutic agent described herein (for example, a vehicle capable of suspending an active compound) and having the properties of being substantially nontoxic and non-inflammatory in a patient.
- the term "subject” refers to any organism to which an agent or composition disclosed herein can be administered, e.g., for experimental, diagnostic, prophylactic, and/or therapeutic purposes. Typical subjects include any animal (e.g., mammals such as mice, rats, rabbits, non-human primates, and humans). A subject can seek or be in need of treatment, require treatment, be receiving treatment, be receiving treatment in the future, or be a human or animal who is under care by a trained professional for a particular disease or condition.
- animal e.g., mammals such as mice, rats, rabbits, non-human primates, and humans.
- a subject can seek or be in need of treatment, require treatment, be receiving treatment, be receiving treatment in the future, or be a human or animal who is under care by a trained professional for a particular disease or condition.
- the terms “treat,” “treated,” and “treating” mean both therapeutic treatment and prophylactic or preventative measures wherein the object is to prevent or slow down (lessen) an undesired physiological condition, disorder, or disease, or obtain beneficial or desired clinical results.
- treating reduces or lessens the symptoms associated with a disease or disorder.
- the treating results in a beneficial or desired clinical result.
- Beneficial or desired clinical results include, but are not limited to, alleviation of symptoms; diminishment of the extent of a condition, disorder, or disease; stabilized (i.e., not worsening) state of condition, disorder, or disease; delay in onset or slowing of condition, disorder, or disease progression; amelioration of the condition, disorder, or disease state or remission (whether partial or total), whether detectable or undetectable; an amelioration of at least one measurable physical parameter, not necessarily discernible by the patient; or enhancement or improvement of condition, disorder, or disease.
- treatment includes eliciting a clinically significant response without excessive levels of side effects.
- treatment includes prolonging survival as compared to expected survival if not receiving treatment.
- the term “amelioration” or “ameliorating” refers to a lessening of severity of at least one indicator of a condition or disease.
- the term “preventing” or “prevention” refers to delaying or forestalling the onset, development or progression of a condition or disease for a period of time, including weeks, months, or years.
- constructs comprising a polynucleotide comprising a promoter operably linked to a nucleic acid encoding a PSME (e.g., a PA28A alone, PA28B, or a PA28A in combination with PA28B).
- PSME e.g., a PA28A alone, PA28B, or a PA28A in combination with PA28B.
- Proteasomes are multi-subunit complexes assembled upon association of two principal components: the 20S core and the regulatory caps.
- the 20S core is responsible for the entire proteolytic process however, its basal activity is very low in the absence of regulatory caps.
- the most common caps are 19S and 1 IS, which facilitate protein degradation in ubiquitin-dependent and ubiquitin-independent manners, respectively.
- a single 20S core may associate with one or two caps (either identical or different) at the sites located at the opposite ends of the 20S barrel-like structure. Binding of the 1 IS cap to the ends of the 20S core facilitates substrate access and proteolytic activities confined inside the 20S barrel-like structure.
- the 1 IS cap can be formed by a hetero-heptameric ring consisting of PA28A and PA28B (also known as the PA28 complex). However, functional 1 IS caps can also be formed by PA28A alone.
- misfolded proteins can accumulate when the proteasome is impaired and/or when the demand for proteasome function surpasses the functional capacity of proteasomes. Such accumulation leads to proteinopathies.
- transgene delivery of a PA28A alone or a combination of a PA28A and PA28B to a cell or tissue can increase proteasome capacity and thereby reduce the amount of aberrant protein aggregation in the targeted cell or tissue.
- delivery of the constructs disclosed herein can be useful for treating proteinopathies (e.g., Huntington’s Disease, Parkinson’s Disease, ocular diseases, and cardiac proteinopathies).
- a polynucleotide encoding a PSME e.g., a PA28A alone,
- PA28B or a PA28A in combination with PA28B
- a viral vector e.g., an AAV vector
- Proteasome activator complex subunit 1 generally abbreviated PA28A or PSME1 herein (also known as 1 IS regulator complex subunit alpha, REG-alpha, activator of multi catalytic protease subunit 1, interferon gamma up-regulated 1-5111 protein, Proteasome activator 28 subunit alpha, P28a, and P28alpha) is a protein which in humans is encoded by the PSME1 gene.
- Th ePSMEl gene is located on chromosome 14 (bases 24,136,194 to 24,138,962;
- PA28A has three isoforms produced by alternative splicing.
- the wild-type amino acid sequences for the PA28A isoforms are shown in Table 1 below.
- the last tyrosine (Y) residue of isoform 1 can be mutated to tryptophan (W), which has been shown to have some increased activity in some in vitro assays (see Song, X. et al, JBC 272(44):P27994-28000 (1997)).
- the PA28A comprises an amino acid sequence having a sequence of at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity to SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, or SEQ ID NO: 4.
- the PA28A comprises an amino acid sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, or SEQ ID NO: 4.
- the nucleic acid encoding PA28A comprises a sequence having at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity to SEQ ID NO: 5, SEQ ID NO: 6, or SEQ ID NO: 7.
- the nucleic acid encoding PA28A comprises SEQ ID NO: 5, SEQ ID NO: 6, or SEQ ID NO: 7.
- the wild-type nucleic acid sequences can be modified to improve expression, manufacturability, and/or reduce immunologic risk.
- the PA28A nucleotide sequences are shown in Table 2 below.
- Proteasome activator complex subunit 2 generally abbreviated PA28B or PSME2 herein (also known as 1 IS regulator complex subunit beta, REG-beta, Activator of multi catalytic protease subunit 2, Proteasome activator 28 subunit beta, PA28b, and PA28beta) is a protein which in humans is encoded by the PSME2 gene.
- the PSME2 gene is located on chromosome 14 (bases 24,143,365 to 24,146,646; NCBI Reference Sequence NC_000014.9).
- the PA28B comprises an amino acid sequence having a sequence of at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity to SEQ ID NO: 8.
- the PA28A comprises an amino acid sequence of SEQ ID NO: 8.
- the nucleic acid encoding PA28B comprises a sequence having at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity to SEQ ID NO: 9.
- the nucleic acid encoding PA28B comprises SEQ ID NO: 9.
- the wild-type nucleic acid sequence can be modified to improve expression, manufacturability, and/or reduce immunologic risk.
- the nucleotide sequence encoding PA28B is shown in Table 4 below.
- nucleic acid construct or an expression construct having a promoter operably linked to a DNA of interest that encodes a PSME disclosed herein (e.g., a PA28A alone or a PA28A in combination with PA28B, with or without a second promoter).
- the constructs comprise a DNA sequence (or the corresponding RNA sequence) can be used in accordance with the disclosure and can be any eukaryotic expression construct containing a DNA or RNA of interest.
- a plasmid or viral construct e.g., an AAV vector
- termini are bound to exogenous DNA having complementary, like ligatable termini to provide a biologically functional DNA molecule having an intact replicon and a desired phenotypic property.
- the construct is capable of replication in both eukaryotic and prokaryotic hosts.
- the constructs disclosed herein contains a promoter to facilitate expression of the DNA of interest within a certain cell type and/or tissue.
- the promoter is a heterologous promoter.
- heterologous promoter refers to a promoter that does is not found to be operatively linked to a given encoding sequence in nature.
- the promoter is a ubiquitous promoter or a tissue specific promoter.
- the promoter is selected from a MecP2 promoter, a GUSB promoter, a CBh promoter, a PGK promoter, a human rhodopsin (hRho) kinase promoter, a cone arrestin promoter, a tyrosine hydroxylase promoter, a dopamine transporter promoter, a GRK1 promoter, a cardiac myosin light chain 2 promoter, a CAMKII promoter, a hDlx promoter, a CMV promoter, a CAG promoter, a bidirectional CAG promoter, a CBA promoter, a human ubiquitin C promoter, synapsin (Syn) promoter, a human synapsin (hSyn) promoter, a EFla promoter, or a rhodopsin (Rhod).
- a MecP2 promoter a GUSB promoter, a CBh promote
- the promoter is a rhodopsin (Rhod) promoter. In some aspects, the promoter is a human rhodopsin (hRho) promoter. In some aspects, the promoter is synapsin (Syn) promoter. In some aspects, the promoter is a human synapsin (hSyn) promoter. In some aspects, the promoter is selected from a hSyn or a pRhod promoter. In some aspects, the promoter is a EFla promoter. In some aspects, the promoter is a CBh promoter.
- the human synapsin promoter has at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or 100% identity to SEQ ID NO: 16. In some aspects, the human synapsin promoter has the sequence of SEQ ID NO: 16.
- the human rhodopsin promoter has at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or 100% identity to SEQ ID NO: 13. In some aspects, the human rhodopsin promoter has the sequence of SEQ ID NO: 13.
- the EFla promoter has at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or 100% identity to SEQ ID NO: 17. In some aspects, the EFla promoter has the sequence of SEQ ID NO: 17.
- the CBh promoter has at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or 100% identity to SEQ ID NO: 18. In some aspects, the CBh promoter has the sequence of SEQ ID NO: 18.
- the nucleic acid construct or an expression construct can further comprise one or more additional elements, for example, an enhancer, a polyadenylation site, a woodchuck hepatitis virus post- transcriptional regulatory element (WPRE), an optimized woodchuck hepatitis virus post- transcriptional regulatory element (oPRE), an intron, a microRNA binding motif, and/or other elements known to affect expression levels of the encoding sequence.
- WPRE woodchuck hepatitis virus post- transcriptional regulatory element
- oPRE optimized woodchuck hepatitis virus post- transcriptional regulatory element
- the nucleic acid construct or expression construct comprises a CMV enhancer.
- the oPRE has at least 50%, at least 55%, at least 60%, at least
- the oPRE has the sequence of SEQ ID NO: 15.
- the CMV enhancer has at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or 100% identity to SEQ ID NO: 19. In some aspects, the CMV enhancer has the sequence of SEQ ID NO: 19.
- the nucleic acid construct comprises a chimeric intron. In some aspects, the nucleic acid construct comprises a beta-globin intron. In some aspects, the nucleic acid construct comprises minute virus of mice (MVM) intron.
- VMM minute virus of mice
- the nucleic acid construct comprises a polyA sequence selected from the group consisting of a beta-globin polyA, a bovine growth hormone polyA (bGH polyA), a human growth hormone polyA (hGH polyA), and a SV40 immediate early poly A (SV40 polyA).
- the nucleic acid construct or expression construct comprises a bovine growth hormone polyA (bGH polyA).
- the bovine growth hormone polyA (bGH poly A) has at least
- bovine growth hormone polyA has the sequence of SEQ ID NO: 14.
- the nucleic acid construct comprises a 5’ and/or a 3’ untranslated region (UTR). In some aspects, the nucleic acid construct can further comprise a microRNA binding motif in the 3’ untranslated region (UTR).
- the nucleic acid construct or an expression construct e.g., comprising an expression cassette
- WPRE woodchuck hepatitis virus post-transcriptional regulatory element
- the nucleic acid construct of an expression construct comprises an optimized woodchuck hepatitis virus post- transcriptional regulatory element (oPRE).
- the nucleic acid construct or an expression construct comprises a polynucleotide comprising a promoter (e.g., hSyn or pRhod promoter), a nucleic acid encoding a PSME (e.g., a PA28A or PA28A in combination with PA28B), and a WPRE.
- a promoter e.g., hSyn or pRhod promoter
- a nucleic acid encoding a PSME e.g., a PA28A or PA28A in combination with PA28B
- WPRE WPRE
- the nucleic acid construct or an expression construct comprises a polynucleotide comprising a promoter (e.g., hSyn or pRhod promoter), a nucleic acid encoding a PSME (e.g., a PA28A or PA28A in combination with PA28B), and an optimized woodchuck hepatitis virus post- transcriptional regulatory element (oPRE).
- a promoter e.g., hSyn or pRhod promoter
- a nucleic acid encoding a PSME e.g., a PA28A or PA28A in combination with PA28B
- oPRE optimized woodchuck hepatitis virus post- transcriptional regulatory element
- the disclosure provides nucleic acids encoding a PSME (e.g., a
- PA28A or PA28A in combination with PA28B that is a multi cistronic (e.g., bicistronic) construct (e.g., comprising PA28A and PA28B).
- the multi cistronic (e.g., bicistronic) construct comprises a bi-directional promoter.
- the multi cistronic (e.g., bicistronic) construct comprises tandem expression cassettes each with a promoter, a transgene (encoding PA28A or PA28B), and polyA.
- the multi cistronic (e.g., bicistronic) construct further comprises a cleavage element between the nucleic acid encoding a PA28A and the nucleic acid encoding PA28B.
- the multicistronic (e.g., bicistronic) construct further comprises a cleavage element between an expression construct comprising a nucleic acid encoding a PA28A and an expression construct comprising the nucleic acid encoding PA28B.
- the cleavage element comprises a nucleic acid encoding self-cleavage peptide.
- the cleavage peptide is a 2A self-cleaving peptide (P2A, T2A, etc.).
- the cleavage site is a furin cleavage site, e.g., with the consensus sequence of RXK(R)R (SEQ ID NO: 10).
- the cleavage site is an intein-F2A peptide. See Zhang et al., Plant Biotechnol J. (2017) 15, pp.718-728.
- the multi cistronic (e.g., bicistronic) construct comprises first expression cassette comprising a polynucleotide encoding PA28A and a second expression cassette comprising a polynucleotide encoding PA28B. See, e.g., Kugler et al., Virology 311 (2003) 89-95.
- the first and second expression cassettes comprise the same promoter.
- the first and second expression cassettes comprise different promoters.
- the first expression cassette comprises a hSyn promoter and the second expression cassette comprises a CMV promoter.
- the first expression cassette comprises a CMV promoter and the second expression cassette comprises a hSyn promoter.
- the first and/or second expression cassettes comprise a polyA (e.g., bGH polyadenylation site, hGH polyadenylation site, or a SV40 polyadenylation site).
- the first and/or second expression cassette can further comprise a WPRE.
- the first and/or second expression cassette can further comprise an optimized WPRE (oPRE).
- the construct is a bicistronic nucleic acid construct comprising a first nucleic acid encoding a PA28A and a second nucleic acid encoding PA28B.
- the construct is a bicistronic nucleic acid construct comprising a first nucleic acid encoding a PA28A, a second nucleic acid encoding PA28B, and a nucleic acid encoding a cleavage peptide located between the first and second nucleic acid.
- the bicistronic construct further comprises a WPRE.
- the bicistronic construct further comprises an optimized WPRE (oPRE).
- the construct is a bicistronic nucleic acid construct comprising a first nucleic acid encoding a PA28A, a second nucleic acid encoding PA28B, a nucleic acid encoding a cleavage peptide located between the first and second nucleic acid, and a WPRE.
- the construct is a bicistronic nucleic acid construct comprising a first nucleic acid encoding a PA28A, a second nucleic acid encoding PA28B, a nucleic acid encoding a cleavage peptide located between the first and second nucleic acid, and an optimized WPRE (oPRE).
- oPRE optimized WPRE
- bicistronic expression constructs are provided in which the expression cassettes are positioned in different ways.
- a multi cistronic expression construct is provided in which a first expression cassette is positioned adjacent to a second expression cassette.
- the first expression cassette and the second expression cassette are operably linked by a bidirectional promoter, wherein the first expression cassette and the second expression cassette are flanked by adeno-associated virus (AAV) inverted terminal repeats (ITRs).
- AAV adeno-associated virus
- bicistronic expression constructs are provided in which the expression cassettes are oriented in different ways.
- bicistronic expression construct is provided comprising a first and a second expression cassette in opposite orientations.
- bicistronic expression constructs are provided allowing for efficient expression of two or more encoding nucleic acid sequences.
- the bicistronic expression construct comprises two expression cassettes.
- the bicistronic expression construct provided is a recombinant AAV (rAAV) construct.
- PA28A and PA28B are expressed in a single open reading frame.
- an amino acid sequence or linker containing a high efficiency cleavage site is disposed between the PA28A and PA28B.
- high cleavage efficiency is defined as more than 50%, more than 70%, more than 80%, or more than 90% of the translated protein is cleaved. Cleavage efficiency can be measured by Western Blot analysis.
- the expression construct disclosed herein encodes PA28A- cleavage sequence-PA28B in 5'-3' orientation. In some aspects, the expression construct encodes PA28B-cleavage sequence-PA28A in 5'-3' orientation. In some aspects, the nucleic acid sequence of the construct comprises a promoter, PA28A coding sequence, a cleavage sequence, and a PA28B coding sequence in 5'-3' orientation. In some aspects, the nucleic acid sequence of the construct comprises a promoter, a PA28B coding sequence, cleavage sequence, and aPA28A coding sequence in 5'-3' orientation. In some aspects the cleavage sequence a P2A, a furin, or an intein-F2A cleavage sequence.
- the expression construct disclosed herein encodes PA28A-P2A-
- the expression construct encodes PA28B- P2A-PA28A in 5'-3' orientation.
- the nucleic acid sequence of the construct comprises a promoter, a PA28A coding sequence, a P2A sequence, and a PA28B coding sequence in 5'-3' orientation.
- the nucleic acid sequence of the construct comprises a promoter, a PA28B coding sequence, a P2A sequence, and a PA28A coding sequence in 5'-3' orientation.
- the promoter is a bi directional promoter.
- the promoter is selected from a MecP2 promoter, a GUSB promoter, a CBh promoter, a PGK promoter, a EFla promoter, a human rhodopsin (hRho) kinase promoter, a cone arrestin promoter, a tyrosine hydroxylase promoter, a dopamine transporter promoter, a GRK1 promoter, a cardiac myosin light chain 2 promoter, a CAMKII promoter, a hDlx promoter, a CMV promoter, a CAG promoter, a bidirectional CAG promoter, a CBA promoter, a human ubiquitin C promoter, a human synapsin (hSyn) promoter, synapsin (Syn) promoter, or a rhodopsin (Rhod).
- a MecP2 promoter a GUSB promoter, a CBh promote
- the expression construct comprises a synapsin promoter. In some aspects, the expression construct comprises a rhodopsin promoter. In some aspects, the expression construct comprises a human rhodopsin promoter. In some aspects, the expression construct comprises a human synapsin (hSyn) promoter. In some aspects, the expression construct comprises a CBh promoter. In some aspects, the expression construct comprises a EFla promoter.
- the expression construct further comprises an enhancer, a polyadenylation site, a woodchuck hepatitis virus post-transcriptional regulatory element (WPRE), an optimized woodchuck hepatitis virus post-transcriptional regulatory element (oPRE), an intron, a microRNA binding motif, and/or other element(s) known to affect expression levels of the encoding sequence.
- WPRE woodchuck hepatitis virus post-transcriptional regulatory element
- oPRE optimized woodchuck hepatitis virus post-transcriptional regulatory element
- an intron a microRNA binding motif, and/or other element(s) known to affect expression levels of the encoding sequence.
- the enhancer is a CMV enhancer.
- the nucleic acid construct comprises a chimeric intron. In some aspects, the nucleic acid construct comprises a beta-globin intron. In some aspects, the nucleic acid construct comprises minute virus of mice (MVM) intron.
- VMM minute virus of mice
- the nucleic acid construct comprises polyA sequence selected from the group consisting of beta-globin polyA, bovine growth hormone polyA (bGH polyA), human growth hormone polyA (hGH polyA), and SV40 immediate early poly A (SV40 polyA). In some aspects, the nucleic acid construct comprises bovine growth hormone polyA (bGH polyA).
- the nucleic acid construct comprises a 5’ and/or a 3’ untranslated region (UTR). In some aspects, the nucleic acid construct comprises a microRNA binding motif in the 3’ untranslated region (UTR).
- the expression construct further comprises a WPRE. In some aspects, the expression construct further comprises an optimized WPRE (oPRE).
- oPRE optimized WPRE
- the constructs of the disclosure can also include other components such as a marker (e.g., an antibiotic resistance gene (such as an ampicillin resistance gene) or b-galactosidase) to aid in selection of cells containing and/or expressing the construct, an origin of replication for stable replication of the construct in a bacterial cell (preferably, a high copy number origin of replication), a nuclear localization signal, or other elements which facilitate production of the DNA construct, the protein encoded thereby, or both.
- a marker e.g., an antibiotic resistance gene (such as an ampicillin resistance gene) or b-galactosidase
- an origin of replication for stable replication of the construct in a bacterial cell preferably, a high copy number origin of replication
- a nuclear localization signal e.g., a nuclear localization signal, or other elements which facilitate production of the DNA construct, the protein encoded thereby, or both.
- the construct can comprise at a minimum a eukaryotic promoter operably linked to a DNA of interest, which is in turn operably linked to a polyadenylation sequence (poly(A)).
- the polyadenylation signal sequence can be selected from any of a variety of polyadenylation signal sequences known in the art.
- the construct e.g., expression construct
- the construct further comprises a polyA sequence selected from the group consisting of a beta-globin polyA, a bovine growth hormone polyA (bGH polyA), a human growth hormone polyA (hGH polyA), and a SV40 immediate early poly A (SV40 polyA).
- the polyA sequence comprises a bovine growth hormone polyA.
- the polyA sequence comprises a SV40 immediate early polyA.
- the polyA comprises b-globin polyA.
- the polyA comprises hGH polyA.
- the constructs disclosed herein are packaged in a delivery vector, e.g., a viral vector, comprising the nucleic acid construct or an expression construct comprising a nucleic acid encoding a PA28A, PA28B, or a combination thereof.
- a delivery vector e.g., a viral vector
- the delivery vector is suitable for delivery to a specific tissue.
- the tissue is selected from brain, eye, or heart muscle.
- the viral vector comprises a construct comprising a polynucleotide comprising a promoter sequence and a nucleic acid sequence encoding Proteasome activator complex subunit 1 (PA28A).
- PA28A Proteasome activator complex subunit 1
- the construct comprises a hSyn promoter, a hRho promoter, or a pRhod promoter.
- the construct comprises a CBh promoter.
- the construct comprises a EFla promoter.
- the polynucleotide further comprises a WPRE.
- the polynucleotide further comprises an optimized WPRE (oPRE).
- the nucleic acid construct or an expression construct comprising a promoter sequence (e.g., hSyn or pRod), a nucleic acid sequence encoding Proteasome activator complex subunit 1 (PA28A), and optionally a WPRE.
- the nucleic acid construct or an expression construct comprising an expression cassette comprising a promoter sequence (e.g., hSyn or pRod), a nucleic acid sequence encoding Proteasome activator complex subunit 1 (PA28A), and optionally an optimized WPRE (oPRE).
- the nucleic acid construct or an expression construct further comprises a nucleic acid sequence encoding Proteasome activator complex subunit 2 (PA28B).
- PA28B Proteasome activator complex subunit 2
- the polynucleotide comprises a promoter sequence, a nucleic acid encoding PA28A, a proteolytic cleavage site, and a nucleic acid encoding PA28B.
- the polynucleotide comprises a promoter sequence, a nucleic acid encoding PA28A, a proteolytic cleavage site, and a nucleic acid encoding PA28B in 5'-3' orientation. In some aspects, wherein the polynucleotide comprises a promoter sequence, a nucleic acid encoding PA28B, a proteolytic cleavage site, and a nucleic acid encoding PA28A in 5'-3' orientation. In some aspects, the proteolytic cleavage site is P2A, furin, or intein-F2A. In some aspects, the promoter is hSyn, hRho, or pRhod.
- the construct comprises a CBh promoter. In some aspects, the construct comprises a EFla promoter. In some aspects, the polynucleotide further comprises a WPRE. In some aspects, the polynucleotide further comprises an optimized WPRE (oPRE). In some aspects, the nucleic acid construct or an expression construct (e.g., comprising an expression cassette) further comprises an ITR (e.g., AAV2 ITRs).
- an ITR e.g., AAV2 ITRs
- nucleic acid construct or an expression construct e.g., comprising an expression cassette
- an expression construct e.g., comprising an expression cassette
- exogenous (i.e., donor) DNA used in the disclosure can be obtained from suitable cells, and the constructs prepared using techniques well known in the art.
- techniques for obtaining expression of exogenous DNA or RNA sequences in a genetically altered host cell are known in the art (see e.g., Kormal et al., Proc. Natl. Acad. Sci. USA, 84:2150-2154 (1987); Sambrook et al. Molecular Cloning: a Laboratory Manual, 2nd Ed., 1989, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.; each of which are hereby incorporated by reference with respect to methods and compositions for eukaryotic expression of a DNA of interest).
- Certain aspects of the disclosure are directed to delivery vectors comprising the nucleic acid construct or an expression construct (e.g., comprising an expression cassette), or gene therapy constructs disclosed herein.
- the delivery vector is a viral vector.
- the delivery vector (e.g., a viral vector) comprises a nucleic acid encoding PA28A disclosed herein. In some aspects, the delivery vector (e.g., a viral vector) comprises a nucleic acid encoding PA28A in combination with PA28B disclosed herein.
- the promoter is selected from a MecP2 promoter, a GUSB promoter, a CBh promoter, a PGK promoter, a human synapsin (Syn) promoter, a human rhodopsin (Rhod) kinase promoter, a cone arrestin promoter, a tyrosine hydroxylase promoter, a dopamine transporter promoter, a GRK1 promoter, a cardiac myosin light chain 2 promoter, a CAMKII promoter, a hDlx promoter, a CMV promoter, a CAG promoter, a bidirectional CAG promoter, a CBA promoter, an EFla promoter, or a human ubiquitin C promoter.
- the delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a polyadenylation site, a woodchuck hepatitis virus post-transcriptional regulatory element (WPRE), an optimized woodchuck hepatitis virus post-transcriptional regulatory element (oPRE), an intron, a microRNA binding motif, or any combination thereof.
- WPRE woodchuck hepatitis virus post-transcriptional regulatory element
- oPRE optimized woodchuck hepatitis virus post-transcriptional regulatory element
- the delivery vector comprises an expression construct comprising a first expression cassette comprising a polynucleotide encoding PA28A and a second expression cassette comprising a polynucleotide encoding PA28B. See Kugler et al., Virology 311 (2003) 89-95.
- the first and second expression cassettes comprise the same promoter. In some aspects, the first and second expression cassettes comprise different promoters. In some aspects, the first expression cassette comprises a hSyn promoter and the second expression cassette comprises a CMV promoter. In some aspects, the first expression cassette comprises a CMV promoter and the second expression cassette comprises a hSyn promoter. In some aspects, the first and/or second expression cassettes comprise a polyA (e.g., bGH polyadenylation site, hGH polyadenylation site, or a SV40 polyadenylation site). In some aspects, the first and/or second expression cassette can further comprise a WPRE. In some aspects, the first and/or second expression cassette can further comprise an optimized WPRE (oPRE).
- oPRE optimized WPRE
- the delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a chimeric intron. In some aspects, the delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a beta-globin intron. In some aspects, the delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising minute virus of mice (MVM) intron.
- VMM minute virus of mice
- the delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a polyA sequence selected from the group consisting of a beta-globin polyA, a bovine growth hormone polyA (bGH polyA), a human growth hormone polyA (hGH polyA), and a SV40 immediate early poly A (SV40 polyA).
- a polyA sequence selected from the group consisting of a beta-globin polyA, a bovine growth hormone polyA (bGH polyA), a human growth hormone polyA (hGH polyA), and a SV40 immediate early poly A (SV40 polyA).
- the polyA is a bovine growth hormone polyA.
- the delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a 5’ and/or a 3’ untranslated region (UTR).
- the delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprises a microRNA binding motif in the 3’ untranslated region (UTR).
- the delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising comprises a WPRE.
- the delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising comprises an optimized WPRE (oPRE).
- the delivery vector comprises the nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a promoter sequence (e.g., hSyn or pRod), a nucleic acid sequence encoding Proteasome activator complex subunit 1 (PA28A), and optionally a WPRE.
- the delivery vector comprises the nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a promoter sequence (e.g., hSyn or pRod), a nucleic acid sequence encoding Proteasome activator complex subunit 1 (PA28A), and optionally an optimized WPRE (oPRE).
- the delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) further comprising a nucleic acid sequence encoding Proteasome activator complex subunit 2 (PA28B).
- the delivery vector comprises a polynucleotide comprising a promoter sequence, a nucleic acid encoding PA28A, a proteolytic cleavage site, and a nucleic acid encoding PA28B.
- delivery vector comprises a polynucleotide comprising a promoter sequence, a nucleic acid encoding PA28A, a proteolytic cleavage site, and a nucleic acid encoding PA28B in 5'-3' orientation.
- the delivery vector comprises a polynucleotide comprising a promoter sequence, a nucleic acid encoding PA28B, a proteolytic cleavage site, and a nucleic acid encoding PA28A in 5'-3' orientation.
- the cleavage sequence a P2A, a furin, or an intein-F2A cleavage sequence.
- the proteolytic cleavage site is P2A.
- the promoter is hSyn or pRhod.
- the polynucleotide further comprises a WPRE.
- the polynucleotide further comprises an optimized WPRE (oPRE).
- the delivery vector is suitable for tissue specific delivery (e.g., to the brain, eye, or heart muscle.
- viral delivery vectors used in accordance with the disclosure can be composed of a viral particle derived from a naturally-occurring virus which has been genetically altered to render the virus replication-defective and to express a recombinant gene of interest in accordance with the disclosure. Once the virus delivers its genetic material to a cell, it does not generate additional infectious virus but does introduce exogenous recombinant genes into the cell, preferably into the genome of the cell.
- the delivery vector e.g., viral vector
- AAV adeno-associated viral
- HSV herpes simplex virus
- CMV cytomegalovirus
- vaccinia vaccinia
- poliovirus vectors e.g., retrovirus, adenovirus, adeno-associated virus (AAV), herpes simplex virus (HSV), cytomegalovirus (CMV), vaccinia and poliovirus vectors.
- the delivery vector e.g., viral vector
- AAV adeno-associated viral vector
- adenoviral vector adenoviral vector
- lentiviral vector a lentiviral vector
- retroviral vector e.g., retroviral vector
- the production of infective virus particles containing either DNA or RNA corresponding to the DNA of interest can be produced by introducing the viral construct into a recombinant cell line which provides the missing components essential for viral replication.
- transformation of the recombinant cell line with the recombinant viral vector will not result in production of replication-competent viruses, e.g., by homologous recombination of the viral sequences of the recombinant cell line into the introduced viral vector.
- nucleic acid of the present disclosure after delivery to or integration in the genomic DNA of a target cell can require a specific promoter, including but not limited to, a promoter that is species specific, inducible, tissue-specific, or cell cycle- specific (Parr et al., Nat. Med.3: 1145-9 (1997); the contents of which are herein incorporated by reference in their entirety).
- a specific promoter including but not limited to, a promoter that is species specific, inducible, tissue-specific, or cell cycle- specific (Parr et al., Nat. Med.3: 1145-9 (1997); the contents of which are herein incorporated by reference in their entirety).
- AAV Adeno-Associated Virus
- isolated adeno-associated viruses comprising a payload, e.g., the nucleic acid construct or an expression construct (e.g., comprising an expression cassette), or gene therapy constructs disclosed herein.
- AAVs adeno-associated viruses
- the term “isolated” refers to an AAV that has been artificially produced or obtained. Isolated AAVs may be produced using recombinant methods. Such AAVs are referred to herein as “recombinant AAVs”.
- Recombinant AAVs preferably have tissue-specific targeting capabilities, such that a nuclease and/or transgene of the rAAV will be delivered specifically to one or more predetermined tissue(s).
- the AAV capsid is an important element in determining these tissue-specific targeting capabilities.
- a rAAV having a capsid appropriate for the tissue being targeted can be selected.
- capsid proteins are structural proteins encoded by the cap gene of an AAV.
- AAVs comprise three capsid proteins, virion proteins 1 to 3 (named VP1, VP2 and VP3), all of which are transcribed from a single cap gene via alternative splicing.
- the molecular weights of VP1, VP2 and VP3 are respectively about 87 kDa, about 72 kDa and about 62 kDa.
- capsid proteins upon translation, form a spherical 60-mer protein shell around the viral genome.
- the functions of the capsid proteins are to protect the viral genome, deliver the genome and interact with the host.
- capsid proteins deliver the viral genome to a host in a tissue specific manner.
- an AAV capsid protein is of an AAV serotype selected from the group consisting of AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAVrh8, AAV9, AAV10, AAVrhlO, and AAV.PHP.B.
- the AAV capsid protein is a variant of AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAVrh8, AAV9, AAV10, AAVrhlO, and AAV.PHP.B.
- the variant comprises sequence modifications resulting from, but not limited to, capsid evolution, selection, shuffling, or directed mutagenesis.
- AAV vector can be derived from various serotypes, including combinations of serotypes (i.e., "pseudotyped” AAV) or from various genomes (e.g., single stranded or self-complementary).
- an AAV capsid protein is of a serotype derived from a non-human primate, for example AAVrh8 serotype.
- an AAV capsid protein is of a serotype derived for CNS transduction, for example AAV.PHP.B.
- the capsid protein is of AAV serotype 9.
- the capsid protein is of AAV serotype 2.
- the capsid protein is of AAV serotype 8.
- the components to be cultured in the host cell to package a rAAV vector in an AAV capsid can be provided to the host cell in trans.
- any one or more of the required components e.g., recombinant AAV vector, rep sequences, cap sequences, and/or helper functions
- a stable host cell which has been engineered to contain one or more of the required components using methods known to those of skill in the art.
- a stable host cell will contain the required component(s) under the control of an inducible promoter.
- the required component(s) may be under the control of a constitutive promoter.
- a selected stable host cell may contain selected component(s) under the control of a constitutive promoter and other selected component s) under the control of one or more inducible promoters.
- a stable host cell can be generated which is derived from 293 cells (which contain El helper functions under the control of a constitutive promoter), but which contain the rep and/or cap proteins under the control of inducible promoters. Still other stable host cells can be generated by one of skill in the art.
- recombinant AAVs may be produced using the triple transfection method (described in detail in U.S. Pat. No. 6,001,650).
- the recombinant AAVs are produced by transfecting a host cell with an AAV vector (comprising a transgene flanked by ITR elements) to be packaged into AAV particles, an AAV helper function vector, and an accessory function vector.
- An AAV helper function vector encodes the “AAV helper function” sequences (e.g., rep and cap), which function in trans for productive AAV replication and encapsidation.
- the AAV helper function vector supports efficient AAV vector production without generating any detectable wild-type AAV virions (e.g., AAV virions containing functional rep and cap genes).
- AAV virions e.g., AAV virions containing functional rep and cap genes.
- vectors suitable for use with the present disclosure include pHLP19, described in U.S. Pat. No. 6,001,650 and pRep6cap6 vector, described in U.S. Pat. No. 6,156,303, the entirety of both incorporated by reference herein.
- the accessory function vector encodes nucleotide sequences for non-AAV derived viral and/or cellular functions upon which AAV is dependent for replication (e.g., “accessory functions”).
- the accessory functions include those functions required for AAV replication, including, without limitation, those moieties involved in activation of AAV gene transcription, stage specific AAV mRNA splicing, AAV DNA replication, synthesis of cap expression products, and AAV capsid assembly.
- Viral-based accessory functions can be derived from any of the known helper viruses such as adenovirus, herpesvirus (other than herpes simplex virus type-1), and vaccinia virus.
- the rAAV vectors of the present disclosure comprise a viral genome with at least one ITR region and a payload region, e.g., the nucleic acid construct or an expression construct (e.g., comprising an expression cassette), or gene therapy constructs comprising a polynucleotide encoding a proteasome regulatory subunit disclosed herein.
- composition comprising a rAAV delivery vector comprises a payload comprising a nucleic acid encoding PA28A, optionally in combination with PA28B disclosed herein.
- the rAAV delivery vector comprises the nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a promoter which is tissue or cell-type specific promoter.
- the promoter is a bi directional promoter.
- the promoter is selected from a MecP2 promoter, a GUSB promoter, a CBh promoter, a PGK promoter, a human synapsin (Syn) promoter, a human rhodopsin (Rhod) kinase promoter, a cone arrestin promoter, a tyrosine hydroxylase promoter, a dopamine transporter promoter, a GRK1 promoter, a cardiac myosin light chain 2 promoter, a CAMKII promoter, a hDlx promoter, a CMV promoter, a CAG promoter, a bidirectional CAG promoter, a CBA promoter, an EFla promoter, or a human ubiquitin C promoter.
- the rAAV delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a polyadenylation site, a woodchuck hepatitis virus post-transcriptional regulatory element (WPRE), an optimized woodchuck hepatitis virus post-transcriptional regulatory element (oPRE), an intron, a microRNA binding motif, or any combination thereof.
- WPRE woodchuck hepatitis virus post-transcriptional regulatory element
- oPRE optimized woodchuck hepatitis virus post-transcriptional regulatory element
- the rAAV delivery vector comprises a first expression cassette comprising a polynucleotide encoding PA28A and a second expression cassette comprising a polynucleotide encoding PA28B. See Kugler et ah, Virology 311 (2003) 89-95.
- the first and second expression cassettes comprise the same promoter. In some aspects, the first and second expression cassettes comprise different promoters. In some aspects, the first expression cassette comprises a hSyn promoter and the second expression cassette comprises a CMV promoter. In some aspects, the first expression cassette comprises a CMV promoter and the second expression cassette comprises a hSyn promoter. In some aspects, the first and/or second expression cassettes comprise a polyA (e.g., bGH polyadenylation site, hGH polyadenylation site, or a SV40 polyadenylation site). In some aspects, the first and/or second expression cassette can further comprise a WPRE. In some aspects, the first and/or second expression cassette can further comprise an optimized WPRE (oPRE).
- oPRE optimized WPRE
- the rAAV delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a chimeric intron. In some aspects, the rAAV delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a beta-globin intron. In some aspects, the rAAV delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising minute virus of mice (MVM) intron.
- VMM minute virus of mice
- the rAAV delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a polyA sequence selected from the group consisting of a beta-globin polyA, a bovine growth hormone polyA (bGH polyA), a human growth hormone polyA (hGH polyA), and a SV40 immediate early poly A (SV40 poly A).
- a polyA sequence selected from the group consisting of a beta-globin polyA, a bovine growth hormone polyA (bGH polyA), a human growth hormone polyA (hGH polyA), and a SV40 immediate early poly A (SV40 poly A).
- the polyA is a bovine growth hormone polyA.
- the rAAV delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a 5’ and/or a 3’ untranslated region (UTR).
- the rAAV delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprises a microRNA binding motif in the 3’ untranslated region (UTR).
- the rAAV delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising comprises a WPRE. In some aspects, the rAAV delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising comprises an optimized WPRE (oPRE).
- oPRE optimized WPRE
- the nucleic acid construct or an expression construct comprises a promoter sequence (e.g., hSyn or pRod), a nucleic acid sequence encoding Proteasome activator complex subunit 1 (PA28A), and optionally a WPRE.
- the nucleic acid construct or an expression construct comprises a promoter sequence (e.g., hSyn or pRod), a nucleic acid sequence encoding Proteasome activator complex subunit 1 (PA28A), and optionally an optimized WPRE (oPRE).
- the rAAV delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) further comprises a nucleic acid sequence encoding Proteasome activator complex subunit 2 (PA28B).
- PA28B Proteasome activator complex subunit 2
- the polynucleotide comprises a promoter sequence, a nucleic acid encoding PA28A, a proteolytic cleavage site, and a nucleic acid encoding PA28B.
- the polynucleotide comprises a promoter sequence, a nucleic acid encoding PA28A, a proteolytic cleavage site, and a nucleic acid encoding PA28B in 5'-3' orientation.
- the polynucleotide comprises a promoter sequence, a nucleic acid encoding PA28B, a proteolytic cleavage site, and a nucleic acid encoding PA28A in 5'-3' orientation.
- the cleavage sequence a P2A, a furin, or an intein-F2A cleavage sequence.
- the proteolytic cleavage site is P2A.
- the promoter is hSyn or pRhod.
- the polynucleotide further comprises a WPRE.
- the polynucleotide further comprises an optimized WPRE (oPRE).
- the rAAV delivery vector is suitable for tissue specific delivery (e.g., to the brain, eye, or heart muscle).
- the AAV vector comprises two ITRs. These two ITRs flank the payload region at the 5' and 3' ends.
- the ITRs function as origins of replication comprising recognition sites for replication.
- ITRs comprise sequence regions, which can be complementary and symmetrically arranged.
- ITRs incorporated into AAV vectors of the disclosure can be comprised of naturally occurring polynucleotide sequences or recombinantly derived polynucleotide sequences.
- the AAV vector is self-complementary.
- the AAV vector comprises two ITRs that facilitate the sc AAV.
- the ITRs can be derived from the same serotype as the capsid, selected from any of the serotypes listed herein, or a derivative thereof.
- the ITR can be of a different serotype from the capsid.
- the AAV vector has more than one ITR.
- the AAV vector has a viral genome comprising two ITRs.
- the ITRs are of the same serotype as one another.
- the ITRs are of different serotypes. Non-limiting examples include zero, one or both of the ITRs having the same serotype as the capsid.
- both ITRs of the AAV vector are AAV2 ITRs.
- the AAV vector of the present disclosure comprises a polyadenylation sequence.
- the AAV vectors of the present disclosure can comprise a polyadenylation sequence between the 3' end of the payload coding sequence and the 5' end of the 3' ITR.
- the AAV vector of the present disclosure comprises an untranslated region (UTR).
- UTR untranslated region
- wild-type untranslated regions (UTRs) of a gene are transcribed but not translated.
- the 5' UTR starts at the transcription start site and ends at the start codon and the 3' UTR starts immediately following the stop codon and continues until the termination signal for transcription.
- Wild-type 3' UTRs are known to have stretches of Adenosines and Uridines embedded therein. These AU rich signatures are particularly prevalent in genes with high rates of turnover. Based on their sequence features and functional properties, the AU rich elements (AREs) can be separated into three classes (Chen et al, 1995, the contents of which are herein incorporated by reference in its entirety). Most proteins binding to the AREs are known to destabilize the messenger, whereas members of the ELAV family, most notably HuR, have been documented to increase the stability of mRNA. HuR binds to AREs of all the three classes. Engineering the HuR specific binding sites into the 3' UTR of nucleic acid molecules will lead to HuR binding and thus, stabilization of the message in vivo.
- AREs AU rich elements
- any UTR from any gene known in the art can be incorporated into an AAV vector of the present disclosure. These UTRs, or portions thereof, can be placed in the same orientation as in the gene from which they were selected or they can be altered in orientation or location.
- the UTR used in an AAV vector of the present disclosure can be inverted, shortened, lengthened, made with one or more other 5' UTRs or 3' UTRs known in the art.
- the term "altered" as it relates to a UTR means that the UTR has been changed in some way in relation to a reference sequence.
- a 3' or 5' UTR can be altered relative to a wild-type or native UTR by the change in orientation or location as taught above or can be altered by the inclusion of additional nucleotides, deletion of nucleotides, swapping or transposition of nucleotides.
- an AAV vector of the present disclosure comprises at least one artificial UTRs, which is not a variant of a wild-type UTR.
- an AAV vector of the present disclosure comprises UTRs, which have been selected from a family of transcripts whose proteins share a common function, structure, feature or property.
- the payload region of an AAV vector of the present disclosure comprises at least one element to enhance the expression.
- nucleic acids described herein further comprise a Woodchuck
- WPRE Hepatitis Virus Posttranscriptional Regulatory Element
- the WPRE sequence can be any wild-type or mutated WPRE sequence. See, e.g., U.S. Patent No. 10,179,918.
- the nucleic acids described herein further comprise an optimized Woodchuck Hepatitis Virus Posttranscriptional Regulatory Element (oPRE).
- the present disclosure provides also methods for the generation of AAV particles, by viral genome replication in a viral replication cell comprising contacting the viral replication cell with an AAV polynucleotide or AAV genome (e.g., an AAV vector of the present disclosure).
- an AAV particle is produced by a method comprising the steps of: (1) co-transfecting competent bacterial cells with a bacmid vector and either a viral construct vector and/or AAV payload construct vector, (2) isolating the resultant viral construct expression vector and AAV payload construct expression vector and separately transfecting viral replication cells, (3) isolating and purifying resultant payload and viral construct particles comprising viral construct expression vector or AAV payload construct expression vector, (4) co-infecting a viral replication cell with both the AAV payload and viral construct particles comprising viral construct expression vector or AAV payload construct expression vector, and (5) harvesting and purifying the viral particle comprising a parvoviral genome.
- the present disclosure provides a method for producing an AAV particle comprising the steps of (1) simultaneously co-transfecting mammalian cells, such as, but not limited to HEK293 cells, with a payload region (e.g., polynucleotide encoding a therapeutic protein or therapeutic peptide of the disclosure), a construct expressing rep and cap genes and a helper construct, and (2) harvesting and purifying the AAV particle comprising a viral genome.
- a payload region e.g., polynucleotide encoding a therapeutic protein or therapeutic peptide of the disclosure
- a construct expressing rep and cap genes and a helper construct e.g., a construct expressing rep and cap genes and a helper construct
- the AAV particles can be produced in a viral replication cell that comprises an insect cell.
- Growing conditions for insect cells in culture, and production of heterologous products in insect cells in culture are well-known in the art, see, e.g., U.S. Patent No. 6,204,059.
- the viral replication cell can be selected from any biological organism, including prokaryotic (e.g., bacterial) cells, and eukaryotic cells, including, insect cells, yeast cells and mammalian cells.
- Viral replication cells can comprise mammalian cells such as A549, WEH1, 3T3, 10T1/2, BHK, MDCK, COS 1, COS 7, BSC 1, BSC 40, BMT 10, VERO. W138, HeLa, HEK293, Saos, C2C12, L cells, HT1080, HepG2 and primary fibroblast, hepatocyte and myoblast cells derived from mammals.
- Viral replication cells comprise cells derived from mammalian species including, but not limited to, human, monkey, mouse, rat, rabbit, and hamster or cell type, including but not limited to fibroblast, hepatocyte, tumor cell, cell line transformed cell, etc.
- Viral production disclosed herein describes processes and methods for producing
- AAV particles that contact a target cell to deliver a payload e.g. a recombinant viral construct, which comprises a polynucleotide sequence encoding a payload such as a therapeutic protein disclosed herein.
- the AAV particles can be produced in a viral replication cell that comprises a mammalian cell.
- Viral replication cells commonly used for production of recombinant AAV particles include, but are not limited to 293 cells, COS cells, HeLa cells, and KB cells.
- AAV particles are produced in mammalian cells wherein all three VP proteins are expressed at a stoichiometry approaching 1:1:10 (VP1 :VP2:VP3).
- the regulatory mechanisms that allow this controlled level of expression include the production of two mRNAs, one for VP1, and the other for VP2 and VP3, produced by differential splicing.
- AAV particles are produced in mammalian cells using a triple transfection method wherein a payload construct, parvoviral Rep and parvoviral Cap and a helper construct are comprised within three different constructs.
- the triple transfection method of the three components of AAV particle production can be utilized to produce small lots of virus for assays including transduction efficiency, target tissue (tropism) evaluation, and stability.
- the viral construct vector and the AAV payload construct vector can be each incorporated by a transposon donor/acceptor system into a bacmid, also known as a baculovirus plasmid, by standard molecular biology techniques known and performed by a person skilled in the art. Transfection of separate viral replication cell populations produces two baculoviruses, one that comprises the viral construct expression vector, and another that comprises the AAV payload construct expression vector. The two baculoviruses can be used to infect a single viral replication cell population for production of AAV particles.
- Baculovirus expression vectors for producing viral particles in insect cells including but not limited to Spodoptera frugiperda (Sf9) cells, provide high titers of viral particle product.
- Recombinant baculovirus encoding the viral construct expression vector and AAV payload construct expression vector initiates a productive infection of viral replicating cells.
- Infectious baculovirus particles released from the primary infection secondarily infect additional cells in the culture, exponentially infecting the entire cell culture population in a number of infection cycles that is a function of the initial multiplicity of infection, see, e.g., Urabe, M. e/a/., J Virol. 2006 Feb; 80 (4): 1874-85, the contents of which are herein incorporated by reference in their entirety.
- AAV particles with baculovirus in an insect cell system can address known baculovirus genetic and physical instability.
- Baculovirus-infected viral producing cells are harvested into aliquots that can be cryopreserved in liquid nitrogen; the aliquots retain viability and infectivity for infection of large-scale viral producing cell culture (Wasilko DJ et al., Protein Expr Purif. 2009 Jun; 65(2): 122-32).
- stable viral replication cells permissive for baculovirus infection are engineered with at least one stable integrated copy of any of the elements necessary for AAV replication and viral particle production including, but not limited to, the entire AAV genome, Rep and Cap genes, Rep genes, Cap genes, each Rep protein as a separate transcription cassette, each VP protein as a separate transcription cassette, the AAP (assembly activation protein), or at least one of the baculovirus helper genes with native or non-native promoters.
- AAV particle production can be modified to increase the scale of production.
- Transfection of replication cells in large-scale culture formats can be carried out according to any methods known in the art.
- cell culture bioreactors can be used for large scale viral production.
- bioreactors comprise stirred tank reactors.
- Cell lysis methods can be chemical or mechanical. Chemical cell lysis typically comprises contacting one or more cells with one or more lysis agent. Mechanical lysis typically comprises subjecting one or more cells to one or more lysis condition and/or one or more lysis force. In some aspects, chemical lysis can be used to lyse cells.
- lysis agent refers to any agent that can aid in the disruption of a cell. In some cases, lysis agents are introduced in solutions, termed lysis solutions or lysis buffers. As used herein, the term "lysis solution” refers to a solution (typically aqueous) comprising one or more lysis agent. In addition to lysis agents, lysis solutions can include one or more buffering agents, solubilizing agents, surfactants, preservatives, cryoprotectants, enzymes, enzyme inhibitors and/or chelators.
- Concentrations of salts can be increased or decreased to obtain an effective concentration for rupture of cell membranes.
- Lysis agents comprising detergents can include ionic detergents or non-ionic detergents.
- Detergents can function to break apart or dissolve cell structures including, but not limited to cell membranes, cell walls, lipids, carbohydrates, lipoproteins and glycoproteins.
- lysis force refers to a physical activity used to disrupt a cell. Lysis forces can include, but are not limited to mechanical forces, sonic forces, gravitational forces, optical forces, electrical forces and the like. Cell lysis carried out by mechanical force is referred to herein as “mechanical lysis.” Mechanical forces that can be used according to mechanical lysis can include high shear fluid forces.
- Clarification refers to initial steps taken in purification of viral particles from cell lysates. Clarification serves to prepare lysates for further purification by removing larger, insoluble debris. Clarification steps can include, but are not limited to centrifugation and filtration.
- AAV particles can be purified from clarified cell lysates by one or more methods of chromatography.
- Chromatography refers to any number of methods known in the art for separating out one or more elements from a mixture. Such methods can include, but are not limited to ion exchange chromatography (e.g. cation exchange chromatography and anion exchange chromatography), immunoaffmity chromatography and size-exclusion chromatography.
- the viral particles comprise a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a polynucleotide comprising a promoter sequence (e.g., hSyn or pRod), a nucleic acid sequence encoding Proteasome activator complex subunit 1 (PA28A), and optionally a WPRE.
- a promoter sequence e.g., hSyn or pRod
- PA28A Proteasome activator complex subunit 1
- the viral particles comprise a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a polynucleotide comprising a promoter sequence, a nucleic acid encoding PA28A, a proteolytic cleavage site, and a nucleic acid encoding PA28B.
- the viral particles comprise a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a polynucleotide comprising a promoter sequence, a nucleic acid encoding PA28A, a proteolytic cleavage site, and a nucleic acid encoding PA28B in 5'-3' orientation.
- the viral particles comprise a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a polynucleotide comprising a promoter sequence, a nucleic acid encoding PA28B, a proteolytic cleavage site, and a nucleic acid encoding PA28A in 5'-3' orientation.
- the viral particles comprise the nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a promoter which is tissue or cell-type specific promoter.
- the promoter is a bi-directional promoter.
- the promoter is selected from a MecP2 promoter, a GUSB promoter, a CBh promoter, a PGK promoter, a human synapsin (Syn) promoter, a human rhodopsin (Rhod) kinase promoter, a cone arrestin promoter, a tyrosine hydroxylase promoter, a dopamine transporter promoter, a GRK1 promoter, a cardiac myosin light chain 2 promoter, a CAMKII promoter, a hDlx promoter, a CMV promoter, a CAG promoter, a bidirectional CAG promoter, a CBA promoter, an EFla promoter, or a human ubiquitin C promoter.
- the viral particles comprise comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a polyadenylation site, a woodchuck hepatitis virus post-transcriptional regulatory element (WPRE), an optimized woodchuck hepatitis virus post-transcriptional regulatory element (oPRE), an intron, a microRNA binding motif, or any combination thereof.
- the viral particles e.g., AAV particles
- the viral particles comprise a first expression cassette comprising a polynucleotide encoding PA28A and a second expression cassette comprising a polynucleotide encoding PA28B.
- the first and second expression cassettes comprise the same promoter. In some aspects, the first and second expression cassettes comprise different promoters. In some aspects, the first expression cassette comprises a hSyn promoter and the second expression cassette comprises a CMV promoter. In some aspects, the first expression cassette comprises a CMV promoter and the second expression cassette comprises a hSyn promoter. In some aspects, the first and/or second expression cassettes comprise a polyA (e.g., bGH polyadenylation site, hGH polyadenylation site, or a SV40 polyadenylation site). In some aspects, the first and/or second expression cassette can further comprise a WPRE. In some aspects, the first and/or second expression cassette can further comprise an optimized WPRE (oPRE).
- oPRE optimized WPRE
- the viral particles comprise comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a chimeric intron.
- the viral particles comprise comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a beta-globin intron.
- the viral particles comprise a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising minute virus of mice (MVM) intron.
- the viral particles comprise a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a polyA sequence selected from the group consisting of a beta-globin poly A, a bovine growth hormone polyA (bGH polyA), a human growth hormone polyA (hGH polyA), and a SV40 immediate early poly A (SV40 polyA).
- a polyA sequence selected from the group consisting of a beta-globin poly A, a bovine growth hormone polyA (bGH polyA), a human growth hormone polyA (hGH polyA), and a SV40 immediate early poly A (SV40 polyA).
- the polyA is a bovine growth hormone polyA.
- the viral particles comprise a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a 5’ and/or a 3’ untranslated region (UTR).
- the viral particles e.g., AAV particles
- AAV particles comprise a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprises a microRNA binding motif in the 3’ untranslated region (UTR).
- an expression construct e.g., comprising an expression cassette
- UTR untranslated region
- the viral particles comprise a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising comprises a WPRE.
- the viral particles comprise a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising comprises an optimized WPRE (oPRE).
- the cleavage sequence a P2A, a furin, or an intein-F2A cleavage sequence.
- the proteolytic cleavage site is P2A.
- the promoter is hSyn or pRhod.
- the polynucleotide further comprises a WPRE.
- the polynucleotide further comprises an optimized WPRE (oPRE).
- the viral particles e.g., AAV particles
- are suitable for tissue specific delivery e.g., to the brain, eye, or heart muscle).
- compositions comprising a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy constructs, or a viral vector (e.g., AAV vector or AAV particle) disclosed herein and a carrier (e.g., pharmaceutically acceptable carrier).
- a construct or vector of the disclosure is formulated for delivery to a cell or tissues.
- the construct or vector of the disclosure is formulated for delivery to a subject.
- pharmaceutically acceptable carriers can be readily selected by one of skill in the art in view of the indication for which the viral vector (e.g., AAV vector or AAV particle) is directed.
- one suitable carrier includes saline, which may be formulated with a variety of buffering solutions (e.g., phosphate buffered saline).
- Other exemplary carriers include sterile saline, lactose, sucrose, calcium phosphate, gelatin, dextran, agar, pectin, peanut oil, sesame oil, and water. The selection of the carrier is not a limitation of the present disclosure.
- compositions of the disclosure may contain, in addition to the viral vector (e.g., AAV vector or AAV particle) and carrier(s), other conventional pharmaceutical ingredients, such as preservatives, or chemical stabilizers.
- suitable exemplary preservatives include chlorobutanol, potassium sorbate, sorbic acid, sulfur dioxide, propyl gallate, the parabens, ethyl vanillin, glycerin, phenol, parachlorophenol, and poloxamers (non-ionic surfactants) such as Pluronic® F-68.
- Suitable chemical stabilizers include gelatin and albumin.
- nucleic acid constructs e.g., comprising an expression cassette
- gene therapy constructs e.g., viral vectors (e.g., AAV vector or AAV particle), or pharmaceutical compositions disclosed herein.
- viral vectors e.g., AAV vector or AAV particle
- pharmaceutical compositions disclosed herein e.g., pharmaceutical compositions disclosed herein.
- the present disclosure provides methods for increasing proteasome activity in a cell and/or tissue comprising administering to the cell or tissue a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical composition disclosed herein.
- a nucleic acid construct e.g., comprising an expression cassette
- a gene therapy construct e.g., a viral vector or AAV particle
- pharmaceutical composition disclosed herein e.g., AAV vector or AAV particle
- the present disclosure provides methods for treating a disease or condition associated with proteinopathy in a subject in need thereof comprising administering to the subject a composition comprising a gene therapy construct or vector disclosed herein.
- the disease or condition is a neurodegenerative disease (e.g., Parkinson’s Disease or Huntington’s Disease), an ocular disease (e.g., retinitis pigmentosa or Kjer’s optic neuropathy), or a cardiac disease.
- the administration is tissue specific.
- the gene therapy construct or vector disclosed herein is administered to the brain, eye, or heart muscle.
- the methods include administering a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical composition comprising a polynucleotide encoding PA28A, optionally in combination with a polynucleotide encoding PA28B disclosed herein.
- an expression construct e.g., comprising an expression cassette
- a gene therapy construct e.g., a viral vector (e.g., AAV vector or AAV particle)
- pharmaceutical composition comprising a polynucleotide encoding PA28A, optionally in combination with a polynucleotide encoding PA28B disclosed herein.
- the method comprises administering a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical composition comprising a polynucleotide comprising a promoter sequence (e.g., hSyn or pRod), a nucleic acid sequence encoding Proteasome activator complex subunit 1 (PA28A), and optionally a WPRE.
- a nucleic acid construct e.g., comprising an expression cassette
- a gene therapy construct e.g., a gene therapy construct
- a viral vector e.g., AAV vector or AAV particle
- pharmaceutical composition comprising a polynucleotide comprising a promoter sequence (e.g., hSyn or pRod), a nucleic acid sequence encoding Proteasome activator complex subunit 1 (PA28A), and optionally a
- the method comprises administering a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical composition comprising a polynucleotide comprising a promoter sequence (e.g., hSyn or pRod), a nucleic acid sequence encoding Proteasome activator complex subunit 1 (PA28A), and optionally an optimized WPRE (oPRE).
- a nucleic acid construct e.g., comprising an expression cassette
- a gene therapy construct e.g., a gene therapy construct
- a viral vector e.g., AAV vector or AAV particle
- pharmaceutical composition comprising a polynucleotide comprising a promoter sequence (e.g., hSyn or pRod), a nucleic acid sequence encoding Proteasome activator complex subunit 1 (PA28A), and
- the method comprises administering a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical composition comprising further comprising a nucleic acid sequence encoding Proteasome activator complex subunit 2 (PA28B).
- a nucleic acid construct e.g., comprising an expression cassette
- a gene therapy construct e.g., a viral vector or AAV particle
- pharmaceutical composition comprising further comprising a nucleic acid sequence encoding Proteasome activator complex subunit 2 (PA28B).
- PA28B Proteasome activator complex subunit 2
- the method comprises a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical composition comprising a polynucleotide comprising a promoter sequence, a nucleic acid encoding PA28A, a proteolytic cleavage site, and a nucleic acid encoding PA28B.
- an expression construct e.g., comprising an expression cassette
- a gene therapy construct e.g., a viral vector or AAV particle
- pharmaceutical composition comprising a polynucleotide comprising a promoter sequence, a nucleic acid encoding PA28A, a proteolytic cleavage site, and a nucleic acid encoding PA28B.
- the method comprises administering a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical composition comprising a polynucleotide comprising a promoter sequence, a nucleic acid encoding PA28A, a proteolytic cleavage site, and a nucleic acid encoding PA28B in 5'-3' orientation.
- an expression construct e.g., comprising an expression cassette
- a gene therapy construct e.g., a viral vector or AAV particle
- pharmaceutical composition comprising a polynucleotide comprising a promoter sequence, a nucleic acid encoding PA28A, a proteolytic cleavage site, and a nucleic acid encoding PA28B in 5'-3' orientation.
- the method comprises administering a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical composition comprising a polynucleotide comprising a promoter sequence, a nucleic acid encoding PA28B, a proteolytic cleavage site, and a nucleic acid encoding PA28A in 5'-3' orientation.
- an expression construct e.g., comprising an expression cassette
- a gene therapy construct e.g., a viral vector or AAV particle
- pharmaceutical composition comprising a polynucleotide comprising a promoter sequence, a nucleic acid encoding PA28B, a proteolytic cleavage site, and a nucleic acid encoding PA28A in 5'-3' orientation.
- the methods include administering a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical composition comprising a promoter which is tissue or cell-type specific promoter.
- the promoter is a bi-directional promoter.
- the promoter is selected from a MecP2 promoter, a GUSB promoter, a CBh promoter, a PGK promoter, a human synapsin (Syn) promoter, a human rhodopsin (Rhod) kinase promoter, a cone arrestin promoter, a tyrosine hydroxylase promoter, a dopamine transporter promoter, a GRK1 promoter, a cardiac myosin light chain 2 promoter, a CAMKII promoter, a hDlx promoter, a CMV promoter, a CAG promoter, a bidirectional CAG promoter, a CBA promoter, an EFla promoter, or a human ubiquitin C promoter.
- the methods include administering a nucleic acid construct, an expression construct (e.g., comprising an expression cassette, or comprising a first expression cassette and a second expression cassette described herein), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical composition comprising a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a polyadenylation site, a woodchuck hepatitis virus post-transcriptional regulatory element (WPRE), an optimized woodchuck hepatitis virus post-transcriptional regulatory element (oPRE), an intron, a microRNA binding motif, or any combination thereof.
- WPRE woodchuck hepatitis virus post-transcriptional regulatory element
- oPRE optimized woodchuck hepatitis virus post-transcriptional regulatory element
- the methods include administering a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical composition comprising a chimeric intron.
- the methods include administering a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical composition comprising a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a beta-globin intron.
- the methods include administering a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical composition comprising minute virus of mice (MVM) intron.
- a nucleic acid construct e.g., comprising an expression cassette
- a gene therapy construct e.g., a viral vector or AAV particle
- a pharmaceutical composition comprising minute virus of mice (MVM) intron e.g., AAV vector or AAV particle
- the methods include administering a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical composition comprising a polyA sequence selected from the group consisting of a beta-globin poly A, a bovine growth hormone polyA (bGH polyA), a human growth hormone polyA (hGH polyA), and a SV40 immediate early poly A (SV40 polyA).
- the polyA is a bovine growth hormone polyA.
- the methods include administering a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical composition comprising a 5’ and/or a 3’ untranslated region (UTR).
- the methods include administering a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical composition comprising a microRNA binding motif in the 3’ untranslated region (UTR).
- the methods include administering a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical composition comprising a WPRE.
- the methods include administering a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical composition comprising an optimized WPRE (oPRE).
- the cleavage sequence a P2A, a furin, or an intein-F2A cleavage sequence.
- the proteolytic cleavage site is P2A.
- the promoter is hSyn or pRhod.
- the polynucleotide further comprises a WPRE.
- the polynucleotide further comprises an optimized WPRE (oPRE).
- method is suitable for tissue specific delivery (e.g., to the brain, eye, or heart muscle).
- the gene therapy composition comprising an AAV vector, an AAV vector, or an AAV particle of the present disclosure for use in therapy, or for use as a medicament, or for use in treating a disease or disorder associated with misfolded or damaged proteins (e.g., neurological, optical or a cardiac diseases or disorders associated with proteinopathy) in a subject in need thereof is contemplated.
- a disease or disorder associated with misfolded or damaged proteins e.g., neurological, optical or a cardiac diseases or disorders associated with proteinopathy
- the diseases or disorders associated with proteinopathy is selected from the group consisting of inherited retinal disease (retinitis pigmentosa, Leber congenital amaurosis, Kjer’s optic neuropathy), neurodegeneration (genetic or idiopathic forms of Parkinson’s Disease, Alzheimer’s Disease, Huntington’s Disease, prion disease, multiple system atrophy, multiple sclerosis, amyotrophic lateral sclerosis, Friedreich ataxia), and cardiovascular diseases (desmin-related cardiomyopathy, ischemia/reperfusion injury).
- retinal disease retinitis pigmentosa, Leber congenital amaurosis, Kjer’s optic neuropathy
- neurodegeneration geneetic or idiopathic forms of Parkinson’s Disease, Alzheimer’s Disease, Huntington’s Disease, prion disease, multiple system atrophy, multiple sclerosis, amyotrophic lateral sclerosis, Friedreich ataxia
- cardiovascular diseases desmin-related cardiomyopathy, ischemia/reperfusion injury.
- the diseases or disorder associated with proteinopathy is selected from the group consisting of Alzheimer’s disease, Dutch hereditary cerebral hemorrhage with amyloidosis, Congophilic angiopathy, Tauopathies, Corticobasal degeneration, Pick’s disease, Progressive supranuclear palsy, Prion diseases, Creutzfeld-Jacob disease, Gerstmann-Straussler-Schneiker syndrome, fatal familia insomnia, Kuru, Bovine spongiform encephalopathy, Scrapie, Chronic wasting disease, Synucleinopathies, Parkinson’s disease, Lewy body variant of Alzheimer’s disease, Diffuse Lewy body disease, Dementia with Lewy bodies, Multiple system atrophy, Neurodegeneration with brain iron accumulation type I, Amyotrophic lateral sclerosis and frontotemporal lobar degeneration, Amyotrophic lateral sclerosis, Huntington’s disease, Hereditary dentatorubral-pallidoluysian atrophy, Kennedy’s
- the disease or disorder is selected from the group consisting of a neurodegenerative disease (e.g., Parkinson’s Disease or Huntington’s Disease), an ocular disease (e.g., retinitis pigmentosa or Kjer’s optic neuropathy), or a cardiac disease.
- a neurodegenerative disease e.g., Parkinson’s Disease or Huntington’s Disease
- an ocular disease e.g., retinitis pigmentosa or Kjer’s optic neuropathy
- a cardiac disease e.g., a retinitis pigmentosa or Kjer’s optic neuropathy
- the administration is tissue specific.
- the gene therapy construct or vector disclosed herein is administered to the brain, eye, or heart muscle.
- nucleic acid constructs e.g., comprising an expression cassette
- gene therapy constructs e.g., viral vectors (e.g., AAV vector or rAAV particle)
- viral vectors e.g., AAV vector or rAAV particle
- pharmaceutical compositions disclosed herein can be administered by any route which results in a therapeutically effective outcome, e.g., for therapeutic expression of a PSME (e.g., PA28A alone or PA28A in combination with PA28B) disclosed herein.
- PSME e.g., PA28A alone or PA28A in combination with PA28B
- the delivery can be intramuscular (IM), intravenous (IV), intraocular (e.g., sub-retinal), intracranial (e.g., striatum, ventricular), or intrathecal (e.g., cistema magna, lumbar).
- IM intramuscular
- IV intravenous
- IV intraocular
- intracranial e.g., striatum, ventricular
- intrathecal e.g., cistema magna, lumbar
- the disclosure is directed to a method of delivering a gene therapy to brain tissue.
- the disclosure is directed to delivering a gene therapy construct to eye tissue.
- the disclosure is directed to delivering a gene therapy construct to heart tissue.
- the delivery vectors disclosed herein can be administered in any suitable form, either as a liquid solution or suspension, as a solid form suitable for liquid solution or suspension in a liquid solution.
- kits, or products of manufacture comprising
- nucleic acid construct e.g., comprising one or more expression cassettes
- expression construct e.g., comprising one or more expression cassettes
- gene therapy construct e.g., a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical compositions of the present disclosure
- viral vector e.g., AAV vector or AAV particle
- optionally instructions for use e.g., a package insert with instructions to perform any of the methods described herein.
- the kit or product of manufacture comprises (i) a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical compositions of the present disclosure, (ii) optionally, an additional therapeutic agent, and (iii) optionally, instructions for use (e.g., a package insert with instructions to perform any of the methods described herein are also contemplated).
- a nucleic acid construct e.g., comprising an expression cassette
- a gene therapy construct e.g., a viral vector or AAV particle
- a viral vector e.g., AAV vector or AAV particle
- instructions for use e.g., a package insert with instructions to perform any of the methods described herein are also contemplated.
- kits or product of manufacture are in one or more containers.
- the kit or product of manufacture comprises (i) a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical compositions of the present disclosure, and (ii) a brochure with instructions to prepare the product for administration.
- Viral vector constructs for expressing PA28A and PA28B subunits are prepared.
- the constructs include: (1) pAAV9- hSyn-PA28alpha(mouse)-WPRE; (2) pAAV2- pRhod-PA28alpha(mouse)-WPRE; (3) pAAV9- hSyn-PA28beta-P2A- PA28alpha(mouse)-WPRE; and (4) pAAV2-pRhod- PA28beta-P2A-PA28alpha(mouse)- WPRE.
- Constructs are prepared by synthesizing genes de novo followed by molecular biology techniques to place into AAV or lentiviral backbones.
- HEK-293T cells (PSME1-KO) cells were transfected with plasmids comprising expression of PA28alpha-only (PSMEl-ZsGreenl in pBi-CMV) (FIG. 1A), bicistronic expression of PA28alpha and PA28beta (PSME1-PSME2 in pBi- CMV) (FIG. IB), or an empty vector control (ZsGreen (Neg Ctrl) in pBi-CMV) (FIG. 1C). 48 hours post-transfection, the cells were harvested and the lysates were used in a proteasomal activity assay using Suc-LLVY-AMC as an artificial substrate.
- Proteasome activity cleaves AMC from the artificial peptide substrate (Suc-LLVY-AMC) and generates free AMC which is fluorescent. Proteasomal activity was kinetically monitored by measuring the increase in fluorescence of the free AMC (FIG. 2A). Epoxomicin, a 20S proteasome inhibitor, was used to determine background non-proteasomal cleavage of the substrate and thereby calculate specific proteasome activity. Formation of 11 S caps with both PA28alpha and PA28beta has stronger proteasomal activation property than 11 S cap formed from PA28alpha alone. (FIGs. 2B-2E). Overall activity increased with more lysate used.
- Plasmids were generated to test bicistronic expression of PA28alpha and PA28beta to achieve formation of active 11 S proteasomal caps.
- PSMEl-KO HEK-293T cells were transfected with either an empty vector plasmid (ZsGreen in pBi-CMV), a PA28alpha and PA28beta bicistronic plasmid (PA28a and RA28b in pBi-CMV (bicistronic)) (FIG. 4C), a 1 : 1 ratio of a PA28alpha and PA28beta plasmid (FIG.
- PA28a- IRES-PA28P (FIGs. 4G and 4H)
- PA28a-R2A-RA28b PA28alpha and PA28beta P2A bicistronic plasmid
- PA28a-Flexible Linker-PA28P PA28a-Flexible Linker-PA28P
- Lysates from the transfected PSME1-KO HEK-293T cells were used to determine the proteasomal activity of the PA28alpha and PA28beta bicistronic plasmid constructs (FIGs. 6A-6B). Only the PA28alpha-PA28beta bicistronic construct, 1 : 1 mix of PA28alpha only & PA28beta only, and PA28alpha only samples showed appreciable proteasomal activity.
- Plasmids were generated to test tandem bicistronic expression of PA28alpha and PA28beta to achieve formation of active 11 S proteasomal caps (FIGs. 7A-7C, 7E-7G).
- PSME1-KO HEK-293T cells were transfected with either a wild-type pBI-CMV vector (ZsGreen in pBi-CMV) (FIG. 1C), a PA28alpha and PA28beta bicistronic plasmid (PA28a and RA28b in pBi-CMV (bicistronic) (FIG.
- Lysates from the transfected PSME1-KO HEK-293T cells were used to determine the proteasomal activity of the PA28alpha and PA28beta constructs (FIGS. 9A-9B). Only The PA28alpha and PA28beta in pBi-CMV, 1 : 1 mix of PA28alpha only & PA28beta only, PA28alpha only, and EF1 alpha tandem samples showed appreciable proteasomal activity.
- mice were administered lpl of self-complementary AAV8 (scAAV8) viral mixture via sub-retinal injection to the left eye.
- the scAAV8 viral mixture comprising a 4:4: 1 (by volume) mixture of scAAV8-hRho-mPMSEl (PA28alpha) (titer: 5.38xl0 13 gc/mL), scAAV8-hRho- mPSME2 (PA28beta) (titer: 3.99xl0 13 gc/mL), and scAAV8-hRho-eGFP (titer: 4.49xl0 13 gc/mL).
- the eyes were harvested 18 days post-injection and protein expression was analyzed by western blot. Lysate from the uninjected right eye was used as a control. PA28alpha, PA28beta, and GFP were all detected in the injected left eyes (FIG. 10A).
- mice were administered lpl of the scAAV8 vectors, AAV8-hRHO-oPRE tandem mPA28alpha/beta (FIG. 7D) or AAV8-CBh tandem mA28alpha/beta (FIG. 7B) via sub-retinal injection to the left eye. Eyes were harvested 21 days post-injection and protein expression was analyzed by western blot. Lysate from the uninjected right eye was used as a control. Western blotting shows successful expression of both PA28alpha and PA28beta in the injected left eyes (FIG. 10B).
- mice were administered 1 pL of an AAV8 expressing both PA28oc and RA28b in the tandem configuration under control of CBh promoter (FIG. 7B). Stronger expression of PA28alpha was detected in the retina pigment epithelium rather than the photoreceptors (except in the extreme left part of the retina) (FIG. 11C).
- Example 4 Mouse Model of Retinitis Pigmentosa
- constructs are administered by sub-retinal injection in a mouse model of Retinitis Pigmentosa, Rho P23H mouse model (Jackson Labs (stock #017628)) of Retinitis Pigmentosa (see Lobanova et ak, Nat Commun. 2018 Apr 30;9(1): 1738).
- constructs of the disclosure are administered to the striatum along with co-infusion of AAV that overexpresses the mutant form of Huntington (66Q, mHTT) ⁇ Cell Reports, 24, 1397-1406, 2018) in mice.
- Tissues are harvested and analyzed by histology for the presence of aggregated huntingtin protein, loss or damage to neurons, and presence of PA28 subunits Tissues may also be measured for proteasomal activity.
- constructs of the disclosure are administered along with co-infusion of an AAV that overexpresses the mutant form of alpha-synuclein (A53T) in the substantia nigra of mice ⁇ Molecular Neurodegeneration 2013, 8:44).
- A53T alpha-synuclein
- Tissues are harvested and analyzed by histology for the presence of dopaminergic neurons, aggregated alpha-synuclein, and presence of PA28 subunits. Tissues are also be measured for proteasomal activity.
- Example 7 Mouse Model Cariac Proteinopathy and ischemia/reperfusion injury
- constructs of the disclosure are administered to mice harboring the ab-crystallin R120G mutation (CryAB(R120G), which is a model of desmin-related cardiomyopathy.
- a mouse model of myocardial I/R injury are tested where a vessel blockage is formed and later released to model I/R injury.
- Mice are evaluated for increases in LV posterior and anterior wall thickness at both end of diastole and systole as well as in calculated LV mass, ejection fraction, and fraction shortening. Histology of cardiac tissue are performed to characterize CryAB aggregates.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Genetics & Genomics (AREA)
- Chemical & Material Sciences (AREA)
- Engineering & Computer Science (AREA)
- Organic Chemistry (AREA)
- Biotechnology (AREA)
- Zoology (AREA)
- Wood Science & Technology (AREA)
- General Health & Medical Sciences (AREA)
- Molecular Biology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- General Engineering & Computer Science (AREA)
- Biomedical Technology (AREA)
- Biophysics (AREA)
- Biochemistry (AREA)
- Physics & Mathematics (AREA)
- Microbiology (AREA)
- Plant Pathology (AREA)
- Medicinal Chemistry (AREA)
- Animal Behavior & Ethology (AREA)
- Pharmacology & Pharmacy (AREA)
- Public Health (AREA)
- Epidemiology (AREA)
- Veterinary Medicine (AREA)
- Virology (AREA)
- Toxicology (AREA)
- Gastroenterology & Hepatology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Abstract
The present disclosure provides gene therapy constructs comprising nucleic acids encoding proteasome subunit PA28A, and optionally PA28B, delivery vectors (e.g., viral vectors), compositions, and methods of using the same to increase proteasomal activity and treat diseases associated with proteinopathy in a subject in need thereof.
Description
VECTOR CONSTRUCTS FOR DELIVERY OF NUCLEIC ACIDS ENCODING THERAPEUTIC PROTEASOME ACTIVATOR COMPLEX SUBUNITS AND
METHODS OF USING THE SAME
CROSS REFERENCE TO RELATED APPLICATIONS
[0001] The present application claims the priority benefit of U.S. Provisional Application
No. 63/223,840, filed July 20, 2021, which is hereby incorporated by reference in its entirety.
REFERENCE TO SEQUENCE LISTING SUBMITTED ELECTRONICALLY
[0002] The content of the electronically submitted sequence listing in ASCII text file
(Name: 4226_042PC01_Seqlisting_ST26.txt; Size: 29,156 bytes; and Date of Creation: July 14, 2022) filed with the application is incorporated herein by reference in its entirety.
FIELD OF DISCLOSURE
[0003] The present disclosure pertains to the medical field, including gene therapy constructs, vectors (e.g., viral vectors), and compositions suitable for delivery of nucleic acids encoding therapeutic proteins, and methods of using the same.
BACKGROUND
[0004] The ubiquitin-proteasome system (UPS) mediates the targeted degradation of abnormal as well as most normal intracellular proteins and generally includes two main steps: ubiquitination of a specific protein and subsequent degradation of the ubiquitinated protein by the proteasome.
[0005] Eukaryotic proteasomes are large multisubunit complexes that degrade the majority of proteins in the cell under normal conditions. The proteasome can be divided into two subcomplexes: a regulatory cap and the 20S catalytic core particle. The catalytic core (20S) is barrel shaped in structure and contains the enzymatic activity to degrade proteins. The activity of the catalytic core is regulated by 2 main types of regulatory caps: 19S and PA28. When combined with the 20S catalytic core, these caps, respectively,
form the 26S proteasome (20S + 19S) that degrades ubiquitin-tagged proteins or the 1 IS proteasome (20S + PA28) that degrades damaged or oxidized proteins independent of ubiquitin.
[0006] The 19S particle in eukaryotes consists of 19 individual proteins and is divisible into two subassemblies, a 9-subunit base that binds directly to the a ring of the 20S core particle, and a 10-subunit lid. Six of the nine base proteins are ATPase subunits from the AAA Family, and an evolutionary homolog of these ATPases exists in archaea, called PAN (Proteasome- Activating Nucleotidase).
[0007] PA28 (also referred to as 1 IS, REG or PSME) is a family of proteasome regulators whose members are present in many eukaryotic supergroups. In jawed vertebrates, they are represented by three paralogs, PA28a (also known as Proteasome activator complex subunit 1, PSME1, PA28A, and REGalpha), RA28b (also known as Proteasome activator complex subunit 2, PSME2, PA28B, and REGbeta), and RA28g, which assemble as heptameric hetero (RA28ab) or homo (RA28g) rings on one or both extremities of the 20S proteasome cylindrical structure. PSME1 (or PA28a) and PSME2 (or RA28b) are encoded by the PSME1 gene and the PSME2 gene, respectively.
[0008] Proteinopathies are diseases caused by protein misfolding and are characterized by aberrant protein aggregation. Terminally misfolded proteins are degraded mainly by the UPS. Ubiquitinated proteins usually accumulate in the cell during proteasome functional insufficiency (PFI). Consequently, PFI and protein aggregation can result in a cycle that can result in proteinopathy and other disorders. Enhancement of proteasomal function has been shown to protect against cardiac proteinopathy and ischemia/reperfusion injury in mice. Li et ak, J Clin Invest. 2011 Sep; 121(9):3689-700. Furthermore, increased proteasomal activity has been shown to support photoreceptor survival in inherited retinal degeneration. Lobanova et ak, Nat Commun. 2018 Apr 30; 9(1): 1738.
BRIEF SUMMARY
[0009] Certain aspects of the present disclosure are directed to constructs (e.g., gene therapy constructs) for use in packaging a PA28 cap (an alpha subunit alone, or alpha plus beta subunits) in a vector (e.g., viral vector such as AAV or lentivirus) for delivery to discrete parts of the body, e.g., the eye, brain, or heart, to increase the capacity of proteasomes to process misfolded or damaged proteins that cause disease. In certain
aspects, the disclosure is directed to gene therapy constructs encoding a PA28A, PA28B, or a combination thereof, vectors (e.g., viral vectors) suitable for delivery of nucleic acids encoding therapeutic a PA28A, PA28B, or a combination thereof, compositions thereof and methods of using the same. Certain aspects of the disclosure are directed to one or more viral vectors (e.g., adeno-associated virus (AAV) or lentivirus) for delivery of nucleic acids encoding a PA28A, PA28B, or a combination thereof to a subject in need thereof.
[0010] In some aspects, the present disclosure provides a construct (e.g., a gene therapy construct) comprising a polynucleotide comprising a promoter and a nucleic acid sequence encoding a Proteasome activator complex subunit 1 (PA28A).
[0011] In some aspects, the present disclosure provides a construct (e.g., a gene therapy construct) comprising a polynucleotide comprising a promoter, a nucleic acid sequence encoding a Proteasome activator complex subunit 1 (PA28A), and a nucleic acid sequence encoding Proteasome activator complex subunit 2 (PA28B). In some aspects the construct is a bicistronic construct.
[0012] In some aspects, the present disclosure provides a gene therapy constructs (e.g., a bicistronic construct) comprising a polynucleotide comprising (a) a first promoter sequence and a nucleic acid sequence encoding a Proteasome activator complex subunit 1 (PA28A), and (b) a second promoter sequence and a nucleic acid sequence encoding Proteasome activator complex subunit 2 (PA28B). In some aspects, the first and second promoter are the same. In some aspects, the first and second promoter are different.
[0013] In some aspects, the present disclosure provides a plurality of gene therapy constructs comprising (a) a polynucleotide comprising a promoter sequence and a nucleic acid sequence encoding a Proteasome activator complex subunit 1 (PA28A), and (b) a polynucleotide comprising a promoter sequence and a nucleic acid sequence encoding a promoter sequence and Proteasome activator complex subunit 2 (PA28B). In some aspects, the promoters are the same. In some aspects, the promoters are different.
[0014] In some aspects, the present disclosure provides an expression cassette comprising a polynucleotide comprising a promoter sequence and a nucleic acid sequence encoding a Proteasome activator complex subunit 1 (PA28A).
[0015] In some aspects, the present disclosure provides an expression cassette comprising a polynucleotide comprising a promoter, a nucleic acid sequence encoding a Proteasome
activator complex subunit 1 (PA28A), and a nucleic acid sequence encoding Proteasome activator complex subunit 2 (PA28B).
[0016] In some aspects, the present disclosure provides a first expression cassette comprising a polynucleotide comprising a promoter, a nucleic acid sequence encoding a Proteasome activator complex subunit 1 (PA28A), and a second expression cassette comprising a polynucleotide comprising a promoter and Proteasome activator complex subunit 2 (PA28B). In some aspects, the promoter of the first and second expression cassette are the same or different.
[0017] In some aspects, the promoter is a heterologous promoter. In some aspects, the promoter is tissue or cell-type specific promoter. In some aspects, the promoter is a bi directional promoter. In some aspects, the promoter is selected from a MecP2 promoter, a GUSB promoter, a CBh promoter, a PGK promoter, a human synapsin (Syn) promoter, a human rhodopsin (Rhod) kinase promoter, a cone arrestin promoter, a tyrosine hydroxylase promoter, a dopamine transporter promoter, a GRK1 promoter, a cardiac myosin light chain 2 promoter, a CAMKII promoter, a hDlx promoter, a CMV promoter, a CAG promoter, a bidirectional CAG promoter, a CBA promoter, an EFla promoter, or a human ubiquitin C promoter.
[0018] In some aspects, the gene therapy construct or expression construct further comprises a polyadenylation site, a woodchuck hepatitis virus post-transcriptional regulatory element (WPRE), an optimized woodchuck hepatitis virus post-transcriptional regulatory element (oPRE), an intron, a microRNA binding motif, or any combination thereof.
[0019] In some aspects, the gene therapy construct or expression construct comprises a chimeric intron. In some aspects, the gene therapy construct or expression construct comprises a beta-globin intron. In some aspects, the gene therapy construct or expression construct comprises minute virus of mice (MVM) intron.
[0020] In some aspects, the gene therapy construct or expression construct comprises a polyA sequence selected from the group consisting of a beta-globin poly A, a bovine growth hormone polyA (bGH polyA), a human growth hormone polyA (hGH polyA), and a SV40 immediate early poly A (SV40 polyA).
[0021] In some aspects, the gene therapy construct or expression construct comprises a 5’ and/or a 3’ untranslated region (UTR). In some aspects, gene therapy construct or
expression construct further comprise a microRNA binding motif in the 3’ untranslated region (UTR).
[0022] In some aspects, the gene therapy construct or expression construct further comprises a WPRE. In some aspects, the gene therapy construct or expression construct further comprises an optimized woodchuck hepatitis virus post-transcriptional regulatory element (oPRE).
[0023] In some aspects, the present disclosure provides a vector comprising gene therapy construct or expression construct disclosed herein. In some aspects, the vector is a viral vector. In some aspects, the viral vector is an adeno-associated virus (AAV) vector. In some aspects, the AAV vector comprises an inverted terminal repeat (ITR), e.g., a pair of ITRs flanking the gene therapy or expression construct. In some aspect, the gene therapy construct, expression construct, or viral vector disclosed herein is packaged in an AAV capsid (e.g., a recombinant AAV (rAAV)). In some aspects, the AAV serotype of the capsid is selected from AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAVrh8, AAV9, AAV10, AAVrhlO, or AAV.PHP.B.
[0024] In some aspects, the present disclosure provides methods for increasing proteasome activity in a cell and/or tissue comprising administering to the cell or tissue a composition comprising a gene therapy construct, expression cassette, viral vector, or rAAV particle disclosed herein.
[0025] In some aspects, the present disclosure provides methods for treating a disease or condition associated with proteinopathy in a subject in need thereof comprising administering to the subject a composition comprising a gene therapy construct or vector disclosed herein. In some aspects, the disease or condition is a neurodegenerative disease (e.g., Parkinson’s Disease or Huntington’s Disease), an ocular disease (e.g., retinitis pigmentosa or Kjer’s optic neuropathy), or a cardiac disease. In some aspect, the administration is tissue specific. In some aspects, the gene therapy construct or vector disclosed herein is administered to the brain, eye, or heart muscle. In some aspects, the disease or condition is Alzheimer’s disease, Dutch hereditary cerebral hemorrhage with amyloidosis, Congophilic angiopathy, Tauopathies, Corticobasal degeneration, Pick’s disease, Progressive supranuclear palsy, Prion diseases, Creutzfeld-Jacob disease, Gerstmann-Straussler-Schneiker syndrome, fatal familia insomnia, Kuru, Bovine spongiform encephalopathy, Scrapie, Chronic wasting disease, Synucleinopathies,
Parkinson’s disease, Lewy body variant of Alzheimer’s disease, Diffuse Lewy body disease, Dementia with Lewy bodies, Multiple system atrophy, Neurodegeneration with brain iron accumulation type I, Amyotrophic lateral sclerosis and frontotemporal lobar degeneration, Amyotrophic lateral sclerosis, Huntington’s disease, Hereditary dentatorubral-pallidoluysian atrophy, Kennedy’s disease or X-linked spinal and bulbar muscular atrophy, Spinocerebellar ataxia 1, Neuronal intranuclear inclusion disease, Spinocerebellar ataxia 2, Spinocerebellar ataxia 3, Spinocerebellar ataxia 6, Spinocerebellar ataxia 7, Spinocerebellar ataxia 17, Alexander’s disease, Cockayne syndrome, or Spinal muscular atrophy.
BRIEF DESCRIPTION OF FIGURES
[0026] FIGs. 1A-1C depict exemplary plasmids for expression of PA28alpha and
PA28beta. FIG. 1A depicts an exemplary plasmid for expression of PA28alpha comprising a CMV enhancer, a minimal CMV promoter, PA28alpha gene, and SV40 polyadenylation signal. FIG. IB depicts an exemplary plasmid for expression of PA28alpha and PA28beta comprising a minimal CMV promoter, a PA28beta gene, a SV40 polyadenylation signal, a CMV enhancer, a second minimal CMV promoter, a PA28alpha gene, and a second SV40 polyadenylation signal. FIG. 1C depicts an exemplary empty vector plasmid which does not express PA28alpha or PA28beta.
[0027] FIGs. 2A-2E show proteasome activity in HEK-293T cells transfected with plasmids expressing PA28alpha-only (FIG. 1A), PA28alpha/beta 11 S caps (FIG. IB), or an empty vector control (FIG. 1C). FIG. 2A depicts the proteasome assay, in which proteasome activity cleaves the artificial substrate Suc-LLVY-AMC (AMC) and generates free AMC which is fluorescent. Proteasome activity was analyzed with either 5 pL (FIG. 2B), 10 pL (FIG. 2C), 25 pL (FIG. 2D), or 50 pL (FIG. 2E) cell lysate.
[0028] FIGs. 3A-3C are western blots showing protein expression of PA28alpha (FIG.
3 A), PA28beta (FIG. 3B), or PA28alpha and PA28beta (FIG. 3C) in HEK-293T cells transfected with plasmids expressing PA28alpha-only (FIG. 1A), PA28alpha/beta 1 IS caps (FIG. IB), or an empty vector control (FIG. 1C).
[0029] FIGs. 4A-4M depict exemplary plasmids for expression of PA28alpha and
PA28beta. FIG. 4A depicts an exemplary plasmid for expression of PA28beta comprising a CMV promoter, PA28beta gene, and SV40 polyadenylation signal. FIG. 4B depicts an
exemplary plasmid for tandem expression of PA28alpha and PA28beta comprising a promoter, PA28alpha gene, an optimized WPRE (oPRE), a polyA tail, a second promoter, a PA28beta gene, a second optimized WPRE (oPRE), and a second polyA tail. FIG. 4C depicts an exemplary bidirectional plasmid for expression of PA28alpha and PA28beta comprising a poly A tail, a PA28alpha gene, a promoter, an enhancer, a second promoter, a PA28beta gene, and a second polyA tail. FIG. 4D depicts exemplary 1:1 coexpression of PA28alpha and PA28beta comprising a first plasmid comprising a promoter, a PA28alpha gene, an optimized WPRE (oPRE), and a poly A tail and a second plasmid comprising a promoter, a PA28beta gene, a second optimized WPRE (oPRE), and a polyA tail. FIG. 4E depicts an exemplary P2A self-cleavage plasmid for expression of PA28alpha and PA28beta comprising a promoter, a PA28alpha gene, a P2A site, a PA28beta gene, and a polyA tail. FIG. 4F depicts an exemplary P2A self-cleavage plasmid for expression of PA28alpha and PA28beta comprising a CMV promoter, a PA28alpha gene, a P2A site, a PA28beta gene, and a SV40 polyadenylation signal. FIG. 4G depicts an exemplary IRES self-cleavage plasmid for expression of PA28alpha and PA28beta comprising a promoter, a PA28alpha gene, a IRES site, a PA28beta gene, and a polyA tail. FIG. 4H depicts an exemplary IRES self-cleavage plasmid for expression of PA28alpha and PA28beta comprising a CMV promoter, a PA28alpha gene, an IRES site, a PA28beta gene, and a SV40 polyadenylation signal. FIG. 41 depicts an exemplary flexible linker plasmid for expression of PA28alpha and PA28beta comprising a promoter, a PA28alpha gene, a linker, a PA28beta gene, and a polyA tail. FIG. 4J depicts an exemplary flexible linker plasmid for expression of PA28alpha and PA28beta comprising a CMV promoter, a PA28alpha gene, a 3xGGGGS flexible linker, a PA28beta gene, and a SV40 polyadenylation signal.
[0030] FIGs. 5A-5C are western blots showing protein expression of PA28alpha (FIG.
5A), PA28beta (FIG. 5B), or PA28alpha and PA28beta (FIG. 5C) in HEK-293T cells transfected with either a PA28alpha and PA28beta bicistronic plasmid (FIG. 4C), a 1 : 1 ratio of a PA28alpha and PA28beta plasmid (FIG. 4D), a PA28alpha only plasmid, a PA28beta only plasmid, a PA28alpha and PA28beta IRES bicistronic plasmid (FIGs. 4G and 4H), a PA28alpha and PA28beta P2A bicistronic plasmid (FIG. 4E and 4F), or a PA28alpha and PA28beta flexible linker bicistronic plasmid (FIG. 41 and 4J).
[0031] FIGs. 6A and 6B are graphs showing the proteasome activity of HEK-293T cells transfected with either a PA28alpha and PA28beta bicistronic plasmid (FIG. 4C), a 1 : 1 ratio of a PA28alpha and PA28beta plasmid (FIG. 4D), a PA28alpha only plasmid, a PA28beta only plasmid, a PA28alpha and PA28beta IRES bicistronic plasmid (FIGs. 4G and 4H), a PA28alpha and PA28beta P2A bicistronic plasmid (FIG. 4E and 4F), or a PA28alpha and PA28beta flexible linker bicistronic plasmid (FIG. 41 and 4J).
[0032] FIGs. 7A-7J depict exemplary AAV expression vectors for expression of
PA28alpha and PA28beta. FIG. 7A depicts an exemplary AAV expression vector for tandem expression of PA28alpha and PA28beta comprising a 5' ITR, CBh promoter, PA28alpha gene, an optimized WPRE (oPRE), bGH polyadenylation signal, a second CBh promoter, a PA28beta gene, a second optimized WPRE (oPRE), a second bGH polyadenylation signal, and a 3' ITR. FIG. 7B depicts an exemplary AAV expression vector for tandem expression of PA28alpha and PA28beta comprising a 5' ITR, CBh promoter, a PA28alpha gene, a bGH polyadenylation signal, a second CBh promoter, a PA28beta gene, a second bGH polyadenylation signal, and a 3' ITR. FIG. 7C depicts an exemplary AAV expression vector for tandem expression of PA28alpha and PA28beta comprising a 5' ITR, hRHO promoter, a PA28alpha gene, a bGH polyadenylation signal, a second hRHO promoter, a PA28beta gene, a second bGH polyadenylation signal, and a 3' ITR. FIG. 7D depicts an exemplary AAV expression vector for tandem expression of PA28alpha and PA28beta comprising a 5' ITR, a hRHO promoter, a PA28alpha gene, an optimized WPRE (oPRE), a bGH polyadenylation signal, a second hRHO promoter, a PA28beta gene, a second optimized WPRE (oPRE), a second bGH polyadenylation signal, and a 3' ITR. FIG. 7E depicts an exemplary AAV expression vector for tandem expression of PA28alpha and PA28beta comprising a 5' ITR, a SYN1 promoter, a PA28alpha gene, a bGH polyadenylation signal, a second SYN 1 promoter, a PA28beta gene, a second bGH polyadenylation signal, and a 3' ITR. FIG. 7F depicts an exemplary AAV expression vector for tandem expression of PA28alpha and PA28beta comprising a 5' ITR, a SYN1 promoter, a PA28alpha gene, an optimized WPRE (oPRE), a bGH polyadenylation signal, a second SYN1 promoter, a PA28beta gene, a second optimized WPRE (oPRE), a second bGH polyadenylation signal, and a 3' ITR. FIG. 7G depicts an exemplary AAV expression vector for tandem expression of PA28alpha and PA28beta comprising a 5' ITR, a EFla promoter, a PA28alpha gene, a bGH polyadenylation signal,
a second EFla promoter, a PA28beta gene, a second bGH polyadenylation signal and a 3' ITR. FIG. 7H depicts an exemplary AAV expression vector for bicistronic expression of PA28alpha and PA28beta comprising an AAV2 ITR, an SV40 polyadenylation signal, a PA28beta gene, an hRHO promoter, a CMV enhancer, a second hRHO promoter, a PA28alpha gene, a second SV40 polyadenylation signal, and a second AAV2 ITR. FIG.
71 depicts an exemplary AAV expression vector for bicistronic expression of PA28alpha and PA28beta comprising an AAV2 ITR, an SV40 polyadenylation signal, an optimized WPRE (oPRE), a PA28beta gene, a SYN1 promoter, a CMV enhancer, a second SYN1 promoter, a PA28alpha gene, a second optimized WPRE (oPRE), a second SV40 polyadenylation signal, and a AAV2 ITR. FIG. 7J depicts an exemplary AAV expression vector for bicistronic expression of PA28alpha and PA28beta comprising an AAV2 ITR, a SV40 polyadenylation signal, a PA28beta gene, a SYN1 promoter, a CMV enhancer, a second SYN1 promoter, a PA28beta gene, a second SV40 polyadenylation signal, and a second AAV2 ITR.
[0033] FIGs. 8A-8C are western blots showing protein expression of PA28alpha (FIG.
8A), PA28beta (FIG. 8B), or PA28alpha and PA28beta (FIG. 8C) in HEK-293T cells transfected with either a wild-type pBI-CMV vector (ZsGreen) (FIG. 1C), a PA28alpha and PA28beta bicistronic plasmid (FIG. IB), a PA28alpha only plasmid, a PA28beta only plasmid, a 1 : 1 ratio of a PA28alpha and PA28beta plasmid (FIG. 4D), a PA28alpha and PA28beta tandem plasmid under the transcriptional control of a EFla promoter (FIG.
7G), a PA28alpha and PA28beta tandem plasmid under the transcriptional control of a hSynapsin promoter (FIG. 7J), a PA28alpha and PA28beta tandem plasmid under the transcriptional control of a hSynapsin promoter with an optimized WPRE (oPRE) sequence (FIG. 71), or a PA28alpha and PA28beta tandem plasmid under the transcriptional control of a CBh promoter (FIG. 7B).
[0034] FIGs. 9A-9B are graphs showing the proteasome activity of HEK-293T cells transfected with either a wild-type pBI-CMV vector (ZsGreen) (FIG. 1C), a PA28alpha and PA28beta bicistronic plasmid (pBi-PSMEl-PSME2) (FIG. IB), a PA28alpha only plasmid (PSMEl only), a PA28beta only plasmid (PSME2 only), a 1:1 ratio of a PA28alpha and PA28beta plasmid (1 : 1 PSMEl :PSME2) (FIG. 4D), a PA28alpha and PA28beta tandem plasmid under the transcriptional control of a EFla promoter (EFla tandem) (FIG. 7G), a PA28alpha and PA28beta tandem plasmid under the transcriptional
control of a hSynapsin promoter (hSyn-tandem) (FIG. IT), a PA28alpha and PA28beta tandem plasmid under the transcriptional control of a hSynapsin promoter with an optimized WPRE (oPRE) sequence (hSyn-OPRE tandem) (FIG. 71), or a PA28alpha and PA28beta tandem plasmid under the transcriptional control of a CBh promoter (CBh- tandem) (FIG. 7B).
[0035] FIGs. 10A-10B are western blots showing in vivo expression of PA28alpha and
PA28beta from mice administered PA28alpha and PA28beta constructs via subretinal injection. FIG. 10A shows expression of HSC70, beta-actin, PA28alpha, PA28beta, and GFP from mice injected in the left eye with an scAAV8 viral mixture comprising a 4:4: 1 (by volume) mixture of scAAV8-hRho-mPMSEl (PA28alpha) (titer: 5.38xl013 gc/mL), scAAV8-hRho-mPSME2 (PA28beta) (titer: 3.99xl013 gc/mL), and scAAV8-hRho-eGFP (titer: 4.49xl013 gc/mL). Lysate from the right eye was used as a control. FIG. 10B shows expression of HSC70, PA28alpha, or PA28beta from mice injected in the left eye with lpl of the scAAV8 vectors, AAV8-hRHO-oPRE tandem mPA28alpha/beta or AAV8- CBh tandem mA28alpha/beta via sub-retinal injection. Uninjected lysate and recombinant PA28alpha and PA28beta were used as controls.
[0036] FIGs. 11 A-l 1C are immunohistochemistry images showing localization of
PA28alpha in retinal tissue after mice administered PA28alpha and PA28beta constructs via subretinal injection. FIG. 11 A shows PA28alpha localization and GFP expression in the retina of mice administered a scAAV8 mixture comprising hRho- mPSMEl(PA28alpha), scAAV8-hRho-mPSME2(PA28beta), scAAV8-hRho-eGFP in a 16:16:1 ratio. FIG. 1 IB shows PA28alpha localization in the retina of mice administered lpl of AAV8 expressing both PA28alpha and PA28beta in the tandem configuration under control of human rhodopsin promoter (hRho) with oPRE elements after the transgenes (FIG. 7D) via sub-retinal injection. DAPI was used as a marker of the nucleus. FIG. llC shows localization of PA28alpha in mice administered 1 pL of an AAV8 expressing both PA28oc and RA28b in the tandem configuration under control of CBh promoter (FIG. 7B).
[0037] FIGs. 12A-12E are immunohistochemistry images showing localization of
PA28alpha and PA28beta in the brain tissue of mice administered PA28alpha and PA28beta constructs via injection into the substantia nigra. FIG. 12A shows localization of PA28alpha in the brain of mice administered a 1:1 mixture of scAAV9-hSyn-mPA28oc
and scAAV9-hSyn-mPA28 . FIGs. 12B and 12C show localization of PA28alpha (FIG. 12B) and PA28beta (FIG. 12C) in the brain of mice injected on one side with scAAV9- hSyn-mPA28oc (right side) and on the other side with scAAV9-hSyn-mPA28P (left side). FIGs. 12D and 12E show localization of PA28alpha (FIG. 12D) and PA28beta (FIG. 12E) in the brain of mice administered AAV9 expressing both PA28oc and RA28b in the tandem configuration under control of CBh promoter (FIG. 7B) (Left side) or EFloc promoter (FIG. 7G). Dopaminergic neurons are indicated by staining of tyrosine hydroxylase (TH).
DETAILED DESCRIPTION
I. Definitions
[0038] In order that the present disclosure can be more readily understood, certain terms are first defined. Additional definitions are set forth throughout the detailed disclosure.
[0039] It is to be noted that the term "a" or "an" entity refers to one or more of that entity; for example, "a nucleic acid sequence," is understood to represent one or more nucleic acid sequences, unless stated otherwise. As such, the terms "a" (or "an"), "one or more," and "at least one" can be used interchangeably herein.
[0040] Furthermore, "and/or", where used herein, is to be taken as specific disclosure of each of the two specified features or components with or without the other. Thus, the term "and/or" as used in a phrase such as "A and/or B" herein is intended to include "A and B," "A or B," "A" (alone), and "B" (alone). Likewise, the term "and/or" as used in a phrase such as "A, B, and/or C" is intended to encompass each of the following aspects: A, B, and C; A, B, or C; A or C; A or B; B or C; A and C; A and B; B and C; A (alone); B (alone); and C (alone).
[0041] It is understood that wherever aspects are described herein with the language
"comprising," otherwise analogous aspects described in terms of "consisting of' and/or "consisting essentially of are also provided.
[0042] The term "about" is used herein to mean approximately, roughly, around, or in the regions of. When the term "about" is used in conjunction with a numerical range, it modifies that range by extending the boundaries above and below the numerical values
set forth. In general, the term "about" can modify a numerical value above and below the stated value by a variance of, e.g., 10 percent, up or down (higher or lower).
[0043] The term "at least" prior to a number or series of numbers is understood to include the number adjacent to the term "at least," and all subsequent numbers or integers that could logically be included, as clear from context. For example, the number of nucleotides in a nucleic acid molecule must be an integer. For example, "at least 18 nucleotides of a 21 -nucleotide nucleic acid molecule" means that 18, 19, 20, or 21 nucleotides have the indicated property. When at least is present before a series of numbers or a range, it is understood that "at least" can modify each of the numbers in the series or range. "At least" is also not limited to integers (e.g., "at least 5%" includes 5.0%, 5.1%, 5.18% without consideration of the number of significant figures).
[0044] "Nucleic acid," "polynucleotide," and "oligonucleotide," are used interchangeably in the present application. These terms refer only to the primary structure of the molecule. Thus, these terms include double- and single-stranded DNA, as well as double- and single-stranded RNA. The terms "nucleic acid," "polynucleotide," and "oligonucleotide," as used herein, are defined as it is generally understood by the skilled person as a molecule comprising two or more covalently linked nucleosides. Such covalently bound nucleosides can also be referred to as nucleic acid molecules or oligomers. Polynucleotides can be made recombinantly, enzymatically, or synthetically, e.g., by solid-phase chemical synthesis followed by purification. When referring to a sequence of the polynucleotide or nucleic acid, reference is made to the sequence or order of nucleobase moieties, or modifications thereof, of the covalently linked nucleotides or nucleosides.
[0045] The term "mRNA," as used herein, refers to a single stranded RNA that encodes the amino acid sequence of one or more polypeptide chains.
[0046] As used herein, the term "polypeptide" is intended to encompass a singular
"polypeptide" as well as plural "polypeptides," and comprises any chain or chains of two or more amino acids. Thus, as used herein, a "peptide," a "peptide subunit," a "protein," an "amino acid chain," an "amino acid sequence," or any other term used to refer to a chain or chains of two or more amino acids, are included in the definition of a "polypeptide," even though each of these terms can have a more specific meaning. The term "polypeptide" can be used instead of, or interchangeably with any of these terms.
The term further includes polypeptides which have undergone post-translational or post synthesis modifications, for example, conjugation of a palmitoyl group, glycosylation, acetylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, or modification by non-naturally occurring amino acids.
The term "peptide," as used herein encompasses full length peptides and fragments, variants or derivatives thereof.
[0047] "Percent (%) sequence identity" with respect to a reference polynucleotide or polypeptide sequence is defined as the percentage of nucleic acids or amino acids in a candidate sequence that are identical to the nucleic acids or amino acids in the reference polynucleotide or polypeptide sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity. Alignment for purposes of determining percent nucleic acid or amino acid sequence identity can be achieved in various ways that are within the capabilities of one of skill in the art, for example, using publicly available computer software such as BLAST, BLAST-2, or Megalign software. Those skilled in the art can determine appropriate parameters for aligning sequences, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared. For example, percent sequence identity values can be generated using the sequence comparison computer program BLAST.
[0048] A "coding sequence" or a sequence "encoding" a particular molecule (e.g., a therapeutic protein or peptide) is a nucleic acid that is transcribed (in the case of DNA) or translated (in the case of mRNA) into polypeptide, in vitro or in vivo , when operably linked to an appropriate regulatory sequence, such as a promoter. The boundaries of the coding sequence are determined by a start codon at the 5' (amino) terminus and a translation stop codon at the 3' (carboxy) terminus. A coding sequence can include, but is not limited to, cDNA from prokaryotic or eukaryotic mRNA, genomic DNA sequences from prokaryotic or eukaryotic DNA, and synthetic DNA sequences. A transcription termination sequence will usually be located 3' to the coding sequence.
[0049] As used herein, the term "promoter" refers to a DNA sequence recognized by the machinery of a cell, or introduced synthetic machinery, required to initiate the specific transcription of a coding sequence (e.g., a gene). The term "promoter" is also meant to encompass those nucleic acid elements sufficient for promoter-dependent gene expression controllable for cell-type specific, tissue-specific or inducible by external signals or
agents; such elements can be located in the 5' or 3' regions of the native gene. In some aspects, the promoter is a constitutively active promoter, a cell-type specific promoter, or an inducible promoter.
[0050] As used herein, the term "enhancer" is a cis-acting element that stimulates or inhibits transcription of adjacent genes. An enhancer that inhibits transcription is also referred to as a "silencer." Enhancers can function (e.g., can be associated with a coding sequence) in either orientation, over distances of up to several kilobase pairs (kb) from the coding sequence and from a position downstream of a transcribed region.
[0051] As used herein, the term "regulatable promoter" is any promoter whose activity is affected by a cis or trans acting factor (e.g., an inducible promoter, such as an external signal or agent).
[0052] As used herein, the term "constitutive promoter" is any promoter that directs RNA production in many or all tissue/cell types at most times, e.g., the human CMV immediate early enhancer/promoter region that promotes constitutive expression of cloned DNA inserts in mammalian cells.
[0053] The terms "transcriptional regulatory protein," "transcriptional regulatory factor," and "transcription factor" are used interchangeably herein, and refer to a nuclear protein that binds a DNA response element and thereby transcriptionally regulates the expression of an associated gene or genes. Transcriptional regulatory proteins generally bind directly to a DNA response element, however in some cases binding to DNA can be indirect by way of binding to another protein that in turn binds to, or is bound to a DNA response element.
[0054] As used herein, the term "termination signal sequence" can be any genetic element that causes RNA polymerase to terminate transcription, such as for example a polyadenylation signal sequence. A polyadenylation signal sequence is a recognition region necessary for endonuclease cleavage of an RNA transcript that is followed by the polyadenylation consensus sequence AATAAA. A polyadenylation signal sequence provides a "polyA site," i.e., a site on a RNA transcript to which adenine residues will be added by post-transcriptional polyadenylation.
[0055] The terms "operatively linked," "operatively inserted," "operatively positioned,"
"under control" or "under transcriptional control" means that the promoter is in the correct location and orientation in relation to the nucleic acid to control RNA polymerase
initiation and expression of the gene. The term "operably linked" means that a DNA sequence and a regulatory sequence(s) are connected in such a way as to permit gene expression when the appropriate molecules (e.g., transcriptional activator proteins) are bound to the regulatory sequence(s). The term "operably inserted" means that the DNA of interest introduced into the cell is positioned adjacent a DNA sequence which directs transcription and translation of the introduced DNA (i.e., facilitates the production of, e.g., a polypeptide encoded by a DNA of interest).
[0056] The term “expression cassette” or “expression construct" means any type of genetic construct containing a nucleic acid in which part or all of the nucleic acid encoding sequence is capable of being transcribed.
[0057] The term “cistron” refers to a nucleic acid cassette sufficient for expression of a gene product. In some aspects, a cistron is an expression cassette. Accordingly, some aspects of this invention provide nucleic acid constructs comprising two or more cistrons (e.g., multi cistronic, e.g., bicistronic), for example, two or more expression cassettes.
[0058] As used herein, the term "delivery vector" or "vector" refers to any vehicle for the transfer of a nucleic acid (e.g., for cloning or transfer into a host cell). In some aspects the vector can be a plasmid, phage, transposon, cosmid, chromosome, artificial chromosome, virus, virion, etc. A vector can be a replicon to which another nucleic acid segment can be attached so as to bring about the replication of the attached segment. A "replicon" refers to any genetic element (e.g., plasmid, phage, cosmid, chromosome, virus) that functions as an autonomous unit of replication in vivo , i.e., capable of replication under its own control. The term "delivery vector" or "vector" includes both viral and nonviral vehicles for introducing the nucleic acid into a cell in vitro, ex vivo or in vivo. In some aspects, insertion of a polynucleotide into a suitable vector can be accomplished by ligating the appropriate polynucleotide fragments into a chosen vector that has complementary cohesive termini. Vectors can be engineered to encode selectable markers or reporters that provide for the selection or identification of cells that have incorporated the vector. In some aspects, the delivery vector is a viral vector (e.g., an AAV vector or lentivirus vector).
[0059] Some aspects of the disclosure are directed to delivery vectors for gene therapy, which can include, e.g., viruses, particularly attenuated and/or replication-deficient viruses. In some aspects, the delivery vector of the disclosure is a viral vector selected
from the group consisting of an adeno-associated viral (AAV) vector, an adenoviral vector, a lentiviral vector, or a retroviral vector. In some aspects, the deliver vector is a non-viral vector, a plasmid, a lipid-DNA micelle or liposome, or an exosome.
[0060] The term "adeno-associated virus vector" or "AAV vector" as used herein refers to any vector that comprises or derives from components of an adeno-associated vector and is suitable to infect mammalian cells, preferably human cells. The term AAV vector typically designates an AAV-type viral particle or virion comprising a payload. The AAV vector can be derived from various serotypes, including combinations of serotypes (i.e., "pseudotyped" AAV) or from various genomes (e.g., single stranded or self- complementary). In addition, the AAV vector can be replication defective and/or targeted. As used herein, the term "adeno-associated virus" (AAV), includes but is not limited to, AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAVrh8, AAV9, AAV10, AAVrhlO, and AAV.PHP.B.
[0061] As used herein, a "recombinant AAV particle" or a "rAAV particle" is an AAV virus capsid that comprises an AAV vector having at least one payload region (e.g., a polynucleotide encoding a therapeutic protein or peptide) and at least one inverted terminal repeat (ITR) region.
[0062] The term "derived from," as used herein, refers to a component that is isolated from or made using a specified molecule or organism, or information (e.g., amino acid or nucleic acid sequence) from the specified molecule or organism.
[0063] In the case of a polynucleotide disclosed herein, the derived species can be obtained by, for example, naturally occurring mutagenesis, artificial directed mutagenesis or artificial random mutagenesis. The mutagenesis used to derive polynucleotides can be intentionally directed or intentionally random, or a mixture of each. The mutagenesis of a polynucleotide to create a different polynucleotide derived from the first can be a random event (e.g., caused by polymerase infidelity) and the identification of the derived polynucleotide can be made by appropriate screening methods.
[0064] As used herein, the term "modified" refers to a changed state or structure of a molecule of the disclosure. Molecules can be modified in many ways including chemically, structurally, and functionally.
[0065] As used herein, the term "mutation" refers to any changing of the structure of a gene, resulting in a variant (also called "mutant") form that can be transmitted to
subsequent generations. Mutations in a gene can be caused by the alternation of single base in DNA, or the deletion, insertion, or rearrangement of larger sections of genes or chromosomes.
[0066] As used herein, the term "synthetic" means produced, prepared, and/or manufactured by the hand of man. Synthesis of polynucleotides or polypeptides or other molecules of the present disclosure can be chemical or enzymatic.
[0067] As used herein, the term "administration" refers to the administration of a composition of the present disclosure (e.g., a gene therapy construct, vector, or composition disclosed herein) to a subject or system. Administration to an animal subject (e.g., to a human) can be by any appropriate route.
[0068] The phrase "contacting a cell" (e.g., contacting a cell with a gene therapy construct, vector, or composition of the disclosure) as used herein, includes contacting a cell directly or indirectly. In some aspects, contacting a cell includes contacting a cell in vitro or in vivo. Thus, for example, the gene therapy construct, vector, or composition can be put into physical contact with the cell by the individual performing the method, or alternatively, the gene therapy construct, vector, or composition can be put into a situation that will permit or cause it to subsequently come into contact with the cell.
[0069] As used herein, the terms "effective amount," "therapeutically effective amount," and a "sufficient amount" of a therapeutic agent, e.g., a gene therapy construct, vector, or composition disclosed herein, refers to a quantity sufficient to, when administered to the subject, including a human, effect beneficial or desired results, including clinical results, and, as such, an "effective amount" or synonym thereto depends on the context in which it is being applied. In some aspects, a therapeutically effective amount of an agent is an amount that results in a beneficial or desired result in a subject as compared to a control.
[0070] The amount of a given agent (e.g., a gene therapy construct, vector, or composition disclosed herein) will correspond to such an amount will vary depending upon various factors, such as the given agent, the pharmaceutical formulation, the route of administration, the type of disease or disorder, the identity of the subject (e.g., age, sex, and/or weight) or host being treated, and the like.
[0071] As used herein, the term "gene therapy" is the insertion of nucleic acid sequences
(e.g., a polynucleotide comprising a promoter operably linked to a nucleic acid encoding a therapeutic molecule as disclosed herein) into an individual's cells and/or tissues to
treat, reduce the symptoms of, or reduce the likelihood of a disease. Gene therapy also includes insertion of transgene that are activating in nature, i.e., that activate, increase or cause expression, activity or function of an endogenous gene or protein, such as a desirable or gene or protein. Such transgenes can be exogenous. An exogenous molecule or sequence is understood to be molecule or sequence not normally occurring in the cell, tissue and/or individual to be treated. Both acquired and congenital diseases are amenable to gene therapy.
[0072] The term "prophylactically effective amount," as used herein, includes the amount of an agent, (e.g., a gene therapy construct, vector or composition disclosed herein) that, when administered to a subject having or predisposed to have a disease or disorder (e.g., a neurological disease or disorder (e.g., Parkinson’s Disease (PD) or Huntington’s Disease (HD) or ocular diseases or disorders (e.g., retinitis pigmentosa)) is sufficient to prevent, reduce the symptoms of, or ameliorate the disease or disorder or one or more symptoms of the disease or disorder. Ameliorating the disease or disorder includes slowing the course of the disease or disorder or reducing the severity of later-developing disease or disorder. The "prophylactically effective amount" can vary depending on the characteristics of the agent, how the agent is administered, the degree of risk of disease, and the history, age, weight, family history, genetic makeup, the types of preceding or concomitant treatments, if any, and other individual characteristics of the patient to be treated.
[0073] As used herein, the term "in vitro " refers to events that occur in an artificial environment, e.g., in a test tube or reaction vessel, in cell culture, in a Petri dish, etc., rather than within an organism (e.g., animal, plant, or microbe).
[0074] As used herein, the term "in vivo " refers to events that occur within an organism
(e.g., animal, plant, or microbe or cell or tissue thereof).
[0075] As used herein, the term "transfection" refers to methods to introduce exogenous nucleic acids into a cell. Methods of transfection include, but are not limited to, chemical methods, physical treatments and cationic lipids or mixtures.
[0076] By "level" is meant a level or activity of a protein, or mRNA encoding the protein, optionally as compared to a reference. The reference can be any useful reference, as defined herein. By a "decreased level" or an "increased level" of a protein or expression is meant a decrease or increase in protein or expression level, as compared to a reference.
[0077] By a "reference" is meant any useful reference used to compare protein or mRNA levels or activity. The reference can be any sample, standard, standard curve, or level that is used for comparison purposes. The reference can be a normal reference sample or a reference standard or level.
[0078] The term "pharmaceutical composition," as used herein, represents a composition comprising a therapeutic agent disclosed herein, e.g., a gene therapy construct, vector or composition disclosed herein, formulated with a pharmaceutically acceptable excipient, and can be manufactured or sold with the approval of a governmental regulatory agency as part of a therapeutic regimen for the treatment of disease in a mammal.
[0079] A "pharmaceutically acceptable excipient," as used herein, refers to any ingredient other than the therapeutic agent described herein (for example, a vehicle capable of suspending an active compound) and having the properties of being substantially nontoxic and non-inflammatory in a patient.
[0080] As used herein, the term "subject" refers to any organism to which an agent or composition disclosed herein can be administered, e.g., for experimental, diagnostic, prophylactic, and/or therapeutic purposes. Typical subjects include any animal (e.g., mammals such as mice, rats, rabbits, non-human primates, and humans). A subject can seek or be in need of treatment, require treatment, be receiving treatment, be receiving treatment in the future, or be a human or animal who is under care by a trained professional for a particular disease or condition.
[0081] As used herein, the terms "treat," "treated," and "treating" mean both therapeutic treatment and prophylactic or preventative measures wherein the object is to prevent or slow down (lessen) an undesired physiological condition, disorder, or disease, or obtain beneficial or desired clinical results. In some aspects, treating reduces or lessens the symptoms associated with a disease or disorder. In some aspects, the treating results in a beneficial or desired clinical result.
[0082] Beneficial or desired clinical results include, but are not limited to, alleviation of symptoms; diminishment of the extent of a condition, disorder, or disease; stabilized (i.e., not worsening) state of condition, disorder, or disease; delay in onset or slowing of condition, disorder, or disease progression; amelioration of the condition, disorder, or disease state or remission (whether partial or total), whether detectable or undetectable; an amelioration of at least one measurable physical parameter, not necessarily discernible
by the patient; or enhancement or improvement of condition, disorder, or disease. In some aspects, treatment includes eliciting a clinically significant response without excessive levels of side effects. In some aspects, treatment includes prolonging survival as compared to expected survival if not receiving treatment. As used herein, the term "amelioration" or "ameliorating" refers to a lessening of severity of at least one indicator of a condition or disease. As used herein, the term "preventing" or "prevention" refers to delaying or forestalling the onset, development or progression of a condition or disease for a period of time, including weeks, months, or years.
II. Proteasome Regulatory Subunits
[0083] The present disclosure provides constructs (e.g., expression constructs) comprising a polynucleotide comprising a promoter operably linked to a nucleic acid encoding a PSME (e.g., a PA28A alone, PA28B, or a PA28A in combination with PA28B).
[0084] Proteasomes are multi-subunit complexes assembled upon association of two principal components: the 20S core and the regulatory caps. The 20S core is responsible for the entire proteolytic process however, its basal activity is very low in the absence of regulatory caps. The most common caps are 19S and 1 IS, which facilitate protein degradation in ubiquitin-dependent and ubiquitin-independent manners, respectively. A single 20S core may associate with one or two caps (either identical or different) at the sites located at the opposite ends of the 20S barrel-like structure. Binding of the 1 IS cap to the ends of the 20S core facilitates substrate access and proteolytic activities confined inside the 20S barrel-like structure. The 1 IS cap can be formed by a hetero-heptameric ring consisting of PA28A and PA28B (also known as the PA28 complex). However, functional 1 IS caps can also be formed by PA28A alone.
[0085] Without being bound by theory, misfolded proteins can accumulate when the proteasome is impaired and/or when the demand for proteasome function surpasses the functional capacity of proteasomes. Such accumulation leads to proteinopathies. As disclosed herein, transgene delivery of a PA28A alone or a combination of a PA28A and PA28B to a cell or tissue, can increase proteasome capacity and thereby reduce the amount of aberrant protein aggregation in the targeted cell or tissue. In some aspects, delivery of the constructs disclosed herein can be useful for treating proteinopathies (e.g., Huntington’s Disease, Parkinson’s Disease, ocular diseases, and cardiac proteinopathies).
[0086] In some aspects, a polynucleotide encoding a PSME (e.g., a PA28A alone,
PA28B, or a PA28A in combination with PA28B) can delivered to the cell or tissue using a viral vector (e.g., an AAV vector) disclosed herein.
[0087] Proteasome activator complex subunit 1, generally abbreviated PA28A or PSME1 herein (also known as 1 IS regulator complex subunit alpha, REG-alpha, activator of multi catalytic protease subunit 1, interferon gamma up-regulated 1-5111 protein, Proteasome activator 28 subunit alpha, P28a, and P28alpha) is a protein which in humans is encoded by the PSME1 gene.
[0088] Th ePSMEl gene is located on chromosome 14 (bases 24,136,194 to 24,138,962;
NCBI Reference Sequence NC_000014.9).
[0089] PA28A has three isoforms produced by alternative splicing. The wild-type amino acid sequences for the PA28A isoforms are shown in Table 1 below. Furthermore, the last tyrosine (Y) residue of isoform 1 can be mutated to tryptophan (W), which has been shown to have some increased activity in some in vitro assays (see Song, X. et al, JBC 272(44):P27994-28000 (1997)).
[0090] In some aspects, the PA28A comprises an amino acid sequence having a sequence of at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity to SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, or SEQ ID NO: 4. In some aspects, the PA28A comprises an amino acid sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, or SEQ ID NO: 4.
[0091] In some aspects, the nucleic acid encoding PA28A comprises a sequence having at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity to SEQ ID NO: 5, SEQ ID NO: 6, or SEQ ID NO: 7. In some aspects, the nucleic acid encoding PA28A comprises SEQ ID NO: 5, SEQ ID NO: 6, or SEQ ID NO: 7. In certain aspects, the wild-type nucleic acid sequences can be modified to improve expression, manufacturability, and/or reduce immunologic risk. In some aspects, the PA28A nucleotide sequences are shown in Table 2 below.
[0092] Proteasome activator complex subunit 2, generally abbreviated PA28B or PSME2 herein (also known as 1 IS regulator complex subunit beta, REG-beta, Activator of multi catalytic protease subunit 2, Proteasome activator 28 subunit beta, PA28b, and PA28beta) is a protein which in humans is encoded by the PSME2 gene. The PSME2 gene is located on chromosome 14 (bases 24,143,365 to 24,146,646; NCBI Reference Sequence NC_000014.9).
[0093] The wild-type amino acid sequence for PA28B is shown in Table 3 below.
[0094] In some aspects, the PA28B comprises an amino acid sequence having a sequence of at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity to SEQ ID NO: 8. In some aspects, the PA28A comprises an amino acid sequence of SEQ ID NO: 8.
[0095] In some aspects, the nucleic acid encoding PA28B comprises a sequence having at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity to SEQ ID NO: 9. In some aspects, the nucleic acid encoding PA28B comprises SEQ ID NO: 9. In certain aspects, the wild-type nucleic acid sequence can be modified to improve expression, manufacturability, and/or reduce immunologic risk. In some aspects, the nucleotide sequence encoding PA28B is shown in Table 4 below.
III. Constructs
[0096] Some aspects of the disclosure are directed to a nucleic acid construct or an expression construct (e.g., comprising one or more expression cassettes) having a promoter operably linked to a DNA of interest that encodes a PSME disclosed herein (e.g., a PA28A alone or a PA28A in combination with PA28B, with or without a second promoter). In some aspects, the constructs comprise a DNA sequence (or the corresponding RNA sequence) can be used in accordance with the disclosure and can be any eukaryotic expression construct containing a DNA or RNA of interest. For example, a plasmid or viral construct (e.g., an AAV vector) can be cleaved to provide linear DNA having ligatable termini. These termini are bound to exogenous DNA having
complementary, like ligatable termini to provide a biologically functional DNA molecule having an intact replicon and a desired phenotypic property. In some aspects, the construct is capable of replication in both eukaryotic and prokaryotic hosts.
[0097] In some aspects, the constructs disclosed herein contains a promoter to facilitate expression of the DNA of interest within a certain cell type and/or tissue. In some aspects, the promoter is a heterologous promoter. The term “heterologous promoter”, as used herein, refers to a promoter that does is not found to be operatively linked to a given encoding sequence in nature. In some aspects, the promoter is a ubiquitous promoter or a tissue specific promoter. In some aspects the promoter is selected from a MecP2 promoter, a GUSB promoter, a CBh promoter, a PGK promoter, a human rhodopsin (hRho) kinase promoter, a cone arrestin promoter, a tyrosine hydroxylase promoter, a dopamine transporter promoter, a GRK1 promoter, a cardiac myosin light chain 2 promoter, a CAMKII promoter, a hDlx promoter, a CMV promoter, a CAG promoter, a bidirectional CAG promoter, a CBA promoter, a human ubiquitin C promoter, synapsin (Syn) promoter, a human synapsin (hSyn) promoter, a EFla promoter, or a rhodopsin (Rhod). In some aspects, the promoter is a rhodopsin (Rhod) promoter. In some aspects, the promoter is a human rhodopsin (hRho) promoter. In some aspects, the promoter is synapsin (Syn) promoter. In some aspects, the promoter is a human synapsin (hSyn) promoter. In some aspects, the promoter is selected from a hSyn or a pRhod promoter. In some aspects, the promoter is a EFla promoter. In some aspects, the promoter is a CBh promoter.
[0098] In some aspects, the human synapsin promoter has at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or 100% identity to SEQ ID NO: 16. In some aspects, the human synapsin promoter has the sequence of SEQ ID NO: 16.
[0099] In some aspects, the human rhodopsin promoter has at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or 100% identity to SEQ ID NO: 13. In some aspects, the human rhodopsin promoter has the sequence of SEQ ID NO: 13.
[0100] In some aspects, the EFla promoter has at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least
95%, or 100% identity to SEQ ID NO: 17. In some aspects, the EFla promoter has the sequence of SEQ ID NO: 17.
[0101] In some aspects, the CBh promoter has at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or 100% identity to SEQ ID NO: 18. In some aspects, the CBh promoter has the sequence of SEQ ID NO: 18.
[0102] In some aspects, the nucleic acid construct or an expression construct (e.g., comprising an expression cassette) can further comprise one or more additional elements, for example, an enhancer, a polyadenylation site, a woodchuck hepatitis virus post- transcriptional regulatory element (WPRE), an optimized woodchuck hepatitis virus post- transcriptional regulatory element (oPRE), an intron, a microRNA binding motif, and/or other elements known to affect expression levels of the encoding sequence. In some aspects, the nucleic acid construct or expression construct comprises an optimized woodchuck hepatitis virus post-transcriptional regulatory element (oPRE). In some aspects, the nucleic acid construct or expression construct comprises a CMV enhancer.
[0103] In some aspects, the oPRE has at least 50%, at least 55%, at least 60%, at least
65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or 100% identity to SEQ ID NO: 15. In some aspects, the oPRE has the sequence of SEQ ID NO: 15.
[0104] In some aspects, the CMV enhancer has at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or 100% identity to SEQ ID NO: 19. In some aspects, the CMV enhancer has the sequence of SEQ ID NO: 19.
[0105] In some aspects, the nucleic acid construct comprises a chimeric intron. In some aspects, the nucleic acid construct comprises a beta-globin intron. In some aspects, the nucleic acid construct comprises minute virus of mice (MVM) intron.
[0106] In some aspects, the nucleic acid construct comprises a polyA sequence selected from the group consisting of a beta-globin polyA, a bovine growth hormone polyA (bGH polyA), a human growth hormone polyA (hGH polyA), and a SV40 immediate early poly A (SV40 polyA). In some aspects, the nucleic acid construct or expression construct comprises a bovine growth hormone polyA (bGH polyA).
[0107] In some aspects, the bovine growth hormone polyA (bGH poly A) has at least
50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or 100% identity to SEQ ID NO: 14. In some aspects, the bovine growth hormone polyA (bGH polyA) has the sequence of SEQ ID NO: 14.
[0108] In some aspects, the nucleic acid construct comprises a 5’ and/or a 3’ untranslated region (UTR). In some aspects, the nucleic acid construct can further comprise a microRNA binding motif in the 3’ untranslated region (UTR). In some aspects, the nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprises a woodchuck hepatitis virus post-transcriptional regulatory element (WPRE). In some aspects, the nucleic acid construct of an expression construct (e.g., comprising an expression cassette) comprises an optimized woodchuck hepatitis virus post- transcriptional regulatory element (oPRE). In some aspects, the nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprises a polynucleotide comprising a promoter (e.g., hSyn or pRhod promoter), a nucleic acid encoding a PSME (e.g., a PA28A or PA28A in combination with PA28B), and a WPRE. In some aspects, the nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprises a polynucleotide comprising a promoter (e.g., hSyn or pRhod promoter), a nucleic acid encoding a PSME (e.g., a PA28A or PA28A in combination with PA28B), and an optimized woodchuck hepatitis virus post- transcriptional regulatory element (oPRE).
[0109] In some aspects, the disclosure provides nucleic acids encoding a PSME (e.g., a
PA28A or PA28A in combination with PA28B) that is a multi cistronic (e.g., bicistronic) construct (e.g., comprising PA28A and PA28B). In some aspects, the multi cistronic (e.g., bicistronic) construct comprises a bi-directional promoter. In some aspects, the multi cistronic (e.g., bicistronic) construct comprises tandem expression cassettes each with a promoter, a transgene (encoding PA28A or PA28B), and polyA. In some aspects, the multi cistronic (e.g., bicistronic) construct further comprises a cleavage element between the nucleic acid encoding a PA28A and the nucleic acid encoding PA28B. In some aspects, the multicistronic (e.g., bicistronic) construct further comprises a cleavage element between an expression construct comprising a nucleic acid encoding a PA28A and an expression construct comprising the nucleic acid encoding PA28B. In some
aspects, the cleavage element comprises a nucleic acid encoding self-cleavage peptide. In some aspects, the cleavage peptide is a 2A self-cleaving peptide (P2A, T2A, etc.). In some aspects, the cleavage site is a furin cleavage site, e.g., with the consensus sequence of RXK(R)R (SEQ ID NO: 10). In some aspects, the cleavage site is an intein-F2A peptide. See Zhang et al., Plant Biotechnol J. (2017) 15, pp.718-728.
[0110] In some aspects, the multi cistronic (e.g., bicistronic) construct comprises first expression cassette comprising a polynucleotide encoding PA28A and a second expression cassette comprising a polynucleotide encoding PA28B. See, e.g., Kugler et al., Virology 311 (2003) 89-95. In some aspects, the first and second expression cassettes comprise the same promoter. In some aspects, the first and second expression cassettes comprise different promoters. In some aspects, the first expression cassette comprises a hSyn promoter and the second expression cassette comprises a CMV promoter. In some aspects, the first expression cassette comprises a CMV promoter and the second expression cassette comprises a hSyn promoter. In some aspects, the first and/or second expression cassettes comprise a polyA (e.g., bGH polyadenylation site, hGH polyadenylation site, or a SV40 polyadenylation site). In some aspects, the first and/or second expression cassette can further comprise a WPRE. In some aspects, the first and/or second expression cassette can further comprise an optimized WPRE (oPRE).
[0111] In some aspects, the construct is a bicistronic nucleic acid construct comprising a first nucleic acid encoding a PA28A and a second nucleic acid encoding PA28B. In some aspects, the construct is a bicistronic nucleic acid construct comprising a first nucleic acid encoding a PA28A, a second nucleic acid encoding PA28B, and a nucleic acid encoding a cleavage peptide located between the first and second nucleic acid. In some aspects, the bicistronic construct further comprises a WPRE. In some aspects, the bicistronic construct further comprises an optimized WPRE (oPRE). In some aspects, the construct is a bicistronic nucleic acid construct comprising a first nucleic acid encoding a PA28A, a second nucleic acid encoding PA28B, a nucleic acid encoding a cleavage peptide located between the first and second nucleic acid, and a WPRE. In some aspects, the construct is a bicistronic nucleic acid construct comprising a first nucleic acid encoding a PA28A, a second nucleic acid encoding PA28B, a nucleic acid encoding a cleavage peptide located between the first and second nucleic acid, and an optimized WPRE (oPRE).
[0112] Some aspects of this invention provide bicistronic expression constructs comprising two or more expression cassettes in various configurations.
[0113] In some aspects, bicistronic expression constructs are provided in which the expression cassettes are positioned in different ways. For example, in some aspects, a multi cistronic expression construct is provided in which a first expression cassette is positioned adjacent to a second expression cassette. In some embodiments, the first expression cassette and the second expression cassette are operably linked by a bidirectional promoter, wherein the first expression cassette and the second expression cassette are flanked by adeno-associated virus (AAV) inverted terminal repeats (ITRs).
[0114] In some aspects, bicistronic expression constructs are provided in which the expression cassettes are oriented in different ways. For example, in some aspects, bicistronic expression construct is provided comprising a first and a second expression cassette in opposite orientations.
[0115] The term “orientation” as used herein in connection with expression cassettes, refers to the directional characteristic of a given cassette or structure. In some aspects, an expression cassette harbors a promoter 5' of the encoding nucleic acid sequence, and transcription of the encoding nucleic acid sequence runs from the 5' terminus to the 3' terminus of the sense strand, making it a directional cassette (e.g., 5'- promoter/(intron)/encoding sequence-3'). Since virtually all expression cassettes are directional in this sense, those of skill in the art can easily determine the orientation of a given expression cassette in relation to a second nucleic acid structure, for example, a second expression cassette, a viral genome, or, if the cassette is comprised in an AAV construct, in relation to an AAV ITR.
[0116] In some aspects, bicistronic expression constructs are provided allowing for efficient expression of two or more encoding nucleic acid sequences. In some aspects, the bicistronic expression construct comprises two expression cassettes. In some aspects, the bicistronic expression construct provided is a recombinant AAV (rAAV) construct.
[0117] In some aspects, PA28A and PA28B are expressed in a single open reading frame.
In some aspects, an amino acid sequence or linker containing a high efficiency cleavage site is disposed between the PA28A and PA28B. As used herein, high cleavage efficiency is defined as more than 50%, more than 70%, more than 80%, or more than 90% of the
translated protein is cleaved. Cleavage efficiency can be measured by Western Blot analysis.
[0118] In some aspects, the expression construct disclosed herein encodes PA28A- cleavage sequence-PA28B in 5'-3' orientation. In some aspects, the expression construct encodes PA28B-cleavage sequence-PA28A in 5'-3' orientation. In some aspects, the nucleic acid sequence of the construct comprises a promoter, PA28A coding sequence, a cleavage sequence, and a PA28B coding sequence in 5'-3' orientation. In some aspects, the nucleic acid sequence of the construct comprises a promoter, a PA28B coding sequence, cleavage sequence, and aPA28A coding sequence in 5'-3' orientation. In some aspects the cleavage sequence a P2A, a furin, or an intein-F2A cleavage sequence.
[0119] In some aspects, the expression construct disclosed herein encodes PA28A-P2A-
PA28B in 5'-3' orientation. In some aspects, the expression construct encodes PA28B- P2A-PA28A in 5'-3' orientation. In some aspects, the nucleic acid sequence of the construct comprises a promoter, a PA28A coding sequence, a P2A sequence, and a PA28B coding sequence in 5'-3' orientation. In some aspects, the nucleic acid sequence of the construct comprises a promoter, a PA28B coding sequence, a P2A sequence, and a PA28A coding sequence in 5'-3' orientation. In some aspects, the promoter is a bi directional promoter. In some aspects the promoter is selected from a MecP2 promoter, a GUSB promoter, a CBh promoter, a PGK promoter, a EFla promoter, a human rhodopsin (hRho) kinase promoter, a cone arrestin promoter, a tyrosine hydroxylase promoter, a dopamine transporter promoter, a GRK1 promoter, a cardiac myosin light chain 2 promoter, a CAMKII promoter, a hDlx promoter, a CMV promoter, a CAG promoter, a bidirectional CAG promoter, a CBA promoter, a human ubiquitin C promoter, a human synapsin (hSyn) promoter, synapsin (Syn) promoter, or a rhodopsin (Rhod). In some aspects, the expression construct comprises a synapsin promoter. In some aspects, the expression construct comprises a rhodopsin promoter. In some aspects, the expression construct comprises a human rhodopsin promoter. In some aspects, the expression construct comprises a human synapsin (hSyn) promoter. In some aspects, the expression construct comprises a CBh promoter. In some aspects, the expression construct comprises a EFla promoter.
[0120] In some aspects, the expression construct further comprises an enhancer, a polyadenylation site, a woodchuck hepatitis virus post-transcriptional regulatory element
(WPRE), an optimized woodchuck hepatitis virus post-transcriptional regulatory element (oPRE), an intron, a microRNA binding motif, and/or other element(s) known to affect expression levels of the encoding sequence. In some aspects, the enhancer is a CMV enhancer.
[0121] In some aspects, the nucleic acid construct comprises a chimeric intron. In some aspects, the nucleic acid construct comprises a beta-globin intron. In some aspects, the nucleic acid construct comprises minute virus of mice (MVM) intron.
[0122] In some aspects, the nucleic acid construct comprises polyA sequence selected from the group consisting of beta-globin polyA, bovine growth hormone polyA (bGH polyA), human growth hormone polyA (hGH polyA), and SV40 immediate early poly A (SV40 polyA). In some aspects, the nucleic acid construct comprises bovine growth hormone polyA (bGH polyA).
[0123] In some aspects, the nucleic acid construct comprises a 5’ and/or a 3’ untranslated region (UTR). In some aspects, the nucleic acid construct comprises a microRNA binding motif in the 3’ untranslated region (UTR).
[0124] In some aspects, the expression construct further comprises a WPRE. In some aspects, the expression construct further comprises an optimized WPRE (oPRE).
[0125] In some aspects, the constructs of the disclosure can also include other components such as a marker (e.g., an antibiotic resistance gene (such as an ampicillin resistance gene) or b-galactosidase) to aid in selection of cells containing and/or expressing the construct, an origin of replication for stable replication of the construct in a bacterial cell (preferably, a high copy number origin of replication), a nuclear localization signal, or other elements which facilitate production of the DNA construct, the protein encoded thereby, or both.
[0126] For eukaryotic expression, the construct can comprise at a minimum a eukaryotic promoter operably linked to a DNA of interest, which is in turn operably linked to a polyadenylation sequence (poly(A)). The polyadenylation signal sequence can be selected from any of a variety of polyadenylation signal sequences known in the art. In some aspects, the construct (e.g., expression construct) further comprises a polyA sequence selected from the group consisting of a beta-globin polyA, a bovine growth hormone polyA (bGH polyA), a human growth hormone polyA (hGH polyA), and a SV40 immediate early poly A (SV40 polyA). In some aspects, the polyA sequence comprises a
bovine growth hormone polyA. In some aspects, the polyA sequence comprises a SV40 immediate early polyA. In some aspects, the polyA comprises b-globin polyA. In some aspects, the polyA comprises hGH polyA.
[0127] In some aspects, the constructs disclosed herein are packaged in a delivery vector, e.g., a viral vector, comprising the nucleic acid construct or an expression construct comprising a nucleic acid encoding a PA28A, PA28B, or a combination thereof. In some aspects, the delivery vector is suitable for delivery to a specific tissue. In some aspects, the tissue is selected from brain, eye, or heart muscle.
[0128] In some aspects, the viral vector comprises a construct comprising a polynucleotide comprising a promoter sequence and a nucleic acid sequence encoding Proteasome activator complex subunit 1 (PA28A). In some aspects, the construct comprises a hSyn promoter, a hRho promoter, or a pRhod promoter. In some aspects, the construct comprises a CBh promoter. In some aspects, the construct comprises a EFla promoter. In some aspects, the polynucleotide further comprises a WPRE. In some aspects, the polynucleotide further comprises an optimized WPRE (oPRE).
[0129] In some aspects, the nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a promoter sequence (e.g., hSyn or pRod), a nucleic acid sequence encoding Proteasome activator complex subunit 1 (PA28A), and optionally a WPRE. In some aspects, the nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a promoter sequence (e.g., hSyn or pRod), a nucleic acid sequence encoding Proteasome activator complex subunit 1 (PA28A), and optionally an optimized WPRE (oPRE). In some aspects, the nucleic acid construct or an expression construct (e.g., comprising an expression cassette) further comprises a nucleic acid sequence encoding Proteasome activator complex subunit 2 (PA28B). In some aspects, the polynucleotide comprises a promoter sequence, a nucleic acid encoding PA28A, a proteolytic cleavage site, and a nucleic acid encoding PA28B.
In some aspects, the polynucleotide comprises a promoter sequence, a nucleic acid encoding PA28A, a proteolytic cleavage site, and a nucleic acid encoding PA28B in 5'-3' orientation. In some aspects, wherein the polynucleotide comprises a promoter sequence, a nucleic acid encoding PA28B, a proteolytic cleavage site, and a nucleic acid encoding PA28A in 5'-3' orientation. In some aspects, the proteolytic cleavage site is P2A, furin, or intein-F2A. In some aspects, the promoter is hSyn, hRho, or pRhod. In some aspects, the
construct comprises a CBh promoter. In some aspects, the construct comprises a EFla promoter. In some aspects, the polynucleotide further comprises a WPRE. In some aspects, the polynucleotide further comprises an optimized WPRE (oPRE). In some aspects, the nucleic acid construct or an expression construct (e.g., comprising an expression cassette) further comprises an ITR (e.g., AAV2 ITRs).
[0130] In some aspects, the nucleic acid construct or an expression construct (e.g., comprising an expression cassette) disclosed herein is a gene therapy construct.
[0131] The exogenous (i.e., donor) DNA used in the disclosure can be obtained from suitable cells, and the constructs prepared using techniques well known in the art. Likewise, techniques for obtaining expression of exogenous DNA or RNA sequences in a genetically altered host cell are known in the art (see e.g., Kormal et al., Proc. Natl. Acad. Sci. USA, 84:2150-2154 (1987); Sambrook et al. Molecular Cloning: a Laboratory Manual, 2nd Ed., 1989, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.; each of which are hereby incorporated by reference with respect to methods and compositions for eukaryotic expression of a DNA of interest).
[0132] Sequences for the possible vector elements is provided in Table 4.
IV. Delivery Vectors
[0133] Certain aspects of the disclosure are directed to delivery vectors comprising the nucleic acid construct or an expression construct (e.g., comprising an expression cassette), or gene therapy constructs disclosed herein. In some aspects, the delivery vector is a viral vector.
[0134] In certain aspects, the delivery vector (e.g., a viral vector) comprises a nucleic acid encoding PA28A disclosed herein. In some aspects, the delivery vector (e.g., a viral vector) comprises a nucleic acid encoding PA28A in combination with PA28B disclosed herein.
[0135] In some aspects, the delivery vector comprises the nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a promoter which is tissue or cell-type specific promoter. In some aspects, the promoter is a bi directional promoter. In some aspects, the promoter is selected from a MecP2 promoter, a GUSB promoter, a CBh promoter, a PGK promoter, a human synapsin (Syn) promoter, a human rhodopsin (Rhod) kinase promoter, a cone arrestin promoter, a tyrosine hydroxylase promoter, a dopamine transporter promoter, a GRK1 promoter, a cardiac myosin light chain 2 promoter, a CAMKII promoter, a hDlx promoter, a CMV promoter, a CAG promoter, a bidirectional CAG promoter, a CBA promoter, an EFla promoter, or a human ubiquitin C promoter.
[0136] In some aspects, the delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a polyadenylation site, a woodchuck hepatitis virus post-transcriptional regulatory element (WPRE), an optimized woodchuck hepatitis virus post-transcriptional regulatory element (oPRE), an intron, a microRNA binding motif, or any combination thereof. In some
aspects, the delivery vector comprises an expression construct comprising a first expression cassette comprising a polynucleotide encoding PA28A and a second expression cassette comprising a polynucleotide encoding PA28B. See Kugler et al., Virology 311 (2003) 89-95. In some aspects, the first and second expression cassettes comprise the same promoter. In some aspects, the first and second expression cassettes comprise different promoters. In some aspects, the first expression cassette comprises a hSyn promoter and the second expression cassette comprises a CMV promoter. In some aspects, the first expression cassette comprises a CMV promoter and the second expression cassette comprises a hSyn promoter. In some aspects, the first and/or second expression cassettes comprise a polyA (e.g., bGH polyadenylation site, hGH polyadenylation site, or a SV40 polyadenylation site). In some aspects, the first and/or second expression cassette can further comprise a WPRE. In some aspects, the first and/or second expression cassette can further comprise an optimized WPRE (oPRE).
[0137] In some aspects, the delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a chimeric intron. In some aspects, the delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a beta-globin intron. In some aspects, the delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising minute virus of mice (MVM) intron.
[0138] In some aspects, the delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a polyA sequence selected from the group consisting of a beta-globin polyA, a bovine growth hormone polyA (bGH polyA), a human growth hormone polyA (hGH polyA), and a SV40 immediate early poly A (SV40 polyA). In some aspects, the polyA is a bovine growth hormone polyA.
[0139] In some aspects, the delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a 5’ and/or a 3’ untranslated region (UTR). In some aspects, the delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprises a microRNA binding motif in the 3’ untranslated region (UTR).
[0140] In some aspects, the delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising comprises a WPRE. In some aspects, the delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising comprises an optimized WPRE (oPRE).
[0141] In some aspects, the delivery vector comprises the nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a promoter sequence (e.g., hSyn or pRod), a nucleic acid sequence encoding Proteasome activator complex subunit 1 (PA28A), and optionally a WPRE. In some aspects, the delivery vector comprises the nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a promoter sequence (e.g., hSyn or pRod), a nucleic acid sequence encoding Proteasome activator complex subunit 1 (PA28A), and optionally an optimized WPRE (oPRE). In some aspects, the delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) further comprising a nucleic acid sequence encoding Proteasome activator complex subunit 2 (PA28B). In some aspects, the delivery vector comprises a polynucleotide comprising a promoter sequence, a nucleic acid encoding PA28A, a proteolytic cleavage site, and a nucleic acid encoding PA28B. In some aspects, delivery vector comprises a polynucleotide comprising a promoter sequence, a nucleic acid encoding PA28A, a proteolytic cleavage site, and a nucleic acid encoding PA28B in 5'-3' orientation. In some aspects, the delivery vector comprises a polynucleotide comprising a promoter sequence, a nucleic acid encoding PA28B, a proteolytic cleavage site, and a nucleic acid encoding PA28A in 5'-3' orientation.
[0142] In some aspects, the cleavage sequence a P2A, a furin, or an intein-F2A cleavage sequence. In some aspects, the proteolytic cleavage site is P2A. In some aspects, the promoter is hSyn or pRhod. In some aspects, the polynucleotide further comprises a WPRE. In some aspects, the polynucleotide further comprises an optimized WPRE (oPRE). In some aspects, the delivery vector is suitable for tissue specific delivery (e.g., to the brain, eye, or heart muscle.
[0143] In general, viral delivery vectors used in accordance with the disclosure can be composed of a viral particle derived from a naturally-occurring virus which has been genetically altered to render the virus replication-defective and to express a recombinant
gene of interest in accordance with the disclosure. Once the virus delivers its genetic material to a cell, it does not generate additional infectious virus but does introduce exogenous recombinant genes into the cell, preferably into the genome of the cell.
[0144] Numerous viral vectors are well known in the art, including, for example, retrovirus, adenovirus, adeno-associated virus (AAV), herpes simplex virus (HSV), cytomegalovirus (CMV), vaccinia and poliovirus vectors. In some aspects, the delivery vector (e.g., viral vector) is selected from the group consisting of an adeno-associated viral (AAV) vector, an adenoviral vector, a lentiviral vector, or a retroviral vector.
[0145] Where a replication-deficient virus is used as the viral vector, the production of infective virus particles containing either DNA or RNA corresponding to the DNA of interest can be produced by introducing the viral construct into a recombinant cell line which provides the missing components essential for viral replication. In some aspects, transformation of the recombinant cell line with the recombinant viral vector will not result in production of replication-competent viruses, e.g., by homologous recombination of the viral sequences of the recombinant cell line into the introduced viral vector. Methods for production of replication-deficient viral particles containing a nucleic acid of interest are well known in the art and are described in, e.g., Rosenfeld et al ., Science 252:431-434 (1991) and Rosenfeld etal. , Cell 68:143-155 (1992) (adenovirus); U.S. Patent No. 5,139,941 (adeno-associated virus); U.S. Patent No. 4,861,719 (retrovirus); and U.S. Patent No. 5,356,806 (vaccinia virus).
[0146] Expression of nucleic acid of the present disclosure after delivery to or integration in the genomic DNA of a target cell can require a specific promoter, including but not limited to, a promoter that is species specific, inducible, tissue-specific, or cell cycle- specific (Parr et al., Nat. Med.3: 1145-9 (1997); the contents of which are herein incorporated by reference in their entirety).
V. Adeno-Associated Virus (AAV)
[0147] Certain aspects of the disclosure provides isolated adeno-associated viruses (AAVs) comprising a payload, e.g., the nucleic acid construct or an expression construct (e.g., comprising an expression cassette), or gene therapy constructs disclosed herein. As used herein with respect to AAVs, the term “isolated” refers to an AAV that has been artificially produced or obtained. Isolated AAVs may be produced using recombinant methods. Such AAVs are referred to herein as “recombinant AAVs”. Recombinant AAVs (rAAVs)
preferably have tissue-specific targeting capabilities, such that a nuclease and/or transgene of the rAAV will be delivered specifically to one or more predetermined tissue(s). The AAV capsid is an important element in determining these tissue-specific targeting capabilities. Thus, a rAAV having a capsid appropriate for the tissue being targeted can be selected.
[0148] Methods for obtaining recombinant AAVs having a desired capsid protein are described, e.g., in US 2003/0138772, the contents of which are incorporated herein by reference in their entirety. Typically the methods involve culturing a host cell which contains a nucleic acid sequence encoding an AAV capsid protein; a functional rep gene; a recombinant AAV vector composed of AAV inverted terminal repeats (ITRs) and a transgene; and sufficient helper functions to permit packaging of the recombinant AAV vector into the AAV capsid proteins. In some aspects, capsid proteins are structural proteins encoded by the cap gene of an AAV. AAVs comprise three capsid proteins, virion proteins 1 to 3 (named VP1, VP2 and VP3), all of which are transcribed from a single cap gene via alternative splicing. In some aspects, the molecular weights of VP1, VP2 and VP3 are respectively about 87 kDa, about 72 kDa and about 62 kDa. In some aspects, upon translation, capsid proteins form a spherical 60-mer protein shell around the viral genome. In some aspects, the functions of the capsid proteins are to protect the viral genome, deliver the genome and interact with the host. In some aspects, capsid proteins deliver the viral genome to a host in a tissue specific manner.
[0149] In some aspects, an AAV capsid protein is of an AAV serotype selected from the group consisting of AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAVrh8, AAV9, AAV10, AAVrhlO, and AAV.PHP.B. In some aspects, the AAV capsid protein is a variant of AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAVrh8, AAV9, AAV10, AAVrhlO, and AAV.PHP.B. In some aspects, the variant comprises sequence modifications resulting from, but not limited to, capsid evolution, selection, shuffling, or directed mutagenesis. In some aspects, AAV vector can be derived from various serotypes, including combinations of serotypes (i.e., "pseudotyped" AAV) or from various genomes (e.g., single stranded or self-complementary). In some aspects, an AAV capsid protein is of a serotype derived from a non-human primate, for example AAVrh8 serotype. In some aspects, an AAV capsid protein is of a serotype derived for CNS transduction, for example AAV.PHP.B. In some aspects, the capsid protein is of AAV
serotype 9. In some aspects, the capsid protein is of AAV serotype 2. In some aspects, the capsid protein is of AAV serotype 8.
[0150] In some aspects, the components to be cultured in the host cell to package a rAAV vector in an AAV capsid can be provided to the host cell in trans. Alternatively, any one or more of the required components (e.g., recombinant AAV vector, rep sequences, cap sequences, and/or helper functions) may be provided by a stable host cell which has been engineered to contain one or more of the required components using methods known to those of skill in the art. Most suitably, such a stable host cell will contain the required component(s) under the control of an inducible promoter. However, the required component(s) may be under the control of a constitutive promoter. Examples of suitable inducible and constitutive promoters are provided herein, in the discussion of regulatory elements suitable for use with the transgene. In some aspects, a selected stable host cell may contain selected component(s) under the control of a constitutive promoter and other selected component s) under the control of one or more inducible promoters. For example, a stable host cell can be generated which is derived from 293 cells (which contain El helper functions under the control of a constitutive promoter), but which contain the rep and/or cap proteins under the control of inducible promoters. Still other stable host cells can be generated by one of skill in the art.
[0151] The recombinant AAV vector, rep sequences, cap sequences, and helper functions required for producing the rAAV of the disclosure can be delivered to the packaging host cell using any appropriate genetic element (vector). The selected genetic element can be delivered by any suitable method, including those described herein. The methods used to construct any aspect of this disclosure are known to those with skill in nucleic acid manipulation and include genetic engineering, recombinant engineering, and synthetic techniques. See, e.g., Sambrook et ah, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press, Cold Spring Harbor, N.Y. Similarly, methods of generating rAAV virions are well known and the selection of a suitable method is not a limitation on the present disclosure. See, e.g., K. Fisher et ak, J. Virol., 70:520-532 (1993) and U.S. Pat. No. 5,478,745.
[0152] In some aspects, recombinant AAVs may be produced using the triple transfection method (described in detail in U.S. Pat. No. 6,001,650). Typically, the recombinant AAVs are produced by transfecting a host cell with an AAV vector (comprising a transgene
flanked by ITR elements) to be packaged into AAV particles, an AAV helper function vector, and an accessory function vector. An AAV helper function vector encodes the “AAV helper function” sequences (e.g., rep and cap), which function in trans for productive AAV replication and encapsidation. Preferably, the AAV helper function vector supports efficient AAV vector production without generating any detectable wild-type AAV virions (e.g., AAV virions containing functional rep and cap genes). Non-limiting examples of vectors suitable for use with the present disclosure include pHLP19, described in U.S. Pat. No. 6,001,650 and pRep6cap6 vector, described in U.S. Pat. No. 6,156,303, the entirety of both incorporated by reference herein. The accessory function vector encodes nucleotide sequences for non-AAV derived viral and/or cellular functions upon which AAV is dependent for replication (e.g., “accessory functions”). The accessory functions include those functions required for AAV replication, including, without limitation, those moieties involved in activation of AAV gene transcription, stage specific AAV mRNA splicing, AAV DNA replication, synthesis of cap expression products, and AAV capsid assembly. Viral-based accessory functions can be derived from any of the known helper viruses such as adenovirus, herpesvirus (other than herpes simplex virus type-1), and vaccinia virus.
[0153] In certain aspects, the rAAV vectors of the present disclosure comprise a viral genome with at least one ITR region and a payload region, e.g., the nucleic acid construct or an expression construct (e.g., comprising an expression cassette), or gene therapy constructs comprising a polynucleotide encoding a proteasome regulatory subunit disclosed herein.
[0154] In some aspects, a composition comprising a rAAV delivery vector comprises a payload comprising a nucleic acid encoding PA28A, optionally in combination with PA28B disclosed herein.
[0155] In some aspects, the rAAV delivery vector comprises the nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a promoter which is tissue or cell-type specific promoter. In some aspects, the promoter is a bi directional promoter. In some aspects, the promoter is selected from a MecP2 promoter, a GUSB promoter, a CBh promoter, a PGK promoter, a human synapsin (Syn) promoter, a human rhodopsin (Rhod) kinase promoter, a cone arrestin promoter, a tyrosine hydroxylase promoter, a dopamine transporter promoter, a GRK1 promoter, a cardiac myosin light chain 2 promoter, a CAMKII promoter, a hDlx promoter, a CMV promoter,
a CAG promoter, a bidirectional CAG promoter, a CBA promoter, an EFla promoter, or a human ubiquitin C promoter.
[0156] In some aspects, the rAAV delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a polyadenylation site, a woodchuck hepatitis virus post-transcriptional regulatory element (WPRE), an optimized woodchuck hepatitis virus post-transcriptional regulatory element (oPRE), an intron, a microRNA binding motif, or any combination thereof. In some aspects, the rAAV delivery vector comprises a first expression cassette comprising a polynucleotide encoding PA28A and a second expression cassette comprising a polynucleotide encoding PA28B. See Kugler et ah, Virology 311 (2003) 89-95. In some aspects, the first and second expression cassettes comprise the same promoter. In some aspects, the first and second expression cassettes comprise different promoters. In some aspects, the first expression cassette comprises a hSyn promoter and the second expression cassette comprises a CMV promoter. In some aspects, the first expression cassette comprises a CMV promoter and the second expression cassette comprises a hSyn promoter. In some aspects, the first and/or second expression cassettes comprise a polyA (e.g., bGH polyadenylation site, hGH polyadenylation site, or a SV40 polyadenylation site). In some aspects, the first and/or second expression cassette can further comprise a WPRE. In some aspects, the first and/or second expression cassette can further comprise an optimized WPRE (oPRE).
[0157] In some aspects, the rAAV delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a chimeric intron. In some aspects, the rAAV delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a beta-globin intron. In some aspects, the rAAV delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising minute virus of mice (MVM) intron.
[0158] In some aspects, the rAAV delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a polyA sequence selected from the group consisting of a beta-globin polyA, a bovine growth hormone polyA (bGH polyA), a human growth hormone polyA (hGH polyA), and a
SV40 immediate early poly A (SV40 poly A). In some aspects, the polyA is a bovine growth hormone polyA.
[0159] In some aspects, the rAAV delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a 5’ and/or a 3’ untranslated region (UTR). In some aspects, the rAAV delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprises a microRNA binding motif in the 3’ untranslated region (UTR).
[0160] In some aspects, the rAAV delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising comprises a WPRE. In some aspects, the rAAV delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising comprises an optimized WPRE (oPRE).
[0161] In some aspects, the nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprises a promoter sequence (e.g., hSyn or pRod), a nucleic acid sequence encoding Proteasome activator complex subunit 1 (PA28A), and optionally a WPRE. In some aspects, the nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprises a promoter sequence (e.g., hSyn or pRod), a nucleic acid sequence encoding Proteasome activator complex subunit 1 (PA28A), and optionally an optimized WPRE (oPRE). In some aspects, the rAAV delivery vector comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) further comprises a nucleic acid sequence encoding Proteasome activator complex subunit 2 (PA28B). In some aspects, the polynucleotide comprises a promoter sequence, a nucleic acid encoding PA28A, a proteolytic cleavage site, and a nucleic acid encoding PA28B. In some aspects, the polynucleotide comprises a promoter sequence, a nucleic acid encoding PA28A, a proteolytic cleavage site, and a nucleic acid encoding PA28B in 5'-3' orientation. In some aspects, wherein the polynucleotide comprises a promoter sequence, a nucleic acid encoding PA28B, a proteolytic cleavage site, and a nucleic acid encoding PA28A in 5'-3' orientation. In some aspects, the cleavage sequence a P2A, a furin, or an intein-F2A cleavage sequence. In some aspects, the proteolytic cleavage site is P2A. In some aspects, the promoter is hSyn or pRhod. In some aspects, the polynucleotide further comprises a WPRE. In some aspects, the polynucleotide further comprises an optimized WPRE (oPRE). In some
aspects, the rAAV delivery vector is suitable for tissue specific delivery (e.g., to the brain, eye, or heart muscle).
[0162] In some aspects, the AAV vector comprises two ITRs. These two ITRs flank the payload region at the 5' and 3' ends. The ITRs function as origins of replication comprising recognition sites for replication. ITRs comprise sequence regions, which can be complementary and symmetrically arranged. ITRs incorporated into AAV vectors of the disclosure can be comprised of naturally occurring polynucleotide sequences or recombinantly derived polynucleotide sequences. In some aspects, the AAV vector is self-complementary. In some aspects, the AAV vector comprises two ITRs that facilitate the sc AAV.
[0163] The ITRs can be derived from the same serotype as the capsid, selected from any of the serotypes listed herein, or a derivative thereof. The ITR can be of a different serotype from the capsid. In some aspects, the AAV vector has more than one ITR. In a non-limiting example, the AAV vector has a viral genome comprising two ITRs. In some aspects, the ITRs are of the same serotype as one another. In some aspects, the ITRs are of different serotypes. Non-limiting examples include zero, one or both of the ITRs having the same serotype as the capsid. In some aspects both ITRs of the AAV vector are AAV2 ITRs.
[0164] In some aspects, the AAV vector of the present disclosure comprises a polyadenylation sequence. The AAV vectors of the present disclosure can comprise a polyadenylation sequence between the 3' end of the payload coding sequence and the 5' end of the 3' ITR.
[0165] In some aspects, the AAV vector of the present disclosure comprises an untranslated region (UTR). By definition, wild-type untranslated regions (UTRs) of a gene are transcribed but not translated. Generally, the 5' UTR starts at the transcription start site and ends at the start codon and the 3' UTR starts immediately following the stop codon and continues until the termination signal for transcription.
[0166] Wild-type 5' untranslated regions (UTRs) include features which play roles in translation initiation. Kozak sequences, which are commonly known to be involved in the process by which the ribosome initiates translation of many genes, are usually included in 5' UTRs. Kozak sequences have the consensus CCR(A/G)CCAUGG (SEQ ID NO: 11), where R is a purine (adenine or guanine) three bases upstream of the start codon (ATG),
which is followed by another 'G. In some aspects, the 5'UTR in a AAV vector of the present disclosure includes a Kozak sequence.
[0167] Wild-type 3' UTRs are known to have stretches of Adenosines and Uridines embedded therein. These AU rich signatures are particularly prevalent in genes with high rates of turnover. Based on their sequence features and functional properties, the AU rich elements (AREs) can be separated into three classes (Chen et al, 1995, the contents of which are herein incorporated by reference in its entirety). Most proteins binding to the AREs are known to destabilize the messenger, whereas members of the ELAV family, most notably HuR, have been documented to increase the stability of mRNA. HuR binds to AREs of all the three classes. Engineering the HuR specific binding sites into the 3' UTR of nucleic acid molecules will lead to HuR binding and thus, stabilization of the message in vivo.
[0168] Any UTR from any gene known in the art can be incorporated into an AAV vector of the present disclosure. These UTRs, or portions thereof, can be placed in the same orientation as in the gene from which they were selected or they can be altered in orientation or location. In some aspects, the UTR used in an AAV vector of the present disclosure can be inverted, shortened, lengthened, made with one or more other 5' UTRs or 3' UTRs known in the art. As used herein, the term "altered" as it relates to a UTR, means that the UTR has been changed in some way in relation to a reference sequence. For example, a 3' or 5' UTR can be altered relative to a wild-type or native UTR by the change in orientation or location as taught above or can be altered by the inclusion of additional nucleotides, deletion of nucleotides, swapping or transposition of nucleotides. In some aspects, an AAV vector of the present disclosure comprises at least one artificial UTRs, which is not a variant of a wild-type UTR. In some aspects, an AAV vector of the present disclosure comprises UTRs, which have been selected from a family of transcripts whose proteins share a common function, structure, feature or property.
[0169] In some aspects, the payload region of an AAV vector of the present disclosure comprises at least one element to enhance the expression.
[0170] In some aspects, the nucleic acids described herein further comprise a Woodchuck
Hepatitis Virus Posttranscriptional Regulatory Element (WPRE). In some aspects, the WPRE sequence can be any wild-type or mutated WPRE sequence. See, e.g., U.S. Patent
No. 10,179,918. In some aspects, the nucleic acids described herein further comprise an optimized Woodchuck Hepatitis Virus Posttranscriptional Regulatory Element (oPRE).
VI. Method for Producing AAV Particles
[0171] The present disclosure provides also methods for the generation of AAV particles, by viral genome replication in a viral replication cell comprising contacting the viral replication cell with an AAV polynucleotide or AAV genome (e.g., an AAV vector of the present disclosure).
[0172] In some aspects, an AAV particle is produced by a method comprising the steps of: (1) co-transfecting competent bacterial cells with a bacmid vector and either a viral construct vector and/or AAV payload construct vector, (2) isolating the resultant viral construct expression vector and AAV payload construct expression vector and separately transfecting viral replication cells, (3) isolating and purifying resultant payload and viral construct particles comprising viral construct expression vector or AAV payload construct expression vector, (4) co-infecting a viral replication cell with both the AAV payload and viral construct particles comprising viral construct expression vector or AAV payload construct expression vector, and (5) harvesting and purifying the viral particle comprising a parvoviral genome.
[0173] In one aspect, the present disclosure provides a method for producing an AAV particle comprising the steps of (1) simultaneously co-transfecting mammalian cells, such as, but not limited to HEK293 cells, with a payload region (e.g., polynucleotide encoding a therapeutic protein or therapeutic peptide of the disclosure), a construct expressing rep and cap genes and a helper construct, and (2) harvesting and purifying the AAV particle comprising a viral genome.
[0174] In some aspects, the AAV particles can be produced in a viral replication cell that comprises an insect cell. Growing conditions for insect cells in culture, and production of heterologous products in insect cells in culture are well-known in the art, see, e.g., U.S. Patent No. 6,204,059.
[0175] The viral replication cell can be selected from any biological organism, including prokaryotic (e.g., bacterial) cells, and eukaryotic cells, including, insect cells, yeast cells and mammalian cells. Viral replication cells can comprise mammalian cells such as A549, WEH1, 3T3, 10T1/2, BHK, MDCK, COS 1, COS 7, BSC 1, BSC 40, BMT 10, VERO. W138, HeLa, HEK293, Saos, C2C12, L cells, HT1080, HepG2 and primary
fibroblast, hepatocyte and myoblast cells derived from mammals. Viral replication cells comprise cells derived from mammalian species including, but not limited to, human, monkey, mouse, rat, rabbit, and hamster or cell type, including but not limited to fibroblast, hepatocyte, tumor cell, cell line transformed cell, etc.
[0176] Viral production disclosed herein describes processes and methods for producing
AAV particles that contact a target cell to deliver a payload, e.g. a recombinant viral construct, which comprises a polynucleotide sequence encoding a payload such as a therapeutic protein disclosed herein.
[0177] In some aspects, the AAV particles can be produced in a viral replication cell that comprises a mammalian cell. Viral replication cells commonly used for production of recombinant AAV particles include, but are not limited to 293 cells, COS cells, HeLa cells, and KB cells.
[0178] In some aspects, AAV particles are produced in mammalian cells wherein all three VP proteins are expressed at a stoichiometry approaching 1:1:10 (VP1 :VP2:VP3). The regulatory mechanisms that allow this controlled level of expression include the production of two mRNAs, one for VP1, and the other for VP2 and VP3, produced by differential splicing.
[0179] In some aspects, AAV particles are produced in mammalian cells using a triple transfection method wherein a payload construct, parvoviral Rep and parvoviral Cap and a helper construct are comprised within three different constructs. The triple transfection method of the three components of AAV particle production can be utilized to produce small lots of virus for assays including transduction efficiency, target tissue (tropism) evaluation, and stability.
[0180] In some aspects, the viral construct vector and the AAV payload construct vector can be each incorporated by a transposon donor/acceptor system into a bacmid, also known as a baculovirus plasmid, by standard molecular biology techniques known and performed by a person skilled in the art. Transfection of separate viral replication cell populations produces two baculoviruses, one that comprises the viral construct expression vector, and another that comprises the AAV payload construct expression vector. The two baculoviruses can be used to infect a single viral replication cell population for production of AAV particles.
[0181] Baculovirus expression vectors for producing viral particles in insect cells, including but not limited to Spodoptera frugiperda (Sf9) cells, provide high titers of viral particle product. Recombinant baculovirus encoding the viral construct expression vector and AAV payload construct expression vector initiates a productive infection of viral replicating cells. Infectious baculovirus particles released from the primary infection secondarily infect additional cells in the culture, exponentially infecting the entire cell culture population in a number of infection cycles that is a function of the initial multiplicity of infection, see, e.g., Urabe, M. e/a/., J Virol. 2006 Feb; 80 (4): 1874-85, the contents of which are herein incorporated by reference in their entirety.
[0182] Production of AAV particles with baculovirus in an insect cell system can address known baculovirus genetic and physical instability. Baculovirus-infected viral producing cells are harvested into aliquots that can be cryopreserved in liquid nitrogen; the aliquots retain viability and infectivity for infection of large-scale viral producing cell culture (Wasilko DJ et al., Protein Expr Purif. 2009 Jun; 65(2): 122-32).
[0183] In some aspects, stable viral replication cells permissive for baculovirus infection are engineered with at least one stable integrated copy of any of the elements necessary for AAV replication and viral particle production including, but not limited to, the entire AAV genome, Rep and Cap genes, Rep genes, Cap genes, each Rep protein as a separate transcription cassette, each VP protein as a separate transcription cassette, the AAP (assembly activation protein), or at least one of the baculovirus helper genes with native or non-native promoters.
[0184] In some aspects, AAV particle production can be modified to increase the scale of production. Transfection of replication cells in large-scale culture formats can be carried out according to any methods known in the art.
[0185] In some aspects, cell culture bioreactors can be used for large scale viral production. In some cases, bioreactors comprise stirred tank reactors.
[0186] Cells of the disclosure, particularly, but not limited to viral production cells, can be subjected to cell lysis according to any methods known in the art. Cell lysis can be carried out to obtain one or more agents (e.g. viral particles) present within any cells of the disclosure.
[0187] Cell lysis methods can be chemical or mechanical. Chemical cell lysis typically comprises contacting one or more cells with one or more lysis agent. Mechanical lysis
typically comprises subjecting one or more cells to one or more lysis condition and/or one or more lysis force. In some aspects, chemical lysis can be used to lyse cells. As used herein, the term "lysis agent" refers to any agent that can aid in the disruption of a cell. In some cases, lysis agents are introduced in solutions, termed lysis solutions or lysis buffers. As used herein, the term "lysis solution" refers to a solution (typically aqueous) comprising one or more lysis agent. In addition to lysis agents, lysis solutions can include one or more buffering agents, solubilizing agents, surfactants, preservatives, cryoprotectants, enzymes, enzyme inhibitors and/or chelators.
[0188] Concentrations of salts can be increased or decreased to obtain an effective concentration for rupture of cell membranes. Lysis agents comprising detergents can include ionic detergents or non-ionic detergents. Detergents can function to break apart or dissolve cell structures including, but not limited to cell membranes, cell walls, lipids, carbohydrates, lipoproteins and glycoproteins.
[0189] In some aspects, mechanical cell lysis is carried out. Mechanical cell lysis methods can include the use of one or more lysis condition and/or one or more lysis force. As used herein, the term "lysis condition" refers to a state or circumstance that promotes cellular disruption. Lysis conditions can comprise certain temperatures, pressures, osmotic purity, salinity and the like. In some aspects, lysis conditions comprise increased or decreased temperatures. In some aspects, lysis conditions comprise changes in temperature to promote cellular disruption. Cell lysis carried out according to such aspects can include freeze-thaw lysis.
[0190] As used herein, the term "lysis force" refers to a physical activity used to disrupt a cell. Lysis forces can include, but are not limited to mechanical forces, sonic forces, gravitational forces, optical forces, electrical forces and the like. Cell lysis carried out by mechanical force is referred to herein as "mechanical lysis." Mechanical forces that can be used according to mechanical lysis can include high shear fluid forces.
[0191] In some aspects, a method for harvesting AAV particles without lysis can be used for efficient and scalable AAV particle production. In a non-limiting example, AAV particles can be produced by culturing an AAV particle lacking a heparin binding site, thereby allowing the AAV particle to pass into the supernatant, in a cell culture, collecting supernatant from the culture; and isolating the AAV particle from the supernatant, as described in US Patent Application 20090275107.
[0192] Cell lysates comprising viral particles can be subjected to clarification.
Clarification refers to initial steps taken in purification of viral particles from cell lysates. Clarification serves to prepare lysates for further purification by removing larger, insoluble debris. Clarification steps can include, but are not limited to centrifugation and filtration.
[0193] In some aspects, AAV particles can be purified from clarified cell lysates by one or more methods of chromatography. Chromatography refers to any number of methods known in the art for separating out one or more elements from a mixture. Such methods can include, but are not limited to ion exchange chromatography (e.g. cation exchange chromatography and anion exchange chromatography), immunoaffmity chromatography and size-exclusion chromatography.
[0194] Certain aspects of the disclosure are directed to viral particles (e.g., AAV particles) comprising a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a polynucleotide encoding PA28A, optionally in combination with a polynucleotide encoding PA28B disclosed herein. In some aspects, the viral particles comprise a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a polynucleotide comprising a promoter sequence (e.g., hSyn or pRod), a nucleic acid sequence encoding Proteasome activator complex subunit 1 (PA28A), and optionally a WPRE. In some aspects, the viral particles comprise a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a polynucleotide comprising a promoter sequence (e.g., hSyn or pRod), a nucleic acid sequence encoding Proteasome activator complex subunit 1 (PA28A), and optionally an optimized WPRE (oPRE). In some aspects, the viral particles (e.g., AAV particles) comprise a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) disclosed herein further comprising a nucleic acid sequence encoding Proteasome activator complex subunit 2 (PA28B). In some aspects, the viral particles (e.g., AAV particles) comprise a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a polynucleotide comprising a promoter sequence, a nucleic acid encoding PA28A, a proteolytic cleavage site, and a nucleic acid encoding PA28B. In some aspects, the viral particles (e.g., AAV particles) comprise a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a polynucleotide
comprising a promoter sequence, a nucleic acid encoding PA28A, a proteolytic cleavage site, and a nucleic acid encoding PA28B in 5'-3' orientation. In some aspects, the viral particles (e.g., AAV particles) comprise a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a polynucleotide comprising a promoter sequence, a nucleic acid encoding PA28B, a proteolytic cleavage site, and a nucleic acid encoding PA28A in 5'-3' orientation.
[0195] In some aspects, the viral particles (e.g., AAV particles) comprise the nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a promoter which is tissue or cell-type specific promoter. In some aspects, the promoter is a bi-directional promoter. In some aspects, the promoter is selected from a MecP2 promoter, a GUSB promoter, a CBh promoter, a PGK promoter, a human synapsin (Syn) promoter, a human rhodopsin (Rhod) kinase promoter, a cone arrestin promoter, a tyrosine hydroxylase promoter, a dopamine transporter promoter, a GRK1 promoter, a cardiac myosin light chain 2 promoter, a CAMKII promoter, a hDlx promoter, a CMV promoter, a CAG promoter, a bidirectional CAG promoter, a CBA promoter, an EFla promoter, or a human ubiquitin C promoter.
[0196] In some aspects, the viral particles (e.g., AAV particles) comprise comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a polyadenylation site, a woodchuck hepatitis virus post-transcriptional regulatory element (WPRE), an optimized woodchuck hepatitis virus post-transcriptional regulatory element (oPRE), an intron, a microRNA binding motif, or any combination thereof. In some aspects, the viral particles (e.g., AAV particles) comprise a first expression cassette comprising a polynucleotide encoding PA28A and a second expression cassette comprising a polynucleotide encoding PA28B. See Kugler et ah, Virology 311 (2003) 89-95. In some aspects, the first and second expression cassettes comprise the same promoter. In some aspects, the first and second expression cassettes comprise different promoters. In some aspects, the first expression cassette comprises a hSyn promoter and the second expression cassette comprises a CMV promoter. In some aspects, the first expression cassette comprises a CMV promoter and the second expression cassette comprises a hSyn promoter. In some aspects, the first and/or second expression cassettes comprise a polyA (e.g., bGH polyadenylation site, hGH polyadenylation site, or a SV40 polyadenylation site). In some aspects, the first and/or
second expression cassette can further comprise a WPRE. In some aspects, the first and/or second expression cassette can further comprise an optimized WPRE (oPRE).
[0197] In some aspects, the viral particles (e.g., AAV particles) comprise comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a chimeric intron. In some aspects, the viral particles (e.g., AAV particles) comprise comprises a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a beta-globin intron. In some aspects, the viral particles (e.g., AAV particles) comprise a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising minute virus of mice (MVM) intron.
[0198] In some aspects, the viral particles (e.g., AAV particles) comprise a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a polyA sequence selected from the group consisting of a beta-globin poly A, a bovine growth hormone polyA (bGH polyA), a human growth hormone polyA (hGH polyA), and a SV40 immediate early poly A (SV40 polyA). In some aspects, the polyA is a bovine growth hormone polyA.
[0199] In some aspects, the viral particles (e.g., AAV particles) comprise a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a 5’ and/or a 3’ untranslated region (UTR). In some aspects, the viral particles (e.g.,
AAV particles) comprise a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprises a microRNA binding motif in the 3’ untranslated region (UTR).
[0200] In some aspects, the viral particles (e.g., AAV particles) comprise a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising comprises a WPRE. In some aspects, the viral particles (e.g., AAV particles) comprise a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising comprises an optimized WPRE (oPRE).
[0201] In some aspects, the cleavage sequence a P2A, a furin, or an intein-F2A cleavage sequence. In some aspects, the proteolytic cleavage site is P2A. In some aspects, the promoter is hSyn or pRhod. In some aspects, the polynucleotide further comprises a WPRE. In some aspects, the polynucleotide further comprises an optimized WPRE
(oPRE). In some aspects, the viral particles (e.g., AAV particles) are suitable for tissue specific delivery (e.g., to the brain, eye, or heart muscle).
VII. Pharmaceutical Compositions
[0202] Certain aspects of the disclosure are directed to a pharmaceutical composition comprising a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy constructs, or a viral vector (e.g., AAV vector or AAV particle) disclosed herein and a carrier (e.g., pharmaceutically acceptable carrier). In some aspects, a construct or vector of the disclosure is formulated for delivery to a cell or tissues. In some aspects, the construct or vector of the disclosure is formulated for delivery to a subject.
[0203] In some aspects, pharmaceutically acceptable carriers can be readily selected by one of skill in the art in view of the indication for which the viral vector (e.g., AAV vector or AAV particle) is directed. For example, one suitable carrier includes saline, which may be formulated with a variety of buffering solutions (e.g., phosphate buffered saline). Other exemplary carriers include sterile saline, lactose, sucrose, calcium phosphate, gelatin, dextran, agar, pectin, peanut oil, sesame oil, and water. The selection of the carrier is not a limitation of the present disclosure.
[0204] Optionally, the compositions of the disclosure may contain, in addition to the viral vector (e.g., AAV vector or AAV particle) and carrier(s), other conventional pharmaceutical ingredients, such as preservatives, or chemical stabilizers. Suitable exemplary preservatives include chlorobutanol, potassium sorbate, sorbic acid, sulfur dioxide, propyl gallate, the parabens, ethyl vanillin, glycerin, phenol, parachlorophenol, and poloxamers (non-ionic surfactants) such as Pluronic® F-68. Suitable chemical stabilizers include gelatin and albumin.
VIII. Methods of Use and Treatment
[0205] Certain aspects of the disclosure are directed to methods of using the nucleic acid constructs, expression constructs (e.g., comprising an expression cassette), gene therapy constructs, viral vectors (e.g., AAV vector or AAV particle), or pharmaceutical compositions disclosed herein. In some aspects,
[0206] In some aspects, the present disclosure provides methods for increasing proteasome activity in a cell and/or tissue comprising administering to the cell or tissue a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a
gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical composition disclosed herein.
[0207] In some aspects, the present disclosure provides methods for treating a disease or condition associated with proteinopathy in a subject in need thereof comprising administering to the subject a composition comprising a gene therapy construct or vector disclosed herein. In some aspects, the disease or condition is a neurodegenerative disease (e.g., Parkinson’s Disease or Huntington’s Disease), an ocular disease (e.g., retinitis pigmentosa or Kjer’s optic neuropathy), or a cardiac disease. In some aspect, the administration is tissue specific. In some aspects, the gene therapy construct or vector disclosed herein is administered to the brain, eye, or heart muscle.
[0208] In some aspects, the methods include administering a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical composition comprising a polynucleotide encoding PA28A, optionally in combination with a polynucleotide encoding PA28B disclosed herein. In some aspects, the method comprises administering a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical composition comprising a polynucleotide comprising a promoter sequence (e.g., hSyn or pRod), a nucleic acid sequence encoding Proteasome activator complex subunit 1 (PA28A), and optionally a WPRE. In some aspects, the method comprises administering a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical composition comprising a polynucleotide comprising a promoter sequence (e.g., hSyn or pRod), a nucleic acid sequence encoding Proteasome activator complex subunit 1 (PA28A), and optionally an optimized WPRE (oPRE). In some aspects, the method comprises administering a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical composition comprising further comprising a nucleic acid sequence encoding Proteasome activator complex subunit 2 (PA28B). In some aspects, the method comprises a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical
composition comprising a polynucleotide comprising a promoter sequence, a nucleic acid encoding PA28A, a proteolytic cleavage site, and a nucleic acid encoding PA28B. In some aspects, the method comprises administering a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical composition comprising a polynucleotide comprising a promoter sequence, a nucleic acid encoding PA28A, a proteolytic cleavage site, and a nucleic acid encoding PA28B in 5'-3' orientation. In some aspects, the method comprises administering a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical composition comprising a polynucleotide comprising a promoter sequence, a nucleic acid encoding PA28B, a proteolytic cleavage site, and a nucleic acid encoding PA28A in 5'-3' orientation.
[0209] In some aspects, the methods include administering a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical composition comprising a promoter which is tissue or cell-type specific promoter. In some aspects, the promoter is a bi-directional promoter. In some aspects, the promoter is selected from a MecP2 promoter, a GUSB promoter, a CBh promoter, a PGK promoter, a human synapsin (Syn) promoter, a human rhodopsin (Rhod) kinase promoter, a cone arrestin promoter, a tyrosine hydroxylase promoter, a dopamine transporter promoter, a GRK1 promoter, a cardiac myosin light chain 2 promoter, a CAMKII promoter, a hDlx promoter, a CMV promoter, a CAG promoter, a bidirectional CAG promoter, a CBA promoter, an EFla promoter, or a human ubiquitin C promoter.
[0210] In some aspects, the methods include administering a nucleic acid construct, an expression construct (e.g., comprising an expression cassette, or comprising a first expression cassette and a second expression cassette described herein), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical composition comprising a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a polyadenylation site, a woodchuck hepatitis virus post-transcriptional regulatory element (WPRE), an optimized woodchuck hepatitis virus post-transcriptional regulatory element (oPRE), an intron, a microRNA binding motif, or any combination thereof.
[0211] In some aspects, the methods include administering a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical composition comprising a chimeric intron. In some aspects, the methods include administering a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical composition comprising a nucleic acid construct or an expression construct (e.g., comprising an expression cassette) comprising a beta-globin intron. In some aspects, the methods include administering a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical composition comprising minute virus of mice (MVM) intron.
[0212] In some aspects, the methods include administering a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical composition comprising a polyA sequence selected from the group consisting of a beta-globin poly A, a bovine growth hormone polyA (bGH polyA), a human growth hormone polyA (hGH polyA), and a SV40 immediate early poly A (SV40 polyA). In some aspects, the polyA is a bovine growth hormone polyA.
[0213] In some aspects, the methods include administering a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical composition comprising a 5’ and/or a 3’ untranslated region (UTR). In some aspects, the methods include administering a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical composition comprising a microRNA binding motif in the 3’ untranslated region (UTR).
[0214] In some aspects, the methods include administering a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical composition comprising a WPRE. In some aspects, the methods include administering a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene
therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical composition comprising an optimized WPRE (oPRE).
[0215] In some aspects, the cleavage sequence a P2A, a furin, or an intein-F2A cleavage sequence. In some aspects, the proteolytic cleavage site is P2A. In some aspects, the promoter is hSyn or pRhod. In some aspects, the polynucleotide further comprises a WPRE. In some aspects, the polynucleotide further comprises an optimized WPRE (oPRE). In some aspects, method is suitable for tissue specific delivery (e.g., to the brain, eye, or heart muscle).
[0216] Based on the methods disclosed herein, the gene therapy composition comprising an AAV vector, an AAV vector, or an AAV particle of the present disclosure for use in therapy, or for use as a medicament, or for use in treating a disease or disorder associated with misfolded or damaged proteins (e.g., neurological, optical or a cardiac diseases or disorders associated with proteinopathy) in a subject in need thereof is contemplated.
[0217] In some aspects, the diseases or disorders associated with proteinopathy is selected from the group consisting of inherited retinal disease (retinitis pigmentosa, Leber congenital amaurosis, Kjer’s optic neuropathy), neurodegeneration (genetic or idiopathic forms of Parkinson’s Disease, Alzheimer’s Disease, Huntington’s Disease, prion disease, multiple system atrophy, multiple sclerosis, amyotrophic lateral sclerosis, Friedreich ataxia), and cardiovascular diseases (desmin-related cardiomyopathy, ischemia/reperfusion injury).
[0218] In some aspects, the diseases or disorder associated with proteinopathy is selected from the group consisting of Alzheimer’s disease, Dutch hereditary cerebral hemorrhage with amyloidosis, Congophilic angiopathy, Tauopathies, Corticobasal degeneration, Pick’s disease, Progressive supranuclear palsy, Prion diseases, Creutzfeld-Jacob disease, Gerstmann-Straussler-Schneiker syndrome, fatal familia insomnia, Kuru, Bovine spongiform encephalopathy, Scrapie, Chronic wasting disease, Synucleinopathies, Parkinson’s disease, Lewy body variant of Alzheimer’s disease, Diffuse Lewy body disease, Dementia with Lewy bodies, Multiple system atrophy, Neurodegeneration with brain iron accumulation type I, Amyotrophic lateral sclerosis and frontotemporal lobar degeneration, Amyotrophic lateral sclerosis, Huntington’s disease, Hereditary dentatorubral-pallidoluysian atrophy, Kennedy’s disease or X-linked spinal and bulbar muscular atrophy, Spinocerebellar ataxia 1, Neuronal intranuclear inclusion disease,
Spinocerebellar ataxia 2, Spinocerebellar ataxia 3, Spinocerebellar ataxia 6, Spinocerebellar ataxia 7, Spinocerebellar ataxia 17, Alexander’s disease, Cockayne syndrome, or Spinal muscular atrophy.
[0219] In some aspects, the disease or disorder is selected from the group consisting of a neurodegenerative disease (e.g., Parkinson’s Disease or Huntington’s Disease), an ocular disease (e.g., retinitis pigmentosa or Kjer’s optic neuropathy), or a cardiac disease.
[0220] In some aspect, the administration is tissue specific. In some aspects, the gene therapy construct or vector disclosed herein is administered to the brain, eye, or heart muscle.
IX. Administration
[0221] The nucleic acid constructs, expression constructs (e.g., comprising an expression cassette), gene therapy constructs, viral vectors (e.g., AAV vector or rAAV particle), and pharmaceutical compositions disclosed herein can be administered by any route which results in a therapeutically effective outcome, e.g., for therapeutic expression of a PSME (e.g., PA28A alone or PA28A in combination with PA28B) disclosed herein.
[0222] In some aspects, the delivery can be intramuscular (IM), intravenous (IV), intraocular (e.g., sub-retinal), intracranial (e.g., striatum, ventricular), or intrathecal (e.g., cistema magna, lumbar). In some aspects, the disclosure is directed to a method of delivering a gene therapy to brain tissue. In some aspects, the disclosure is directed to delivering a gene therapy construct to eye tissue. In some aspects, the disclosure is directed to delivering a gene therapy construct to heart tissue.
[0223] The delivery vectors disclosed herein (e.g., AAV vectors or AAV particles) can be administered in any suitable form, either as a liquid solution or suspension, as a solid form suitable for liquid solution or suspension in a liquid solution.
X. Kits
[0224] The present disclosure also provides kits, or products of manufacture, comprising
(i) a nucleic acid construct, an expression construct (e.g., comprising one or more expression cassettes), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical compositions of the present disclosure, and (ii) optionally instructions for use (e.g., a package insert with instructions to perform any of the methods described herein).
[0225] In some aspects, the kit or product of manufacture comprises (i) a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical compositions of the present disclosure, (ii) optionally, an additional therapeutic agent, and (iii) optionally, instructions for use (e.g., a package insert with instructions to perform any of the methods described herein are also contemplated).
[0226] In some aspects, the components of a kit or product of manufacture disclosed herein are in one or more containers. In some aspects, the kit or product of manufacture comprises (i) a nucleic acid construct, an expression construct (e.g., comprising an expression cassette), a gene therapy construct, a viral vector (e.g., AAV vector or AAV particle), or pharmaceutical compositions of the present disclosure, and (ii) a brochure with instructions to prepare the product for administration.
[0227] One skilled in the art will readily recognize that vectors, polynucleotides, and pharmaceutical compositions of the present disclosure, or combinations thereof, can be readily incorporated into one of the established kit formats which are well known in the art.
[0228] The practice of the present disclosure will employ, unless otherwise indicated, conventional techniques of cell biology, cell culture, molecular biology, transgenic biology, microbiology, recombinant DNA, and immunology, which are within the skill of the art. Such techniques are explained fully in the literature.
[0229] All of the references cited above, as well as all references cited herein, are incorporated herein by reference in their entireties.
[0230] The following examples are offered by way of illustration and not by way of limitation.
EXAMPLES
Example 1 - PA28 Subunit Constructs
[0231] Viral vector constructs for expressing PA28A and PA28B subunits are prepared. The constructs include: (1) pAAV9- hSyn-PA28alpha(mouse)-WPRE; (2) pAAV2- pRhod-PA28alpha(mouse)-WPRE; (3) pAAV9- hSyn-PA28beta-P2A- PA28alpha(mouse)-WPRE; and (4) pAAV2-pRhod- PA28beta-P2A-PA28alpha(mouse)-
WPRE. Constructs are prepared by synthesizing genes de novo followed by molecular biology techniques to place into AAV or lentiviral backbones.
Example 2 - In vitro Expression and Proteasome Activity of PA28alpha and
PA28beta constructs
[0232] Bi-directional CMV promoter expression plasmids were generated to test the ability of PA28alpha-only (FIG. 1A) or PA28 alpha/beta 11 S caps (FIG. IB) to enhance proteasomal activity.
[0233] To test proteasome activity, HEK-293T cells (PSME1-KO) cells were transfected with plasmids comprising expression of PA28alpha-only (PSMEl-ZsGreenl in pBi-CMV) (FIG. 1A), bicistronic expression of PA28alpha and PA28beta (PSME1-PSME2 in pBi- CMV) (FIG. IB), or an empty vector control (ZsGreen (Neg Ctrl) in pBi-CMV) (FIG. 1C). 48 hours post-transfection, the cells were harvested and the lysates were used in a proteasomal activity assay using Suc-LLVY-AMC as an artificial substrate. Proteasome activity cleaves AMC from the artificial peptide substrate (Suc-LLVY-AMC) and generates free AMC which is fluorescent. Proteasomal activity was kinetically monitored by measuring the increase in fluorescence of the free AMC (FIG. 2A). Epoxomicin, a 20S proteasome inhibitor, was used to determine background non-proteasomal cleavage of the substrate and thereby calculate specific proteasome activity. Formation of 11 S caps with both PA28alpha and PA28beta has stronger proteasomal activation property than 11 S cap formed from PA28alpha alone. (FIGs. 2B-2E). Overall activity increased with more lysate used.
[0234] Western blotting shows PA28alpha and PA28beta expression in wild-type or transfected PA28alpha knockout (PSMEl-KO) HEK-293T cell lysates (FIG. 3A-3E). The antibodies were selective for their respective antigens. Endogenous PA28beta was increased when PA28alpha was overexpressed.
[0235] Plasmids were generated to test bicistronic expression of PA28alpha and PA28beta to achieve formation of active 11 S proteasomal caps. PSMEl-KO HEK-293T cells were transfected with either an empty vector plasmid (ZsGreen in pBi-CMV), a PA28alpha and PA28beta bicistronic plasmid (PA28a and RA28b in pBi-CMV (bicistronic)) (FIG. 4C), a 1 : 1 ratio of a PA28alpha and PA28beta plasmid (FIG. 4D), a PA28alpha only plasmid, a PA28beta only plasmid, a PA28alpha and PA28beta IRES bicistronic plasmid (PA28a- IRES-PA28P (FIGs. 4G and 4H), a PA28alpha and PA28beta P2A bicistronic plasmid
(RA28a-R2A-RA28b) (FIG. 4E and 4F), or a PA28alpha and PA28beta flexible linker bicistronic plasmid (PA28a-Flexible Linker-PA28P) (FIG. 41 and 4J) and protein expression was detected by western blotting (FIG. 5A-5C).
[0236] Lysates from the transfected PSME1-KO HEK-293T cells were used to determine the proteasomal activity of the PA28alpha and PA28beta bicistronic plasmid constructs (FIGs. 6A-6B). Only the PA28alpha-PA28beta bicistronic construct, 1 : 1 mix of PA28alpha only & PA28beta only, and PA28alpha only samples showed appreciable proteasomal activity.
[0237] Plasmids were generated to test tandem bicistronic expression of PA28alpha and PA28beta to achieve formation of active 11 S proteasomal caps (FIGs. 7A-7C, 7E-7G). PSME1-KO HEK-293T cells were transfected with either a wild-type pBI-CMV vector (ZsGreen in pBi-CMV) (FIG. 1C), a PA28alpha and PA28beta bicistronic plasmid (PA28a and RA28b in pBi-CMV (bicistronic) (FIG. IB), a PA28alpha only plasmid, a PA28beta only plasmid, a 1 : 1 ratio of a PA28alpha and PA28beta plasmid (FIG. 4D), a PA28alpha and PA28beta tandem plasmid under the transcriptional control of a EF la promoter (EFla- P A28a-EF 1 a-P A28b (tandem)) (FIG. 7G), a PA28alpha and PA28beta tandem plasmid under the transcriptional control of a hSynapsin promoter (1^h-RA28a-1^h-RA28b (tandem)) (FIG. 7E), a PA28alpha and PA28beta tandem plasmid under the transcriptional control of a hSynapsin promoter with an optimized WPRE (oPRE) sequence (hSyn-PA28a- oRKE-1^h-RA28b-oRKE (tandem)) (FIG. 7F), or a PA28alpha and PA28beta tandem plasmid under the transcriptional control of a CBh promoter (OB1i-RA28a-(¾1i-RA28b (tandem)) (FIG. 7B) and protein expression was detected by western blotting (FIGs. 8A- 8C).
[0238] Lysates from the transfected PSME1-KO HEK-293T cells were used to determine the proteasomal activity of the PA28alpha and PA28beta constructs (FIGS. 9A-9B). Only The PA28alpha and PA28beta in pBi-CMV, 1 : 1 mix of PA28alpha only & PA28beta only, PA28alpha only, and EF1 alpha tandem samples showed appreciable proteasomal activity.
Example 3 -In vivo Expression and Localization of PA28alpha and PA28beta AAV constructs
[0239] To test in vivo expression of PA28alpha and PA28beta subunits, mice were administered lpl of self-complementary AAV8 (scAAV8) viral mixture via sub-retinal injection to the left eye. The scAAV8 viral mixture comprising a 4:4: 1 (by volume) mixture
of scAAV8-hRho-mPMSEl (PA28alpha) (titer: 5.38xl013 gc/mL), scAAV8-hRho- mPSME2 (PA28beta) (titer: 3.99xl013 gc/mL), and scAAV8-hRho-eGFP (titer: 4.49xl013 gc/mL). The eyes were harvested 18 days post-injection and protein expression was analyzed by western blot. Lysate from the uninjected right eye was used as a control. PA28alpha, PA28beta, and GFP were all detected in the injected left eyes (FIG. 10A).
[0240] To test in vivo tandem expression of PA28alpha and PA28beta subunits, mice were administered lpl of the scAAV8 vectors, AAV8-hRHO-oPRE tandem mPA28alpha/beta (FIG. 7D) or AAV8-CBh tandem mA28alpha/beta (FIG. 7B) via sub-retinal injection to the left eye. Eyes were harvested 21 days post-injection and protein expression was analyzed by western blot. Lysate from the uninjected right eye was used as a control. Western blotting shows successful expression of both PA28alpha and PA28beta in the injected left eyes (FIG. 10B).
[0241] In vivo localization of PA28alpha was examined by immunohistochemistry. Mice were administered lpl of a mixture of scAAV8 vectors via sub-retinal injection to the left eye. The scAAV8 mixture comprised hRho-mPSMEl(PA28alpha), scAAV8-hRho- mPSME2(PA28beta), scAAV8-hRho-eGFP in a 16:16:1 ratio. Expression of PA28alpha was detected in the photoreceptor layer of the injected eye (FIG. 11 A).
[0242] In vivo localization of PA28alpha in retinal tissue of AAV8 expressing both PA28oc and RA28b in the tandem configuration under control of human rhodopsin promoter (hRho) with oPRE elements after the transgenes was examined. Mice were administered lpl of AAV8 expressing both PA28alpha and PA28beta in the tandem configuration under control of human rhodopsin promoter (hRho) with oPRE elements after the transgenes (FIG. 7D) via sub-retinal injection to the left eye. Expression of PA28alpha was detected in the photoreceptor layer of the injected eye (FIG. 1 IB).
[0243] A second set of mice were administered 1 pL of an AAV8 expressing both PA28oc and RA28b in the tandem configuration under control of CBh promoter (FIG. 7B). Stronger expression of PA28alpha was detected in the retina pigment epithelium rather than the photoreceptors (except in the extreme left part of the retina) (FIG. 11C).
[0244] In vivo localization of PA28alpha in the brain tissue of mice stereotactically inj ected on one side with a 1:1 mixture of scAAV9-hSyn-mPA28oc and 8ϋAAn9-1^h-ihRA28b targeting the dopaminergic substantia nigra neurons was examined. Coronal brain sections containing the target injection site (substantia nigra) were removed from fixed mice and
stained for PA28oc and tyrosine hydroxylase (TH, marker of dopaminergic neurons). Expression of PA28alpha was detected in the dopaminergic substantia nigra neurons (FIG. 12A).
[0245] In vivo localization of PA28alpha in the brain tissue of mice stereotactically inj ected on one side with scAAV9-hSyn-mPA28oc (right side) and on the other side with scAAV9- hSyn-mPA28 (left side) targeting the dopaminergic substantia nigra neurons was examined. Coronal brain sections containing the target injection site (substantia nigra) were removed from fixed mice and stained for PA28oc and tyrosine hydroxylase (TH, marker of dopaminergic neurons). Expression of PA28alpha (FIG. 12B) and PA28beta (FIG. 12C) was detected in the dopaminergic substantia nigra neurons.
[0246] In vivo localization of PA28alpha and PA28beta in the brain tissue of mice administered AAV9 expressing both PA28oc and RA28b in the tandem configuration under control of CBh promoter (FIG. 7B) (Left side) or EFloc promoter (FIG. 7G) (Right side) after the transgenes. Mice were administered the AAV9 vectors via stereotactic injection of the substantia nigra. Coronal brain sections containing the target injection site (substantia nigra) were removed from fixed mice and stained for PA28oc and tyrosine hydroxylase (TH, marker of dopaminergic neurons) (FIG. 12D) or RA28b and tyrosine hydroxylase (FIG. 12E). Expression of both PA28alpha and PA28beta was detected.
[0247] A table summarizing the results of the bicistronic AAV vector designs is provided in Table 6.
Example 4 - Mouse Model of Retinitis Pigmentosa [0248] To assess the effects of the constructs of the disclosure on proteasome activity and treatment of Retinitis Pigmentosa in vivo , constructs are administered by sub-retinal injection in a mouse model of Retinitis Pigmentosa, RhoP23H mouse model (Jackson Labs (stock #017628)) of Retinitis Pigmentosa (see Lobanova et ak, Nat Commun. 2018 Apr 30;9(1): 1738).
[0249] Histology for the number of photoreceptors is assessed as well as behavioral visual assays to determine if visual acuity is preserved ( See e.g., Visual function testing: a quantifiable visually guided behavior in mice, Vision Research , 48:3, pages 346-352, 2008).
Example 5 - Mouse Model of Huntington’s Disease [0250] To assess the effects of the constructs of the disclosure on proteasome activity and treatment of Huntington’s disease in vivo , constructs of the disclosure are administered to the striatum along with co-infusion of AAV that overexpresses the mutant form of Huntington (66Q, mHTT) {Cell Reports, 24, 1397-1406, 2018) in mice.
[0251] Tissues are harvested and analyzed by histology for the presence of aggregated huntingtin protein, loss or damage to neurons, and presence of PA28 subunits Tissues may also be measured for proteasomal activity.
Example 6 - Mouse Model of Parkinson’s Disease [0252] To assess the effects of the constructs of the disclosure on proteasome activity and treatment of Parkinson’s Disease in vivo , constructs of the disclosure are administered
along with co-infusion of an AAV that overexpresses the mutant form of alpha-synuclein (A53T) in the substantia nigra of mice {Molecular Neurodegeneration 2013, 8:44).
[0253] Tissues are harvested and analyzed by histology for the presence of dopaminergic neurons, aggregated alpha-synuclein, and presence of PA28 subunits. Tissues are also be measured for proteasomal activity.
Example 7 - Mouse Model Cariac Proteinopathy and ischemia/reperfusion injury [0254] To assess the effects of the constructs of the disclosure on proteasome activity and treatment of cardiac dysfunction and/or myocardial I/R injury, constructs of the disclosure are administered to mice harboring the ab-crystallin R120G mutation (CryAB(R120G), which is a model of desmin-related cardiomyopathy. A mouse model of myocardial I/R injury are tested where a vessel blockage is formed and later released to model I/R injury. [0255] Mice are evaluated for increases in LV posterior and anterior wall thickness at both end of diastole and systole as well as in calculated LV mass, ejection fraction, and fraction shortening. Histology of cardiac tissue are performed to characterize CryAB aggregates.
Claims
1. A gene therapy construct comprising a polynucleotide comprising a promoter operably linked to a nucleic acid sequence encoding Proteasome activator complex subunit 1 (PA28A).
2. The gene therapy construct of claims 1, wherein the promoter is a heterologous promoter.
3. The gene therapy construct of claim 1 or 2, wherein the promoter is a tissue specific promoter.
4. The gene therapy construct of any of the previous claims wherein the promoter is selected from the group consisting of a MecP2 promoter, a GUSB promoter, a CBh promoter, a PGK promoter, a human rhodopsin kinase promoter, a cone arrestin promoter, a tyrosine hydroxylase promoter, a dopamine transporter promoter, a GRK1 promoter, a cardiac myosin light chain 2 promoter, a CAMKII promoter, a hDlx promoter, a CMV promoter, a CAG promoter, a bidirectional CAG promoter, a CBA promoter, a human ubiquitin C promoter, a synapsin (Syn) promoter, a human synapsin (hSyn) promoter, an EFla promoter, a rhodopsin (Rhod), and any combination thereof.
5. The gene therapy construct any of the previous claims, wherein the polynucleotide further comprises a polyadenylation (poly A) site, a woodchuck hepatitis virus post-transcriptional regulatory element (WPRE), an optimized woodchuck hepatitis virus post-transcriptional regulatory element (oPRE), an intron, a microRNA binding motif, or any combination thereof.
6. The gene therapy construct of claim 5, wherein the polyA site selected from the group consisting of a beta-globin polyA, a bovine growth hormone polyA (bGH polyA), a human growth hormone polyA (hGH polyA), and a SV40 immediate early poly A (SV40 polyA).
7. The gene therapy construct of any of the previous claims, wherein the polynucleotide comprises a woodchuck hepatitis virus post-transcriptional regulatory element (WPRE).
8. The gene therapy construct of any of the previous claims, wherein the polynucleotide comprises an optimized woodchuck hepatitis virus post-transcriptional regulatory element (oPRE).
9. The gene therapy construct any of the previous claims, wherein the polynucleotide comprises an intron.
10. The gene therapy construct of claim 9, wherein the intron is a chimeric intron.
11. The gene therapy construct of claim 9 or 10, wherein the intron is selected from a beta- globin intron or a minute virus of mice (MVM) intron.
12. The gene therapy construct of any of the previous claims, wherein the polynucleotide comprises a 5’ and/or a 3’ untranslated region (UTR).
13. The gene therapy construct of claim 12, wherein the polynucleotide further comprises a microRNA binding motif in the 3’ untranslated region (UTR).
14. The gene therapy construct of any of the previous claims, wherein the PA28A is human isoform 1, human isoform 2, human isoform 3, or a functional mutant thereof.
15. The gene therapy construct of any of the previous claims, wherein the PA28A comprises an amino acid sequence having a sequence of at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity to any of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, or SEQ ID NO: 4.
16. The gene therapy construct any of the previous claims further comprising a nucleic acid sequence encoding Proteasome activator complex subunit 2 (PA28B).
17. The gene therapy construct of claim 16, wherein the construct comprises a first expression cassette comprising the nucleic acid sequence encoding PA28A and a second expression cassette comprising the nucleic acid sequence encoding PA28B.
18. The gene therapy construct of claim 17, wherein the first expression cassette and the second expression cassette comprise the same promoter or different promoters.
19. The gene therapy construct of any of claims 16-18 further comprising a nucleic acid sequence encoding a self-cleaving peptide.
20. The gene therapy construct of claim 18, wherein the self-cleaving peptide is P2A, furin, or intein-F2A.
21. The gene therapy construct of any one of claims 1-20 further comprising two inverted terminal repeats (ITRs).
22. A bicistronic construct comprising a polynucleotide comprising a promoter, a nucleic acid sequence encoding Proteasome activator complex subunit 1 (PA28A), and a nucleic acid sequence encoding Proteasome activator complex subunit 2 (PA28B).
23. The bicistronic construct of claim 22, comprising a first expression cassette comprising the nucleic acid sequence encoding Proteasome activator complex subunit 1 (PA28A) and a second expression cassette comprising the nucleic acid sequence encoding Proteasome activator complex subunit 2 (PA28B).
24. The bicistronic construct of claim 23, wherein the first expression cassette and the second expression cassette comprise the same or different promoters.
25. The bicistronic construct of any of claims 22-24, wherein the polynucleotide further comprises a nucleic acid sequence encoding a self-cleaving peptide.
26. The bicistronic construct of claims 25, wherein the self-cleaving peptide is P2A, furin, or intein-F2A.
27. The bicistronic construct of claim 25 or 26, wherein the polynucleotide comprises the nucleic acid encoding PA28A, the self-cleaving peptide, and the nucleic acid encoding PA28B in 5'-3' orientation.
28. The bicistronic construct of claim 25 or 26, wherein the polynucleotide comprises the nucleic acid encoding PA28B, the self-cleaving peptide, and the nucleic acid encoding PA28A in 5'-3' orientation.
29. The bicistronic construct of any of claims 22-28, wherein the promoter is a heterologous promoter.
30. The bicistronic construct of any of claims 22-29, wherein the promoter is a bi-directional promoter.
31. The bicistronic construct of any of claims 22-30, wherein the promoter is a tissue specific promoter.
32. The bicistronic construct of any of claims 22-31, wherein the promoter is a rhodopsin (Rhod) promoter, synapsin (Syn) promoter, a MecP2 promoter, a GUSB promoter, a CBh promoter, a
PGK promoter, a cone arrestin promoter, a tyrosine hydroxylase promoter, a dopamine transporter promoter, a GRK1 promoter, a cardiac myosin light chain 2 promoter, a CAMKII promoter, a hDlx promoter, a CMV promoter, a CAG promoter, a bidirectional CAG promoter, a CBA promoter, a human rhodopsin (hRho) promoter, a human synapsin (hSyn) promoter, a EFla promoter, or a human ubiquitin C promoter.
33. The bicistronic construct of any of claims 22-32, wherein the polynucleotide further comprises a polyadenylation (poly A) site, a woodchuck hepatitis virus post-transcriptional regulatory element (WPRE), an optimized woodchuck hepatitis virus post-transcriptional regulatory element (oPRE), an intron, a microRNA binding motif, or any combination thereof.
34. The bicistronic construct of claim 33, wherein the polyA site selected from the group consisting of a beta-globin polyA, a bovine growth hormone polyA (bGH polyA), a human growth hormone polyA (hGH polyA), and a SV40 immediate early poly A (SV40 polyA).
35. The bicistronic construct of any of claims 22-34, wherein the polynucleotide comprises a woodchuck hepatitis virus post-transcriptional regulatory element (WPRE).
36. The bicistronic construct of any of claims 22-34, wherein the polynucleotide comprises an optimized woodchuck hepatitis virus post-transcriptional regulatory element (oPRE).
37. The bicistronic construct of any of claims 22-36, wherein the polynucleotide comprises an intron.
38. The bicistronic construct of claim 37, wherein the intron is a chimeric intron.
39. The bicistronic construct of claim 37 or 38, wherein the intron is selected from a beta-globin intron or a minute virus of mice (MVM) intron.
40. The bicistronic construct of any of claims 22-39, wherein the polynucleotide comprises a 5’ and/or a 3’ untranslated region (UTR)
41. The bicistronic construct of claim 40, wherein the polynucleotide further comprises a microRNA binding motif in the 3’ untranslated region (UTR).
42. The bicistronic construct of any of claims 22-41, wherein the PA28A is human isoform 1, human isoform 2, human isoform 3, or a functional mutant thereof.
43. The bicistronic construct of any of claims 22-42, wherein the PA28A comprises an amino acid sequence having a sequence of at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity to any of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, or SEQ ID NO: 4.
44. The bicistronic construct of any of claims 22-43, wherein the PA28B comprises an amino acid sequence having a sequence of at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity to any of SEQ ID NO: 8.
45. The bicistronic construct of any of claims 22-44 which further comprises two ITRs.
46. A vector comprising the gene therapy construct of any one of claims 1-21 or the bicistronic construct of any of claims 22-45.
47. The vector of claim 46, wherein the vector is a viral vector, non-viral vector, or plasmid.
48. The vector of claim 47, wherein the viral vector is adeno-associated viral (AAV) vector, an adenoviral vector, a lentiviral vector, or a retroviral vector.
49. The vector of claim 48, wherein AAV serotype is selected from AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAVrh8, AAV9, AAV10, AAVrhlO, or AAV.PHP.B.
50. A recombinant AAV (rAAV) particle, comprising an AAV capsid, and a vector genome comprising the gene therapy construct of any one of claims 1-21 or the bicistronic construct of any of claims 22-45.
51. The AAV vector of claim 47, wherein AAV serotype is selected from AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAVrh8, AAV9, AAV10, AAVrhlO, or AAV.PHP.B.
52. A host cell comprising the gene therapy construct, the bicistronic construct, the vector, or the rAAV particle of any of the previous claims.
53. A pharmaceutical composition comprising the gene therapy construct, the bicistronic construct, the vector, or the rAAV particle of any of the previous claims.
54. A method of increasing proteasomal activity in a cell, comprising administering to the cell the gene therapy construct, the bicistronic construct, the vector, the rAAV particle, or
pharmaceutical composition of any of the previous claims, thereby increasing proteasomal activity in the cell.
55. The method of claim 54, wherein the cell is a brain cell, ocular cell, or heart muscle cell.
56. A method of treating retinitis pigmentosa, comprising administering to a subject in need thereof the gene therapy construct, the bicistronic construct, the vector, the rAAV particle, or pharmaceutical composition of any of claims 1-51 and 53.
57. A method of treating Huntington’s disease, comprising administering to a subject in need thereof the gene therapy construct, the bicistronic construct, the vector, the rAAV particle, or pharmaceutical composition of any of claims 1-51 and 53.
58. A method of treating Parkinson’s disease, comprising administering to a subject in need thereof the gene therapy construct, the bicistronic construct, the vector, the rAAV particle, or pharmaceutical composition of any of claims 1-51 and 53.
59. A method of treating a disease or condition associated with proteinopathy in a subject comprising administering to the subject the gene therapy construct, the bicistronic construct, the vector, the rAAV particle, or pharmaceutical composition of any of claims 1-47 and 49, wherein the disease or condition is selected from the group consisting of Alzheimer’s disease, Dutch hereditary cerebral hemorrhage with amyloidosis, Congophilic angiopathy, Tauopathies, Corticobasal degeneration, Pick’s disease, Progressive supranuclear palsy, Prion diseases, Creutzfeld-Jacob disease, Gerstmann-Straussler-Schneiker syndrome, fatal familia insomnia, Kuru, Bovine spongiform encephalopathy, Scrapie, Chronic wasting disease, Synucleinopathies, Parkinson’s disease, Lewy body variant of Alzheimer’s disease, Diffuse Lewy body disease, Dementia with Lewy bodies, Multiple system atrophy, Neurodegeneration with brain iron accumulation type I, Amyotrophic lateral sclerosis and frontotemporal lobar degeneration, Amyotrophic lateral sclerosis, Huntington’s disease, Hereditary dentatorubral-pallidoluysian atrophy, Kennedy’s disease or X-linked spinal and bulbar muscular atrophy, Spinocerebellar ataxia 1, Neuronal intranuclear inclusion disease, Spinocerebellar ataxia 2, Spinocerebellar ataxia 3, Spinocerebellar ataxia 6, Spinocerebellar ataxia 7, Spinocerebellar ataxia 17, Alexander’s disease, Cockayne syndrome, or Spinal muscular atrophy.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US202163223840P | 2021-07-20 | 2021-07-20 | |
US63/223,840 | 2021-07-20 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2023004365A1 true WO2023004365A1 (en) | 2023-01-26 |
Family
ID=84979785
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2022/073965 WO2023004365A1 (en) | 2021-07-20 | 2022-07-20 | Vector constructs for delivery of nucleic acids encoding therapeutic proteasome activator complex subunits and methods of using the same |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2023004365A1 (en) |
Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US6767705B2 (en) * | 2000-05-26 | 2004-07-27 | The Board Of Trustees Of The University Of Illinois | Reagents and methods for identifying and modulating expression of genes regulated by retinoids |
US20050266561A1 (en) * | 2003-11-21 | 2005-12-01 | Revivicor, Inc. | Use of interfering RNA in the production of transgenic animals |
US20190241633A1 (en) * | 2016-05-04 | 2019-08-08 | Curevac Ag | Rna encoding a therapeutic protein |
WO2020041791A1 (en) * | 2018-08-24 | 2020-02-27 | Locana, Inc. | Fasl immunomodulatory gene therapy compositions and methods for use |
-
2022
- 2022-07-20 WO PCT/US2022/073965 patent/WO2023004365A1/en unknown
Patent Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US6767705B2 (en) * | 2000-05-26 | 2004-07-27 | The Board Of Trustees Of The University Of Illinois | Reagents and methods for identifying and modulating expression of genes regulated by retinoids |
US20050266561A1 (en) * | 2003-11-21 | 2005-12-01 | Revivicor, Inc. | Use of interfering RNA in the production of transgenic animals |
US20190241633A1 (en) * | 2016-05-04 | 2019-08-08 | Curevac Ag | Rna encoding a therapeutic protein |
WO2020041791A1 (en) * | 2018-08-24 | 2020-02-27 | Locana, Inc. | Fasl immunomodulatory gene therapy compositions and methods for use |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
KR102373765B1 (en) | Capsid-free aav vectors, compositions, and methods for vector production and gene delivery | |
US6346415B1 (en) | Transcriptionally-activated AAV inverted terminal repeats (ITRS) for use with recombinant AAV vectors | |
US20210095313A1 (en) | Adeno-associated virus (aav) systems for treatment of genetic hearing loss | |
KR20180091863A (en) | Extensible method for producing recombinant adeno-associated virus (AAV) vectors in serum-free suspension cell culture systems suitable for clinical use | |
EP3393522A1 (en) | Improved hybrid dual recombinant aav vector systems for gene therapy | |
WO2023284879A1 (en) | Modified aav capsid for gene therapy and methods thereof | |
CN115298316A (en) | Method for producing adeno-associated virus vector | |
WO2018187552A9 (en) | Minigene therapy | |
US20230089490A1 (en) | Raav-mediated in vivo delivery of suppressor trnas | |
JP2023534037A (en) | Compositions Useful for Treating Charcot-Marie-Tooth Disease | |
CA2304801C (en) | Transcriptionally-activated aav inverted terminal repeats (itrs) for use with recombinant aav vectors | |
CN114402075A (en) | Gene therapy for Uschel syndrome (USH2A) | |
EP3953484A1 (en) | Aav-mediated gene therapy for maple syrup urine disease (msud) | |
EP3972610A1 (en) | Minigene therapy | |
US20030190746A1 (en) | Gene expression control system and its use in recombinant virus packaging cell lines | |
WO2023147584A2 (en) | Compositions and methods for treating sialidosis | |
WO2023004365A1 (en) | Vector constructs for delivery of nucleic acids encoding therapeutic proteasome activator complex subunits and methods of using the same | |
WO2022187679A1 (en) | Viral vector constructs incorporating dna for inhibiting toll like receptors and methods of using the same | |
TW202208407A (en) | Gene therapy with dysferlin dual vectors | |
IL293285A (en) | Gene therapy for neurodegenerative disorders | |
US20240269328A1 (en) | Recombinant adeno-associated viruses for lesch-nyhan disorders and uses thereof | |
JP2024515612A (en) | Compositions useful for treating spinal-bulbar muscular atrophy (SBMA) | |
AU2003203790B2 (en) | Transcriptionally-activated AAV inverted terminal repeats (ITRs) for use with recombinant AAV vectors | |
WO2024161032A1 (en) | Gene therapy vectors for use in parkinson's disease | |
KR20230043181A (en) | AAV Vectors Encoding Parkin and Their Uses |
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: 22846824 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
32PN | Ep: public notification in the ep bulletin as address of the adressee cannot be established |
Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205A DATED 13/06/2024) |