WO2018134168A1 - Methods of expressing a polynucleotide of interest in the cone photoreceptors - Google Patents

Methods of expressing a polynucleotide of interest in the cone photoreceptors Download PDF

Info

Publication number
WO2018134168A1
WO2018134168A1 PCT/EP2018/050925 EP2018050925W WO2018134168A1 WO 2018134168 A1 WO2018134168 A1 WO 2018134168A1 EP 2018050925 W EP2018050925 W EP 2018050925W WO 2018134168 A1 WO2018134168 A1 WO 2018134168A1
Authority
WO
WIPO (PCT)
Prior art keywords
polynucleotide
cone
interest
promoter
retinal
Prior art date
Application number
PCT/EP2018/050925
Other languages
French (fr)
Inventor
Deniz DALKARA
José-Alain Sahel
Thierry Leveillard
Jens Duebel
Hanen KHABOU
Original Assignee
INSERM (Institut National de la Santé et de la Recherche Médicale)
Université Pierre Et Marie Curie (Paris 6)
Centre National De La Recherche Scientifique (Cnrs)
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by INSERM (Institut National de la Santé et de la Recherche Médicale), Université Pierre Et Marie Curie (Paris 6), Centre National De La Recherche Scientifique (Cnrs) filed Critical INSERM (Institut National de la Santé et de la Recherche Médicale)
Priority to EP18701288.5A priority Critical patent/EP3570895A1/en
Publication of WO2018134168A1 publication Critical patent/WO2018134168A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal 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
    • A61K48/0058Nucleic acids adapted for tissue specific expression, e.g. having tissue specific promoters as part of a contruct
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector

Definitions

  • the present inventions relate to methods of expressing a polynucleotide of interest in the cone photoreceptors of a subject comprising the intravitreal delivery of a therapeutically effective amount of a recombinant AAV2-derived vector.
  • the fovea -located at the center of the macula- is a specialized region of the retina that dominates the visual perception of primates by providing high acuity color vision (1).
  • the highest density of cones is found at the center of the fovea ( ⁇ 0.3 mm from the foveal center), devoid of rod photoreceptors (2).
  • Cone density decreases by up to 100 fold with distance from the fovea (3).
  • Cone cells in the fovea are the primary targets of gene therapies aiming to treat inherited retinal diseases like mid-stage retinitis pigmentosa (4, 5) and achromatopsia (6).
  • the present inventions relates to methods of expressing a polynucleotide of interest in the cone photoreceptors of a subject comprising the intravitreal delivery of a therapeutically effective amount of a recombinant AAV2 -variant vector.
  • the present invention is defined by the claims.
  • AAV adeno-associated viral
  • the first object of the present invention relates to a method of expressing a polynucleotide of interest in the cone photoreceptors of a subject comprising the intravitreal delivery of a therapeutically effective amount of a recombinant AAV2-variant vector comprising the VP1 capsid protein as set forth in SEQ ID NO: l encoding a polynucleotide of interest under the control of the PR1.7 promoter as set forth in SEQ ID NO:2.
  • the term "subject" is typically intended for a human. Typically the subject is affected or likely to be affected with a retinal disease affecting cone photoreceptors. Accordingly a wide variety of retinal diseases impacting retinal cone photoreceptors may thus be treated given the teachings provided herein and typically include age-related macular degeneration, Bassen-kornzweig syndrome, choroideremia, gyrate atrophy, Refsum syndrome, Usher syndrome, color blindness, blue cone monochromacy, achromatopsia, incomplete achromatopsia, oligocone trichromacy, retinitis pigmentosa (RP), macular degeneration, Stargardt's Disease, Bardet-Biedl syndrome, Bornholm eye disease, Best's Disease and Leber's congenital amaurosis.
  • age-related macular degeneration Bassen-kornzweig syndrome, choroideremia, gyrate atrophy, Refsum syndrome, Usher syndrome, color blindness, blue cone monochro
  • a further object of this invention is to provide a method for treating a retinal disease affecting cone photoreceptors in a subject in need thereof comprising the intravitreal delivery of a therapeutically effective amount of a recombinant AAV2 -variant vector comprising the VP1 capsid protein as set forth in SEQ ID NO: l and the polynucleotide of interest under the control of the PR1.7 promoter as set forth in SEQ ID NO:2 wherein the polynucleotide of interest which when expressed in cone photoreceptor has a beneficial effect on the retinal disease.
  • treatment refers to both prophylactic or preventive treatment as well as curative or disease modifying treatment, including treatment of patient at risk of contracting the disease or suspected to have contracted the disease as well as patients who are ill or have been diagnosed as suffering from a disease or medical condition, and includes suppression of clinical relapse.
  • the treatment may be administered to a subject having a medical disorder or who ultimately may acquire the disorder, in order to prevent, cure, delay the onset of, reduce the severity of, or ameliorate one or more symptoms of a disorder or recurring disorder.
  • therapeutic regimen is meant the pattern of treatment of an illness, e.g., the pattern of dosing used during therapy.
  • a therapeutic regimen may include an induction regimen and a maintenance regimen.
  • induction regimen or “induction period” refers to a therapeutic regimen (or the portion of a therapeutic regimen) that is used for the initial treatment of a disease.
  • the general goal of an induction regimen is to provide a high level of drug to a patient during the initial period of a treatment regimen.
  • An induction regimen may employ (in part or in whole) a "loading regimen", which may include administering a greater dose of the drug than a physician would employ during a maintenance regimen, administering a drug more frequently than a physician would administer the drug during a maintenance regimen, or both.
  • maintenance regimen refers to a therapeutic regimen (or the portion of a therapeutic regimen) that is used for the maintenance of a patient during treatment of an illness, e.g., to keep the patient in remission for long periods of time (months or years).
  • a maintenance regimen may employ continuous therapy (e.g., administering a drug at regular intervals, e.g., weekly, monthly, yearly, etc.) or intermittent therapy (e.g., interrupted treatment, intermittent treatment, treatment at relapse, or treatment upon achievement of a particular predetermined criteria [e.g., disease manifestation, etc.]).
  • cone photoreceptors has its general meaning in the art and are one of three types of photoreceptor cells in the retina of mammalian eyes. They are responsible for color vision and function best in relatively bright light, as opposed to rod cells, which work better in dim light.
  • the combination between the specific vector and the PR1.7 promoter specially drives the expression of the polynucleotide of interest in cone photoreceptors.
  • vitreous humour also called vitreous body or simply vitreous
  • the clear gel fills the space between the lens and the retina of the eyeball of humans and other vertebrates.
  • polynucleotide of interest designates any nucleotide sequence coding for any polypeptide, structural protein, enzyme etc., the expression of which is wanted in a target cell, for any kind of reason. It can also designate a non-coding sequence, for example an antisense sequence or the sequence of an interfering RNA aimed at decreasing the expression of a gene.
  • a non-coding sequence for example an antisense sequence or the sequence of an interfering RNA aimed at decreasing the expression of a gene.
  • Gene therapy can be performed either by introducing in cone photoreceptor a functional copy of a polynucleotide of interest (e.g. a gene) that is deficient therein (gene replacement therapy), or by delivering to cone photoreceptors a polynucleotide of interest which will have a beneficial effect on the eye disease to be treated (symptomatic therapy).
  • a polynucleotide of interest e.g. a gene
  • gene replacement therapy e.g. a functional copy of a polynucleotide of interest
  • RPGRORF15 retinitis pigmentosa GTPase regulator
  • GNAT2 cone specific alpha subunit of transducin
  • CNGA3 alpha subunit of the cone cyclic nucleotide-gated cation channel
  • the polynucleotide of interest may encode for a neurotrophic factor.
  • the "neurotrophic factor” is a generic term of proteins having a physiological action such as survival and maintenance of nerve cells, promotion of neuronal differentiation.
  • the neurotrophic factor is RdCVF.
  • RdCVF has its general meaning in the art and refers to rod-derived cone viability factor (Leveillard T, Mohand-Sa ' id S, Lorentz O, Hicks D, Fintz A C, Clerin E et al. Identification and characterization of rod-derived cone viability factor. Nat Genet 2004; 36: 755-759.).
  • the polynucleotide of interest encodes both a long form (RdCVF-L, 217 aa, Q8VC33) having a putative thiol-oxydoreductase activity (JEFFERY, Trends Biochem. Sci., vol.24(l):8-l 1, 1999; JEFFERY, Trends Genet, vol.19(8) :415-417, 2003) and a short form (RdCVF-S, 109 aa, Q91W38) with trophic activity for cones but no redox activity.
  • the neurotrophic factor is RdCVF2, which shares many similarities with RdCVF in terms of gene structure, expression in a rod-dependent manner and protein 3D structure (see e.g. WO2008148860 and Chalmel F, Leveillard T, Jaillard C, Lardenois A, Berdugo N, Morel E, Koehl P, Lambrou G, Holmgren A, Sahel JA, Poch O.
  • Rod-derived Cone Viability Factor-2 is a novel bifunctional-thioredoxin-like protein with therapeutic potential. BMC Mol Biol. 2007 Aug 31;8:74.).
  • the RdCVF2 short isoform exhibits cone rescue activity that is independent of its putative thiol-oxydoreductase activity.
  • the polynucleotide of interest encodes for RdCVFL2.
  • the polynucleotide product of interest is an opsin.
  • the opsin sequence can be derived from any suitable single- or multicellular- organism, including human, algae and bacteria.
  • the opsin is rhodopsin, photopsin, L/M wavelength (red/green) cone-opsin, or short wavelength (S) cone-opsin (blue).
  • the opsin is channelrhodopsin or halorhodopsin or cruxhalorhodopsin.
  • the opsin is a light-responsive opsin as described in U.S. Patent Publication No. 2007/0261127 (e.g., ChR2; Chop2); U.S.
  • opsins include NpHR, eNpHR 1.0, eNpHR 2.0, eNpHR 3.0, SwiChR, SwiChR 2.0, SwiChR 3.0, Mac, Mac 3.0, Arch, ArchT, Arch 3.0, ArchT 3.0, iChR, ChR2, C1V1-T, C1V1-TT, Chronos, Chrimson, ChrimsonR, CatCh, VChRl-SFO, ChR2-SFO, ChR2-SSFO, ChEF, ChlEF, Jaws, ChloC, Slow ChloC, iClC2, iClC2 2.0, and iClC2 3.0.
  • the opsin consists of the amino acid sequence as set forth in SEQ ID NO: 3 or SEQ ID NO:
  • the polynucleotide product of interest is a site-specific endonuclease that provides for site-specific knock-down of gene function, e.g., where the endonuclease knocks out an allele associated with a retinal disease.
  • a site-specific endonuclease can be targeted to the defective allele and knock out the defective allele.
  • the vector thus comprises a polynucleotide that encodes a site-specific endonuclease; and a polynucleotide that encodes a functional copy of a defective allele, where the functional copy encodes a functional retinal protein.
  • Site-specific endonucleases that are suitable for use include, e.g., zinc finger nucleases (ZFNs); transcription activator-like effector nucleases (TALENs), and CRISPR-associated endonuclease.
  • CRISPR-associated endonuclease has its general meaning in the art and refers to clustered regularly interspaced short palindromic repeats associated which are the segments of prokaryotic DNA containing short repetitions of base sequences.
  • the CRISPR-associated endonuclease is Cas9 or a derivative thereof.
  • the Cas9 nuclease can have a nucleotide sequence identical to the wild type Streptococcus pyrogenes sequence. Alternatively, the wild type Streptococcus pyrogenes Cas9 sequence can be modified.
  • the Cas9 nuclease sequence can be for example, the sequence contained within a commercially available vector such as PX330 or PX260 from Addgene (Cambridge, MA).
  • the Cas9 endonuclease can have an amino acid sequence that is a variant or a fragment of any of the Cas9 endonuclease sequences of Genbank accession numbers KM099231.1 GL669193757; KM099232.1; GL669193761; or KM099233.1 GL669193765 or Cas9 amino acid sequence of PX330 or PX260 (Addgene, Cambridge, MA).
  • the Cas9 nucleotide sequence can be modified to encode biologically active variants of Cas9, and these variants can have or can include, for example, an amino acid sequence that differs from a wild type Cas9 by virtue of containing one or more mutations (e.g., an addition, deletion, or substitution mutation or a combination of such mutations).
  • the Cas9 nuclease can be mutated in the conserved FiNH and RuvC domains, which are involved in strand specific cleavage.
  • the vector comprises one or more guide RNA.
  • the term "one or more guide RNA” refers to the RNAs that guide the insertion or deletion of residues. In the context of the invention, the guide RNA is used for recruiting Cas9 to specific genomic loci.
  • the guide RNA can be a sequence complementary to a coding or a non-coding sequence.
  • the subject is administered with a combination of at least one vectors comprising one polynucleotide encoding for a Cas9 endonuclease and at least one vector comprising the guide RNA.
  • the polynucleotide product is an interfering RNA (RNAi), in particular a siRNA.
  • RNAi interfering RNA
  • a "small interfering” or “short interfering RNA” or siRNA is a RNA duplex of nucleotides that is targeted to a gene interest (a "target gene”).
  • An “RNA duplex” refers to the structure formed by the complementary pairing between two regions of a RNA molecule.
  • siRNA is "targeted” to a gene in that the nucleotide sequence of the duplex portion of the siRNA is complementary to a nucleotide sequence of the targeted gene.
  • the length of the duplex of siRNAs is less than 30 nucleotides.
  • the polynucleotide product is an antisense oligonucleotide.
  • antisense oligonucleotide is understood to refer to a nucleotide sequence which is substantially complementary to a target nucleotide sequence in a pre- mRNA molecule, hnRNA (heterogenous nuclear RNA) or mRNA molecule.
  • the degree of complementarity (or substantial complementarity) of the antisense sequence is preferably such that a molecule comprising the antisense sequence can form a stable hybrid with the target nucleotide sequence in the RNA molecule under physiological conditions.
  • AAV refers to more than 30 naturally occurring and available adeno-associated viruses, as well as artificial AAVs.
  • AAV capsid, ITRs, and other selected AAV components described herein may be readily selected from among any AAV, including, without limitation, AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV6.2, AAV7, AAV8, AAV9, rhlO, AAVrh64Rl, AAVrh64R2, rh8, variants of any of the known or mentioned AAVs or AAVs yet to be discovered or variants or mixtures thereof. See, e.g., WO 2005/033321.
  • GenBank and PDB Accession Numbers NC_002077 and 3NG9 (AAV-1), AF043303 and 1LP3 (AAV-2), NC_001729 (AAV-3), U89790 and 2G8G (AAV- 4), NC_006152 and 3NTT (AAV-5), 30AH (AAV6), AF513851 (AAV-7), NC_006261 and 2QA0 (AAV-8), AY530579 and 3UX1 (AAV-9 (isolate hu.14)); the disclosures of which are incorporated by reference herein for teaching AAV nucleic acid and amino acid sequences. See also, e.g., Srivistava et al. (1983) J.
  • the term "recombinant AAV2-derived vector” refers to an AAV2 -based vector comprising the VPl capsid protein as set forth in SEQ ID NO: l and the polynucleotide sequence of interest (i.e., a polynucleotide heterologous to AAV).
  • the native VPl capsid protein of AAV2 is substituted by the VPl capsid protein as set forth in SEQ ID NO: l .
  • the recombinant AAV2- derived vector of the present invention typically comprises 5' and 3' adeno-associated virus inverted terminal repeats (ITRs), the polynucleotide of interest (i.e a heterologous polynucleotide) operatively linked to the promoter PR1.7.
  • ITRs adeno-associated virus inverted terminal repeats
  • the vectors of the invention are produced using methods known in the art.
  • the methods generally involve (a) the introduction of the DNA necessary for AAV replication and synthesis of the capsid, (b) the introduction of a helper construct into the producer cell line, wherein the helper construct comprises the viral functions missing from the AAV vector (c) introducing a helper virus into the producer cell line, (d) the plasmid construct containing the genome of the AAV vector, e.g. ITRs, promoter and transgene sequences, etc.... All functions for AAV virion replication and packaging need to be present, to achieve replication and packaging of the AAV vector into AAV virions.
  • the introduction into the producer host cell can be carried out using standard virology techniques simultaneously or sequentially.
  • the host cells are cultured to produce AAV virions and are purified using standard techniques such as iodixanol or CsCl gradients or other purification methods.
  • the purified AAV virion is then ready for use in the methods.
  • promoter has its general meaning in the art and refers to a nucleic acid fragment that controls the transcription of one or more genes, located upstream with respect to the direction of transcription of the transcription initiation site of the gene, and is structurally identified by the presence of a binding site for DNA-dependent R A polymerase, transcription initiation sites and any other DNA sequences, including, but not limited to transcription factor binding sites, repressor and activator protein binding sites, and any other sequences of nucleotides known to one of skill in the art to act directly or indirectly to regulate the amount of transcription from the promoter.
  • PR1.7 promoter refers to the 1.7-kb L-opsin promoter described in Hum Gene Ther. 2016 Jan;27(l):72-82 and characterized by the nucleic acid sequence as set forth in SEQ ID NO:2.
  • the promoter and the polynucleotide of interest are operatively linked.
  • operably linked refers to two or more nucleic acid or amino acid sequence elements that are physically linked in such a way that they are in a functional relationship with each other.
  • a promoter is operably linked to a coding sequence if the promoter is able to initiate or otherwise control/regulate the transcription and/or expression of a coding sequence, in which case the coding sequence should be understood as being "under the control of the promoter.
  • the coding sequence should be understood as being "under the control of the promoter.
  • two nucleic acid sequences when operably linked, they will be in the same orientation and usually also in the same reading frame. They will usually also be essentially contiguous, although this may not be required.
  • a “therapeutically effective amount” is meant a sufficient amount of the vector to treat the retinal disease at a reasonable benefit/risk ratio. It will be understood that the total daily usage of the vector will be decided by the attending physician within the scope of sound medical judgment.
  • the specific therapeutically effective dose level for any particular patient will depend upon a variety of factors including the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific polypeptide employed; and like factors well known in the medical arts.
  • the doses of vectors may be adapted depending on the disease condition, the subject (for example, according to his weight, metabolism, etc.), the treatment schedule, etc.
  • a preferred effective dose within the context of this invention is a dose allowing an optimal transduction of the cone photoreceptors.
  • from 10 8 to 10 10 viral genomes (vg) are administered per dose in mice.
  • the doses of AAV vectors to be administered in humans may range from 10 9 to 10 12 vg.
  • compositions may comprise, in addition to the vector, a pharmaceutically acceptable excipient, carrier, buffer, stabiliser or other materials well known to those skilled in the art. Such materials should be non-toxic and should not interfere with the efficacy of the active ingredient (i.e. the vector of the invention).
  • a pharmaceutically acceptable excipient such as water, petroleum, animal or vegetable oils, mineral oil or synthetic oil.
  • Physiological saline solution magnesium chloride, dextrose or other saccharide solution or glycols such as ethylene glycol, propylene glycol or polyethylene glycol may be included.
  • the active ingredient will be in the form of an aqueous solution, which is pyrogen-free and has suitable pH, isotonicity and stability.
  • isotonic vehicles such as Sodium Chloride Injection, Ringer's Injection, Lactated Ringer's Injection.
  • Preservatives, stabilisers, buffers, antioxidants and/or other additives may be included, as required.
  • the vector may be included in a pharmaceutical composition, which is formulated for slow release, such as in microcapsules formed from biocompatible polymers or in liposomal carrier systems according to methods known in the art.
  • a pharmaceutical composition of the present invention is supplied in a prefilled syringe.
  • a "ready- to-use syringe” or “prefilled syringe” is a syringe which is supplied in a filled state, i.e. the pharmaceutical composition to be administered is already present in the syringe and ready for administration.
  • Prefilled syringes have many benefits compared to separately provided syringe and vial, such as improved convenience, affordability, accuracy, sterility, and safety.
  • the pH of the liquid pharmaceutical composition of the present invention is in the range of 5.0 to 7.0, 5.1 to 6.9, 5.2 to 6.8, 5.3 to 6.7 or 5.4 to 6.6.
  • Figure 1 Expression in all layers of the mouse retina is obtained with the combination of AAV2-7m8-GFP and the mouse cone arrestin (mCAR) promoter (A), PR2.1 promoter (B) while most cone-specific expression pattern is achieved with PR1.7 promoter (C) -after intravitreal injections.
  • mCAR mouse cone arrestin
  • FIG. 2 Cone-specific expression in the retinal degeneration 10 (rdlO) mouse model for Retinitis Pigmentosa.
  • Figure 3 Reporter gene expression pattern using ubiquitous versus cone-specific promoter in vivo in monkeys after intravitreal injections.
  • Retinal flatmount images showing that AAV2-7m8-CMV leads to pan-retinal GFP expression (A) while it is foveal cone- specific with AAV2-7m8-PR1.7 (B). Close up to the fovea with AAV2-7m8-CMV (C, left) and AAV2-7m8-PR1.7 (C, right).
  • Figure 4 Foveal cone-specific expression of Jaws-GFP under the control of PR1.7 promoter in combination with AAV2-7m8 capsid in the non-human primate, after intravitreal injections.
  • A In vivo eye fundus image showing foveal expression
  • B flatmount image of a half fovea (arrow: foveola)
  • C retinal cryosections at the level of the fovea.
  • Figure 5 Cone-specific expression of GFP under the control of PR1.7 promoter and with AAV2-7m8 capsid in human tissue.
  • A Retinal organoids derived from human induced pluripotent stem cells expres GFP;
  • B GFP-expressing cells in the organoids are exclusively cones (cryosections),
  • C expression in cones of a human post-mortem retinal explant (cryosections).
  • AAV vectors were produced as previously described using the co-transfection method and purified by iodixanol gradient ultracentrifugation (49). AAV vector stocks were titered by quantitative PCR (50) using SYBR Green (Thermo Fischer Scientific). Animals and intraocular injections
  • mice Wild-type C57BL6/j or rdlO mice, and cynomolgus macaques were used for this study.
  • mice were anesthetized by isofluorane inhalation. Pupils were dilated and a 33 -gauge needle was inserted into the eye to deliver 2 ⁇ , of AAV vector solution intravitreally or ⁇ ⁇ subretinally.
  • macaques were first selected based on absence neutralizing antibody titers against AAV. Primates were anesthetized with 10: 1 mg/kg ketamine/xylazine. After subretinal or intravitreal vector administration, opthtalmic steroid and antibiotic ointment were applied to the corneas post injections.
  • Spectralis HRA+OCT system Heidelberg Engineering, Germany
  • fluorescent images of GFP using the "Fundus Autofluoresence mode" which consists of and excitation wavelength of 488 nm and barrier filter of 500 nm.
  • a two-photon microscope equipped with a 40x water immersion objective (LUMPLFLN40x/W/0.80, Olympus) with a pulsed femto-second laser (InSightTM DeepSeeTM - Newport Corporation) was used for imaging GFP-positive retinal cells from whole-mount retinas (with photoreceptor-cell-side up) or retina slices (vertical sections).
  • AAV-treated macaque retinas were isolated and later imaged in oxygenized (95% 02, 5% C02) Ames medium (Sigma-Aldrich).
  • retinas were placed in the recording chamber of the microscope, and z-stacks were acquired using the excitation laser at a wavelength of 930 nm. Images were processed offline using Image J.
  • Electrodes were made from borosilicate glass (BF100- 50-10, Sutter Instruments) and pulled to 6-9 ⁇ . Pipettes were filled with 115 mM K Gluconate, 10 mM KC1, 1 mM MgC12, 0.5 mM CaC12, 1.5 mM EGTA, 10 mM HEPES, and 4 mM ATP-Na2 (pH 7.2). Cells were clamped at a potential of -40 mV in voltage-clamp configuration, or recorded in current-clamp (current zero) configuration. Retinas were dark-adapted at least half an hour in the recording chamber prior to recordings.
  • the retinal organoids were infected at day 28 of differentiation at a dose of 5xl0 10 vg/organoid with AAV2-7m8 vectors carrying the GFP gene under the control of pR1.7 promoter. 10 ⁇ DAPT (Selleck) was added to the medium for a week from day 28 on to promote cell cycle arrest of the existent cell populations. Fluorescence intensity was observed for the first time 5 days after infection and continued to increase up to day 43.
  • Human retinal explants were prepared using a previously described protocol (38). Briefly, eyes were dissected in C02-independent-medium (Thermo Fischer Scientific). The anterior parts were removed, retina was isolated and cut into small pieces. These explants were placed photoreceptor side-up on a Transwell cell culture insert (Corning), and 2mL of Neurobasal medium (Thermo Fischer Scientific) supplemented with B27 (Thermo Fischer Scientific) were added to each well below each explant. The following day, each explant was infected with a single 0,5 drop of AAV-pRl .7-GFP containing 10 10 viral particles. Vector- infected explants were incubated for 10-15 days to allow GFP expression, which was checked using an epifiuorescence macroscope.
  • mice were enucleated and immediately fixed in 10% formalin - 4% formaldehyde for 2 hours for cryosections.
  • Macaque retinas were fixed after dissection in 4% formaldehyde for 3 hours.
  • Retinal organoids and human retinal explants were rinsed in PBS at the end of their culture periods and fixed in 4% paraformaldehyde for 10 minutes.
  • mouse and macaque retinas, retinal organoids and human retinal explants were immersed in PBS-30% sucrose overnight at 4°C.
  • TF binding site analysis was performed on red opsin gene promoter sequence -pR2.1 and pR1.7 sequences- and the cone arrestin 3 genomic region.
  • the TRANSFAC database 8.3 http://alggen.lsi.upc.es/) was used for TF binding site prediction.
  • Each TF from the predicted list was analyzed using the Knowledge Base for Sensory System (KBASS, http://kbass.institut-vision.org/KBaSS/transcriptomics/index.php) to select those expressed in human retina using the transcriptomic experiment RNG209 (51).
  • a filter was used to retain TFs with a signal intensity value superior to 40 units in the sample prepared from the experiment RNG209 after normalization by Robust Multi-array Average (RMA) as previously described (52).
  • RMA Robust Multi-array Average
  • human retinal specimens used as controls were post-mortem specimens collected within 12 hours following death of patients with no past medical history of eye disease or diabetes.
  • Nineteen samples were collected from 19 eyes representing 17 patients. Sex ratio was 12 men / 7 women with a mean age of 61 years (range 25-78 years).
  • mCAR promoter lead to GFP expression in some cones, but was leaky towards rods as well as cells of the inner nuclear layer (INL) and ganglion cell layer (GCL) ( Figure 1). Both PR2.1 and PR 1.7 promoters lead to more cone labeling than mCAR promoter ( Figure 1). PR2.1 transduced more cones than PR1.7 but, it also produced non-specific GFP expression in the INL and GCL. Only the PR1.7 promoter showed GFP expression in cones with minimal expression in rods and was not leaky towards the inner retina (data not shown).
  • AAV2-7m8-PR1.7 vector-promoter combination in a mouse model of retinal degeneration.
  • AAV2- 7m8-PR1.7-GFP intravitreally in the rdlO mouse model of retinitis pigmentosa.
  • GFP expression was restricted to cones ( Figure 2) supporting the suitability of this vector for cone-directed gene therapy via both intravitreal and subretinal injections.
  • COUP-TFI has been shown to suppress green opsin gene (Opnlmw) expression in the mouse retina (22) and might thus be accountable for lower expression with PR2.1 promoter in macaque cones when AAV is delivered subretinally as previously shown (18).
  • COUP-TFI has been shown to suppress green opsin gene (Opnlmw) expression in the mouse retina (22) and might thus be accountable for lower expression with PR2.1 promoter in macaque cones when AAV is delivered subretinally as previously shown (18).
  • COUP-TFI has been shown to suppress green opsin gene (Opnlmw) expression in the mouse retina (22) and might thus be accountable for lower expression with PR2.1 promoter in macaque cones when AAV is delivered subretinally as previously shown (18).
  • OFPB green opsin gene
  • GTF2I ubiquitous activator TFs
  • the short sequence consists of a 521bp portion of the genomic proximal CAR promoter (data not shown) presenting a TATA-box, a TATA-like box, as well as binding sites for CRX (Cone-rod homeobox protein) and SP (Specificity Protein) TFs (23) (data not shown).
  • CRX Cone-rod homeobox protein
  • SP Specificity Protein
  • CRX and SP TFs interact with each other and with RARA (Retinoic Acid Receptor Alpha), RXRA (Retinoid X Receptor Alpha) and THRB (Thyroid Hormone Receptor Beta) TFs (data not shown).
  • RARA Retinoic Acid Receptor Alpha
  • RXRA Retinoid X Receptor Alpha
  • THRB thyroid Hormone Receptor Beta
  • PRl.7 promoter drives strong and highly specific gene expression in human cones
  • the fovea accounts for less than 1% of the retinal surface area in primates yet it provides the input to about 50% of the cells in the primary visual cortex (1).
  • the high concentration of cones in the fovea, the thinnest and most delicate part of the retina allows for high acuity vision and it is of utmost importance to preserve the unique functions (39) and architecture (40) of the cones in this area during therapeutic interventions.
  • Foveal cones can be targeted via different administration routes, using either subretinal or intravitreal injections (35, 41, 42) but detaching the fovea might lead to mechanical damage, especially in the degenerating retina (43). For all of these reasons, ways to deliver therapeutics to the fovea, without detaching this region are needed.
  • Intravitreal injections are a surgically simple way to deliver therapeutics without retinal detachment.
  • Gene therapy vectors can target the outer retina via intravitreal injections in rodents without damage to the photoreceptors (17, 35).
  • safe and efficient gene delivery to primate cones via intravitreal injection had not been achieved so far, likely due to the substantial dilution of the vector in the vitreous and resulting loss of efficacy.
  • the use of cell-type specific promoters that provide high-level gene expression with a lower local concentration is critical to overcome this challenge (29, 44).
  • mCAR and PR2.1 gave rise to non-specific expression in inner retinal cells, making them unsuitable for optogenetic applications where any expression in downstream neurons would cancel out the response from the photoreceptors.
  • Subsequent in silico analysis of TF binding sites within each promoter sequence proposed basis for more specific transduction with PR1.7 and the observed the lack of specificity with mCAR promoter.
  • AAV2-7m8 equipped with PR1.7 promoter to transduce foveal cones.
  • Table 1 AAV vector administration strategies for cone-directed gene therapy.
  • High acuity vision that includes the fovea expression pattern
  • SEQ ID NO: 1 VP1 capsid of the recombinant AAV2-derived vector
  • Wikler KC Williams RW, Rakic P. Photoreceptor mosaic: Number and distribution of rods and cones in the rhesus monkey retina. J. Comp. Neurol. 1990;297(4):499-508.
  • Vandenberghe LH et al. AAV9 targets cone photoreceptors in the nonhuman primate retina.

Abstract

Intraocular injection of adeno-associated viral (AAV) vectors has been an evident route for delivering gene drugs into the retina. Currently, the vectors need to be injected into the subretinal space in order to provide gene delivery to cones. In this approach, gene delivery is limited to cells that contact the local "bleb" of injected fluid. Furthermore, retinal detachment that occurs during subretinal injections is a concern in eyes with retinal degeneration. Here, the inventors establish several new vector-promoter combinations to overcome the limitations associated with AAV-mediated cone transduction in the fovea with supporting studies in mouse models, human induced pluripotent stem cell-derived organoids, post-mortem human retinal explants and living macaques. They show that an engineered AAV2 variant provides gene delivery to foveal cones with a well-tolerated dose administered intravitreally. The delivery modality relies on a cone-specific promoter and result in high-level transgene expression compatible with optogenetic vision restoration. Accordingly, the present invention relates to method of expressing a polynucleotide of interest in the cone photoreceptors of a subject comprising intravitreal delivery of a therapeutically effective amount of a recombinant AAV2-derived vector comprising a VP1 capsid protein as set forth in SEQ ID NO: 1 and the polynucleotide of interest under the control of the PR1.7 promoter as set forth in SEQ ID NO:2.

Description

METHODS OF EXPRESSING A POLYNUCLEOTIDE OF INTEREST IN THE
CONE PHOTORECEPTORS
FIELD OF THE INVENTION:
The present inventions relate to methods of expressing a polynucleotide of interest in the cone photoreceptors of a subject comprising the intravitreal delivery of a therapeutically effective amount of a recombinant AAV2-derived vector.
BACKGROUND OF THE INVENTION:
The fovea -located at the center of the macula- is a specialized region of the retina that dominates the visual perception of primates by providing high acuity color vision (1). The highest density of cones is found at the center of the fovea (<0.3 mm from the foveal center), devoid of rod photoreceptors (2). Cone density decreases by up to 100 fold with distance from the fovea (3). Cone cells in the fovea are the primary targets of gene therapies aiming to treat inherited retinal diseases like mid-stage retinitis pigmentosa (4, 5) and achromatopsia (6). Currently, viral vectors encoding therapeutic proteins need to be injected into the subretinal space between the photoreceptors and the retinal pigment epithelium (RPE) cells in order to provide gene delivery to cones. In this approach, gene delivery is limited to cells that contact the local "bleb" of injected fluid. Furthermore, retinal detachment that occurs during subretinal injections is a concern in eyes with retinal degeneration. The earliest clinical trials using subretinal delivery of adeno-associated virus (AAV) to deliver a healthy RPE65 gene in Leber's Congenital Amaurosis patients (7-9) led to some improvements in vision despite the detachment of the macula to deliver the viral vector (10, 11). However, the treatment was in certain cases complicated by macular holes and increased macular thinning in the case of sub-foveal injections (11). Furthermore, contrary to the surrounding regions, there were no treatment benefits in the fovea (12). Gene therapy using AAV has also been studied for patients with choroideremia in which the macula was the target for gene delivery (13). The 6 month follow up results from this latter study thus far suggest that sub-foveal retinal detachment does not cause vision reduction in this region but, one of the patients in this trial had visual acuity loss in the treated eye compared to his untreated eye (13). With more gene therapies reaching clinical stages of application there is a growing need to find new methods for delivering gene therapy to the fovea without detaching this brittle region (14). This can be achieved by engineering the viral vectors to permit gene delivery away from the injection site. AAV capsids that can provide gene delivery to foveal cones after injection into the vitreous humor are one possible option.
SUMMARY OF THE INVENTION:
The present inventions relates to methods of expressing a polynucleotide of interest in the cone photoreceptors of a subject comprising the intravitreal delivery of a therapeutically effective amount of a recombinant AAV2 -variant vector. In particular, the present invention is defined by the claims.
DETAILED DESCRIPTION OF THE INVENTION:
Intraocular injection of adeno-associated viral (AAV) vectors has been an evident route for delivering gene drugs into the retina. However, gaps in our understanding of AAV transduction patterns within the anatomically unique environments of the intravitreal space of the primate eye impeded the establishment of non-invasive and efficient gene delivery to foveal cones in the clinic. Here, the inventors establish several new vector-promoter combinations to overcome the limitations associated with AAV-mediated cone transduction in the fovea with supporting studies in mouse models, human induced pluripotent stem cell- derived organoids, post-mortem human retinal explants and living macaques. They show that an engineered AAV2 variant provides gene delivery to foveal cones with a well-tolerated dose administered intravitreally . This delivery modality relies on a cone-specific promoter and result in high-level transgene expression compatible with optogenetic vision restoration.
Accordingly the first object of the present invention relates to a method of expressing a polynucleotide of interest in the cone photoreceptors of a subject comprising the intravitreal delivery of a therapeutically effective amount of a recombinant AAV2-variant vector comprising the VP1 capsid protein as set forth in SEQ ID NO: l encoding a polynucleotide of interest under the control of the PR1.7 promoter as set forth in SEQ ID NO:2.
As used herein, the term "subject" is typically intended for a human. Typically the subject is affected or likely to be affected with a retinal disease affecting cone photoreceptors. Accordingly a wide variety of retinal diseases impacting retinal cone photoreceptors may thus be treated given the teachings provided herein and typically include age-related macular degeneration, Bassen-kornzweig syndrome, choroideremia, gyrate atrophy, Refsum syndrome, Usher syndrome, color blindness, blue cone monochromacy, achromatopsia, incomplete achromatopsia, oligocone trichromacy, retinitis pigmentosa (RP), macular degeneration, Stargardt's Disease, Bardet-Biedl syndrome, Bornholm eye disease, Best's Disease and Leber's congenital amaurosis. Accordingly, a further object of this invention is to provide a method for treating a retinal disease affecting cone photoreceptors in a subject in need thereof comprising the intravitreal delivery of a therapeutically effective amount of a recombinant AAV2 -variant vector comprising the VP1 capsid protein as set forth in SEQ ID NO: l and the polynucleotide of interest under the control of the PR1.7 promoter as set forth in SEQ ID NO:2 wherein the polynucleotide of interest which when expressed in cone photoreceptor has a beneficial effect on the retinal disease.
As used herein, the term "treatment" or "treat" refer to both prophylactic or preventive treatment as well as curative or disease modifying treatment, including treatment of patient at risk of contracting the disease or suspected to have contracted the disease as well as patients who are ill or have been diagnosed as suffering from a disease or medical condition, and includes suppression of clinical relapse. The treatment may be administered to a subject having a medical disorder or who ultimately may acquire the disorder, in order to prevent, cure, delay the onset of, reduce the severity of, or ameliorate one or more symptoms of a disorder or recurring disorder. By "therapeutic regimen" is meant the pattern of treatment of an illness, e.g., the pattern of dosing used during therapy. A therapeutic regimen may include an induction regimen and a maintenance regimen. The phrase "induction regimen" or "induction period" refers to a therapeutic regimen (or the portion of a therapeutic regimen) that is used for the initial treatment of a disease. The general goal of an induction regimen is to provide a high level of drug to a patient during the initial period of a treatment regimen. An induction regimen may employ (in part or in whole) a "loading regimen", which may include administering a greater dose of the drug than a physician would employ during a maintenance regimen, administering a drug more frequently than a physician would administer the drug during a maintenance regimen, or both. The phrase "maintenance regimen" or "maintenance period" refers to a therapeutic regimen (or the portion of a therapeutic regimen) that is used for the maintenance of a patient during treatment of an illness, e.g., to keep the patient in remission for long periods of time (months or years). A maintenance regimen may employ continuous therapy (e.g., administering a drug at regular intervals, e.g., weekly, monthly, yearly, etc.) or intermittent therapy (e.g., interrupted treatment, intermittent treatment, treatment at relapse, or treatment upon achievement of a particular predetermined criteria [e.g., disease manifestation, etc.]).
As used herein, the term "cone photoreceptors" has its general meaning in the art and are one of three types of photoreceptor cells in the retina of mammalian eyes. They are responsible for color vision and function best in relatively bright light, as opposed to rod cells, which work better in dim light. The combination between the specific vector and the PR1.7 promoter specially drives the expression of the polynucleotide of interest in cone photoreceptors.
As used herein, the term "intravitreal delivery" refers to the administration of the vector directly into the vitreous humour (also called vitreous body or simply vitreous) which is the clear gel that fills the space between the lens and the retina of the eyeball of humans and other vertebrates.
As used herein, the term "polynucleotide of interest" herein designates any nucleotide sequence coding for any polypeptide, structural protein, enzyme etc., the expression of which is wanted in a target cell, for any kind of reason. It can also designate a non-coding sequence, for example an antisense sequence or the sequence of an interfering RNA aimed at decreasing the expression of a gene. One skilled in the art knows, by its knowledge of the scientific literature in his field, which are the polynucleotides that may be more appropriate to treat a specific retinal disease.
Gene therapy can be performed either by introducing in cone photoreceptor a functional copy of a polynucleotide of interest (e.g. a gene) that is deficient therein (gene replacement therapy), or by delivering to cone photoreceptors a polynucleotide of interest which will have a beneficial effect on the eye disease to be treated (symptomatic therapy). Examples of polynucleotides of interest that can be used for gene replacement therapy are genes that are specifically or preferentially expressed in cone photoreceptors, such as retinitis pigmentosa GTPase regulator (RPGRORF15) (refs: 1. B. S. Pawlyk, O. V. Bulgakov, X. Sun, M. Adamian, X. Shu, A. J. Smith, E. L. Berson, R. R. Ali, S. Khani, A. F. Wright, M. A. Sandberg, T. Li, Photoreceptor rescue by an abbreviated human RPGR gene in a murine model of X-linked retinitis pigmentosa. Gene Ther 23, 196-204 (2016).; 2. D. H. Hong, B. S. Pawlyk, M. Adamian, M. A. Sandberg, T. Li, A single, abbreviated RPGR-ORF15 variant reconstitutes RPGR function in vivo. Invest Ophthalmol Vis Sci 46, 435-441 (2005).; 3. W. A. Beltran, A. V. Cideciyan, A. S. Lewin, W. W. Hauswirth, S. G. Jacobson, G. D. Aguirre, Gene augmentation for X-linked retinitis pigmentosa caused by mutations in RPGR. Cold Spring Harbor perspectives in medicine 5, a017392 (2014).; 4. W. T. Deng, F. M. Dyka, A. Dinculescu, J. Li, P. Zhu, V. A. Chiodo, S. L. Boye, T. J. Conlon, K. Erger, T. Cossette, W. W. Hauswirth, Stability and Safety of an AAV Vector for Treating RPGR-ORF15 X-Linked Retinitis Pigmentosa. Hum Gene Ther 26, 593-602 (2015). 5. Z. Wu, S. Hiriyanna, H. Qian, S. Mookherjee, M. M. Campos, C. Gao, R. Fariss, P. A. Sieving, T. Li, P. Colosi, A. Swaroop, A long-term efficacy study of gene replacement therapy for RPGR-associated retinal degeneration. Hum Mol Genet 24, 3956-3970 (2015).; 6. W. A. Beltran, A. V. Cideciyan, A. S. Lewin, S. Iwabe, H. Khanna, A. Sumaroka, V. A. Chiodo, D. S. Fajardo, A. J. Roman, W.-T. Deng, M. Swider, T. S. Aleman, S. L. Boye, S. Genini, A. Swaroop, W. W. Hauswirth, S. G. Jacobson, G. D. Aguirre, in Proc. Natl. Acad. Sci. U.S.A. (2012), vol. 109, pp. 2132-2137), CNGB3 (beta subunit of the cone cyclic nucleotide-gated cation channel) (see, e.g., Kohl et al.(2005) Eur J Hum Genet. 13(3):302), GNAT2 (cone specific alpha subunit of transducin) and CNGA3 (alpha subunit of the cone cyclic nucleotide-gated cation channel) (see, e.g., GenBank Accession No. NP 001289; and Booij et al. (2011) Ophthalmology 118: 160-167).
In some embodiments, the polynucleotide of interest may encode for a neurotrophic factor. As used herein, the "neurotrophic factor" is a generic term of proteins having a physiological action such as survival and maintenance of nerve cells, promotion of neuronal differentiation. In some embodiments, the neurotrophic factor is RdCVF. As used herein the term "RdCVF" has its general meaning in the art and refers to rod-derived cone viability factor (Leveillard T, Mohand-Sa'id S, Lorentz O, Hicks D, Fintz A C, Clerin E et al. Identification and characterization of rod-derived cone viability factor. Nat Genet 2004; 36: 755-759.). The polynucleotide of interest encodes both a long form (RdCVF-L, 217 aa, Q8VC33) having a putative thiol-oxydoreductase activity (JEFFERY, Trends Biochem. Sci., vol.24(l):8-l 1, 1999; JEFFERY, Trends Genet, vol.19(8) :415-417, 2003) and a short form (RdCVF-S, 109 aa, Q91W38) with trophic activity for cones but no redox activity. In some embodiments, the neurotrophic factor is RdCVF2, which shares many similarities with RdCVF in terms of gene structure, expression in a rod-dependent manner and protein 3D structure (see e.g. WO2008148860 and Chalmel F, Leveillard T, Jaillard C, Lardenois A, Berdugo N, Morel E, Koehl P, Lambrou G, Holmgren A, Sahel JA, Poch O. Rod-derived Cone Viability Factor-2 is a novel bifunctional-thioredoxin-like protein with therapeutic potential. BMC Mol Biol. 2007 Aug 31;8:74.). Like RdCVF, the RdCVF2 short isoform exhibits cone rescue activity that is independent of its putative thiol-oxydoreductase activity. In some embodiments, the polynucleotide of interest encodes for RdCVFL2.
In some embodiments, the polynucleotide product of interest is an opsin. The opsin sequence can be derived from any suitable single- or multicellular- organism, including human, algae and bacteria. In some embodiments, the opsin is rhodopsin, photopsin, L/M wavelength (red/green) cone-opsin, or short wavelength (S) cone-opsin (blue). In some embodiments, the opsin is channelrhodopsin or halorhodopsin or cruxhalorhodopsin. In some embodiments, the opsin is a light-responsive opsin as described in U.S. Patent Publication No. 2007/0261127 (e.g., ChR2; Chop2); U.S. Patent Publication No. 2001/0086421; U.S. Patent Publication No. 2010/0015095; and Diester et al. (2011) Nat. Neurosci. 14:387. Other examples of opsins include NpHR, eNpHR 1.0, eNpHR 2.0, eNpHR 3.0, SwiChR, SwiChR 2.0, SwiChR 3.0, Mac, Mac 3.0, Arch, ArchT, Arch 3.0, ArchT 3.0, iChR, ChR2, C1V1-T, C1V1-TT, Chronos, Chrimson, ChrimsonR, CatCh, VChRl-SFO, ChR2-SFO, ChR2-SSFO, ChEF, ChlEF, Jaws, ChloC, Slow ChloC, iClC2, iClC2 2.0, and iClC2 3.0. In some embodiments, the opsin consists of the amino acid sequence as set forth in SEQ ID NO: 3 or SEQ ID NO:4, or SEQ ID NO:5.
In some embodiments, the polynucleotide product of interest is a site-specific endonuclease that provides for site-specific knock-down of gene function, e.g., where the endonuclease knocks out an allele associated with a retinal disease. For example, where a dominant allele encodes a defective copy of a gene that, when wild-type, is a retinal structural protein and/or provides for normal retinal function, a site-specific endonuclease can be targeted to the defective allele and knock out the defective allele. In some embodiments, the vector thus comprises a polynucleotide that encodes a site-specific endonuclease; and a polynucleotide that encodes a functional copy of a defective allele, where the functional copy encodes a functional retinal protein. Site-specific endonucleases that are suitable for use include, e.g., zinc finger nucleases (ZFNs); transcription activator-like effector nucleases (TALENs), and CRISPR-associated endonuclease. As used herein, the term "CRISPR- associated endonuclease" has its general meaning in the art and refers to clustered regularly interspaced short palindromic repeats associated which are the segments of prokaryotic DNA containing short repetitions of base sequences. In particular, the CRISPR-associated endonuclease is Cas9 or a derivative thereof. The Cas9 nuclease can have a nucleotide sequence identical to the wild type Streptococcus pyrogenes sequence. Alternatively, the wild type Streptococcus pyrogenes Cas9 sequence can be modified. For instance, the Cas9 nuclease sequence can be for example, the sequence contained within a commercially available vector such as PX330 or PX260 from Addgene (Cambridge, MA). In some embodiments, the Cas9 endonuclease can have an amino acid sequence that is a variant or a fragment of any of the Cas9 endonuclease sequences of Genbank accession numbers KM099231.1 GL669193757; KM099232.1; GL669193761; or KM099233.1 GL669193765 or Cas9 amino acid sequence of PX330 or PX260 (Addgene, Cambridge, MA). The Cas9 nucleotide sequence can be modified to encode biologically active variants of Cas9, and these variants can have or can include, for example, an amino acid sequence that differs from a wild type Cas9 by virtue of containing one or more mutations (e.g., an addition, deletion, or substitution mutation or a combination of such mutations). For example the Cas9 nuclease can be mutated in the conserved FiNH and RuvC domains, which are involved in strand specific cleavage. For example, an aspartate-to-alanine (D10A) mutation in the RuvC catalytic domain allows the Cas9 nickase mutant (Cas9n) to nick rather than cleave DNA to yield single - stranded breaks, and the subsequent preferential repair through HDR can potentially decrease the frequency of unwanted indel mutations from off-target double-stranded breaks. In some embodiments, the vector comprises one or more guide RNA. As used herein, the term "one or more guide RNA" refers to the RNAs that guide the insertion or deletion of residues. In the context of the invention, the guide RNA is used for recruiting Cas9 to specific genomic loci. In some embodiments, the guide RNA can be a sequence complementary to a coding or a non-coding sequence. In some embodiments, the subject is administered with a combination of at least one vectors comprising one polynucleotide encoding for a Cas9 endonuclease and at least one vector comprising the guide RNA.
In some embodiments, the polynucleotide product is an interfering RNA (RNAi), in particular a siRNA. A "small interfering" or "short interfering RNA" or siRNA is a RNA duplex of nucleotides that is targeted to a gene interest (a "target gene"). An "RNA duplex" refers to the structure formed by the complementary pairing between two regions of a RNA molecule. siRNA is "targeted" to a gene in that the nucleotide sequence of the duplex portion of the siRNA is complementary to a nucleotide sequence of the targeted gene. In some embodiments, the length of the duplex of siRNAs is less than 30 nucleotides.
In some embodiments, the polynucleotide product is an antisense oligonucleotide. As used herein, the term "antisense oligonucleotide" is understood to refer to a nucleotide sequence which is substantially complementary to a target nucleotide sequence in a pre- mRNA molecule, hnRNA (heterogenous nuclear RNA) or mRNA molecule. The degree of complementarity (or substantial complementarity) of the antisense sequence is preferably such that a molecule comprising the antisense sequence can form a stable hybrid with the target nucleotide sequence in the RNA molecule under physiological conditions.
As used herein the term "AAV" refers to more than 30 naturally occurring and available adeno-associated viruses, as well as artificial AAVs. Typically the AAV capsid, ITRs, and other selected AAV components described herein, may be readily selected from among any AAV, including, without limitation, AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV6.2, AAV7, AAV8, AAV9, rhlO, AAVrh64Rl, AAVrh64R2, rh8, variants of any of the known or mentioned AAVs or AAVs yet to be discovered or variants or mixtures thereof. See, e.g., WO 2005/033321. The genomic and proteic sequences of various serotypes of AAV, as well as the sequences of the native terminal repeats (TRs), Rep proteins, and capsid subunits including VPl protein are known in the art. Such sequences may be found in the literature or in public databases such as GenBank or Protein Data Bank (PDB). See, e.g., GenBank and PDB Accession Numbers NC_002077 and 3NG9 (AAV-1), AF043303 and 1LP3 (AAV-2), NC_001729 (AAV-3), U89790 and 2G8G (AAV- 4), NC_006152 and 3NTT (AAV-5), 30AH (AAV6), AF513851 (AAV-7), NC_006261 and 2QA0 (AAV-8), AY530579 and 3UX1 (AAV-9 (isolate hu.14)); the disclosures of which are incorporated by reference herein for teaching AAV nucleic acid and amino acid sequences. See also, e.g., Srivistava et al. (1983) J. Virology 45:555; Chiorini et al. (1998) J. Virology 71 :6823; Chiorini et al. (1999) J. Virology 73: 1309; Bantel-Schaal et al. (1999) J. Virology 73:939; Xiao et al. (1999) J. Virology 73:3994; Muramatsu et al. (1996) Virology 221 :208; Shade et al.,(1986) J. Virol. 58:921; Gao et al. (2002) Proc. Nat. Acad. Sci. USA 99: 11854; Moris et al. (2004) Virology 33:375-383; international patent publications WO 00/28061, WO 99/61601, WO 98/11244; and U.S. Pat. No. 6,156,303 and US7906111.
As used herein the term "recombinant AAV2-derived vector" refers to an AAV2 -based vector comprising the VPl capsid protein as set forth in SEQ ID NO: l and the polynucleotide sequence of interest (i.e., a polynucleotide heterologous to AAV). Thus, in the recombinant AAV2-derived vector of the present invention the native VPl capsid protein of AAV2 is substituted by the VPl capsid protein as set forth in SEQ ID NO: l . The recombinant AAV2- derived vector of the present invention typically comprises 5' and 3' adeno-associated virus inverted terminal repeats (ITRs), the polynucleotide of interest (i.e a heterologous polynucleotide) operatively linked to the promoter PR1.7. The vectors of the invention are produced using methods known in the art. In short, the methods generally involve (a) the introduction of the DNA necessary for AAV replication and synthesis of the capsid, (b) the introduction of a helper construct into the producer cell line, wherein the helper construct comprises the viral functions missing from the AAV vector (c) introducing a helper virus into the producer cell line, (d) the plasmid construct containing the genome of the AAV vector, e.g. ITRs, promoter and transgene sequences, etc.... All functions for AAV virion replication and packaging need to be present, to achieve replication and packaging of the AAV vector into AAV virions. The introduction into the producer host cell can be carried out using standard virology techniques simultaneously or sequentially. Finally, the host cells are cultured to produce AAV virions and are purified using standard techniques such as iodixanol or CsCl gradients or other purification methods. The purified AAV virion is then ready for use in the methods. As used herein, the term "promoter" has its general meaning in the art and refers to a nucleic acid fragment that controls the transcription of one or more genes, located upstream with respect to the direction of transcription of the transcription initiation site of the gene, and is structurally identified by the presence of a binding site for DNA-dependent R A polymerase, transcription initiation sites and any other DNA sequences, including, but not limited to transcription factor binding sites, repressor and activator protein binding sites, and any other sequences of nucleotides known to one of skill in the art to act directly or indirectly to regulate the amount of transcription from the promoter. As used herein, the term "PR1.7 promoter" refers to the 1.7-kb L-opsin promoter described in Hum Gene Ther. 2016 Jan;27(l):72-82 and characterized by the nucleic acid sequence as set forth in SEQ ID NO:2. The promoter and the polynucleotide of interest are operatively linked. As used herein, the term "operably linked" refers to two or more nucleic acid or amino acid sequence elements that are physically linked in such a way that they are in a functional relationship with each other. For instance, a promoter is operably linked to a coding sequence if the promoter is able to initiate or otherwise control/regulate the transcription and/or expression of a coding sequence, in which case the coding sequence should be understood as being "under the control of the promoter. Generally, when two nucleic acid sequences are operably linked, they will be in the same orientation and usually also in the same reading frame. They will usually also be essentially contiguous, although this may not be required.
By a "therapeutically effective amount" is meant a sufficient amount of the vector to treat the retinal disease at a reasonable benefit/risk ratio. It will be understood that the total daily usage of the vector will be decided by the attending physician within the scope of sound medical judgment. The specific therapeutically effective dose level for any particular patient will depend upon a variety of factors including the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific polypeptide employed; and like factors well known in the medical arts. For example, it is well known within the skill of the art to start doses of the compound at levels lower than those required to achieve the desired therapeutic effect and to gradually increase the dosage until the desired effect is achieved. Thus, the doses of vectors may be adapted depending on the disease condition, the subject (for example, according to his weight, metabolism, etc.), the treatment schedule, etc. A preferred effective dose within the context of this invention is a dose allowing an optimal transduction of the cone photoreceptors. Typically, from 108 to 1010 viral genomes (vg) are administered per dose in mice. Typically, the doses of AAV vectors to be administered in humans may range from 109 to 1012 vg.
The vector of the invention is thus formulated into pharmaceutical compositions. These compositions may comprise, in addition to the vector, a pharmaceutically acceptable excipient, carrier, buffer, stabiliser or other materials well known to those skilled in the art. Such materials should be non-toxic and should not interfere with the efficacy of the active ingredient (i.e. the vector of the invention). The precise nature of the carrier or other material may be determined by the skilled person according to the route of administration, i.e. here intravitreal injection. The pharmaceutical composition is typically in liquid form. Liquid pharmaceutical compositions generally include a liquid carrier such as water, petroleum, animal or vegetable oils, mineral oil or synthetic oil. Physiological saline solution, magnesium chloride, dextrose or other saccharide solution or glycols such as ethylene glycol, propylene glycol or polyethylene glycol may be included. For injection, the active ingredient will be in the form of an aqueous solution, which is pyrogen-free and has suitable pH, isotonicity and stability. Those of relevant skill in the art are well able to prepare suitable solutions using, for example, isotonic vehicles such as Sodium Chloride Injection, Ringer's Injection, Lactated Ringer's Injection. Preservatives, stabilisers, buffers, antioxidants and/or other additives may be included, as required. For delayed release, the vector may be included in a pharmaceutical composition, which is formulated for slow release, such as in microcapsules formed from biocompatible polymers or in liposomal carrier systems according to methods known in the art. Typically, the pharmaceutical composition of the present invention is supplied in a prefilled syringe. A "ready- to-use syringe" or "prefilled syringe" is a syringe which is supplied in a filled state, i.e. the pharmaceutical composition to be administered is already present in the syringe and ready for administration. Prefilled syringes have many benefits compared to separately provided syringe and vial, such as improved convenience, affordability, accuracy, sterility, and safety. The use of prefilled syringes results in greater dose precision, in a reduction of the potential for needle sticks injuries that can occur while drawing medication from vials, in pre- measured dosage reducing dosing errors due to the need to reconstituting and/or drawing medication into a syringe, and in less overfilling of the syringe helping to reduce costs by minimising drug waste. In some embodiments the pH of the liquid pharmaceutical composition of the present invention is in the range of 5.0 to 7.0, 5.1 to 6.9, 5.2 to 6.8, 5.3 to 6.7 or 5.4 to 6.6. The invention will be further illustrated by the following figures and examples. However, these examples and figures should not be interpreted in any way as limiting the scope of the present invention. FIGURES:
Figure 1: Expression in all layers of the mouse retina is obtained with the combination of AAV2-7m8-GFP and the mouse cone arrestin (mCAR) promoter (A), PR2.1 promoter (B) while most cone-specific expression pattern is achieved with PR1.7 promoter (C) -after intravitreal injections.
Figure 2: Cone-specific expression in the retinal degeneration 10 (rdlO) mouse model for Retinitis Pigmentosa. AAV2-7m8-PR1.7-GFP expression pattern on a retinal flatmount (A) and observed specifically in cones with a higher magnification (B) after intravitreal injections.
Figure 3: Reporter gene expression pattern using ubiquitous versus cone-specific promoter in vivo in monkeys after intravitreal injections. Retinal flatmount images showing that AAV2-7m8-CMV leads to pan-retinal GFP expression (A) while it is foveal cone- specific with AAV2-7m8-PR1.7 (B). Close up to the fovea with AAV2-7m8-CMV (C, left) and AAV2-7m8-PR1.7 (C, right).
Figure 4: Foveal cone-specific expression of Jaws-GFP under the control of PR1.7 promoter in combination with AAV2-7m8 capsid in the non-human primate, after intravitreal injections. (A) In vivo eye fundus image showing foveal expression, (B) flatmount image of a half fovea (arrow: foveola), (C) retinal cryosections at the level of the fovea.
Figure 5: Cone-specific expression of GFP under the control of PR1.7 promoter and with AAV2-7m8 capsid in human tissue. (A) Retinal organoids derived from human induced pluripotent stem cells expres GFP; (B) GFP-expressing cells in the organoids are exclusively cones (cryosections), (C) expression in cones of a human post-mortem retinal explant (cryosections).
EXAMPLE:
Material & Methods:
AAV production
AAV vectors were produced as previously described using the co-transfection method and purified by iodixanol gradient ultracentrifugation (49). AAV vector stocks were titered by quantitative PCR (50) using SYBR Green (Thermo Fischer Scientific). Animals and intraocular injections
Wild-type C57BL6/j or rdlO mice, and cynomolgus macaques were used for this study. For eye injections (n=6 eyes/condition), mice were anesthetized by isofluorane inhalation. Pupils were dilated and a 33 -gauge needle was inserted into the eye to deliver 2 μΐ, of AAV vector solution intravitreally or ΙμΙ^ subretinally. Regarding macaque eye injections, macaques were first selected based on absence neutralizing antibody titers against AAV. Primates were anesthetized with 10: 1 mg/kg ketamine/xylazine. After subretinal or intravitreal vector administration, opthtalmic steroid and antibiotic ointment were applied to the corneas post injections.
In vivo macaque eye imaging
After pupil dilation, a Spectralis HRA+OCT system (Heidelberg Engineering, Germany) was used to acquire OCT images, and fluorescent images of GFP using the "Fundus Autofluoresence mode" which consists of and excitation wavelength of 488 nm and barrier filter of 500 nm.
Two-photon imaging and ex-vivo electrophysiological recordings of macaque retinas
A two-photon microscope equipped with a 40x water immersion objective (LUMPLFLN40x/W/0.80, Olympus) with a pulsed femto-second laser (InSight™ DeepSee™ - Newport Corporation) was used for imaging GFP-positive retinal cells from whole-mount retinas (with photoreceptor-cell-side up) or retina slices (vertical sections). AAV-treated macaque retinas were isolated and later imaged in oxygenized (95% 02, 5% C02) Ames medium (Sigma-Aldrich). For live two-photon imaging, retinas were placed in the recording chamber of the microscope, and z-stacks were acquired using the excitation laser at a wavelength of 930 nm. Images were processed offline using Image J. For whole-cell patch- clamp recordings, an Axon Multiclamp 700B amplifier was used. Electrodes were made from borosilicate glass (BF100- 50-10, Sutter Instruments) and pulled to 6-9 ΜΩ. Pipettes were filled with 115 mM K Gluconate, 10 mM KC1, 1 mM MgC12, 0.5 mM CaC12, 1.5 mM EGTA, 10 mM HEPES, and 4 mM ATP-Na2 (pH 7.2). Cells were clamped at a potential of -40 mV in voltage-clamp configuration, or recorded in current-clamp (current zero) configuration. Retinas were dark-adapted at least half an hour in the recording chamber prior to recordings.
Human iPSC cultures
We have generated retinal organoids from human iPSCs based on a previously published protocol (37). Clone hiPSC-2 was expanded and differentiated on fibroblast feeders from postnatal human foreskins (ATCC CRL 2429) in "proneural medium" as already described (37). Starting from highly confluent adherent iPS cell cultures and in the absence of fibroblast growth factor 2 (FGF2), self-forming retinal organoids can be identified after 2 weeks. At this point the organoids were mechanically isolated and cultured in 3D conditions for up to 43 days. FGF2 was supplemented to the medium in 3 conditions for 7 days after the mechanical isolation of the organoids to promote their growth. The retinal organoids were infected at day 28 of differentiation at a dose of 5xl010 vg/organoid with AAV2-7m8 vectors carrying the GFP gene under the control of pR1.7 promoter. 10 μΜ DAPT (Selleck) was added to the medium for a week from day 28 on to promote cell cycle arrest of the existent cell populations. Fluorescence intensity was observed for the first time 5 days after infection and continued to increase up to day 43.
Human post-mortem retinal explants
Human retinal explants were prepared using a previously described protocol (38). Briefly, eyes were dissected in C02-independent-medium (Thermo Fischer Scientific). The anterior parts were removed, retina was isolated and cut into small pieces. These explants were placed photoreceptor side-up on a Transwell cell culture insert (Corning), and 2mL of Neurobasal medium (Thermo Fischer Scientific) supplemented with B27 (Thermo Fischer Scientific) were added to each well below each explant. The following day, each explant was infected with a single 0,5 drop of AAV-pRl .7-GFP containing 1010 viral particles. Vector- infected explants were incubated for 10-15 days to allow GFP expression, which was checked using an epifiuorescence macroscope.
Histology, immunohistochemistry and microscopy
Mouse eyes were enucleated and immediately fixed in 10% formalin - 4% formaldehyde for 2 hours for cryosections. Macaque retinas were fixed after dissection in 4% formaldehyde for 3 hours. Retinal organoids and human retinal explants were rinsed in PBS at the end of their culture periods and fixed in 4% paraformaldehyde for 10 minutes. For cryosections, mouse and macaque retinas, retinal organoids and human retinal explants were immersed in PBS-30% sucrose overnight at 4°C. Mouse eyecups, human retinal explants, macaque retinas were embedded in OCT medium and frozen in liquid nitrogen, while retinal organoids were embedded in gelatin- sucrose and frozen in dry ice-cold isopentane. 10 μιη- thick vertical sections were cut with a Microm cryostat. After incubation in the blocking buffer, sections were incubated with primary antibodies overnight at 4°C: human Cone Arrestin antibody (gift from Cheryl Craft); M/L opsin antibody (Millipore AB5405), mouse Cone Arrestin antibody (Millipore, AB 15282). After multiple washes of the sections, the secondary antibodies (Alexa Fluor 488, 594 or 647, ThermoFisher) were added, followed by several washes. Retinal flatmounts or cryosections were mounted in Vectashield mounting medium (Vector Laboratories) for fluorescence microscopy. Retinal sections were visualized using an Olympus Upright confocal microscope.
In silico identification of potential regulatory elements and transcriptomic analysis TF binding site analysis was performed on red opsin gene promoter sequence -pR2.1 and pR1.7 sequences- and the cone arrestin 3 genomic region. The TRANSFAC database 8.3 (http://alggen.lsi.upc.es/) was used for TF binding site prediction. Each TF from the predicted list was analyzed using the Knowledge Base for Sensory System (KBASS, http://kbass.institut-vision.org/KBaSS/transcriptomics/index.php) to select those expressed in human retina using the transcriptomic experiment RNG209 (51). A filter was used to retain TFs with a signal intensity value superior to 40 units in the sample prepared from the experiment RNG209 after normalization by Robust Multi-array Average (RMA) as previously described (52). In this experiment, human retinal specimens used as controls were post-mortem specimens collected within 12 hours following death of patients with no past medical history of eye disease or diabetes. Nineteen samples were collected from 19 eyes representing 17 patients. Sex ratio was 12 men / 7 women with a mean age of 61 years (range 25-78 years).
Statistics
Data were analyzed using a one-way Anova test in Graphpad Prism (multiple comparison, Tukey correction). Error bars on the graphs show the Standard Error of the Mean (SEM). p values are expressed as the following *p<0,033, ns: non significant.
Study approval
For animals, the experiments were realized in accordance with the National Institutes of Health Guide for Care and Use of Laboratory Animals. The protocols were approved by the Local Animal Ethics Committees and conducted in accordance with Directive 2010/63/EU of the European Parliament. Postmortem human ocular globes from donors were acquired from the School of Surgery (Ecole de Chirugie, Assitance Publique Hopitaux de Paris). The protocol was approved by the institutional review boards of the School of Surgery and the Quinze-Vingts National Ophtalmology Hospital. All experiments on postmortem human retinal explants were performed according to the local regulations as well as the guidelines of the Declaration of Helsinki.
Results:
Selection of a strong and specific cone-cell specific promoter in murine models In order to find vector-promoter combinations suitable for strong and specific cone targeting away from the injection site, we compared several AAVs after intravitreal and subretinal delivery in mouse retinas. To enable efficient cone photoreceptor targeting, we used an engineered AAV variant called AAV2-7m8, which has been shown to target photoreceptors efficiently via both administration routes (16, 17). Specific targeting of cone cells has never been attempted using vitreally administered AAV. In order to find suitable promoter sequences for restricted gene expression in cones applicable in the clinic, we focused on promoters that have previously been validated in either non-human primate (NHP) (18) or human tissue (4). We generated AAV2-7m8 vectors encoding GFP under the control of mCAR (mouse Cone Arrestin), PR2.1 and PR1.7 promoters (synthetic promoters based on the human red opsin gene enhancer and promoter sequences) and injected them at equal titers into eyes of six weeks old wild-type mice. Three weeks after subretinal injections, retinal cross-sections were stained with cone arrestin and GFP expression was examined (data not shown). We found high GFP expression in both rod and cone photoreceptors with mCAR promoter while PR2.1 and PR1.7 lead to strong expression mostly in cones, as reported previously (18, 19). Using the same vectors, we obtained strikingly different expression patterns after intravitreal delivery (Figure 1). mCAR promoter lead to GFP expression in some cones, but was leaky towards rods as well as cells of the inner nuclear layer (INL) and ganglion cell layer (GCL) (Figure 1). Both PR2.1 and PR 1.7 promoters lead to more cone labeling than mCAR promoter (Figure 1). PR2.1 transduced more cones than PR1.7 but, it also produced non-specific GFP expression in the INL and GCL. Only the PR1.7 promoter showed GFP expression in cones with minimal expression in rods and was not leaky towards the inner retina (data not shown). Finally, as retinal disease state can influence AAV-mediated gene delivery and transgene expression patterns (20, 21) we validated AAV2-7m8-PR1.7 vector-promoter combination in a mouse model of retinal degeneration. We injected AAV2- 7m8-PR1.7-GFP intravitreally in the rdlO mouse model of retinitis pigmentosa. Two months after injection, GFP expression was restricted to cones (Figure 2) supporting the suitability of this vector for cone-directed gene therapy via both intravitreal and subretinal injections.
Bioinformatic analysis of mCAR, pR1.7 and pR2.1 promoter sequences
Before moving onto further studies in other species, we aimed to better understand the reasons behind the divergent expression patterns obtained with the three promoters. To do so, we analyzed transcription factor (TF) binding sites within each promoter sequence using bioinformatics (data not shown). The present analysis aimed to answer the following questions: (i) why is pR1.7 more efficient than PR2.1 in cones (18)? (ii) why do pR2.1 and mCAR promoters lead to off-target expression after intravitreal administration? We hypothesized that the differential expression patterns observed between PR1.7 and PR2.1 are due to additional TF binding sites found in the 337 bp sequence located in the 5' region of PR2.1 promoter but not in PR1.7 promoter (data not shown). Interestingly, we found a COUP-TFI binding site within this 337 bp sequence (data not shown). COUP-TFI has been shown to suppress green opsin gene (Opnlmw) expression in the mouse retina (22) and might thus be accountable for lower expression with PR2.1 promoter in macaque cones when AAV is delivered subretinally as previously shown (18). Within the same specific 337bp region we also found multiple binding sites for generic, ubiquitous activator TFs (data not shown), such as CEBPB and GTF2I. These additional binding sites of TFs that enhance binding and basal transcriptional machinery assembly and that are not specifically expressed in cones might be responsible for some of the off-target expression observed with PR2.1 compared to PR1.7 (data not shown). We also analyzed TF binding sites in the genomic mCAR promoter sequence to explain the lack of specificity using the short version of this promoter used in the AAV constructs. The short sequence consists of a 521bp portion of the genomic proximal CAR promoter (data not shown) presenting a TATA-box, a TATA-like box, as well as binding sites for CRX (Cone-rod homeobox protein) and SP (Specificity Protein) TFs (23) (data not shown). However, the 'Reg' sequence modulating CAR promoter activity (23) located directly upstream of the 521bp region is excluded from the short sequence (data not shown). Based on the interactome of the TFs binding mCAR promoter obtained from the STRING database (24), CRX and SP TFs interact with each other and with RARA (Retinoic Acid Receptor Alpha), RXRA (Retinoid X Receptor Alpha) and THRB (Thyroid Hormone Receptor Beta) TFs (data not shown). These three nuclear receptors (NR) are involved in cell- type specific regulation of gene expression via MEDl (25) (Mediator Complex Subunit 1) by forming a cell-specific transcription co-activator complex (26, 27) (data not shown). CRX and SP binding sites are located on the 521bp region, while RXRA, RARA and THRB binding sites are positioned on the Reg region (data not shown). Moreover, the Reg region contains five binding sites for THR 2, an important nuclear receptor expressed in cones (28) (data not shown). For all of these reasons, removal of the Reg region is likely responsible for the off- target expression observed with the short mCAR promoter.
Safe gene delivery to macaque foveal cones via intravitreal administration of AAV Others and we have shown transduction of macaque cones using AAV variants with ubiquitous promoters (16, 29-31), but achieving cone transduction by vitreally administered AAV has only been possible at high doses leading to inflammation (16, 29). We reasoned that foveal cone targeting could be achieved if we use a strong cone-specific promoter at lower intravitreally injected AAV doses compatible with safety (29, 32). To test if such "dose sparing" is possible, we injected two macaque eyes with AAV2-7m8-PRl .7-GFP and two other eyes with AAV2-7m8-CMV-GFP at a dose of 1011 viral genome (vg). Using in vivo eye fundus imaging, we observed GFP expression as early as two weeks post-injection with CMV (Figure 3). GFP fluorescence was predominantly in the periphery and in the parafoveal region. GFP expression with PRl .7 became detectable 6 to 8 weeks after administration and was restricted to the fovea (Figure 3). There was no detectable damage to the fovea as assessed by Optical Coherence Tomography (OCT) (data not shown). After sacrifice, flatmounts of the maculas were prepared (Figure 3) and retinal cryosections at the level of the fovea (data not shown) and imaged GFP fluorescence using confocal microscopy, with equal acquisition settings for each eye. These images corroborated the in vivo findings and show specific and robust cone transduction from the vitreous, at a dose of 1011 particles, using AAV2-7m8-PR1.7. Using a similar dose, it is not possible to transduce cones with CMV.
Therapeutic gene delivery to foveal cones for vision restoration using ontogenetics
We next aimed to evaluate the possibility of using this promoter for therapeutic gene delivery. There is no existing blind macaque or primate model of retinal degeneration to test functional outcomes after gene replacement (i.e. CNGB3 for treatment of achromatopsia). However, it is possible to evaluate vision restoration in wild-type macaques using optogenetic strategies since we can distinguish between optogenetic-mediated light responses versus endogenous cone opsin-mediated responses (4). We evaluated the potential of optogenetic vision restoration by expression of Jaws, a hyperpolarizing microbial opsin (15), in foveal cones. We injected one macaque with 1011 vg of AAV2-7m8-PR1.7-Jaws-GFP in the vitreous to evaluate its therapeutic potential for reactivation of dormant cones in mid-stage retinitis pigmentosa as described previously in mice (4,15). We found high-level Jaws-GFP expression restricted to the foveal cones in the injected eyes (Figure 4) similar to GFP expression alone (Figure 3). The animal was then sacrificed two months post injection and half of the retina was processed for histology. Retinal flat-mounts showed typical anatomy of cones in the foveola, the region of the fovea that contains densely packed cones responsible our high acuity vision. Immunostaining for cone arrestin was used to quantify transduced cones (data not shown). About 50 percent of the cone arrestin positive cells were found to express detectable levels of Jaws-GFP in this foveola.
The other half of the retina was conserved as explants (33) for characterization of optogenetic light responses arising from the hyperpolarizing pump Jaws (data not shown). Electrophysiological recordings were performed on transduced cones expressing Jaws and in control cones without GFP expression (data not shown). Whole-cell patch-clamp recordings in GFP positive Jaws-cones exhibited robust light responses to orange light flashes (n=4) (data not shown), while control cones never responded to the same light stimuli (data not shown). Action spectrum of recorded cells showed that highest light responses were obtained using orange light between 575 and 600nm (data not shown) as previously shown for Jaws (15). Cones recorded in current-clamp configuration displayed light-elicited hyperpolarizations followed by short depolarizations (data not shown).
Finally, we injected intravitreally another macaque eye with 1010 particles of AAV2- 7m8-pR1.7-Jaws-GFP to evaluate feasibility of foveal transduction at even lower doses. We obtained detectable foveal Jaws expression even with this lower dose (data not shown), although expression levels were lower. Altogether, all three macaque eyes injected with AAV2-7m8-pPvl .7-GFP (n=2) or Jaws-GFP (n=2) show reproducibility and strength of intravitreal approach compatible with optogenetic reactivation of cones.
PRl.7 promoter drives strong and highly specific gene expression in human cones
Altogether our data in non-human primates show for the first time non-invasive, specific and high-level primate foveal cone transduction compatible with optogenetic applications for vision restoration. However as promoter activity shows important variations across species (29, 35, 36) we deemed it necessary to validate PRl .7 in human cells and tissues. Due to the lack of a good human photoreceptor cell line or other model that could be used to test efficiency of cone promoter activity, we used 3D retinal organoids derived from human induced Pluripotent Stem (iPS) cells (37). We generated photoreceptor-enriched retinal organoids and infected them with AAV2-7m8 vectors encoding GFP under the control of PRl .7 promoter (Figure 5). GFP expression was observed as early as 5 days post infection and continued to increase until the experiment was terminated for analysis on day 43. GFP expression in these organoids colocalized with human Cone Arrestin (data not shown). Lastly, as human retinal organoids do not represent all features of mature human retina, we validated the efficacy and specificity of PRl .7 promoter in post-mortem human retinal explants. Human retinal explants were cultured as described previously (38) and infected with a single drop of AAV2-7m8-pR1.7-GFP (Figure 5). Ten days after infection, GFP expression was analyzed on cryosections. The expression was restricted to the ONL (data not shown) and co-localized with M/L-opsin, a cone cell marker (data not shown). These data collectively point towards high efficiency and specificity of PRl .7 in leading to restricted gene expression in human cones. Discussion:
The fovea accounts for less than 1% of the retinal surface area in primates yet it provides the input to about 50% of the cells in the primary visual cortex (1). The high concentration of cones in the fovea, the thinnest and most delicate part of the retina allows for high acuity vision and it is of utmost importance to preserve the unique functions (39) and architecture (40) of the cones in this area during therapeutic interventions. Foveal cones can be targeted via different administration routes, using either subretinal or intravitreal injections (35, 41, 42) but detaching the fovea might lead to mechanical damage, especially in the degenerating retina (43). For all of these reasons, ways to deliver therapeutics to the fovea, without detaching this region are needed. Intravitreal injections are a surgically simple way to deliver therapeutics without retinal detachment. Gene therapy vectors can target the outer retina via intravitreal injections in rodents without damage to the photoreceptors (17, 35). However, safe and efficient gene delivery to primate cones via intravitreal injection had not been achieved so far, likely due to the substantial dilution of the vector in the vitreous and resulting loss of efficacy. The use of cell-type specific promoters that provide high-level gene expression with a lower local concentration is critical to overcome this challenge (29, 44).
In this study, we sought to first achieve strong and exclusive transduction of cones via non-invasive, intravitreal injection using various promoters in combination with AAV2-7m8 capsid. We selected three previously described promoters in view of their utility in driving gene expression in primate cones (4, 18, 19, 45, 46) and tested them for specificity and strength of cone transduction side-by-side. All promoters tested in vivo in mouse retinas lead to transgene expression in the photoreceptor layer when delivered subretinally. mCAR promoter led to expression in rods and cones. Surprisingly after intravitreal delivery; only PR1.7 maintained its specificity towards cones, while PR2.1 and mCAR gave rise to non- specific gene expression in inner retinal neurons. mCAR and PR2.1 gave rise to non-specific expression in inner retinal cells, making them unsuitable for optogenetic applications where any expression in downstream neurons would cancel out the response from the photoreceptors. Subsequent in silico analysis of TF binding sites within each promoter sequence proposed basis for more specific transduction with PR1.7 and the observed the lack of specificity with mCAR promoter. Next, to study the ability of AAV2-7m8 equipped with PR1.7 promoter to transduce foveal cones, we conducted gene delivery studies in macaque eyes. Complete restriction of gene expression to primate cones was achieved using AAV2- 7m8-PR1.7 in the fovea via intravitreal administration. One shortcoming with intravitreal injection route is the higher susceptibility of AAVs administered into this compartment to interactions with the immune system compared to subretinal administration (32). It has been shown that antibody neutralization poses a barrier to intravitreal AAV vector mediated gene delivery in non-human primates and this will likely pose a challenge for human application. We thus aimed to develop another gene delivery approach for patients who have neutralizing antibodies towards AAV2. To this aim we tested gene delivery to foveal cones by subretinal administration of AAV9-7m8 at a distal site. We demonstrated that robust light responses could be obtained with this new delivery approach, thanks to the vector's ability to diffuse laterally and mediate expression outside of the bleb. Using the same optogenetic cone reactivation strategy, we showed that this approach also affords robust light responses mediated by Jaws but in a higher percentage of cones compared to intravitreal route (4, 15).
Our in vivo findings collectively point to three important considerations in retinal gene delivery. First, enhanced AAV vectors, whether obtained via directed evolution (AAV2-7m8 (35)) or rational design (AAV9-7m8 (17)), can achieve therapeutic objectives where parental serotypes fail to provide sufficient gene delivery. Indeed AAV2 and AAV9 cannot perform efficient non-invasive foveal targeting (30, 31) while 7m8 modified vectors bridge this gap. Second, strong cell-type specific promoters allow dose sparing important for the safety of gene therapy (i.e avoiding immune response). Third, our study shows the non-negligible impact of vector administration route on transgene expression patterns. Finally, to complement our in vivo results in animals, we performed a battery of ex vivo tests in human tissues; that in combination with in vivo experiments constitute a versatile platform for validating gene therapy for clinical application. The vector-promoter combinations described here will find utility in all retinal diseases where cone targeting is desired. Each administration route and vector can be considered based on the serological state of the patient and natural history of the targeted disease (see Table 1). The combination of PR1.7 and AAV2-7m8 is ideal for therapeutic gene expression in human foveal cones when delivered into the vitreous and can be an ideal way to reanimate remaining dormant cones with optogenetics in retinitis pigmentosa (4). Since cones subsist in both the center and the periphery in achromatopsia, gene delivery in the periphery, using AAV9-7m8-PR1.7 can be more efficacious, as it would deliver the therapeutic gene into both the foveal and peripheral cones.
Table 1: AAV vector administration strategies for cone-directed gene therapy. Injection Peripheral SR Central SR Peripheral SR (near Intravitreal route macula)
Therapeutic Peripheral Central (macula- Peripheral and central Central
gene fovea)
expression
Potential AAV2-3YF(18, 47) AAV2 in clinical AAV9-7m8 as used in AAV2-7m8 as capsids AAV9(18, 30, 47) trials(8, 9, 48) this study used in this study
Advantages Immune privilege Immune privilege Immune privilege Non invasive
High level High level therapeutic High level therapeutic Potential high therapeutic gene gene expression gene expression acuity vision expression Foveal transduction Larger expression area Controlled area of
High acuity vision that includes the fovea expression pattern
Not invasive to the
fovea
Disadvantages Invasive No Invasive, risk of Presence of NAbs foveal adverse effects such in the vitreous (use transduction as macular of
Low acuity vision thinning(l 1) glucocorticoids^ 9) could prevent anti- vector immune response if patient is seropositive for AAV2)
Lower gene expression than SR
Potential Retinitis Pigmentosa: optogenetic vision restoration (Jaws) (4, 15)
target Achromatopsia: CNGA3 or CNGB3(47)
diseases and
applications
SEQUENCES:
SEQ ID NO: 1: VP1 capsid of the recombinant AAV2-derived vector
MAADGYLPDWLEDTLSEGIRQWWKLKPGPPPPKPAERHKDDSRGLVLPGYK YLGPFNGLDKGEPVNEADAAALEHDKAYDRQLDSGDNPYLKYNHADAEFQERLKE DTSFGGNLGRAVFQAKKRVLEPLGLVEEPVKTAPGK RPVEHSPVEPDSSSGTGKAG QQPARKRLNFGQTGDADSVPDPQPLGQPPAAPSGLGTNTMATGSGAPMAD NEGAD GVGNSSGNWHCDSTWMGDRVITTSTRTWALPTYNNHLYKQISSQSGASNDNHYFGY STPWGYFDFNRFHCHFSPRDWQRLINNNWGFRPKRLNFKLFNIQVKEVTQNDGTTTI A NLTSTVQVFTDSEYQLPYVLGSAHQGCLPPFPADVFMVPQYGYLTL NGSQAVG RSSFYCLEYFPSQMLRTG NFTFSYTFEDVPFHSSYAHSQSLDRLMNPLIDQYLYYLS RTNTPSGTTTQSRLQFSQAGASDIRDQSRNWLPGPCYRQQRVSKTSAD NNSEYSWT GATKYHLNGRDSLVNPGPAMASHKDDEEKFFPQSGVLIFGKQGSEKTNVDIEKVMIT DEEEIRTTNPVATEQYGSVSTNLQRGLALGETTRPARQAATADVNTQGVLPGMVW QDRDVYLQGPIWAKIPHTDGHFHPSPLMGGFGLKHPPPQILIK TPVPANPSTTFS AA KFASFITQYSTGQVSVEIEWELQKENSKRWNPEIQYTSNYNKSVNVDFTVDTNGVYS EPRPIGTRYLTR SEQ ID NO:2: PR1.7 promoter
ggaggctgaggggtggggaaagggcatgggtgtttcatgaggacagagcttccgtttcatgcaatgaaaagagtttggaga cggatggtggtgactggactatacacttacacacggtagcgatggtacactttgtattatgtatattttaccacgatctttttaaagtgtcaaa ggcaaatggccaaatggttccttgtcctatagctgtagcagccatcggctgttagtgacaaagcccctgagtcaagatgacagcagccc ccataactcctaatcggctctcccgcgtggagtcatttaggagtagtcgcattagagacaagtccaacatctaatcttccaccctggccag ggccccagctggcagcgagggtgggagactccgggcagagcagagggcgctgacattggggcccggcctggcttgggtccctct ggcctttccccaggggccctctttccttggggctttcttgggccgccactgctcccgctcctctccccccatcccaccccctcaccccctc gttcttcatatccttctctagtgctccctccactttcatccacccttctgcaagagtgtgggaccacaaatgagttttcacctggcctgggga cacacgtgcccccacaggtgctgagtgactttctaggacagtaatctgctttaggctaaaatgggacttgatcttctgttagccctaatcat caattagcagagccggtgaaggtgcagaacctaccgcctttccaggcctcctcccacctctgccacctccactctccttcctgggatgtg ggggctggcacacgtgtggcccagggcattggtgggattgcactgagctgggtcattagcgtaatcctggacaagggcagacaggg cgagcggagggccagctccggggctcaggcaaggctgggggcttcccccagacaccccactcctcctctgctggacccccacttca tagggcacttcgtgttctcaaagggcttccaaatagcatggtggccttggatgcccagggaagcctcagagttgcttatctccctctagac agaaggggaatctcggtcaagagggagaggtcgccctgttcaaggccacccagccagctcatggcggtaatgggacaaggctggc cagccatcccaccctcagaagggacccggtggggcaggtgatctcagaggaggctcacttctgggtctcacattcttggatccggttc caggcctcggccctaaatagtctccctgggctttcaagagaaccacatgagaaaggaggattcgggctctgagcagtttcaccaccca ccccccagtctgcaaatcctgacccgtgggtccacctgccccaaaggcggacgcaggacagtagaagggaacagagaacacataa acacagagagggccacagcggctcccacagtcaccgccaccttcctggcggggatgggtggggcgtctgagtttggttcccagcaa atccctctgagccgcccttgcgggctcgcctcaggagcaggggagcaagaggtgggaggaggaggtctaagtcccaggcccaatta agagatcaggtagtgtagggtttgggagcttttaaggtgaagaggcccgggctgatcccacaggccagtataaagcgccgtgaccctc aggtgatgcgccagggccggctgccgtcggggacagggctttccatagcc
SEQ ID NO:3 : Jaws-GFP= Halo57 +2 mutations + KGC + GFP + ER2
MTAVSTTATTVLQATQSDVLQEIQSNFLLNSSIWVNIALAGVVILLFVAMGRD LESPRAKLIWVATMLVPLVSISSYAGLASGLTVGFLQMPPGHALAGQEVLSPWGRYL TWTFSTPMILLALGLLADTDIASLFTAITMDIGMCVTGLAAALITSSHLLRWVFYGISC AFFVAVLYVLLVQWPADAEAAGTSEIFGTLRILTVVLWLGYPILFALGSEGVALLSVG VTSWGYSGLDILAKYVFAFLLLRWVAANEGTVSGSGMGIGSGGAAPADDRPVVKSR ITSEGEYIPLDQIDINVAPAGAVSKGEELFTGVVPILVELDGDVNGHKFSVSGEGEGDA TYGKLTLKFICTTGKLPVPWPTLVTTLTYGVQCFSRYPDHMKQHDFFKSAMPEGYVQ ERTIFFKDDGNYKTRAEVKFEGDTLVNRIELKGIDFKEDGNILGHKLEYNYNSHNVYI MADKQK GIKVNFKIRHNIEDGSVQLADHYQQNTPIGDGPVLLPDNHYLSTQSALSK DPNEKRDHMVLLEFVTAAGITLGMDELYKVAFCYENEV SEQ ID NO: 4: Jaws = Halo57 + 2 mutations + KGC + ER2
MTAVSTTATTVLQATQSDVLQEIQSNFLLNSSIWVNIALAGVVILLFVAMGRD LESPRAKLIWVATMLVPLVSISSYAGLASGLTVGFLQMPPGHALAGQEVLSPWGRYL TWTFSTPMILLALGLLADTDIASLFTAITMDIGMCVTGLAAALITSSHLLRWVFYGISC AFFVAVLYVLLVQWPADAEAAGTSEIFGTLRILTVVLWLGYPILFALGSEGVALLSVG VTSWGYSGLDILAKYVFAFLLLRWVAANEGTVSGSGMGIGSGGAAPADDRPVVKSR ITSEGEYIPLDQIDINVAPAGAVAFCYENEV SEQ ID NO: 5: Halo57 + 2 mutations + KGC
MTAVSTTATTVLQATQSDVLQEIQSNFLLNSSIWVNIALAGVVILLFVAMGRD LESPRAKLIWVATMLVPLVSISSYAGLASGLTVGFLQMPPGHALAGQEVLSPWGRYL TWTFSTPMILLALGLLADTDIASLFTAITMDIGMCVTGLAAALITSSHLLRWVFYGISC AFFVAVLYVLLVQWPADAEAAGTSEIFGTLRILTVVLWLGYPILFALGSEGVALLSVG VTS WGYSGLDILAKYVFAFLLLRWVAANEGTVSGSGMGIGSGGAAPADDRPVVKSR ITSEGEYIPLDQIDINV
REFERENCES:
Throughout this application, various references describe the state of the art to which this invention pertains. The disclosures of these references are hereby incorporated by reference into the present disclosure.
1. Wassle H, Grunert U, Rohrenbeck J, Boycott BB. Cortical magnification factor and the ganglion cell density of the primate retina. Nature 1989;341(6243):643-646.
2. Kolb H, Zhang L, Dekorver L, Cuenca N. A new look at calretinin-immunoreactive amacrine cell types in the monkey retina.. J. Comp. Neurol. 2002;453(2): 168-84.
3. Wikler KC, Williams RW, Rakic P. Photoreceptor mosaic: Number and distribution of rods and cones in the rhesus monkey retina. J. Comp. Neurol. 1990;297(4):499-508.
4. Busskamp V et al. Genetic reactivation of cone photoreceptors restores visual responses in retinitis pigmentosa. [Internet]. Science 2010;329(5990):413-7.
5. Byrne LC et al. Viral-mediated RdCVF and RdCVFL expression protects cone and rod photoreceptors in retinal degeneration. [Internet]. J. Clin. Invest. 2015; 125(1): 105-16.
6. Komaromy AM et al. Gene therapy rescues cone function in congenital achromatopsia. Hum. Mol. Genet. 2010;19:2581-2593. 7. Maguire AM et al. Safety and Efficacy of Gene Transfer for Leber's Congenital Amaurosis. N. Engl. J. Med. 2008;358(21):2240-2248.
8. Bainbridge JWB et al. Effect of gene therapy on visual function in Leber's congenital amaurosis.. N. Engl. J. Med. 2008;358(21):2231-9.
9. Cideciyan A V et al. Human gene therapy for RPE65 isomerase deficiency activates the retinoid cycle of vision but with slow rod kinetics.. Proc. Natl. Acad. Sci. U. S. A. 2008;105(39): 15112-7.
10. Bainbridge JWB et al. Long-Term Effect of Gene Therapy on Leber's Congenital Amaurosis. N. Engl. J. Med. 2015;150504083137004.
11. Jacobson SG et al. Improvement and Decline in Vision with Gene Therapy in
Childhood Blindness. N. Engl. J. Med. 2015;150503141523009.
12. Jacobson SG et al. Gene therapy for leber congenital amaurosis caused by RPE65 mutations: safety and efficacy in 15 children and adults followed up to 3 years.. Arch. Ophthalmol. 2012;130(l):9-24.
13. MacLaren RE et al. Retinal gene therapy in patients with choroideremia: Initial fi ndings from a phase 1/2 clinical trial. Lancet 2014;383(9923): 1129-1137.
14. Duncan JL. Visual Consequences of Delivering Therapies to the Subretinal Space. JAMA Ophthalmol. 2017;135(3):242-243. doi: 10.100 l/jamaophthalmol.2016.5659.
15. Chuong AS et al. Noninvasive optical inhibition with a red-shifted microbial rhodopsin [Internet]. Nat N euro sci 2014;17(8): 1123-1129.
16. Dalkara D et al. In Vivo-Directed Evolution of a New Adeno-Associated Virus for Therapeutic Outer Retinal Gene Delivery from the Vitreous. Sci. Transl. Med. 2013;5(189): 189ra76-189ra76.
17. Khabou H et al. Insight into the mechanisms of enhanced retinal transduction by the engineered AAV2 capsid variant -7m8.. Biotechnol. Bioeng. [published online ahead of print: June 2016]; doi:10.1002/bit.26031
18. Ye G-J et al. Cone-Specific Promoters for Gene Therapy of Achromatopsia and Other Retinal Diseases. Hum. Gene Ther. 2016;27(l):72-82.
19. Ye G et al. Safety and Biodistribution Evaluation in CNGB3 -Deficient Mice of rAAV2tYF-PRl . 7-hCNGB3 , a Recombinant AAV Vector for Treatment of
Achromatopsia2016;27(l):27-37.
20. Kolstad KD et al. Changes in adeno-associated virus-mediated gene delivery in retinal degeneration.. Hum. Gene Ther. 2010;21(5):571-578. 21. Vacca O et al. AAV-mediated gene delivery in Dp71-null mouse model with compromised barriers. [Internet]. Glia 2014;62(3):468-76.
22. Satoh S et al. The spatial patterning of mouse cone opsin expression is regulated by bone morphogenetic protein signaling through downstream effector COUP-TF nuclear receptors.. J. Neurosci. 2009;29(40): 12401-11.
23. Pickrell SW, Zhu X, Wang X, Craft CM. Deciphering the contribution of known cis-elements in the mouse cone arrestin gene to its cone-specific expression. Investig. Ophthalmol. Vis. Sci. 2004;45(11):3877-3884.
24. von Mering C et al. STRING 7 - Recent developments in the integration and prediction of protein interactions. Nucleic Acids Res. 2007;35(SUPPL. 1):358— 362.
25. Blazek E, Mittler G, Meisterernst M. The mediator of RNA polymerase II. Chwmosoma 2005;113(8):399-408.
26. Regulators LT. Letter to the Editor A Unified Nomenclature for Protein Subunits of Mediator Complexes. Mol. Cell 2004;14:553-557.
27. Boyd JM, Verma S, Uhlmann E. 14584 Corrections. Annu. Rev. Microbiol.
1998;95:2-3.
28. Viets K, Eldred KC, Jr RJJ. Mechanisms of Photoreceptor Patterning in Vertebrates and Invertebrates. Trends Genet. 2016;32(10):638-659.
29. Ramachandran PS et al. Evaluation of Dose and Safety of AAV7m8 and AAV8BP2 in the Non-Human Primate Retina.. Hum. Gene Ther. [published online ahead of print: October 2016]; doi: 10.1089/hum.2016.111
30. Vandenberghe LH et al. AAV9 targets cone photoreceptors in the nonhuman primate retina.. PLoS One 2013;8(l):e53463.
31. Vandenberghe LH et al. Dosage thresholds for AAV2 and AAV8 photoreceptor gene therapy in monkey.. Sci. Transl. Med. 201 l;3(88):88ra54.
32. Kotterman M a et al. Antibody neutralization poses a barrier to intravitreal adeno- associated viral vector gene delivery to non-human primates.. Gene Ther. 2014;(August): l- 11.
33. Sengupta A et al. Red-shifted channelrhodopsin stimulation restores light responses in blind mice , macaque retina , and human retina. EMBO Mol Med 2016; 1-17.
34. Bell CL, Gurda BL, Van Vliet K, Agbandje-McKenna M, Wilson JM. Identification of the Galactose Binding Domain of the Adeno-Associated Virus Serotype 9 Capsid. J. Virol. 2012;86(13):7326-7333. 35. Dalkara D et al. In vivo-directed evolution of a new adeno-associated virus for therapeutic outer retinal gene delivery from the vitreous. [Internet]. Sci. Transl. Med. 2013;5: 189ra76.
36. Yin L et al. Intravitreal injection of AAV2 transduces macaque inner retina.. Invest. Ophthalmol. Vis. Sci. 2011;52(5):2775-83.
37. Reichman S, Terray A, Slembrouck A, Nanteau C, Orieux G. From confluent human iPS cells to self-forming neural retina and retinal pigmented epithelium[published online ahead of print: 2014]; doi: 10.1073/pnas.1324212111
38. Fradot M, Busskamp V, Bennett J. Gene Therapy in Ophthalmology : Validation on Cultured201 l;593(May):587-593.
39. Sinha R et al. Cellular and Circuit Mechanisms Shaping the Perceptual Properties of the Primate Fovea. Cell 2017;168(3):413-426.el2.
40. Anderson DH, Fisher SK. The relationship of primate foveal cones to the pigment epithelium.. J. Ultrastruct. Res. 1979;67(l):23-32.
41. Allocca M et al. Novel adeno-associated virus serotypes efficiently transduce murine photoreceptors.. J. Virol. 2007;81(20):11372-80.
42. Kay CN et al. Targeting photoreceptors via intravitreal delivery using novel, capsid-mutated AAV vectors.. PLoS One 2013;8(4):e62097.
43. Jacobson SG et al. Gene Therapy for Leber Congenital Amaurosis caused by RPE65 mutations: Safety and Efficacy in Fifteen Children and Adults Followed up to Three
Years. Arch. Ophthalmol. 2012;130(l):9-24.
44. Tenenbaum L, Lehtonen E, Monahan PE. Evaluation of risks related to the use of adeno-associated virus-based vectors.. Curr. Gene Ther. 2003;3(6):545-65.
45. Boyd RF et al. Photoreceptor-targeted gene delivery using intravitreally administered AAV vectors in dogs. Gene Ther. 2016;23(2):223-230.
46. Ye G-J et al. Safety and Biodistribution Evaluation in Cynomolgus Macaques of rAAV2tYF-PR1.7-hCNGB3, a Recombinant AAV Vector for Treatment of Achromatopsia. Hum. Gene Ther. Clin. Dev. 2016;27(l):hum.2015.164.
47. Maguire AM et al. Safety and efficacy of gene transfer for Leber's congenital amaurosis.. N. Engl. J. Med. 2008;358(21):2240-8.
48. Gernoux G, Wilson JM, Mueller C. Regulatory and Exhausted T Cell Responses to AAV Capsid. Hum. Gene Ther. 2017;28(4):338-349.
49. Choi VW, Asokan A, Haberman R a, Samulski RJ. Production of recombinant adeno-associated viral vectors.. Curr. Protoc. Hum. Genet. 2007;Chapter 12:Unit 12.9. 50. Aurnhammer C et al. Universal real-time PCR for the detection and quantification of adeno-associated virus serotype 2-derived inverted terminal repeat sequences.. Hum. Gene Ther. Methods 2012;23(l): 18-28.
51. Delyfer M-N et al. Transcriptomic Analysis of Human Retinal Detachment Reveals Both Inflammatory Response and Photoreceptor Death. PLoS One
2011;6(12):e28791.
52. Reichman S et al. The homeobox gene CHX10/VSX2 regulates RdCVF promoter activity in the inner retina.. Hum. Mol. Genet. 2010;19(2):250-61.

Claims

CLAIMS:
1. A method of expressing a polynucleotide of interest in the cone photoreceptors of a subject comprising the intravitreal delivery of a therapeutically effective amount of a recombinant AAV2-derived vector comprising a VP1 capsid protein as set forth in SEQ ID NO: l and the polynucleotide of interest under the control of the PR1.7 promoter as set forth in SEQ ID NO: 2.
2. The method of claim 1 wherein the subject is affected or likely to be affected with a retinal disease affecting cone photoreceptors.
3. The method of claim 1 wherein the subject suffers from a retinal disease selected from the group consisting of age-related macular degeneration, Bassen-kornzweig syndrome, choroideremia, gyrate atrophy, Refsum syndrome, Usher syndrome, color blindness, blue cone monochromacy, achromatopsia, incomplete achromatopsia, oligocone trichromacy, retinitis pigmentosa (RP), macular degeneration, Stargardt's Disease, Bardet-Biedl syndrome, Bornholm eye disease, Best's Disease and Leber's congenital amaurosis.
4. The method of claim 1 wherein the polynucleotide of interest encodes for retinitis pigmentosa GTPase regulator (RPGRORF15), CNGB3 (beta subunit of the cone cyclic nucleotide-gated cation channel), CNGA3 (alpha subunit of the cone cyclic nucleotide-gated cation channel) or GNAT2.
5. The method of claim 1 wherein the polynucleotide of interest encodes for a neurotrophic factor.
6. The method of claim 1 wherein the polynucleotide of interest encodes for RdCVF, RdCVF2, RdCVFL or RdCVFL2.
7. The method of claim 1 wherein the polynucleotide of interest encodes for an opsin such as rhodopsin like halorhodopsin or cruxhalorhodopsin, photopsin, L/M wavelength (red/green) cone-opsin, or short wavelength (S) cone-opsin (blue).
8. The method of claim 7 wherein the polynucleotide of interest encodes for an opsin consisting of an amino acid sequence as set forth in SEQ ID NO:3, SEQ ID NO:4 or SEQ ID NO:5.
9. The method of claim 1 wherein the polynucleotide of interest encodes for a site- specific endonuclease that provides for site-specific knock-down of gene function selected from the group consisting of zinc finger nucleases (ZFNs); transcription activator-like effector nucleases (TALENs), and CRISPR-associated endonucleases.
10. The method of claim 1 wherein the polynucleotide of interest encodes for Cas9 nuclease.
11. The method of claim 1 wherein the polynucleotide of interest encodes for an interfering RNA (RNAi), in particular a siRNA.
12. The method of claim 1 wherein the polynucleotide of interest encode for an antisense oligonucleotide.
13. A recombinant AAV2-derived vector comprising a VP1 capsid protein as set forth in SEQ ID NO: l and a polynucleotide of interest under the control of the PR1.7 promoter as set forth in SEQ ID NO:2.
14. The recombinant AAV2-derived vector of claim 13 for use in a method for the treatment of a retinal disease affecting cone photoreceptors.
PCT/EP2018/050925 2017-01-17 2018-01-16 Methods of expressing a polynucleotide of interest in the cone photoreceptors WO2018134168A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP18701288.5A EP3570895A1 (en) 2017-01-17 2018-01-16 Methods of expressing a polynucleotide of interest in the cone photoreceptors

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
EP17305050 2017-01-17
EP17305050.1 2017-01-17
EP17306429.6 2017-10-20
EP17306429 2017-10-20

Publications (1)

Publication Number Publication Date
WO2018134168A1 true WO2018134168A1 (en) 2018-07-26

Family

ID=61024752

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2018/050925 WO2018134168A1 (en) 2017-01-17 2018-01-16 Methods of expressing a polynucleotide of interest in the cone photoreceptors

Country Status (2)

Country Link
EP (1) EP3570895A1 (en)
WO (1) WO2018134168A1 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019094904A1 (en) * 2017-11-13 2019-05-16 The Regents Of The University Of California Compositions and methods for enhancing visual function
EP3448437A4 (en) * 2016-04-29 2019-11-06 Adverum Biotechnologies, Inc. Evasion of neutralizing antibodies by a recombinant adeno-associated virus
WO2020079464A1 (en) * 2018-10-17 2020-04-23 INSERM (Institut National de la Santé et de la Recherche Médicale) Method for treating retinal degeneration disease by administering nucleolin polynucleotide or polypeptide
EP3892738A1 (en) 2020-04-10 2021-10-13 Sorbonne Université G-protein-gated-k+ channel-mediated enhancements in light sensitivity in rod-cone dystrophy (rcd)
WO2021204407A1 (en) 2020-04-10 2021-10-14 Sorbonne Université G-protein-gated-k+ channel-mediated enhancements in light sensitivity in rod-cone dystrophy (rcd)
EP4083203A1 (en) * 2016-06-16 2022-11-02 Adverum Biotechnologies, Inc. Compositions and methods for reducing ocular neovascularization
WO2022248634A1 (en) 2021-05-26 2022-12-01 Sparingvision G-protein-gated-k+ channel-mediated enhancements in light sensitivity in rod-cone dystrophy (rcd)
EP4163296A1 (en) 2021-10-11 2023-04-12 Sparingvision G-protein-gated-k+ channel-mediated enhancements in light sensitivity in patients with rod-cone dystrophy (rcd)
EP3989982A4 (en) * 2019-06-27 2023-06-28 University Of Florida Research Foundation, Incorporated Enhancing aav-mediated transduction of ocular tissues with hyaluronic acid
EP4357359A1 (en) 2022-10-20 2024-04-24 Sparingvision G-protein-gated-k+ channel-mediated enhancements in light sensitivity in rod-cone dystrophy (rcd)

Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998011244A2 (en) 1996-09-11 1998-03-19 The Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Aav4 vector and uses thereof
WO1999061601A2 (en) 1998-05-28 1999-12-02 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Aav5 vector and uses thereof
WO2000028061A2 (en) 1998-11-05 2000-05-18 The Trustees Of The University Of Pennsylvania Adeno-associated virus serotype 1 nucleic acid sequences, vectors and host cells containing same
US6156303A (en) 1997-06-11 2000-12-05 University Of Washington Adeno-associated virus (AAV) isolates and AAV vectors derived therefrom
WO2005033321A2 (en) 2003-09-30 2005-04-14 The Trustees Of The University Of Pennsylvania Adeno-associated virus (aav) clades, sequences, vectors containing same, and uses therefor
US20070261127A1 (en) 2005-07-22 2007-11-08 Boyden Edward S Light-activated cation channel and uses thereof
WO2008148860A1 (en) 2007-06-05 2008-12-11 Institut National De La Sante Et De La Recherche Medicale (Inserm) New neuronal viability factor and use thereof
US20100015095A1 (en) 2006-05-04 2010-01-21 Wayne State University Restoration of visual responses by in vivo delivery of rhodopsin nucleic acids
US20110086421A1 (en) 2002-04-11 2011-04-14 Max-Planck-Gesel.Zur Foerderung Der Wissen. E.V. Use of biological photoreceptors as directly light-activated ion channels
US20130317091A1 (en) * 2011-01-07 2013-11-28 Guo-jie Ye Promoters, expression cassettes, vectors, kits, and methods for the treatment of achromatopsia and other diseases
WO2015142941A1 (en) * 2014-03-17 2015-09-24 Avalanche Biotechnologies, Inc. Compositions and methods for enhanced gene expression in cone cells
WO2016141078A1 (en) * 2015-03-02 2016-09-09 Avalanche Biotechnologies, Inc. Compositions and methods for intravitreal delivery of polynucleotides to retinal cones
WO2017210664A1 (en) * 2016-06-03 2017-12-07 Massachusetts Institute Of Technology Somatic opsins for single cell resolution optogenetics

Patent Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998011244A2 (en) 1996-09-11 1998-03-19 The Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Aav4 vector and uses thereof
US6156303A (en) 1997-06-11 2000-12-05 University Of Washington Adeno-associated virus (AAV) isolates and AAV vectors derived therefrom
WO1999061601A2 (en) 1998-05-28 1999-12-02 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Aav5 vector and uses thereof
WO2000028061A2 (en) 1998-11-05 2000-05-18 The Trustees Of The University Of Pennsylvania Adeno-associated virus serotype 1 nucleic acid sequences, vectors and host cells containing same
US20110086421A1 (en) 2002-04-11 2011-04-14 Max-Planck-Gesel.Zur Foerderung Der Wissen. E.V. Use of biological photoreceptors as directly light-activated ion channels
US7906111B2 (en) 2003-09-30 2011-03-15 The Trustees Of The University Of Pennsylvania Adeno-associated virus (AAV) clades, sequences, vectors containing same, and uses therefor
WO2005033321A2 (en) 2003-09-30 2005-04-14 The Trustees Of The University Of Pennsylvania Adeno-associated virus (aav) clades, sequences, vectors containing same, and uses therefor
US20070261127A1 (en) 2005-07-22 2007-11-08 Boyden Edward S Light-activated cation channel and uses thereof
US20100015095A1 (en) 2006-05-04 2010-01-21 Wayne State University Restoration of visual responses by in vivo delivery of rhodopsin nucleic acids
WO2008148860A1 (en) 2007-06-05 2008-12-11 Institut National De La Sante Et De La Recherche Medicale (Inserm) New neuronal viability factor and use thereof
US20130317091A1 (en) * 2011-01-07 2013-11-28 Guo-jie Ye Promoters, expression cassettes, vectors, kits, and methods for the treatment of achromatopsia and other diseases
WO2015142941A1 (en) * 2014-03-17 2015-09-24 Avalanche Biotechnologies, Inc. Compositions and methods for enhanced gene expression in cone cells
WO2016141078A1 (en) * 2015-03-02 2016-09-09 Avalanche Biotechnologies, Inc. Compositions and methods for intravitreal delivery of polynucleotides to retinal cones
WO2017210664A1 (en) * 2016-06-03 2017-12-07 Massachusetts Institute Of Technology Somatic opsins for single cell resolution optogenetics

Non-Patent Citations (87)

* Cited by examiner, † Cited by third party
Title
"GENBANK", Database accession no. GL669193757
"GENBANK", Database accession no. GL669193761
"GENBANK", Database accession no. GL669193765
"Genbank", Database accession no. KM099231.1
"GENBANK", Database accession no. KM099232.1
"GENBANK", Database accession no. KM099233.1
"GenBank", Database accession no. NP 001289
ALLOCCA M ET AL.: "Novel adeno-associated virus serotypes efficiently transduce murine photoreceptors.", J. VIROL., vol. 81, no. 20, 2007, pages 11372 - 80, XP002528463, DOI: doi:10.1128/JVI.01327-07
ANDERSON DH; FISHER SK: "The relationship of primate foveal cones to the pigment epithelium.", J. ULTRASTRUCT. RES., vol. 67, no. l, 1979, pages 23 - 32, XP024873258, DOI: doi:10.1016/S0022-5320(79)80014-3
AURNHAMMER C ET AL.: "Universal real-time PCR for the detection and quantification of adeno-associated virus serotype 2-derived inverted terminal repeat sequences.", HUM. GENE THER. METHODS, vol. 23, no. 1, 2012, pages 18 - 28, XP055346603, DOI: doi:10.1089/hgtb.2011.034
B. S. PAWLYK; O. V. BULGAKOV; X. SUN; M. ADAMIAN; X. SHU; A. J. SMITH; E. L. BERSON; R. R. ALI; S. KHANI; A. F. WRIGHT: "Photoreceptor rescue by an abbreviated human RPGR gene in a murine model of X-linked retinitis pigmentosa", GENE THER, vol. 23, 2016, pages 196 - 204, XP055310551, DOI: doi:10.1038/gt.2015.93
BAINBRIDGE JWB ET AL.: "Effect of gene therapy on visual function in Leber's congenital amaurosis.", N. ENGL. J. MED., vol. 358, no. 21, 2008, pages 2231 - 9, XP002718542, DOI: doi:10.1056/NEJMoa0802268
BAINBRIDGE JWB ET AL.: "Long-Term Effect of Gene Therapy on Leber's Congenital Amaurosis", N. ENGL. J. MED., 2015
BANTEL-SCHAAL ET AL., J. VIROLOGY, vol. 73, 1999, pages 939
BELL CL; GURDA BL; VAN VLIET K; AGBANDJE-MCKENNA M; WILSON JM: "Identification of the Galactose Binding Domain of the Adeno-Associated Virus Serotype 9 Capsid", J. VIROL., vol. 86, no. 13, 2012, pages 7326 - 7333, XP055165759, DOI: doi:10.1128/JVI.00448-12
BLAZEK E; MITTLER G; MEISTERERNST M: "The mediator of RNA polymerase II", CHROMOSOMA, vol. 113, no. 8, 2005, pages 399 - 408, XP019342025, DOI: doi:10.1007/s00412-005-0329-5
BOOIJ ET AL., OPHTHALMOLOGY, vol. 118, 2011, pages 160 - 167
BOYD JM; VERMA S; UHLMANN E: "14584 Corrections", ANNU. REV. MICROBIOL., vol. 95, 1998, pages 2 - 3
BOYD RF ET AL.: "Photoreceptor-targeted gene delivery using intravitreally administered AAV vectors in dogs", GENE THER., vol. 23, no. 2, 2016, pages 223 - 230
BUSSKAMP V ET AL.: "Genetic reactivation of cone photoreceptors restores visual responses in retinitis pigmentosa. [Internet", SCIENCE, vol. 329, no. 5990, 2010, pages 413 - 7, XP002679057, DOI: doi:10.1126/SCIENCE.1190897
BYRNE LC ET AL.: "Viral-mediated RdCVF and RdCVFL expression protects cone and rod photoreceptors in retinal degeneration. [Internet", J. CLIN. INVEST., vol. 125, no. 1, 2015, pages 105 - 16, XP055230095, DOI: doi:10.1172/JCI65654
CHALMEL F; LEVEILLARD T; JAILLARD C; LARDENOIS A; BERDUGO N; MOREL E; KOEHL P; LAMBROU G; HOLMGREN A; SAHEL JA: "Poch O. Rod-derived Cone Viability Factor-2 is a novel bifunctional-thioredoxin-like protein with therapeutic potential", BMC MOL BIOL, vol. 8, 31 August 2007 (2007-08-31), pages 74, XP021028244, DOI: doi:10.1186/1471-2199-8-74
CHIORINI ET AL., J. VIROLOGY, vol. 71, 1998, pages 6823
CHIORINI ET AL., J. VIROLOGY, vol. 73, 1999, pages 1309
CHOI VW; ASOKAN A; HABERMAN R A; SAMULSKI RJ: "Curr. Protoc. Hum. Genet.", 2007, article "Production of recombinant adeno-associated viral vectors."
CHUONG AS ET AL.: "Noninvasive optical inhibition with a red-shifted microbial rhodopsin [Internet", NAT NEUROSCI, vol. 17, no. 8, 2014, pages 1123 - 1129
CIDECIYAN A V ET AL.: "Human gene therapy for RPE65 isomerase deficiency activates the retinoid cycle of vision but with slow rod kinetics.", PROC. NATL. ACAD. SCI. U. S. A., vol. 105, no. 39, 2008, pages 15112,7, XP055390820, DOI: doi:10.1073/pnas.0807027105
D. H. HONG; B. S. PAWLYK; M. ADAMIAN; M. A. SANDBERG; T. LI; A SINGLE: "abbreviated RPGR-ORF15 variant reconstitutes RPGR function in vivo", INVEST OPHTHALMOL VIS SCI, vol. 46, 2005, pages 435 - 441, XP055218709, DOI: doi:10.1167/iovs.04-1065
DALKARA D ET AL.: "In Vivo-Directed Evolution of a New Adeno-Associated Virus for Therapeutic Outer Retinal Gene Delivery from the Vitreous", SCI. TRANSL. MED., vol. 5, no. 189, 2013, pages 189ra76 - 189ra76, XP055452295
DALKARA D ET AL.: "In vivo-directed evolution of a new adeno-associated virus for therapeutic outer retinal gene delivery from the vitreous. [Internet", SCI. TRANSL. MED., vol. 5, 2013, pages 189ra76, XP055452295
DELYFER M-N ET AL.: "Transcriptomic Analysis of Human Retinal Detachment Reveals Both Inflammatory Response and Photoreceptor Death", PLOS ONE, vol. 6, no. 12, 2011, pages e28791
DIESTER ET AL., NAT. NEUROSCI., vol. 14, 2011, pages 387
DUNCAN JL: "Visual Consequences of Delivering Therapies to the Subretinal Space", JAMA OPHTHALMOL, vol. 135, no. 3, 2017, pages 242 - 243
FRADOT M; BUSSKAMP V; BENNETT J, GENE THERAPY IN OPHTHALMOLOGY : VALIDATION ON CULTURED, vol. 593, May 2011 (2011-05-01), pages 587 - 593
GAO ET AL., PROC. NAT. ACAD. SCI. USA, vol. 99, 2002, pages 11854
GERNOUX G; WILSON JM; MUELLER C.: "Regulatory and Exhausted T Cell Responses to AAV Capsid", HUM. GENE THER., vol. 28, no. 4, 2017, pages 338 - 349
GUO-JIE YE ET AL: "Cone-Specific Promoters for Gene Therapy of Achromatopsia and Other Retinal Diseases", HUMAN GENE THERAPY, vol. 27, no. 1, 1 January 2016 (2016-01-01), US, pages 72 - 82, XP055266044, ISSN: 1043-0342, DOI: 10.1089/hum.2015.130 *
GUO-JIE YE ET AL: "Safety and Biodistribution Evaluation in Cynomolgus Macaques of rAAV2tYF-PR1.7-hCNGB3, a Recombinant AAV Vector for Treatment of Achromatopsia", HUMAN GENE THERAPY CLINICAL DEVELOPMENT, vol. 27, no. 1, 1 March 2016 (2016-03-01), pages 37 - 48, XP055372702, ISSN: 2324-8637, DOI: 10.1089/humc.2015.164 *
HANEN KHABOU ET AL: "Insight into the mechanisms of enhanced retinal transduction by the engineered AAV2 capsid variant -7m8 : Mechanisms of Enhanced Transduction by AAV2-7m8", BIOTECHNOLOGY AND BIOENGINEERING, vol. 113, no. 12, 30 June 2016 (2016-06-30), pages 2712 - 2724, XP055372484, ISSN: 0006-3592, DOI: 10.1002/bit.26031 *
HANEN KHABOU ET AL: "Noninvasive gene delivery to foveal cones for vision restoration", JCI INSIGHT, vol. 3, no. 2, 25 January 2018 (2018-01-25), XP055454489, DOI: 10.1172/jci.insight.96029 *
JACOBSON SG ET AL.: "Gene therapy for leber congenital amaurosis caused by RPE65 mutations: safety and efficacy in 15 children and adults followed up to 3 years", ARCH. OPHTHALMOL, vol. 130, no. 1, 2012, pages 9 - 24
JACOBSON SG ET AL.: "Gene Therapy for Leber Congenital Amaurosis caused by RPE65 mutations: Safety and Efficacy in Fifteen Children and Adults Followed up to Three Years", ARCH. OPHTHALMOL., vol. 130, no. 1, 2012, pages 9 - 24
JACOBSON SG ET AL.: "Improvement and Decline in Vision with Gene Therapy in Childhood Blindness", N. ENGL. J. MED., 2015
JEFFERY, TRENDS BIOCHEM. SCI., vol. 24, no. l, 1999, pages 8 - l 1
JEFFERY, TRENDS GENET., vol. 19, no. 8, 2003, pages 415 - 417
KAY CN ET AL.: "Targeting photoreceptors via intravitreal delivery using novel, capsid-mutated AAV vectors.", PLOS ONE, vol. 8, no. 4, 2013, pages e62097, XP055372490, DOI: doi:10.1371/journal.pone.0062097
KHABOU H ET AL.: "Insight into the mechanisms of enhanced retinal transduction by the engineered AAV2 capsid variant -7m8", BIOTECHNOL. BIOENG., June 2016 (2016-06-01)
KOHL ET AL., EUR J HUM GENET, vol. 13, no. 3, 2005, pages 302
KOLB H; ZHANG L; DEKORVER L; CUENCA N: "A new look at calretinin-immunoreactive amacrine cell types in the monkey retina.", J. COMP. NEUROL., vol. 453, no. 2, 2002, pages 168 - 84
KOLSTAD KD ET AL.: "Changes in adeno-associated virus-mediated gene delivery in retinal degeneration", HUM. GENE THER., vol. 21, no. 5, 2010, pages 571 - 578, XP055356163, DOI: doi:10.1089/hum.2009.194
KOMAROMY AM ET AL.: "Gene therapy rescues cone function in congenital achromatopsia", HUM. MOL. GENET., vol. 19, 2010, pages 2581 - 2593, XP002615067, DOI: doi:10.1093/HMG/DDQ136
KOTTERMAN M A ET AL.: "Antibody neutralization poses a barrier to intravitreal adeno-associated viral vector gene delivery to non-human primates", GENE THER., August 2014 (2014-08-01), pages l-11
LEVEILLARD T; MOHAND-SAID S; LORENTZ O; HICKS D; FINTZ A C; CLERIN E ET AL.: "Identification and characterization of rod-derived cone viability factor", NAT GENET, vol. 36, 2004, pages 755 - 759, XP002313018, DOI: doi:10.1038/ng1386
MACLAREN RE ET AL., RETINAL GENE THERAPY IN PATIENTS WITH CHOROIDEREMIA: INITIAL FI NDINGS FROM A PHASE 1/2 CLINICAL TRIAL. LANCET, vol. 383, no. 9923, 2014, pages 1129,1137
MAGUIRE AM ET AL.: "Safety and Efficacy of Gene Transfer for Leber's Congenital Amaurosis", N. ENGL. J. MED., vol. 358, no. 21, 2008, pages 2240 - 2248, XP055027817, DOI: doi:10.1056/NEJMoa0802315
MAGUIRE AM ET AL.: "Safety and efficacy of gene transfer for Leber's congenital amaurosis.", N. ENGL. J. MED., vol. 358, no. 21, 2008, pages 2240 - 8, XP055027817, DOI: doi:10.1056/NEJMoa0802315
MORIS ET AL., VIROLOGY, vol. 33, 2004, pages 375 - 383
MURAMATSU ET AL., VIROLOGY, vol. 221, 1996, pages 208
PICKRELL SW; ZHU X; WANG X; CRAFT CM: "Deciphering the contribution of known cis-elements in the mouse cone arrestin gene to its cone-specific expression", INVESTIG. OPHTHALMOL. VIS. SCI., vol. 45, no. 11, 2004, pages 3877 - 3884
RAMACHANDRAN PS ET AL.: "Evaluation of Dose and Safety of AAV7m8 and AAV8BP2 in the Non-Human Primate Retina.", HUM. GENE THER., October 2016 (2016-10-01)
REGULATORS LT: "Letter to the Editor A Unified Nomenclature for Protein Subunits of Mediator Complexes", MOL. CELL, vol. 14, 2004, pages 553 - 557
REICHMAN S ET AL.: "The homeobox gene CHX10/VSX2 regulates RdCVF promoter activity in the inner retina", HUM. MOL. GENET., vol. 19, no. 2, 2010, pages 250 - 61
REICHMAN S; TERRAY A; SLEMBROUCK A; NANTEAU C; ORIEUX G, FROM CONFLUENT HUMAN IPS CELLS TO SELF-FORMING NEURAL RETINA AND RETINAL PIGMENTED EPITHELIUM, 2014
SANDY HUNG ET AL: "AAV-Mediated CRISPR/Cas Gene Editing of Retinal Cells In Vivo", INVESTIGATIVE OPHTHALMOLOGY & VISUAL SCIENCE, 1 June 2016 (2016-06-01), United States, pages 3470 - 3476, XP055373371, Retrieved from the Internet <URL:http://iovs.arvojournals.org/pdfaccess.ashx?url=/data/journals/iovs/935339/i1552-5783-57-7-3470.pdf> DOI: 10.1167/iovs.16-19316 *
SATOH S ET AL.: "The spatial patterning of mouse cone opsin expression is regulated by bone morphogenetic protein signaling through downstream effector COUP-TF nuclear receptors.", J. NEUROSCI., vol. 29, no. 40, 2009, pages 12401 - 11
SENGUPTA A ET AL.: "Red-shifted channelrhodopsin stimulation restores light responses in blind mice , macaque retina , and human retina", EMBO MOL MED, 2016, pages 1 - 17
SHADE ET AL., J. VIROL., vol. 58, 1986, pages 921
SINHA R ET AL.: "Cellular and Circuit Mechanisms Shaping the Perceptual Properties of the Primate Fovea", CELL, vol. 168, no. 3, 2017, pages 413 - 426, XP029897819, DOI: doi:10.1016/j.cell.2017.01.005
SRIVISTAVA ET AL., J. VIROLOGY, vol. 45, 1983, pages 555
TENENBAUM L; LEHTONEN E; MONAHAN PE: "Evaluation of risks related to the use of adeno-associated virus-based vectors", CURR. GENE THER., vol. 3, no. 6, 2003, pages 545 - 65
THIERRY LÉVEILLARD ET AL: "Rod-Derived Cone Viability Factor for Treating Blinding Diseases: From Clinic to Redox Signaling", SCIENCE TRANSLATIONAL MEDICINE,, vol. 2, no. 26, 7 April 2010 (2010-04-07), pages 1 - 5, XP009160751, ISSN: 1946-6234, DOI: 10.1126/SCITRANSLMED.3000866 *
VACCA O ET AL.: "AAV-mediated gene delivery in Dp71-null mouse model with compromised barriers. [Internet", GLIA, vol. 62, no. 3, 2014, pages 468 - 76, XP055141047, DOI: doi:10.1002/glia.22617
VANDENBERGHE LH ET AL.: "AAV9 targets cone photoreceptors in the nonhuman primate retina.", PLOS ONE, vol. 8, no. 1, 2013, pages e53463
VANDENBERGHE LH ET AL.: "Dosage thresholds for AAV2 and AAV8 photoreceptor gene therapy in monkey", SCI. TRANSL. MED., vol. 3, no. 88, 2011, pages 88ra54
VIETS K; ELDRED KC; JR RJJ: "Mechanisms of Photoreceptor Patterning in Vertebrates and Invertebrates", TRENDS GENET., vol. 32, no. 10, 2016, pages 638 - 659, XP029737885, DOI: doi:10.1016/j.tig.2016.07.004
VON MERING C ET AL.: "STRING 7 - Recent developments in the integration and prediction of protein interactions", NUCLEIC ACIDS RES., vol. 35, no. 1, 2007, pages 358 - 362
W. A. BELTRAN; A. V. CIDECIYAN; A. S. LEWIN; S. IWABE; H. KHANNA; A. SUMAROKA; V. A. CHIODO; D. S. FAJARDO; A. J. ROMAN; W.-T. DEN, PROC. NATL. ACAD. SCI. U.S.A., vol. 109, 2012, pages 2132 - 2137
W. A. BELTRAN; A. V. CIDECIYAN; A. S. LEWIN; W. W. HAUSWIRTH; S. G. JACOBSON; G. D. AGUIRRE: "Gene augmentation for X-linked retinitis pigmentosa caused by mutations in RPGR", COLD SPRING HARBOR PERSPECTIVES IN MEDICINE, vol. 5, 2014, pages a017392
W. T. DENG; F. M. DYKA; A. DINCULESCU; J. LI; P. ZHU; V. A. CHIODO; S. L. BOYE; T. J. CONLON; K. ERGER; T. COSSETTE: "Stability and Safety of an AAV Vector for Treating RPGR-ORF15 X-Linked Retinitis Pigmentosa", HUM GENE THER, vol. 26, 2015, pages 593 - 602, XP055313051, DOI: doi:10.1089/hum.2015.035
WASSLE H; GRUNERT U; ROHRENBECK J; BOYCOTT BB: "Cortical magnification factor and the ganglion cell density of the primate retina", NATURE, vol. 341, no. 6243, 1989, pages 643 - 646
WIKLER KC; WILLIAMS RW; RAKIC P.: "Photoreceptor mosaic: Number and distribution of rods and cones in the rhesus monkey retina", J. COMP. NEUROL., vol. 297, no. 4, 1990, pages 499 - 508
XIAO ET AL., J. VIROLOGY, vol. 73, 1999, pages 3994
YE G ET AL.: "Safety and Biodistribution Evaluation in CNGB3-Deficient Mice of rAAV2tYF-PRl . 7-hCNGB3", A RECOMBINANT AAV VECTOR FOR TREATMENT OF ACHROMATOPSIA, vol. 27, no. 1, 2016, pages 27 - 37, XP055372695, DOI: doi:10.1089/humc.2015.163
YE G-J ET AL.: "Cone-Specific Promoters for Gene Therapy of Achromatopsia and Other Retinal Diseases", HUM. GENE THER., vol. 27, no. l, 2016, pages 72 - 82, XP055266044, DOI: doi:10.1089/hum.2015.130
YE G-J ET AL.: "Safety and Biodistribution Evaluation in Cynomolgus Macaques of rAAV2tYF-PR1.7-hCNGB3, a Recombinant AAV Vector for Treatment of Achromatopsia", HUM. GENE THER. CLIN. DEV., vol. 27, no. 1, 2016, XP055372702, DOI: doi:10.1089/humc.2015.164
YIN L ET AL.: "Intravitreal injection of AAV2 transduces macaque inner retina.", INVEST. OPHTHALMOL. VIS. SCI., vol. 52, no. 5, 2011, pages 2775 - 83, XP002714132, DOI: doi:10.1167/iovs.10-6250
Z. WU; S. HIRIYANNA; H. QIAN; S. MOOKHERJEE; M. M. CAMPOS; C. GAO; R. FARISS; P. A. SIEVING; T. LI; P. COLOSI: "A long-term efficacy study of gene replacement therapy for RPGR-associated retinal degeneration", HUM MOL GENET, vol. 24, 2015, pages 3956 - 3970, XP055218847, DOI: doi:10.1093/hmg/ddv134

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP4049683A1 (en) * 2016-04-29 2022-08-31 Adverum Biotechnologies, Inc. Evasion of neutralizing antibodies by a recombinant adeno-associated virus
EP3448437A4 (en) * 2016-04-29 2019-11-06 Adverum Biotechnologies, Inc. Evasion of neutralizing antibodies by a recombinant adeno-associated virus
EP4083203A1 (en) * 2016-06-16 2022-11-02 Adverum Biotechnologies, Inc. Compositions and methods for reducing ocular neovascularization
US10745453B2 (en) 2017-11-13 2020-08-18 The Regents Of The University Of California Compositions and methods for enhancing visual function
WO2019094904A1 (en) * 2017-11-13 2019-05-16 The Regents Of The University Of California Compositions and methods for enhancing visual function
WO2020079464A1 (en) * 2018-10-17 2020-04-23 INSERM (Institut National de la Santé et de la Recherche Médicale) Method for treating retinal degeneration disease by administering nucleolin polynucleotide or polypeptide
EP3989982A4 (en) * 2019-06-27 2023-06-28 University Of Florida Research Foundation, Incorporated Enhancing aav-mediated transduction of ocular tissues with hyaluronic acid
US11744851B2 (en) 2019-06-27 2023-09-05 University Of Florida Research Foundation, Incorporated Enhancing AAV-mediated transduction of ocular tissues with hyaluronic acid
EP3892738A1 (en) 2020-04-10 2021-10-13 Sorbonne Université G-protein-gated-k+ channel-mediated enhancements in light sensitivity in rod-cone dystrophy (rcd)
WO2021204407A1 (en) 2020-04-10 2021-10-14 Sorbonne Université G-protein-gated-k+ channel-mediated enhancements in light sensitivity in rod-cone dystrophy (rcd)
WO2022248634A1 (en) 2021-05-26 2022-12-01 Sparingvision G-protein-gated-k+ channel-mediated enhancements in light sensitivity in rod-cone dystrophy (rcd)
EP4163296A1 (en) 2021-10-11 2023-04-12 Sparingvision G-protein-gated-k+ channel-mediated enhancements in light sensitivity in patients with rod-cone dystrophy (rcd)
EP4357359A1 (en) 2022-10-20 2024-04-24 Sparingvision G-protein-gated-k+ channel-mediated enhancements in light sensitivity in rod-cone dystrophy (rcd)

Also Published As

Publication number Publication date
EP3570895A1 (en) 2019-11-27

Similar Documents

Publication Publication Date Title
WO2018134168A1 (en) Methods of expressing a polynucleotide of interest in the cone photoreceptors
US11723988B2 (en) Methods of expressing a polynucleotide of interest in the cone photoreceptors of a subject comprising the subretinal delivery of a therapeutically effective amount of a recombinant AAV9-derived vector
US20230323398A1 (en) Viral vectors for the treatment of retinal dystrophy
Kong et al. Self-complementary AAV5 vector facilitates quicker transgene expression in photoreceptor and retinal pigment epithelial cells of normal mouse
US20210188927A1 (en) Compositions and methods for treating age-related macular degeneration
JP2011516049A (en) How to treat genetic disorders
US20210371480A1 (en) Compositions and methods for treating age-related macular degeneration and other diseases
US20220347320A1 (en) Regeneration of retinal ganglion cells
JP2021510301A (en) Compositions and Methods for Treating Retinopathy
Fischer On retinal gene therapy
EP4061831A1 (en) Materials and methods for treatment of disorders associated with the ighmbp2 gene
US20220143217A1 (en) Neuroprotective gene therapy targeting the akt pathway
EA046019B1 (en) COMPOSITIONS AND METHODS FOR TREATING RETINA DISORDERS

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: 18701288

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2018701288

Country of ref document: EP

Effective date: 20190819