TREATMENT OF DEAFNESS BY GENE THERAPY

Information

  • Patent Application
  • 20210023242
  • Publication Number
    20210023242
  • Date Filed
    February 21, 2019
    5 years ago
  • Date Published
    January 28, 2021
    3 years ago
Abstract
The present invention relates to a viral vector, especially an adeno-associated virus (AAV) vector, and the use thereof in the gene-therapy treatment of hearing impairment, especially of hearing impairment based on one or more mutations in the otoferlin gene (OTOF).
Description
TECHNICAL FIELD OF THE INVENTION

The present invention relates to a viral vector, especially an adeno-associated virus (AAV) vector, and the use thereof in the gene-therapy treatment of hearing impairment, especially of hearing impairment based on one or more mutations in the otoferlin gene (OTOF).


TECHNICAL BACKGROUND OF THE INVENTION

Hearing is a complex process through which acoustic signals are perceived and processed via the ears and in the brain. Hearing impairment is a reduction in hearing ability culminating in deafness. According to WHO classification, hearing impairment can be mild (faintest perceptible sounds between 25-40 dB; WHO 1), middling/moderate (faintest perceptible sounds between 41-60 dB; WHO 2), severe/extreme (faintest perceptible sounds between 61-80 dB; WHO 3) and profound or bordering on deafness (faintest perceptible sounds over 81 dB; WHO 4). There are many different causes and these encompass, for example, excessive noise exposure (the most common cause), age, various diseases and also genetic causes. The incidence of hereditary hearing impairment is approx. 1 in 650-1000, with currently over 100 different genes being known in which defects/mutations are associated with hearing impairment. Globally, about 360 million people are hearing impaired according to WHO classification, about 16% of the population in Germany over 18 years of age. Over the age of 65, this is every second man and every third woman.


Mutations affecting the otoferlin gene (OTOF), which encodes the multi-C2 domain protein otoferlin, lead to severe, nonsyndromic forms of prelingual DFNB9 hearing impairment or temperature-sensitive auditory synaptopathy (see, for example, Yasunaga et al. Nat. Genet. 1999, 21:363-369; Varga et al. J. Med. Genet. 2006, 43:576-581; Shearer and Smith GeneReviews® Seattle (Wash.): University of Washington, Seattle; 1993-2018; Santarelli et al. Hear Res. 2015, 330:200-212; Pangršič et al. Trends Neurosci. 2012, 35(11):671-680; Rodriguez-Ballesteros et al. Hum Mutat 2008, 29:823-831). From a clinical perspective, 5-8% of all cases of autosomal recessive nonsyndromic hearing loss in some Western populations is OTOF-mediated deafness (Rodríguez-Ballesteros et al. Hum Mutat 2008, 29:823-831), which thus occupies a place under the top five of all genetic hearing disorders for which there is still no treatment option (Angeli et al. Anat Rec (Hoboken) 2012, 295:1812-1829).


Concerning the molecular function of otoferlin, it is now well-known that otoferlin is expressed in sensory inner hair cells (IHC) of the cochlea and plays a key role in the final steps of presynaptic vesicle fusion at synapses of the inner hair cells of the cochlea with afferent neurons of the spiral ganglion (Roux et al. Cell 2006, 127:277-289). Here, multiple functions have been proposed for otoferlin, such as, for instance, a role in (i) presynaptic Ca2+ binding for triggering vesicular exocytosis after hair cell depolarization (see, for example, Roux et al. Hum Mol Genet 2009, 18:4615-4628; Beurg et al. J Neurosci 2010, 30:13281-13290; Johnson et al. Nat Neurosci 2010, 13:45-52; Michalski et al. Elife 2017, 6), and (ii) vesicular priming and refilling of the synaptic vesicles at the release site in order to ensure an untiring and temporally precise neurotransmitter release even under relatively long stimulation (see, for example, Pangršič et al. Nat Neurosci 2010, 13:869-876; Vogl et al. J Cell Sci 2015, 128:638-644; Jung et al. EMBO J 2015, 34:2686-2702; Strenzke et al. EMBO J 2016, 35:2519-2535; Vogl et al. EMBO J 2016, 35:2536-2552).


Patients with hereditary impairments in hearing ability, such as, for example, hearing impairment based on mutations in the otoferlin gene (OTOF), must generally resort to hearing aids or cochlear implants, since medicaments which can restore hearing ability and other cause-controlling treatment methods are so far not available. Gene therapy, i.e., the introduction of intact copies of defective genes into the affected cell populations in the ear, could be a major advance toward restoring hearing ability in cases of monogenic (but also acquired) hearing loss—for example, animal studies show a partial improvement in hearing ability in the case of gene therapy in mutated mice (see, for example, Akil et al. Neuron 2012, 75:283-293; Askew et al. Sci. Transl. Med. 2015, 7:295ra108; Jung et al. EMBO J 2015, 34:2686-2702; Landegger et al. Nat Biotechnol 2017, 35:280-284). Any monogenic hearing disease requires the development of an individual gene-therapy (viral) construct which introduces the repaired gene in question into the affected cells, and such constructs are still not generally available.


To date, multiple Otof mouse mutants (Roux et al. Cell 2006, 127:277-289; Pangršič et al. Nat Neurosci 2010, 13:869-876; Strenzke et al. EMBO J 2016, 35:2519-2535)—and also other mouse models in which the otoferlin protein level in sensory hair cells was lowered to a large extent (e.g., AP2μ, Jung et al. EMBO J 2015, 34:2686-2702; Wrb, Vogl et al. EMBO J 2016, 35:2536-2552)—have been generated, this emphasizing the physiological significance of otoferlin in the presynaptic release at hair cell presynapses. In fact, Otof deletion mutants (Otof knockout) are completely deaf and are therefore a useful model system for investigating genetic restoration (rescue) by corrective viral treatments with the wild-type sequence. The inventors of the present invention have already established viral injections into postnatal murine inner ears, a methodology which allows a subsequent in vivo analysis (i.e., with regard to auditory brainstem recording [ABR] thresholds and ABR amplitudes, etc.) and ex vivo single-cell analysis (presynaptic patch-clamp electrophysiology, immunohistochemistry, etc.) for determining and validating the therapeutic potential of defined rescue constructs.


Owing to the size of the otoferlin-encoding sequence (>5.5 kb) and the limited packaging size of standard adeno-associated viral vectors (AAV; <4.4 kb; Grieger and Samulski J Virol 2005, 79:9933-9944), which are commonly used for gene-therapy applications because of their favorable safety profile, it is a huge challenge to establish gene therapy using full-length otoferlin.


It was therefore a goal of the present invention to provide a gene-therapy viral vector and a corresponding gene-therapy method for treating hearing impairment based on mutations in the otoferlin gene (OTOF).


BRIEF DESCRIPTION OF THE INVENTION

The present invention provides a viral vector comprising a nucleic acid comprising a promoter and an operatively associated encoding sequence which encodes otoferlin or a functional fragment or a variant thereof.


In one embodiment, the viral vector is selected from the group consisting of adeno-associated virus vector (AAV vector), adenovirus vector, lentivirus vector, herpes simplex virus (HSV) vector, vaccinia virus vector and Sendai virus vector.


In one embodiment, the nucleic acid is DNA or RNA, preferably DNA.


In one embodiment, the viral vector is an AAV vector or an adenovirus vector.


In one embodiment, the AAV vector is selected from the group consisting of AAV-PHP.B, AAV-PHP.eB, AAV-PHP.S and AAV-Anc80.


In one embodiment, the AAV vector is selected from the group consisting of AAV-8, AAV-9 and AAV-1/2.


In one embodiment, the promoter is selected from the group consisting of cytomegalovirus (CMV) promoter, human β-actin/CMV hybrid promoter, chicken β-actin/CMV hybrid promoter, CMV-actin-globin (CAG) hybrid promoter, Math1 promoter, VGLUT3 promoter, parvalbumin promoter, calretinin promoter, calbindin 28k promoter, prestin promoter, otoferlin promoter and myosin II, V, VI, VIIa or XVa promoter.


In one embodiment, the promoter is a human β-actin/CMV hybrid promoter.


In one embodiment, the encoding sequence encodes full-length otoferlin.


In one embodiment, the viral vector is an exosome-associated viral vector.


In one embodiment, the viral vector is an exo-AAV vector.


In a further aspect, the present invention provides a pharmaceutical composition comprising a viral vector of the present invention and a pharmaceutically compatible carrier or excipient.


In a further aspect, the present invention provides a viral vector of the present invention or a pharmaceutical composition of the present invention for use as a medicament.


In a further aspect, the present invention provides a viral vector of the present invention or a pharmaceutical composition of the present invention for use in a method for treating hearing impairment.


In one embodiment, the hearing impairment is a hearing impairment based on one or more mutations in the otoferlin gene (OTOF).


In one embodiment, the hearing impairment is DFNB9 hearing impairment.


In one embodiment, the method comprises the administration of the viral vector into the inner ear, especially into the cochlea, especially into inner hair cells of the cochlea.


In one embodiment, the administration comprises injection through the round window, injection into the scala vestibuli via a stapedotomy, injection into the scala tympani via a cochleostomy and/or application as a repository into the round window niche, for example as a constituent of a gel, of a sponge or via an application catheter.


In one embodiment, the administration leads to an expression of otoferlin or the functional fragment or the variant thereof in inner hair cells of the cochlea.


In a further aspect, the present invention provides for the use of a viral vector of the present invention in the production of a medicament for treating hearing impairment.


In one embodiment, the hearing impairment is a hearing impairment based on one or more mutations in the otoferlin gene (OTOF).


In one embodiment, the hearing impairment is DFNB9 hearing impairment.


In a further aspect, the present invention provides a method for treating hearing impairment, comprising the administration of the viral vector of the present invention into the inner ear, especially into the cochlea, especially into inner hair cells of the cochlea.


In one embodiment, the hearing impairment is a hearing impairment based on one or more mutations in the otoferlin gene (OTOF).


In one embodiment, the hearing impairment is DFNB9 hearing impairment.


In one embodiment, the administration comprises injection through the round window, injection into the scala vestibuli via a stapedotomy, injection into the scala tympani via a cochleostomy and/or application as a repository into the round window niche, for example as a constituent of a gel, of a sponge or via an application catheter.


In one embodiment, the administration leads to an expression of otoferlin or the functional fragment or the variant thereof in inner hair cells of the cochlea.





DESCRIPTION OF THE FIGURES


FIG. 1 shows a cloning strategy scheme for full-length otoferlin (A) and also the vector map of the cis-plasmid containing full-length otoferlin (B).



FIG. 2 shows the vector maps for the plasmids used in the production of virus particles in HEK293T cells, pHelper (Takara/Clontech; A) and tTA-iCAP-PHPb (trans-plasmid; B), which provides the viral capsid PHP.B (Deverman et al. Nat. Biotechnol. 2016, 34:204-209).



FIG. 3 shows the virus-mediated restoration of hearing ability in otoferlin knockout mice, where A is a schematic representation of the otoferlin rescue construct used for the AAV-PHP.B vector (C2A-F-C2A-F domains; CC—coiled coil domain; FerB—ferlin B domain; TM—transmembrane domain), B is a schematic representation of the AAV-PHP.B vector construct bearing the otoferlin cDNA under the control of the human β-actin/CMV hybrid promoter, B′ and B″ show the postnatal (p5-7) method for injecting virus into the cochlea of mice, C shows the AAV-PHP.B-mediated exogeneous expression of otoferlin in the inner hair cells of OTOF KO mice (calretinin as counterdye for specific labeling of inner hair cells; scale bar 5 μm), and D, D′ and D″ show auditory brainstem recordings (ABR) on adult, postnatally injected OTOF KO mice, which show the successful restoration of ABR amplitudes and average click thresholds due to the AAV-PHP.B-otoferlin vector (**p<0.005).





DETAILED DESCRIPTION OF THE INVENTION

The present invention provides a viral vector comprising a nucleic acid comprising a promoter and an operatively associated encoding sequence which encodes otoferlin or a functional fragment or a variant thereof.


In one embodiment, the nucleic acid is DNA or RNA, preferably DNA. The nucleic acid can also be referred to herein as a genetic construct.


The term “viral vector” refers to a virus particle which is used to channel genetic material (e.g., an encoding sequence which encodes otoferlin or a functional fragment or a variant thereof) into target cells. The transport of said genetic material is referred to as “transduction”.


In one embodiment, the viral vector is selected from the group consisting of adeno-associated virus vector (AAV vector), adenovirus vector, lentivirus vector, herpes simplex virus (HSV) vector, vaccinia virus vector and Sendai virus vector. Suitable viral vectors are, for example, described in Sacheli et al. Gene Therapy 2012, 1-11.


In one embodiment, the viral vector is an AAV vector or an adenovirus vector (e.g., Ad5, Ad28 or Hd-Ad).


In a preferred embodiment, the viral vector is an AAV vector.


The term “AAV vector” includes AAV vectors of all serotypes, and also AAV vectors based on the combination of different serotypes (also referred to as “hybrid AAV vectors” or “pseudotype AAV vectors”). In one embodiment, the AAV vector is an AAV vector having a serotype selected from the group consisting of AAV-1, AAV-2, AAV-3, AAV-4, AAV-5, AAV-6, AAV-7, AAV-8, AAV-9, AAV-10, AAV-11, AAV-7m8 and combinations thereof. The synthetic AAV variant AAV-7m8 is, for example, described in Dalkara et al. Sci Transl Med 2013, 5(189):189ra76. Likewise included are other synthetic AAV vectors, such as, for instance, AAV-PHP.B, AAV-PHP.B2, AAV-PHP.B3, AAV-PHP.A, AAV-PHP.eB and AAV-PHP.S (Deverman et al. Nature Biotechnol 2016, 34:204-209; Chan et al. Nature Neuroscience 2017, 20:1172-1179) or AAV-Anc80 (WO 2017/100791 A1; Landegger et al. Nat Biotechnol 2017, 35:280-284; Zinn et al. Cell Rep. 2015, 12(6):1056-1068). Suitable AAV vectors are also commercially available, for example from Penn Vector Core (PA, USA) and SignaGen Laboratories (MD, USA).


In one embodiment, the AAV vector is an AAV-8 or an AAV-9 vector.


In one embodiment, the AAV vector is an AAV-1/2 vector. The term “AAV-1/2 vector” refers to an AAV vector, the genome of which is packaged into the capsid of serotype 1 and 2 in mosaic form (see, for example, Choi et al. Curr Gene Ther. 2005, 5(3):299-310). In a preferred embodiment, this is the genome of serotype 2 (AAV-2).


In one embodiment, the AAV vector is a synthetic AAV vector. In a preferred embodiment, the AAV vector is selected from the group consisting of AAV-PHP.B, AAV-PHP.eB, AAV-PHP.S and AAV-Anc80. In one embodiment, the AAV vector is an AAV-7m8 vector or a combination of AAV-7m8 with another serotype.


In one embodiment, the viral vector is an individual viral vector. In one embodiment, the viral vector is not a dual viral vector, for example not a dual AAV vector.


In one embodiment, the viral vector is an exosome-associated viral vector, preferably an exo-AAV vector. Methods for producing exosome-associated viral vectors are known to a person skilled in the art and include, for example, the isolation of exosome-associated viral vectors from the conditioned medium of producer cells by means of ultracentrifugation (see, for example, Hudry et al. Gene Ther. 2016 23(4):380-392). Exosome-associated viral vectors are, for example, distinguished by an increased resistance to neutralizing anti-AAV antibodies, which are formed after virus infection in the body, and this contributes to an increased transduction efficiency.


In one embodiment, the promoter is a constitutive promoter. The term “constitutive promoter”, as used herein, refers to a nonregulated promoter which allows a continuous expression of its associated gene.


In one embodiment, the promoter is selected from the group consisting of cytomegalovirus (CMV) promoter, human β-actin/CMV hybrid promoter, chicken β-actin/CMV hybrid promoter, CMV-actin-globin (CAG) hybrid promoter, Math1 promoter, VGLUT3 promoter, parvalbumin promoter, calretinin promoter, calbindin 28k promoter, prestin promoter, otoferlin promoter and myosin II, V, VI, VIIa or XVa promoter.


In one embodiment, the promoter is a human β-actin/CMV hybrid promoter.


In one embodiment, otoferlin is wild-type otoferlin. In one embodiment, otoferlin is human otoferlin (see, for example, UniProt database ID: Q9HC10). In one embodiment, human otoferlin comprises an amino acid sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7 and SEQ ID NO: 9, or consists of said amino acid sequence. In one embodiment, human otoferlin comprises the amino acid sequence SEQ ID NO: 1 or SEQ ID NO: 3 or consists of said amino acid sequence. In a preferred embodiment, human otoferlin comprises the amino acid sequence SEQ ID NO: 1 or consists of said amino acid sequence.


The term “functional fragment” refers to a fragment of otoferlin that has the same functional activity as otoferlin or a substantially similar (e.g., +/−20% or +/−10%) functional activity to otoferlin. In one embodiment, the functional fragment is an N-terminally and/or C-terminally truncated form of otoferlin. In one embodiment, the functional fragment comprises at least 500, 600, 700, 800, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800 or 1900 continuous amino acid residues of otoferlin. The functional fragment of otoferlin is preferably a fragment having an amino acid sequence which is sufficiently long to identify the fragment as a fragment of otoferlin and to rule out that it is the fragment of a protein which is not otoferlin.


In one embodiment, the variant is a functional variant of otoferlin, for example a variant of otoferlin that has the same functional activity as otoferlin or a substantially similar (e.g., +/−20% or +/−10%) functional activity to otoferlin.


In one embodiment, the variant comprises one or more amino acid insertions, amino acid additions, amino acid deletions and/or amino acid substitutions. In one embodiment, the variant comprises the insertion, addition, deletion and/or substitution (e.g., conservative substitution) of up to 30, 25, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3 or 2 amino acids.


The term “variant”, as used herein, can also refer to naturally occurring mutants, variants and homologs (e.g., orthologs) of otoferlin. In one embodiment, the naturally occurring homolog is mouse otoferlin. In one embodiment, mouse otoferlin comprises an amino acid sequence selected from the group consisting of SEQ ID NO: 11, SEQ ID NO: 13, SEQ ID NO: 15 and SEQ ID NO: 17, or consists of said amino acid sequence. In a preferred embodiment, mouse otoferlin comprises the amino acid sequence SEQ ID NO: 11 or consists of said amino acid sequence.


In one embodiment, the variant comprises an amino acid sequence which is at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identical to an amino acid sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9, SEQ ID NO: 11, SEQ ID NO: 13, SEQ ID NO: 15 and SEQ ID NO: 17, preferably SEQ ID NO: 1 or SEQ ID NO: 3, more preferably SEQ ID NO: 1, or consists of said amino acid sequence.


In one embodiment, the abovementioned functional activity of otoferlin is the ability to restore the presynaptic neurotransmitter release by inner hair cells of an Otof knockout mouse in full or in part, for example through the depolarization-induced increase in membrane capacity that is measured electrophysiologically (equivalent to the fusion of synaptic messenger-substance vesicles with the presynaptic plasma membrane, i.e., “exocytosis”; Roux et al. Cell 2006, 127:277-289; Vogl et al. EMBO J 2016, 35:2536-2552). In one embodiment, the abovementioned functional activity of otoferlin is the ability to restore otoferlin expression in inner hair cells of Otof knockout mice, for example detectable in the cytosol or the hair-cell plasma membrane using immunohistochemical single-cell or tissue RNA sequencing or single-cell or tissue PCR analysis. In one embodiment, the abovementioned functional activity of otoferlin is the ability to restore the hearing ability of an Otof knockout mouse in full or in part, for example determined by auditory brainstem recording (ABR), for example essentially as described in Example 2.


In one embodiment, the encoding sequence comprises a nucleotide sequence which is at least 70%, at least 75%, at least 80%, at least 85%, at least 90% or at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identical to a nucleotide sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO: 16 and SEQ ID NO: 18, preferably SEQ ID NO: 2 or SEQ ID NO: 4, more preferably SEQ ID NO: 2, or consists of said nucleotide sequence. In one embodiment, the encoding sequence comprises the nucleotide sequence of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 10, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO: 16 and SEQ ID NO: 18, preferably SEQ ID NO: 2 or SEQ ID NO: 4, more preferably SEQ ID NO: 2, or consists of said nucleotide sequence.


The similarity between two nucleotide or amino acid sequences, for example expressed by the percentage of their identity, can be determined via sequence alignments. Such alignments can be carried out using various algorithms known to a person skilled in the art, preferably using the mathematical algorithms by Karlin and Altschul (Karlin & Altschul Proc. Natl. Acad. Sci. U.S.A. 1993, 90:5873-5877), for example using hmmalign (HMMER Package, http://hmmer.wustl.edu/), or using the CLUSTAL algorithm (Thompson J.D. et al. Nucleic Acids Res. 1994, 22:4673-80), which is, for example, available at http://www.ebi.ac.uk/Tools/clustalw/or at http://www.ebi.ac.uk/Tools/clustalw2/index.html or at http://npsa-pbil.ibcp.fr/cgi-bin/npsa_automat.pl?page=/NPSA/npsa_clustalw.html.


In a preferred embodiment of the viral vector of the present invention, the encoding sequence encodes full-length otoferlin.


In one embodiment, the nucleic acid present in the viral vector according to the invention, for example AAV vector, comprises further sequence elements. Such sequence elements encompass, for example, inverted terminal repeats (ITRs; for example AAV-2 ITRs), Kozak sequences, resistance genes (e.g., AmpR), polyadenylation sequences (e.g., the polyadenylation sequence of bovine growth hormone, bGH) and regulatory elements, such as the post-transcriptional regulatory element of the woodchuck hepatitis virus (WPRE). In addition, the nucleic acid can contain further encoding sequences. Such further encoding sequences can, for example, encode additional therapeutically active peptides/proteins, or marker proteins (e.g., fluorescent proteins, such as, for instance, EGFP). In one embodiment, the nucleic acid present in the viral vector according to the invention, for example AAV vector, comprises ITRs, a promoter and an operatively associated encoding sequence which encodes otoferlin or a functional fragment or a variant thereof, a WPRE sequence and a polyadenylation sequence (see, for example, FIG. 3B).


In a further aspect, the present invention provides a nucleic acid (or a genetic construct) as described herein.


In a further aspect, the present invention provides a host cell comprising a viral vector of the present invention or a nucleic acid (or a genetic construct) of the present invention. Said host cell can be of a prokaryotic nature (e.g., a bacterial cell) or a eukaryotic nature (e.g., a fungal cell, plant cell or animal cell). Preferably, the host cell has been isolated. In one embodiment, the host cell is a producer cell or producer cell line which allows the production of the viral vector according to the invention (e.g., AAV vector), for example on the basis of the nucleic acid (or the genetic construct) of the present invention and by means of cotransfection of suitable helper constructs, for example helper plasmids (see, for example, US 2004/0235174 A1). Suitable producer cells or producer cell lines are known to a person skilled in the art and include, for example, HEK293 cells or HEK293T cells.


In a further aspect, the present invention provides a nonhuman transgenic animal comprising a viral vector of the present invention or a nucleic acid (or a genetic construct) of the present invention. The term “nonhuman transgenic animal” refers especially to nonhuman primates or other animals, especially a mammal such as cow, horse, pig, sheep, goat, dog, cat, simian, prosimian, bird such as chicken, or rodent such as mouse, rat, guinea pig, hamster and Mongolian gerbil.


Methods for producing viral vectors are known to a person skilled in the art. A method for producing, for example, an AAV vector consists in the triple transfection of a suitable producer cell line, for example HEK293 or HEK293T, and subsequent purification across iodixanol or cesium chloride gradients. Here, the producer cells are transfected with three vectors: encoded on a first vector/plasmid is the gene of interest (in this case: otoferlin) flanked by appropriate packaging signals (see FIG. 1B); encoded on a second vector/plasmid are the required AAV proteins, especially Rep and Cap (e.g., tTA-iCAP-PHPb; see FIG. 2B); and a third vector/plasmid provides adenoviral helper functions, without which AAV particle production is not possible (e.g., pHelper, Takara/Clontech; see FIG. 2A). Suitable methods are also described in Grieger et al. (Nature Protocols 2006, 1(3):1412-1428).


In a further aspect, the present invention provides a pharmaceutical composition comprising a viral vector of the present invention and a pharmaceutically compatible carrier or excipient.


The pharmaceutical composition according to the invention is preferably sterile and contains a therapeutically effective amount of the viral vector.


A “therapeutically effective amount” concerns the amount which, alone or together with further doses, achieves a desired reaction or a desired effect, for example an improvement in or partial or full restoration of hearing ability. A therapeutically effective amount is dependent on the status to be treated, the severity of the disease, the individual parameters of the patient, including age, physiological status, height and weight, the duration of the treatment, the nature of a concomitant therapy (if present), the specific route of administration, and similar factors.


In one embodiment, approx. 108 to approx. 1013 viral particles are administered, suspended in a suitable volume of a carrier.


Possible carriers (e.g., solvents) are, for example, artificial perilymphs, sterile water, Ringer's solution, Ringer's lactate solution, physiological saline solution, bacteriostatic saline solution (e.g., 0.9% benzyl alcohol-containing saline solution), phosphate-buffered saline solution (PBS), Hanks' solution, fixed oils, polyalkylene glycols, hydrogenated naphthalenes and biocompatible polylactides, lactide/glycolide copolymers or polyoxyethylene/polyoxypropylene copolymers. The resultant solutions or suspensions are preferably isotonic. Suitable carriers and their formulation are also described in detail in Remington's Pharmaceutical Sciences, 17th edition, 1985, Mack Publishing Co.


In a preferred embodiment, the carrier is artificial perilymphs.


The term “excipient”, as used herein, includes all substances which can be present in a pharmaceutical composition and are themselves not active ingredients, such as, for example, salts, binders (e.g., lactose, dextrose, sucrose, trehalose, sorbitol, mannitol), lubricants, thickeners, surface-active substances, preservatives, emulsifiers, buffer substances, stabilizers, flavorings or colorants.


The term “pharmaceutically compatible” concerns a nontoxic material which preferably does not interact with the action of the active constituent of the pharmaceutical composition. In particular, the term “pharmaceutically compatible” means that the substance in question has been authorized by a state regulatory authority for use in animals and especially humans or is listed in the U.S. Pharmacopeia, European Pharmacopeia or other recognized pharmacopeia for use in animals and especially humans.


In a further aspect, the present invention provides a viral vector of the present invention or a pharmaceutical composition of the present invention for use as a medicament.


The term “medicament”, as used herein, refers to a substance or composition which is used therapeutically, i.e., in the treatment, improvement or prevention of a disease or health disorder.


In a further aspect, the present invention provides a viral vector of the present invention or a pharmaceutical composition of the present invention for use in a method for treating hearing impairment.


According to the invention, the patient treated or the individual treated is a human being, nonhuman primate or some other animal, especially a mammal such as cow, horse, pig, sheep, goat, dog, cat, simian, prosimian, bird such as chicken, or rodent such as mouse, rat, guinea pig, hamster and Mongolian gerbil. In a particularly preferred embodiment, the patient treated or the individual treated is a human being.


In one embodiment, the hearing impairment is a hearing impairment based on one or more mutations in the otoferlin gene (OTOF).


In one embodiment, the patient treated or the individual treated has one or more mutations in the otoferlin gene, especially mutations which inhibit or block the expression and/or the function of otoferlin.


In one embodiment, the hearing impairment is DFNB9 hearing impairment.


In one embodiment, the method comprises the administration of the viral vector into the inner ear, especially into the cochlea, especially into inner hair cells of the cochlea.


In one embodiment, the administration comprises injection through the round window, injection into the scala vestibuli via a stapedotomy, injection into the scala tympani via a cochleostomy and/or application as a repository into the round window niche, for example as a constituent of a gel, of a sponge or via an application catheter.


In one embodiment, the administration comprises intratympanic injection.


In one embodiment, the administration leads to an expression of otoferlin or the functional fragment or the variant thereof in inner hair cells of the cochlea, for example in inner hair cells of the apical turn of the cochlea.


According to the invention, the term “expression” is used in its most general meaning and encompasses, for example, the production of RNA or of RNA and protein.


In one embodiment, the viral vector according to the invention, the pharmaceutic composition according to the invention and the methods and uses according to the invention allow an expression of otoferlin or the functional fragment or the variant thereof in at least 50%, in at least 60%, in at least 70%, in at least 80%, in at least 90% or in at least 95% of the inner hair cells of the cochlea, preferably of the inner hair cells of the apical turn of the cochlea.


The viral vector according to the invention and the pharmaceutic composition according to the invention are administered in therapeutically effective amounts.


In a further aspect, the present invention provides for the use of a viral vector of the present invention in the production of a medicament for treating hearing impairment.


In one embodiment, the hearing impairment is a hearing impairment based on one or more mutations in the otoferlin gene (OTOF).


In one embodiment, the hearing impairment is DFNB9 hearing impairment.


In a further aspect, the present invention provides a method for treating hearing impairment, comprising the administration of the viral vector of the present invention into the inner ear, especially into the cochlea, especially into inner hair cells of the cochlea.


In one embodiment, the hearing impairment is a hearing impairment based on one or more mutations in the otoferlin gene (OTOF).


In one embodiment, the hearing impairment is DFNB9 hearing impairment.


In one embodiment, the administration comprises injection through the round window, injection into the scala vestibuli via a stapedotomy, injection into the scala tympani via a cochleostomy and/or application as a repository into the round window niche, for example as a constituent of a gel, of a sponge or via an application catheter.


In one embodiment, the administration comprises intratympanic injection.


In one embodiment, the administration leads to an expression of otoferlin or the functional fragment or the variant thereof in inner hair cells of the cochlea, for example in inner hair cells of the apical turn of the cochlea.


EXAMPLES
Example 1: Production of a Full-Length Otoferlin-Expressing Viral Vector
Cloning Method and Strategy

Full-length otoferlin (flOtoferlin) from mouse (SEQ ID NO: 11/12) was subcloned from a previously generated cDNA clone (pcDNA3-mOtof-IRES-EGFP) into a pAAV vector.


Owing to the absence of suitable restriction sites within the otoferlin-encoding nucleotide sequence and the considerable length of the full-length otoferlin cDNA, the use of traditional T4 ligase-based cloning techniques was dispensed with. What was used instead was a fusion cloning strategy in which the target vector (pAAV) was first linearized by digestion using the restriction enzymes NheI and HindIII (Fermentas). This was followed by amplifying three otoferlin fragments which complemented flOtoferlin and which contained sequences overlapping with one another and with the linearized target vector. This step required the optimization of fragments and primers, for example by the determination of optimal fragment sizes and ratios and the use of “split overlaps” for reducing the primer lengths. The NheI/HindIII-linearized pAAV target vector, the otoferlin fragments A, B, C and the position of the primers used are depicted in FIG. 1A. Primer A_F2 comprised, at the 5′ end, a 15 bp overlap with the target vector and also the start codon for the expression of otoferlin, whereas C_R2 comprised the stop codon and a 15 bp overlap with the AAV vector at the 3′ end (see Table 1).









TABLE 1





Primers used for the cloning. The primers are


depicted in 5′-3′ orientation, with overlapping


sequences underlined and start and stop codons for


transcription in bold and italics.







Fragment A


A_F2: AATTCAAGCTGCTAGCcustom-character GCCCTGATTGTTCACC


(SEQ ID NO: 19)





A_R1: CGTTTGTTGTTGCTCATCATCCAAATG


(SEQ ID NO: 20)





Fragment B


B_F1 GAGCAACAACAAACGTATCGCCTATGC


(SEQ ID NO: 21)





B_R1 GCAGCTCGTACTTCTTGGGTTTCCTG


(SEQ ID NO: 22)





Fragment C


C_F1 AGAAGTACGAGCTGCGGGTCATCGTG


(SEQ ID NO: 23)





C_R2 GATTATCGATAAGCTTcustom-character GGCCCCTAGGAGCTT


(SEQ ID NO: 24)









Thereafter, the linearized vector and the three fragments were fused with the aid of the In-Fusion HD Cloning Kit (Takara/Clontech), the instructions from the manufacturer being followed. This approach yielded the final full-length otoferlin virus vector (pAAV-flOtoferlin), which was used for the subsequent virus production. The corresponding vector map of pAAV-flOtoferlin with the ubiquitous human (3-actin/CMV hybrid promoter is depicted in FIG. 1B. Restriction enzyme digestion and Sanger sequencing (SeqLab, Germany) were used to check that the flOtoferlin insert was correct.


Virus Production and Purification

AAVs were generated in HEK293T cells (ATCC) by means of polyethylenimine transfection (25 000 MW, Polysciences, USA) (Gray et al. Current Protocols in Neuroscience 2011, Hoboken N.J., USA: John Wiley & Sons, Inc.; Deverman et al. Nat. Biotechnol. 2016, 34:204-209). In summary, what was carried out was the triple transfection of HEK293T cells with the aid of the pHelper plasmid (Takara/Clontech, see FIG. 2A), a trans-plasmid providing the viral capsid PHP.B (Deverman et al. Nat. Biotechnol. 2016, 34:204-209; see FIG. 2B), and a cis-plasmid providing flOtoferlin (see FIG. 1B). The cell line was regularly tested for mycoplasma. Virus particles were obtained from the medium 72 hours after transfection and from the cells and the medium 120 hours after transfection. Virus particles from the medium were precipitated using 40% polyethylene glycol 8000 (Acros Organics, Germany) in 500 mM NaCl at 4° C. for 2 hours and then, after centrifugation at 4000 g for 30 min, combined with cell pellets for processing. The cell pellets were suspended in 500 mM NaCl, 40 mM Tris, 2.5 mM MgCl2, pH 8 and 100 U/mL salt active nuclease (Arcticzymes, USA) at 37° C. for 30 min. Thereafter, the cell lysates were clarified by centrifugation at 2000 g for 10 min and then purified across iodixanol stepped gradients (Optiprep, Axis Shield, Norway; 15%, 25%, 40% and 60%) at 58 400 rpm for 2.25 hours (Zolotukhin et al. Gene Ther. 1999, 6:973-985; Nature Protocols 2006, 1(3):1412-1428). The viruses were concentrated using Amicon filters (EMD, UFC910024) and formulated in sterile phosphate-buffered saline solution (PBS) supplemented with 0.001% Pluronic F-68 (Gibco, USA). The virus titers were measured using the AAV titration kit (Takara/Clontech) according to the manufacturer's instructions, by determining the number of DNase I-resistant vg using qPCR (StepOne, Applied Biosystems). The purity of the viruses produced was routinely checked by silver staining (Pierce, Germany) after gel electrophoresis (Novex™ 4-12% Tris-glycine, Thermo Fisher Scientific) according to the manufacturers' instructions. The presence of viral capsid proteins was positively confirmed in all virus preparations. The virus stocks were kept at −80° C. until the day of experimentation.


Example 2: In Vivo Application of the Full-Length Otoferlin-Expressing Viral Vector
Animals and Viral Transmission

The postnatal AAV injection (approx. 1-1.5 μl of the virus formulation; 1.29×1012 GC/ml) into the tympanic duct (scala tympani) of the left ear via the round window was done at p5-p7 essentially as described in the study by Akil et al. (Akil et al. Neuron 2012, 75:283-293). What were used were otoferlin knockout (Otof−/−) mice, which have extreme hearing impairment and do not exhibit ABR responses to sound pressure levels up to 120 dB (Reisinger et al. J. Neurosci. 2011, 31:4886-4895). Four weeks after the injection, hearing ability (function of the inner hair cells) was tested by means of auditory brainstem recording (ABR). The animals were then painlessly killed under anesthesia, and the extracted cochleae were extracted and used further for an immunohistochemical analysis. All experiments were carried out in accordance with national animal care guidelines and approved by the animal welfare board of the University of Göttingen and the animal welfare office of the federal state of Niedersachsen (AZ: 33.4-42502-04-14/1391).


Auditory Brainstem Recording (ABR)

For the ABR analysis, mice were anesthetized i.p. using a combination of ketamine (125 mg/kg) and xylazine (2.5 mg/kg). The core temperature was kept constant at 37° C. with the aid of a heated blanket (Hugo Sachs Elektronik-Harvard Apparatus GmbH). For stimulus generation, presentation and data acquisition, use was made of the TDT III system (Tucker Davis Technologies) operated by custom-written Matlab software (Mathworks). Tone bursts (4/6/8/12/16/24/32 kHz, 10 ms plateau, 1 ms cos2 rise/fall) or 0.03 ms clicks were produced ipsilaterally using a JBL 2402 loudspeaker at 40 Hz (tone bursts) or 20 Hz (clicks) in an open space. The difference potential between vertex and mastoid needles was boosted 50 000-fold, filtered (400-4000 Hz) and sampled 1300-fold at a rate of 50 kHz for 20 ms in order to obtain two average ABR traces for each sound intensity. The hearing thresholds were determined with an accuracy of 10 dB as lowest stimulus intensity, which caused a reproducible response waveform in both traces through visual inspection by two independent observers. Tone burst thresholds exceeding the maximum loudspeaker output (100 dB SPL) were rated with a value of 110 dB. For rescue experiments, the injected ear of the Otof knockout mouse was first recorded. Then, the left ear was closed using electrode gel (Pauli-Magnus et al. Neuroscience 2007, 149:673-684) and small cellulose fabric strips, thereby achieving a conductive hearing impairment of 30-40 dB (Pauli-Magnus et al. Neuroscience 2007, 149:673-684), and the noninjected ear was recorded.


Immunohistochemistry and Confocal Microscopy

Cochlea explants were fixed in 4% formaldehyde in PBS on ice (either for 10 min or 1 hour, depending on the molecular target), as previously described (Khimich et al. Nature 2005, 434:889-894; Meyer et al. Nat. Neurosci. 2009, 12:444-453). After washing and a blocking step using a goat serum-containing buffer (16% normal goat serum, 450 mM NaCl, 0.3% Triton X-100 and 20 mM phosphate buffer at pH 7.4), the following primary antibodies were applied at 4° C. overnight: mouse anti-otoferlin (cat. No. ab53233; Abcam), chicken anti-calretinin (cat. No. 214 106; Synaptic Systems). For visualization, secondary AlexaFluor 488, 568 and 647-conjugated antibodies (cat. No. A-11034, A-11011 or A-11075 and A-21236; Thermo Fisher Scientific) were applied at room temperature for 1 h. After fixation of the sample between coverslip and slide in Mowiol, image acquisition was carried out on an Abberior Instruments Expert Line STED microscope (based on an inverted Olympus IX83 microscope) in confocal mode, controlled by Imspector software, using excitation lasers at 485 nm and 640 nm and a 1.4 NA UPlanSApo 100× oil immersion objective. Image stacks with xy pixel sizes of 60×60 nm and a z step width of 200 nm were acquired.


RESULTS

The experimental data presented in FIG. 3 show that a gene-therapy approach using otoferlin-encoding viral vectors is actually feasible and has clinical potential.


The early postnatal transduction of otoferlin knockout mice with full-length otoferlin achieved clinically relevant transduction rates in inner hair cells (IHC) of the cochlea (FIG. 3C) and resulted in adult animals having significantly reduced ABR thresholds (FIG. 3D-D″), this being equivalent to a strong improvement in hearing ability. After AAV injection into the left ear, ABRs were derivable on the injected ear and the noninjected ear, whereas in the noninjected animals, ABRs could not be triggered even at the highest levels. ABR triggering on the noninjected side might be caused by overhearing onto the left inner ear (despite the induced conductive hearing impairment). In addition, there were no observable undesired adverse effects.

Claims
  • 1. A viral vector comprising a nucleic acid comprising a promoter and an operatively associated encoding sequence which encodes full-length otoferlin or a functional fragment or a functional variant thereof, the functional fragment comprising at least 1500 continuous amino acid residues of full-length otoferlin, and the functional variant comprising an amino acid sequence which is at least 75% identical to an amino acid sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9, SEQ ID NO: 11, SEQ ID NO: 13, SEQ ID NO: 15 and SEQ ID NO: 17.
  • 2. The viral vector of claim 1, wherein the viral vector is selected from the group consisting of adeno-associated virus vector (AAV vector), adenovirus vector, lentivirus vector, herpes simplex virus (HSV) vector, vaccinia virus vector and Sendai virus vector, the viral vector preferably being an AAV vector or an adenovirus vector.
  • 3. The viral vector of claim 2, wherein the AAV vector is selected from the group consisting of AAV-PHP.B, AAV-PHP.eB, AAV-PHP.S and AAV-Anc80.
  • 4. The viral vector of claim 2, wherein the AAV vector is selected from the group consisting of AAV-8, AAV-9 and AAV-1/2.
  • 5. The viral vector of claim 1, wherein the promoter is selected from the group consisting of cytomegalovirus (CMV) promoter, human β-actin/CMV hybrid promoter, chicken β-actin/CMV hybrid promoter, CMV-actin-globin (CAG) hybrid promoter, Math1 promoter, VGLUT3 promoter, parvalbumin promoter, calretinin promoter, calbindin 28k promoter, prestin promoter, otoferlin promoter and myosin II, V, VI, VIIa or XVa promoter.
  • 6. The viral vector of claim 1, wherein the promoter is a human β-actin/CMV hybrid promoter.
  • 7. The viral vector of claim 1, wherein the viral vector is an exosome-associated viral vector.
  • 8. The viral vector of claim 7, wherein the viral vector is an exo-AAV vector.
  • 9. A pharmaceutical composition comprising a viral vector as claimed in claim 1 and a pharmaceutically compatible carrier or excipient.
  • 10. The viral vector of claim 1 for use as a medicament.
  • 11. The viral vector of claim 1 for use in a method for treating hearing impairment based on one or more mutations in the otoferlin gene (OTOF).
  • 12. The viral vector or pharmaceutical composition claim 11, wherein the hearing impairment is DFNB9 hearing impairment.
  • 13. The viral vector or pharmaceutical composition claim 11, wherein the method comprises the administration of the viral vector into the inner ear.
  • 14. The viral vector or pharmaceutical composition claim 13, wherein the administration comprises injection through the round window, injection into the scala vestibuli via a stapedotomy, injection into the scala tympani via a cochleostomy and/or application as a repository into the round window niche, for example as a constituent of a gel, of a sponge or via an application catheter.
  • 15. The viral vector or pharmaceutical composition of claim 11, wherein the administration leads to an expression of full-length otoferlin or the functional fragment or the functional variant thereof in inner hair cells of the cochlea.
  • 16. The pharmaceutical composition of claim 9 for use as a medicament.
  • 17. The pharmaceutical composition of claim 9 for use in a method for treating hearing impairment based on one or more mutations in the otoferlin gene (OTOF).
Priority Claims (1)
Number Date Country Kind
10 2018 103 924.7 Feb 2018 DE national
PCT Information
Filing Document Filing Date Country Kind
PCT/EP2019/054352 2/21/2019 WO 00