CN113952474A - Compositions and methods for treating ocular diseases - Google Patents

Compositions and methods for treating ocular diseases Download PDF

Info

Publication number
CN113952474A
CN113952474A CN202010706658.XA CN202010706658A CN113952474A CN 113952474 A CN113952474 A CN 113952474A CN 202010706658 A CN202010706658 A CN 202010706658A CN 113952474 A CN113952474 A CN 113952474A
Authority
CN
China
Prior art keywords
promoter
sequence
composition
aav
linker
Prior art date
Legal status (The legal status 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 status listed.)
Pending
Application number
CN202010706658.XA
Other languages
Chinese (zh)
Inventor
赵巍
施中东
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Insperry Co ltd
Original Assignee
Insperry Co ltd
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 Insperry Co ltd filed Critical Insperry Co ltd
Priority to CN202010706658.XA priority Critical patent/CN113952474A/en
Priority to AU2021313839A priority patent/AU2021313839A1/en
Priority to PCT/IB2021/000498 priority patent/WO2022018516A1/en
Priority to JP2023512663A priority patent/JP2023540464A/en
Priority to EP21845723.2A priority patent/EP4185333A1/en
Priority to CA3186830A priority patent/CA3186830A1/en
Priority to CN202180064547.6A priority patent/CN116323949A/en
Publication of CN113952474A publication Critical patent/CN113952474A/en
Priority to US18/157,599 priority patent/US20230295243A1/en
Pending legal-status Critical Current

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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/162Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/52Isomerases (5)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • 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
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y503/00Intramolecular oxidoreductases (5.3)
    • C12Y503/03Intramolecular oxidoreductases (5.3) transposing C=C bonds (5.3.3)
    • 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
    • C12N2750/14121Viruses as such, e.g. new isolates, mutants or their genomic sequences
    • 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
    • C12N2750/14122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Genetics & Genomics (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Biochemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Diabetes (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Virology (AREA)
  • Molecular Biology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Wood Science & Technology (AREA)
  • Biophysics (AREA)
  • Microbiology (AREA)
  • Biomedical Technology (AREA)
  • Endocrinology (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Ophthalmology & Optometry (AREA)
  • Urology & Nephrology (AREA)
  • Cardiology (AREA)
  • Vascular Medicine (AREA)
  • Emergency Medicine (AREA)
  • Peptides Or Proteins (AREA)
  • Hematology (AREA)
  • Obesity (AREA)

Abstract

The present invention relates to a system for treating ocular diseases, and methods of using the system to treat ocular diseases.

Description

Compositions and methods for treating ocular diseases
Background
Angiogenesis refers to the new formation or growth of blood vessels in tissues, or the further formation or growth of existing capillaries or blood vessels, which has important roles in disease and health. Pathologic ocular angiogenesis or neovascularization can occur in the retina, choroid, and cornea and can lead to severe visual impairment. Ocular angiogenesis is associated with a wide range of diseases including wet age-related macular degeneration (wet AMD), diabetic retinopathy, macular edema, and the like.
Antibody-based drugs, such as anti-VEGF antibodies Lucentis and Avastin, have been developed for the treatment of angiogenesis-related disorders. However, these antibodies are expensive. Therefore, new therapies targeting angiogenesis are needed for the treatment of ocular diseases associated with angiogenesis.
Disclosure of Invention
There is a need in the art to develop drugs and methods that are effective in treating ocular diseases associated with angiogenesis. The present invention provides compositions, systems and methods that address the above-mentioned needs.
In one aspect, the present invention provides a composition comprising: (i) a first polynucleotide, wherein the first nucleotide comprises a first sequence encoding an adeno-associated virus (AAV) capsid protein operably linked to a first promoter and a second sequence encoding an AAV rep protein operably linked to a second promoter, the first and second promoters being suitable for expression in insect cells, and (ii) a second polynucleotide, wherein the second polynucleotide comprises a third sequence encoding a Vascular Endothelial Growth Factor (VEGF) inhibitor operably linked to a CMV, CAG, MNDU3, PGK, EF1a promoter or an eye-specific promoter.
In some embodiments, the VEGF inhibitor is a VEGF antibody or an antigen-binding fragment thereof. In some embodiments, the VEGF inhibitor comprises the sequences of SEQ ID NOs 1, 2, and 3. In some embodiments, the VEGF inhibitor comprises the sequences of SEQ ID NOs 4, 5, and 6. In some embodiments, the VEGF inhibitor comprises the sequence of SEQ ID NO 7. In some embodiments, the VEGF inhibitor comprises the sequence of SEQ ID NO 8.
In some embodiments, the insect cell is an Sf9 cell. In some embodiments, the first promoter or the second promoter is a p10 promoter. In some embodiments, the first promoter or the second promoter is a PolH promoter.
In some embodiments, the ocular-specific promoter is selected from the group consisting of an RPE 65 gene promoter, a human retinal binding protein (CRALBP) gene promoter, a murine 11-cis Retinol Dehydrogenase (RDH) gene promoter, a rhodopsin kinase promoter (rhodopsin kinase promoter), a tissue inhibitor of metalloproteinase 3 (Timp3) promoter, a photoreceptor retinoid binding protein promoter, and a vitroplastic macular dystrophy 2 (vitroplastic muscular 2) promoter, an inter-photoreceptor retinoid-binding protein (IRBP) promoter.
In some embodiments, the first sequence further comprises a poly a sequence at the 3' terminus. In some embodiments, the second sequence further comprises a poly a sequence at the 3' terminus. In some embodiments, the first sequence and the second sequence are linked by a sequence encoding a linker. In some embodiments, the linker is a cleavable linker. In some embodiments, the linker comprises the sequence of the 2A peptide. In some embodiments, the linker encoding sequence further comprises a promoter. In some embodiments, the promoter is an FMDV promoter.
In some embodiments, the third sequence further comprises a poly a sequence at the 3' terminus. In some embodiments, the second polynucleotide further comprises a stuffer sequence. In some embodiments, the second polynucleotide further comprises an Inverted Terminal Repeat (ITR) sequence.
In some embodiments, the second polynucleotide further comprises a fourth sequence encoding an additional therapeutic protein. In some embodiments, the additional therapeutic protein is selected from the group consisting of: VEGF inhibitors, PDGF inhibitors, integrin inhibitors, mTOR inhibitors, angiogenin inhibitors, and TGF β inhibitors. In some embodiments, the third sequence and the fourth sequence are linked by a sequence encoding a linker. In some embodiments, the linker is a cleavable linker. In some embodiments, the linker comprises the sequence of the 2A peptide.
In another aspect, the present invention provides a recombinant adeno-associated virus (rAAV) particle prepared by transfecting a composition of the invention into an insect cell. In some embodiments, the insect cell is an Sf9 cell.
In another aspect, the invention provides a system for treating an ocular disease in a subject in need thereof, comprising a rAAV particle of the invention and a pharmaceutically acceptable carrier.
In another aspect, the present invention provides a method for treating an ocular disease in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the system of the present invention. In some embodiments, the ocular disease is selected from: wet age-related macular degeneration (wet AMD), diabetic retinopathy, diabetic macular edema, proliferative diabetic retinopathy, non-proliferative diabetic retinopathy and macular edema.
Detailed Description
While various embodiments of the present invention have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the invention. It should be understood that various alternatives to the embodiments of the invention described herein may be employed.
The practice of some embodiments disclosed herein employs, unless otherwise indicated, conventional techniques of immunology, biochemistry, chemistry, molecular biology, microbiology, cell biology, genomics, and recombinant DNA. See, e.g., Sambrook and Green, Molecular Cloning: A Laboratory Manual,4th Edition (2012); the series Current Protocols in Molecular Biology (F.M. Ausubel, et al. eds.); the series Methods In Enzymology (Academic Press, Inc.), PC 2: A Practical Approach (M.J. MacPherson, B.D. hames and G.R. Taylor eds. (1995)), Harlow and Lane, eds. (1988) Antibodies, A Laboratory Manual, and Culture of Animal Cells: A Manual of Basic technology and Specialized Applications,6th Edition (R.I. Freeshness, ed. (2010)).
Definition of
As used in the specification and in the claims, the singular form of "a", "an", and "the" include plural referents unless the context clearly dictates otherwise. For example, the term "immune activator" includes one or more immune activators.
The term "about" or "approximately" means within an acceptable error range for the particular value as determined by one of ordinary skill in the art, which will depend in part on how the value is measured or determined, i.e., the limitations of the measurement system. For example, "about" can mean within 1 or a standard deviation of greater than 1, according to practice in the art. Alternatively, "about" may represent a range of up to 20%, up to 10%, up to 5%, or up to 1% of a given value. Alternatively, particularly for biological systems or processes, the term may denote an order of magnitude, preferably within 5-fold, more preferably within 2-fold, of the value. Where particular values are described in the application and claims, unless otherwise stated, it should be assumed that the term "about" means within an acceptable error range for the particular value.
As used herein, "treatment" refers to an attempt to alter the natural course of disease in a treated individual, and may be a clinical intervention performed for prophylaxis or during the course of clinical pathology. Desirable effects of treatment include, but are not limited to, preventing occurrence or recurrence of disease, alleviating symptoms, reducing any direct or indirect pathological consequences of the disease, preventing metastasis, slowing the rate of disease progression, improving or alleviating the disease state, and/or improving prognosis.
As used herein, the terms "polypeptide," "peptide," and "protein" are used interchangeably herein to refer to a polymer of amino acids of any length. The polymer may be linear, cyclic or branched, it may comprise modified amino acids, and it may be interrupted by non-amino acids. The term also includes amino acid polymers that have been modified, for example, by sulfation, glycosylation, lipidation, acetylation, phosphorylation, iodination, methylation, oxidation, proteolytic processing, phosphorylation, prenylation, racemization, selenization, RNA transfer-mediated addition of amino acids to proteins (e.g., arginylation), ubiquitination, or any other manipulation, such as conjugation to a labeling component. As used herein, the term "amino acid" refers to natural and/or unnatural or synthetic amino acids, including glycine and D or L optical isomers, as well as amino acid analogs and peptidomimetics. A polypeptide or amino acid sequence "derived" from a given protein refers to the origin of the polypeptide. Preferably, the polypeptide has an amino acid sequence which is substantially identical to the amino acid sequence of the polypeptide encoded in the sequence, or a portion thereof, wherein the portion consists of at least 10-20 amino acids or at least 20-30 amino acids or at least 30-50 amino acids, or it can be immunologically identified with the polypeptide encoded in the sequence. The term also includes polypeptides expressed from a given nucleic acid sequence. As used herein, the term "domain" refers to a portion of a protein that is physically or functionally distinct from other portions of the protein or peptide. Physically defined domains include amino acid sequences that are extremely hydrophobic or hydrophilic, such as those that are membrane-bound or cytoplasmic-bound. Domains can also be defined by internal homology, for example, due to gene replication. Functionally defined domains have different biological functions. For example, an antigen binding domain refers to the portion of an antigen binding unit or antibody that binds to an antigen. Functionally defined domains need not be encoded by contiguous amino acid sequences, and functionally defined domains may contain one or more physically defined domains.
As used herein, the term "amino acid" refers to natural and/or unnatural or synthetic amino acids, including but not limited to D or L optical isomers, as well as amino acid analogs and peptidomimetics. The standard single or three letter code is used to refer to amino acids. Amino acids are generally referred to herein by the single and three letter abbreviations commonly known in the art. For example, alanine can be represented by A or Ala.
As used herein, in the context of a polypeptide, a "sequence" is the order of amino acids in a polypeptide in the direction from the amino terminus to the carboxy terminus, wherein residues that are adjacent to each other in the sequence are contiguous in the primary structure of the polypeptide. The sequence may also be a linear sequence of a portion of a polypeptide known to contain additional residues in one or both orientations.
As used herein, "identity," "homology," or "sequence identity" refers to sequence similarity or interchangeability between two or more polynucleotide sequences or between two or more polypeptide sequences. When determining sequence identity, similarity or homology between two different amino acid sequences using programs such as the Emboss Needle or BestFit, default settings may be used, or an appropriate scoring matrix, such as blosum45 or blosum80, may be selected to optimize the identity, similarity or homology score. Preferably, homologous polynucleotides are those that hybridize under stringent conditions as defined herein and have at least 70%, preferably at least 80%, more preferably at least 90%, more preferably 95%, more preferably 97%, more preferably 98% and even more preferably 99% sequence identity to these sequences. Homologous polypeptides preferably have at least 80%, or at least 90%, or at least 95%, or at least 97%, or at least 98% sequence identity, or at least 99% sequence identity, when optimally aligned for sequences of comparable length.
For the purposes of the antigen binding units identified herein, "percent (%) sequence identity" is defined as the percentage of amino acid residues in the query sequence that are identical to the amino acid residues of a second, reference polypeptide sequence, or portion thereof, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignments directed to determining percent amino acid sequence identity can be achieved in various ways within the skill in the art, for example, using publicly available computer software, such as BLAST, BLAST-2, ALIGN, needlet, or megalign (dnastar) software. One skilled in the art can determine suitable parameters for measuring alignment, including any algorithms required to achieve maximum alignment over the full length of the sequences being compared. Percent identity may be measured over the length of the entire defined polypeptide sequence, or may be measured over a shorter length, e.g., over the length of a fragment taken from a larger, defined polypeptide sequence, e.g., a fragment of at least 5, at least 10, at least 15, at least 20, at least 50, at least 100, or at least 200 contiguous residues. These lengths are exemplary only, and it should be understood that any fragment length supported by the sequences shown in the tables, figures, or sequence listing herein can be used to describe the length over which the percent identity can be measured.
The proteins described herein may have one or more modifications relative to a reference sequence. The modification may be deletion, insertion or addition, or substitution of an amino acid residue. "deletion" refers to a change in the amino acid sequence due to the absence of one or more amino acid residues. "insertion" or "addition" refers to an amino acid sequence change that results in the addition of one or more amino acid residues as compared to a reference sequence. "substitution" or "substitution" refers to the replacement of one or more amino acids with a different amino acid. In this context, the mutation of an antigen binding unit relative to a reference sequence can be determined by comparing the antigen binding unit to the reference sequence. Optimal alignment of sequences for comparison can be performed according to any method known in the art.
As used herein, the term "isolated" refers to a separation from cellular and other components to which polynucleotides, peptides, polypeptides, proteins, antibodies or fragments thereof are normally associated in nature. One skilled in the art will appreciate that a non-naturally occurring polynucleotide, peptide, polypeptide, protein, antibody, or fragment thereof need not be "isolated" to distinguish it from its naturally occurring counterpart. In addition, a "concentrated," "isolated," or "diluted" polynucleotide, peptide, polypeptide, protein, antibody, or fragment thereof is distinguishable from its naturally-occurring counterpart in that the concentration or number of molecules per unit volume is greater ("concentrated") or less than from its naturally-occurring counterpart ("isolated"). Enrichment can be measured on an absolute basis, such as the weight of the solution per unit volume, or it can be measured relative to a second, potentially interfering substance present in the source mixture.
The terms "polynucleotide", "nucleic acid", "nucleotide" and "oligonucleotide" are used interchangeably. They refer to polymeric forms of nucleotides of any length (whether deoxyribonucleotides or ribonucleotides) or analogs thereof. The polynucleotide may have any three-dimensional structure and may perform any known or unknown function. The following are non-limiting examples of polynucleotides: coding or non-coding regions of a gene or gene fragment, loci determined from linkage analysis, exons, introns, messenger RNA (mrna), transfer RNA, ribosomal RNA, ribozymes, cDNA, recombinant polynucleotides, branched polynucleotides, plasmids, vectors, isolated DNA of any sequence, isolated RNA of any sequence, nucleic acid probes, primers, oligonucleotides, or synthetic DNA. Polynucleotides may comprise modified nucleotides, such as methylated nucleotides and nucleotide analogs. Modifications to the nucleotide structure, if present, may be imparted before or after assembly of the polymer. The sequence of nucleotides may be interrupted by non-nucleotide components. The polynucleotide may be further modified after polymerization, for example by conjugation with a labeling component.
"recombinant" when applied to a polynucleotide means that the polynucleotide is the product of various combinations of cloning, restriction digestion, and/or ligation steps, as well as other procedures that produce constructs different from those found in nature.
The terms "gene" or "gene fragment" are used interchangeably herein. They refer to polynucleotides comprising at least one open reading frame capable of encoding a particular protein following transcription and translation. The gene or gene fragment may be genomic, cDNA, or synthetic, so long as the polynucleotide contains at least one open reading frame, which may cover the entire coding region or a segment thereof.
The terms "operably linked" or "operatively linked" refer to a juxtaposition wherein the components so described are in a relationship permitting them to function in their intended manner. For example, a promoter sequence is operably linked to a coding sequence if it promotes transcription of the coding sequence.
As used herein, "expression" refers to the process by which a polynucleotide is transcribed into mRNA, and/or the process by which transcribed mRNA (also referred to as "transcript") is subsequently translated into a peptide, polypeptide or protein. The transcripts and the encoded polypeptides are collectively referred to as gene products. If the polynucleotide is derived from genomic DNA, expression may include splicing of mRNA in eukaryotic cells.
As used herein, the term "vector" refers to a nucleic acid delivery vehicle into which a polynucleotide can be inserted. When a vector is capable of expressing a protein encoded by an inserted polynucleotide, the vector is referred to as an expression vector. The vector may be introduced into a host cell by transformation, transduction, or transfection, and the genetic material elements carried thereby are expressed in the host cell. Vectors are well known to those skilled in the art and include, but are not limited to: a plasmid; phagemid; artificial chromosomes such as Yeast Artificial Chromosomes (YACs), Bacterial Artificial Chromosomes (BACs), or artificial chromosomes (PACs) derived from P1; bacteriophage such as lambda phage or M13 phage, animal virus, etc. Animal viruses that may be used as vectors include, but are not limited to, retroviruses (including lentiviruses), adenoviruses, adeno-associated viruses, herpes viruses (e.g., herpes simplex virus), poxviruses, baculoviruses, papilloma viruses, papilloma polyoma vacuolatum viruses (e.g., SV 40). A vector may contain a variety of elements that control expression, including, but not limited to, promoter sequences, transcription initiation sequences, enhancer sequences, selection elements, and reporter genes. In addition, the vector may contain a replication initiation site.
As used herein, the term "antibody" refers to an immunoglobulin molecule that is typically composed of two pairs of polypeptide chains, each pair having one "light" (L) chain and one "heavy" (H) chain. Antibody light chains can be classified as kappa and lambda light chains. Heavy chains can be classified as μ, δ, γ, α or ε, and the antibody isotypes are defined as IgM, IgD, IgG, IgA, and IgE, respectively. Within the light and heavy chains, the variable and constant regions are connected by a "J" region of about 12 or more amino acids, and the heavy chain also contains a "D" region of about 3 or more amino acids. Each heavy chain consists of a heavy chain variable region (VH) and a heavy chain constant region (CH). The heavy chain constant region consists of 3 domains (CH1, CH2, and CH 3). Each light chain consists of a light chain variable region (VL) and a light chain constant region (CL). The light chain constant region consists of one domain CL. The constant region of the antibody may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component of the classical complement system (C1 q). The VH and VL regions can also be subdivided into regions of high denaturation, called Complementarity Determining Regions (CDRs), interspersed with regions that are more conserved, called Framework Regions (FRs). Each VH and VL are composed of, in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4 are composed of 3 CDRs and 4 FRs arranged from amino terminus to carboxy terminus. The variable regions (VH and VL) of each heavy/light chain pair form the antibody binding sites, respectively. The assignment of amino acids to the various regions or domains follows either Kabat Sequences of Proteins of Immunological Interest (National Institutes of Health, Bethesda, Md. (1987and 1991)), or Chothia & Lesk (1987) J.mol.biol.196: 901-; chothia et al (1989) Nature 342: 878-883. The term "antibody" is not limited by any particular method of producing an antibody. For example, it includes recombinant antibodies, monoclonal antibodies and polyclonal antibodies. The antibody may be of a different isotype, for example, an IgG (e.g., IgG1, IgG2, IgG3, or IgG4 subtype), IgA1, IgA2, IgD, IgE, or IgM antibody.
As used herein, the term "antigen-binding fragment" of an antibody refers to a polypeptide comprising a fragment of a full-length antibody that retains the ability to specifically bind to the same antigen to which the full-length antibody binds, and/or competes with the full-length antibody for specific binding to the antigen, which is also referred to as an "antigen-binding portion". See generally, Fundamental Immunology, ch.7(Paul, w., ed., 2 nd edition, Raven Press, n.y. (1989), which is incorporated herein by reference in its entirety for all purposes, antigen-binding fragments of antibodies can be generated by recombinant DNA techniques or by enzymatic or chemical cleavage of intact antibodies in some cases, antigen-binding fragments include Fab, Fab ', F (ab')2, Fd, Fv, dAb and Complementarity Determining Region (CDR) fragments, single chain antibodies (e.g., scFv), chimeric antibodies, diabodies (diabodies), and polypeptides comprising at least a portion of an antibody sufficient to confer specific antigen-binding capability to the polypeptide in some cases, antigen-binding fragments of antibodies are single chain antibodies (e.g., scFv), wherein the VL and VH domains form monovalent molecules through a linker that enables them to be produced as a single polypeptide chain (see, e.g., bird et al, Science 242: 423426 (1988) and Huston et al, Proc. Natl. Acad. Sci. USA 85: 58795883 (1988)). Such scFv molecules can have the general structure: NH 2-VL-linker-VH-COOH or NH 2-VH-linker-VL-COOH. Suitable prior art linkers consist of repeated GGGGS amino acid sequences or variants thereof. For example, a linker having the amino acid sequence (GGGGS)4 may be used, but variants thereof may also be used (Holliger et al (1993), Proc. Natl. Acad. Sci. USA 90: 6444-. Other linkers useful in the context of the present invention are described by Alfthan et al (1995), Protein Eng.8: 725-.
Antigen-binding fragments of antibodies (e.g., antibody fragments described above) can be obtained from a given antibody (e.g., an antibody provided herein) using conventional techniques known to those skilled in the art (e.g., recombinant DNA techniques or enzymatic or chemical fragmentation methods), and the antigen-binding fragments of antibodies are specifically screened for in the same manner as for intact antibodies. Herein, when the term "antibody" is referred to, it includes not only intact antibodies, but also antigen-binding fragments of antibodies, unless the context clearly indicates otherwise.
As used herein, the term "host cell" refers to a cell that can be used for introducing a vector, and includes, but is not limited to, prokaryotic cells such as Escherichia coli or Bacillus subtilis, fungal cells such as yeast cells or Aspergillus, insect cells such as S2 Drosophila cells or Sf9, or animal cells such as fibroblast, CHO cells, COS cells, NSO cells, HeLa cells, BHK cells, HEK293 cells, or human cells.
The terms "antagonist" and "inhibitor" are used interchangeably herein and refer to a molecule capable of inhibiting a biological function of a target protein by inhibiting the activity or expression of the target protein. Thus, the terms "antagonist" and "inhibitor" are defined in the context of the biological effects of the target protein. Although the preferred antagonists herein specifically interact with (e.g., bind to) the target, molecules that inhibit the biological activity of the target protein by interacting with other members of the signaling pathway of which the target protein is a member are also expressly included within this definition.
As used herein, "effective amount" refers to at least the minimum amount required to achieve a measurable improvement or prevention of a particular condition. The effective amount herein may vary with such factors as the disease state, age, sex and weight of the patient. An effective amount is also an amount where the therapeutically beneficial effect exceeds any toxic or adverse effects of the treatment. In the treatment of cancer or tumors, an effective amount of the drug may have the following effects: reducing the number of cancer cells, reducing the size of a tumor, inhibiting infiltration of cancer cells into peripheral organs, inhibiting tumor metastasis, inhibiting tumor growth to some extent, and/or alleviating one or more symptoms associated with the disorder to some extent. An effective amount may be administered in one or more administrations.
As used herein, the terms "recipient," "individual," "subject," "host," and "patient" are used interchangeably herein and refer to any mammalian subject, particularly a human, for which diagnosis, treatment, or therapy is desired.
As used herein, the terms "treat," "treating," and the like are used herein to generally refer to obtaining a desired pharmacological and/or physiological effect. The effect may be prophylactic in terms of completely or partially preventing the disease or symptoms thereof, and/or may be therapeutic in terms of partially or completely stabilizing or curing the disease and/or adverse effects due to the disease. As used herein, "treatment" encompasses any treatment of a disease in a mammal, e.g., mouse, rat, rabbit, pig, primate, including human and other apes, particularly humans, and the term includes: (a) preventing the disease or condition from occurring in a subject who may be predisposed to the disease or condition but has not yet been diagnosed; (b) suppression of disease symptoms; (C) arrest of disease progression; (d) relieving the symptoms of the disease; (e) causing regression of the disease or condition; or any combination thereof. The term "kit" as used herein refers to a combination packaged for co-use or marketed. For example, a kit of the present disclosure can comprise a composition of the present disclosure, and instructions for using the composition or kit. The term "instructions" refers to the instruction insert typically contained in commercial packages of therapeutic products containing information about the indications, uses, dosages, administrations, combination therapies, contraindications and/or warnings concerning the use of such therapeutic products.
Wet macular degeneration
The macula is the central portion of the retina. Macular degeneration, also known as retinal degeneration, is an eye disease involving macular degeneration.
Age-related macular degeneration (AMD) is one of the leading causes of irreversible visual impairment in people over the age of 50. AMD is clinically divided into two types, "dry" and "wet". In the wet form of AMD, new blood vessels form and alter the blood supply to retinal tissue, particularly that underlying the macula. However, new blood vessels are easily damaged, their rupture leading to bleeding and injury of surrounding tissues, scarring of retinal tissue, and rapid loss of vision. The disease progresses rapidly and often leads to blindness. Wet macular degeneration usually begins with a distortion in the center of the visual field, accounting for about 90% of the blindness associated with macular degeneration.
A number of cytokines have been found to play important roles in the regulation of angiogenesis including, but not limited to, Vascular Endothelial Growth Factor (VEGF), VEGF receptor (VEGFR), Platelet Derived Growth Factor (PDGF), Hypoxia Inducible Factor (HIF), angiopoietin (Ang) and other cytokines, mitogen activated protein kinase (MPK), and the like.
Vascular Endothelial Growth Factor (VEGF) is a glycoprotein of 46kDa in size, which is expressed in ocular cells including pigment epithelial cells, pericytes, Vascular endothelial cells, glia and ganglion cells. VEGF is known to be associated with a variety of ocular diseases including, but not limited to, ischemic retinopathy, intraocular neovascularization, age-related macular degeneration (AMD), wet AMD, dry AMD, retinal neovascularization, diabetic macular edema, diabetic retinal ischemia, diabetic retinal edema, proliferative diabetic retinopathy, retinal vein occlusion, central retinal vein occlusion, branched retinal vein occlusion, and the like.
Figure BDA0002594993810000141
Is approvedA medicament for the treatment of wet AMD which is an anti-VEGF antibody that prevents ocular neovascularization and thereby treats wet AMD. Clinical studies have shown that administration is in progress
Figure BDA0002594993810000142
About 95% of patients have improved or stabilized vision. However, this antibody is expensive. Therefore, new therapies are needed for targeting VEGF to treat related ocular diseases.
Recombinant AAV vectors
Adeno-associated virus (AAV) belongs to the parvoviridae family, and is a single-stranded dna (ssdna) virus. The AAV has a genome of about 4.7 kilobases in total, and comprises Inverted Terminal Repeats (ITRs) at both ends of a DNA strand and two Open Reading Frames (ORFs) called rep and cap.
An "AAV Inverted Terminal Repeat (ITR)" sequence is a sequence of about 145 nucleotides found at both ends of the native single-stranded AAV genome. ITRs are symmetric nucleic acid sequences in the adeno-associated viral genome for efficient replication, which can serve as origins of replication for viral DNA synthesis, and are essential structural components of recombinant AAV vectors.
"rep" comprises polynucleotide sequences encoding the four rep proteins, rep78, rep68, rep52, and rep40, required for the life cycle of AAV. "cap" then comprises polynucleotide sequences encoding the AAV capsid proteins VP1, VP2, and VP3, wherein the AAV capsid proteins VP1, VP2, and VP3 are capable of interacting to form a twenty-four-sided symmetrical AAV capsid.
AAV is capable of efficiently infecting dividing as well as non-dividing human cells whose genome can integrate into a single chromosomal site in the host cell genome. Most importantly, while AAV is present in many human bodies, current research suggests that AAV is not associated with any disease. Based on the characteristics of high safety, low immunogenicity, wide host range, capability of mediating long-term stable expression of exogenous genes in animal bodies and the like, AAV has become the most promising vector system in gene therapy.
To date, 13 different AAVs have been identified, AAV1-AAV13, depending on the AAV serotype or the tissue or cell infected. Also, as shown in table 2 below, different AAVs have been developed as advantageous vector systems for transfection of specific cell types. Among the many AAV serotypes, serotype 2(AAV2), which is the most widely studied and used, is capable of infecting retinal epithelium, photoreceptor cells, skeletal muscle, central nerves, and liver cells, etc., and has been used as a vector in a variety of clinical studies.
TABLE 2 AAV serotypes and tissues thereof as vectors for delivery in gene therapy
Figure BDA0002594993810000161
The term "recombinant AAV vector (rAAV vector)" as used herein refers to a polynucleotide vector containing one or more heterologous sequences (i.e., nucleic acid sequences not of AAV origin) flanked by two AAV Inverted Terminal Repeats (ITRs). The rAAV vector can replicate and package into an AAV viral particle when present in a host cell that expresses AAV rep and Cap proteins.
A "recombinant AAV (rAAV) virus" or "rAAV viral particle" refers to an AAV viral particle consisting of at least one AAV capsid protein encapsulating a rAAV vector. The host cells currently used for rAAV viral particle production are all cell types from mammals, such as 293 cells, COS cells, HeLa cells, KB cells, and other mammalian cell lines. rAAV viral particles can be produced in the mammalian cell culture system by providing a rAAV plasmid. However, the yields of most of the above mammalian cell culture systems are difficult to achieve for clinical trials and commercial scale production. For this reason, rAAV viral particle production systems using insect cells such as Sf9 cells have recently been developed. However, in the production of AAV in insect cells, some modifications must be made to obtain the correct stoichiometric ratio of AAV capsid proteins.
Baculoviruses (baculoviruses) belong to the family baculoviridae and are double-stranded circular DNA viruses with a genome size between 90kb and 230 kb. Baculovirus is parasitic specifically in arthropods and is known to infect over 600 insects. In 1983, Smith et al successfully expressed human interferon-beta in Spodoptera frugiperda Sf9 using Autographa Californica Multicapsid Nuclear Polyhedrosis Virus (AcMNPV), and initiated a baculovirus expression system (Mol Cell Biol,1983,3: 2156-. Since then, baculovirus expression systems have been developed and developed, and have become a very widely used eukaryotic expression system. In 2002, Urabe et al have demonstrated that baculovirus-infected Sf9 insect cells can support AAV replication, and have successfully produced rAAV viral particles by co-infecting Sf9 cells with three recombinant baculoviruses carrying the rep gene, Cap gene and ITR core expression elements of AAV, respectively. On this basis, researchers have developed systems that are more suitable for large-scale production of rAAV viral particles.
At present, the method for preparing rAAV virus particles on a large scale by using a baculovirus expression system mainly comprises the following two methods: two baculovirus systems (Two Bac systems) and One baculovirus system (One Bac system) that relies on packaging cell lines. The main process for preparing rAAV viral particles using the two-baculovirus system is to integrate the rep gene and Cap gene of AAV into one baculovirus genome, integrate the ITR core expression element and the target gene of interest into another baculovirus genome, and then use the two recombinant baculoviruses to co-infect host cells to produce rAAV viral particles carrying the target gene. The main process of preparing rAAV virus particle with one packaging cell line dependent baculovirus system includes establishing packaging cell line for inducing expression of rep gene and Cap gene, setting the rep gene and Cap gene expression element separately under the control of late baculovirus gene expression strong promoter PH, and adding hr2 enhancer sequence and AAV rep protein combining sequence in the upstream of the PH promoter. Upon infection with a recombinant baculovirus containing AAV ITRs and a gene of interest, expression of the rep and Cap genes in the packaging cell line is induced, thereby producing rAAV viral particles incorporating the gene of interest.
In some embodiments, the rAAV vector for carrying a gene of interest in a rAAV viral particle may further comprise one or more "expression regulatory elements. The term "expression control element" as used herein refers to a nucleic acid sequence that affects the expression of an operably linked polynucleotide, including polynucleotide sequences that facilitate transcription and translation of heterologous polynucleotides. Expression regulatory elements useful in the present invention include, but are not limited to, promoters, enhancers, intron splicing signals, poly A, Inverted Terminal Repeats (ITRs), and the like.
A "promoter" is a DNA sequence located adjacent to a heterologous polynucleotide sequence encoding a product of interest, which is typically operably linked to adjacent sequences, such as a heterologous polynucleotide. A promoter generally increases the amount of expression of the heterologous polynucleotide compared to the amount expressed in the absence of the promoter.
An "enhancer" is a sequence that enhances promoter activity. Unlike promoters, enhancers do not have promoter activity and generally can function independent of their position relative to the promoter (i.e., upstream or downstream of the promoter). Non-limiting examples of enhancer elements (or portions thereof) that can be used in the present invention include baculovirus enhancers and enhancer elements found in insect cells.
"stuffer" refers to a nucleotide sequence contained within a larger nucleic acid molecule (such as a vector) that is typically used to create a desired separation between two nucleic acid features (such as between a promoter and a coding sequence), or to extend a nucleic acid molecule to have a desired length. The stuffer sequence contains no protein-coding information and may be of unknown/synthetic origin and/or unrelated to other nucleic acid sequences within the larger nucleic acid molecule.
Composition comprising a metal oxide and a metal oxide
In one aspect, the invention provides a composition comprising a first polynucleotide and a second polynucleotide, wherein the first polynucleotide comprises a first sequence operably linked to a first promoter and a second sequence operably linked to a second promoter.
In some embodiments, the first sequence encodes an adeno-associated virus (AAV) cap protein. The cap protein can be any structural protein known in the art that is capable of forming a functional AAV capsid (i.e., capable of packaging DNA and infecting a target cell). In some embodiments, the cap protein comprises VP1, VP2, and VP 3. In some embodiments, the cap protein need not include all of VP1, VP2, VP3, so long as it is capable of producing a functional AAV capsid. In some embodiments, the cap protein comprises VP1 and VP 2. In some embodiments, the cap protein comprises VP1 and VP 3. In some embodiments, the cap protein comprises VP2 and VP 3. In some embodiments, the cap protein comprises VP 1. In some embodiments, the cap protein comprises VP 2. In some embodiments, the cap protein comprises VP 3.
The VP1, VP2, VP3 can be derived from any AAV serotype. In some embodiments, the VP1 may be derived from AAV serotype 1(AAV1), AAV serotype 2(AAV2), AAV serotype 3(AAV3, including serotypes 3A and 3B), AAV serotype 4(AAV4), AAV serotype 5(AAV5), AAV serotype 6(AAV6), AAV serotype 7(AAV7), AAV serotype 8(AAV8), AAV serotype 9(AAV9), AAV serotype 10(AAV10), AAV serotype 11(AAV11), AAV serotype 12(AAV12), AAV serotype 13(AAV13), AAV-Rh10, AAV-Rh74, AAV-2i8, and any other AAV known. In some embodiments, the VP1 is at least 75%, 80%, 85%, 90%, 95% or more identical to wild-type VP1 derived from AAV1, AAV2, AAV3, (including AAV3A and 3B), AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAV13, AAV-Rh10, AAV-Rh74, or AAV-2i 8. In some embodiments, the VP1 has one or more amino acid substitutions, deletions, and/or additions compared to wild-type VP1 derived from AAV1, AAV2, AAV3, (including AAV3A and 3B), AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAV13, AAV-Rh10, AAV-Rh74, or AAV-2i 8.
In some embodiments, the VP2 may be derived from AAV1, AAV2, AAV3, (including AAV3A and 3B), AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAV13, AAV-Rh10, AAV-Rh74, AAV-2i8, and any other AAV known. In some embodiments, the VP2 is at least 75%, 80%, 85%, 90%, 95% or more identical to wild-type VP2 derived from AAV1, AAV2, AAV3, (including AAV3A and 3B), AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAV13, AAV-Rh10, AAV-Rh74, or AAV-2i 8. In some embodiments, the VP2 has one or more amino acid substitutions, deletions, and/or additions compared to wild-type VP2 derived from AAV1, AAV2, AAV3, (including AAV3A and 3B), AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAV13, AAV-Rh10, AAV-Rh74, or AAV-2i 8.
The VP3 may be derived from AAV1, AAV2, AAV3, (including AAV3A and 3B), AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAV13, AAV-Rh10, AAV-Rh74, AAV-2i8, and any other AAV known. In some embodiments, the VP3 is at least 75%, 80%, 85%, 90%, 95% or more identical to wild-type VP3 derived from AAV1, AAV2, AAV3, (including AAV3A and 3B), AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAV13, AAV-Rh10, AAV-Rh74, or AAV-2i 8. In some embodiments, the VP3 has one or more amino acid substitutions, deletions, and/or additions compared to wild-type VP3 derived from AAV1, AAV2, AAV3, (including AAV3A and 3B), AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAV13, AAV-Rh10, AAV-Rh74, or AAV-2i 8.
In some embodiments, the cap comprises VP1, VP2, and/or VP3 derived from an AAV of the same serotype, e.g., the cap can comprise VP1, VP2, and/or VP3 derived from AAV 2. In some embodiments, the cap comprises VP1, VP2, and/or VP3 derived from AAV of different serotypes, e.g., the cap can comprise VP1, VP2, and/or VP3 derived from any one or more of AAV1, AAV2, AAV3, (including AAV3A and 3B), AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAV13, AAV-Rh10, AAV-Rh74, AAV-2i 8. A
In some embodiments, the first sequence encoding the cap is operably linked to a first promoter. The first promoter may be any suitable promoter known in the art capable of driving expression of the cap. In some embodiments, the first promoter can be a tissue specific promoter, a constitutive promoter, a regulatable promoter. In some embodiments, the first promoter may be selected from different sources, for example the first promoter may be a viral promoter, a plant promoter, and a mammalian promoter.
Examples of such first promoters include, but are not limited to, the human Cytomegalovirus (CMV) immediate early enhancer/promoter, SV40 early enhancer/promoter, JC polyomavirus promoter, Myelin Basic Protein (MBP) or Glial Fibrillary Acidic Protein (GFAP) promoter, herpes simplex virus (HSV-1) Latency Associated Promoter (LAP), Rous Sarcoma Virus (RSV) Long Terminal Repeat (LTR) promoter, neuron specific promoter (NSE), platelet-derived growth factor (PDGF) promoter, hsin, melanin-aggregating hormone (MCH) promoter, CBA, matrix metalloprotein promoter (MPP), chicken beta-actin promoter, CAG, MNDU3, PGK, and EF1a promoters.
In some embodiments, the first promoter is a promoter suitable for expression in an insect cell. In some embodiments, the promoter suitable for expression in insect cells includes, but is not limited to, PolH promoter, p10 promoter, alkaline promoter, inducible promoter, E1 promoter, or Δ E1 promoter. In some embodiments, the first promoter is a PolH promoter. In some embodiments, the first promoter is a p10 promoter.
In some embodiments, the 3' end of the first sequence further comprises a polyadenylation sequence or "poly a sequence". In some embodiments, the polyadenylation sequence or "poly a sequence" may range in length from about 1 to 500 bp. In some embodiments, the polyadenylation sequence or "poly a sequence" may be, but is not limited to, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 50, 10, 200, or 500 nucleotides in length.
In some embodiments, the second sequence encodes an AAV rep protein, wherein the rep protein can be a replication protein necessary for replication and packaging of any rAAV vector into a rAAV viral particle. In some embodiments, the rep proteins include rep78, rep68, rep52, and rep 40. In some embodiments, the rep protein need not include all of rep78, rep68, rep52, and rep40, so long as it is capable of allowing replication and packaging of the rAAV vector into rAAV viral particles. In some embodiments, the rep proteins include any three of rep78, rep68, rep52, and rep 40. In some embodiments, the rep proteins include any two of rep78, rep68, rep52, and rep 40. In some embodiments, the rep protein comprises any one of rep78, rep68, rep52, and rep 40. In some embodiments, the rep proteins include rep78 and rep 52. In some embodiments, the rep proteins include rep78 and rep 40. In some embodiments, the rep proteins include rep68 and rep 52. In some embodiments, the rep proteins include rep68 and rep 40.
The rep78, rep68, rep52, and rep40 can be derived from any AAV serotype. In some embodiments, the rep78 can be derived from AAV1, AAV2, AAV3, (including AAV3A and 3B), AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAV13, AAV-Rh10, AAV-Rh74, AAV-2i8, and any other AAV known. In some embodiments, the rep78 is at least 75%, 80%, 85%, 90%, 95% or more identical to a wild-type rep78 derived from AAV1, AAV2, AAV3, (including AAV3A and 3B), AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAV13, AAV-Rh10, AAV-Rh74, or AAV-2i 8. In some embodiments, the rep78 has a substitution, deletion, and/or addition of one or more amino acids compared to wild-type rep78 derived from AAV1, AAV2, AAV3, (including AAV3A and 3B), AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAV13, AAV-Rh10, AAV-Rh74, or AAV-2i 8.
In some embodiments, the rep68 can be derived from AAV1, AAV2, AAV3, (including AAV3A and 3B), AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAV13, AAV-Rh10, AAV-Rh74, AAV-2i8, and any other AAV known. In some embodiments, the rep68 is at least 75%, 80%, 85%, 90%, 95% or more identical to a wild-type rep68 derived from AAV1, AAV2, AAV3, (including AAV3A and 3B), AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAV13, AAV-Rh10, AAV-Rh74, or AAV-2i 8. In some embodiments, the rep68 has a substitution, deletion, and/or addition of one or more amino acids compared to wild-type rep68 derived from AAV1, AAV2, AAV3, (including AAV3A and 3B), AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAV13, AAV-Rh10, AAV-Rh74, or AAV-2i 8.
In some embodiments, the rep52 can be derived from AAV1, AAV2, AAV3, (including AAV3A and 3B), AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAV13, AAV-Rh10, AAV-Rh74, AAV-2i8, and any other AAV known. In some embodiments, the rep52 is at least 75%, 80%, 85%, 90%, 95% or more identical to a wild-type rep52 derived from AAV1, AAV2, AAV3, (including AAV3A and 3B), AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAV13, AAV-Rh10, AAV-Rh74, or AAV-2i 8. In some embodiments, the rep52 has a substitution, deletion, and/or addition of one or more amino acids compared to wild-type rep52 derived from AAV1, AAV2, AAV3, (including AAV3A and 3B), AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAV13, AAV-Rh10, AAV-Rh74, or AAV-2i 8.
In some embodiments, the rep40 can be derived from AAV1, AAV2, AAV3, (including AAV3A and 3B), AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAV13, AAV-Rh10, AAV-Rh74, AAV-2i8, and any other AAV known. In some embodiments, the rep40 is at least 75%, 80%, 85%, 90%, 95% or more identical to a wild-type rep52 derived from AAV1, AAV2, AAV3, (including AAV3A and 3B), AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAV13, AAV-Rh10, AAV-Rh74, or AAV-2i 8. In some embodiments, the rep40 has a substitution, deletion, and/or addition of one or more amino acids compared to wild-type rep52 derived from AAV1, AAV2, AAV3, (including AAV3A and 3B), AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAV13, AAV-Rh10, AAV-Rh74, or AAV-2i 8.
In some embodiments, the rep comprises rep78, rep68, rep52, and/or rep40 derived from an AAV of the same serotype, e.g., the rep can comprise rep78, rep68, rep52, and/or rep40 derived from AAV 2. In some embodiments, the rep comprises rep78, rep68, rep52, and/or rep40 derived from AAV of different serotypes, e.g., the rep can comprise rep78, rep68, rep52, and/or rep40 derived from any one or more of AAV1, AAV2, AAV3, (including AAV3A and 3B), AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAV13, AAV-Rh10, AAV-Rh74, AAV-2i8, and any other AAV known.
In some embodiments, the second sequence encoding the rep protein is operably linked to a second promoter. The second promoter may be any suitable promoter known in the art capable of driving expression of the cap. In some embodiments, the second promoter can be a tissue specific promoter, a constitutive promoter, a regulatable promoter. In some embodiments, the second promoter may be selected from different sources, for example the second promoter may be a viral promoter, a plant promoter, and a mammalian promoter.
Examples of such second promoters include, but are not limited to, the human Cytomegalovirus (CMV) immediate early enhancer/promoter, SV40 early enhancer/promoter, JC polyomavirus promoter, Myelin Basic Protein (MBP) or Glial Fibrillary Acidic Protein (GFAP) promoter, herpes simplex virus (HSV-1) Latency Associated Promoter (LAP), Rous Sarcoma Virus (RSV) Long Terminal Repeat (LTR) promoter, neuron specific promoter (NSE), platelet-derived growth factor (PDGF) promoter, hsin, melanin-aggregating hormone (MCH) promoter, CBA, matrix metalloprotein promoter (MPP), chicken beta-actin promoter, CAG, MNDU3, PGK, and EF1a promoters.
In some embodiments, the second promoter is a promoter suitable for expression in an insect cell. In some embodiments, the promoter suitable for expression in insect cells includes, but is not limited to, PolH promoter, p10 promoter, alkaline promoter, inducible promoter, E1 promoter, or Δ E1 promoter. In some embodiments, the second promoter is a PolH promoter. In some embodiments, the second promoter is a p10 promoter.
In some embodiments, the 3' end of the second sequence further comprises a polyadenylation sequence or "poly a sequence". In some embodiments, the polyadenylation sequence or "poly a sequence" may range in length from about 1 to 500 bp. In some embodiments, the polyadenylation sequence or "poly a sequence" may be, but is not limited to, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 50, 10, 200, or 500 nucleotides in length.
In some embodiments, the cap and the rep may be derived from the same AAV serotype. For example, the cap and rep may be derived from the same AAV1, AAV2, AAV3, (including AAV3A and 3B), AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAV13, AAV-Rh10, AAV-Rh74, AAV-2i8, or any other AAV known.
In some embodiments, the cap and the rep may be derived from different AAV serotypes, e.g., the cap and the rep may be derived from AAV1, AAV2, AAV3, (including AAV3A and 3B), AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAV13, AAV-Rh10, AAV-Rh74, AAV-2i8, or any other AAV known. For example, in some embodiments, the cap can be derived from AAV2 and the rep is derived from AAV 5.
In some embodiments, the first promoter and the second promoter may be the same promoter. For example, the first promoter and the second promoter are any one selected from the group consisting of PolH promoter, p10 promoter, alkaline promoter, inducible promoter, E1 promoter, or Δ E1 promoter. For example, in some embodiments, the first promoter and the second promoter are both PolH promoters. In some embodiments, the first promoter and the second promoter are both p10 promoters.
In some embodiments, the first promoter and the second promoter may be different promoters. For example, the first promoter and the second promoter may be any two promoters selected from PolH promoter, p10 promoter, alkaline promoter, inducible promoter, E1 promoter, or Δ E1 promoter, respectively. For example, in some embodiments, said first promoter is a PolH promoter and said second promoter is a p10 promoter. In some embodiments, said first promoter is a p10 promoter and said second promoter is a PolH promoter.
In some embodiments, the first sequence and the second sequence are linked by a sequence encoding a linker. In some embodiments, the linker is a cleavable linker. In some embodiments, the cleavable linker is a sequence comprising a 2A peptide. In some embodiments, the 2A peptide may be a 2A peptide selected from a 2A peptide derived from the genus aphtovirus or cardiovirus, such as a 2A peptide derived from Foot and Mouth Disease Virus (FMDV), Equine Rhinitis A Virus (ERAV), thoseasgagnna virus (TaV), or porcine teschovirus (PTV-1). In some embodiments, the sequence encoding the linker further comprises a promoter sequence. In some embodiments, the promoter is an FMDV promoter.
In some embodiments, the second polynucleotide in the compositions of the invention comprises a third sequence operably linked to a CMV, CAG, MNDU3, PGK, EF1a promoter, or an eye-specific promoter, wherein the third sequence encodes a VEGF inhibitor.
The VEGF inhibitor can be any polypeptide or protein that is capable of inhibiting the activity or expression of a VEGF protein and thus its biological function. In some embodiments, the VEGF inhibitor is an anti-VEGF antibody or an antigen-binding fragment thereof. In some embodiments, the antigen-binding fragments include, but are not limited to, Fab ', F (ab')2, Fd, Fv, dAb, and Complementarity Determining Region (CDR) fragments, single chain antibodies (scFv), chimeric antibodies, and diabodies (diabodies). In some embodiments, the anti-VEGF inhibitor is selected from ranibizumab (ranibizumab), bevacizumab (bevacizumab).
In some embodiments, the VEGF inhibitor comprises SEQ ID NO: 1.2 and 3 or a sequence having at least 90% homology thereto. In some embodiments, the VEGF inhibitor comprises SEQ ID NO: 1 or a sequence having at least 90% homology thereto. In some embodiments, the VEGF inhibitor comprises SEQ ID NO: 2 or a sequence having at least 90% homology thereto. In some embodiments, the VEGF inhibitor comprises SEQ ID NO: 3 or a sequence having at least 90% homology thereto.
In some embodiments, the VEGF inhibitor comprises SEQ ID NO: 4.5 and 6 or a sequence having at least 90% homology thereto. In some embodiments, the VEGF inhibitor comprises SEQ ID NO: 4 or a sequence having at least 90% homology thereto. In some embodiments, the VEGF inhibitor comprises SEQ ID NO: 5 or a sequence having at least 90% homology thereto. In some embodiments, the VEGF inhibitor comprises SEQ ID NO: 6 or a sequence having at least 90% homology thereto.
In some embodiments, the VEGF inhibitor comprises SEQ ID NO: 7, or a sequence having at least 70%, 80%, 90%, 95%, 96%, 97%, 98%, 99% homology thereto. In some embodiments, the VEGF inhibitor comprises SEQ ID NO: 8, or a sequence having at least 70%, 80%, 90%, 95%, 96%, 97%, 98%, 99% homology thereto. In some embodiments, the VEGF inhibitor comprises SEQ ID NO: 7 or a sequence identical to SEQ ID NO: 7, and SEQ ID NO: 8 or a sequence identical to SEQ ID NO: 8, at least 70%, 80%, 90%, 95%, 96%, 97%, 98%, 99% homologous.
In some embodiments, the third sequence is operably linked to a CMV, CAG, MNDU3, PGK, EF1a promoter, or an eye-specific promoter. In some embodiments, the eye-specific promoter is a Retinal Pigment Epithelium (RPE) cell-specific promoter. Examples of the RPE cell-specific promoter include, but are not limited to, an RPE 65 gene promoter, a human retinal binding protein (CRALBP) gene promoter, a murine 11-cis Retinol Dehydrogenase (RDH) gene promoter, a rhodopsin kinase promoter (rhodopsin kinase promoter), a tissue inhibitor of metalloproteinase 3 (Timp3) promoter, a photoreceptor retinoid binding protein promoter, and a vitroplastic macular dystrophy 2 (vitroplastic mammalian dynamics 2) promoter, an inter-photoreceptor retinoid-binding protein (IRBP) promoter.
In some embodiments, the second polynucleotide further comprises other regulatory sequences including, but not limited to, Inverted Terminal Repeats (ITRs), enhancers, splicing signals, polyadenylation signals, stuffer sequences, terminators, protein degradation signals, internal ribosome entry elements (IRES), 2A sequences and the like.
In some embodiments, the second polynucleotide further comprises an enhancer region. In some embodiments, the enhancer region includes the SV40 enhancer, the immediate early cytomegalovirus enhancer, the IRBP enhancer, an enhancer derived from an immunoglobulin gene. In some embodiments, the enhancer region is located upstream of the CMV, CAG, MNDU3, PGK, EF1a promoter. In some embodiments, the enhancer is upstream of the eye-specific promoter. In some embodiments, the enhancer region is downstream of the CMV, CAG, MNDU3, PGK, EF1a promoter. In some embodiments, the enhancer is located downstream of the eye-specific promoter.
In some embodiments, the second polynucleotide further comprises an Inverted Terminal Repeat (ITR). In some embodiments, the second polynucleotide comprises at least one Inverted Terminal Repeat (ITR). In some embodiments, the second polynucleotide comprises two Inverted Terminal Repeats (ITRs). In some embodiments, the two ITRs are the same as each other. In some embodiments, the two ITRs are different from each other. In some embodiments, the Inverted Terminal Repeat (ITR) is an ITR derived from AAV. In some embodiments, the ITRs may be derived from the ITRs of AAV1, AAV2, AAV3, (including AAV3A and 3B), AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAV13, AAV-Rh10, AAV-Rh74, AAV-2i8, and any other AAV known. In some embodiments, the ITRs have a mutation, insertion or deletion of one or more bases compared to wild-type ITRs derived from AAV1, AAV2, AAV3, (including AAV3A and 3B), AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV12, AAV13, AAV-Rh10, AAV-Rh74, AAV-2i8, and any other AAV known, so long as they retain the desired terminal repeat function, such as replication of the gene of interest, packaging and/or integration of the viral particle, and the like.
In some embodiments, the second polynucleotide further comprises one or more stuffer sequences. In some embodiments, the stuffer sequence is upstream of the CMV, CAG, MNDU3, PGK, EF1a promoter sequence. In some embodiments, the stuffer sequence is downstream of the CMV, CAG, MNDU3, PGK, EF1a promoter sequence. In some embodiments, the stuffer sequence is upstream of the eye-specific promoter. In some embodiments, the stuffer sequence is located downstream of the eye-specific promoter. In some embodiments, the stuffer sequence is located 5 'of the 5' ITR sequence. In some embodiments, the stuffer sequence is located 3 'of the 5' ITR sequence. In some embodiments, the stuffer sequence is located 5 'of the 5' ITR sequence. In some embodiments, the stuffer sequence is located 5 'of the 3' ITR sequence. In some embodiments, the stuffer sequence is located 3 'of the 3' ITR sequence.
In some embodiments, the length of the stuffer sequence may be about 0.1kb to 5kb, such as, but not limited to, 0.1kb, 0.2kb, 0.3kb, 0.4kb, 0.5kb, 0.6kb, 0.7kb, 0.8kb, 0.9kb, 1kb, 1.1kb, 1.2kb, 1.3kb, 1.4kb, 1.5kb, 1.6kb, 1.7kb, 1.8kb, 1.9kb, 2kb, 2.1kb, 2.2kb, 2.3kb, 2.4kb, 2.5kb, 2.6kb, 2.7kb, 2.8kb, 2.9kb, 3kb, 3.1kb, 3.2kb, 3.3kb, 3.4kb, 3.5kb, 3.6kb, 3.7, 3.8kb, 3.9kb, 4.0.0 kb, 4.7kb, 4.4kb, 4.5kb, 4.6kb, 4.7kb, 4.8kb, 4.9kb, 4.4kb, or 4.4 kb.
In some embodiments, the second polynucleotide further comprises a fourth sequence encoding another therapeutic protein. In some embodiments, the therapeutic protein is selected from the group consisting of: VEGF inhibitors, PDGF inhibitors, integrin inhibitors, mTOR inhibitors, angiogenin inhibitors, and TGF β inhibitors.
In some embodiments, the fourth sequence is linked to the third sequence by a sequence encoding a linker. In some embodiments, the linker is a cleavable linker. In some embodiments, the cleavable linker comprises the sequence of the 2A peptide. In some embodiments, the 2A peptide may be a 2A peptide selected from a 2A peptide derived from the genus aphtovirus or cardiovirus, such as a 2A peptide derived from Foot and Mouth Disease Virus (FMDV), Equine Rhinitis A Virus (ERAV), thoseasgagnna virus (TaV), or porcine teschovirus (PTV-1).
Recombinant AAV viral particles
In another aspect, the present invention provides a recombinant adeno-associated virus (rAAV) particle prepared by transfecting a composition of the invention as described above into an insect cell. In some embodiments, the insect cell is an Sf9 cell.
In some embodiments, the compositions of the invention can be delivered into the insect cells by any method known in the art. In some embodiments, the methods include, but are not limited to, electroporation, calcium phosphate precipitation, liposome-mediated, and the like. In some embodiments, the composition is stably transfected into the insect cell. In some embodiments, the composition is transiently transfected into the insect cell. In some embodiments, the insect cell is used to produce the rAAV viral particle.
If desired, the rAAV viral particles can be isolated and purified from the insect cells according to conventional methods known to those skilled in the art. For example, the rAAV viral particles can be purified using centrifugation, HPLC, Hydrophobic Interaction Chromatography (HIC), anion exchange chromatography, cation exchange chromatography, size exclusion chromatography, ultrafiltration, gel electrophoresis, affinity chromatography, and/or other purification techniques.
System for controlling a power supply
In another aspect, the invention provides a system for treating an ocular disease in a subject in need thereof, comprising a rAAV particle of the invention and a pharmaceutically acceptable carrier or excipient.
As used herein, "pharmaceutically or therapeutically acceptable carrier or excipient" refers to a carrier medium that does not interfere with the effectiveness of the biological activity of the active ingredient and is not toxic to the host or patient. The type of carrier employed in the pharmaceutical formulation will depend on the method of administering the therapeutic compound. Many methods of preparing pharmaceutical compositions for various routes of administration are known in the art. By "pharmaceutically acceptable ophthalmic vector" is meant a pharmaceutically acceptable carrier or excipient that can be used to deliver the rAAV viral particles of the invention directly or indirectly to, on, or near the eye.
In some embodiments of the invention, the system is prepared by dissolving the rAAV viral particles of the invention in an appropriate solvent. Suitable solvents include, but are not limited to, water, saline solution (e.g., NaCl), buffered solutions, ointments, gels, or other solvents. In certain embodiments, the solvent is sterile.
The aqueous solution and diluent for suspension used in preparing the eye drop may include distilled water, physiological saline, and the like. Various additives may be contained in the eye drops, the ophthalmic gel and/or the ophthalmic ointment as needed. These additives may include additional ingredients, additives or carriers suitable for contact with or use around the eye without undue toxicity, incompatibility, instability, irritation, allergic response, and the like. Additives such as solvents, bases, solubilizing agents, suspending agents, thickening agents, emulsifiers, stabilizers, buffers, isotonicity adjusting agents, pH adjusting agents, chelating agents, soothing agents, preservatives, flavoring agents, coloring agents, excipients, binders, lubricants, surfactants, absorption promoters, dispersing agents, preservatives, solubilizers and the like may be added to the formulation as appropriate.
For example, eye drops can be formulated by dissolving rAAV viral particles in sterile water with a surfactant dissolved therein, and optionally adding appropriate pharmaceutical additives such as preservatives, stabilizers, buffers, antioxidants, and viscosity modifiers.
For example, a buffer is added to keep the pH constant, and the buffer may include a pharmaceutically acceptable buffer such as a borate buffer, a citrate buffer, a tartrate buffer, a phosphate buffer, an acetate buffer, or a Tris-HCl buffer (including Tris (hydroxymethyl) aminomethane and HCl).
In addition to the buffer, an isotonic agent may be added to the eye drops to prepare a formulation isotonic with tear fluid. Isotonic agents include, but are not limited to, sugars such as dextrose, glucose, sucrose, and fructose; sugar alcohols such as mannitol and sorbitol; polyols such as glycerol, polyethylene glycol and propylene glycol; and salts, such as sodium chloride, sodium citrate, benzalkonium chloride, ephedrine hydrochloride, potassium chloride, procaine hydrochloride, chloramphenicol, and sodium succinate. The isotonic agent is added in an amount such that the osmotic pressure of the eye drops is equal to the osmotic pressure of tears.
Preservatives may be added to maintain the integrity of the eye drops and/or ophthalmic ointment. Examples of preservatives include, but are not limited to, sorbic acid, benzalkonium chloride, benzyl dodecyl dimethyl ammonium bromide (benzodiciclium bromide), parabens, chlorobutanol, benzyl alcohol, phenethyl alcohol, edetate disodium, sorbic acid, polyquaternium-1, or other agents known to those skilled in the art.
In some embodiments, thickening agents are used to increase the viscosity of ophthalmic formulations such as eye drops, ophthalmic gels, and/or ophthalmic ointments. Thickeners that may be used include, but are not limited to, glycerin, polyethylene glycol, carboxymethyl cellulose, and carboxyvinyl polymers.
In addition to the foregoing, in some embodiments it may be desirable to use additional agents, including but not limited to stabilizers such as sodium sulfite, sodium carbonate, and propylene glycol; antioxidants, such as ascorbic acid, sodium ascorbate, Butylated Hydroxytoluene (BHT), Butylated Hydroxyanisole (BHA), tocopherol, sodium thiosulfate; and/or chelating agents such as ethylenediaminetetraacetic acid (EDTA), ethylene glycol-bis- (2-aminoethyl) -N, N-tetraacetic acid (EGTA), and sodium citrate.
The eye drops, ophthalmic gel and/or ophthalmic ointment may be prepared by aseptic manipulation or may alternatively be sterilized at a suitable stage of preparation. For example, sterile pharmaceutical compositions can be prepared by aseptically mixing sterile ingredients. Alternatively, the sterile pharmaceutical composition may be prepared by first mixing the ingredients and then sterilizing the final formulation. Sterilization methods may include, but are not limited to, heat sterilization, radiation, and filtration.
Ophthalmic ointments (eye ointments) may be prepared aseptically by mixing the active ingredient into a base for preparing the eye ointment, followed by formulating into a pharmaceutical preparation using any method known in the art. Examples of typical bases for eye ointments are petrolatum, jelene 50, plastibase and polyethylene glycol. In addition, a surfactant may be added to increase hydrophilicity.
A variety of effective methods for controlled release of the active agent may be used. See, e.g., Wagh V.D., Inamdar B., Samanta M.K., Polymers used in annular devices and drug delivery systems. Asian J Pharm 2,2008,12-17 and references cited therein, the contents of which are incorporated herein by reference. Polymers (e.g., cellulose derivatives such as hydroxypropylmethyl cellulose (HPMC) and hydroxypropyl cellulose (HPC), poly (acrylic acid) (PAA), polyacrylates, cyclodextrins and natural gums, Polyorthoesters (POE) and mucoadhesive polymers) are particularly contemplated; semi-solids, such as gels, films, and other inserts; resins, such as ion exchange resins; iontophoretic delivery; and colloidal particles such as microspheres and nanoparticles.
The rAAV viral particles of the invention may also be provided in combination with other therapeutic agents. In some embodiments, the compounds of the present invention may be co-formulated with other active agents including, but not limited to, anti-infective agents, antibiotics, antiviral agents, antifungal agents, antiprotozoal agents, anti-inflammatory agents, antiallergic agents (including antihistamines), artificial tear vasoconstrictors, vasodilators, local anesthetics, analgesics, ocular hypotensive agents, immunomodulators, antioxidants, vitamins and minerals, enzyme inhibitors or alternative proteases and peptidases, cytokine inhibitors, and the like.
In various embodiments, the compounds of the present invention may also be provided in combination with an ocular therapeutic agent selected from the group consisting of anagla (Acular) (ketorolac tromethamine ophthalmic solution) 0.5%, Acuvail (ketorolac tromethamine), AK-Con-a (naphazoline ophthalmic), Akten (lidocaine hydrochloride), Alamast, Alphagan (brimonidine), Alrex, Astepro (nitrogen hydrochloride)
Figure BDA0002594993810000361
Nasal sprays of statin), Azasite (azithromycin), Bepreve (Betasastin ophthalmic solution besilate), Besivance (Bexifloxacin ophthalmic suspension), Betaxon, BSS sterile lavage fluid, Cosopt, Durezol (difluprednate), Lotemax, Lucentis (ranibizumab), Lumigan (bimatoprost ophthalmic solution), Macugen (Pergatatinib), Ocufox (Oxofloxacin ophthalmic solution) 0.3%, OcuHist, Ozurdex (dexamethasone), Quixin (levofloxacin), Rescula (unoprostone isopropyl ophthalmic solution) 0.15%, Restasis (cyclosporine ophthalmic emulsion), Vissagen tablets, Travatan (travoprost ophthalmic solution), Valcyve (valganciclovir hydrochloride), trifluorothymidine (Vitroptide), Visstive (Cidovudine), Vituporisin (Vitrovirapine), Viertsuvin implant (Tortusin), Vitrovafloxacin (Zloxacin), Zygsin (Zygorfloxacin), Zygorfloxacin (Zygen ophthalmic solution), Zygorubicin (Zygoxin) and Zygorubicin (Zygorubicin), Zygorubicin) as, Atropine, flurbiprofen, physostine (physiostatin), parimine (Azopt), gentamicin, pilocarpine (Proparacaine), bacitracin, hypromellose eye fluid (Goniosol), polymyxin B, povidone iodine (Betadine), gramicidin, prednisolone, betaxolol, Humorsol, Proparacaine, betaxolol eye fluid (Betoptic), hylatin, Propine, brinzolamide, hypertonic NaCl, Puralube, BSS, indocyanine Green (indocynine Green), Rose Bengal (Rose Bengal), carbachol, itraconazole, sodium hyaluronate, cefazolin, latanoprost, suprofen, carvacrol (Celluvisc), mannitol, oxytetracycline, chloramphenicol, methazolamide, tiazolol, ciloxacin, miconazole, ciprofloxacin, lotoxin, cotrion, muramyl 128 tro, muraglicol, muramyl, and soFloxuridine, Demecarium, neomycin, tropicamide, dexamethasone, methazolamide (Neptazane), Trusopt, dipivefrin, Ocuflox, vidarabine, dorzolamide, ofloxacin, Vira-a, epinephrine, oxytetracycline, trifluorothymidine, fluorescein, phenylephrine, and alitan (Xalatan).
Examples of drugs may include anti-angiogenic agents such as angiostatin (angiostatin), anecortave acetate, thrombospondin, VEGF receptor tyrosine kinase inhibitors and anti-vascular endothelial growth factor (anti-VEGF) drugs such as ranibizumab (ranibizumab) and bevacizumab (bevacizumab), pegaptanib (pegaptanib), sunitinib (sunitinib) and sorafenib (sorafenib) and any of a variety of known small molecules and transcriptional inhibitors with anti-angiogenic effects; various classes of known ophthalmic drugs include: glaucoma agents, such as adrenergic antagonists, including, for example, beta-blockers such as acebutolol (acetobutolol), atenolol (atenolol), bisoprolol (bisoprolol), carvedilol (carvedilol), esmolol (asmolol), labetalol (labetalol), nadolol (nadolol), penbutolol (penbutolol), pindolol (pindolol), propranolol (propranolol), metiralol (metiranolol), betaxolol (betaxolol), carteolol (carteolol), levobetaxolol (levobetabetolol), levobunolol (levobunolol), and timolol (levobunolol); adrenergic agonists or sympathomimetics such as epinephrine, dipivefrin, clonidine, apraclonidine, and brimonidine; parasympathomimetic or cholinergic receptor agonists such as pilocarpine (pilocarpine), carbachol (carbachol), phosphorylcholine iodide (phospholine iodide), and physostigmine (physostigmine), salicylic acid, acetylcholine chloride, escein (eserine), diisopropylfluorophosphate, and dimeglumine (demecainium bromide); muscarinic species; carbonic anhydrase inhibitor agents, including topical and/or systemic agents, such as acetazolamide (acetozolamide), brinzolamide (brinzolamide), dorzolamide (dorzolamide), and methazolamide (methazolamide), ethoxzolamide (ethoxzolamide), acetazolamide (diamox), and dichlorphenamide (dichlorphenamide); cycloplegic mydriatic-cyclopegiants such as atropine, cyclopentolate, succinylcholine, homatropine, phenylephrine, scopolamine, and tropicamide; prostanoids such as prostaglandin F2 a, anti-prostaglandins, prostaglandin precursors, or prostaglandin analog agents such as bimatoprost, latanoprost, travoprost, and unoprostone.
Additional examples of medicaments may also include anti-inflammatory agents, including, for example, glucocorticoids and corticosteroids such as betamethasone, cortisone, dexamethasone 21-phosphate, methylprednisolone, prednisolone 21-phosphate, prednisolone acetate, prednisolone, fluoromethalone, loteprednol, medrysone, fluocinonide, triamcinolone acetonide, triamcinolone acetate, beclomethasone, budesonide, flunisolide, fluoromethalone, fluticasone, fludrocortisone, hydrocortisone acetate, loteprednol, rimexolone, and nonsteroidal anti-inflammatory drugs including, for example, aspirin, diclofen, flurbiprofen, ibuprofen, bromfenac, nepafenac, and ketorolac, salicylates, indomethacin, naproxen (naxopren), piroxicam, and nabumetone diflunisal (nabumeiflol), Etodolac, fenoprofen, flurbiprofen, indomethacin, ketoprofen (ketoprofen), meclofenamate, mefenamic acid, meloxicam, nabumetone, oxaprozin (oxaprozin), piroxicam, salsalate, sulindac, and tolmetin; COX-2 inhibitors such as celecoxib, rofecoxib, and valdecoxib; anti-infective or antimicrobial agents such as antibiotics including, for example, tetracycline, chlortetracycline, bacitracin, neomycin, polymyxin, gramicin, cephalexin, oxytetracycline, chloramphenicol, rifampin, ciprofloxacin, tobramycin, gentamicin, erythromycin, penicillin, sulfonamides, sulfadiazine, sulfacetamide, sulfamethylthiadiazole, sulfaisoxazole, nitrofurazone (nitrofurazone), sodium propionate, aminoglycosides such as gentamicin, tobramycin, amikacin, and streptomycin; fluoroquinolones such as ciprofloxacin, gatifloxacin, levofloxacin, moxifloxacin, norfloxacin, ofloxacin; bacitracin, erythromycin, fusidic acid, neomycin, polymyxin B, gramicidin, trimethoprim, and sulfacetamide; antifungal agents such as amphotericin B, caspofungin, clotrimazole, fluconazole, itraconazole, ketoconazole, voriconazole, terbinafine, nystatin, and miconazole; antimalarial agents such as chloroquine, atovaquone, mefloquine, primaquine, quinidine, and quinine; antimycobacterial agents such as ethambutol, isoniazid, pyrazinamide, rifampin and rifabutin; antiparasitic agents such as albendazole, mebendazole, thiobendazole, metronidazole, pyrantel, atovaquone, diiodoquinol, ivermectin, paromomycin, praziquantel, and trimetrexate.
Method
In another aspect, the present application provides a method for treating an ocular disease comprising administering a therapeutically effective amount of the system of the present invention to a subject in need thereof.
In some embodiments, the system may be administered to the subject by any suitable method known in the art. In some embodiments, the system may be administered topically to the eye, e.g., subconjunctival, retrobulbar, periocular, subretinal, suprachoroidal, or intraocular administration.
In some embodiments, the ocular disease includes, but is not limited to, age-related macular degeneration (AMD), wet AMD, dry AMD, retinal neovascularization, choroidal neovascularization, and diabetic retinopathy, proliferative diabetic retinopathy, retinal vein occlusion, central retinal vein occlusion, branch retinal vein occlusion, diabetic macular edema, diabetic retinal ischemia, ischemic retinopathy, and diabetic retinal edema.
In some embodiments, the system comprising the rAAV viral particle is provided in a therapeutically effective amount that achieves a desired biological effect at a medically acceptable level of toxicity. The dosage may vary depending on the route of administration and the severity of the disease. The dosage may also be adjusted according to the weight, age, sex and/or extent of symptoms of each patient to be treated. The precise dosage and route of administration will ultimately be at the discretion of the attendant physician or veterinarian. It will be appreciated that routine variations in dosage may be required depending on the age and weight of the patient and the severity of the condition to be treated.
In some embodiments, the therapeutically effective amount is generally about 1X 105-1×1013A rAAV viral particle. In some embodiments, the therapeutically effective amount is generally about 1X 106-1×1012A rAAV viral particle. In some embodiments, the therapeutically effective amount is generally about 1X 107-1×1012A rAAV viral particle. In some embodiments, the therapeutically effective amount is generally about 1X 108-1×1012A rAAV viral particle. In some embodiments, the therapeutically effective amount is generally about 1X 109-1×1012A rAAV viral particle. In some embodiments, the therapeutically effective amount is generally about 1X 1010-1×1012A rAAV viral particle.
In some embodiments, the delivered volume is about 0.01mL to 1 mL. In some embodiments, the delivered volume is about 0.05mL to 1 mL. In some embodiments, the delivered volume is about 0.1mL to 1 mL. In some embodiments, the delivered volume is about 0.5mL to 1 mL. In some embodiments, the delivered volume is about 0.1mL to about 0.5 mL. In some embodiments, the delivered volume is about 0.01mL to about 0.5 mL. In some embodiments, the delivered volume is about 0.05mL to about 0.5 mL. In some embodiments, the delivered volume is about 0.05mL to 1 mL.
In some embodiments, the dosing frequency may be at least once daily administration, including 2, 3, 4, or 5 times daily. In some embodiments, the treatment can last for 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days, 16 days, 17 days, 18 days, 19 days, 20 days, 21 days, 22 days, 23 days, 24 days, 25 days, 26 days, 27 days, 28 days, 29 days, 30 days, 31 days, 32 days, 33 days, 34 days, 35 days, 36 days, 37 days, 38 days, 39 days, 40 days, 41 days, 42 days, 43 days, 44 days, 45 days, 46 days, 47 days, 48 days, 49 days, 50 days, 60 days, 70 days, 80 days, 90 days, 100 days, 150 days, 200 days, 250 days, 300 days, 400 days, 500 days, 750 days, 1000 days, or more than 1000 days.
Reagent kit
In another aspect, the invention provides a kit for treating an ocular disease comprising the system of the invention and instructions. In some embodiments, the instructions are for a method of administering the system to treat an ocular disease.
In some embodiments, the kit further comprises a container. In some embodiments, the container is configured to deliver a system described herein. In some embodiments, the container comprises a vial, dropper, bottle, tube, and syringe. In some embodiments, the container is a dropper for applying the system. In some embodiments, the container is a syringe for administering the system.
Some embodiments of the invention are further illustrated by the following examples, which should not be construed as limiting. It will be appreciated by those of skill in the art that the techniques disclosed in the examples which follow represent techniques discovered by the inventor to function well in the practice of the embodiments of the invention described herein, and thus can be considered to constitute preferred modes for its practice. However, those of skill in the art should, in light of the present disclosure, appreciate that many changes can be made in the specific embodiments which are disclosed and still obtain a like or similar result without departing from the spirit and scope of the invention.
Examples
The following examples further illustrate the invention. These examples are intended only to illustrate the invention and should not be construed as limiting the invention.
EXAMPLE 1 design and cloning of recombinant AAV vectors
The cap and rep coding sequences from AAV2, along with their corresponding promoters, were cloned into baculovirus plasmid vectors, respectively, to obtain the first polynucleotide comprising the coding sequence for the cap and rep proteins in this application.
The nucleotide sequence encoding Green Fluorescent Protein (GFP) and the nucleotide sequence encoding VEGF inhibitor shown in SEQ ID No.9, respectively, together with their corresponding promoters were cloned into baculovirus plasmid vectors to obtain second polynucleotides comprising the coding sequences of GFP and rep65, respectively.
Example 2 preparation of recombinant AAV viral particles
The first and second polynucleotides obtained in example 1 are mixed to form a composition of the present application and used to transfect Sf9 cells to obtain packaged rAAV2/VEGF inhibitor and rAAV2/GFP viral particles. Isolating and purifying the recombinant AAV2/VEGF inhibitor and AAV2/GFP viral particles from Sf9 cells.
Example 3 delivery and expression of reporter genes in mouse eyes
In this example, mice were divided into experimental and control groups, and purified rAAV2/GFP and PBS obtained in example 2 were injected into the eyes of the experimental and control mice, respectively. After a period of time, fluorescence expression in mouse retinal pigment epithelial cells was observed.
The results showed that green fluorescence was observed in the retinas of the experimental mice compared to the control group. This result indicates that the first polynucleotide in the present application was able to successfully package rAAV vectors comprising GFP coding sequences and generate rAAV2/GFP viral particles for delivery of GFP coding sequences, and that the delivered GFP coding sequences were able to be successfully expressed in mouse retinal pigment epithelium.
Example 4 delivery and expression of VEGF inhibitors in mice
Mice were divided into two groups, in which control and experimental groups were intraocular injected with rAAV2/GFP and AAV2/VEGF inhibitor viral particles purified in example 2, respectively. The eyes of the mice were observed after injection. Observations indicate that GFP was successfully expressed on mouse retinal pigment epithelium, indicating that the recombinant VEGF inhibitor coding sequence can be expressed on the retina.
Example 5 in vivo efficacy of the compositions of the present application
2-arm clinical experiments were performed using a control, system of the present application comprising rAAV2/VEGF inhibitor viral particles to verify the effectiveness of the system described herein.
Sequence listing
Figure BDA0002594993810000431
Figure BDA0002594993810000441

Claims (31)

1. A composition, comprising:
(i) a first polynucleotide, wherein the first nucleotide comprises a first sequence encoding an adeno-associated virus (AAV) capsid protein operably linked to a first promoter and a second sequence encoding an AAV rep protein operably linked to a second promoter, and the first promoter and the second promoter are suitable for expression in insect cells, and
(ii) a second polynucleotide, wherein said second polynucleotide comprises a third sequence encoding a Vascular Endothelial Growth Factor (VEGF) inhibitor operably linked to a CMV, CAG, MNDU3, PGK, EF1a promoter or an eye-specific promoter.
2. The composition of claim 1, wherein the VEGF inhibitor is a VEGF antibody or an antigen-binding fragment thereof.
3. The composition of claim 1, wherein the VEGF inhibitor comprises the sequences of SEQ ID NOs 1, 2, and 3.
4. The composition of claim 1, wherein the VEGF inhibitor comprises the sequences of SEQ ID NOs 4, 5, and 6.
5. The composition of claim 1, wherein the VEGF inhibitor comprises the sequence of SEQ ID NO 7.
6. The composition of claim 1, wherein the VEGF inhibitor comprises the sequence of SEQ ID NO 8.
7. The composition of claim 1, wherein said insect cell is an Sf9 cell.
8. The composition of claim 1, wherein the first promoter or second promoter is a p10 promoter.
9. The composition of claim 1, wherein said first promoter or second promoter is a PolH promoter.
10. The composition of claim 1, wherein the eye-specific promoter is selected from the group consisting of an RPE 65 gene promoter, a human retinal binding protein (CRALBP) gene promoter, a murine 11-cis Retinol Dehydrogenase (RDH) gene promoter, a rhodopsin kinase promoter (rhodopsin kinase promoter), a tissue inhibitor of metalloproteinase 3 (Timp3) promoter, a photoreceptor retinoid binding protein promoter, and a vitreous macular dystrophy 2 (vitroplastic dystrophy 2) promoter, an inter-photoreceptor retinoid-binding protein (IRBP) promoter.
11. The composition of claim 1, wherein the first sequence further comprises a poly-a sequence at the 3' end.
12. The composition of claim 1, wherein the second sequence further comprises a poly-a sequence at the 3' end.
13. The composition of claim 1, wherein the first sequence and the second sequence are linked by a sequence encoding a linker.
14. The composition of claim 13, wherein the linker is a cleavable linker.
15. The composition of claim 13, wherein the linker comprises the sequence of a 2A peptide.
16. The composition of claim 13, wherein the linker encoding sequence further comprises a promoter.
17. The composition of claim 16, wherein the promoter is an FMDV promoter.
18. The composition of claim 1, wherein the third sequence further comprises a poly-a sequence at the 3' end.
19. The composition of claim 1, wherein the second polynucleotide further comprises a stuffer sequence.
20. The composition of claim 1, wherein the second polynucleotide further comprises an Inverted Terminal Repeat (ITR) sequence.
21. The composition of claim 1, wherein the second polynucleotide further comprises a fourth sequence encoding an additional therapeutic protein.
22. The composition of claim 21, wherein the additional therapeutic protein is selected from the group consisting of: VEGF inhibitors, PDGF inhibitors, integrin inhibitors, mTOR inhibitors, angiogenin inhibitors, and TGF β inhibitors.
23. The composition of claim 21, wherein the third sequence and the fourth sequence are linked by a sequence encoding a linker.
24. The composition of claim 23, wherein the linker is a cleavable linker.
25. The composition of claim 23, wherein the linker comprises the sequence of a 2A peptide.
26. A recombinant adeno-associated virus (rAAV) particle prepared by transfecting the composition of any one of claims 1-25 into an insect cell.
27. The rAAV particle of claim 26, wherein the insect cell is an Sf9 cell.
28. A system for treating an ocular disease in a subject in need thereof, comprising combining the rAAV particle of any one of claims 26-27 and a pharmaceutically acceptable carrier.
29. A method for treating an ocular disease in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the system of claim 28.
30. The method of claim 29, wherein the ocular disease is selected from: wet age-related macular degeneration (wet AMD), diabetic retinopathy, diabetic macular edema, proliferative diabetic retinopathy and macular edema.
31. A kit comprising the system of claim 28 and instructions.
CN202010706658.XA 2020-07-21 2020-07-21 Compositions and methods for treating ocular diseases Pending CN113952474A (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
CN202010706658.XA CN113952474A (en) 2020-07-21 2020-07-21 Compositions and methods for treating ocular diseases
AU2021313839A AU2021313839A1 (en) 2020-07-21 2021-07-20 Composition and method for treating eye diseases
PCT/IB2021/000498 WO2022018516A1 (en) 2020-07-21 2021-07-20 Composition and method for treating eye diseases
JP2023512663A JP2023540464A (en) 2020-07-21 2021-07-20 Compositions and methods for the treatment of eye diseases
EP21845723.2A EP4185333A1 (en) 2020-07-21 2021-07-20 Composition and method for treating eye diseases
CA3186830A CA3186830A1 (en) 2020-07-21 2021-07-20 Composition and method for treating eye diseases
CN202180064547.6A CN116323949A (en) 2020-07-21 2021-07-20 Compositions and methods for treating ocular disorders
US18/157,599 US20230295243A1 (en) 2020-07-21 2023-01-20 Composition and method for treating eye diseases

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010706658.XA CN113952474A (en) 2020-07-21 2020-07-21 Compositions and methods for treating ocular diseases

Publications (1)

Publication Number Publication Date
CN113952474A true CN113952474A (en) 2022-01-21

Family

ID=79459859

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010706658.XA Pending CN113952474A (en) 2020-07-21 2020-07-21 Compositions and methods for treating ocular diseases

Country Status (1)

Country Link
CN (1) CN113952474A (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109641065A (en) * 2016-06-16 2019-04-16 阿德夫拉姆生物技术股份有限公司 AMD is treated using AAV2 variant and VEGF Trap
US20190127455A1 (en) * 2016-04-15 2019-05-02 Regenxbio Inc. Treatment of Ocular Diseases with Fully- Human Post-Translationally Modified Anti-VEGF Fab
CN110423281A (en) * 2019-07-31 2019-11-08 成都金唯科生物科技有限公司 For treating the fusion protein, viral vectors and drug of senile macular degeneration

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20190127455A1 (en) * 2016-04-15 2019-05-02 Regenxbio Inc. Treatment of Ocular Diseases with Fully- Human Post-Translationally Modified Anti-VEGF Fab
CN109641065A (en) * 2016-06-16 2019-04-16 阿德夫拉姆生物技术股份有限公司 AMD is treated using AAV2 variant and VEGF Trap
CN110423281A (en) * 2019-07-31 2019-11-08 成都金唯科生物科技有限公司 For treating the fusion protein, viral vectors and drug of senile macular degeneration

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
RUSLAN GRISHANIN等: "Preclinical Evaluation of ADVM-022, a Novel Gene Therapy Approach to Treating Wet Age-Related Macular Degeneration", 《MOLECULAR THERAPY》, vol. 27, no. 1, 2 January 2019 (2019-01-02), pages 118 - 129, XP055643284, DOI: 10.1016/j.ymthe.2018.11.003 *
THALES ANTONIO CABRAL DE GUIMARAES等: "Gene therapy for neovascular age-related macular degeneration: rationale, clinical trials and future directions", 《BR J OPHTHALMOL》, vol. 105, no. 2, 8 April 2020 (2020-04-08), pages 151 - 157 *

Similar Documents

Publication Publication Date Title
JP7492556B2 (en) Compositions for the treatment of wet age-related macular degeneration
US20180353614A1 (en) VE-PTP Extracellular Domain Antibodies Delivered by a Gene Therapy Vector
US20220143221A1 (en) Gene Therapy For Eye Pathologies
US20210093734A1 (en) Compositions for treatment of wet age-realted macular degeneration
US20230295243A1 (en) Composition and method for treating eye diseases
CA3182080A1 (en) Viral vectors for the treatment of retinal dystrophy
US20150182638A1 (en) Virus-mediated delivery of bevacizumab for therapeutic applications
US20230321280A1 (en) Compositions and methods for the treatment of ocular diseases
US20230321281A1 (en) Compositions and methods for the treatment of eye diseases
WO2024002076A1 (en) Aav drug for treating angiogenesis-related fundus diseases
KR20210148333A (en) Treatment of ocular lysosomal storage diseases through administration of AAV expressing TPP1
WO2023201308A1 (en) Gene therapy for treating an ocular disease
CN113952474A (en) Compositions and methods for treating ocular diseases
CN113952473A (en) Compositions and methods for treating ocular diseases
CN113952471A (en) Compositions and methods for treating ocular diseases
JPWO2022018516A5 (en)
US20230381343A1 (en) Treatment for Intraocular Pressure Related Disorders
JPWO2022018518A5 (en)
TW202423974A (en) AAV vector for infecting retina, adalimumab and application thereof
CN118186005A (en) Expression cassette suitable for adalimumab, and vector and application thereof
NZ787237A (en) Compositions For Treatment of Wet Age-Related Macular Degeneration
NZ787256A (en) Compositions For Treatment of Wet Age-Related Macular Degeneration
NZ746729B2 (en) Compositions for treatment of wet age-related macular degeneration

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination