WO2025074268A1 - Méthodes pour traiter la dégénérescence maculaire - Google Patents
Méthodes pour traiter la dégénérescence maculaire Download PDFInfo
- Publication number
- WO2025074268A1 WO2025074268A1 PCT/IB2024/059630 IB2024059630W WO2025074268A1 WO 2025074268 A1 WO2025074268 A1 WO 2025074268A1 IB 2024059630 W IB2024059630 W IB 2024059630W WO 2025074268 A1 WO2025074268 A1 WO 2025074268A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- protein
- scd59
- pharmaceutical composition
- vector
- dose
- 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
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/177—Receptors; Cell surface antigens; Cell surface determinants
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/56—Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
- A61K31/57—Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone
- A61K31/573—Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone substituted in position 21, e.g. cortisone, dexamethasone, prednisone or aldosterone
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/7088—Compounds having three or more nucleosides or nucleotides
- A61K31/713—Double-stranded nucleic acids or oligonucleotides
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P27/00—Drugs for disorders of the senses
- A61P27/02—Ophthalmic agents
Definitions
- the present disclosure relates generally to methods of treating geographic atrophy (GA) that is secondary to age-related macular degeneration (AMD).
- GA geographic atrophy
- AMD age-related macular degeneration
- Age-related macular degeneration is the leading cause of blindness in patients over 60 years of age in developed countries, affecting approximately 1 in 4 people over 70 years of age. Globally, AMD accounts for approximately 9% of all blindness, affects approximately 196 million people and is projected to affect 288 million by 2040. Age-related macular degeneration is a slow and progressive disease of the macula.
- the dry (nonexudative) form of AMD comprises the majority (-90%) of AMD patients.
- the wet (exudative) form of AMD accounts for the other -10% of cases. The most significant loss of vision occurs in advanced forms of AMD such as wet AMD and GA.
- GA Geographic atrophy is characterized by progressive and irreversible loss of photoreceptors, RPE and choriocapillaris. It is typically bilateral, and enlargement of the atrophy leads to irreversible visual function loss. It can affect central visual acuity as well as reading, driving, and low- light vision. Globally, GA affects more than 5 million people. There is a need for improved approaches for treating patients with AMD and GA. The disclosed methods are directed to these and other important needs.
- the present disclosure is directed to methods of treating geographic atrophy (GA) in a subject, the methods comprising administering a pharmaceutical composition into a GA-affected eye of a subject by ocular injection, wherein the pharmaceutical composition comprises a nucleic acid encoding a soluble CD59 (sCD59) protein.
- the GA is secondary to age-related macular degeneration, i.e., the methods involve treating GA in a subject who also suffers from age-related macular degeneration.
- the pharmaceutical composition comprises a dose of viral particles of about 8.2 x 10 10 vector genome (vg).
- the pharmaceutical composition comprises a dose of viral particles of about 4. 1 x 10 n vg.
- the GA covers an area of the affected eye measuring 2.5 mm 2 to 17.5 mm 2 .
- the GA is non-subfoveal.
- the GA is multifocal with at least one focal lesion of >1.25 mm 2 .
- the GA lesion is measured as the square root of GA lesion area by fundus autofluorescence.
- the subject is 60 years of age or older.
- the pharmaceutical composition is injected into only one eye.
- the pharmaceutical composition is administered by a single intravitreal injection in the GA-affected eye.
- the treatment is assessed at about 18 months following administration of the pharmaceutical composition.
- the pharmaceutical composition is administered with prednisone.
- the prednisone is administered orally for 20 days at a dose of about 40 mg/day.
- the dose of prednisone is tapered down by about 10 mg every 5 days.
- the pharmaceutical composition is administered with a corticosteroid.
- corticosteroid is administered by single periocular injection.
- the corticosteroid is triamcinolone acetonide (TA).
- TA is administered at a dose of about 40 mg.
- the sCD59 protein lacks a glycosyl phosphatidyl inositol (GPI) anchoring domain.
- the nucleic acid encoding the sCD59 protein comprises SEQ ID NO: 1.
- the sCD59 protein comprises SEQ ID NO: 2.
- the nucleic acid encoding the sCD59 protein is operably linked to a promoter sequence for expression and secretion of the sCD59 protein in the cells of the GA-affected eye.
- the nucleic acid encoding the sCD59 protein is packaged into a delivery vector.
- the delivery vector is an adeno-associated virus (AAV) vector.
- AAV vector is AAV2.
- the promoter is a CAG promoter.
- the present disclosure is directed to methods of improving GA lesion growth in a subject, the methods comprising administering a pharmaceutical composition into a GA-affected eye of a subject by ocular injection, wherein the pharmaceutical composition comprises a nucleic acid encoding a sCD59 protein.
- the improvement a change in the square root transformed GA lesion area (sqrt-GAA) from baseline at months 3, 6, 9, 12, 15, and 18 in the GA-affected eye.
- the improvement is relative to a sham control.
- the improvement is measured at about 18 months after injection.
- the gradations used in a series of values may be used to determine the intended range available to the term “about” or “substantially” for each value. Where present, all ranges are inclusive and combinable. That is, references to values stated in ranges include every value within that range. [0003] When a list is presented, unless stated otherwise, it is to be understood that each individual element of that list and every combination of that list is to be interpreted as a separate embodiment. For example, a list of embodiments presented as “A, B, or C” is to be interpreted as including the embodiments, “A,” “B,” “C,” “A or B,” “A or C,” “B or C,” or “A, B, or C.”
- compositions can be administered systemically either orally, buccally, parenterally, topically, by inhalation or insufflation (i.e., through the mouth or through the nose), or rectally in dosage unit formulations containing conventional nontoxic pharmaceutically acceptable carriers, adjuvants, and vehicles as desired, or can be locally administered by means such as, but not limited to, injection, implantation, grafting, topical application, or parenterally.
- CD59 refers to a membrane-bound glycoprotein found associated with membranes of cells including both human hematopoietic and non- hematopoietic cells, for example on endothelial cells, peripheral nerve fibers, neurons, microglia, oligodendrocytes, astrocytes, ependymal cells, epithelial cells, acinar cells of the salivary glands, bronchial epithelium, renal tubules and squamous epithelium.
- CD59 protein inhibits assembly of functional membrane attack complexes (MACs) and thus protects cells from complement- mediated activation and/or lysis.
- MACs functional membrane attack complexes
- the protein structure of CD59 includes a single cysteine-rich domain, a hydrophobic core with three loops and a small fourth helical loop (Yu et al. 1997 Journal of Experimental Medicine 185(4): 745-753).
- Human CD59 includes 26 amino acids located at the C terminus, which specifies a signal sequence for attachment of a glycosyl phosphatidyl inositol anchor (GPI anchor) at amino acid asparagine at position 77.
- GPI anchor glycosyl phosphatidyl inositol anchor
- doctor unit form refers to a physically discrete unit of active agent appropriate for the patient to be treated.
- drusen refers to yellow deposits under the retina.
- impairment refers to an increase or to intensify in quality or quantity, or to make better or augment.
- a polypeptide functionally equivalent to SEQ ID NO: 2 may have a biologic activity, e.g., an inhibitory activity, kinetic parameters, salt inhibition, a cofactor-dependent activity, and/or a functional unit size that is substantially similar or identical to the expressed polypeptide of SEQ ID NO: 2.
- a biologic activity e.g., an inhibitory activity, kinetic parameters, salt inhibition, a cofactor-dependent activity, and/or a functional unit size that is substantially similar or identical to the expressed polypeptide of SEQ ID NO: 2.
- FAF fundus autofluorescence
- cSLO confocal laser scanning ophthalmoscopy
- inhibitor and its various grammatical forms, including, but not limited to, “inhibiting” or “inhibition”, are used herein to refer to reducing the amount or rate of a process, to stopping the process entirely, or to decreasing, limiting, or blocking the action or function thereof.
- Inhibition can include a reduction or decrease of the amount, rate, action function, or process of a substance by at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, or at least 99%.
- membrane attack complex and “MAC” are used interchangeably herein to refer to an effector of the immune system comprising a complex of proteins typically formed on the surface of pathogen cell membranes as a result of activation of a host's complement system.
- Antibody-mediated complement activation leads to MAC deposition on the surface of infected cells, leading to pores that disrupt the cell membrane of the infected cells, resulting in cell lysis and death.
- the MAC is composed of complement components C5b, C6, C7, C8 and several C9 molecules.
- modify means to change, vary, adjust, temper, alter, affect or regulate to a certain measure or proportion in one or more particulars.
- multifocal when used in reference to GA means two or more atrophic lesions in the eye.
- nucleic acid is used herein to refer to a deoxyribonucleotide or ribonucleotide polymer in either single- or double-stranded form, and unless otherwise limited, encompasses known analogues having the essential nature of natural nucleotides in that they hybridize to single-stranded nucleic acids in a manner similar to naturally occurring nucleotides (e.g., peptide nucleic acids).
- nucleotide is used herein to refer to a chemical compound that consists of a heterocyclic base, a sugar, and one or more phosphate groups.
- the base is a derivative of purine or pyrimidine
- the sugar is the pentose deoxyribose or ribose.
- Nucleotides are the monomers of nucleic acids, with three or more bonding together in order to form a nucleic acid.
- Nucleotides are the structural units of RNA, DNA, and several cofactors, including, but not limited to, CoA, FAD, DMN, NAD, and NADP.
- Purines include adenine (A), and guanine (G); pyrimidines include cytosine (C), thymine (T), and uracil (U).
- sequence relationships between two or more nucleic acids or polynucleotides are used herein to describe the sequence relationships between two or more nucleic acids or polynucleotides: (a) “reference sequence”, (b) “comparison window”, (c) “sequence identity”, (d) “percentage of sequence identity”, and (e) “substantial identity.”
- reference sequence refers to a sequence used as a basis for sequence comparison.
- a reference sequence may be a subset or the entirety of a specified sequence; for example, as a segment of a full-length cDNA or gene sequence, or the complete cDNA or gene sequence.
- comparison window refers to a contiguous and specified segment of a polynucleotide sequence, wherein the polynucleotide sequence may be compared to a reference sequence and wherein the portion of the polynucleotide sequence in the comparison window may comprise additions or deletions (i.e., gaps) compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment of the two sequences.
- the comparison window is at least 20 contiguous nucleotides in length, and optionally can be at least 30 contiguous nucleotides in length, at least 40 contiguous nucleotides in length, at least 50 contiguous nucleotides in length, at least 100 contiguous nucleotides in length, or longer.
- a gap penalty typically is introduced and is subtracted from the number of matches.
- the BLAST family of programs which can be used for database similarity searches, includes: BLASTN for nucleotide query sequences against nucleotide database sequences; BLASTX for nucleotide query sequences against protein database sequences; BLASTP for protein query sequences against protein database sequences; TBLASTN for protein query sequences against nucleotide database sequences; and TBLASTX for nucleotide query sequences against nucleotide database sequences.
- sequence identity/similarity values refer to the value obtained using the BLAST 2.0 suite of programs using default parameters.
- Altschul et al. Nucleic Acids Res. 25:3389-3402 (1997).
- Software for performing BLAST analyses is publicly available, e.g., through the National Center for Biotechnology- Information.
- This algorithm involves first identifying high scoring sequence pairs (HSPs) by identifying short words of length W in the query sequence, which either match or satisfy some positive-valued threshold score T when aligned with a word of the same length in a database sequence. T is referred to as the neighborhood word score threshold (Altschul et al., supra).
- the BLAST algorithm parameters W, T, and X determine the sensitivity and speed of the alignment.
- the BLAST algorithm In addition to calculating percent sequence identity, the BLAST algorithm also performs a statistical analysis of the similarity between two sequences (see, e.g., Karlin & Altschul, Proc. Natl. Acad. Set. USA 90:5873-5787 (1993)).
- One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N)), which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance.
- P(N) the smallest sum probability
- BLAST searches assume that proteins may be modeled as random sequences. However, many real proteins comprise regions of nonrandom sequences which may be homopolymeric tracts, short-period repeats, or regions enriched in one or more amino acids.
- Such low-complexity regions may be aligned between unrelated proteins even though other regions of the protein are entirely dissimilar.
- a number of low-complexity filter programs may be employed to reduce such low-complexity alignments. Lor example, the SEG (Wooten and Lederhen, Comput. Chem., 17: 149-163 (1993)) and XNU (Claverie and States, Comput. Chem., 17: 191-201 (1993)) low-complexity filters may be employed alone or in combination.
- sequence identity or “identity” in the context of two nucleic acid or polypeptide sequences is used herein to refer to the residues in the two sequences that are the same when aligned for maximum correspondence over a specified comparison window.
- sequence identity When percentage of sequence identity is used in reference to proteins it is recognized that residue positions that are not identical often differ by conservative amino acid substitutions, i.e., where amino acid residues are substituted for other amino acid residues with similar chemical properties (e.g. charge or hydrophobicity) and therefore do not change the functional properties of the molecule. Where sequences differ in conservative substitutions, the percent sequence identity may be adjusted upwards to correct for the conservative nature of the substitution.
- Sequences that differ by such conservative substitutions are said to have “sequence similarity” or “similarity.” Means for making this adjustment are well-known to those of skill in the art. Typically this involves scoring a conservative substitution as a partial rather than a full mismatch, thereby increasing the percentage sequence identity. Thus, for example, where an identical amino acid is given a score of 1 and a non-conservative substitution is given a score of zero, a conservative substitution is given a score between zero and 1. The scoring of conservative substitutions is calculated, e.g., according to the algorithm of Meyers and Miller, Computer Applic. Biol. Set., 4: 11-17 (1988) e.g., as implemented in the program PC/GENE (Intelligenetics, Mountain View, Calif, USA).
- the term “percentage of sequence identity” is used herein mean the value determined by comparing two optimally aligned sequences over a comparison window, wherein the portion of the polynucleotide sequence in the comparison window may comprise additions or deletions (i.e., gaps) as compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment of the two sequences. The percentage is calculated by determining the number of positions at which the identical nucleic acid base or amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window of comparison, and multiplying the result by 100 to yield the percentage of sequence identity.
- polynucleotide sequences means that a polynucleotide comprises a sequence that has at least 70% sequence identity, at least 80% sequence identity, at least 90% sequence identity and at least 95% sequence identity, compared to a reference sequence using one of the alignment programs described using standard parameters.
- sequence identity compared to a reference sequence using one of the alignment programs described using standard parameters.
- One of skill will recognize that these values may be adjusted appropriately to determine corresponding identity of proteins encoded by two nucleotide sequences by taking into account codon degeneracy, amino acid similarity, reading frame positioning and the like.
- Substantial identity of amino acid sequences for these purposes normally means sequence identity of at least 60%, or at least 70%, at least 80%, at least 90%, or at least 95%.
- nucleotide sequences are substantially identical is if two molecules hybridize to each other under stringent conditions. However, nucleic acids that do not hybridize to each other under stringent conditions are still substantially identical if the polypeptides that they encode are substantially identical. This may occur, e.g., when a copy of a nucleic acid is created using the maximum codon degeneracy permitted by the genetic code.
- One indication that two nucleic acid sequences are substantially identical is that the polypeptide that the first nucleic acid encodes is immunologically cross reactive with the polypeptide encoded by the second nucleic acid.
- substantially identical of protein sequences refers to a first amino acid sequence that contains a sufficient or minimum number of amino acid residues that are identical to aligned amino acid residues in a second amino acid sequence such that the first and second amino acid sequences can have a common structural domain and/or common functional activity.
- amino acid sequences that contain a common structural domain having at least about 60% identity, or at least 75%, 80%, 85%, 90%, 95%, 96%, 98%, or 99% identity.
- parenteral refers to introduction into the body by way of an injection (i.e., administration by injection), including, for example, intraocularly (also known as intravitreally) (i.e., an injection into the vitreous of the eye), subretinally (i.e., an injection into the subretinal space which exists between the photoreceptors of the retina and the retinal pigment epithelium (RPE) layer), subcutaneously (i.e., an injection beneath the skin), intramuscularly (i.e., an injection into a muscle); intravenously (i.e., an injection into a vein), intrathecally (i.e., an injection into the space around the spinal cord), intrastemal injection, or infusion techniques.
- intraocularly also known as intravitreally
- subretinally i.e., an injection into the subretinal space which exists between the photoreceptors of the retina and the retinal pigment epithelium (RPE) layer
- subcutaneously i.e.
- a parenterally administered composition of the described invention is delivered using a needle, e.g., a surgical needle.
- a surgical needle refers to any needle adapted for delivery of fluid (i.e., capable of flow) compositions of the described invention into a selected anatomical structure.
- injectable preparations such as sterile injectable aqueous or oleaginous suspensions, can be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
- pharmaceutically acceptable carrier refers to any substantially non-toxic carrier conventionally useable for administration of pharmaceuticals in which the isolated polypeptide of the present invention will remain stable and bioavailable.
- the pharmaceutically acceptable carrier must be of sufficiently high purity and of sufficiently low toxicity to render it suitable for administration to the mammal being treated. It further should maintain the stability and bioavailability of an active agent.
- the pharmaceutically acceptable carrier can be liquid or solid and is selected, with the planned manner of administration in mind, to provide for the desired bulk, consistency, etc., when combined with an active agent and other components of a given composition.
- pharmaceutically acceptable salt means those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like and are commensurate with a reasonable benefit/risk ratio.
- polypeptide “peptide” and “protein” are used interchangeably herein to refer to a polymer of amino acid residues.
- the terms apply to amino acid polymers in which one or more amino acid residue is an artificial chemical analogue of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers.
- the essential nature of such analogues of naturally occurring amino acids is that, when incorporated into a protein, that protein is specifically reactive to antibodies elicited to the same protein but consisting entirely of naturally occurring amino acids.
- polypeptide and protein also are used herein in their broadest sense to refer to a sequence of subunit amino acids, amino acid analogs, or peptidomimetics. The subunits are linked by peptide bonds, except where noted.
- the polypeptides described herein may be chemically synthesized or recombinantly expressed. Polypeptides of the described invention also can be synthesized chemically. Synthetic polypeptides, prepared using the well-known techniques of solid phase, liquid phase, or peptide condensation techniques, or any combination thereof, can include natural and unnatural amino acids.
- Amino acids used for peptide synthesis may be standard Boc (N-a-amino protected N-a-t- butyloxycarbonyl) amino acid resin with the standard deprotecting, neutralization, coupling and wash protocols of the original solid phase procedure of Merrifield (1963, J. Am. Chem. Soc. 85:2149-2154), or the base-labile N-a-amino protected 9-fluorenylmethoxy carbonyl (Fmoc) amino acids first described by Carpino and Han (1972, J. Org. Chem. 37:3403-3409). Both Fmoc and Boc N-a-amino protected amino acids can be obtained from Sigma, Cambridge Research Biochemical, or other chemical companies familiar to those skilled in the art.
- polypeptides can be synthesized with other N-a-protecting groups that are familiar to those skilled in this art.
- Solid phase peptide synthesis may be accomplished by techniques familiar to those in the art and provided, for example, in Stewart and Young, 1984, Solid Phase Synthesis, Second Edition, Pierce Chemical Co., Rockford, Ill.; Fields and Noble, 1990, Int. J. Pept. Protein Res. 35: 161-214, or using automated synthesizers.
- the polypeptides of the invention may comprise D-amino acids (which are resistant to L-amino acid-specific proteases in vivo), a combination of D- and L-amino acids, and various “designer” amino acids (e.g., P-methyl amino acids, C-a- methyl amino acids, and N-a- methyl amino acids, etc.) to convey special properties.
- D-amino acids which are resistant to L-amino acid-specific proteases in vivo
- various “designer” amino acids e.g., P-methyl amino acids, C-a- methyl amino acids, and N-a- methyl amino acids, etc.
- synthetic amino acids include ornithine for lysine, and norleucine for leucine or isoleucine.
- the polypeptides can have peptidomimetic bonds, such as ester bonds, to prepare peptides with novel properties.
- a peptide may be generated that incorporates a reduced peptide bond, i.e., RI- CH2-NH- R2, where R1 and R2 are amino acid residues or sequences.
- a reduced peptide bond may be introduced as a dipeptide subunit.
- Such a polypeptide would be resistant to protease activity, and would possess an extended half-live in vivo. Accordingly, these terms also apply to amino acid polymers in which one or more amino acid residue is an artificial chemical analogue of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers.
- the essential nature of such analogues of naturally occurring amino acids is that, when incorporated into a protein, the protein is specifically reactive to antibodies elicited to the same protein but consisting entirely of naturally occurring amino acids.
- polypeptide also are inclusive of modifications including, but not limited to, glycosylation, lipid attachment, sulfation, gammacarboxylation of glutamic acid residues, hydroxylation, and ADP-ribosylation. It will be appreciated, as is well known and as noted above, that polypeptides may not be entirely linear. For instance, polypeptides may be branched as a result of ubiquitination, and they may be circular, with or without branching, generally as a result of posttranslational events, including natural processing event and events brought about by human manipulation which do not occur naturally.
- Circular, branched and branched circular polypeptides may be synthesized by non-translation natural process and by entirely synthetic methods, as well.
- the peptide is of any length or size.
- the term "recombinant” refers to a cell or vector that has been modified by the introduction of a heterologous nucleic acid or the cell that is derived from a cell so modified.
- Recombinant cells express genes that are not found in identical form within the native (nonrecombinant) form of the cell or express native genes that are otherwise abnormally expressed, under-expressed or not expressed at all as a result of deliberate human intervention.
- the term "recombinant” as used herein does not encompass the alteration of the cell or vector by naturally occurring events (e.g., spontaneous mutation, natural transformation transduction/transposition) such as those occurring without deliberate human intervention.
- recombinant expression cassette refers to a nucleic acid construct, generated recombinantly or synthetically, with a series of specified nucleic acid elements which permit transcription of a particular nucleic acid in a host cell.
- the recombinant expression cassette can be incorporated into a plasmid, chromosome, mitochondrial DNA, virus, or nucleic acid fragment.
- the recombinant expression cassette portion of an expression vector includes, among other sequences, a nucleic acid to be transcribed, a promoter, and a transcription termination signal such as a poly -A signal.
- the term "recombinant host” refers to any prokaryotic or eukaryotic cell that contains either a cloning vector or an expression vector. This term also includes those prokaryotic or eukaryotic cells that have been genetically engineered to contain the cloned genes, or gene of interest, in the chromosome or genome of the host cell.
- recombinant protein refers to a protein produced by genetic engineering, for example, by manipulation of genetically modified organisms such as microorganisms.
- reduce or “reducing” as used herein refers to the limiting of an occurrence of a disorder in individuals at risk of developing the disorder.
- sham refers to a procedure performed using a syringe without a needle or peri/intraocular penetration.
- solution refers to a homogeneous mixture of two or more substances. It is frequently, though not necessarily, a liquid. In a solution, the molecules of the solute (or dissolved substance) are uniformly distributed among those of the solvent.
- soluble CD59 refers to a CD59 amino acid sequence that lacks a GPI anchor or has a modified GPI anchor that lacks function and ability to bind to a cell membrane or a cell- membrane- associated structure such as a membrane-bound protein.
- suspension refers to a dispersion (mixture) in which a finely- divided species is combined with another species, with the former being so finely divided and mixed that it doesn't rapidly settle out.
- the most common suspensions are those of solids in liquid.
- subfoveal refers to as involving the center point of the fovea whereas the term “non-subfoveal” refers to not involving the center point of the fovea.
- the terms “subject” or “individual” or “patient” or “participant” are used interchangeably to refer to a member of an animal species of mammalian origin, including humans.
- the term “a subject in need thereof is used to refer to a subject having, or at risk of progression to heart failure, including a subject having an AMI that leads to a disease manifestation of left ventricular remodeling.
- subject in need of such treatment refers to a patient who suffers from a disease, disorder, condition, or pathological process.
- the term “subject in need of such treatment” also is used to refer to a patient who (i) will be administered at least one dose of the adenovirus vector construct expressing human sCD59 of the described invention; (ii) is receiving at least one dose of the adenovirus vector construct expressing human sCD59 of the described invention; or (iii) has received at least one dose of the adenovirus vector construct expressing human sCD59 of the described invention, unless the context and usage of the phrase indicates otherwise.
- symptom refers to a phenomenon that arises from and accompanies a particular disease or disorder and serves as an indication of it.
- therapeutic agent refers to a drug, molecule, nucleic acid, protein, metabolite, composition or other substance that provides a therapeutic effect.
- active refers to the ingredient, component or constituent of the compositions of the described invention responsible for the intended therapeutic effect.
- therapeutic agent and “active agent” are used interchangeably herein.
- therapeutic component refers to a therapeutically effective dosage (i.e., dose and frequency of administration) that eliminates, reduces, or prevents the progression of a particular disease manifestation in a percentage of a population.
- An example of a commonly used therapeutic component is the ED50 which describes the dose in a particular dosage that is therapeutically effective for a particular disease manifestation in 50% of a population.
- terapéuticaally effective amount refers to an amount that is sufficient to provide the intended benefit of treatment.
- An effective amount of the active agent(s) that can be employed according to the described invention generally ranges from about 8.2 x 10 10 vector genome (vg) to 4. 1 x 10 11 vg per dose.
- dosage levels are based on a variety of factors, including the type of injury, the age, weight, sex, medical condition of the patient, the severity of the condition, the route of administration, and the particular active agent employed. Thus, the dosage regimen may vary widely, but can be determined routinely by a physician using standard methods.
- compositions of the described invention includes prophylactic or preventative amounts of the compositions of the described invention.
- pharmaceutical compositions or medicaments are administered to a patient susceptible to, or otherwise at risk of, a disease, disorder or condition in an amount sufficient to eliminate or reduce the risk, lessen the severity, or delay the onset of the disease, disorder or condition, including biochemical, histologic and/or behavioral symptoms of the disease, disorder or condition, its complications, and intermediate pathological phenotypes presenting during development of the disease, disorder or condition. It is generally preferred that a maximum dose be used, that is, the highest safe dose according to some medical judgment.
- dose and “dosage” are used interchangeably herein.
- the therapeutically effective amount may be initially determined from preliminary in vitro studies and/or animal models.
- a therapeutically effective dose may also be determined from human data.
- the applied dose may be adjusted based on the relative bioavailability and potency of the administered compound. Adjusting the dose to achieve maximal efficacy based on the methods described above and other well-known methods is within the capabilities of the ordinarily skilled artisan.
- Pharmacokinetic principles provide a basis for modifying a dosage regimen to obtain a desired degree of therapeutic efficacy with a minimum of unacceptable adverse effects. In situations where the drug's plasma concentration can be measured and related to the therapeutic window, additional guidance for dosage modification can be obtained.
- Drug products are considered to be pharmaceutical equivalents if they contain the same active ingredients and are identical in strength or concentration, dosage form, and route of administration. Two pharmaceutically equivalent drug products are considered to be bioequivalent when the rates and extents of bioavailability of the active ingredient in the two products are not significantly different under suitable test conditions.
- treat or “treating” includes abrogating, substantially inhibiting, slowing or reversing the progression of a disease, condition or disorder, substantially ameliorating clinical or esthetical symptoms of a condition, substantially preventing the appearance of clinical or esthetical symptoms of a disease, condition, or disorder, and protecting from harmful or annoying symptoms.
- Treating further refers to accomplishing one or more of the following: (a) reducing the severity of the disorder; (b) limiting development of symptoms characteristic of the disorder(s) being treated; (c) limiting worsening of symptoms characteristic of the disorder(s) being treated; (d) limiting recurrence of the disorder(s) in patients that have previously had the disorder(s); and (e) limiting recurrence of symptoms in patients that were previously asymptomatic for the disorder(s).
- variants are used herein to refer to nucleotide or polypeptide sequences with substantial identity to a reference nucleotide or polypeptide sequence.
- the differences in the sequences may be the result of changes, either naturally or by design, in sequence or structure. Natural changes may arise during the course of normal replication or duplication in nature of the particular nucleic acid sequence. Designed changes may be specifically designed and introduced into the sequence for specific purposes. Such specific changes may be made in vitro using a variety of mutagenesis techniques. Such sequence variants generated specifically may be referred to as “mutants” or “derivatives” of the original sequence.
- a skilled artisan likewise can produce polypeptide variants of polypeptide SEQ ID NO: 2 having single or multiple amino acid substitutions, deletions, additions or replacements, but functionally equivalent to SEQ ID NO: 2.
- These variants may include inter alia: (a) variants in which one or more amino acid residues are substituted with conservative or non-conservative amino acids; (b) variants in which one or more amino acids are added; (c) variants in which at least one amino acid includes a substituent group; (d) variants in which amino acid residues from one species are substituted for the corresponding residue in another species, either at conserved or non-conserved positions; and (d) variants in which a target protein is fused with another peptide or polypeptide such as a fusion partner, a protein tag or other chemical moiety, that may confer useful properties to the target protein, for example, an epitope for an antibody.
- mutation refers to a change of the DNA sequence within a gene or chromosome of an organism resulting in the creation of a new character or trait not found in the parental type, or the process by which such a change occurs in a chromosome, either through an alteration in the nucleotide sequence of the DNA coding for a gene or through a change in the physical arrangement of a chromosome.
- Three mechanisms of mutation include substitution (exchange of one base pair for another), addition (the insertion of one or more bases into a sequence), and deletion (loss of one or more base pairs).
- vector refers to a carrier that is genetically engineered to deliver a gene to a cell.
- viral vector refers to a virus used as a vector to deliver a gene of interest by infecting a cell. Such viruses are modified so they cannot cause disease when used in humans. Types of viruses include, but are not limited to, retroviruses, which integrate their genetic material (including the gene interest) into a chromosome in a cell, and adenoviruses, which introduce their DNA (including the gene of interest) into the nucleus of a cell without integrating the genetic material into a chromosome.
- vehicle refers to a substance that facilitates the use of a drug or other material that is mixed with it.
- the present disclosure is directed to methods of treating geographic atrophy (GA) in a subject.
- the method comprises contacting cells or tissue with a pharmaceutical composition comprising a source of human sCD59 protein.
- the pharmaceutical composition comprises a nucleotide sequence encoding human sCD59 operably linked to a promoter.
- the pharmaceutical composition comprises a human sCD59 expression construct.
- the pharmaceutical composition comprises a nucleotide sequence encoding human sCD59 operably linked to a promoter that is packaged into a delivery vector.
- the pharmaceutical composition comprises a human sCD59 expression construct that is packaged into a delivery vector.
- the methods comprising, consisting of, or consisting essentially of administering a pharmaceutical composition into a GA-affected eye of a subject, wherein the pharmaceutical composition comprises a nucleic acid encoding a soluble CD59 (sCD59) protein, thereby treating the GA.
- a pharmaceutical composition comprises a nucleic acid encoding a soluble CD59 (sCD59) protein
- the GA is secondary to age-related macular degeneration.
- Age-related Macular Degeneration refers to a slow, progressive disease of the macula that impairs the ability of individuals to read, drive a car, recognize faces, and see objects in fine detail.
- the appearance of AMD begins with lipoproteinaceous yellow deposits (drusen) in the region of the macula and can progress towards advanced AMD where the most significant vision loss occurs.
- Advanced AMD is generally divided into 2 categories, which are not mutually exclusive: “wet” or “dry” AMD, also referred to as “exudative” or “non-exudative,” respectively.
- Wet AMD is associated with choroidal neovascularization (CNV) formation.
- CNV choroidal neovascularization
- Dry AMD is associated with drusen and pigment changes in the macula.
- the majority of vision loss occurs in advanced cases of dry AMD with the formation and enlargement of GA that is associated with loss of photoreceptors, retinal pigment epithelium (RPE), and choriocapillaris.
- the AMD is wet or exudative AMD.
- the AMD is dry AMD.
- the present disclosure is directed to methods of treating wet AMD in a subject.
- the method comprises contacting cells or tissue with a pharmaceutical composition comprising a source of human sCD59 protein.
- the pharmaceutical composition comprises a nucleotide sequence encoding human sCD59 operably linked to a promoter.
- the pharmaceutical composition comprises a human sCD59 expression construct.
- the pharmaceutical composition comprises a nucleotide sequence encoding human sCD59 operably linked to a promoter that is packaged into a delivery vector.
- the pharmaceutical composition comprises a human sCD59 expression construct that is packaged into a delivery vector.
- the methods comprising, consisting of, or consisting essentially of administering a pharmaceutical composition into a AMD-affected eye of a subject, wherein the pharmaceutical composition comprises a nucleic acid encoding a sCD59 protein, thereby treating the wet AMD.
- CD59 plasma membranes of cells are normally protected from the effects of complement by cell-surface proteins, e.g., CD59, that specifically inhibit activation of the C5b-9 pore upon C9 complement protein binding to membrane C5b-8
- CD59 competes with C9 complement protein for binding to C8 complement protein in the C5b-8 complex, thereby decreasing or preventing the formation of the C5b-9 membrane attack complex. CD59 thus acts to reduce both cell activation and cell lysis by terminal complement MACs.
- Diseases associated with uncontrolled complement activity include bacterial infection such as with Haemophilus influenza, Streptococcus pnemoniae, Neisseria meningitidis; angiodema; renal disease for example atypical haemolytic uremic syndrome; paroxysmal nocturnal hemoglobinuria; systemic lupus erythematosus; central nervous system diseases including Alzheimer's disease, Huntington's disease and diseases of the retina including, but not limited to, AMD.
- bacterial infection such as with Haemophilus influenza, Streptococcus pnemoniae, Neisseria meningitidis; angiodema; renal disease for example atypical haemolytic uremic syndrome; paroxysmal nocturnal hemoglobinuria; systemic lupus erythematosus; central nervous system diseases including Alzheimer's disease, Huntington's disease and diseases of the retina including, but not limited to, AMD.
- human sCD59 is effective to inhibit MAC formation.
- MAC formation is inhibited by delivering into a cell a vector containing a human sCD59-encoding nucleic acid.
- human sCD59 is effective to treat GA that is secondary to AMD.
- GA is treated by delivering into a cell a vector containing a human sCD59-encoding nucleic acid.
- human sCD59 is effective to prevent the onset of GA.
- the onset of GA is prevented by delivering into a cell a vector containing a human sCD59-encoding nucleic acid.
- human sCD59 is effective to prevent the progression of GA.
- the progression of GA is prevented by delivering into a cell a vector containing a human sCD59-encoding nucleic acid.
- human sCD59 is effective to reverse the progression of GA.
- the progression of GA is reversed by delivering into a cell a vector containing a human sCD59-encoding nucleic acid.
- the present disclosure is directed to methods of improving GA lesion growth in a subject, the methods comprising, consisting of, or consisting essentially of administering a pharmaceutical composition into a GA-affected eye of a subject by ocular injection, wherein the pharmaceutical composition comprises a nucleic acid encoding sCD59 protein.
- the present disclosure is directed to methods of slowing the progression of GA lesion growth in a subject, the methods comprising, consisting of, or consisting essentially of administering a pharmaceutical composition into a GA- affected eye of a subject by ocular injection, wherein the pharmaceutical composition comprises a nucleic acid encoding a sCD59 protein.
- the present disclosure is directed to methods of improving low luminance visual acuity (LLVA) in a subject suffering from GA, the methods comprising, consisting of, or consisting essentially of administering a pharmaceutical composition into a GA-affected eye of a subject by ocular injection, wherein the pharmaceutical composition comprises a nucleic acid encoding a sCD59 protein.
- the improvement in LLVA is determined by an improvement in an Early Treatment Diabetic Retinopathy Study (ETDRS) letter score.
- EDRS Early Treatment Diabetic Retinopathy Study
- the methods of improving LLVA in a subject suffering from GA comprise contacting the GA-affected eye with a composition including a vector carrying a nucleotide sequence encoding a recombinantly engineered human sCD59 protein operably linked to a promoter sequence causing expression of the protein in a cell, such that the protein includes at least one mutation resulting in loss of function of GIP anchoring domain, such that the protein is recombinant membrane-independent sCD59 and is not membrane targeting; and, observing an improvement in ETDRS after contacting, in comparison to the ETDRS observed prior to contacting
- the present disclosure is directed to methods of improving visual function and retinal function in a subject suffering from GA, the methods comprising, consisting of, or consisting essentially of administering a pharmaceutical composition into a GA-affected eye of a subject by ocular injection, wherein the pharmaceutical composition comprises a nucleic acid encoding a sCD59 protein.
- the improvement in visual function and retinal function is determined by an improvement in reading speed (Radner Chart).
- the improvement in visual function and retinal function is determined by an improvement in retinal sensitivity by mesopic microperimetry (macular integrity assessment (MAIA)).
- the methods of improving visual function and retinal function in a subject suffering from GA comprise contacting the GA-affected eye with a composition including a vector carrying a nucleotide sequence encoding a recombinantly engineered human sCD59 protein operably linked to a promoter sequence causing expression of the protein in a cell, such that the protein includes at least one mutation resulting in loss of function of GPI anchoring domain, such that the protein is recombinant membrane-independent CD59 (sCD59) and is not membrane targeting; and, observing an improvement in reading speed after contacting, in comparison to the reading speed observed prior to contacting.
- a composition including a vector carrying a nucleotide sequence encoding a recombinantly engineered human sCD59 protein operably linked to a promoter sequence causing expression of the protein in a cell, such that the protein includes at least one mutation resulting in loss of function of GPI anchoring domain, such that the protein is recombinant membrane-independent CD59 (sCD
- the methods of improving visual acuity in a subject suffering from GA comprise contacting the GA-affected eye with a composition including a vector carrying a nucleotide sequence encoding a recombinantly engineered human sCD59 protein operably linked to a promoter sequence causing expression of the protein in a cell, such that the protein includes at least one mutation resulting in loss of function of GIP anchoring domain, such that the protein is recombinant membraneindependent sCD59 and is not membrane targeting; and, observing an improvement in BCVA or ETDRS after contacting, in comparison to the BCVA or ETDRS observed prior to contacting.
- the present disclosure is directed to methods of improving functional reading independence and vision-related quality of life in a subject suffering from GA, the methods comprising, consisting of, or consisting essentially of administering a pharmaceutical composition into a GA-affected eye of a subject by ocular injection, wherein the pharmaceutical composition comprises a nucleic acid encoding a sCD59 protein.
- the improvement in functional reading independence and vision-related quality of life is determined by an improvement in the Functional Reading Independence (FRI) Index.
- the improvement in functional reading independence and vision-related quality of life is determined by an improvement in the National Eye Institute Visual Functioning Questionnaire-25 (NEI-VFQ- 25) composite score.
- the methods of improving functional reading independence and vision-related quality of life in a subject suffering from GA comprise contacting the GA-affected eye with a composition including a vector carrying a nucleotide sequence encoding a recombinantly engineered human sCD59 protein operably linked to a promoter sequence causing expression of the protein in a cell, such that the protein includes at least one mutation resulting in loss of function of GIP anchoring domain, such that the protein is recombinant membrane-independent sCD59 and is not membrane targeting; and, observing an improvement in FRI Index or NEI-VFQ-25 composite score after contacting, in comparison to the FRI Index or NEI-VFQ-25 observed prior to contacting
- the improvement a change in the square root transformed GA lesion area (sqrt-GAA) from baseline at months 3, 6, 9, 12, 15, and 18 in the GA-affected eye.
- the improvement is relative to a subject or a subject population who is not suffering from GA.
- the improvement is relative to a sham control injection.
- the sham control injection is performed using a syringe without a needle or peri/intraocular penetration.
- the subject is 80 years of age or older. In certain embodiments, the subject is 70 years of age or older. In further embodiments, the subject is 60 years of age or older. In certain embodiments, the subject is 50 years of age or older.
- cDNA sequences encoding human CD59 are known in the art. For example, cDNA sequences have been reported by Sawada, R. et al. 1989 Nucleic Acids Res 17(16): 6728 and are available from the American Type Tissue Culture Collection (ATCC, Manassas, Va.). A cDNA encoding CD59 has also been cloned from human T-cell leukemia (YT) and human erythroleukemia (K562) cell lines, and CD59 has been transiently expressed in COS cells (Walsh, L. A. et al. 1990 Eur J. Immol 21(3): 847-850).
- YT human T-cell leukemia
- K562 human erythroleukemia
- the sCD59 protein lacks a GPI anchoring domain.
- the human sCD59 comprises a modified GPI anchor domain amino acid sequence that is functionally defective and lacks the ability to target a membrane.
- the modified GPI anchor domain amino acid sequence comprises a variation. Such variations include, but are not limited to, substitution and deletion of nucleic acids encoding amino acids at omega positions used to reduce or eliminate the attachment of the GPI anchor or reduce or eliminate the effective functionality of the GPI anchor. Omega amino acids are amino acids to which GPI is transferred.
- such a variation includes, but is not limited to, substituting the nucleic acids encoding hydrophobic leucine (e.g., nucleic acids CTG) and alanine (e.g., nucleic acids GCA) with nucleic acids encoding glycine (e.g., nucleic acids CAG) and glutamate (e.g., nucleic acids GAA), which are less hydrophobic (i.e., more hydrophilic) amino acids.
- a variation may include substituting the omega residue with another amino acid, such as a glycine for a tyrosine.
- the nucleic acid encoding the sCD59 protein comprises or consists essentially of SEQ ID NO: 1. In certain embodiments, the nucleic acid encoding the sCD59 protein comprises or consists essentially of a sequence that is at least 90% identical to SEQ ID NO: 1. In certain embodiments, the nucleic acid encoding the sCD59 protein comprises or consists essentially of a sequence that is at least 95% identical to SEQ ID NO: 1.
- the sCD59 protein comprises or consists essentially of SEQ ID NO: 2. In certain embodiments, the sCD59 protein comprises or consists essentially of a sequence that is at least 90% identical to SEQ ID NO: 2. In certain embodiments, the sCD59 protein comprises or consists essentially of a sequence that is at least 95% identical to SEQ ID NO: 2.
- the human sCD59 of the described invention is a recombinant protein.
- a variety of commercially available expression vector/host systems are useful to contain and express a CD59 protein encoding sequence. These include but are not limited to microorganisms such as bacteria transformed with recombinant bacteriophage, plasmid or cosmid DNA expression vectors; yeast transformed with yeast expression vectors; insect cell systems contacted with virus expression vectors (e.g., baculovirus); plant cell systems transfected with virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with bacterial expression vectors (e.g., Ti, pBR322, or pET25b plasmid); or animal cell systems. See Ausubel et al., Current Protocols in Molecular Biology, John Wiley & Sons, New York, N.Y., 1989.
- Types of cells used in recombinant protein expression include, but are not limited to, prokaryotic and eukaryotic cells.
- Prokaryotic cells include, but are not limited to, bacterial cells.
- a non -limiting example of a bacterial cell includes Escherichia coli.
- Eukaryotic cells include, but are not limited to, mammalian, insect, yeast and algae cells.
- Non-limiting examples of mammalian cells include human embryonic kidney cells (e.g., HEK293, HEK293T), baby hamster kidney cells (e.g., BHK21), Chinese hamster ovary (CHO) cells, mouse myeloma cells (e.g., NSO) and murine non-producing hybridoma cells (e.g., SP2/O-Agl4).
- Non-limiting examples of insect cells include, Spodoptera frugiperda pupa ovarian cells (e.g., Sf9, Sf21).
- a non-limiting example of a yeast cell includes Saccharomyces cerevisiae.
- a non-limiting example of an algae cell includes Chlamydomonas reinhardtii.
- the human sCD59 is a synthetic protein.
- Methods of preparing synthetic proteins are well-known in the art. See, for example, Peptide Synthesis Protocols, Methods in Molecular Biology, vol. 35, Pennington, M. W. and Dunn, B. M., 1995, XII, Humana Press, Inc. Totowa, New Jersey.
- Synthetic proteins, prepared using well-known techniques such as solid phase, liquid phase, or peptide condensation techniques, or any combination thereof, can include natural and unnatural amino acids.
- Amino acids used for peptide synthesis may be standard Boc amino acid resin with the standard deprotecting, neutralization, coupling and wash protocols of original solid phase procedure of Merrifield (1963, J. Am. Chem. Soc.
- the nucleotide sequence encoding human sCD59 is used to construct an expression vector.
- the described invention provides a human sCD59 expression construct.
- Methods used to construct expression vectors are well known to those skilled in the art. For example, such methods can be used to construct expression vectors containing the nucleotide sequence encoding the human sCD59 protein operably linked to appropriate transcriptional and translational control elements. These methods include, but are not limited to, in vitro recombinant DNA techniques, synthetic techniques and in vivo recombination or genetic recombination. Such techniques are described in Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press, Plainview, N.Y., 1989.
- the nucleic acid encoding the sCD59 protein is operably linked to a promoter sequence for expression and secretion of the sCD59 protein in the cells of the GA-affected eye.
- the promoter is a constitutive promoter.
- the promoter is a cell cycle-specific promoter.
- the promoter is a ubiquitous promoter.
- the promoter is a tissue-specific promoters. Examples of tissue-specific promoters include, but are not limited to, human rhodopsin kinase (hRK) promoter and retinal pigmemt epithelium specific promoter (e.g., RPE65 promoter).
- the promoter is a metabolically regulated promoter.
- the promoter is an inducible promoter.
- the promoter is a hybrid promoter.
- a non-limiting example of a hybrid promoter is cytomegalovirus (CMV) early enhancer element/the first exon and the first intron of chicken beta-actin gene/the splice acceptor of the rabbit beta-globin gene (CAG).
- CMV cytomegalovirus
- CAG rabbit beta-globin gene
- Nonlimiting examples of promoters are shown in Evans et al. U.S. Pat. No. 6,677,311 Bl issued Jan. 13, 2004; Clark et al. U.S. Pat. No. 7,109,029 B2 issued Sep. 19, 2006; and Hallenbeck et al. U.S. Pat. No. 5,998,205 issued Dec. 7, 1999, each of which is incorporated herein by reference in its entirety.
- the delivery vector is a viral vector.
- the vector is at least one of: an engineered viral vector recombinantly linked to the nucleotide sequence encoding the sCD59 protein; and a synthetic gene delivery vector for delivery of the nucleotide sequence encoding human sCD59.
- the viral vector is selected from the group consisting of adenovirus, adeno- associated virus (AAV), a herpesvirus, a poxvirus, a lentivirus, and helper-dependent adenovirus vectors.
- the synthetic gene delivery vector is selected from the group consisting of a liposome, a lipid/polycation (LPD), a peptide, a nanoparticle, a gold particle, and a polymer.
- Adenovirus vectors are commercially available from American Type Tissue Culture Collection (Manassas, Va.). Methods of constructing adenovirus vectors and using adenovirus vectors are described in Klein et al. 2007 Ophthalmology 114: 253-262, and van Lecuwen et al. 2003 Eur. J. Epidemiol. 18: 845-854. Adenovirus vectors have been used in eukaryotic gene expression (Levrero et al. 1991 Gene, 101: 195-202) and vaccine development (Graham et al. 1991 Methods in Molecular Biology: Gene Transfer and Expression Protocols 7, (Murray, Ed.), Humana Press, Clifton, N.J., 109-128). Further, recombinant adenovirus vectors are used for gene therapy (Wu et al. U.S. Pat. No. 7,235,391 issued Jun. 26, 2007 which is incorporated herein by reference in its entirety).
- Recombinant adenovirus vectors are generated, for example, from homologous recombination between a shuttle vector and a pro virus vector (Wu et al., U.S. Pat. No. 7,235,391 issued Jun. 26, 2007).
- the adenovirus vectors used herein are replication defective.
- the adenovirus vectors are conditionally defective, lacking adenovirus El region.
- a polynucleotide encoding a protein of interest, human sCD59 for example, is introduced at the position from which the El -coding sequences have been removed.
- the polynucleotide encoding the protein of interest e.g., human sCD59
- Defective adenovirus vectors can be generated and propagated using a helper cell line.
- Helper cell lines may be derived from human cells such as, 293 human embryonic kidney cells (HEK293), muscle cells, hematopoietic cells or other human embryonic mesenchymal or epithelial cells.
- the helper cells may be derived from the cells of other mammalian species that are permissive for human adenovirus, e.g., Vero cells or other monkey embryonic mesenchymal or epithelial cells. Generation and propagation of these replication defective adenovirus vectors using a helper cell line is described in Graham et al 1977 J. Gen. Virol. 36: 59-72.
- Lentiviral packaging vectors are commercially available from Invitrogen Corporation (Carlsbad Calif).
- An HIV-based packaging system for the production of lentiviral vectors is prepared using constructs described in Naldini et al. 1996 Science 272: 263-267; Zufferey et al. 1997 Nature Biotechnol. 15: 871-875; and Dull et al. 1998 J. Virol. 72: 8463-8471.
- a number of vector constructs are available to be packaged using a system, based on third-generation lentiviral SIN vector backbone (Dull et al. 1998 J. Virol. 72: 8463- 8471).
- the vector construct pRRLsinCMVGFPpre contains a 5' LTR in which the HIV promoter sequence has been replaced with that of Rous sarcoma virus (RSV), a selfinactivating 3' LTR containing a deletion in the U3 promoter region, the HIV packaging signal, RRE sequences linked to a marker gene cassette consisting of the Aequora jellyfish green fluorescent protein (GFP) driven by the CMV promoter, and the woodchuck hepatitis virus PRE element, which appears to enhance nuclear export.
- the GFP marker gene allows quantitation of transfection or transduction efficiency by direct observation of UV fluorescence microscopy or flow cytometry (Kafri et al. 1997 Nature Genet. 17: 314-317; and Sakoda et al. 1999 J. Mol. Cell. Cardiol. 31 : 2037-2047).
- Retroviral nucleic acids to construct a retroviral vector containing a gene of interest (e.g., gene that encodes for human sCD59 protein) and packaging cells is accomplished using techniques known in the art (See, e.g., Ausubel, et al., 1992, Volume 1, Section III (units 9.10.1-9.14.3); Sambrook, et al., 1989. Molecular Cloning: A Laboratory Manual. Second Edition. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.; Miller, et al., Biotechniques. 7:981-990, 1989; Eglitis, et al., Biotechniques. 6:608-614, 1988; U.S. Pat. Nos.
- a retroviral vector can be constructed and packaged into non-infectious transducing viral particles (virions) using an amphotropic packaging system. Examples of such packaging systems are described in Miller et al. 1986 Mol. Cell Biol. 6 : 2895-2902; Markowitz et al. 1988 J. Virol. 62: 1120-1124; Cosset et al. 1990 J. Virol. 64: 1070-1078; U.S. Pat. Nos.
- producer cells can be accomplished by introducing retroviral vectors into the packaging cells. Examples of such retroviral vectors are found in, for example, Korman et al. 1987 Proc. Natl. Acad. Sci. USA.
- Herpesvirus packaging vectors are commercially available from Invitrogen Corporation, (Carlsbad, Calif).
- Exemplary herpesviruses include, but are not limited to, an a- herpesvirus, such as Varicella-Zoster vims or pseudorabies vims; a herpes simplex vims such as HSV-1 or HSV-2; or a herpesvirus such as Epstein-Barr vims.
- a method for preparing empty herpesvirus particles that can be packaged with a desired nucleotide segment for example a human sCD59 nucleotide or polynucleotide sequence, in the absence of a helper vims that is capable to most herpesviruses is described in Fraefel et al. (U.S. Pat. No. 5,998,208, issued Dec. 7, 1999 which is incorporated by reference in its entirety).
- the herpesvirus DNA vector can be constmcted using techniques known to the skilled artisan. For example, DNA segments encoding the entire genome of a herpesvirus is divided among a number of vectors capable of carrying large DNA segments, e.g., cosmids (Evans, et al., Gene 79, 9-20, 1989), yeast artificial chromosomes (YACS) (Sambrook, J. et al., MOLECULAR CLONING: A LABORATORY MANUAL, 2nd Edition, Cold Spring Harbor Press, Cold Spring Harbor, N.Y., 1989) or E. coli F element plasmids (O'Conner et al. 1989 Science 244: 1307- 1313).
- cosmids Evans, et al., Gene 79, 9-20, 1989
- yeast artificial chromosomes YACS
- E. coli F element plasmids O'Conner et al. 1989 Science 244: 1307- 1313.
- sets of cosmids have been isolated which contain overlapping clones that represent the entire genomes of a variety of herpesviruses including Epstein-Barr virus, Varicella-Zoster virus, pseudorabies virus and HSV-1.
- Epstein-Barr virus Varicella-Zoster virus
- pseudorabies virus pseudorabies virus
- HSV-1 herpesviruses
- Adeno-associated virus is a dependent parvovirus in that it depends on coinfection with another virus (either adenovirus or a member of the herpes virus family) to undergo a productive infection in cultured cells (Muzyczka 1992 Curr. Top. Microbiol. Immunol., 158:97 129).
- recombinant AAV (rAAV) virus can be made by cotransfecting a plasmid containing a gene of interest (e.g., human sCD59 gene), flanked by the two AAV terminal repeats (McLaughlin et al. 1988 J. Virol., 62(6): 1963-1973; Samulski et al. 1989 J.
- AAVs are innately nonpathogenic, poorly immunogenic, and broadly tropic, making them attractive gene delivery candidates for virusbased gene therapies.
- Most naturally occurring AAVs utilize glycan moieties for initial attachment to a cell surface, and these interactions have been well characterized for a number of serotypes.
- the interacting glycan moieties identified include AAV serotype 2 (AAV2), AAV3, AAV6 and AAV8; N- terminal galactose for AAV9; and specific N- or O-linked sialic acid moieties for AAV1, -4, -5, and -6.
- Serotypes differ by the types of cells they infect, making AAV a very useful system for preferentially transducing specific cell types.
- Adeno-associated virus (AAV) packaging vectors are commercially available from GeneDetect (Auckland, New Zealand). AAV has a broad host range for infectivity (Tratschin et al. 1984 Mol. Cell. Biol. 4: 2072-2081; Laughlin et al. 1986 J. Virol., 60(2): 515-524; Lebkowski et al. 1988 Mol. Cell. Biol. 8(10): 3988-3996; McLaughlin et al. 1988 J. Virol. 62(6): 1963- 1973).
- the nucleotide sequence encoding human sCD59 operably linked to a promoter is packaged into an adeno-associated virus (AAV) vector.
- AAV vector is AAV2.
- the AAV vector is AAV5.
- the AAV vector is AAV8.
- the nucleotide sequence encoding human sCD59 is packaged between inverted terminal repeat (ITR) sequences within an AAV vector.
- the nucleotide sequence encoding human sCD59 operably linked to a promoter is packaged between inverted terminal repeat (ITR) sequences within an AAV vector.
- the human sCD59 expression vector is packaged between inverted terminal repeat (ITR) sequences within an AAV vector.
- the ITR sequences are AAV2 sequences.
- the ITR sequences are AAV5 sequences.
- the ITR sequences are AAV8 sequences.
- the AAV vector is a hybrid vector.
- Hybrid vectors contain ITR sequences from one AAV serotype and a capsid protein from a different AAV serotype.
- the hybrid vector comprises ITR sequences from AAV2 and a capsid protein from AAV5 (AAV2/5).
- the hybrid vector comprises ITR sequences from AAV2 and a capsid protein from AAV8 (AAV2/8).
- the hybrid vector comprises ITR sequences from AAV5 and a capsid protein from AAV2 (AAV5/2).
- the hybrid vector comprises ITR sequences from AAV5 and a capsid protein from AAV8 (AAV5/8). According to some embodiments, the hybrid vector comprises ITR sequences from AAV8 and a capsid protein from AAV2 (AAV8/2). According to some embodiments, the hybrid vector comprises ITR sequences from AAV8 and a capsid protein from AAV5 (AAV8/5).
- the delivery vectors are non-viral vectors.
- the delivery vectors are synthetic gene delivery vehicles or vectors that are not related to a virus particle and that specifically deliver the gene material to the target cells or tissue.
- non-viral vectors include, but are not limited to, liposomes, peptides, nanoparticles, emulsions, or encapsulated two or more phase systems or other suitable preparation.
- the described invention provides a non- viral vector with nucleic acid that is loaded and contacted to a tissue or cell.
- a liposome containing naked DNA encoding a human sCD59 protein having a modified GPI anchor that does not target a membrane, or a gene encoding a human sCD59 protein having no GPI anchor is encapsulated in the liposome and the liposome is contacted to the tissue or cell such that the nucleic acid is effectively delivered to the tissue or cell.
- delivery of human sCD59 to the RA- affected eyes of using an AAV vector is effective for long-term transgene expression.
- the pharmaceutical composition is administered by a route selected from the group consisting of intravitreal, subretinal, subconjunctival, subtenon; subcutaneous and intravenous.
- the pharmaceutical composition is administered by injection.
- the tissues contacted by the pharmaceutical composition comprises at least one tissue selected from the group consisting of retinal pigment epithelium, retina, choroid, sclera, Bruch's membrane and choroidal blood vessels.
- the pharmaceutical composition is injected into only one eye. In certain embodiments, the pharmaceutical composition is injected into both eyes. In some embodiments, the pharmaceutical composition is administered by a single intravitreal injection in the GA-affected eye.
- the administering step comprises administering the pharmaceutical composition as a single dose or as multiple doses.
- the administering step comprises administering the pharmaceutical composition as a single dose.
- the single dose is administered to the eye of a subject in need thereof.
- the subject in need thereof is suffering from AMD.
- the subject in need thereof is suffering from wet or exudative AMD.
- the subject in need thereof is suffering from GA.
- contacting cells with a vector containing a human sCD59-encoding nucleic acid produces a subset of cells that are 'factories' for local production and secretion of sCD59, which may protect adjacent ocular cells including retinal pigment epithelium (RPE) cells and choroidal blood vessels.
- RPE retinal pigment epithelium
- the exact dosage of the pharmaceutical composition may be chosen by the individual physician in view of the patient to be treated. Dosage and administration are adjusted to provide sufficient levels of the active agent(s) or to maintain the desired effect. Additional factors which may be taken into account include the severity of the disease state, e.g., intermediate or advanced stage of AMD; age, weight and gender of the patient; diet, time and frequency of administration; route of administration; drug combinations; reaction sensitivities; and tolerance/response to therapy. Long-acting pharmaceutical compositions may be administered one time, hourly, twice hourly, every 3 to four hours, once daily, twice daily, every 3 to 4 days, every week, or once every two weeks depending on half-life and clearance rate of the particular composition.
- the active agents of the described invention can be formulated in dosage unit form for ease of administration and uniformity of dosage. It will be understood, however, that the total daily usage of the compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment.
- the therapeutically effective dose can be estimated initially either in cell culture assays or in animal models, as provided herein, usually mice, but also potentially from rats, rabbits, dogs, or pigs. Such information can then be used to determine useful doses and routes of administration for humans. Pharmaceutical compositions which exhibit large therapeutic indices are preferred. The data obtained from cell culture assays and animal studies are used in formulating a range of dosage for human use.
- a therapeutically effective dose refers to that amount of active agent that ameliorates the symptoms or condition or prevents progression of GA.
- Therapeutic efficacy and toxicity of active agents can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., ED50 (the dose is therapeutically effective in 50% of the population) and LD50 (the dose is lethal to 50% of the population).
- the dose ratio of toxic to therapeutic effects is the therapeutic index, and it can be expressed as the ratio, LD50/ED50.
- a source of expression of a human sCD59 protein is administered as a dose of a viral vector or a nucleic acid vector.
- the pharmaceutical composition comprises a dose of viral particles of about 8.2 x 10 10 vector genome (vg).
- the pharmaceutical composition comprises a dose of viral particles of about 4. 1 x 10 11 vg.
- the dose of viral particles is alternatively expressed as DNAse-resistant particles (DRP).
- the pharmaceutical composition is administered with prednisone or prednisolone.
- the prednisone or prednisolone is administered orally.
- the prednisone or prednisolone is administered orally for 20 days at a dose of about 40 mg/day.
- the dose of prednisone or prednisolone is tapered down by about 10 mg every 5 days.
- the prednisone or prednisolone is administered orally at a dose of 40 mg/day, at day 5 the prednisone or prednisolone is administered orally at a dose of 30 mg/day, at day 10 the prednisone or prednisolone is administered orally at a dose of 20 mg/day, and at day 15 the prednisone or prednisolone is administered orally at a dose of 15 mg/day.
- the prednisone or prednisolone is administered starting on the same day that the pharmaceutical composition comprising a nucleotide sequence encoding human sCD59 is administered.
- the prednisone or prednisolone is taken in the morning with or after a meal.
- the pharmaceutical composition comprising a nucleotide sequence encoding human sCD59 is administered with a corticosteroid.
- corticosteroid is administered by single periocular injection.
- the corticosteroid is triamcinolone acetonide (TA).
- TA is administered at a dose of about 40 mg.
- the prednisone or prednisolone is administered starting four days after the pharmaceutical composition comprising a nucleotide sequence encoding human sCD59 is administered.
- the pharmaceutical composition comprising a nucleotide sequence encoding human sCD59 is administered with a VEGF treatment.
- the VEGF treatment is initiated if the subject has suspected onset or presence of new exudative MNV (also referred to as active CNV or wet AMD).
- the VEGF treatment is ranibizumab or aflibercept.
- the pharmaceutical composition comprising a nucleotide sequence encoding human sCD59 is not administered with a complement inhibitor.
- the described invention provides a pharmaceutical composition.
- the pharmaceutical composition comprises a human sCD59 protein comprising a full-length nucleic acid of CD59 that was modified to remove the signal sequence for attachment of the GPI anchor at the nucleotides encoding amino acid asparagine at position 77.
- the nucleic acid sequence of human sCD59 protein is modified by point mutations, substitutions or deletions to obtain a nucleic acid sequence that encodes an amino acid sequence that has a modified amino acid sequence at the GPI anchor location, such that the protein is unable to attach to a membrane of a cell.
- the pharmaceutical composition is formulated sterile for ocular delivery.
- the pharmaceutical composition formulated for sterile ocular delivery is in a dose effective to treat GA.
- the pharmaceutical composition formulated for ocular delivery further includes at least one of a pharmaceutically acceptable buffer, a pharmaceutically acceptable salt and a pharmaceutically acceptable emollient suitable for delivery by at least one route selected from: intra-ocular injection, subconjunctival injection, subtenon injection, eye drop, and ointment.
- the described invention provides a pharmaceutical composition that comprises a CD59-encoding nucleic acid or a source of human sCD59 protein expression.
- the CD59 protein includes a membrane -independent (i.e., soluble) CD59 protein.
- the pharmaceutical composition is compounded as an ophthalmologic formulation for administration to the eye.
- the pharmaceutical composition is compounded to enhance delivery to the fundus.
- the pharmaceutical composition is compounded to provide sustained release locally to the retina.
- the pharmaceutical composition is formulated to provide effective treatment of vessels and/or tissue involved in ocular diseases.
- the ocular disease is age-related macular degeneration (AMD).
- AMD age-related macular degeneration
- the AMD is wet or exudative AMD.
- the AMD is dry AMD or GA.
- the pharmaceutical composition of the described invention is formulated sufficiently pure for administration to a human subject, e.g., to the eye of a human subject.
- the pharmaceutical composition includes one or more additional therapeutic agent(s).
- the additional therapeutic agent or agents are selected from the group consisting of growth factors, anti-inflammatory agents, vasopressor agents, including, but not limited to, nitric oxide and calcium channel blockers, collagenase inhibitors, steroids (e.g., prednisolone), matrix metalloproteinase inhibitors, ascorbates, angiotensin H, angiotensin III, calreticulin, tetracyclines, fibronectin, collagen, thrombospondin, transforming growth factors (TGF), keratinocyte growth factor (KGF), fibroblast growth factor (FGF), insulin-like growth factors (IGFs), IGF binding proteins (IGFBPs), epidermal growth factor (EGF), platelet derived growth factor (PDGF), neu differentiation factor (NDF), hepatocyte growth factor (HGF), vascular endothelial growth factor (VEGF), heparin-binding EGF (HBEGF), thrombospondins, von Willebrand Fact
- the additional therapeutic agent or agents include, without limitation, anti-tumor, antiviral, antibacterial, anti- mycobacterial, anti-fungal, antiproliferative or anti-apoptotic agents.
- Therapeutic agents that are included in the pharmaceutical composition of the described invention are well known in the art. See for example, Goodman & Gilman's The Pharmacological Basis of Therapeutics, 9th Ed., Hardman, et al., eds., McGraw-Hill, 1996, the contents of which are herein incorporated by reference herein.
- the additional therapeutic agent or agents is a compound, composition, biologic or the like. According to some embodiments, the additional therapeutic agent or agents potentiate, stabilize, synergize or substitute for the ability of human sCD59 protein to protect cells from MAC deposition. According to some embodiments, the additional therapeutic agent or agents are provided at the same time as the pharmaceutical composition that comprises the human sCD59 protein. According to some embodiments, the additional therapeutic agent or agents are provided after the pharmaceutical composition that comprises the human sCD59 protein. According to some embodiments, the additional therapeutic agent or agents are provided before the pharmaceutical composition that comprises the human sCD59 protein. According to some embodiments, the additional therapeutic agent or agents are used to treat the same, a concurrent or a related symptom, condition or disease.
- the pharmaceutical composition of the described invention comprises a pharmaceutically acceptable carrier.
- Pharmaceutical acceptable carriers include, but are not limited to, any and all solvents, diluents, or other liquid vehicle, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired.
- Remington's Pharmaceutical Sciences Ed. by Gennaro, Mack Publishing, Easton, Pa., 1995 provides various carriers used in formulating pharmaceutical compositions and known techniques for the preparation thereof.
- materials which can serve as pharmaceutically acceptable carriers include, but are not limited to, sugars such as glucose and sucrose; excipients such as cocoa butter and suppository waxes; oils such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, com oil, and soybean oil; glycols such a propylene glycol; esters such as ethyl oleate and ethyl laurate; agar; buffering agents such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; Ringer's solution; ethyl alcohol; and phosphate buffer solutions, as well as other non-toxic compatible lubricants such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, releasing agents, coating agents, preservatives and antioxidants can also be present in the composition, according to the judgment of the formulator.
- sugars such as glucose and sucrose
- the pharmaceutical composition further includes a peptide for overall delivery (POD), the pharmaceutical composition operably linked to the compound to obtain a conjugated compound, such that the POD includes a protein transduction domain (PTD).
- POD protein transduction domain
- the POD composition is one shown in Kumar-Singh et al. PCT7US2008/010179 filed Aug. 28, 2008 or Kumar-Singh et al. U.S. publication 2010/0209447 published Aug. 19, 2010, each of which is incorporated herein by reference in its entirety.
- the pharmaceutical composition comprises a dose of viral vector particles administered to an affected eye.
- the pharmaceutical composition comprises a dose of viral particles of about 8.2 x 10 10 vector genome (vg).
- the pharmaceutical composition comprises a dose of viral particles of about 4. 1 x 10 11 vg.
- the pharmaceutical composition further includes at least one therapeutic agent selected from the group consisting of an antiinflammatory, an antitumor, an antiviral, an antibacterial, an anti-mycobacterial, an antifungal, an anti-proliferative and an anti-apoptotic.
- the described invention provides a method for formulating a composition for treating GA in a subject, the method comprising engineering a vector to deliver and express a human sCD59 nucleotide sequence encoding an amino acid sequence corresponding to human sCD59, such that the nucleotide sequence includes a mutation encoding for amino acids of a GPI anchoring domain of the protein, such that the resulting vector encodes an engineered recombinant membrane- independent (i.e., soluble) CD59 (sCD59) protein, and the vector is a viral vector or a synthetic gene delivery vector; and, contacting at least one ocular tissue of the subject with the composition, such that the cells of the tissue express and secrete the CD59 locally, thereby treating the subject for GA.
- a human sCD59 nucleotide sequence encoding an amino acid sequence corresponding to human sCD59
- the nucleotide sequence includes a mutation encoding for amino acids of a GPI anchoring domain of the protein
- the resulting vector
- the source of expression of human sCD59 protein is administered by ocular injections.
- Injectable preparations such as sterile injectable aqueous or oleaginous suspensions, may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
- the injectable preparation is a sterile injectable preparation.
- the sterile injectable preparation may be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3- butanediol.
- the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P.
- sterile, fixed oils are conventionally employed as a solvent or suspending medium. Any bland, fixed oil can be employed including, but not limited to, synthetic mono- or diglycerides.
- fatty acids such as oleic acid are used in the preparation of injectables.
- the injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
- the sterile injectable preparation includes excipients.
- excipients include, without limitation, suspending agents (e.g., sodium carboxymethylcellulose, methylcellulose, hydroxy-propylmethylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth, and gum acacia), dispersing or wetting agents including, a naturally-occurring phosphatide (e.g., lecithin), or condensation products of an alkylene oxide with fatty acids (e.g., polyoxyethylene stearate), or condensation products of ethylene oxide with long chain aliphatic alcohols (e.g., heptadecaethyl-eneoxycetanol), or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol (e.g., polyoxyethylene sorbitol monooleate), or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol (e.g.,
- the sterile injectable preparation may also be a sterile injectable solution or suspension in a nontoxic parenterally-acceptable or diluent or solvent, for example, as a solution in 1, 3- butanediol.
- a solution generally is considered as a homogeneous mixture of two or more substances; it is frequently, though not necessarily, a liquid.
- the molecules of the solute (or dissolved substance) are uniformly distributed among those of the solvent.
- a suspension is a dispersion (mixture) in which a finely- divided species is combined with another species, with the former being so finely divided and mixed that it does not rapidly settle out. In everyday life, the most common suspensions are those of solids in liquid water.
- Suitable vehicles and solvents that may be employed are water, Ringer’s solution, and isotonic sodium chloride solution.
- sterile, fixed oils are conventionally employed as a solvent or suspending medium.
- suitable vehicles consist of solutions, preferably oily or aqueous solutions, as well as suspensions, emulsions, or implants.
- Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes.
- Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran.
- the suspension also may contain suitable stabilizers or agents, which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
- the active compounds may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
- the liquid dosage forms may contain inert diluents commonly used in the art such as water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, com, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
- ocular- delivered pharmaceutical compositions can also include adjuvants such as wetting agents and emulsifying and suspending agents.
- the pharmaceutical composition of the described invention may be in the form of a sterile injectable aqueous or oleaginous suspension.
- injectable preparations such as sterile injectable aqueous or oleaginous suspensions, may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
- the described invention comprises ophthalmological devices, surgical devices, audiological devices or products which contain disclosed compositions (e.g., gauze bandages or strips), and methods of making or using such devices or products. These devices may be coated with, impregnated with, bonded to, or otherwise treated with the pharmaceutical composition described herein.
- the composition is administered in a pharmaceutically acceptable solution, which may routinely contain pharmaceutically acceptable concentrations of salt, buffering agents, preservatives, compatible carriers, adjuvants, and optionally other therapeutic agents.
- the pharmaceutical composition is an aqueous suspension or emulsion in admixture with excipients suitable for the manufacture of aqueous suspensions and emulsions.
- excipients include, but are not limited to, suspending agents, such as sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth, and gum acacia; dispersing or wetting agents may be a naturally-occurring phosphatide such as lecithin, or condensation products of an alkylene oxide with fatty acids, for example, polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example, heptadecaethyl-eneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of
- Solutions or suspensions used for parenteral, intradermal, subcutaneous, intrathecal, or topical application may include, but are not limited to, a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose.
- the parenteral preparation may be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic. Administered intravenously, particular carriers are physiological saline or phosphate buffered saline (PBS).
- PBS physiological saline or phosphate buffered sa
- the injectable formulations may be sterilized, for example, by filtration through a bacterial-retaining filter or by incorporating sterilizing agents in the form of sterile solid compositions that may be dissolved or dispersed in sterile water or other sterile injectable medium just prior to use.
- Injectable preparations for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
- the sterile injectable preparation also may be a sterile injectable solution, suspension or emulsion in a nontoxic, parenterally acceptable diluent or solvent such as a solution in 1,3-butanediol.
- Suitable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P. and isotonic sodium chloride solution.
- sterile, fixed oils conventionally are employed or as a solvent or suspending medium.
- any bland fixed oil may be employed including synthetic mono- or diglycerides.
- fatty acids such as oleic acid are used in the preparation of injectables.
- Formulations for parenteral administration include aqueous and nonaqueous sterile injection solutions that may contain anti-oxidants, buffers, bacteriostats and solutes, which render the formulation isotonic with the blood of the intended recipient; and aqueous and nonaqueous sterile suspensions, which may include suspending agents and thickening agents.
- the formulations may be presented in unit-dose or multi -dose containers, for example sealed ampules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example, saline, water- for-inj ection, immediately prior to use.
- Suspensions in addition to the active compounds, may contain suspending agents, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar, tragacanth, and mixtures thereof.
- suspending agents for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar, tragacanth, and mixtures thereof.
- the pharmaceutical composition of the described invention may further include conventional excipients, i.e., pharmaceutically acceptable organic or inorganic carrier substances suitable for parenteral application which do not deleteriously react with the active compounds.
- suitable pharmaceutically acceptable carriers include, but are not limited to, water, salt solutions, alcohol, vegetable oils, polyethylene glycols, gelatin, lactose, amylose, magnesium stearate, talc, silicic acid, viscous paraffin, perfume oil; fatty acid monoglycerides and diglycerides, petroethral fatty acid esters, hydroxymethylcellulose, polyvinylpyrrolidone, etc.
- the pharmaceutical composition of the described invention may be sterilized and if desired, mixed with auxiliary agents, e.g., lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, colorings, flavoring and/or aromatic substances and the like which do not deleteriously react with the active compounds.
- suitable vehicles include solutions, such as oily or aqueous solutions, as well as suspensions, emulsions, or implants.
- Aqueous suspensions may contain substances which increase the viscosity of the suspension and include, for example, but not limited to, sodium carboxymethyl cellulose, sorbitol and/or dextran.
- the suspension also may contain stabilizers.
- These compositions also may contain adjuvants including preservative agents, wetting agents, emulsifying agents, and dispersing agents. Prevention of the action of microorganisms may be ensured by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, and the like. It also may be desirable to include isotonic agents, for example, sugars, sodium chloride and the like. Prolonged absorption of the injectable pharmaceutical form may be brought about by the use of agents delaying absorption, for example, aluminum monostearate and gelatin.
- the pharmaceutical composition of the described invention comprises a therapeutically effective amount of human sCD59 and optionally other therapeutic agents included in a pharmaceutically-acceptable carrier.
- the components of the pharmaceutical composition also are capable of being commingled in a manner such that there is no interaction which would substantially impair the desired pharmaceutical efficacy.
- the pharmaceutical composition of the described invention includes a pharmaceutically acceptable salt.
- Pharmaceutically acceptable salts are those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like and are commensurate with a reasonable benefit/risk ratio.
- Pharmaceutically acceptable salts are well-known in the art. For example, P. H. Stahl, et al. describe pharmaceutically acceptable salts in detail in "Handbook of Pharmaceutical Salts: Properties, Selection, and Use" (Wiley VCH, Zurich, Switzerland: 2002).
- EXAMPLE 1 A Phase 2b Clinical Trial to Evaluate Intravitreal JNJ-81201887 (AAVCAGsCD59) Compared to Sham Procedure for the Treatment of Geographic Atrophy (GA) Secondary to Age-related Macular Degeneration (AMD)
- JNJ-81201887 is a recombinant adeno-associated virus serotype 2 (rAAV2) with a transgene encoding soluble CD59 (sCD59).
- Participants meeting all eligibility criteria will be randomized in a 1 : 1 : 1 ratio (stratified by lesion contiguity [multifocal vs non-multifocal] and baseline GA area [2.5 to 10 mm2 or >10 to 17.5 mm2]) to 1 of 2 dose levels of JNJ-81201887 (low dose: 8.2 x 10 10 vg dose; high dose: 4. 1 x 10 11 vg dose) or a sham arm.
- vg vector genome.
- Study participants will undergo an up to 28-day screening period during which all baseline procedures/tests will be performed on or prior to Day 1 /randomization (also start steroid/placebo course after randomization), after which dose administration will occur on Day 4. Participants will be followed for a period of 18 months. The study duration is approximately 19 months.
- At least one focal lesion must be >1.25 mm 2 (0.5 disc area).
- GA can be photographed in its entirety by FAF, using a 30-degree image centered on the fovea.
- GA can be measured separately from any areas of peripapillary atrophy, as assessed by the CRC.
- BCVA Best corrected distance visual acuity
- a woman of childbearing potential must have a negative highly sensitive serum (b-human chorionic gonadotropin) test for the sample collected at Screening and negative urine pregnancy test on Day 4 before receiving the study intervention.
- b-human chorionic gonadotropin b-human chorionic gonadotropin
- a woman must be (a) not of childbearing potential or (b) of childbearing potential and practicing a highly effective method of contraception (failure rate of ⁇ 1% per year when used consistently and correctly) and agrees to remain on a highly effective method throughout the study.
- a male participant must wear a condom when engaging in any activity that allows for passage of ejaculate to a woman of childbearing potential for a period of 18 months from dosing.
- Male participants should also be advised of the benefit for a female partner of childbearing potential to use a highly effective method of contraception, as a condom may break or leak.
- a male participant must agree not to father a child or donate sperm for the purpose of reproduction for a period of 18 months from dosing.
- retinal disease other than GA diabetic retinopathy, central serous chorioretinopathy, inherited retinal degeneration, toxic maculopathies (i.e., hydroxychloroquine maculopathy), arterial and venous occlusive disease, macular hole that is present or has been previously repaired, or choroidal melanoma.
- toxic maculopathies i.e., hydroxychloroquine maculopathy
- arterial and venous occlusive disease macular hole that is present or has been previously repaired, or choroidal melanoma.
- Uncontrolled hypertension defined as systolic blood pressure >170 mm Hg or diastolic blood pressure >100 mm Hg.
- Predisposition or history of increased risk of infection e.g., history of splenectomy or chronic immunosuppression.
- Medical e.g., uncontrolled diabetes, known congestive heart failure, clinically significant valvular heart disease, clinically significant cardiac rhythm or conduction abnormalities
- psychiatric conditions or any condition that may compromise the well-being of the participant or that could prevent, limit, or confound the protocol-specified assessments.
- Group/Arm C Single sham procedure (syringe with no needle) to mimic intravitreal injection in the study eye (placebo) Prophylaxis of intraocular inflammation:
- SD-OCT Spectral Domain Optical Coherence Tomography
- BCVA Best Corrected Visual Acuity
- PROs Functional Reading Independence
- NAI-VFQ-25 National Eye Institute Visual Functioning Questionnaire-25
- Safety evaluations include ocular adverse events of interest (including intraocular inflammation and new onset exudative macular neovascularization [MNV] among others as described in the body of the protocol), systemic adverse events, physical examination (including vital signs), ocular examination, ocular imaging parameters (for example, FAF, SD-OCT, fluorescein angiography, and color fundus photography), laboratory evaluations (clinical chemistry and hematologic measures), and pregnancy testing, if applicable.
- MNV new onset exudative macular neovascularization
- the estimated mean and standard deviation for the change from baseline in GA area (square root transformed) at 18 months is 0.48 mm and 0.27 mm, respectively. Assuming 15% drop out by 18 months, it is estimated that 100 participants per arm will provide 87% power to detect at least one active treatment showing a between-group difference of 0.12 mm (corresponding to a 25% reduction in growth) at significance level of 0.025 (1-sided).
- a mixed model for repeated measures will be used for the primary efficacy analysis of change in sqrt-GAA in the study eye, will include treatment, time (via a categorical variable for visit), treatment-by-time interaction, one indicator variable for stratification factor (GA lesion contiguity) as fixed effects and baseline sqrt-GAA as a co variate.
- An unstructured covariance will be used to model the within subject variability.
- the between-treatment differences in least-squares means and their 2-sided 95% confidence intervals at Month 18 will be presented.
- a Dunnett’s step-down procedure which controls the overall Type 1 error rate (two-sided alpha level of 0.0499 at the final analysis) will be used to test the primary endpoint.
- Safety will be summarized by change from baseline in physical examination (including vital signs), ocular examination, clinical chemistry, hematologic measures, and ocular and systemic treatment-emergent adverse events and SAEs, including ocular adverse events of interest (intraocular inflammation and new onset exudative MNV and others as outlined in the body of this report).
- certain imaging parameters will be assessed on a regular basis to monitor post-injection changes to the retina, including FAF, SD-OCT, fluorescein angiography and color fundus photography and changes in and abnormal ocular examination findings will be summarized descriptively. Observed values and change from baseline in clinical laboratory tests and vital signs will be summarized through Month 18 using descriptive statistics.
- a listing of participants who are positive for antibodies to sCD59 transgene protein and anti-AAV2 viral capsid protein will be provided.
- the titers of antibodies to sCD59 transgene protein or anti-AAV2 viral capsid protein will be summarized for participants who are positive for antibodies to sCD59 transgene protein or anti-AAV2 viral capsid protein.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Epidemiology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Engineering & Computer Science (AREA)
- Ophthalmology & Optometry (AREA)
- Molecular Biology (AREA)
- Biochemistry (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Organic Chemistry (AREA)
- Cell Biology (AREA)
- Zoology (AREA)
- Gastroenterology & Hepatology (AREA)
- Immunology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Abstract
L'invention concerne des méthodes pour traiter l'atrophie géographique (AG) qui est une forme secondaire de la dégénérescence maculaire liée à l'âge (DMLA).
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US202363587230P | 2023-10-02 | 2023-10-02 | |
| US63/587,230 | 2023-10-02 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2025074268A1 true WO2025074268A1 (fr) | 2025-04-10 |
Family
ID=93211862
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/IB2024/059630 Pending WO2025074268A1 (fr) | 2023-10-02 | 2024-10-02 | Méthodes pour traiter la dégénérescence maculaire |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2025074268A1 (fr) |
Citations (29)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US4405712A (en) | 1981-07-01 | 1983-09-20 | The United States Of America As Represented By The Department Of Health And Human Services | LTR-Vectors |
| WO1985005629A1 (fr) | 1984-05-25 | 1985-12-19 | Dana-Farber Cancer Institute | Vecteurs ltr, leurs procedes de preparation et leur utilisation |
| US4650764A (en) | 1983-04-12 | 1987-03-17 | Wisconsin Alumni Research Foundation | Helper cell |
| US4797368A (en) | 1985-03-15 | 1989-01-10 | The United States Of America As Represented By The Department Of Health And Human Services | Adeno-associated virus as eukaryotic expression vector |
| WO1989007150A1 (fr) | 1988-02-05 | 1989-08-10 | The Trustees Of Columbia University In The City Of | Lignees de cellules de conservation de retrovirus et leurs procedes d'utilisation |
| US4861719A (en) | 1986-04-25 | 1989-08-29 | Fred Hutchinson Cancer Research Center | DNA constructs for retrovirus packaging cell lines |
| WO1990002797A1 (fr) | 1988-09-15 | 1990-03-22 | North Carolina State University | Vecteurs retroviraux, auto-inactivants, defectifs au niveau de la replication, et cellules d'assistance pour la production de tels vecteurs |
| WO1990002806A1 (fr) | 1988-09-01 | 1990-03-22 | Whitehead Institute For Biomedical Research | Retrovirus de recombinaison a gammes d'hotes amphotropiques et ecotropiques |
| WO1990013641A1 (fr) | 1989-05-10 | 1990-11-15 | Sloan-Kettering Institute For Cancer Research | Cellules eucaryotes transformees de maniere stable comprenant un adn etranger susceptible d'etre transcrit sous la direction d'un promoteur pol iii |
| US4980289A (en) | 1987-04-27 | 1990-12-25 | Wisconsin Alumni Research Foundation | Promoter deficient retroviral vector |
| WO1992005266A2 (fr) | 1990-09-21 | 1992-04-02 | Viagene, Inc. | Cellules d'encapsidation |
| US5112767A (en) | 1988-03-04 | 1992-05-12 | University Of Southern California | Vectors with enhancer domains |
| WO1992007943A1 (fr) | 1990-10-31 | 1992-05-14 | Somatix Therapy Corporation | Vecteurs de retrovirus efficaces en therapie genique |
| US5122767A (en) | 1991-01-10 | 1992-06-16 | Northern Telecom Limited | Saw device tapped delay lines |
| US5124263A (en) | 1989-01-12 | 1992-06-23 | Wisconsin Alumni Research Foundation | Recombination resistant retroviral helper cell and products produced thereby |
| US5139941A (en) | 1985-10-31 | 1992-08-18 | University Of Florida Research Foundation, Inc. | AAV transduction vectors |
| WO1992014829A1 (fr) | 1991-02-19 | 1992-09-03 | The Regents Of The University Of California | Particules virales agissant sur une gamme d'hotes modifiee |
| WO1993014188A1 (fr) | 1992-01-17 | 1993-07-22 | The Regents Of The University Of Michigan | Virus cible |
| US5624837A (en) | 1994-03-03 | 1997-04-29 | Alexion Pharmaceuticals, Inc. | Nucleic acid encoding chimeric complement inhibitor proteins |
| US5998205A (en) | 1994-11-28 | 1999-12-07 | Genetic Therapy, Inc. | Vectors for tissue-specific replication |
| US5998208A (en) | 1995-07-26 | 1999-12-07 | Children's Medical Center Corporation | Helper virus-free herpesvirus vector packaging system |
| US6677311B1 (en) | 1988-07-20 | 2004-01-13 | The Salk Institute For Biological Studies | Inducible HSV-TK in transformed cell populations |
| US7109029B2 (en) | 2001-02-23 | 2006-09-19 | Cell Genesys, Inc. | Vector constructs |
| US7235391B2 (en) | 1998-11-16 | 2007-06-26 | Introgen Therapeutics, Inc. | Formulation of adenovirus for gene therapy |
| US20070196367A1 (en) | 2006-02-22 | 2007-08-23 | Valentin Dinu | Methods of preventing and treating Alzheimer's disease, age related macular degeneration and other diseases involving extra-cellular debris through the inhibition of the complement system |
| US20100209447A1 (en) | 2007-08-29 | 2010-08-19 | Tufts University | Methods of making and using a cell penetrating peptide for enhanced delivery of nucleic acids, proteins, drugs, and adenovirus to tissues and cells, and compositions and kits |
| US20110015136A1 (en) * | 2008-02-15 | 2011-01-20 | Tufts University | humanized model of membrane attack complex (MAC) formation on murine retina and compositions, kits and methods for treatment of macular degeneration |
| WO2012021891A2 (fr) * | 2010-08-13 | 2012-02-16 | Tufts University | Compositions, kits et méthodes de traitement de pathologies associées au complément |
| WO2023089564A1 (fr) * | 2021-11-19 | 2023-05-25 | Janssen Biotech, Inc. | Méthode de traitement d'une atrophie géographique avec un vecteur de thérapie génique exprimant cd59 soluble |
-
2024
- 2024-10-02 WO PCT/IB2024/059630 patent/WO2025074268A1/fr active Pending
Patent Citations (29)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US4405712A (en) | 1981-07-01 | 1983-09-20 | The United States Of America As Represented By The Department Of Health And Human Services | LTR-Vectors |
| US4650764A (en) | 1983-04-12 | 1987-03-17 | Wisconsin Alumni Research Foundation | Helper cell |
| WO1985005629A1 (fr) | 1984-05-25 | 1985-12-19 | Dana-Farber Cancer Institute | Vecteurs ltr, leurs procedes de preparation et leur utilisation |
| US4797368A (en) | 1985-03-15 | 1989-01-10 | The United States Of America As Represented By The Department Of Health And Human Services | Adeno-associated virus as eukaryotic expression vector |
| US5139941A (en) | 1985-10-31 | 1992-08-18 | University Of Florida Research Foundation, Inc. | AAV transduction vectors |
| US4861719A (en) | 1986-04-25 | 1989-08-29 | Fred Hutchinson Cancer Research Center | DNA constructs for retrovirus packaging cell lines |
| US4980289A (en) | 1987-04-27 | 1990-12-25 | Wisconsin Alumni Research Foundation | Promoter deficient retroviral vector |
| WO1989007150A1 (fr) | 1988-02-05 | 1989-08-10 | The Trustees Of Columbia University In The City Of | Lignees de cellules de conservation de retrovirus et leurs procedes d'utilisation |
| US5112767A (en) | 1988-03-04 | 1992-05-12 | University Of Southern California | Vectors with enhancer domains |
| US6677311B1 (en) | 1988-07-20 | 2004-01-13 | The Salk Institute For Biological Studies | Inducible HSV-TK in transformed cell populations |
| WO1990002806A1 (fr) | 1988-09-01 | 1990-03-22 | Whitehead Institute For Biomedical Research | Retrovirus de recombinaison a gammes d'hotes amphotropiques et ecotropiques |
| WO1990002797A1 (fr) | 1988-09-15 | 1990-03-22 | North Carolina State University | Vecteurs retroviraux, auto-inactivants, defectifs au niveau de la replication, et cellules d'assistance pour la production de tels vecteurs |
| US5124263A (en) | 1989-01-12 | 1992-06-23 | Wisconsin Alumni Research Foundation | Recombination resistant retroviral helper cell and products produced thereby |
| WO1990013641A1 (fr) | 1989-05-10 | 1990-11-15 | Sloan-Kettering Institute For Cancer Research | Cellules eucaryotes transformees de maniere stable comprenant un adn etranger susceptible d'etre transcrit sous la direction d'un promoteur pol iii |
| WO1992005266A2 (fr) | 1990-09-21 | 1992-04-02 | Viagene, Inc. | Cellules d'encapsidation |
| WO1992007943A1 (fr) | 1990-10-31 | 1992-05-14 | Somatix Therapy Corporation | Vecteurs de retrovirus efficaces en therapie genique |
| US5122767A (en) | 1991-01-10 | 1992-06-16 | Northern Telecom Limited | Saw device tapped delay lines |
| WO1992014829A1 (fr) | 1991-02-19 | 1992-09-03 | The Regents Of The University Of California | Particules virales agissant sur une gamme d'hotes modifiee |
| WO1993014188A1 (fr) | 1992-01-17 | 1993-07-22 | The Regents Of The University Of Michigan | Virus cible |
| US5624837A (en) | 1994-03-03 | 1997-04-29 | Alexion Pharmaceuticals, Inc. | Nucleic acid encoding chimeric complement inhibitor proteins |
| US5998205A (en) | 1994-11-28 | 1999-12-07 | Genetic Therapy, Inc. | Vectors for tissue-specific replication |
| US5998208A (en) | 1995-07-26 | 1999-12-07 | Children's Medical Center Corporation | Helper virus-free herpesvirus vector packaging system |
| US7235391B2 (en) | 1998-11-16 | 2007-06-26 | Introgen Therapeutics, Inc. | Formulation of adenovirus for gene therapy |
| US7109029B2 (en) | 2001-02-23 | 2006-09-19 | Cell Genesys, Inc. | Vector constructs |
| US20070196367A1 (en) | 2006-02-22 | 2007-08-23 | Valentin Dinu | Methods of preventing and treating Alzheimer's disease, age related macular degeneration and other diseases involving extra-cellular debris through the inhibition of the complement system |
| US20100209447A1 (en) | 2007-08-29 | 2010-08-19 | Tufts University | Methods of making and using a cell penetrating peptide for enhanced delivery of nucleic acids, proteins, drugs, and adenovirus to tissues and cells, and compositions and kits |
| US20110015136A1 (en) * | 2008-02-15 | 2011-01-20 | Tufts University | humanized model of membrane attack complex (MAC) formation on murine retina and compositions, kits and methods for treatment of macular degeneration |
| WO2012021891A2 (fr) * | 2010-08-13 | 2012-02-16 | Tufts University | Compositions, kits et méthodes de traitement de pathologies associées au complément |
| WO2023089564A1 (fr) * | 2021-11-19 | 2023-05-25 | Janssen Biotech, Inc. | Méthode de traitement d'une atrophie géographique avec un vecteur de thérapie génique exprimant cd59 soluble |
Non-Patent Citations (69)
| Title |
|---|
| "Remington's Pharmaceutical Sciences", 1995, MACK PUBLISHING |
| ALTSCHUL ET AL., NUCLEIC ACIDS RES., vol. 25, 1997, pages 3389 - 3402 |
| ANDWATERMAN, ADV. APPL. MATH, vol. 2, 1981, pages 482 |
| CARPINOHAN, J. ORG. CHEM., vol. 37, 1972, pages 3403 - 3409 |
| CLAVERIESTATES, COMPUT. CHEM, vol. 17, 1993, pages 191 - 201 |
| CLINICALTRIALS.GOV: "Intravitreal AAVCAGsCD59 for Advanced Dry Age-related Macular Degeneration (AMD) With Geographic Atrophy (GA)", 1 January 2021 (2021-01-01), XP093234387, Retrieved from the Internet <URL:https://clinicaltrials.gov/study/NCT04358471> * |
| COHEN ET AL., PROC. NAT'L ACAD. SCI. U.S.A., vol. 90, 1993, pages 7376 |
| CORPET ET AL., NUCLEIC ACIDS RESEARCH, vol. 16, 1988, pages 10881 - 90 |
| COSSET ET AL., J. VIROL, vol. 64, 1990, pages 1070 - 1078 |
| CUKRAS CATHERINE ET AL: "Retinal AAV8-RS1 Gene Therapy for X-Linked Retinoschisis: Initial Findings from a Phase I/IIa Trial by Intravitreal Delivery", vol. 26, no. 9, 5 September 2018 (2018-09-05), pages 2282 - 2294, XP009511982, ISSN: 1525-0016, Retrieved from the Internet <URL:https://www.sciencedirect.com/journal/molecular-therapy> [retrieved on 20180707], DOI: 10.1016/J.YMTHE.2018.05.025 * |
| CUNNINGHAM ET AL., VIROLOGY, vol. 197, 1993, pages 116 |
| DAVIES ET AL., J. EXP. MED., vol. 170, pages 637 - 654 |
| DEUTSCHER, METHODS IN ENZYMOLOGY, 1990, pages 182 |
| DULL ET AL., J. VIROL, vol. 72, 1998, pages 8463 - 8471 |
| EGLITIS ET AL., BIOTECHNIQUES, vol. 6, 1988, pages 608 - 614 |
| EVANS ET AL., GENE, vol. 79, 1989, pages 9 - 20 |
| FIELDSNOBLE, INT. J. PEPT. PROTEIN RES, vol. 35, 1990, pages 161 - 214 |
| FLOTTE ET AL., AM. J. RESPIR. CELL MOL. BIOL, vol. 7, no. 3, 1992, pages 349 - 356 |
| GOODMANGILMAN'S ET AL.: "The Pharmacological Basis of Therapeutics", 1996, MCGRAW-HILL |
| GRAHAM ET AL., J. GEN. VIROL, vol. 36, 1977, pages 59 - 72 |
| HAMILTON ET AL., BLOOD, vol. 76, 1990, pages 2572 - 2577 |
| HENIKOFFHENIKOFF, PROC. NATL. ACAD. SET. USA, vol. 89, 1989, pages 10915 |
| HIGGINSSHARP, CABIOS, vol. 5, 1989, pages 151 - 153 |
| HIGGINSSHARP, GENE, vol. 73, 1988, pages 237 - 244 |
| HOLGUIN ET AL., J. CLIN. INVEST, vol. 84, 1989, pages 7 - 17 |
| HUANG ET AL.: "Computer Applications in the Biosciences", SASSOS, 1992 |
| KAFRI ET AL., NATURE GENET., vol. 17, 1997, pages 314 - 317 |
| KAPLITT ET AL., NAT GENET., vol. 8, no. 2, 1994, pages 148 - 154 |
| KARLINALTSCHUL, PROC. NATL. ACAD. SET. USA, vol. 90, 1993, pages 5873 - 5787 |
| KLEIN ET AL., OPHTHALMOLOGY, vol. 114, 2007, pages 253 - 262 |
| KORMAN ET AL., PROC. NATL. ACAD. SCI. USA., vol. 84, 1987, pages 2150 - 2154 |
| LAFACE ET AL., VIROLOGY, vol. 162, no. 2, 1988, pages 486 |
| LAUGHLIN ET AL., J. VIROL, vol. 60, no. 2, 1986, pages 515 - 524 |
| LEBKOWSKI ET AL., MOL. CELL. BIOL, vol. 8, no. 10, 1988, pages 3988 - 3996 |
| LEVRERO ET AL., GENE, vol. 101, 1991, pages 195 - 202 |
| M. VAN ZIJI ET AL., J. VIROL, vol. 62, no. 6, 1988, pages 1963 - 1973 |
| MERRIFIELD, J. AM. CHEM. SOC., vol. 85, 1963, pages 2149 - 2154 |
| MEYERSMILLER, COMPUTER APPLIC. BIOL. SET, vol. 4, 1988, pages 11 - 17 |
| MILLER ET AL., BIOTECHNIQUES, vol. 7, 1989, pages 981 - 990 |
| MILLER ET AL., MOL. CELL BIOL, vol. 6, 1986, pages 2895 - 2902 |
| MORGENSTERN ET AL., NUCLEIC ACIDS RES., vol. 18, pages 3587 - 3596 |
| MUZYCZKA, CURR. TOP. MICROBIOL. IMMUNOL, vol. 158, no. 97, 1992, pages 129 |
| NALDINI ET AL., SCIENCE, vol. 272, 1996, pages 263 - 267 |
| NEEDLEMANWUNSCH, J. MOL. BIOL., vol. 48, 1970, pages 443 |
| NICHOLAS A. MOORE ET AL: "Gene therapy for age-related macular degeneration", EXPERT OPINION ON BIOLOGICAL THERAPY, vol. 17, no. 10, 20 July 2017 (2017-07-20), pages 1235 - 1244, XP055404429, ISSN: 1471-2598, DOI: 10.1080/14712598.2017.1356817 * |
| O'CONNER, SCIENCE, vol. 244, 1989, pages 1307 - 1313 |
| OHI ET AL., GENE, vol. 89, no. 2, 1990, pages 279 - 282 |
| P. H. STAHL ET AL.: "Handbook of Pharmaceutical Salts: Properties, Selection, and Use", 2002, WILEY VCH |
| PEARSON ET AL., METHODS IN MOLECULAR BIOLOGY, vol. 24, 1994, pages 307 - 331 |
| PEARSONLIPMAN, PROC. NATL. ACAD. SCI., vol. 85, 1988, pages 2444 |
| PENNINGTON, M. WDUNN, B. M: "Peptide Synthesis Protocols", vol. 35, 1995, HUMANA PRESS, INC, article "Methods in Molecular Biology" |
| ROLLINS ET AL., J. IMMUNOL., vol. 144, 1990, pages 3478 - 3483 |
| SAKODA ET AL., J. MOL. CELL. CARDIOL, vol. 31, 1999, pages 2037 - 2047 |
| SAMULSKI ET AL., EMBO J., vol. 10, 1991, pages 3941 - 3950 |
| SAMULSKI ET AL., J. VIROL, vol. 63, 1989, pages 3822 - 3828 |
| SAWADA, R. ET AL., NUCLEIC ACIDS RES, vol. 17, no. 16, 1989, pages 6728 |
| SCOPES: "Protein Purification: Principles and Practice", 1982, SPRINGER- VERLAG |
| SHELLINGSMITH, GENE THERAPY, vol. 1, 1994, pages 261 268 - 169 |
| SIMS ET AL., J. BIOL. CHEM., vol. 264, 1989, pages 19228 - 19235 |
| TOMKINSON ET AL., J. VIROL, vol. 67, 1993, pages 7298 |
| TRATSCHIN ET AL., MOL. CELL. BIOL, vol. 4, 1984, pages 2072 - 2081 |
| TRATSCHIN ET AL., MOL. CELL. BIOL, vol. 5, 1985, pages 3258 - 3260 |
| VAN LECUWEN ET AL., EUR. J. EPIDEMIOL, vol. 18, 2003, pages 845 - 854 |
| WALSH ET AL., J. CLIN. INVEST, vol. 94, 1994, pages 1440 - 1448 |
| WALSH, L. A ET AL., EUR J. IMMOL, vol. 21, no. 3, 1990, pages 847 - 850 |
| YODER ET AL., BLOOD, vol. 82, 1994, pages 347A |
| YU ET AL., JOURNAL OF EXPERIMENTALMEDICINE, vol. 185, no. 4, 1997, pages 745 - 753 |
| ZHOU ET AL., EXP. HEMATOL, vol. 21, 1993, pages 928 - 933 |
| ZUFFEREY ET AL., NATURE BIOTECHNOL., vol. 15, 1997, pages 871 - 875 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| AU2021225178B2 (en) | Treatment of AMD using AAV2 variant with aflibercept | |
| EP4083203B1 (fr) | Compositions et procédés destinés à réduire la néovascularisation oculaire | |
| CN104994882A (zh) | 使用aavsflt-1治疗amd | |
| EP4027983B1 (fr) | Traitement de maladies néovasculaires oculaires à l'aide de variants d'aav2 codant pour l'aflibercept | |
| US20250018063A1 (en) | Method of Treating Geographic Atrophy with a Gene Therapy Vector Expressing Soluble CD59 | |
| US20210100856A1 (en) | Methods of treating ocular neovascular diseases using aav2 variants encoding aflibercept | |
| WO2021050094A1 (fr) | Méthodes de traitement de maladies néovasculaires oculaires à l'aide de variants d'aav2 codant pour l'aflibercept | |
| WO2025074268A1 (fr) | Méthodes pour traiter la dégénérescence maculaire | |
| US20240197915A1 (en) | Methods of treating ocular diseases using aav2 variants encoding aflibercept | |
| JP2024148177A (ja) | アフリベルセプトをコードするaav2バリアントを使用する眼内血管新生疾患を処置する方法 | |
| EP4479098A1 (fr) | Procédé de réduction de fluctuation de cst dans une dmla néovasculaire par un virus adéno-associé recombinant | |
| WO2025061000A1 (fr) | Procédé de traitement d'une maladie oculaire associée à l'angiogenèse chez un sujet | |
| JP2025534347A (ja) | 抗vegf fabをコードする組換えウイルスベクターを用いる眼疾患の処置 | |
| CN118946370A (zh) | 重组腺相关病毒减少新生血管性amd中的cst波动的方法 |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 24793907 Country of ref document: EP Kind code of ref document: A1 |