WO2020102680A1 - Antibody-based therapeutics for targeting htra1 and methods of use - Google Patents

Antibody-based therapeutics for targeting htra1 and methods of use Download PDF

Info

Publication number
WO2020102680A1
WO2020102680A1 PCT/US2019/061740 US2019061740W WO2020102680A1 WO 2020102680 A1 WO2020102680 A1 WO 2020102680A1 US 2019061740 W US2019061740 W US 2019061740W WO 2020102680 A1 WO2020102680 A1 WO 2020102680A1
Authority
WO
WIPO (PCT)
Prior art keywords
antigen
antibody
binding fragment
seq
acid sequence
Prior art date
Application number
PCT/US2019/061740
Other languages
French (fr)
Inventor
Scott Lauder
Original Assignee
Gemini Therapeutics Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Gemini Therapeutics Inc. filed Critical Gemini Therapeutics Inc.
Publication of WO2020102680A1 publication Critical patent/WO2020102680A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding

Definitions

  • Age-related macular degeneration is a medical condition and is the leading cause of legal blindness in Western societies. AMD typically affects older adults and results in a loss of central vision due to degenerative and neovascu!ar changes to the macula, a pigmented region at the center of the retina which is responsible for visual acuity.
  • AMD typically affects older adults and results in a loss of central vision due to degenerative and neovascu!ar changes to the macula, a pigmented region at the center of the retina which is responsible for visual acuity.
  • AMD is identified by the focal hyperpigmentation of the retinal pigment epithelium (RPE) and accumulation of drusen deposits. The size and number of drusen deposits typically correlates with AMD se verity.
  • RPE retinal pigment epithelium
  • AMD occurs in up to 8% of individuals over the age of 60, and the prevalence of AMD continues to increase with age.
  • the U.S. is anticipated to have nearly 22 million cases of AMD by the year 2050, while global cases of AMD are expected to be nearly 288 million by the year 2040.
  • the disclosure provides for an antibody or antigen-binding fragment thereof that binds to a HTRA1 protein, or a functional fragment thereof.
  • the antibody or antigen-binding fragment also binds to one or more of HTRA2, HTRA3, or HTRA4. In some embodiments, the antibody or antigen-binding fragment also binds to HTRA4. In some embodiments, the antibody or antigen-binding fragment does not bind to, or displays weak binding to, HTRA2, HTRA3 and HTRA4.
  • the HTRA1 protein is a human HTRA1 protein, or functional fragment thereof. In some embodiments, the HTRA 1 protein comprises an amino acid sequence that is at least 80%, 85%, 90%, 92%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to the ammo acid sequence of SEQ ID NO: 121 or a functional fragment thereof.
  • the antibody or antigen-binding fragment thereof is capable of inhibiting any one or more HTRA1 function. In some embodiments, the antibody or antigen-binding fragment thereof is capable of inhibiting HTRA 1 proteolytic activity by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the proteolytic activity of a wildtype HTRA1 protein in the absence of the HTRA1 antibody or antigen-binding fragment thereof.
  • the antibody or antigen-binding fragment thereof is capable of inhibiting HTRA1 proteolytic activity in a cell by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the proteolytic activity of a wildtype HTRA 1 protein in the same cell type in the absence of the HTRA 1 antibody or antigen-binding fragment thereof.
  • the antibody or antigen-binding fragment thereof is capable of inhibiting HTRA1 proteolytic activity in any eye by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%,
  • the antibody or antigen-binding fragment is capable of reducing HTRAl’s ability to cleave any one or more HTRA1 substrate.
  • the HTRA1 substrate is selected from the group consisting of: fibromodulin, elastin, clusterin, ADAM9, vitronectin, a2 -macroglobulin, talin- 1, fascin, LTBP-1, EFEMPI, fibulin 5, tau, RseA, and chloride intracellular channel protein.
  • the antibody or antigen-binding fragment is capable of inhibiting HTRA l’s ability to cleave a regulator of the complement cascade (e.g., vitronectin, fibromodulin or clusterin).
  • the antibody or antigen-binding fragment is capable of inhibiting HTRAl such that HTRA l’s ability to cleave an HTRA1 substrate and/or regulator of the complement cascade is reduced by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the ability of the HTRAl to cleave the HTRAl substrate and/or regulator of the complement cascade in the absence of the anti -HTRAl antibodies or antigen-binding fragments thereof.
  • the antibody or antigen-binding fragment is capable of inhibiting the ability of the HTRAl protein to tnmerize.
  • the antibody or antigen-binding fragment is capable of inhibiting the ability of the HTRA1 protein to trimerize by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the ability of the HTRA1 protein to trimerize in the absence of the antibody or antigen-binding fragment.
  • the antibody or antigen-binding fragment thereof comprises any one of, or combination of, the CDR sequences indicated in Figure 2.
  • the antibody or antigen-binding fragment thereof comprises any one of, or any combination of, the CDR a o acid sequences of SEQ ID NOs: 1-120.
  • the antibody or antigen-binding fragment thereof comprises a set of CDRS as forth as any one of the following sets of SEQ ID NOs: 1-6, 7-12, 13-18, 19-24, 25-30, 31 -36, 37-42, 43-48, 49-54, 55-60, 61-66, 67-72, 73-78, 79-84, 85-90, 91-96, 97-102, 103-108, 109-114, or 1 15-120.
  • the antibody or antigen-binding fragment thereof comprises the CDRs set forth as SEQ ID NOs: 1-6.
  • the antibody or antigen-binding fragment thereof comprises the CDRs set forth as SEQ ID NOs: 7-12.
  • the antibody or antigen-binding fragment thereof comprises the CDRs set forth as SEQ ID NOs: 19-24. In some embodiments, the antibody or antigen-binding fragment thereof comprises the CDRs set forth as SEQ ID NOs: 31-36. In some embodiments, the antibody or antigen-binding fragment thereof comprises the CDRs set forth as SEQ ID NOs: 37-42 In some embodiments, the antibody or antigen-binding fragment thereof comprises the CDRs set forth as SEQ ID NOs: 61-66. In some embodiments, the antibody or antigen-binding fragment thereof comprises the CDRs set forth as SEQ ID NOs: 73-78 In some
  • the antibody or antigen-binding fragment thereof comprises the CDRs set forth as SEQ ID NOs: 103-108. In some embodiments, the antibody or antigen-binding fragment thereof comprises the CDRs set forth as SEQ ID NOs: 109-114. In some embodiments, the antibody or antigen-binding fragment thereof comprises the CDRs set forth as SE1Q ID NOs:
  • the antibody or antigen-binding fragment thereof comprises any one of or combination of the amino acid sequences of SEQ ID NO: 1-120, but wherein the amino acid sequences comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 amino acid substitutions.
  • the antibody or antigen-binding fragment thereof comprises a set of CDRS as forth as any one of tire following sets of SEQ ID NOs: 1-6, 7-12, 13-18, 19-24, 25-30, 31-36, 37-42, 43-48, 49-54, 55-60, 61-66, 67-72, 73-78, 79-84, 85-90, 91-96, 97-102, 103-108, 109-114, or 1 15-120 but wherein the amino acid sequences comprise at least 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , or 12 amino acid substitutions.
  • die substitutions are conservative substitutions.
  • die substitutions either increase the binding affinity of the antibody or antigen-binding fragment to the HTRA1 epitope, or the substitutions result in a reduction of binding affinity of no more than 5%,
  • the antibody or antigen-binding fragment is a full-length antibody. In some embodiments, the antibody or antigen-binding fragment is a monoclonal antibody. In some embodiments, the antibody or antigen-binding fragment is a humanized antibody. In some embodiments, the antibody or antigen-binding fragment is an antigen-binding fragment thereof. In some embodiments, the antigen-binding fragment is an scFv.
  • tire antigen-binding fragment is selected from the group consisting of: a Fab fragment, a F(ab')2 fragment, a Fab ' fragment, dAb fragment and/or a dsFv.
  • the antibody or antigen-binding fragment binds to one or more of the following HTRA1 domains: an insulin-like growth factor binding domain, a kazal domain, a trypsin-like peptidase domain, and/or a PDZ domain.
  • the antibody or antigen-binding fragment is capable of binding to a non-human species HTRA1.
  • the non-human species HTRA I is mouse, rat, rabbit, cow, monkey (e.g., eynomo!gus monkey), or ape (e.g. chimpanzee) HTRA! protein.
  • the disclosure provides for a polynucleotide encoding any of the antibodies or antigen-binding fragments disclosed herein.
  • the disclosure provides for a vector comprising any of the polynucleotides.
  • the disclosure provides for a host cell comprising any of the vectors disclosed herein and capable of expressing the polynucleotide.
  • the disclosure provides for a host cell expressing one or more of any of the polynucleotides disclosed herein encoding any of the antibody or antigen- binding fragments disclosed herein.
  • the disclosure provides for a method of treating a disease or disorder in a subject in need thereof, wherein the disease or disorder is associated with aberrantly expressed HTRA1, wherein the method comprises administering to the subject any of the antibodies or antigen-binding fragments disclosed herein. In some embodiments, the disclosure provides for a method of treating a disease or disorder in a subject in need thereof, wherein HTRA!
  • the disclosure provides for a method of treating age-related m acular degeneration or polyploidal choroidal vaseu!opathy in a subject in need thereof, wherein the disease or disorder is associated with aberrantly expressed HTRA1, wherein the method comprises administering to the subject any of the antibodies or antigen-binding fragments disclosed herein.
  • control subject is a subject of the same sex and/or of similar age as the subject having the disease or disorder.
  • the subject has one or more mutations in the HTRA i gene.
  • the one or more mutations are not in the coding sequence for the H ' TRAl gene.
  • the one or more mutations are in 10q26 in a human subject.
  • the one or more mutations correspond to any one or more of tlie following polymorphisms in a human subject: rs61871744; rs59616332; rsl 1200630; rs61871745; rsl 1200632: rsl 1200633: rs61871746; rs61871747; rs370974631; rs200227426; rs201396317; rsl99637836; rsl 1200634; rs75431719; rs!0490924; rs!44224550;
  • the subject has age-related macular degeneration. In some embodiments, the subject has wet AMD. In some embodiments, the subject has dry AMD. In some embodiments, the subject has polyploidal choroidal vasculopathy. In some embodiments, the subject is a human. In some
  • the human is at least 40 years of age. In some embodiments, the human is at least 50 years of age. In some embodiments, the human is at least 65 years of age. In some embodiments, the antibody or antigen-binding fragment is administered locally. In some embodiments, the antibody or antigen-binding fragment is administered intravitreaily. In some embodiments, the antibody or antigen-binding fragment is administered subretinally.
  • the antibody or antigen-binding fragment is administered
  • the disclosure provides for a composition comprising any of the antibodies or antigen -binding fragments disclosed herein and a pharmaceutically acceptable carrier.
  • the composition is substantially pyrogen free.
  • Figure 1 provides the experimental summary data from Example 1 for various specific anti- HTKA1 antibody /antigen-bmding fragment clones.
  • Figure 2 provides the heavy chain and light chain CDR amino acid sequences for the clones referenced in Figure 1.
  • FIG 3A and Figure 3B are graphs showing HTRA1 -mediated proteolysis of F AM-labeled elastin over time in the presence and absence of anti-HTRAl monoclonal antibodies (mAbs). Proteolysis is indicated by an increase in relative fluorescence units (RFU) over time.
  • Figure 3C show's a bar graph summarizing the results from Figures 3 A and 3B relative to the antibod -free HTRA1 positive control reaction.
  • Recombinant HTRA1 used in this assay contained a StreplI affinity tag and deletion of the HTRA1 N-terminal Mac domain (StrepII-AMac-HTRAl).
  • Figure 4 A shows Western Blot gel images from four time points for StrepII-AMac-HTRAl -mediated proteol sis of Fibuiin 5 in die presence and absence of anti-HTRAl mAbs over die course of 22 hours.
  • Each proteolysis reaction is shown as a lane on a 12% SDS-PAGE gel. The contents of each reaction are tabulated above the gels.
  • bands representing Fibuiin 5, anti-HTRAl mAb heavy chain, HTRA1, and anti-HTRAl mAb light chain are shown.
  • Figure 4B show's a summary table correlating each anti-HTRAl mAb to its respective number indicated in the gels of Figure 4 A.
  • Figure 5 A shows Western Blot gel images from three time points for StrepII-AMac-HTRA 1 -mediated proteolysis of Tan in the presence and absence of anti-HTRA 1 mAbs over the course of 22 hours.
  • Each proteolysis reaction is shown as a lane on a 12% SDS-PAGE gel. The contents of each reaction are tabulated above the gels. In each lane, bands representing Tan, anti-HTRAl mAb heavy chain, HTRAL and anti-HTRAl mAb light drain are shown.
  • Figure 5B shows a summary table correlating each anti-HTRAl mAb to its respective number indicated in the gels of Figure 5 A.
  • Figure 6A show's Western Blot gel images from three time points for StrepII-AMac-HTRAl -mediated proteolysis of RseA in the presence and absence of anti-HTRAl mAbs over tire course of seven hours.
  • Each proteolysis reaction is shown as a lane on a 12% SDS-PAGE gel. The contents of each reaction are tabulated above the gels. In each lane, bands representing anti-HTRAl mAb heavy chain,
  • HTRA 1, anti-HTRAl mAb light chain, and RseA are shown.
  • Figure 6B show's a summary table correlating each anti-HTRAl mAb to its respective number indicated in the gels of Figure 6A.
  • Figure 7 A shows Western Blot gel images from three time points for StrepII-AMac-HTRA 1 -mediated proteolysis of b-Casein in the presence and absence of anti-HTRAl niAbs over the course of thirty minutes. Each proteolysis reaction is shown as a lane on a 12% SDS-PAGE gel. The contents of each reaction are tabulated above the gels.
  • FIG. 7B shows a summary' table correlating each anti-HTRA l mAb to its respective number indicated in the gels of Figure 7 A.
  • Figure 8 is a table summarizing the results of eadi HTRAl proteolysis assay correlated to their respective anti-HTRAl antibody name and lot number.
  • coluimi 2 the percent inhibition of elastin proteolysis is shown for each antibody used relative to the antibody -free HTRA1 positive control reaction.
  • Inhibition of b-Casein, Tau, RseA, or Fibuiin 5 proteolysis is represented by +.
  • Below or above average inhibition of proteolysis is represented by +/- or ++ respectively.
  • No inhibition of proteolysis is represented by -.
  • the anti-HTRA l SR8047-23 mAb activated StrepII-AMac-HTRA 1- mediated proteolysis of Tau, and this effect is indicated as“Activation”.
  • Figure 9 A is a graph showing StreplI-AMac-HTRAl-mediated proteolysis of F AM-labeled elastin over time in the presence and absence of anti-HTRAl Fab antibody fragments, or anti-HTRA l IgG antibodies. Proteolysis is indicated by an increase in relative fluorescence units (RFU) over time.
  • Figure 9B shows a graph of the lA ax for each reaction shown in Figure 9A.
  • the disclosure provides compositions and methods for treating, preventing, or inhibiting diseases of the eye.
  • the disclosure provides anti-HTRAl antibodies or antigen binding fragments thereof.
  • the disclosure provides methods of treating, preventing, or inhibiting diseases of the eye by intraocularly (e.g., intravitreaily) administering an effective amount of any of the anti-HTRA l antibodies or antigen-binding fragments thereof disclosed herein.
  • a wide variety of diseases of the eye may be treated or prevented using any of the anti- HTRA 1 antibodies or antigen-binding fragments thereof and methods provided herein.
  • Diseases of the eye that may be treated or prevented using the anti-HTRAl antibodies or antigen-binding fragments thereof and methods of the disclosure include but are not limited to, glaucoma, macular degeneration (e.g., age-related macular degeneration), diabetic retinopathies, inherited retinal degeneration such as retinitis pigmentosa, retinal detachment or injury and retinopathies (such as retinopathies that are inherited, induced by surgery, trauma, an underlying aetiology such as severe anemia, SLE, hypertension, blood dyscrasias, systemic infections, or underlying carotid disease, a toxic compound or agent, or photically).
  • macular degeneration e.g., age-related macular degeneration
  • diabetic retinopathies e.g., diabetic
  • the present disclosure encompasses not only the entire group listed as a whole, but each member of the group individually and all possible subgroups of the main group, but also the main group absent one or more of the group members.
  • the present disclosure also envisages the explicit exclusion of one or more of any of the group members in the disclosure.
  • residue refers to a position in a protein and its associated amino acid identity.
  • polynucleotide refers to chains of nucleotides of any length, and include DNA and RNA.
  • the nucleotides can be deoxyribonucleotides, ribonucleotides, modified nucleotides or bases, and/or their analogs, or any substrate that can be incorporated into a chain by DNA or RNA polymerase.
  • a polynucleotide may comprise modified nucleotides, such as methylated nucleotides and their analogs if present, modification to the nucleotide structure may be imparted before or after assembly of the chain.
  • the sequence of nucleotides may be interrupted by non-nucleotide components.
  • a polynucleotide may be further modified after polymerization, such as by conjugation with a labeling component.
  • Other types of modifications include, for example, “caps”, substitution of one or more of the naturally occurring nucleotides with an analog, intemucleotide modifications such as, for example, those with uncharged linkages (e.g., methyl phosphonates, phosphotriesters, phosphoamidates, carbamates, etc.) and with charged linkages (e.g., phosphorothioates, phosphorodithioates, etc.), those containing pendant moiefies, such as, for example, proteins (e.g , nucleases, toxins, antibodies, signal peptides, poly-L-lysine, etc.), those with intercalators (e.g., acridine, psoralen, etc.), those containing chelators (e.g.
  • any of the hydroxyl groups ordinarily present in the sugars may be replaced, for example, by phosphonate groups, phosphate groups, protected by standard protecting groups, or activated to prepare additional linkages to additional nucleotides, or may be conjugated to solid supports.
  • the 5’ and 3’ terminal OH can be phosphorylated or substituted with amines or organic capping group moieties of from 1 to 20 carbon atoms.
  • Other hydroxyls may also be derivatized to standard protecting groups.
  • Polynucleotides can also contain analogous forms of ribose or deoxyribose sugars that are generally known in the art, including, for example, 2 , -0-methyl-, 2’-0-allyl, 2’-fluoro- or 2 - azido-ribose, carbocyclic sugar analogs, alpha- or beta-anomeric sugars, epimeric sugars such as arabmose, xyloses or lyxoses, pyranose sugars, furanose sugars, sedoheptuioses, acyclic analogs and abasic nucleoside analogs such as methyl riboside.
  • One or snore pliosphodiester linkages may be replaced by alternative linking groups. These alternative linking groups include, but are not limited to, embodiments wherein phosphate is replaced by
  • each R or R is independently H or substituted or unsubstituted alkyl (1-20 C) optionally containing an ether (-0-) linkage, aryl, alkenyl, cycloalkyl, cycloalkenyl or ara!dyi. Not all linkages in a polynucleotide need be identical. The preceding description applies to all polynucleotides referred to herein, including RNA and DNA.
  • a“base”,“nucleotide base,” or“nucleobase,” is a heterocyclic pyrimidine or purine compound, which is a standard constituent of all nucleic acids, and includes the bases that form the nucleotides adenine (A), guanine (G), cytosine (C), thymine (T), and uracil (U).
  • a nucleobase may further be modified to include, without limitation, universal bases, hydrophobic bases, promiscuous bases, size -expanded bases, and fluorinated bases.
  • nucleotide can include a modified nucleotide (such as, for example, a nucleotide mimic, abasic residue (Ab), or a surrogate replacement moiety).
  • sequence and“nucleotide sequence” mean a succession or order of nucleobases or nucleotides, described with a succession of leters using standard nomenclature.
  • polypeptide oligopeptide
  • peptide protein
  • Tire chain may be linear or branched, it may comprise modified amino acids, and/or may be interrupted by non-amino acids.
  • the terms also encompass an a mo acid chain that has been modified naturally or by
  • polypeptides containing one or more analogs of an amino acid including, for example, unnatural amino acids, etc.
  • the polypeptides can occur as single chains or associated chains.
  • the term“homologous,” when modified with an adverb such as“highly,” may refer to sequence similarity and may or may not relate to a common evolutionary origin.
  • sequence similarity in all its grammatical forms, refers to the degree of identity or correspondence between nucleic acid or a mo acid sequences that may or may not share a common evolutionary' origin.
  • Percent (%) sequence identity or“percent (%) identical to” with respect to a reference polypeptide (or nucleotide) sequence is defined as the percentage of amino acid residues (or nucleic acids) in a candidate sequence that are identical with the ammo acid residues (or nucleic acids) in the reference polypeptide (nucleotide) sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for aligning sequences, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared.
  • a“host cell” includes an individual cell or cell culture that comprises one or more polynucleotides capable of expressing any of the anti-HTRAl antibodies or antigen binding fragments thereof disclosed herein.
  • a "vector,” refers to a recombinant plasmid comprising one or more polynucleotides encoding any of, or any component of, the anti-HTRAl antibodies or antigen-binding fragments thereof disclosed herein.
  • purify refers to the removal, whether completely or partially, of at least one impurity from a mixture containing the polypeptide and one or more impurities, which thereby improves the level of purity of the polypeptide in the composition (i.e., by decreasing the amount (ppm) of impurity(ies) in the composition).
  • substantially pure refers to material which is at least 50% pure (i.e., free from contaminants), more preferably, at least 90% pure, more preferably, at least 95% pure, yet more preferably, at least 98% pure, and most preferably, at least 99% pure.
  • the temis “patient”,“subject”, or“individual” are used interchangeably herein and refer to either a human or a non-human animal. These terms include mammals, such as humans, non-human primates, laboratory animals, livestock animals (including bovines, porcines, camels, etc.), companion animals (e.g., canines, felines, other domesticated animals, etc.) and rodents (e.g., mice and rats).
  • the subject is a human that is at least 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90 or 95 years of age.
  • the subject has, or is at risk of developing a disease of the eye.
  • a disease of the eye includes, without limitation, AMD, retinitis pigmentosa, rod-cone dystrophy, Leber’s congenital amaurosis, Usher's syndrome, Bardet-Biedl Syndrome, Best disease, retinoschisis, Stargardt disease (autosomal dominant or autosomal recessive), untreated retinal detachment, pattern dystrophy, cone-rod dystrophy, achromatopsia, ocular albinism, enhanced S cone syndrome, diabetic retinopathy, age-related macular degeneration, retinopathy of prematurity, sickle ceil retinopathy, Congenital Stationary Night Blindness, glaucoma, or retinal vein occlusion.
  • the subject has, or is at risk of developing glaucoma, Leber's hereditary optic neuropathy, lysosomal storage disorder, or peroxisomal disorder.
  • the subject is in need of optogenetic therapy.
  • the subject has shown clinical signs of a disease of the eye.
  • the subject has, or is at risk of developing AMD.
  • the AMD is Early AMD; Intermediate AMD; Advanced non-neovascular (“Dry ' ”) AMD; or Advanced neovascular (“Wet”) AMD.
  • Clinical signs of a disease of the eye include, but are not limited to, decreased peripheral vision, decreased central (reading) visi on, decreased night visi on, loss of color perception, reduction in visual acuity, decreased photoreceptor function, and pigmentary changes.
  • the subject shows degeneration of the outer nuclear layer (ONL).
  • the subject has been diagnosed with a disease of the eye.
  • the subject has not yet shown clinical signs of a disease of the eye.
  • the terms“prevent”,“preventing” and“prevention” refer to the prevention of the recurrence or onset of, or a reduction in one or more symptoms of a disease or condition (e.g., a disease of the eye) in a subject as result of the administration of a therapy (e.g., a prophylactic or therapeutic agent).
  • a therapy e.g., a prophylactic or therapeutic agent
  • “prevent”,“preventing” and“prevention” refer to the inhibition or a reduction in the development or onset of a disease or condition (e.g., a disease of the eye), or the prevention of the recurrence, onset, or development of one or more symptoms of a disease or condition (e.g., a disease of the eye), in a subject resulting from the administration of a therapy (e.g., a prophylactic or therapeutic agent), or the administration of a combination of therapies (e.g., a combination of prophylactic or therapeutic agents).
  • “Treating” a condition or patient refers to taking steps to obtain beneficial or desired results, including clinical results.
  • treatment refers to the reduction or amelioration of the progression, severity, and/or duration of an infection (e.g., a disease of the eye or symptoms associated therewith), or the amelioration of one or more symptoms resulting from the administration of one or more therapies (including, but not limited to, the administration of one or more prophylactic or therapeutic agents).
  • administering or“administration of’ a substance, a compound or an agent (e.g., any of the antibodies or antigen-binding fragments disclosed herein) to a subject can be carried out using one of a variety of methods known to those skilled in the art.
  • a compound or an agent can be administered intravitreally or subretinally.
  • the compound or agent is administered intravitreally.
  • administration may be local. In other embodiments, administration may be systemic.
  • Administering can also be performed, for example, once, a plurality of times, and/or over one or more extended periods.
  • the administration includes both direct administration, including self-administration, and indirect administration, including the act of prescribing a drug.
  • direct administration including self-administration
  • indirect administration including the act of prescribing a drug.
  • a physician who instructs a patient to self- administer a drug, or to have the drug administered by another and/or who provides a patient with a prescription for a drug is administering the drug to the patient.
  • the term“ocular cells” refers to any cell in, or associated with tire function of, the eye.
  • the term may refer to any one or more of photoreceptor cells, including rod, cone and photosensitive ganglion cells, retinal pigment epithelium (RPE) cells, glial cells, Muller cells, bipolar cells, horizontal cells, amacrine cells.
  • the ocular cells are bipolar cells.
  • the ocular cells are horizontal cells.
  • the ocular cells are ganglion cells.
  • tire cells are RPE cells.
  • the term“capable of’ means that the referenced composition (e.g.
  • any of the antibodies or antigen-binding fragments disclosed herein has the capability to perform a specific function, but that it is not required to be performing that specific function at any specific moment in time.
  • the term“capable of’ encompasses instances where the composition is actively performing a specific function.
  • Each embodiment described herein may be used individually or in combination with any oilier embodiment described herein.
  • HTRA 1 is a serine protease that targets a variety of proteins, including extracellular matrix proteins such as fibronectin. Fibronectin fragments resulting from HTRA1 cleavage are able to further induce synovial cells to up-regulate MMP1 and MMP3 production. There is evidence that HTRA1 may also degrade proteoglycans, such as aggrecan, decorin and fibromodulin. By cleaving proteoglycans, HTRA1 may release soluble FGF- glycosaminoglycan complexes that promote the range and intensity of FGF signals in the extracellular space HTRA 1 also regulates the availability of insulin-like growth factors (IGFs) by cleaving IGF-binding proteins. Intracellularly, HTRA1 degrades TSC2, leading to the activation of TSC2 downstream targets.
  • IGFs insulin-like growth factors
  • HTRA1 alters the integrity of Bruch’s membrane, which permits choroid capillaries to invade across the extracellular matrix in conditions such as wet age-erlated macular degeneration. Tong et ah, 2010, Mol . Vis , 16: 1958-81. HTR.41 also inhibits signaling mediated by TGF-beta family members, which may regulate many physiological processes, including retinal angiogenesis and neuronal survival and maturation during development. It has been previously determined that a single-nucleotide polymorphism (is 11200638) in the promoter region of the HTRA I gene was found to be significantly associated with susceptibility to AMD in various patient populations. Tong et al., 2010.
  • the antibody or antigen-binding fragment also binds to one or more of HTRA2, HTRA3, or HTRA4. In some embodiments, the antibody or antigen-binding fragment also binds to HTRA4. In some embodiments, the antibody or antigen -binding fragment does not bind to, or displays 'eak binding to, HTRA2, HTRA3 and HTRA4.
  • any of the anti-HTRA l antibodies or antigen-binding fragments thereof disclosed herein is capable of inhibiting any one or more HTRAi function.
  • the anti-HTRA 1 antibodies or antigen-binding fragments thereof are capable of inhibiting all functions of an HTRA 1 protein.
  • the anti-HTRAl antibodies or antigen-binding fragments thereof are capable of binding to an HTRAi protein and inhibiting the function of the HTRAI protein.
  • any of the anti-HTRAl antibodies or antigen-binding fragments thereof disclosed herein is capable of inhibiting proteolytic activity of an HTRAI protein.
  • the anti-HTRAl antibodies or antigen-binding fragments thereof is capable of inhibiting HTRAI proteolytic activity' by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the proteolytic activity of a wildtype HTRAI protein (e.g., an HTRA I protein having the amino acid sequence of SEQ ID NO: 121) in the absence of the HTRAI antibody' or antigen- binding fragment thereof.
  • a wildtype HTRAI protein e.g., an HTRA I protein having the amino acid sequence of SEQ ID NO: 121
  • the anti-HTRAl antibodies or antigen- binding fragments thereof are capable of inhibiting HTRA1 proteolytic activity in a cell by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the proteolytic activity of a wildtype HTRAl protein in the same cell type in the absence of the antibody or antigen-binding fragment.
  • the anti-HTRAl antibodies or antigen-binding fragments thereof are capable of inhibiting HTRAl proteolytic activity in an eye by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the proteolytic activity of a wildtype HTRA l protein in another eye in the absence of the antibody or antigen-binding fragment.
  • any of the anti-HTRA l antibodies or antigen-binding fragments thereof disclosed herein is capable of inhibiting HTRAl’ s ability to cleave any one or more HTRAl substrate.
  • the HTRAl substrate is selected from the group consisting of: fibromodulin, clusterin, ADAM9, elastin, vitronectin, a2-macroglobulin, talin- 1, fascin, LTBP-1, EFEMPl, fihulin 5, tau, RseA, and chloride intracellular channel protein.
  • the anti-HTRAl antibodies or antigen-binding fragments thereof are capable of inhibiting HTRAl’s ability to cleave a regulator of the complement cascade (e.g., vitronectin, fibromodulin or clusterin).
  • a regulator of the complement cascade e.g., vitronectin, fibromodulin or clusterin.
  • the anti-HTRAl antibodies or antigen-binding fragments thereof are capable of inhibiting HTRA I such that HTRA 1’ s ability to cleave an HTRAl substrate and/or regulator of the complement cascade is reduced by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the ability 7 of the HTRAl to cleave the HTRAl substrate and/or regulator of the complement cascade m the absence of the anti- HTRA 1 antibodies or antigen-binding fragments thereof.
  • immunoglobulin means a tetramer comprising two heavy (H) chains (about 50-70 kDa) and two light (L) chains (about 25 kDa) inter-connected by disulfide bonds. There are two types of light chain: l and K. In humans they are similar, but only one type is present in each antibody. Heavy chains are classified as mu, delta, gamma, alpha, or epsilon, and define the antibody's isotype as IgM, IgD, IgG, IgA, and IgEl respectively. See generally,
  • Each heavy chain (herein sometimes referred to as H-chain or He) is compri sed of a heavy chain variable domain (VH, or H-variable domain) and a heavy chain constant region (CH).
  • the heavy chain constant region is comprised of three domains, CHI, CH2 and CH3.
  • Each light chain (herein sometimes referred to as L-chain or Lc) is comprised of a light chain variable domain (VL, or L-variable domain) and a light chain constant region.
  • the light chain constant region is comprised of one domain, CL.
  • variable and constant regions are joined by a“J” region of about 12 or more amino acids, with the heavy chain also including a“D” region of about 3 or more amino acids.
  • VH and VI, regions can be further subdivided into regions of hypervariability', termed“complementarity determining regions”’ (CDR), interspersed with regions that are more conserved, termed “framework regions” (FR).
  • CDR complementarity determining regions
  • Each VH and VL is composed of three CDRs (H-CDR herein designates a CDR from the heavy chain; and L-CDR herein designates a CDR from the light chain) and four FRs, arranged from amino-terminus to carboxyl- terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDRS, FR4.
  • any of the antibodies or antigen-binding fragments disclosed herein comprises 1, 2, 3, 4, 5, or 6 of the CDRs from any of the antibodies or antigen-binding fragments disclosed herein. In some embodiments, any of the antibodies or antigen-binding fragments disclosed herein comprises 1 , 2 or 3 of the CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to any one of SEQ ID NOs: 125-144. In some embodiments, the antibody' or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 125.
  • the antibody or antigen-binding fragment comprises 1 , 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 126. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain am o acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 127. In some embodiments, the antibody or antigen-binding fragment comprises i, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 128.
  • the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 129. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 130. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 131.
  • the antibody or antigen -binding fragment comprises 1 , 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 132. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 133 In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 134.
  • the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 135. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 136. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 137.
  • the antibody or antigen-binding fragment comprises 1 , 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 138. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 139. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 140.
  • the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain ammo acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 141. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 142. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 143.
  • the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 144.
  • any of the antibodies or antigen-binding fragments disclosed herein comprises 1, 2 or 3 of the CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to any one of SEQ ID NOs: 145-164.
  • the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 145. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 146. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain ammo acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 147.
  • the antibody or antigen- binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 148. In some embodiments, the antibody or antigen-binding fragment comprises 1 , 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 149. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 150.
  • the antibody or antigen-bindmg fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 151. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 152. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 153.
  • tire antibody or antigen-binding fragment comprises 1 , 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 154. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 155. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 156.
  • the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 157. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 158. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 159.
  • the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 160. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 161. In some embodiments, the antibody or antigen-binding fragment comprises 1 , 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 162.
  • the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to S EQ ID NO: 163. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 164. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from the sequence of SEQ ID NO: 143. In some embodiments, the antibody or antigen-binding fragment comprises 3 CDRs from the sequence of SEQ ID NO: 143.
  • the antibody or antigen-binding fragment comprises l, 2 or 3 CDRs from tire sequence of SEQ ID NO: 144. In some embodiments, the antibody or antigen-binding fragment comprises 3 CDRs from the sequence of SEQ ID NO: 144. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from the sequence of SEQ ID NO: 163. In some embodiments, the antibody or antigen-binding fragment comprises 3 CDRs from the sequence of SEQ ID NO: 163. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from the sequence of SEQ ID NO: 164.
  • the antibody or antigen-binding fragment comprises 3 CDRs from the sequence of SEQ ID NO: 164. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from the sequence of SEQ ID NO: 143 and 1, 2 or 3 CDRs from the sequence of SEQ ID NO: 163. In some embodiments, the antibody or antigen-binding fragment comprises 3 CDRs from the sequence of SEQ ID NO: 143 and 3 CDRs from the sequence of SEQ ID NO: 163. In some embodiments, the antibody or antigen-binding fragment comprises 1 , 2 or 3 CDRs from the sequence of SEQ ID NO: 144 and comprises 1 , 2 or 3 CDRs from the sequence of SEQ ID NO: 164. In some
  • the antibody or antigen-binding fragment comprises 3 CDRs from the sequence of SEQ ID NO: 144 and comprises 3 CDRs from the sequence of SEQ ID NO: 164.
  • the assignment of amino acids to each domain is in accordance with the definitions of Rabat, Sequences of Proteins of Immunological Interest (National Institutes of Health, Bethesda, MD (1987 and 1991 )). In some embodiments, the assignment of amino acids to each domain is in accordance with Chothia & Lesk, j. Mol. Biol. 196:901-917
  • the assignment of amino acids to each domain is in accordance with MacCailum. In some embodiments, the assignment of amino acids to each domain is in accordance with AbM. In some
  • any of the antibodies or antigen-binding fragments disclosed herein comprises a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to any of SEQ ID NOs: 125-144.
  • the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 125.
  • the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 126. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 127.
  • the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 128.
  • the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 129 In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 130.
  • the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SI Q ID NO: 131. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 132.
  • the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 133. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 134.
  • the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 135. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 136.
  • the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 137. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 138.
  • the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 139. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 140.
  • the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 141. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 142.
  • the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 143 In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 144.
  • any of the antibodies or antigen-binding fragments disclosed herein comprises a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to any of SEQ ID NOs: 145-164.
  • the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 145.
  • the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 146. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 147.
  • the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 148. In some embodiments, the antibody or antigen -binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 149.
  • the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 150. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 151.
  • the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 152. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 153.
  • the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 154. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 155.
  • the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 156. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 157.
  • the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 158.
  • the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 159
  • the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 160.
  • the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 161. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 162.
  • the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 163. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 164.
  • the antibody or antigen-binding fragment comprises a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 143, and further comprises a variable light chain am o acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 163.
  • the antibody or antigen-binding fragment comprises a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 144, and further comprises a variable light chain ammo acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 164.
  • the term“antigen-binding fragment” refers to one or more fragmen ts of an antibody that retain the ability to specifically bind to an antigen.
  • binding fragments encompassed within the term “antigen-binding fragment” include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CHI domains: (ii) a F(ab')2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fab' fragment which is obtained by cleaving a disulfide bond of the hinge region of the F(ab')2; (iv) a Fd fragmen t consisting of the VH and CHI domains; (v) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (vi) a dAb fragment (Ward et af, (1989) Nature 341 :344-546), which consists of a V
  • VL and VH are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv)); see e.g., Bird et al. Science 242:423-426 (1988) and Huston et al. Proc. Natl. Acad. Sci. USA 85:5879-5883 (1988)).
  • antigen-binding molecules comprising a VN and/or a VL, In the case of a VH, the molecule may also comprise one or more of a CH 1, hinge, CH2 or CH3 region.
  • single chain antibodies are also intended to be encompassed within the term“antigen-binding fragment” of an antibody.
  • Other forms of single chain antibodies, such as diabodies are also encompassed
  • Diabodies are bivalent, bispecific antibodies in which VH and VL domains are expressed on a single polypeptide chain, but using a linker that is too short to allow for pairing between the two domains on the same chain, thereby forcing the domains to pair with complementary’ domains of another chain and creating two antigen binding sites (see e.g., Holliger et al. Proc. Natl. Acad. Sci. USA 90:6444-6448 (1993); Po!jak et al. Structure 2: 1121-1123 (1994)).
  • the term“antigen-binding fragment” also includes, e.g., single domain antibodies such as camelized single domain antibodies. See, e.g., Muyldermans et al. (2001) Trends Biochem Sci 26:230-235; Nutta!l et al. (2000) Curr Pharm Biotech 1 :253-263;
  • the disclosure provides single domain antibodies comprising two VH domains with modifications such that single domain antibodies are formed.
  • epitope refers to a site on an antigen (e.g., HTRAi) to which an immunoglobulin or antibody specifically binds.
  • An epitope can be formed both from contiguous ammo acids or noncontiguous amino acids juxtaposed by tertiary folding of a protein. An epitope formed from contiguous amino acids is typically retained on exposure to denaturing solvents, whereas an epitope formed by tertiary folding is typically lost on treatment with denaturing solvents.
  • An epitope typically includes at least 3,
  • epitope mapping Methods for determining what epitopes are bound by a given antibody (i.e., epitope mapping) are well knowm in the art and include, for example, immunob!otting and immunoprecipitation assays, wherein overlapping or contiguous peptides from HTRAI are tested for reactivity with the given anti -HTRAI antibody.
  • Methods of determining spatial conformation of epitopes include techniques in the art and those described herein, for example, x-ray crystallography and 2-dimensional nuclear magnetic resonance (see, e.g., Epitope Mapping Protocols in Methods in Molecular Biology, Vol. 66, G E. Morris, Ed (1996))
  • any of the antibodies or antigen binding-fragments disclosed herein binds to an epitope in human HTRAI.
  • the antibody or antigen binding fragment binds to an epitope in the amino acid sequence of SEQ ID NO: 121, or a functional fragment thereof.
  • any of the antibodies or antigen binding-fragments disclosed herein comprises any one of, or any combination of, the CDR amino acid sequences indicated in Figure 2. In some embodiments, any of the antibodies or antigen-binding fragments disclosed herein comprises any one of, or any combination of, the CDR ammo acid sequences of SEQ ID NGs: 1-120.
  • the antibody or antigen binding fragment comprises a set of CDRS as forth as any one of the following sets: SEQ ID NOs: 1-6, 7-12, 13-18, 19-24, 25-30, 31-36, 37-42, 43-48, 49-54, 55-60, 61-66, 67-72, 73-78, 79-84, 85-90, 91-96, 97-102, 103-108, 109-114, or 115-12.0.
  • the antibody or antigen binding fragment comprises the CDRs set forth as SEQ ID NOs: 1 -6.
  • the antibody or antigen binding fragment comprises the CDRs set forth as SEQ ID NOs: 7- 12.
  • the antibody or antigen binding fragment comprises the CDRs set forth as SEQ ID NOs: 19-24. In some embodiments, the antibody or antigen binding fragment comprises the CDRs set forth as SEQ ID NOs: 31-36. In some embodiments, the antibody or antigen binding fragment comprises the CDRs set forth as SEQ ID NOs: 37-42. In some embodiments, the antibody or antigen binding fragment comprises the CDRs set forth as SEQ ID NOs: 61-66. In some embodiments, the antibody or antigen binding fragment comprises the CDRs set forth as SEQ ID NOs: 73-78. In some embodiments, the antibody or antigen binding fragment comprises the CDRs set forth as SEQ ID NOs: 103- 108.
  • the antibody or antigen binding fragment comprises the CDRs set forth as SEQ ID NOs: 109-114. In some embodiments, the antibody or antigen binding fragment comprises the CDRs set forth as SEQ ID NOs: 115-120. In some embodiments, the antibody or antigen binding fragment comprises any one of or combination of any of the foregoing CDR amino acid sequences, but with 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 amino acid substitutions. In some embodiments, the substitutions are conservative substitutions.
  • the substitutions either increase the binding affinity of the antibody or antigen-binding fragment to the HTRA1 epitope, or the substitutions result in a reduction of binding affinity of no more than 5%, 10%, 20%, 30%, 40%, or 50% as compared to the antibody or antigen-binding fragment lacking the substitutions.
  • the following groups each contain ammo acids that are conservative substitutions for one another. These groups are exemplary' as other conservative substitutions are known to those of skill in the art.
  • any of the CDRs disciosd herein lacks one or two, but no more than three amino acids at the N-terminus and/or C-terminus of any of the CDR sequences disclosed herein.
  • any of the antibodies or antigen-binding fragments disclosed herein comprises any of the heavy chain CDRs disclosed herein.
  • any of the antibodies or antigen binding fragments disclosed herein comprises any of the combinations of heavy chain CDRi, CDR2, and CDR3 disclosed herein.
  • the antibody or antigen-binding fragment thereof comprises any of the following sets of three heavy chain CDRs: SEQ ID NOs: 1-3, 7-9, 13-15, 19-21, 25-27, 31-33, 37-39, 43-45, 49-51, 55-57, 61-63, 67-69, 73-75, 79-81, 85-87, 91-93, 97-99, 103-105, 109-11 1 or 115-1 17, or any of the foregoing CDR combinations comprising one or more (e.g. , 1 , 2, 3, 4, 5, 6, 7, 8, 9, or 10) conservative substitutions.
  • tire antibody or antigen-binding fragment thereof comprises the heavy chain CDRs of SEQ ID NOs: 109-111.
  • the antibody or antigen-binding fragment thereof comprises the heavy chain CDRs of SEQ ID NOs: 109-1 1 1.
  • the antibody or antigen binding fragment thereof comprises the heavy chain CDRs of SEQ ID N Os: 109-111 comprising one or more (e.g. , 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) conservative substitutions.
  • the antibody or antigen-binding fragment thereof comprises the heavy chain CDRs of SEQ ID NOs: 115-117.
  • the antibody or antigen- binding fragment thereof comprises the heavy chain CDRs of SEQ ID NOs: 115-117 comprising one or more (e.g. , 1 , 2, 3, 4, 5, 6, 7, 8, 9, or 10) conservative substitutions.
  • the antibody or antigen-binding fragment comprises a common light chain.
  • the antibody or antigen-binding fragment is a full-length antibody.
  • the antibody is a monoclonal antibody.
  • the antibody is a human antibody.
  • the antibody is a humanized antibody.
  • the antibody or antigen -binding fragment is a chimeric antibody. In some embodiments, the antibody or antigen-binding fragment is a humanized antibody or antigen-binding fragment. In some embodiments, the antibody or antigen-binding fragment is an antigen-binding fragment. In some embodiments, the antigen-binding fragment is selected from the group consisting of a Fab fragment, a F(ab')2 fragment, a Fab' fragment, a dAb, or an scFv. In some embodiments, any of the antibodies or antigen-binding fragments disclosed herein is capable of binding to and inhibiting one or more function of HTRA1.
  • the antibody or antigen-binding fragment binds to one or more of the following HTRA1 domains: die insulin-like growth factor binding domain, die kazal domain, the trypsin-like peptidase domain, and/or the PDZ domain.
  • any of tire antibodies or antigen-bind g fragments thereof disclosed herein bind to a human HTRA1 protein with a binding affinity (KD) of at least 50 nM, at least 25 nM, at least 10 nM, at least 7 nM, at least 5 nM, at least 3 nM, at least 2 nM, at least 1 nM, at least 500 pM, at least 100 pM, at least 50 pM or at least 10 pM.
  • KD binding affinity
  • the antibody or antigen-binding fragment is capable of binding to HTRA1 such that one or more HTRA1 functions are inhibited. In some embodiments, the antibody or antigen-binding fragment binds to HTRA1 such that HTRAl’s ability to cleave any one or more HTRA 1 substrate is reduced. In some embodiments, the HTRA 1 substrate is selected from the group consisting of: fibromodulin, elastin, clusterin, LTBP-1, EFEMP1, ADAM9, vitronectin, a2-macroglobulin, talin-1, fascin, fibulin 5, tan, RseA, and chloride intracellular channel protein.
  • the antibody or antigen-binding fragment binds to HTRA1 such that HTRAl’s ability to cleave a regulator of the complement cascade (e g., vitronectin, fibromodulin or clusterin) is reduced.
  • a regulator of the complement cascade e g., vitronectin, fibromodulin or clusterin
  • the antibody or antigen-binding fragment binds to HTRA1 such that HTRAl’s ability to cleave an HTRA1 substrate and/or regulator of the complement cascade is reduced by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the ability of the HTRA1 to cleave the HTRA1 substrate and/or regulator of the complem ent cascade in the absence of the antibody or antigen-binding fragment thereof.
  • the antibody or antigen -binding fragment is capable of inhibiting proteolytic activity of an HTRA1 protein.
  • the antibody or antigen-binding fragm nt is capable of inhibiting HTRA! proteolytic activity by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the proteolytic activity of a wildtype HTRA1 protein in the absence of the antibody or antigen-binding fragment.
  • the antibody or antigen-binding fragment is capable of inhibiting HTRA I proteolytic activity' in a cell by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the proteolytic activity of a wildtype HTRA1 protein m the same cell type in the absence of the antibody or antigen-binding fragment.
  • the antibody' or antigen-binding fragm nt is capable of inhibiting HTRA1 proteolytic activity in an eye by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the proteolytic activity of a wildtype HTRA1 protein in an eye in the absence of the antibody or antigen-binding fragment.
  • any of the antibodies or antigen-binding fragments disclosed herein is capable of inhibiting HTRA1 trimerization.
  • the antibody or antigen binding fragment is capable of inhibiting HTRA1 trimerization by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the trimerization of wildtype HTRA1 protein in the absence of the antibody or antigen-binding fragment.
  • any of the antibodies or antigen-binding fragments disclosed herein is capable of binding human HTRA1.
  • the antibody or antigen-binding fragment is capable of binding to a non-human species H ' TRAl .
  • the non-human species HTRA1 is mouse, rat, rabbit, cow, monkey (e.g , cynomolgus monkey), and/or ape (e.g. chimpanzee) HTRA 1 protein.
  • Antibodies that recognize the same or overlapping epitope as a known antibody or compete for binding with a known antibody can be identified using routine techniques. Such techniques include, for exampl e, an immunoassay, which shows the ability of one antibody to block the binding of another antibody to a target antigen, i.e., a competitive binding assay. Competitive binding is determined in an assay in which tire immunoglobulin under test inhibits specific binding of a reference antibody to a common antigen, such as H ' TRAl .
  • such an assay involves the use of purified antigen bound to a solid surface or cells bearing either of these, an unlabeled test immunoglobulin and a labeled reference immunoglobulin.
  • Competitive inhibition is measured by determining the amount of label bound to the solid surface or cells in the presence of the test immunoglobulin.
  • the test immunoglobulin is present in excess.
  • a competing antibody is present in excess, it will inhibit specific binding of a reference antibody to a common antigen by at least about 50-55%, 55-60%, 60-65%, 65-70%, 70-75% or more.
  • epitope mapping methods such as, x-ray analyses of crystals of antigen: antibody complexes which provides atomic resolution of the epitope and mass spectrometry combined with hydrogen/deuterium (H/D) exchange which studies the conformation and dynamics of antigen: antibody interactions.
  • H/D hydrogen/deuterium
  • Other methods monitor the binding of the antibody to antigen fragments or mutated variations of the antigen where loss of binding due to a modification of an amino acid residue within the antigen sequence is often considered an indication of an epitope component.
  • computational combinatorial methods for epitope mapping can also be used. These methods rely on the ability of the antibody of interest to affinity isolate specific short peptides from combinatorial phage display peptide libraries. The peptides are then regarded as leads for the definition of the epitope corresponding to the antibody used to screen the peptide library'.
  • computational algorithms have also been developed which have been shown to map conformational discontinuous epitopes.
  • methods for preparing an antibody described herein can include immunizing a subject (e.g , a non-human mammal) with an appropriate immunogen (e.g., an HTRA1 polypeptide having the amino acid sequence of SEQ ID NO: 121 or a fragment thereof). Suitable immunogens for generating any of the antibodies described herein are set forth herein.
  • a skilled artisan can immunize a suitable subject (e.g., a non- human mammal such as a rat, a mouse, a gerbil, a hamster, a dog, a cat, a pig, a goat, a horse, or a non-human primate) with human HTRA1.
  • a suitable subject e.g., a non- human mammal such as a rat, a mouse, a gerbil, a hamster, a dog, a cat, a pig, a goat, a horse, or a non-human primate
  • a suitable subject e.g., a non-human mammal
  • the immunogen can be administered to a subject (e.g., a non-human mammal) with an adjuvant.
  • adjuvants useful in producing an antibody in a subject include, but are not limited to, protein adjuvants; bacterial adjuvants.
  • BCG whole bacteria
  • MER methanol extractable residue
  • complete or incomplete Freund’s adjuvant viral adjuvants
  • chemical adjuvants e.g., aluminum hydroxide, and iodoacetate and cholesteryl hemisuccinate.
  • Other adjuvants that can be used in the methods for inducing an immune response include, e.g., cholera toxin and parapoxvims proteins. See also Bieg et al.
  • the methods include preparing a hybridoma cell line that secretes a monoclonal antibody that binds to the immunogen.
  • a suitable mammal such as a laboratory mouse is immunized with a HTRAl polypeptide as described above.
  • Antibody- producing cells e.g., B cells of the spleen
  • B cells of the spleen can be isolated two to four days after at least one booster immunization of the immunogen and then grown briefly in culture before fusion with cells of a suitable myeloma cell line.
  • the cells can be fused in the presence of a fusion promoter such as, e.g., vaccinia vinis or polyethylene glycol.
  • Tire hybrid cells obtained in die fusion are cloned, and cell clones secreting die desired antibodies are selected.
  • spleen cells of Balb/c mice immunized with a suitable immunogen can be fused with cells of the myeloma cell line PAI or the myeloma cell line Sp2/0-Ag 14.
  • the cells are expanded in suitable culture medium, which is supplemented with a selection medium, for example HAT medium, at regular intervals in order to prevent normal myeloma cells from overgrowing the desired hybridoma cells.
  • suitable culture medium which is supplemented with a selection medium, for example HAT medium, at regular intervals in order to prevent normal myeloma cells from overgrowing the desired hybridoma cells.
  • the obtained hybrid ceils are then screened for secretion of the desired antibodies, e.g., an antibody that binds to human HTRA1 as described herein.
  • a skilled artisan can identify an anti-HTRA 1 antibody from a non- immune biased library as described in, e.g., U.S. patent no. 6,300,064 (to Knappik et al.; Morphosys AG) and Schoonbroodt et al. (2005) Nucleic Acids Res 33(9):e81.
  • the methods described herein can involve, or be used in conjunction with, e.g., phage display technologies, bacterial display, yeast surface display, eukaryotic viral display, mammalian cell display, and cell-free (e.g., ribosomal display) antibody screening techniques (see, e.g., Etz et al. (2001) J Bactenol 183:6924-6935; Georgias (2000) Curr Opin Biotechnol 1 1 :450-454; Klemm et al. (2000) Microbiology 146:3025-3032;Kieke et al. (1997) Protein Eng 10: 1303-1310;Yeung et al.
  • phage display technologies e.g., phage display technologies, bacterial display, yeast surface display, eukaryotic viral display, mammalian cell display, and cell-free (e.g., ribosomal display) antibody screening techniques
  • Etz et al. 2001) J Bactenol 183:69
  • phage display methods functional antibody domains are displayed on the surface of phage particles which cany- the polynucleotide sequences encoding them.
  • phage can be utilized to display antigen-binding domains of antibodies, such as Fab, Fv, or disulfide-bond stabilized Fv antibody fragments, expressed from a repertoire or combinatorial antibody library (e.g., human or murine).
  • Phage used in these methods are typically filamentous phage such as fd and M13.
  • the antigen binding domains are expressed as a recombinantly fused protein to any of the phage coat proteins pill, pVIII, or pIX. See, e.g., Shi et al. (2010) JMB 397:385-396. Examples of phage display methods that can be used to make the
  • immunoglobulins, or fragments thereof, described herein include those disclosed in Brinkman et al. (1995) J Immunol Methods 182:41-50; Ames et al. (1995) J Immunol Methods 184: 177-186; Kettleborougli et al. (1994) Bur I Immunol 24:952-958; Persic et al. (1997) Gene 187:9-18; Burton et al. (1994) Advances m Immunology 57: 191-280; and PCT publication nos. WO 90/02809, WO 91/10737, WO 92/01047, WO 92/18619, WO 93/1 1236, WO 95/15982, and WO 95/20401.
  • the phage display antibody libraries can be generated using mRNA collected from B cells from the immunized mammals.
  • a splenic cell sample comprising B cells can be isolated from mice immunized with a HTRAl polypeptide as described above.
  • mRNA can be isolated from the cells and converted to cDNA using standard molecular biology techniques. See, e.g., Sambrook et al. (1989)‘"Molecular Cloning: A Laboratory Manual, 2nd Edition,” Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.; Harlow and Lane (1988), supra; Benny K. C. Lo (2004), supra; and Borrebaek (1995), supra.
  • the cDNA coding for the variable regions of the heavy chain and light chain polypeptides of immunoglobulins are used to construct the phage display library. Methods for generating such a library' are described in, e.g., Merz et al. (1995) J Neurosci Methods 62(l-2):213-9; Di Niro et al. (2005) Biochem J 388(Pt 3):889-894; and Engherg et al. (1995) Methods Mol Biol 51 :355-376.
  • a combination of selection and screening can be employed to identify an antibody of interest from, e.g., a population of hybridoma-derived antibodies or a phage display antibody library.
  • Suitable methods are known in the art and are described in, e.g., Hoogenboom (1997) Trends in Biotechnology 15:62-70; Brinkman et al. (1995), supra; Ames et ai. (1995), supra; Kettleborough et al. (1994), supra; Persic et ai. (1997), supra; and Burton et al. (1994), supra.
  • a plurality of phage mid vectors each encoding a fusion protein of a bacteriophage coat protein (e.g., pill, pVIII, or pIX of M13 phage) and a different antigen-combining region are produced using standard molecular biology techniques and then introduced into a population of bacteria (e.g., E. coli).
  • Expression of the bacteriophage in bacteria can, in some embodiments, require use of a helper phage. In some embodiments, no helper phage is required (see, e.g., Chasteen et ah, (2006) Nucleic Acids Res 34(21):el45).
  • Phage produced from tire bacteria are recovered and then contacted to, e.g., a target antigen bound to a solid support (immobilized). Phage may also be contacted to antigen in solution, and the complex is subsequently bound to a solid support.
  • a subpopulation of antibodies screened using the above methods can be characterized for their specificity and binding affinity for a particular antigen (e.g., human HTRA 1) using any immunological or biochemical based method known in the art.
  • immunoradiometric assays fluorescent immunoassays, and protein A immunoassays.
  • fluorescent immunoassays fluorescent immunoassays
  • protein A immunoassays Such assays are routine and well known in the art.
  • nucleic acids encoding the CDRs can be chemically synthesized as described in, e.g., Shiraishi et al. (2007) Nucleic Acids Symposium Series 51(1): 129-130 and U.S. Patent No. 6,995,259.
  • the region of the nucleic acid sequence encoding the CDRs can be replaced with the chemically synthesized nucleic acids using standard molecular biology techniques.
  • the 5’ and 3' ends of the chemically synthesized nucleic acids can be synthesized to comprise sticky end restriction enzyme sites for use in cloning tire nucleic acids into the nucleic acid encoding the variable region of the donor antibody.
  • the anti-HTRAl antibodies described herein comprise an altered heavy chain constant region that has reduced (or no) effector function relative to its corresponding unaltered constant region.
  • Effector functions involving the constant region of tire anti-HTRAl antibody may be modulated by altering properties of the constant or Fc region.
  • Altered effector functions include, for example, a modulation in one or more of the following activities: antibody-dependent cellular cytotoxicity (ADCC), complement- dependent cytotoxicity (CDC), apoptosis, binding to one or more Fc-receptors, and pro- inflammatory' responses.
  • Modulation refers to an increase, decrease, or elimination of an effector function activity exhibited by a subject antibody containing an altered constant region as compared to the activity of the unaltered fomi of the constant region.
  • modulation includes situations in which an activity is abolished or completely absent.
  • An altered constant region with altered FcR binding affinity and/or ADCC activity and/or altered CDC activity is a polypeptide which has either an enhanced or diminished FcR binding activity and/or ADCC activity and/or CDC activity compared to the unaltered form of the constant region.
  • An altered constant region which displays increased binding to an FcR binds at least one FcR with greater affinity than the unaltered polypeptide.
  • An altered constant region which displays decreased binding to an FcR binds at least one FcR with lower affinity than the unaltered form of the constant region.
  • Such variants which display- decreased binding to an FcR may possess little or no appreciable binding to an FcR, e.g., 0 to 50% (e.g , less than 50, 49, 48, 47, 46, 45, 44, 43, 42, 41 , 40, 39, 38, 37, 36, 35, 34, 33, 32,
  • an altered constant region that displays modulated ADCC and/or CDC activity- may exhibit either increased or reduced ADCC and/or CDC activity compared to the unaltered constant region.
  • the anti-HTRAl antibody comprises an altered constant region can exhibit approximately 0 to 50% (e.g., less than 50, 49, 48, 47, 46, 45, 44, 43, 42, 41, 40, 39, 38, 37, 36, 35, 34, 33, 32, 31, 30, 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1%) of the ADCC and/or CDC activity of the unaltered form of the constant region.
  • An anti-HTRAl antibody described herein comprising an altered constant region displaying reduced ADCC and/or CDC may exhibit reduced or no ADCC and/or CDC activity 7 .
  • an anti-HTRAl antibody or antigen-binding fragment described herein exhibits reduced or no effector function.
  • an anti-HTRA 1 antibody comprises a hybrid constant region, or a portion thereof, such as a G2/G4 hybrid constant region (see e.g., Burton et al . (1992) Adv Immun 51 : 1-18; Canfield et al. (1991) I Exp Med 173: 1483-1491 ; and Mueller et al . ( 1997) Mol Immunol 34(6):441 -452). See above.
  • an anti-HTRAl antibody or antigen-binding fragment may contain an altered constant region exhibiting enhanced or reduced complement dependent cytotoxicity (CDC).
  • Modulated CDC activity may he achieved by introducing one or more amino acid substitutions, insertions, or deletions in an Fc region of the antibody. See, e.g., IJ.S. patent no. 6,194,551. Alternatively or additionally, cysteine residue(s) may be introduced in the Fc region, thereby allowing interchain disulfide bond formation in this region.
  • the homodimeric antibody thus generated may have improved or reduced internalization capability and/or increased or decreased complement-mediated cell killing. See, e.g., Caron et al.
  • the antibodies or antigen-binding fragments thereof described herein can be produced using a variety of techniques known in the art of molecular biology and protein chemistry .
  • a nucleic acid encoding one or both of the heavy and light chain polypeptides of an antibody can be inserted into an expression vector that contains transcriptional and translational regulatory sequences, which include, e.g., promoter sequences, ribosomal binding sites, transcriptional start and stop sequences, translational start and stop sequences, transcription terminator signals, polyadenylation signals, and enhancer or activator sequences.
  • the regulatory sequences include a promoter and transcriptional start and stop sequences.
  • the expression vector can include more than one replication system such that it can be maintained in two different organisms, for example in mammalian or insect cells for expression and in a prokaryotic host for cloning and amplification.
  • Several possible vector systems are available for the expression of cloned heavy chain and light chain polypeptides from nucleic acids in mammalian cells.
  • One class of vectors relies upon the integration of the desired gene sequences into the host cell genome.
  • Cells which have stably integrated DNA can be selected by simultaneously introducing drug resistance genes such as E. coli gpt (Mulligan and Berg (1981) Proc Natl Acad Sei USA 78:2072) or Tn5 neo (Southern and Berg (1982) Mol Appl Genet 1 :327).
  • the selectable marker gene can be either linked to the DNA gene sequences to be expressed, or introduced into the same cell by co-transfection (Wigler et al. ( 1979) Cell 16:77).
  • a second class of vectors utilizes DNA elements which confer autonomously replicating capabilities to an extrachromosomal plasmid.
  • These vectors can be derived from animal viruses, such as bovine papillomavirus (Sa er et al. (1982) Proc Natl Acad Sci USA, 79:7147), cytomegalovirus, polyoma virus (Deans et al. (1984) Proc Natl Acad Sci USA 81 : 1292), or SV40 virus (Lusky and Botchan (1981) Nature 293:79).
  • the expression vectors can be introduced into cells in a manner suitable for subsequent expression of the nucleic acid.
  • the method of introduction is largely dictated by the targeted ceil type, discussed below.
  • Exemplary methods include CaPCri precipitation, liposome fusion, cationic liposomes, electroporation, viral infection, dextran-mediated transfection, polybrene-mediated transfection, protoplast fusion, and direct microinjection.
  • Appropriate host cells for the expression of antibodies or antigen-binding fragments thereof include yeast, bacteria, insect, plant, and mammalian cells. Of particular interest are bacteria such as E. coli, fungi such as Saccharomyces cerevisiae and Pichia pastons, insect cells such as SF9, mammalian cell lines (e.g., human cell lines), as well as primary cell lines.
  • an antibody or fragment thereof can be expressed in, and purified from, transgenic animals (e.g., transgenic mammals).
  • transgenic animals e.g., transgenic mammals
  • an antibody can be produced in transgenic non-human mammals (e.g., rodents) and isolated from milk as described in, e.g., Houdebine (2002) Curr Opin Biotechnol 13(6):625-629; van Kuik- Romeijn et al . (2000) Transgenic Res 9(2): 155-159; and Pollock et al. (1999) J Immunol Methods 231(1-2): 147-157.
  • the antibodies and fragments thereof can be produced from the cells by culturing a host cell transformed with the expression vector containing nucleic acid encoding the antibodies or fragments, under conditions, and for an amount of time, sufficient to allow expression of the proteins.
  • Such conditions for protein expression will vary with the choice of the expression vector and the host cell, and will be easily ascertained by one skilled in the art through routine experimentation.
  • antibodies expressed in E. coil can be refolded from inclusion bodies (see, e.g., Hou et a!. (1998) Cytokine 10:319-30).
  • an antibody or fragment thereof can be isolated or purified in a variety' of ways known to those skilled in the art depending on what other components are present in the sample.
  • Standard purification methods include electrophoretic, molecular, immunological, and chromatographic techniques, including ion exchange, hydrophobic, affinity, and reverse- phase HPLC chromatography.
  • an antibody can be purified using a standard anti-antibody column (e.g., a protein -A or protein-G column).
  • Ultrafiltration and diafiltration techniques, in conjunction with protein concentration are also useful. See, e.g.. Scopes (1994)“Protein Purification, 3rd edition, , Springer-Verlag, New York City, New York. The degree of purification necessary will vary depending on the desired use. In some instances, no purification of the expressed antibody or fragments thereof will be necessary.
  • Methods for determining the yield or purity of a purified antibody or fragment thereof include, e.g., Bradford assay, UV spectroscopy, Biuret protein assay, Lowry protein assay, amido black protein assay, high pressure liquid chromatography (HPLC), mass spectrometry (MS), and gel electrophoretic methods (e.g., using a protein stain such as Coomassie Blue or colloidal silver stain).
  • the antibodies or antigen -binding fragments thereof can be modified following their expression and purification. The modifications can be covalent or non- covalent modifications.
  • Such modifications can be introduced into the antibodies or fragments by, e.g , reacting targeted amino acid residues of the polypeptide with an organic derivatizing agent that is capable of reacting with selected side chains or terminal residues.
  • Suitable sites for modification can be chosen using any of a variety of criteria including, e.g., structural analysis or amino acid sequence analysis of the antibodies or fragments.
  • the antibodies or antigen-binding fragments thereof can be conjugated to a heterologous moiety.
  • Tire heterologous moiety can be, e.g., a heterologous polypeptide, a therapeutic agent (e.g., a drug), or a detectable label such as, but not limited to, a radioactive label, an enzymatic label, a fluorescent label, a heavy metal label, a luminescent label, or an affinity tag such as biotin or streptavidin.
  • a therapeutic agent e.g., a drug
  • a detectable label such as, but not limited to, a radioactive label, an enzymatic label, a fluorescent label, a heavy metal label, a luminescent label, or an affinity tag such as biotin or streptavidin.
  • Suitable heterologous polypeptides include, e.g., an antigenic tag (e.g., FLAG (DYKDDDDK (SEQ ID NO: 122)), polyhistidine (6-His; HHHHHH (SEQ ID NO: 123), hemagglutinin (HA: YPYDVPDYA (SEQ ID NO: 124)), ghitathione-S-transferase (GST), or maltose-binding protein (MBP)) for use in purifying the antibodies or fragments.
  • an antigenic tag e.g., FLAG (DYKDDDDK (SEQ ID NO: 122)
  • polyhistidine (6-His; HHHHHH (SEQ ID NO: 123), hemagglutinin (HA: YPYDVPDYA (SEQ ID NO: 124)
  • GST ghitathione-S-transferase
  • MBP maltose-binding protein
  • Heterologous polypeptides also include polypeptides (e.g., enzymes) that are useful as diagnostic or detectable markers, for example, luciferase, a fluorescent protein (e.g., green fluorescent protein (GFP)), or chloramphenicol acetyl transferase (CAT).
  • Suitable radioactive labels include, e.g., 32P, 33P, 14C, 1251, 1311, 35S, and 3H.
  • Suitable fluorescent labels include, without limitation, fluorescein, fluorescein isothiocyanate (FITC), green fluorescent protein (GFP), DyLightTM 488, phycoerythrin (PE), propidium iodide (PI), PerCP, PE-Alexa Fluor® 700, Cy5, allophycocyanin, and Cy7 Luminescent labels include, e.g., any of a variety of luminescent lanthanide (e.g., europium or terbium) chelates.
  • suitable europium chelates include the europium chelate of diethyienetiiaminepentaaceticacid (DTPA)or tetraazacyclododecane- i ,4,7, 10-tetraacetic acid (DOTA).
  • Enzymatic labels include, e.g., alkaline phosphatase, CAT, luciferase, and horseradish peroxidase.
  • Two proteins can be cross-linked using any of a number of known chemical cross linkers.
  • cross linkers are those which link two amino acid residues via a linkage that includes a“hindered’ disulfide bond.
  • a disulfide bond within the cross-linking unit is protected (by hindering groups on ei ther side of the disulfide bond) from reduction by the action, for example, of reduced glutathione or die enzyme disulfide reductase.
  • SMPT 4-succinimidyl oxycarbonyl-a-methyl- a(2-pyridyldithio) toluene
  • cross-linkers include, without limitation, reagents which link two am o groups (e.g., N-5-azido-2-nitrobenzoyloxysuccinimide), two sulfliydryi groups (e.g., 1,4-bis-maleimidobutane), an amino group and a sulfliydryi group (e.g., m- maleimido benzoyl-N-hydroxy succinimide ester), an amino group and a carboxyl group (e.g., 4-jp-azidosalicylamido] butylamine), and an amino group and a guanidinium group that is present in the side chain of arginine (e.g., p-azidophenyl glyoxal monohydrate).
  • reagents which link two am o groups e.g., N-5-azido-2-nitrobenzoyloxysuccinimide
  • a radioactive label can he directly conjugated to the amino acid backbone of the antibody.
  • the radioactive label can be included as
  • partofalargermolecule e .g . , 125 Iinmeta- [ 1251] iodophenyl-N -hy droxysuccinimide
  • Such methods involve incubating the proteins with the radioactive label under conditions (e.g., pH, salt concentration, and/or temperature) that facilitate binding of the radioactive label or chelate to the protein (see, e.g., U.S. Patent No. 6,001,329).
  • conditions e.g., pH, salt concentration, and/or temperature
  • fluorophores can be conjugated to free ammo groups (e.g., of lysines) or sulfhydryl groups (e.g., cysteines) of proteins using succinimidyl (NHS) ester or tetrafluorophenyl (TFP) ester moieties attached to the fluorophores.
  • free ammo groups e.g., of lysines
  • sulfhydryl groups e.g., cysteines
  • the fluorophores can be conjugated to a heterobifunctional cross-linker moiety such as suifo-SMCC Suitable conjugation methods involve incubating an antibody protein, or fragment thereof, with the fluorophore under conditions that facilitate binding of the fluorophore to the protein. See, e.g., Welch and Redvaniy (2003)‘Handbook of Radiopharmaceuticals: Radiochemistry and
  • the antibodies or fragments can be modified, e.g., with a moiety that improves the stabilization and/or retention of the antibodies in circulation, e.g., in blood, serum, or other tissues.
  • the antibody or fragment can be PEGylated as described in, e.g., Lee et al. (1999) Bioconjug Chem 10(6): 973-8; Kinstler et al. (2002) Advanced Drug Deliveries Reviews 54:477-485; and Roberts et al.
  • the stabilization moiety can improve the stability, or retention of, the antibody (or fragment) by at least about 1.5 (e.g., at least about 2, 5, 10,
  • the antibodies or antigen -binding fragments thereof described herein can be glycosylated.
  • an antibody or antigen-binding fragment thereof described herein can be subjected to enzymatic or chemical treatment, or produced from a ceil, such that the antibody or fragment has reduced or absent glycosylation.
  • Methods for producing antibodies with reduced glycosylation are known in the art and described in, e.g., U.S. patent no 6,933,368; Wright et al (1991) EMBO .1 10( 10) :2717-2723 ; and Co et al. (1993) Mol Immunol 30: 1361.
  • the disclosure also provides for one or more polynucleotides that encode any of the anti- HTRAl antibodies or antigen-binding fragments thereof.
  • the disclosure provides for one or more vectors encoding any one or more of the polynucleotides disclosed herein.
  • the one or more vectors further comprise one or more promoters that may facilitate the expression of the one or more polynucleotides.
  • the disclosure provides for a host cell capable of expressing any one or more of the polynucleotides disclosed herein.
  • the disclosure provides for vector comprising any of die
  • the disclosure provides for a method of administering any of the vectors disclosed herein to a subject in need thereof.
  • the vector is a viral vector.
  • the viral vector is a retrovirus, lentivirus, or baculovims vector.
  • the viral vector is an adenoviral vector.
  • the viral vector is an AAV vector.
  • a variety of rAAV vectors may be used to deliver the polynucleotides to the eye and to direct its expression. More than 30 naturally occurring serotypes of AAV from humans and non human primates are known. Many natural variants of the AAV capsid exist, and an rAAV vector of the disclosure may be designed based on an AAV with properties specifically suited for ocular cells.
  • Recombinant AAV vectors of the present disclosure may be generated from a variety of adeno-associated viruses.
  • ITRs from any AAV serotype are expected to have similar structures and functions with regard to replication, integration, excision and transcriptional mechanisms.
  • AAV serotypes include AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV 11 and AAV12.
  • the rAAV vector is generated from serotype AAV1, AAV2, AAV4, AAV5, or AAV8. These serotypes are known to target photoreceptor cells or the retinal pigment epithelium.
  • the rAAV vector is generated from serotype AAV2
  • the AAV serotypes include AAVrhS, AAVrhBR or AAVrhl O. It will also be understood that the rAAV vectors may be chimeras of two or more serotypes selected from serotypes AAV1 through AAV12. The tropism of the vector may be altered by packaging the recombinant genome of one serotype into capsids derived from another AAV serotype.
  • the ITRs of the rAAV vims may be based on the ITRs of any one of AAV1-12 and may be combined with an AAV capsid selected from any one of AAV1-12, AAV-DJ, AAV-DJ8, AAV-DJ9 or other modified serotypes.
  • any AAV capsid serotype may be used with the vectors of the disclosure.
  • AAV serotypes examples include AAV1, AAV2, AAV 3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV 10, AAVl !, AAV 12, AAV-DJ, AAV-DJ8, AAV-DJ9, AAVrhS, AAVrhSR or AAVrh lO.
  • the AAV capsid serotype is AAV2.
  • Desirable AAV fragments for assembly into vectors may include the cap proteins, including the vpl, vp2, vp3 and hypervariable regions, the rep proteins, including rep 78, rep 68, rep 52, and rep 40, and the sequences encoding these proteins. These fragments may be readily utilized in a variety of vector systems and host cells. Such fragments maybe used, alone, in combination with other AAV serotype sequences or fragments, or in combination with elements from other AAV or non-AAV viral sequences.
  • artificial AAV serotypes include, without limitation, AAV with a non-naturally occurring capsid protein.
  • Such an artificial capsid may be generated by any suitable technique using a selected AAV sequence (e.g , a fragment of a vpl capsid protein) in combination with heterologous sequences which may be obtained from a different selected AAV serotype, non-contiguous portions of the same AAV serotype, from a non-AAV viral source, or from a non-viral source.
  • An artificial AAV serotype may be, without limitation, a pseudotyped AAV, a chimeric AAV capsid, a recombinant AAV capsid, or a "humanized" AAV capsid
  • the AAV is AAV2/5.
  • the AAV is AAV2/8.
  • the sequences encoding each of the essential rep proteins may be supplied by different AAV sources (e.g., AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8).
  • the rep78/68 sequences may be from AA V2
  • the rep52/40 sequences may he from AAV8.
  • the vectors of the disclosure contain, at a minimum, sequences encoding a selected AAV serotype capsid, e.g., an AAV2 capsid or a fragment thereof. In another embodiment, the vectors of the disclosure contain, at a minimum, sequences encoding a selected AAV serotype rep protein, e.g., AAV2 rep protein, or a fragment thereof.
  • such vectors may contain both AAV cap and rep proteins.
  • the AAV rep and AAV cap sequences can both be of one serotype origin, e.g., all AAV2 origin.
  • the vectors may comprise rep sequences from an AAV serotype which differs from that which is providing the cap sequences.
  • the rep and cap sequences are expressed from separate sources (e.g., separate vectors, or a host cell and a vector). In some embodiments, these rep sequences are fused in frame to cap sequences of a different AAV serotype to form a chimeric AAV vector, such as AAV2/8 described in US Patent No.
  • AAV serotypes include AAV1 , AAV2, AAV3, AAV4, AAV 5, AAV6, AAV7, AAV8, AAV9, AAVIO, AAV11, AAV 12, AAV-DJ, AAV-DJ 8, AAV-DJ9, AAVrh8, AAVrh8R or AAVrhlO.
  • the cap is derived from AAV2.
  • any of the vectors disclosed herein includes a spacer, i.e., a DNA sequence interposed between the promoter and the rep gene ATG start site.
  • the spacer may be a random sequence of nucleotides, or alternatively, it may encode a gene product, such as a marker gene.
  • the spacer may contain genes which typically incorporate start/ stop and poly A sites.
  • the spacer may be a non-coding DNA sequence from a prokaryote or eukaryote, a repetitive non- coding sequence, a coding sequence without transcriptional controls or a coding sequence with transcriptional controls.
  • the spacer is a phage ladder sequences or a yeast ladder sequence. In some embodiments, the spacer is of a size sufficient to reduce expression of the rep78 and rep68 gene products, leaving the rep52, rep40 and cap gene products expressed at normal levels. In some embodiments, the length of the spacer may therefore range from about 10 bp to about 10.0 kbp, preferably in the range of about 100 bp to about 8.0 kbp. In some embodiments, the spacer is less than 2 kbp in length.
  • the capsid is modified to improve therapy.
  • the capsid may be modified using conventional molecular biology techniques.
  • the capsid is modified for minimized immunogenicity, better stability' and particle lifetime, efficient degradation, and/or accurate deli very of any of the polynucleotides disclosed herein encoding any of the antibodies or antigen-binding fragments disclosed herein.
  • the modification or mutation is an amino acid deletion, insertion, substitution, or any combination thereof in a capsid protein.
  • a modified polypeptide may comprise 1, 2, 3, 4, 5, up to 10, or more amino acid substitutions and/or deletions and/or insertions.
  • “deletion” may comprise the deletion of individual amino acids, deletion of small groups of amino acids such as 2, 3, 4 or 5 amino acids, or deletion of larger amino acid regions, such as the deletion of specific ammo acid domains or other features.
  • An“insertion” may comprise the insertion of individual amino acids, insertion of small groups of amino acids such as 2, 3, 4 or 5 amino acids, or insertion of larger a mo acid regions, such as the insertion of specific amino acid domains or other features.
  • A“substitution” comprises replacing a wild type amino acid with another (e.g., a non-wild type amino acid).
  • the another (e.g., non-wild type) or inserted amino acid is Ala (A), His (H), Lys (K), Phe (F), Met (M), Thr (T), Gin (Q), Asp (D), or Giu (E).
  • the another (e.g., non-wild type) or inserted amino acid is A.
  • the another (e.g., non-wild type) amino acid is Arg (R), Asn (N), Cys (C), Giy (G), He (I), Leu (L), Pro (P), Ser (S), Trp (W), Tyr (Y), or Val (V).
  • non-polar Norleucine, Met, Ala, Val, Leu, He
  • polar without charge Cys, Ser, Thr, Asn, Gin
  • acidic negatively charged
  • Asp, Glu Asp, Glu
  • basic positively charged
  • Lys Lys
  • Arg Lys
  • Arg residues that influence chain orientation
  • Gly, Pro residues that influence chain orientation
  • (6) aromatic Tip, Tyr, Phe, His.
  • Conventional amino acids include L or D stereochemistry.
  • the another (e.g., non-wild type) amino acid is a member of a different group (e.g., an aromatic amino acid is substituted for a non-polar amino acid).
  • Substantial modifications in the biological properties of the polypeptide are accomplished by selecting substitutions that differ significantly in their effect on maintaining (a) the structure of the polypeptide backbone in the area of the substitution, for example, as a b-sheet or helical conformation, (b) the charge or hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain .
  • Naturally occurring residues are divided into groups based on common side-chain properties: (1 ) Non-polar: Norleucine, Met, Ala, Val, Leu, Ile;(2) Polar without charge: Cys, Ser, Thr, Asn, Gln;(3) Acidic (negatively charged): Asp, G3u;(4) Basic (positively charged): Lys, Arg;(5) Residues that influence chain orientation: Gly, Pro; and(6) Aromatic: Trp, Tyr, Phe, His.
  • the another (e.g., non wild type) ammo acid is a member of a different group (e.g., a hydrophobic amino acid for a hydrophilic amino acid, a charged amino acid for a neutral amino acid, an acidic amino acid for a basic amino acid, etc.).
  • the another (e.g., non-wild type) amino acid is a member of the same group (e.g., another basic amino acid, another acidic amino acid, another neutral ammo acid, another charged amino acid, another hydrophilic amino acid, another hydrophobic amino acid, another polar amino acid, another aromatic amino acid or another aliphatic amino acid).
  • the another (e.g., non-wild type) amino acid is an unconventional amino acid.
  • Unconventional amino acids are non-naturally occurring amino acids.
  • Examples of an unconventional amino acid include, but are not limited to, aminoadipic acid, beta-alanine, beta-aminopropionic acid, aminobutyric acid, piperidinic acid, aminocaprioic acid, aminoheptanoic acid, aminoisobutyric acid, aminopimelic acid, citrulline, diaminobutyric acid, desmosine, diaminopimelic acid, diaminopropionic acid, N-ethylglycine, N-ethylaspargine, hyroxylysine, allo-hydroxylysine, hydroxyproline, isodesmosine, allo-isoleucine, N-methylglycine, sarcosine, N- methylisoieucine, N-methyl valine, non aline, norleucine, orithine, 4-
  • one or more amino acid substitutions are introduced into one or more of VP1, VP2 and VP3.
  • a modified capsid protein comprises 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14, or 15 conservative or non-conservative substitutions relative to the wild-type polypeptide.
  • the modified capsid polypeptide of the disclosure comprises modified sequences, wherein such modifications can include both conservative and non conservative substitutions, deletions, and/or additions, and typically include peptides that share at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 87%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% sequence identity to the corresponding wild-type capsid protein.
  • the recombinant AAV vector, rep sequences, cap sequences, and helper functions required for producing the rAA V of the disclosure may be delivered to the packaging host cell using any appropriate genetic element (vector).
  • a single nucleic acid encoding all three capsid proteins e.g., VP1, VP2 and VP3 is delivered into the packaging host cell in a single vector.
  • nucleic acids encoding the capsid proteins are delivered into the packaging host cell by two vectors; a first vector comprising a first nucleic acid encoding two capsid proteins (e.g., VP I and VP2) and a second vector comprising a second nucleic acid encoding a single capsid protein (e.g., VPS).
  • three vectors, each comprising a nucleic acid encoding a different capsid protein are delivered to the packaging host cell.
  • the selected genetic element may be delivered by any suitable method, including those described herein. The methods used to construct any embodiment of this disclosure are known to those with skill in nucleic acid manipulation and include genetic engineering, recombinant engineering, and synthetic techniques.
  • Two polynucleotide or polypeptide sequences are said to be “identical” if the sequence of nucleotides or ammo acids in the two sequences is the same when aligned for maximum correspondence as described below . Comparisons between two sequences are typically performed by comparing the sequences over a comparison window to identify and compare local regions of sequence similarity.
  • a “comparison window” as used herein refers to a segment of at least about 20 contiguous positions, usually 30 to about 75, or 40 to about 50, in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned.
  • Optimal alignment of sequences for comparison may be conducted using the MegAlign ® program in the Lasergene ® suite of bioinformatics software (DNASTAR ® , Inc , Madison, WI), using default parameters.
  • This program embodies several alignment schemes described in the following references: Dayhoff, M.Q., 1978, A model of evolutionary change in proteins - Matrices for detecting distant relationships. In Dayhoff, M.O. (ed.) Atlas of Protein Sequence and Structure, National Biomedical Research Foundation, Washington DC Vol. 5, Suppl. 3, pp. 345-358; Hein J., 1990, Unified Approach to Alignment and Phylogenes pp. 626-645 Methods in Enzymology vol.
  • the "percentage of sequence identity” is determined by comparing two optimally aligned sequences over a window of comparison of at least 20 positions, wherein the portion of the polynucleotide or polypeptide sequence in the comparison window may comprise additions or deletions (i.e., gaps) of 20 percent or less, usually 5 to 15 percent, or 10 to 12 percent, as compared to the reference sequences (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 bases 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 reference sequence (i.e., the window size) and multiplying the results by 100 to yield the percentage of sequence identity.
  • Suitable“moderately stringent conditions” include prewashing in a solution of 5 X SSC, 0.5% SDS, 1 .0 mM EDTA (pH 8.0); hybridizing at 50°C-65°C, 5 X SSC, overnight; followed by washing twice at 65°C for 20 minutes with each of 2X, 0.5X and 0.2X SSC containing 0.1 % SDS.
  • high stringent conditions or “high stringency conditions” are those that: (1) employ low ionic strength and high temperature for washing, for example 0.015 M sodium
  • a denaturing agent such as formamide, for example, 50% (v/v) formamide with 0.1 % bovine serum albumin/0.1 % Ficoil/0.1% polyvinylpyrrolidone/50 mM sodium phosphate buffer at pH 6.5 with 750 mM sodium chloride, 75 mM sodium citrate at 42°C; or (3) employ 50% formamide, 5 x SSC (0.75 M NaCl, 0.075 M sodium citrate), 50 mM sodium phosphate (pH 6.8), 0.1% sodium pyrophosphate, 5 x Denhardt’s solution, sonicated salmon sperm DNA (50 pg/ml), 0.1% SDS, and 10% dextran sulfate at 42°C, with washes at 42°C in 0.2 x SSC (sodium chloride/sodium citrate) and 50% formamide at 55°C;
  • formamide for example, 50% (v/v) formamide with 0.1 % bovine serum albumin/0.1 % Fico
  • nucleotide sequences that encode a polypeptide as described herein. Some of these polynucleotides bear minimal homology to the nucleotide sequence of any native gene. Nonetheless, polynucleotides that vary due to differences in codon usage are specifically contemplated by the present disclosure. Further, alleles of the genes comprising the polynucleotide sequences provided herein are within the scope of the present disclosure. Alleles are endogenous genes that are altered as a result of one or more mutations, such as deletions, additions and/or substitutions of nucleotides. The resulting mRNA and protein may, but need not, have an altered structure or function. Alleles may be identified using standard techniques (such as hybridization, amplification and/or database sequence comparison).
  • nucleic acids/polynucleotides of this disclosure can be obtained using chemical synthesis, recombinant methods, or PCR. Methods of chemical polynucleotide synthesis are well known in the art and need not be described in detail herein. One of skill in the art. can use the sequences provided herein and a commercial DNA synthesizer to produce a desired DNA sequence. In some embodiments, the disclosure provides for a polynucleotide that hybridizes under highly stringent conditions with any of the specific nucleotide sequences disclosed herein. One of ordinary ' skill in the art will readily understand that appropriate stringency conditions which promote DNA hybridization can be varied.
  • the hybridization at 6.0 x sodium chloride/sodium citrate (SSC) at about 45 °C, followed by a wash of 2.0 x SSC at 50 °C.
  • the salt concentration in the wash step can be selected from a low stringency of about 2.0 x SSC at 50 °C to a high stringency of about 0.2 x SSC at 50 °C.
  • the temperature in the wash step can be increased from low stringency conditions at room temperature, about 22 °C, to high stringency conditions at about 65 °C. Both temperature and salt may be varied, or temperature or salt concentration may he held constant while the other variable is changed.
  • the disclosure provides nucleic acids which hybridize under low stringency conditions of 6 x SSC at room temperature followed by a wash at 2 x SSC at room temperature.
  • Isolated nucleic acids which differ due to degeneracy in the genetic code are also within the scope of the disclosure.
  • a number of amino acids are designated by more than one triplet. Codons that specify the same amino acid, or synonyms (for example, CAU and CAC are synonyms for histidine) may result in‘silent” mutations which do not affect the amino acid sequence of the protein.
  • CAU and CAC are synonyms for histidine
  • these variations m one or more nucleotides (up to about 3-5% of the nucleotides) of the nucleic acids encoding a particular protein may exist among members of a given species due to natural allelic variation. Any and all such nucleotide variations and resulting amino acid polymorphisms are within the scope of this disclosure.
  • compositions comprising any of the antibodies or antigen-binding fragments disclosed herein, and a pharmaceutically acceptable earner.
  • the pharmaceutical compositions may be suitable for any mode of administration described herein; for example, by intravhreai or intravenous administration.
  • any of the antibodies or antigen-binding fragments disclosed herein for treating retinal diseases such as LCA, retinitis pigmentosa, and age-related macular degeneration require the localized delivery of the anti-HTRAI antibody or antigen binding fragment to the cells in the retina.
  • the cells that will be the treatment target in these diseases are either the photoreceptor cells in the retina or the cells of the RPE underlying the neurosensory retina.
  • the pharmaceutical compositions comprising any of the antibodies or antigen-binding fragment described herein and a pharmaceutically acceptable carrier are suitable for administration to a human subject.
  • Such carriers are well known in the art (see, e.g., Remington's Pharmaceutical Sciences, 15th Edition, pp. 1035-1038 and 1570-1580).
  • the pharmaceu tical compositions comprising any of the an tibodies or antigen-binding fragment described herein and a pharmaceutically acceptable carrier is suitable for ocular injection.
  • the pharmaceutical composition is suitable for intravitreal injection.
  • the pharmaceutical composition is suitable for subretinal delivery.
  • Such pharmaceutically acceptable carriers can be sterile liquids, such as water and oil, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, and the like. Saline solutions and aqueous dextrose, polyethylene glycol (PEG) and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions.
  • the pharmaceutical composition may further comprise additional ingredients, for example preservatives, buffers, tonicity agents, antioxidants and stabilizers, nonionic wetting or clarifying agents, viscosity-increasing agents, and the like.
  • the pharmaceutical compositions described herein can be packaged in single unit dosages or in multidosage forms. The compositions are generally formulated as sterile and substantially isotonic solution.
  • any of the antibodies or antigen-binding fragments disclosed herein is formulated into a pharmaceutical composition intended for subretinal or intravitreal injection.
  • a pharmaceutically and/or physiologically acceptable vehicle or carrier particularly one suitable for administration to the eye, e.g., by subretinal injection, such as buffered saline or other buffers, e.g., HEPES, to maintain pH at appropriate physiological levels, and, optionally, other medicinal agents, pharmaceutical agents, stabilizing agents, buffers, carriers, adjuvants, diluents, etc.
  • the carrier will typically be a liquid.
  • physiologically acceptable carriers include sterile, pyrogen- free water and sterile, pyrogen-free, phosphate buffered saline.
  • the carrier is an isotonic sodium chloride solution.
  • the carrier is balanced salt solution.
  • the carrier includes tween. If the antibody or antigen-binding fragment is to be stored long-term, it may be frozen in the presence of glycerol or Tween20.
  • the pharmaceutically acceptable carrier comprises a surfactant, such as perfluorooctane (Perfluoron liquid).
  • the pharmaceutical composition described above is administered to the subject by subretinal injection. I n other embodiments, the pharmaceutical composition is administered by intravitreal injection.
  • Other forms of administration that may be useful in the methods described herein include, but are not limited to, direct delivery to a desired organ (e.g., the eye), oral, inhalation, intranasal, intratracheal intravenous, intramuscular, subcutaneous, intradermal, and other parental routes of administration. Routes of administration may be combined, if desired.
  • any of tire antibodies or antigen-binding fraginents/pharmaceutical compositions disclosed herein are administered to a patient such that they target cells of any one or more layers or regions of the retina or macula.
  • the compositions disclosed herein target cells of any one or more layers of the retina, including the inner limiting membrane, the nerve fiber layer, the ganglion cell layer (GCL), the inner plexiform layer, the inner nuclear layer, the outer plexiform layer, the outer nuclear layer, the external limiting membrane, the layer of rods and cones, or the retinal pigment epi thelium (RPE)
  • the compositions disclosed herein target glial cells of the GCL, Muller cells, and/or retinal pigment epithelial cells.
  • the compositions disclosed herein target vascular endothelial cells.
  • the compositions disclosed herein targets cells of any one or more regions of the macula including, for example, the umbo, the foveolar, the fo veal avascular zone, the fovea, the parafovea, or the perifovea.
  • the route of administration does not specifi cally target neurons.
  • the route of administration is chosen such that it reduces the risk of retinal detachment in the patient (e.g., intravitreal rather than subretinal
  • intravitreal administration is chosen if the antibody or antigen-binding fragment/composition is to be administered to an elderly adult (e.g., at least 60 years of age).
  • any of the antibodies/antigen-binding fragments/compositions disclosed herein are administered to a subject intravitreally.
  • Procedures for intravitreal injection are known in the art (see, e.g., Peyman, G.A., et al.
  • a subject for intravitreal injection rnay be prepared for the procedure by pupillary dilation, sterilization of the eye, and administration of anesthetic. Any suitable mydriatic agent known in the art may be used for pupillary dilation. Adequate pupillary dilation may be confirmed before treatment. Sterilization may be achieved by applying a sterilizing eye treatment, e.g., an iodide -containing solution such as Povidone- Iodine (BETADINE ⁇ ).
  • a sterilizing eye treatment e.g., an iodide -containing solution such as Povidone- Iodine (BETADINE ⁇ ).
  • a similar solution may also be used to clean the eyelid, eyelashes, and any other nearby tissues ⁇ e.g., skin).
  • Any suitable anesthetic may be used, such as lidocaine or proparacaine, at any suitable concentration.
  • Anesthetic may be administered by any method known in the art, including without limitation topical drops, gels or jellies, and subconjuctival application of anesthetic.
  • a sterilized eyelid speculum may be used to clear the eyelashes from die area.
  • the site of the injection may be marked with a syringe.
  • the site of the injection may be chosen based on the lens of the patient.
  • the injection site may be 3-3.5 m from the limns in pseudophakic or aphakic patients, and 3.5-4 mm from the limbus in phakic patients.
  • the patient may look in a direction opposite the injection site.
  • the needle may be inserted perpendicular to the sclera and pointed to the center of the eye.
  • the needle may be inserted such that the tip ends m the vitreous, rather than the subretinal space. Any suitable volume known in the art for injection may be used.
  • the eye may be treated with a sterilizing agent such as an antiobiotic.
  • the eye may also be rinsed to remove excess sterilizing agent.
  • ophthalmoscopy may include electroretinography (ERG) (particularly the b-wave measurement), perimetry, topographical mapping of the layers of the retina and measurement of the thickness of its layers by means of confocal scanning laser ophthalmoscopy (cSLO) and optical coherence tomography (OCT), topographical mapping of cone density via adaptive optics (AO), functional eye exam, etc.
  • ERG electroretinography
  • OCT optical coherence tomography
  • the composition may be delivered in a volume of from about 0.1 pL to about 1 mL, including all numbers within the range, depending on the size of the area to be treated, the amount of antibody oar antigen -binding fragment used, the route of administration, and the desired effect of the method.
  • the volume is about 50 pL. in another embodiment, the volume is about 70 pL. In a preferred embodiment, the volume is about 100 pL. In another embodiment, the volume is about 125 pL. In another embodiment, the volume is about 150 pL. In another embodiment, the volume is about 175 pL. In yet another embodiment, the volume is about 200 pL. In another embodiment, the volume is about 250 pL. In another embodiment, the volume is about 300 pL. In another embodiment, the volume is about 450 pL. In another embodiment, the volume is about 500 pL. In another
  • the volume is about 600 pL. In another embodiment, the volume is about 750 pL. In another embodiment, the volume is about 850 pL, In another embodiment, the volume is about 1000 pL,
  • Methods of treatment/prophylaxis are various methods of preventing, treating, arresting progression of or ameliorating the ocular disorders and retinal changes associated therewith.
  • the methods include administering to a mammalian subject in need thereof, an effective amount of a composition comprising any of the anti-HTRAl antibodies or antigen-binding fragments disclosed herein.
  • a composition comprising any of the anti-HTRAl antibodies or antigen-binding fragments disclosed herein.
  • Any of the antibodies or antigen-binding fragments described herein, or any of the vectors disclosed herein encoding any of the antibodies or antigen-binding fragments disclosed herein, are useful in the methods described below.
  • any of the antibodies or antigen-binding fragments disclosed herein, or any of the vectors disclosed herein encoding any of the antibodies or antigen -binding fragments disclosed herein are for use in treating retinal diseases, such as LCA, retinitis pigmentosa, and age-related macular degeneration may require the localized delivery of the antibody or antigen-binding fragment to the cells in the retina.
  • the cells that will be the treatment target in these diseases are either tire photoreceptor cells in the retina or the cells of the RPE underlying the neurosensory retina.
  • deli v ering any of the antibodies or antigen-binding fragments disclosed herein to these cells requires injection into the subretinal space between the retina and the RPE.
  • any of the antibodies or antigen-binding fragments disclosed herein are administered intravitreally or intravenously.
  • the disclosure provides a method of treating a subject having age-related macular degeneration (AMD), comprising the step of administering to the subject any of the antibodies or antigen-binding fragments of the disclosure.
  • AMD age-related macular degeneration
  • the AMD is any one of Early AMD; Intermediate AMD; Advanced non-neovascular (“Dr”) AMD; or Advanced neovascuiar (“Wet”) AMD.
  • the disclosure provides for methods of treating a subject with Wet AMD.
  • the disclosure provides for methods of treating a subject with Dry AMD.
  • the disclosure provides for methods of treating a subject with polyploidal choroidal vasculopathy (PCV).
  • the subject has geographic atrophy.
  • the pharmaceutical compositions of the disclosure comprise a pharmaceutically acceptable carrier.
  • the pharmaceutical compositions of the disclosure comprise PBS.
  • the pharmaceutical compositions of the disclosure comprise piuronic.
  • the pharmaceutical compositions of the disclosure comprise PBS.
  • compositions of the disclosure comprise PBS, NaCl and piuronic.
  • any of the antibodies or antigen-binding fragments disclosed herein is capable of inhibiting proteolytic activity of an HTRA1 protein in a subject in need thereof.
  • the antibody or antigen-binding fragment thereof is capable of inhibiting HTRA ! proteolytic activity by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the proteolytic activity of a wildtype HTRAl protein m the absence of the antibody or antigen-binding fragment thereof.
  • the antibody or antigen-binding fragment thereof is capable of inhibiting HTRA 1 proteolytic activity in a cell by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the proteolytic activity of a wildtype HTRAl protein in the same cell type in the absence of the antibody or antigen-binding fragment thereof.
  • the antibody or antigen-binding fragment thereof is capable of inhibiting HTRA 1 proteolytic activity in an eye by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the proteolytic activity of a wildtype HTRAl protein in an eye in the absence of the antibody or antigen-binding fragment thereof.
  • the antibody or antigen-binding fragment thereof is capable of inhibiting HTRAl’s ability to cleave any one or more HTRAl substrate is reduced in a subject.
  • the HTRAl substrate is selected from the group consisting of: fibromodulin, clusterin, ADAM9, eiastin, vitronectin, a2 -macroglobulin, taiin-1, fasein, LTBP-1, EFEMP1, fibulin 5, tau, RseA, and chloride intracellular channel protein.
  • the antibody or antigen-binding fragment thereof is capable of inhibiting HTRA l’s ability to cleave a regulator of the complement cascade (e.g., vitronectin, fibromodulin or clusterin).
  • tire antibody or antigen-binding fragment thereof is capable of inhibiting HTRAl such that HTRAl’s ability to cleave an HTRAl substrate and/or regulator of the complement cascade is reduced by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the ability of the HTRAl to cleave the HTRAl substrate and/or regulator of the complement cascade in the absence of the anti body or antigen-binding fragment thereof.
  • the antibody or antigen -binding fragment thereof is capable of inhibiting HTRAl such that HTRAl’s ability to trimerize is reduced by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the ability of the HTRA1 to trimerize in the absence of the antibody or antigen-binding fragment thereof.
  • any of tire antibodies or antigen binding fragments disclosed herein is administered to celi(s) or tissue(s) in a test subject.
  • the edits) or tissue(s) in the test subject express a higher level of HTRA1 than expressed in the same cell type or tissue type in a reference control subject or population of reference control subjects.
  • the reference control subject is of the same age and/or sex as the test subject. In some embodiments, the reference control subject is a healthy subject, e.g., the subject does not have a disease or disorder of the eye. In some embodiments, the reference control subject does not have a disease or disorder of the eye associated with activation of the complement cascade. In some embodiments, the reference control subject does not have macular degeneration. In some embodiments, the eye or a specific cell type of the eye (e.g, ceils in the foveal region) in the test subject express at least 300%, 250%, 200%, 150%,
  • the eye or a specific cell type of the eye (e.g., cells in the foveal region) in the test subject express an HTRA1 gene having any of the mutations disclosed herein.
  • the eye or a specific cell type of the eye (e.g., cells in the foveal region) in the reference control subject do not express a H ' TRA 1 gene having any of the HTRAl mutations disclosed herein.
  • administration of any of the antibodies or antigen-binding fragments disclosed herein in the ceil(s) or tissue(s) of the test subject results in an decrease in levels of HTRAl protein or functional HTRAl protein.
  • administration of any of the antibodies or antigen -binding fragments disclosed herein in the cell(s) or tissue(s) of the test subject is capable of decreasing levels of HTRAl protein or functional HTRAl protein such that the decreased levels are within 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 10%, 5%, or 1% of, or are the same as, the levels of HTRAl protein or functional HTRA l protein expressed by the same cell type or tissue type in the reference control subject or population of reference control subjects.
  • administering results in a decrease in levels of HTRAl protein or functional HTRAl protein, but the decreased levels of HTRAl protein or functional HTRAl protein are not below the levels of HTRAl protein or functional HTRAl protein expressed by the same cell type or tissue type in the reference control subject or population of reference control subjects.
  • administration of any of the antibodies or antigen -binding fragments disclosed herein in the cell(s) or tissue(s) of the test subject is capable of decreasing levels of H ' TRAl protein or functional HTRA1 protein, but the decreased levels of HTRA l protein or functional HTRA1 protein are below the levels ofHTRAl protein or functional HTRAl protein by no more than 1%, 5%, 10%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or 100% of the levels expressed by the same cell type or tissue type in the reference control subject or population of reference control subjects.
  • any of the treatment and/or prophylactic methods disclosed herein are applied to a subject.
  • the subject is a mammal.
  • the subject is a human.
  • the human is an adult.
  • the human is an elderly adult.
  • the human is at least 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, or 95 years of age.
  • the human is at least 60 or 65 years of age.
  • any of the treatment and/or prophylactic methods disclosed herein is for use in treatment of a patient having one or more mutations that causes macular degeneration (AMD) or that increases the likelihood that a patient develops AMD.
  • the one or more mutations are in the patient’s HTRAl gene.
  • any of the treatment and/or prophylactic methods di sclosed herein is for use in treatment of a subject having one or more mutations in the patient’s HTRAl gene.
  • “mutations” encompasses polymorphisms that are associated with increased HTRAl expression.
  • the mutation is associated with AMD or PC V.
  • the one or more mutations result in overexpression of the HTRAl gene.
  • HTRAl is expressed at a level at least 25%, 50%, 75%, 100%, 150%, 200%, 250%, 300%, 350%, 400%, 450%, or 500% greater in the subject having the disease or disorder as compared to the level in a control subject not having the disease or disorder.
  • control subject is a subject of the same sex and/or of similar age as the subject having the disease or disorder.
  • the one or more mutations are not in the coding sequence for the HTRAl gene.
  • the one or more mutations are in 10q26 in a human patient.
  • the one or more mutations correspond to any one or more of the following human polymorphisms: rs61871744;
  • rs2284665 rs60401382; rsl 1200643; rs58077526; rs932275 and/or rs2142308.
  • the retinal diseases described above are associated with various retinal changes. These may include a loss of photoreceptor structure or function; thinning or thickening of the ou ter nuclear layer (ONL); thinning or thickening of the outer plexiform layer (OPL);
  • a method of preventing, arresting progression of or ameliorating any of the retinal changes associated with these retinal diseases is provided. As a result, the subject's vision is improved, or vision loss is arrested and/or ameliorated.
  • Vision loss associated with an ocular disorder refers to any decrease in peripheral vision, central (reading) vision, night vision, day vision, loss of color perception, loss of contrast sensitivity, or reduction in visual acuity.
  • a method of targeting one or more type(s) of ocular cells for gene augmentation therapy in a subject in need thereof is provided.
  • a method of targeting one or more type of ocular cells for gene suppression therapy in a subject in need thereof is provided.
  • a method of targeting one or more type of ocular cells for gene knockdown/augmentation therapy in a subject in need thereof is provided.
  • a method of targeting one or more type of ocular cells for gene correction therapy in a subject in need thereof is provided.
  • a method of targeting one or more type of ocular cells for neurotropic factor gene therapy in a subject in need thereof is provided.
  • the targeted ceil may be an ocular cell.
  • the targeted cell is a glial cell. In one embodiment, the targeted cell is an RPE cell. In another embodiment, the targeted cell is a photoreceptor. In another embodiment, the photoreceptor is a cone cell. In another embodiment, the targeted cell is a Muller cell. In another embodiment, tire targeted cell is a bipolar cell. In yet another embodiment, the targeted cell is a horizontal cell. In another embodiment, the targeted cell is an amacrme cell. In still another embodiment, the targeted cell is a ganglion cell. In still another embodiment, the gene may be expressed and delivered to an intracellular organelle, such as a
  • any of the methods disclosed herein increase photoreceptor function.
  • photoreceptor function loss means a decrease in photoreceptor function as compared to a normal, non-diseased eye or the same eye at an earlier time point.
  • increase photoreceptor function means to improve the function of the
  • photoreceptors or increase the number or percentage of functional photoreceptors as compared to a diseased eye (having the same ocular disease), the same eye at an earlier time point, a non-treated portion of the same eye, or the contralateral eye of the same patient.
  • Photoreceptor function may be assessed using the functional studies described above and in the examples below, e.g., ERG or perimetry, winch are conventional in the art.
  • the treatment may be used to pre vent the occurrence of retinal damage or to rescue eyes having mild or advanced disease.
  • the term "rescue" means to prevent progression of the disease to total blindness, prevent spread of damage to uninjured ocular cells, improve damage in injured ocular cells, or to provide enhanced vision.
  • the composition is administered before the disease becomes symptomatic or prior to photoreceptor loss.
  • symptomatic is meant onset of any of the various retinal changes described above or vision loss.
  • the composition is administered after disease becomes symptomatic.
  • the composition is administered after initiation of photoreceptor loss.
  • the composition is administered after outer nuclear layer (ONE) degeneration begins.
  • ONE outer nuclear layer
  • the composition is administered after initiation of photoreceptor loss. In yet another embodiment, the composition is administered when less than 90% of the photoreceptors are functioning or remaining, as compared to a non-diseased eye. In another embodiment, the composition is administered when less than 80% of the photoreceptors are functioning or remaining. In another embodiment, the composition is administered when less than 70% of the photoreceptors are functioning or remaining. In another embodiment, the composition is administered when less than 60% of the photoreceptors are functioning or remaining. In another embodiment, the composition is administered when less than 50% of the photoreceptors are functioning or remaining. In another embodiment, the composition is administered when less than 40% of the photoreceptors are functioning or remaining. In another embodiment, the composition is administered when less than 30% of the
  • composition is administered when less than 20% of the photoreceptors are functioning or remaining.
  • tire composition is administered when less than 10% of the
  • the composition is administered only to one or more regions of the eye. in another embodiment, the composition is administered to the entire eye.
  • the method includes performing functional and imaging studies to determine the efficacy of the treatment. These studies include ERG and in vivo retinal imaging, as described in the examples below. In addition visual field studies, perimetry and microperimetry, pupillometry, mobility testing, visual acuity, contrast sensitivity, color vision testing may be performed.
  • any of the above described methods is perfonned in combination with another, or secondary, therapy.
  • the therapy may be any now known, or as yet unknown, therapy which helps prevent, arrest or ameliorate any of the described retinal changes and/or vision loss.
  • the secondary therapy is encapsulated cell therapy (such as that delivering Ciliary Neurotrophic Factor (CNTF)). See, Sieving, P.A. et al, 2006. Proc Natl Acad Sci USA, 103 (10): 3896-3901 , which is hereby incorporated by reference.
  • CNTF Ciliary Neurotrophic Factor
  • the secondary therapy is a neurotrophic factor therapy (such as pigment epithelium-derived factor, PEDF; ciliary' neurotrophic factor 3; rod-derived cone viability factor (RdCVF) or glial -derived neurotrophic factor).
  • the secondary therapy is anti-apoptosis therapy (such as that deli vering X-iinked inhibitor of apoptosis,
  • the secondary therapy is rod derived cone viability factor 2.
  • the secondary therapy can be administered before, concurrent with, or after administration of the antibodies/antigen-binding fragments described above.
  • any of tire antibodies, antigen-binding fragments or compositions disclosed herein is administered to a subject in combination with another therapeutic agent or therapeutic procedure.
  • the additional therapeutic agent is an anti- VEGF therapeutic agent (e.g., such as an anti-VEGF antibody or fragment thereof such as ranibizumab, bevacizumab or afhbercept), a vitamin or mineral (e.g ., vitamin C, vitamin E, lutein, zeaxanthin, zinc or copper), omega-3 fatty acids, and/or VisudyneTM.
  • the other therapeutic procedure is a diet having reduced omega-6 fatty acids, laser surgery, laser photocoagulation, submacular surgery, retinal translocation, and/or photodynamic therapy.
  • any of the antibodies or antigen-binding fragments thereof disclosed herein is assembled into a pharmaceutical or diagnostic or research kit to facilitate their use in therapeutic, diagnostic or research applications.
  • a kit may include one or more containers housing any of the antibodies or antigen-binding fragments thereof disclosed herein and instructions for use.
  • the kit may be designed to facilitate use of the methods described herein by researchers and can take many forms.
  • Each of the compositions of the kit may he provided in liquid fonn (e.g., in solution), or in solid form, (e.g., a dry' powder).
  • some of the compositions may be constitutable or otherwise processable (e.g., to an active form), for example, by the addition of a suitable solvent or other species (for example, water or a cell culture medium), which may or may not be provided with the kit.
  • a suitable solvent or other species for example, water or a cell culture medium
  • “instructions” can define a component of instruction and/or promotion, and typically involve written instructions on or associated with packaging of the disclosure.
  • Instructions also can include any oral or electronic instructions provided in any manner such that a user will clearly recognize that the instructions are to be associated with the kit, for example, audiovisual (e.g., videotape, DVD, etc.), Internet, and/or web-based communications, etc.
  • audiovisual e.g., videotape, DVD, etc.
  • the written instructions may be in a form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which instructions can also reflects approval by the agency of manufacture, use or sale for animal administration.
  • Bead-based panning was employed with biotinylated HTRA captured on streptavidin beads.
  • Four rounds of phage panning was performed with varying stringencies A total of 4 conditions (varying antigen concentration and washes) were tested.
  • the different antigens tested were 0.5 mg of biotinylated HTRA1; biotinylated L2 peptide 201 (19-mer); and biotinylated L3 peptide 202 (19-mer).
  • the panning rounds were performed as outlined below:
  • Phage ELISA on rescued panning outputs showed increased enrichment. A total of 14 plates from rounds 3 and rounds 4 outputs were screened for HTRA 1 binding. Binding was determined by detecting scFv in PPE expression with anti-v5 mAb. Three-hundred and eighty-seven clones were initially identified as ELISA binders and re-arrayed for scFv sequencing. Ninety-one unique sequences and ELISA re-confirmed clones w'ere identified. PPEs of these clones were tested for competition binding with two different control anti- HTRA 1 IgGs (BM1 and BM2) in ELISA format to determine epitope binders.
  • BM1 and BM2 control anti- HTRA 1 IgGs
  • Plate 1 was treated w ith HRP anti-human Fc (detects BM IgG), and plate 2 was treated with anti-v5 mAb and HRP anti-mouse IgG (detects scFV) If scFv binds the same epitope as BM IgG, ELISA signal of scFv will be lower than BM IgG. If scFv binds different epitopes than BM IgG, ELISA signal of scFv is not affected by BM IgG. Forty-five clones appear to bind independently of BMl and BM2.
  • Binding was determined by detecting scFv in PPE expression with anti ⁇ v5 mAb. Forty-two clones were initially identified as ELISA binders and re-arrayed for scFv sequencing. Fifteen sequences were unique and ELISA re-confirmed clones were identified. PPEs of these clones were tested for HTRA1 binding via ELISA but none of these clones bound. A total of 12 plates from rounds 3 and 4 outputs were screened for Pep202 binding. Binding was determined by detecting scFv in PPE expression with anti-v5 mAb.
  • Binders were tested in epitope binning experiments. Briefly, binning experiments were performed on Octet HTX at 25°C In tandem, the binning assay format was set up.
  • Biotinylated antigen bt-HTRAl (10 nM) was loaded onto Streptavidin sensors for 240 seconds. Loaded sensors were dipped into saturating antibody (20 ug/mL) for 900 seconds followed by competing antibody (5 ug/mL for 300 seconds).
  • Epitope binning experiments were completed by evaluating the degree of binding competition in a full pairwise comparison.
  • the first antibody was bound to HTRA1 at a saturating concentration.
  • the Octet sensor tips were then exposed to a solution of the potential competitor binder and the kinetics and level of binding compared to assess whether the two antibodies bound to a similar epitope. Based on these data six bins were defined along with an additional group where the epitope bin was uncertain. Binders were tested in an AlphaLISA assay format, using AlphaScreen signal inhibition. Briefly, an N-terminal Flag RseA-C-termmal His substrate associates with a donor bead (anti-Flag) and an acceptor bead (Nickel chelator) .
  • HTRA1 is capable of cleaving RseA and reducing the output signal.
  • the optimized assay conditions were as follows.
  • the buffer used included 150m M NaHrPOr pH 8.3, 380mM NaCi, 0.05% Triton X-100, 0.3% BSA
  • Five uL of diluted HTR.41 (400 nM final) and 5 ul of buffer/antibody (400 nM final) were added and incubated for 30 minutes at room temperature.
  • Five ul of diluted RseA (25 nM final) was added and then incubated for 1 or 4 hours at room temperature.
  • Example 1 for treating patients with AMD Patients with AMD will be treated with any of the antibodies or antigen-binding fragments, or a control.
  • the antibodies or antigen-binding fragments will be administered by intravitreal, subretinal or intravenous injection. Patients will be monitored for improvements in AMD symptoms.
  • Recombinant HTRA1 was generated containing a Strepil affinity tag and with a deletion of the HTRA1 N-terminal Mac domain (StrepIl-AMac-HTRAl).
  • Antibody specificity for HTRA! w'as confirmed using western blots.
  • Antibodies SR8047-38, SR8047-40, SR8047-17, SR8047-44, SR8047-16, SR8047-43, SR8047-31, SR8047-06, SR8047-23, and SR8047-42 were ail determined to bind to HTRA1.
  • SR8Q47-38 and SR8047-40 also bound to HTRA4. None of the antibodies showed any detectable binding to HTR A2 or HTRA3.
  • HTRA1 -mediated proteolysis of eiastin was measured in the presence and absence of anti-HTRAl antibodies in order to determine whether these antibodies inhibit HTRA1 serine protease activity.
  • HTRA1 -mediated proteolysis of eiastin was measured utilizing the Sensolyte ® Green Elastase Kit (Anaspec, Inc.). Reaction solutions were prepared and earned out in opaque 96-well pates. Each solution w'as prepared by mixing StrepII-AMac-HTRAl (200 iiM) with of 1.2 mM anti-HTRAl antibody in assay buffer (pH 8.0), followed by subsequent incubation at 37°C for 45 minutes.
  • HTRA1 -mediated proteolysis of eiastin generated an increase in the fluorescence of the 5-FAM fluorophore with an intensity proportional to the proteolytic activity of SixepIT-AMac -HTRAl
  • Fluorescence intensity' was measured every 60 seconds and plotted as a function of time ( Figure 3 A, Figure 3B). Measurements were performed at least in duplicate for each sample.
  • Anti-HTRAl antibodies SR8047-44 and SR8047-43 generated a 70% and 50% (respectively) reduction in HTRA1 -mediated proteolysis of elastin relative to the fully active positive controls (Figure 3C), suggesting that these particular antibodies are inhibitors of HTRA1 proteolytic activity.
  • the HTRA1 -mediated proteolysis of Fibulin 5 was measured in the presence and absence of anti- HTRA1 antibodies in order to test whether such antibodies would exert an inhibitory effect.
  • Reaction solutions containing StrepTI-AMac-HTRAl (100 nM) and an anti-HTRAl antibody (600 nM) were prepared in a buffer containing 25 niM HEPES pH 7.5, and 75 niM NaCl, and allowed to incubate at 37°C for 45 minutes at 300 rpm.
  • the proteolysis reaction was initiated upon addition of Fibulin 5 (1 mM) at 37°C and 300 rpm.
  • the antibodies were further tested for their capacity' to inhibit HTRA1 -mediated proteolysis of Tau using substantially the same SDS-PAGE method as described in the Fibulin 5 assay.
  • Reaction solutions containing StreplT-AMac-HTRAl (100 nM) and an anti-HTRAl antibody (600 nM) were prepared in a buffer containing 50 niM Tris pH 8 0, and allowed to incubate at 37°C for 45 minutes. The reaction was initiated upon addition of Tau (800 nM) at 37°C and 300 rpm. Reaction time-points were taken at 0, 7, and 22 hours by quenching a reaction sample with SDS-PAGE gel loading buffer and DTT (1 M).
  • the antibodies were further screened for their ability to inhibit HTRA 1 -mediated proteolysis of RseA.
  • the SDS-PAGE based screen described in the Fibulin 5 and Tau digest assay s was utilized, and reaction solutions containing StrepII-AMac-HTRAi (100 nM) and an anti-HTRAl antibody (600 nM) were prepared in a buffer containing 150 mM NaTfrPCh (Sodium Phosphate) pH 8.0, and 380 mM Nad. These reaction solutions were allowed to incubate at 37°C for 45 minutes. Following incubation, the reactions were initiated upon addition of RseA (1 mM) at 37°C and 300 rpm.
  • the antibodies were further screened for their ability to inhibit HTRA 1 -mediated proteolysis of b-casein using a similar SDS-PAGE screen as utilized above.
  • Reaction solutions containing Strepil-AMae- HTRA1 (100 nM) and an anti-HTRAl antibody (600 nM) were prepared in a buffer containing 50 mM Tris pH 8.0, and 150 mM NaCl. These reaction solutions were allowed to incubate at 37°C for 15 minutes. Following incubation, the reactions were initiated upon addition of b-casein (2 mM) at 37°C and 300 rpm. Reaction time-points were taken at 0, 5, and 30 minutes by quenching a reaction sample with SDS-PAGE gel loading buffer and DTT (1 M).
  • YTFTRQYMH SEQ ID NO: 26— SR8047-16 HCDR2 Amino Acid Sequence GWINPN SGNTGY A
  • FTFSNSDMN SEQ ID NO: 86— SR8047-36 HCDR2 Amino Acid Sequence
  • SEQ ID NO: 121 Human HTRA 1 Amino Acid Sequence- GenBank Accession No.
  • SEQ ID NO: 122 FLAG DYKDDDDK

Abstract

The present disclosure provides compositions and methods for treating, preventing, or inhibiting diseases of the eye. In one aspect, the disclosure provides anti-HTRA1 antibodies or antigen-binding fragments and methods of using the same.

Description

ANTIBODY-BASED THERAPEUTICS FOR TARGETING HTRA1 AND METHODS
OF USE
CROSS-REFERENCE TO RELATED APPLICATIONS
This application claims the benefit of priority from U.S. Provisional Application No.
62/768,557, fried November 16, 2018. The specification of the foregoing application is incorporated herein by reference in its entirety.
BACKGROUND OF THE DISCLOSURE
Age-related macular degeneration (AMD) is a medical condition and is the leading cause of legal blindness in Western societies. AMD typically affects older adults and results in a loss of central vision due to degenerative and neovascu!ar changes to the macula, a pigmented region at the center of the retina which is responsible for visual acuity. There are four major AMD subtypes: Early AMD; Intermediate AMD; Advanced non-neovaseular (“Dry”) AMD; and Advanced neovascular (“Wet”) AMD. Typically, AMD is identified by the focal hyperpigmentation of the retinal pigment epithelium (RPE) and accumulation of drusen deposits. The size and number of drusen deposits typically correlates with AMD se verity.
AMD occurs in up to 8% of individuals over the age of 60, and the prevalence of AMD continues to increase with age. The U.S. is anticipated to have nearly 22 million cases of AMD by the year 2050, while global cases of AMD are expected to be nearly 288 million by the year 2040.
There is a need for novel treatments for preventing progression from early to intermediate and/or from intermediate to advanced stages of AMD to prevent loss of vision.
SUMMARY OF THE DISCLOSURE
In some embodiments, the disclosure provides for an antibody or antigen-binding fragment thereof that binds to a HTRA1 protein, or a functional fragment thereof In some
embodiments, the antibody or antigen-binding fragment also binds to one or more of HTRA2, HTRA3, or HTRA4. In some embodiments, the antibody or antigen-binding fragment also binds to HTRA4. In some embodiments, the antibody or antigen-binding fragment does not bind to, or displays weak binding to, HTRA2, HTRA3 and HTRA4. In some embodiments, the HTRA1 protein is a human HTRA1 protein, or functional fragment thereof. In some embodiments, the HTRA 1 protein comprises an amino acid sequence that is at least 80%, 85%, 90%, 92%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to the ammo acid sequence of SEQ ID NO: 121 or a functional fragment thereof. In some embodiments, the antibody or antigen-binding fragment thereof is capable of inhibiting any one or more HTRA1 function. In some embodiments, the antibody or antigen-binding fragment thereof is capable of inhibiting HTRA 1 proteolytic activity by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the proteolytic activity of a wildtype HTRA1 protein in the absence of the HTRA1 antibody or antigen-binding fragment thereof. In some embodiments, the antibody or antigen-binding fragment thereof is capable of inhibiting HTRA1 proteolytic activity in a cell by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the proteolytic activity of a wildtype HTRA 1 protein in the same cell type in the absence of the HTRA 1 antibody or antigen-binding fragment thereof. In some embodiments, the antibody or antigen-binding fragment thereof is capable of inhibiting HTRA1 proteolytic activity in any eye by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%,
45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the proteolytic activity of a wildtype HTRA1 protein in another eye in the absence of the HTRAl antibody or antigen-binding fragment thereof. In some embodiments, the antibody or antigen-binding fragment is capable of reducing HTRAl’s ability to cleave any one or more HTRA1 substrate. In some embodiments, the HTRA1 substrate is selected from the group consisting of: fibromodulin, elastin, clusterin, ADAM9, vitronectin, a2 -macroglobulin, talin- 1, fascin, LTBP-1, EFEMPI, fibulin 5, tau, RseA, and chloride intracellular channel protein. In some embodiments, the antibody or antigen-binding fragment is capable of inhibiting HTRA l’s ability to cleave a regulator of the complement cascade (e.g., vitronectin, fibromodulin or clusterin). In some embodiments, the antibody or antigen-binding fragment is capable of inhibiting HTRAl such that HTRA l’s ability to cleave an HTRA1 substrate and/or regulator of the complement cascade is reduced by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the ability of the HTRAl to cleave the HTRAl substrate and/or regulator of the complement cascade in the absence of the anti -HTRAl antibodies or antigen-binding fragments thereof. In some embodiments, the antibody or antigen-binding fragment is capable of inhibiting the ability of the HTRAl protein to tnmerize. In some embodiments, the antibody or antigen-binding fragment is capable of inhibiting the ability of the HTRA1 protein to trimerize by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the ability of the HTRA1 protein to trimerize in the absence of the antibody or antigen-binding fragment. In some embodiments, the antibody or antigen-binding fragment thereof comprises any one of, or combination of, the CDR sequences indicated in Figure 2. In some embodiments, the antibody or antigen-binding fragment thereof comprises any one of, or any combination of, the CDR a o acid sequences of SEQ ID NOs: 1-120. In some embodiments, the antibody or antigen-binding fragment thereof comprises a set of CDRS as forth as any one of the following sets of SEQ ID NOs: 1-6, 7-12, 13-18, 19-24, 25-30, 31 -36, 37-42, 43-48, 49-54, 55-60, 61-66, 67-72, 73-78, 79-84, 85-90, 91-96, 97-102, 103-108, 109-114, or 1 15-120. In some embodiments, the antibody or antigen-binding fragment thereof comprises the CDRs set forth as SEQ ID NOs: 1-6. In some embodiments, the antibody or antigen-binding fragment thereof comprises the CDRs set forth as SEQ ID NOs: 7-12. In some embodiments, the antibody or antigen-binding fragment thereof comprises the CDRs set forth as SEQ ID NOs: 19-24. In some embodiments, the antibody or antigen-binding fragment thereof comprises the CDRs set forth as SEQ ID NOs: 31-36. In some embodiments, the antibody or antigen-binding fragment thereof comprises the CDRs set forth as SEQ ID NOs: 37-42 In some embodiments, the antibody or antigen-binding fragment thereof comprises the CDRs set forth as SEQ ID NOs: 61-66. In some embodiments, the antibody or antigen-binding fragment thereof comprises the CDRs set forth as SEQ ID NOs: 73-78 In some
embodiments, the antibody or antigen-binding fragment thereof comprises the CDRs set forth as SEQ ID NOs: 103-108. In some embodiments, the antibody or antigen-binding fragment thereof comprises the CDRs set forth as SEQ ID NOs: 109-114. In some embodiments, the antibody or antigen-binding fragment thereof comprises the CDRs set forth as SE1Q ID NOs:
1 15-120. In some embodiments, the antibody or antigen-binding fragment thereof comprises any one of or combination of the amino acid sequences of SEQ ID NO: 1-120, but wherein the amino acid sequences comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 amino acid substitutions. In some embodiments, the antibody or antigen-binding fragment thereof comprises a set of CDRS as forth as any one of tire following sets of SEQ ID NOs: 1-6, 7-12, 13-18, 19-24, 25-30, 31-36, 37-42, 43-48, 49-54, 55-60, 61-66, 67-72, 73-78, 79-84, 85-90, 91-96, 97-102, 103-108, 109-114, or 1 15-120 but wherein the amino acid sequences comprise at least 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , or 12 amino acid substitutions. In some embodiments, die substitutions are conservative substitutions. In some embodiments, die substitutions either increase the binding affinity of the antibody or antigen-binding fragment to the HTRA1 epitope, or the substitutions result in a reduction of binding affinity of no more than 5%,
10%, 20%, 30%, 40%, or 50% as compared to the antibody or antigen-binding fragment lacking the substitutions. In some embodiments, the antibody or antigen-binding fragment is a full-length antibody. In some embodiments, the antibody or antigen-binding fragment is a monoclonal antibody. In some embodiments, the antibody or antigen-binding fragment is a humanized antibody. In some embodiments, the antibody or antigen-binding fragment is an antigen-binding fragment thereof. In some embodiments, the antigen-binding fragment is an scFv. In some embodiments, tire antigen-binding fragment is selected from the group consisting of: a Fab fragment, a F(ab')2 fragment, a Fab ' fragment, dAb fragment and/or a dsFv. In some embodiments, the antibody or antigen-binding fragment binds to one or more of the following HTRA1 domains: an insulin-like growth factor binding domain, a kazal domain, a trypsin-like peptidase domain, and/or a PDZ domain. In some embodiments, the antibody or antigen-binding fragment is capable of binding to a non-human species HTRA1. In some embodiments, the non-human species HTRA I is mouse, rat, rabbit, cow, monkey (e.g., eynomo!gus monkey), or ape (e.g. chimpanzee) HTRA! protein.
In some embodiments, the disclosure provides for a polynucleotide encoding any of the antibodies or antigen-binding fragments disclosed herein.
In some embodiments, the disclosure provides for a vector comprising any of the polynucleotides.
In some embodiments, the disclosure provides for a host cell comprising any of the vectors disclosed herein and capable of expressing the polynucleotide.
In some embodiments, the disclosure provides for a host cell expressing one or more of any of the polynucleotides disclosed herein encoding any of the antibody or antigen- binding fragments disclosed herein.
In some embodiments, the disclosure provides for a method of treating a disease or disorder in a subject in need thereof, wherein the disease or disorder is associated with aberrantly expressed HTRA1, wherein the method comprises administering to the subject any of the antibodies or antigen-binding fragments disclosed herein. In some embodiments, the disclosure provides for a method of treating a disease or disorder in a subject in need thereof, wherein HTRA! is expressed at a level at least 5%, 10%, 25%, 50%, 75%, 100%, 150%, 200%, 250%, 300%, 350%, 400%, 450%, or 500% greater in the subject having the disease or disorder as compared to the level in a control subject not having the disease or disorder, wherein the method comprises administering to the subject any of the antibodies or antigen- binding fragments disclosed herein. In some embodiments, the disclosure provides for a method of treating age-related m acular degeneration or polyploidal choroidal vaseu!opathy in a subject in need thereof, wherein the disease or disorder is associated with aberrantly expressed HTRA1, wherein the method comprises administering to the subject any of the antibodies or antigen-binding fragments disclosed herein. In some embodiments, the control subject is a subject of the same sex and/or of similar age as the subject having the disease or disorder. In some embodiments, the subject has one or more mutations in the HTRA i gene. In some embodiments, the one or more mutations are not in the coding sequence for the H'TRAl gene. In some embodiments, the one or more mutations are in 10q26 in a human subject. In some embodiments, the one or more mutations correspond to any one or more of tlie following polymorphisms in a human subject: rs61871744; rs59616332; rsl 1200630; rs61871745; rsl 1200632: rsl 1200633: rs61871746; rs61871747; rs370974631; rs200227426; rs201396317; rsl99637836; rsl 1200634; rs75431719; rs!0490924; rs!44224550;
rs36212731; rs36212732; rs36212733; rs3750848; rs3750847: rs3750846; rs566108895; rs3793917; rs3763764; rsl 1200638: rsl049331; rs2293870; rs2284665; rs60401382;
rsl 1200643; rs58077526; rs932275 and/or rs2142308. In some embodiments, the subject has age-related macular degeneration. In some embodiments, the subject has wet AMD. In some embodiments, the subject has dry AMD. In some embodiments, the subject has polyploidal choroidal vasculopathy. In some embodiments, the subject is a human. In some
embodiments, the human is at least 40 years of age. In some embodiments, the human is at least 50 years of age. In some embodiments, the human is at least 65 years of age. In some embodiments, the antibody or antigen-binding fragment is administered locally. In some embodiments, the antibody or antigen-binding fragment is administered intravitreaily. In some embodiments, the antibody or antigen-binding fragment is administered subretinally.
In some embodiments, the antibody or antigen-binding fragment is administered
systemicaily.
In some embodiments, the disclosure provides for a composition comprising any of the antibodies or antigen -binding fragments disclosed herein and a pharmaceutically acceptable carrier. In some embodiments, the composition is substantially pyrogen free.
BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 provides the experimental summary data from Example 1 for various specific anti- HTKA1 antibody /antigen-bmding fragment clones.
Figure 2 provides the heavy chain and light chain CDR amino acid sequences for the clones referenced in Figure 1.
Figure 3A and Figure 3B are graphs showing HTRA1 -mediated proteolysis of F AM-labeled elastin over time in the presence and absence of anti-HTRAl monoclonal antibodies (mAbs). Proteolysis is indicated by an increase in relative fluorescence units (RFU) over time. Figure 3C show's a bar graph summarizing the results from Figures 3 A and 3B relative to the antibod -free HTRA1 positive control reaction. Recombinant HTRA1 used in this assay contained a StreplI affinity tag and deletion of the HTRA1 N-terminal Mac domain (StrepII-AMac-HTRAl). Antibody assignments are as follows: 71= SR8047-38; 72= SR8047-40; 73= SR8047-17; 74= SR8047-44; 76= BM1 ; 77= SR8047-43; 78= SR8047-31; 79= SR8047-06; 80= SR8047-42; 82= BM2.
Figure 4 A shows Western Blot gel images from four time points for StrepII-AMac-HTRAl -mediated proteol sis of Fibuiin 5 in die presence and absence of anti-HTRAl mAbs over die course of 22 hours. Each proteolysis reaction is shown as a lane on a 12% SDS-PAGE gel. The contents of each reaction are tabulated above the gels. In each lane, bands representing Fibuiin 5, anti-HTRAl mAb heavy chain, HTRA1, and anti-HTRAl mAb light chain are shown. Figure 4B show's a summary table correlating each anti-HTRAl mAb to its respective number indicated in the gels of Figure 4 A.
Figure 5 A shows Western Blot gel images from three time points for StrepII-AMac-HTRA 1 -mediated proteolysis of Tan in the presence and absence of anti-HTRA 1 mAbs over the course of 22 hours.
Each proteolysis reaction is shown as a lane on a 12% SDS-PAGE gel. The contents of each reaction are tabulated above the gels. In each lane, bands representing Tan, anti-HTRAl mAb heavy chain, HTRAL and anti-HTRAl mAb light drain are shown. Figure 5B shows a summary table correlating each anti-HTRAl mAb to its respective number indicated in the gels of Figure 5 A.
Figure 6A show's Western Blot gel images from three time points for StrepII-AMac-HTRAl -mediated proteolysis of RseA in the presence and absence of anti-HTRAl mAbs over tire course of seven hours. Each proteolysis reaction is shown as a lane on a 12% SDS-PAGE gel. The contents of each reaction are tabulated above the gels. In each lane, bands representing anti-HTRAl mAb heavy chain,
HTRA 1, anti-HTRAl mAb light chain, and RseA are shown. Figure 6B show's a summary table correlating each anti-HTRAl mAb to its respective number indicated in the gels of Figure 6A. Figure 7 A shows Western Blot gel images from three time points for StrepII-AMac-HTRA 1 -mediated proteolysis of b-Casein in the presence and absence of anti-HTRAl niAbs over the course of thirty minutes. Each proteolysis reaction is shown as a lane on a 12% SDS-PAGE gel. The contents of each reaction are tabulated above the gels. In each lane, bands representing anti-HTR Al mAb heavy chain, HTRA1, b-Casein, and anti-HTRA 1 mAh light chain are shown. Figure 7B shows a summary' table correlating each anti-HTRA l mAb to its respective number indicated in the gels of Figure 7 A.
Figure 8 is a table summarizing the results of eadi HTRAl proteolysis assay correlated to their respective anti-HTRAl antibody name and lot number. In coluimi 2, the percent inhibition of elastin proteolysis is shown for each antibody used relative to the antibody -free HTRA1 positive control reaction. Inhibition of b-Casein, Tau, RseA, or Fibuiin 5 proteolysis is represented by +. Below or above average inhibition of proteolysis is represented by +/- or ++ respectively. No inhibition of proteolysis is represented by -. The anti-HTRA l SR8047-23 mAb activated StrepII-AMac-HTRA 1- mediated proteolysis of Tau, and this effect is indicated as“Activation”.
Figure 9 A is a graph showing StreplI-AMac-HTRAl-mediated proteolysis of F AM-labeled elastin over time in the presence and absence of anti-HTRAl Fab antibody fragments, or anti-HTRA l IgG antibodies. Proteolysis is indicated by an increase in relative fluorescence units (RFU) over time. Figure 9B shows a graph of the lAax for each reaction shown in Figure 9A.
DETAILED DESCRIPTION OF THE DISCLOSURE
The disclosure provides compositions and methods for treating, preventing, or inhibiting diseases of the eye. In one aspect, the disclosure provides anti-HTRAl antibodies or antigen binding fragments thereof. In another aspect, the disclosure provides methods of treating, preventing, or inhibiting diseases of the eye by intraocularly (e.g., intravitreaily) administering an effective amount of any of the anti-HTRA l antibodies or antigen-binding fragments thereof disclosed herein.
A wide variety of diseases of the eye may be treated or prevented using any of the anti- HTRA 1 antibodies or antigen-binding fragments thereof and methods provided herein. Diseases of the eye that may be treated or prevented using the anti-HTRAl antibodies or antigen-binding fragments thereof and methods of the disclosure include but are not limited to, glaucoma, macular degeneration (e.g., age-related macular degeneration), diabetic retinopathies, inherited retinal degeneration such as retinitis pigmentosa, retinal detachment or injury and retinopathies (such as retinopathies that are inherited, induced by surgery, trauma, an underlying aetiology such as severe anemia, SLE, hypertension, blood dyscrasias, systemic infections, or underlying carotid disease, a toxic compound or agent, or photically).
General Techniques
Unless otherwise defined herein, scientific and technical terms used in this application shall have the meanings that are commonly understood by those of ordinary skill in the art.
Generally, nomenclature used in connection with, and techniques of, pharmacology, cell and tissue culture, molecular biology, cell and cancer biology, neurobiology, neurochemistry', virology, immunology, microbiology, genetics and protein and nucleic acid chemistry, described herein, are those well known and commonly used in the art. In case of conflict, the present specification, including definitions, will control.
The practice of the present disclosure will employ, unless otherwise indicated, conventional techniques of molecular biology (including recombinant techniques), microbiology, cell biology, biochemistry and immunology, which are within the skill of the art. Such techniques are explained fully in the literature, such as, Molecular Cloning: A Laboratory' Manual, second edition (Sambrook et a!., 1989) Cold Spring Harbor Press; Oligonucleotide Synthesis (M.J. Gait, ed., 1984); Methods in Molecular Biology, Humana Press; Cell Biology: A Laboratory Notebook (J.E. Cellis, ed., 1998) Academic Press; Animal Cell Culture (R.I. Freshney, ed., 1987); Introduction to Cell and Tissue Culture (J.P. Mather and P.E. Roberts, 1998) Plenum Press; Cell and Tissue Culture: Laboratory Procedures (A.
Doyle, J.B. Griffiths, and D.G. Newell, eds., 1993-1998) J. Wiley and Sons; Methods in Enzymology (Academic Press, Inc.); Gene Transfer Vectors for Mammalian Cells (J.M. Miller and M.P. Ca!os, eds., 1987); Current Protocols in Molecular Biology (F.M. Ausubel et ah, eds., 1987); PCR: Tire Polymerase Chain Reaction, (Muilis et ah, eds., 1994); Sambrook and Russell, Molecular Cloning: A Laboratory Manual, 3rd. ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY (2001); Ausubel et ah, Current Protocols in Molecular Biology, John Wiley & Sons, NY (2002); Harlow and Lane Using Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, AW ( 1998); Co!igan et a!., Short Protocols in Protein Science, John Wiley & Sons, NY' (2003); Short Protocols in Molecular Biology (Wiley and Sons, 1999). Enzymatic reactions and purification techniques are performed according to manufacturer's specifications, as commonly accomplished in the art or as described herein. The
nomenclatures used in connection with, and the laboratory procedures and techniques of, analytical chemistry, biochemistry, immunology, molecular biology, synthetic organic chemistry, and medicinal and pharmaceutical chemistry described herein are those well known and commonly used in the art. Standard techniques are used for chemical syntheses, and chemical analyses.
Throughout this specification and embodim nts, the word "comprise," or variations such as "comprises" or "comprising," will be understood to imply the inclusion of a stated integer or group of integers but not the exclusion of any other integer or group of integers.
It is understood that wherever embodiments are described herein with the language “comprising,” otherwise analogous embodiments described in terms of“consisting of’ and/or “consisting essentially of’ are also provided.
The term“including” is used to mean“including but not limited to.”“Including” and “including but not limited to” are used interchangeably.
Any example(s) following the term“e.g.” or“for example” is not meant to be exhaustive or limiting.
Unless otherwise required by context, singular tenns shall include pluralities and plural terms shall include the singular.
The articles "a" and "an" are used herein to refer to one or to more than one (i.e., to at least one) of the grammatical object of the article. By way of example, "an element" means one element or more than one element. Reference to“about” a value or parameter herein includes (and describes) embodiments that are directed to that value or parameter per se. For example, description referring to“about X” includes description of“X.” Numeric ranges are inclusive of the numbers defining the range.
Notwithstanding that the numerical ranges and parameters setting forth the broad scope of the disclosure are approximations, the numerical values set forth in the specific examples are reported as precisely as possible. Any numerical value, however, inherently contains certain errors necessarily resulting from the standard deviation found in their respective testing measurements. Moreover, all ranges disclosed herein are to be understood to encompass any and all subranges subsumed therein. For example, a stated range of“1 to 10” should be considered to include any and all subranges between (and inclusive of) the minimum value of 1 and the maximum value of 10; that is, all subranges beginning with a minimum value of 1 or more, e.g., 1 to 6.1, and ending with a maximum value of 10 or less, e.g., 5.5 to 10.
Where aspects or embodiments of the disclosure are described in terms of a Markush group or other grouping of alternatives, the present disclosure encompasses not only the entire group listed as a whole, but each member of the group individually and all possible subgroups of the main group, but also the main group absent one or more of the group members. The present disclosure also envisages the explicit exclusion of one or more of any of the group members in the disclosure.
Exemplary methods and materials are described herein, although methods and materials similar or equivalent to those described herein can also be used in the practice or testing of the present disclosure. The materials, methods, and examples are illustrative only and not intended to he limiting.
Definitions
The following terms, unless otherwise indicated, shall be understood to have the following meanings: As used herein, "residue" refers to a position in a protein and its associated amino acid identity.
As known in the art,“polynucleotide,” or“nucleic acid,” as used interchangeably herein, refer to chains of nucleotides of any length, and include DNA and RNA. The nucleotides can be deoxyribonucleotides, ribonucleotides, modified nucleotides or bases, and/or their analogs, or any substrate that can be incorporated into a chain by DNA or RNA polymerase. A polynucleotide may comprise modified nucleotides, such as methylated nucleotides and their analogs if present, modification to the nucleotide structure may be imparted before or after assembly of the chain. The sequence of nucleotides may be interrupted by non-nucleotide components. A polynucleotide may be further modified after polymerization, such as by conjugation with a labeling component. Other types of modifications include, for example, “caps”, substitution of one or more of the naturally occurring nucleotides with an analog, intemucleotide modifications such as, for example, those with uncharged linkages (e.g., methyl phosphonates, phosphotriesters, phosphoamidates, carbamates, etc.) and with charged linkages (e.g., phosphorothioates, phosphorodithioates, etc.), those containing pendant moiefies, such as, for example, proteins (e.g , nucleases, toxins, antibodies, signal peptides, poly-L-lysine, etc.), those with intercalators (e.g., acridine, psoralen, etc.), those containing chelators (e.g., metals, radioactive metals, boron, oxidative metals, etc.), those containing alkylators, those with modified linkages (e.g., alpha anomeric nucleic acids, etc.), as well as unmodified forms of the polvnuc!eotide(s). Further, any of the hydroxyl groups ordinarily present in the sugars may be replaced, for example, by phosphonate groups, phosphate groups, protected by standard protecting groups, or activated to prepare additional linkages to additional nucleotides, or may be conjugated to solid supports. The 5’ and 3’ terminal OH can be phosphorylated or substituted with amines or organic capping group moieties of from 1 to 20 carbon atoms. Other hydroxyls may also be derivatized to standard protecting groups. Polynucleotides can also contain analogous forms of ribose or deoxyribose sugars that are generally known in the art, including, for example, 2,-0-methyl-, 2’-0-allyl, 2’-fluoro- or 2 - azido-ribose, carbocyclic sugar analogs, alpha- or beta-anomeric sugars, epimeric sugars such as arabmose, xyloses or lyxoses, pyranose sugars, furanose sugars, sedoheptuioses, acyclic analogs and abasic nucleoside analogs such as methyl riboside. One or snore pliosphodiester linkages may be replaced by alternative linking groups. These alternative linking groups include, but are not limited to, embodiments wherein phosphate is replaced by
P(0)S(“thioate”), P(S)S (“ditliioate”), (0)NR2 (“amidate”), P(0)R, P(0)OR\ CO or CH2 (“formacetal”), in which each R or R is independently H or substituted or unsubstituted alkyl (1-20 C) optionally containing an ether (-0-) linkage, aryl, alkenyl, cycloalkyl, cycloalkenyl or ara!dyi. Not all linkages in a polynucleotide need be identical. The preceding description applies to all polynucleotides referred to herein, including RNA and DNA.
As used herein, a“base”,“nucleotide base,” or“nucleobase,” is a heterocyclic pyrimidine or purine compound, which is a standard constituent of all nucleic acids, and includes the bases that form the nucleotides adenine (A), guanine (G), cytosine (C), thymine (T), and uracil (U). A nucleobase may further be modified to include, without limitation, universal bases, hydrophobic bases, promiscuous bases, size -expanded bases, and fluorinated bases. As used herein, the term“nucleotide” can include a modified nucleotide (such as, for example, a nucleotide mimic, abasic residue (Ab), or a surrogate replacement moiety). As used herein, the terms“sequence” and“nucleotide sequence” mean a succession or order of nucleobases or nucleotides, described with a succession of leters using standard nomenclature.
The terms "polypeptide", "oligopeptide", "peptide" and "protein" are used interchangeably herein to refer to chains of amino acids of any length. Tire chain may be linear or branched, it may comprise modified amino acids, and/or may be interrupted by non-amino acids. The terms also encompass an a mo acid chain that has been modified naturally or by
intervention; for example, disulfide bond formation, giycosylation, lipidation, acetylation, phosphorylation, or any other manipulation or modification, such as conjugation with a labeling component. Also included within die definition are, for example, polypeptides containing one or more analogs of an amino acid (including, for example, unnatural amino acids, etc.), as well as other modifications known in the art. It is understood that the polypeptides can occur as single chains or associated chains.
“Homologous,” in all its grammatical forms and spelling variations, refers to the relationship between two proteins that possess a“common evolutionary origin,” including proteins from superfamilies in the same species of organism, as well as homologous proteins from different species of organism. Such proteins (and their encoding nucleic acids) have sequence homology, as reflected by their sequence similarity, whether in terms of percent identity or by the presence of specific residues or motifs and conserved positions.
However, in common usage and in the instant application, the term“homologous,” when modified with an adverb such as“highly,” may refer to sequence similarity and may or may not relate to a common evolutionary origin.
The term“sequence similarity,” in all its grammatical forms, refers to the degree of identity or correspondence between nucleic acid or a mo acid sequences that may or may not share a common evolutionary' origin.
“Percent (%) sequence identity” or“percent (%) identical to” with respect to a reference polypeptide (or nucleotide) sequence is defined as the percentage of amino acid residues (or nucleic acids) in a candidate sequence that are identical with the ammo acid residues (or nucleic acids) in the reference polypeptide (nucleotide) sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for aligning sequences, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared.
As used herein, a“host cell” includes an individual cell or cell culture that comprises one or more polynucleotides capable of expressing any of the anti-HTRAl antibodies or antigen binding fragments thereof disclosed herein. As used herein, a "vector," refers to a recombinant plasmid comprising one or more polynucleotides encoding any of, or any component of, the anti-HTRAl antibodies or antigen-binding fragments thereof disclosed herein.
As used herein,“purify,” and grammatical variations thereof, refers to the removal, whether completely or partially, of at least one impurity from a mixture containing the polypeptide and one or more impurities, which thereby improves the level of purity of the polypeptide in the composition (i.e., by decreasing the amount (ppm) of impurity(ies) in the composition).
As used herein,“substantially pure” refers to material which is at least 50% pure (i.e., free from contaminants), more preferably, at least 90% pure, more preferably, at least 95% pure, yet more preferably, at least 98% pure, and most preferably, at least 99% pure. The temis “patient”,“subject”, or“individual” are used interchangeably herein and refer to either a human or a non-human animal. These terms include mammals, such as humans, non-human primates, laboratory animals, livestock animals (including bovines, porcines, camels, etc.), companion animals (e.g., canines, felines, other domesticated animals, etc.) and rodents (e.g., mice and rats). In some embodiments, the subject is a human that is at least 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90 or 95 years of age.
In one embodiment, the subject has, or is at risk of developing a disease of the eye. A disease of the eye, includes, without limitation, AMD, retinitis pigmentosa, rod-cone dystrophy, Leber’s congenital amaurosis, Usher's syndrome, Bardet-Biedl Syndrome, Best disease, retinoschisis, Stargardt disease (autosomal dominant or autosomal recessive), untreated retinal detachment, pattern dystrophy, cone-rod dystrophy, achromatopsia, ocular albinism, enhanced S cone syndrome, diabetic retinopathy, age-related macular degeneration, retinopathy of prematurity, sickle ceil retinopathy, Congenital Stationary Night Blindness, glaucoma, or retinal vein occlusion. In another embodiment, the subject has, or is at risk of developing glaucoma, Leber's hereditary optic neuropathy, lysosomal storage disorder, or peroxisomal disorder. In another embodiment, the subject is in need of optogenetic therapy. In another embodiment, the subject has shown clinical signs of a disease of the eye.
In some embodiments, the subject has, or is at risk of developing AMD. In some embodiments, the AMD is Early AMD; Intermediate AMD; Advanced non-neovascular (“Dry'”) AMD; or Advanced neovascular (“Wet”) AMD.
Clinical signs of a disease of the eye include, but are not limited to, decreased peripheral vision, decreased central (reading) visi on, decreased night visi on, loss of color perception, reduction in visual acuity, decreased photoreceptor function, and pigmentary changes. In one embodiment, the subject shows degeneration of the outer nuclear layer (ONL). In another embodiment, the subject has been diagnosed with a disease of the eye. In yet another embodiment, the subject has not yet shown clinical signs of a disease of the eye. As used herein, the terms“prevent”,“preventing” and“prevention” refer to the prevention of the recurrence or onset of, or a reduction in one or more symptoms of a disease or condition (e.g., a disease of the eye) in a subject as result of the administration of a therapy (e.g., a prophylactic or therapeutic agent). For example, in the context of the administration of a therapy to a subject for an infection,“prevent”,“preventing” and“prevention” refer to the inhibition or a reduction in the development or onset of a disease or condition (e.g., a disease of the eye), or the prevention of the recurrence, onset, or development of one or more symptoms of a disease or condition (e.g., a disease of the eye), in a subject resulting from the administration of a therapy (e.g., a prophylactic or therapeutic agent), or the administration of a combination of therapies (e.g., a combination of prophylactic or therapeutic agents). “Treating” a condition or patient refers to taking steps to obtain beneficial or desired results, including clinical results. With respect to a disease or condition (e.g., a disease of the eye), treatment refers to the reduction or amelioration of the progression, severity, and/or duration of an infection (e.g., a disease of the eye or symptoms associated therewith), or the amelioration of one or more symptoms resulting from the administration of one or more therapies (including, but not limited to, the administration of one or more prophylactic or therapeutic agents). “Administering” or“administration of’ a substance, a compound or an agent (e.g., any of the antibodies or antigen-binding fragments disclosed herein) to a subject can be carried out using one of a variety of methods known to those skilled in the art. For example, a compound or an agent can be administered intravitreally or subretinally. In particular embodiments, the compound or agent is administered intravitreally. In some embodiments, administration may be local. In other embodiments, administration may be systemic.
Administering can also be performed, for example, once, a plurality of times, and/or over one or more extended periods. In some aspects, the administration includes both direct administration, including self-administration, and indirect administration, including the act of prescribing a drug. For example, as used herein, a physician who instructs a patient to self- administer a drug, or to have the drug administered by another and/or who provides a patient with a prescription for a drug is administering the drug to the patient.
As used herein, the term“ocular cells” refers to any cell in, or associated with tire function of, the eye. The term may refer to any one or more of photoreceptor cells, including rod, cone and photosensitive ganglion cells, retinal pigment epithelium (RPE) cells, glial cells, Muller cells, bipolar cells, horizontal cells, amacrine cells. In one embodiment, the ocular cells are bipolar cells. In another embodiment, the ocular cells are horizontal cells. In another embodiment, the ocular cells are ganglion cells. In particular embodiments, tire cells are RPE cells. As used herein, the term“capable of’ means that the referenced composition (e.g. , any of the antibodies or antigen-binding fragments disclosed herein) has the capability to perform a specific function, but that it is not required to be performing that specific function at any specific moment in time. The term“capable of’ encompasses instances where the composition is actively performing a specific function. Each embodiment described herein may be used individually or in combination with any oilier embodiment described herein.
Anti-HTRAl Antibodies or Antigen-Binding Fragments Thereof
HTRA 1 is a serine protease that targets a variety of proteins, including extracellular matrix proteins such as fibronectin. Fibronectin fragments resulting from HTRA1 cleavage are able to further induce synovial cells to up-regulate MMP1 and MMP3 production. There is evidence that HTRA1 may also degrade proteoglycans, such as aggrecan, decorin and fibromodulin. By cleaving proteoglycans, HTRA1 may release soluble FGF- glycosaminoglycan complexes that promote the range and intensity of FGF signals in the extracellular space HTRA 1 also regulates the availability of insulin-like growth factors (IGFs) by cleaving IGF-binding proteins. Intracellularly, HTRA1 degrades TSC2, leading to the activation of TSC2 downstream targets.
Overexpression of HTRA1 alters the integrity of Bruch’s membrane, which permits choroid capillaries to invade across the extracellular matrix in conditions such as wet age-erlated macular degeneration. Tong et ah, 2010, Mol . Vis , 16: 1958-81. HTR.41 also inhibits signaling mediated by TGF-beta family members, which may regulate many physiological processes, including retinal angiogenesis and neuronal survival and maturation during development. It has been previously determined that a single-nucleotide polymorphism (is 11200638) in the promoter region of the HTRA I gene was found to be significantly associated with susceptibility to AMD in various patient populations. Tong et al., 2010.
In some embodiments, the antibody or antigen-binding fragment also binds to one or more of HTRA2, HTRA3, or HTRA4. In some embodiments, the antibody or antigen-binding fragment also binds to HTRA4. In some embodiments, the antibody or antigen -binding fragment does not bind to, or displays 'eak binding to, HTRA2, HTRA3 and HTRA4.
In some embodiments, any of the anti-HTRA l antibodies or antigen-binding fragments thereof disclosed herein is capable of inhibiting any one or more HTRAi function. In some embodiments, the anti-HTRA 1 antibodies or antigen-binding fragments thereof are capable of inhibiting all functions of an HTRA 1 protein. In some embodiments, the anti-HTRAl antibodies or antigen-binding fragments thereof are capable of binding to an HTRAi protein and inhibiting the function of the HTRAI protein.
In some embodiments, any of the anti-HTRAl antibodies or antigen-binding fragments thereof disclosed herein is capable of inhibiting proteolytic activity of an HTRAI protein. In some embodiments, the anti-HTRAl antibodies or antigen-binding fragments thereof is capable of inhibiting HTRAI proteolytic activity' by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the proteolytic activity of a wildtype HTRAI protein (e.g., an HTRA I protein having the amino acid sequence of SEQ ID NO: 121) in the absence of the HTRAI antibody' or antigen- binding fragment thereof. In some embodiments, the anti-HTRAl antibodies or antigen- binding fragments thereof are capable of inhibiting HTRA1 proteolytic activity in a cell by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the proteolytic activity of a wildtype HTRAl protein in the same cell type in the absence of the antibody or antigen-binding fragment. In some embodiments, the anti-HTRAl antibodies or antigen-binding fragments thereof are capable of inhibiting HTRAl proteolytic activity in an eye by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the proteolytic activity of a wildtype HTRA l protein in another eye in the absence of the antibody or antigen-binding fragment.
In some embodiments, any of the anti-HTRA l antibodies or antigen-binding fragments thereof disclosed herein is capable of inhibiting HTRAl’ s ability to cleave any one or more HTRAl substrate. In some embodiments, the HTRAl substrate is selected from the group consisting of: fibromodulin, clusterin, ADAM9, elastin, vitronectin, a2-macroglobulin, talin- 1, fascin, LTBP-1, EFEMPl, fihulin 5, tau, RseA, and chloride intracellular channel protein. In some embodiments, the anti-HTRAl antibodies or antigen-binding fragments thereof are capable of inhibiting HTRAl’s ability to cleave a regulator of the complement cascade (e.g., vitronectin, fibromodulin or clusterin). In some embodiments, the anti-HTRAl antibodies or antigen-binding fragments thereof are capable of inhibiting HTRA I such that HTRA 1’ s ability to cleave an HTRAl substrate and/or regulator of the complement cascade is reduced by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the ability7 of the HTRAl to cleave the HTRAl substrate and/or regulator of the complement cascade m the absence of the anti- HTRA 1 antibodies or antigen-binding fragments thereof.
As used herein, the term“antibody” (Ab), which is synonymous with the term
“immunoglobulin” (Ig), means a tetramer comprising two heavy (H) chains (about 50-70 kDa) and two light (L) chains (about 25 kDa) inter-connected by disulfide bonds. There are two types of light chain: l and K. In humans they are similar, but only one type is present in each antibody. Heavy chains are classified as mu, delta, gamma, alpha, or epsilon, and define the antibody's isotype as IgM, IgD, IgG, IgA, and IgEl respectively. See generally,
Fundamental Immunology Ch. 7 (Paul, W., ed., 2nd ed. Raven Press, N .Y. (1989)). Each heavy chain (herein sometimes referred to as H-chain or He) is compri sed of a heavy chain variable domain (VH, or H-variable domain) and a heavy chain constant region (CH). The heavy chain constant region is comprised of three domains, CHI, CH2 and CH3. Each light chain (herein sometimes referred to as L-chain or Lc) is comprised of a light chain variable domain (VL, or L-variable domain) and a light chain constant region. The light chain constant region is comprised of one domain, CL. Within light and heavy chains, the variable and constant regions are joined by a“J” region of about 12 or more amino acids, with the heavy chain also including a“D” region of about 3 or more amino acids. The VH and VI, regions can be further subdivided into regions of hypervariability', termed“complementarity determining regions"’ (CDR), interspersed with regions that are more conserved, termed “framework regions” (FR). Each VH and VL is composed of three CDRs (H-CDR herein designates a CDR from the heavy chain; and L-CDR herein designates a CDR from the light chain) and four FRs, arranged from amino-terminus to carboxyl- terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDRS, FR4.
In some embodiments, any of the antibodies or antigen-binding fragments disclosed herein comprises 1, 2, 3, 4, 5, or 6 of the CDRs from any of the antibodies or antigen-binding fragments disclosed herein. In some embodiments, any of the antibodies or antigen-binding fragments disclosed herein comprises 1 , 2 or 3 of the CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to any one of SEQ ID NOs: 125-144. In some embodiments, the antibody' or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 125. In some embodiments, the antibody or antigen-binding fragment comprises 1 , 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 126. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain am o acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 127. In some embodiments, the antibody or antigen-binding fragment comprises i, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 128. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 129. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 130. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 131. In some embodiments, the antibody or antigen -binding fragment comprises 1 , 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 132. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 133 In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 134. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 135. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 136. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 137. In some embodiments, the antibody or antigen-binding fragment comprises 1 , 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 138. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 139. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 140. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain ammo acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 141. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 142. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 143. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 144. In some embodiments, any of the antibodies or antigen-binding fragments disclosed herein comprises 1, 2 or 3 of the CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to any one of SEQ ID NOs: 145-164. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 145. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 146. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain ammo acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 147. In some embodiments, the antibody or antigen- binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 148. In some embodiments, the antibody or antigen-binding fragment comprises 1 , 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 149. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 150. In some embodiments, the antibody or antigen-bindmg fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 151. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 152. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 153. In some embodiments, tire antibody or antigen-binding fragment comprises 1 , 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 154. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 155. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 156. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 157. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 158. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 159. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 160. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 161. In some embodiments, the antibody or antigen-binding fragment comprises 1 , 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 162. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to S EQ ID NO: 163. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 164. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from the sequence of SEQ ID NO: 143. In some embodiments, the antibody or antigen-binding fragment comprises 3 CDRs from the sequence of SEQ ID NO: 143. In some embodiments, the antibody or antigen-binding fragment comprises l, 2 or 3 CDRs from tire sequence of SEQ ID NO: 144. In some embodiments, the antibody or antigen-binding fragment comprises 3 CDRs from the sequence of SEQ ID NO: 144. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from the sequence of SEQ ID NO: 163. In some embodiments, the antibody or antigen-binding fragment comprises 3 CDRs from the sequence of SEQ ID NO: 163. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from the sequence of SEQ ID NO: 164. In some embodiments, the antibody or antigen-binding fragment comprises 3 CDRs from the sequence of SEQ ID NO: 164. In some embodiments, the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from the sequence of SEQ ID NO: 143 and 1, 2 or 3 CDRs from the sequence of SEQ ID NO: 163. In some embodiments, the antibody or antigen-binding fragment comprises 3 CDRs from the sequence of SEQ ID NO: 143 and 3 CDRs from the sequence of SEQ ID NO: 163. In some embodiments, the antibody or antigen-binding fragment comprises 1 , 2 or 3 CDRs from the sequence of SEQ ID NO: 144 and comprises 1 , 2 or 3 CDRs from the sequence of SEQ ID NO: 164. In some
embodiments, the antibody or antigen-binding fragment comprises 3 CDRs from the sequence of SEQ ID NO: 144 and comprises 3 CDRs from the sequence of SEQ ID NO: 164. In some embodiments, the assignment of amino acids to each domain is in accordance with the definitions of Rabat, Sequences of Proteins of Immunological Interest (National Institutes of Health, Bethesda, MD (1987 and 1991 )). In some embodiments, the assignment of amino acids to each domain is in accordance with Chothia & Lesk, j. Mol. Biol. 196:901-917
(1987); Chothia et ai.. Nature 342:878-883 (1989) In some embodiments, the assignment of amino acids to each domain is in accordance with MacCailum. In some embodiments, the assignment of amino acids to each domain is in accordance with AbM. In some
embodiments, the assignment of amino acids to each domain is in accordance with IMGT. In some embodiments, any of the antibodies or antigen-binding fragments disclosed herein comprises a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to any of SEQ ID NOs: 125-144. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 125. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 126. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 127. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 128. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 129 In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 130. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SI Q ID NO: 131. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 132. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 133. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 134. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 135. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 136. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 137. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 138. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 139. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 140. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 141. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 142. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 143 In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 144.
In some embodiments, any of the antibodies or antigen-binding fragments disclosed herein comprises a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to any of SEQ ID NOs: 145-164. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 145. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 146. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 147. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 148. In some embodiments, the antibody or antigen -binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 149. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 150. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 151. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 152. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 153. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 154. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 155. in some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 156. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 157. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 158. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 159 In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 160. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 161. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 162. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 163. In some embodiments, the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 164.
In some embodiments, the antibody or antigen-binding fragment comprises a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 143, and further comprises a variable light chain am o acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 163. In some embodiments, the antibody or antigen-binding fragment comprises a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 144, and further comprises a variable light chain ammo acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 164. As used herein, the term“antigen-binding fragment" refers to one or more fragmen ts of an antibody that retain the ability to specifically bind to an antigen. It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody. Non-limiting examples of binding fragments encompassed within the term “antigen-binding fragment” include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CHI domains: (ii) a F(ab')2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fab' fragment which is obtained by cleaving a disulfide bond of the hinge region of the F(ab')2; (iv) a Fd fragmen t consisting of the VH and CHI domains; (v) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (vi) a dAb fragment (Ward et af, (1989) Nature 341 :344-546), which consists of a VH domain; (vii) an isolated complementarity determining region (CDR); and (viii) a dsFv, which consists of a VH: :VL heterodimer stabilized by a disulfide bond. Furthermore, although tire two domains of the Fv fragment,
VL and VH, are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv)); see e.g., Bird et al. Science 242:423-426 (1988) and Huston et al. Proc. Natl. Acad. Sci. USA 85:5879-5883 (1988)). Also within the scope of this disclosure are antigen-binding molecules comprising a VN and/or a VL, In the case of a VH, the molecule may also comprise one or more of a CH 1, hinge, CH2 or CH3 region. Such single chain antibodies are also intended to be encompassed within the term“antigen-binding fragment” of an antibody. Other forms of single chain antibodies, such as diabodies are also encompassed Diabodies are bivalent, bispecific antibodies in which VH and VL domains are expressed on a single polypeptide chain, but using a linker that is too short to allow for pairing between the two domains on the same chain, thereby forcing the domains to pair with complementary’ domains of another chain and creating two antigen binding sites (see e.g., Holliger et al. Proc. Natl. Acad. Sci. USA 90:6444-6448 (1993); Po!jak et al. Structure 2: 1121-1123 (1994)).
As used herein, the term“antigen-binding fragment” also includes, e.g., single domain antibodies such as camelized single domain antibodies. See, e.g., Muyldermans et al. (2001) Trends Biochem Sci 26:230-235; Nutta!l et al. (2000) Curr Pharm Biotech 1 :253-263;
Reichrnarm et al. (1999) J Immunol Meth 231 :25-38; PCT application publication nos. WO 94/04678 and WO 94/25591; and U.S. patent no. 6,005,079, all of which are incorporated herein by reference in their entireties. In some embodiments, the disclosure provides single domain antibodies comprising two VH domains with modifications such that single domain antibodies are formed.
As used herein, the term‘epitope” or“antigenic determinant” refers to a site on an antigen (e.g., HTRAi) to which an immunoglobulin or antibody specifically binds. An epitope can be formed both from contiguous ammo acids or noncontiguous amino acids juxtaposed by tertiary folding of a protein. An epitope formed from contiguous amino acids is typically retained on exposure to denaturing solvents, whereas an epitope formed by tertiary folding is typically lost on treatment with denaturing solvents. An epitope typically includes at least 3,
4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 ammo acids a unique spatial conformation. Methods for determining what epitopes are bound by a given antibody (i.e., epitope mapping) are well knowm in the art and include, for example, immunob!otting and immunoprecipitation assays, wherein overlapping or contiguous peptides from HTRAI are tested for reactivity with the given anti -HTRAI antibody. Methods of determining spatial conformation of epitopes include techniques in the art and those described herein, for example, x-ray crystallography and 2-dimensional nuclear magnetic resonance (see, e.g., Epitope Mapping Protocols in Methods in Molecular Biology, Vol. 66, G E. Morris, Ed (1996))
In some embodiments, any of the antibodies or antigen binding-fragments disclosed herein binds to an epitope in human HTRAI. In some embodiments, the antibody or antigen binding fragment binds to an epitope in the amino acid sequence of SEQ ID NO: 121, or a functional fragment thereof.
In some embodiments, any of the antibodies or antigen binding-fragments disclosed herein comprises any one of, or any combination of, the CDR amino acid sequences indicated in Figure 2. In some embodiments, any of the antibodies or antigen-binding fragments disclosed herein comprises any one of, or any combination of, the CDR ammo acid sequences of SEQ ID NGs: 1-120. In some embodiments, the antibody or antigen binding fragment comprises a set of CDRS as forth as any one of the following sets: SEQ ID NOs: 1-6, 7-12, 13-18, 19-24, 25-30, 31-36, 37-42, 43-48, 49-54, 55-60, 61-66, 67-72, 73-78, 79-84, 85-90, 91-96, 97-102, 103-108, 109-114, or 115-12.0. In some embodiments, the antibody or antigen binding fragment comprises the CDRs set forth as SEQ ID NOs: 1 -6. In some embodiments, the antibody or antigen binding fragment comprises the CDRs set forth as SEQ ID NOs: 7- 12. In some embodiments, the antibody or antigen binding fragment comprises the CDRs set forth as SEQ ID NOs: 19-24. In some embodiments, the antibody or antigen binding fragment comprises the CDRs set forth as SEQ ID NOs: 31-36. In some embodiments, the antibody or antigen binding fragment comprises the CDRs set forth as SEQ ID NOs: 37-42. In some embodiments, the antibody or antigen binding fragment comprises the CDRs set forth as SEQ ID NOs: 61-66. In some embodiments, the antibody or antigen binding fragment comprises the CDRs set forth as SEQ ID NOs: 73-78. In some embodiments, the antibody or antigen binding fragment comprises the CDRs set forth as SEQ ID NOs: 103- 108. In some embodiments, the antibody or antigen binding fragment comprises the CDRs set forth as SEQ ID NOs: 109-114. In some embodiments, the antibody or antigen binding fragment comprises the CDRs set forth as SEQ ID NOs: 115-120. In some embodiments, the antibody or antigen binding fragment comprises any one of or combination of any of the foregoing CDR amino acid sequences, but with 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 amino acid substitutions. In some embodiments, the substitutions are conservative substitutions. In preferred embodiments, the substitutions either increase the binding affinity of the antibody or antigen-binding fragment to the HTRA1 epitope, or the substitutions result in a reduction of binding affinity of no more than 5%, 10%, 20%, 30%, 40%, or 50% as compared to the antibody or antigen-binding fragment lacking the substitutions. The following groups each contain ammo acids that are conservative substitutions for one another. These groups are exemplary' as other conservative substitutions are known to those of skill in the art.
1 ) Alanine (A), Glycine (G);
2) Aspartic acid (D), Glutamic acid (E);
3) Asparagine (N), Glutamine (Q);
4) Arginine (R), Lysine (K);
5) Isoleucine (I), Leucine (L), Methionine (M), Valine (V);
6) Phenylalanine (F), Tyrosine (Y), Tryptophan (W);
7) Serine (S), Threonine (T); and
8) Cysteine (C), Methionine (M)
By way of example, when an aspartic acid at a specific residue is mentioned, also contemplated is a conservative substitution at the residue, for example, glutamic acid.
Nonconservative substitutions, for example, substituting a proline with glycine, are also contemplated. In some embodiments, any of the CDRs disciosd herein lacks one or two, but no more than three amino acids at the N-terminus and/or C-terminus of any of the CDR sequences disclosed herein. In some embodiments, any of the antibodies or antigen-binding fragments disclosed herein comprises any of the heavy chain CDRs disclosed herein. In some embodiments, any of the antibodies or antigen binding fragments disclosed herein comprises any of the combinations of heavy chain CDRi, CDR2, and CDR3 disclosed herein. In some embodiments, the antibody or antigen-binding fragment thereof comprises any of the following sets of three heavy chain CDRs: SEQ ID NOs: 1-3, 7-9, 13-15, 19-21, 25-27, 31-33, 37-39, 43-45, 49-51, 55-57, 61-63, 67-69, 73-75, 79-81, 85-87, 91-93, 97-99, 103-105, 109-11 1 or 115-1 17, or any of the foregoing CDR combinations comprising one or more (e.g. , 1 , 2, 3, 4, 5, 6, 7, 8, 9, or 10) conservative substitutions. In a particular embodiment, tire antibody or antigen-binding fragment thereof comprises the heavy chain CDRs of SEQ ID NOs: 109-111. In a particular embodiment, the antibody or antigen-binding fragment thereof comprises the heavy chain CDRs of SEQ ID NOs: 109-1 1 1. In a particular embodiment, the antibody or antigen binding fragment thereof comprises the heavy chain CDRs of SEQ ID N Os: 109-111 comprising one or more (e.g. , 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) conservative substitutions. In another embodiment, the antibody or antigen-binding fragment thereof comprises the heavy chain CDRs of SEQ ID NOs: 115-117. In a particular embodiment, the antibody or antigen- binding fragment thereof comprises the heavy chain CDRs of SEQ ID NOs: 115-117 comprising one or more (e.g. , 1 , 2, 3, 4, 5, 6, 7, 8, 9, or 10) conservative substitutions. In some embodiments, the antibody or antigen-binding fragment comprises a common light chain.
In some embodiments, the antibody or antigen-binding fragment is a full-length antibody. In some embodiments, the antibody is a monoclonal antibody. In some embodiments, the antibody is a human antibody. In some embodiments, the antibody is a humanized antibody.
In some embodiments, the antibody or antigen -binding fragment is a chimeric antibody. In some embodiments, the antibody or antigen-binding fragment is a humanized antibody or antigen-binding fragment. In some embodiments, the antibody or antigen-binding fragment is an antigen-binding fragment. In some embodiments, the antigen-binding fragment is selected from the group consisting of a Fab fragment, a F(ab')2 fragment, a Fab' fragment, a dAb, or an scFv. In some embodiments, any of the antibodies or antigen-binding fragments disclosed herein is capable of binding to and inhibiting one or more function of HTRA1. In some embodiments, the antibody or antigen-binding fragment binds to one or more of the following HTRA1 domains: die insulin-like growth factor binding domain, die kazal domain, the trypsin-like peptidase domain, and/or the PDZ domain.
In some embodiments, any of tire antibodies or antigen-bind g fragments thereof disclosed herein bind to a human HTRA1 protein with a binding affinity (KD) of at least 50 nM, at least 25 nM, at least 10 nM, at least 7 nM, at least 5 nM, at least 3 nM, at least 2 nM, at least 1 nM, at least 500 pM, at least 100 pM, at least 50 pM or at least 10 pM.
In some embodiments, the antibody or antigen-binding fragment is capable of binding to HTRA1 such that one or more HTRA1 functions are inhibited. In some embodiments, the antibody or antigen-binding fragment binds to HTRA1 such that HTRAl’s ability to cleave any one or more HTRA 1 substrate is reduced. In some embodiments, the HTRA 1 substrate is selected from the group consisting of: fibromodulin, elastin, clusterin, LTBP-1, EFEMP1, ADAM9, vitronectin, a2-macroglobulin, talin-1, fascin, fibulin 5, tan, RseA, and chloride intracellular channel protein. In some embodiments, the antibody or antigen-binding fragment binds to HTRA1 such that HTRAl’s ability to cleave a regulator of the complement cascade (e g., vitronectin, fibromodulin or clusterin) is reduced. In some embodiments, the antibody or antigen-binding fragment binds to HTRA1 such that HTRAl’s ability to cleave an HTRA1 substrate and/or regulator of the complement cascade is reduced by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the ability of the HTRA1 to cleave the HTRA1 substrate and/or regulator of the complem ent cascade in the absence of the antibody or antigen-binding fragment thereof. In some embodiments, the antibody or antigen -binding fragment is capable of inhibiting proteolytic activity of an HTRA1 protein. In some embodiments, the antibody or antigen-binding fragm nt is capable of inhibiting HTRA! proteolytic activity by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the proteolytic activity of a wildtype HTRA1 protein in the absence of the antibody or antigen-binding fragment. In some embodiments, the antibody or antigen-binding fragment is capable of inhibiting HTRA I proteolytic activity' in a cell by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the proteolytic activity of a wildtype HTRA1 protein m the same cell type in the absence of the antibody or antigen-binding fragment. In some embodiments, the antibody' or antigen-binding fragm nt is capable of inhibiting HTRA1 proteolytic activity in an eye by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the proteolytic activity of a wildtype HTRA1 protein in an eye in the absence of the antibody or antigen-binding fragment. In some embodiments, any of the antibodies or antigen-binding fragments disclosed herein is capable of inhibiting HTRA1 trimerization. In some embodiments, the antibody or antigen binding fragment is capable of inhibiting HTRA1 trimerization by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the trimerization of wildtype HTRA1 protein in the absence of the antibody or antigen-binding fragment.
In some embodiments, any of the antibodies or antigen-binding fragments disclosed herein is capable of binding human HTRA1. In some embodiments, the antibody or antigen-binding fragment is capable of binding to a non-human species H'TRAl . In some embodiments, the non-human species HTRA1 is mouse, rat, rabbit, cow, monkey (e.g , cynomolgus monkey), and/or ape (e.g. chimpanzee) HTRA 1 protein.
Antibodies that recognize the same or overlapping epitope as a known antibody or compete for binding with a known antibody can be identified using routine techniques. Such techniques include, for exampl e, an immunoassay, which shows the ability of one antibody to block the binding of another antibody to a target antigen, i.e., a competitive binding assay. Competitive binding is determined in an assay in which tire immunoglobulin under test inhibits specific binding of a reference antibody to a common antigen, such as H'TRAl . Numerous types of competitive binding assays are known, for example: solid phase direct or indirect radioimmunoassay(RIA), solid phase direct or indirect enzyme immunoassay(EIA), sandwich competition assay (see Stahli et a , Methods in Enzymology 9:242 (1983)); solid phase direct biotin-avidin EIA (see Kirkland et ai., J. Immunol. 137:3614 (1986)); solid phase direct labeled assay, solid phase direct labeled sandwich assay (see Harlow and Lane, Antibodies:A Laboratory Manual, Cold Spring Harbor Press (1988)); solid phase direct label RIA using 1-125 label (see Morel et a , Mol. Immunol. 25(I):7 (1988)); solid phase direct biotin-avidin EIA (Cheung et a! , Virology 176:546 ( 1990)); and direct labeled RIA.
(Moldenhauer et a , Scand. I. Immunol. 32:77 (1990)). Typically, such an assay involves the use of purified antigen bound to a solid surface or cells bearing either of these, an unlabeled test immunoglobulin and a labeled reference immunoglobulin. Competitive inhibition is measured by determining the amount of label bound to the solid surface or cells in the presence of the test immunoglobulin. Usually the test immunoglobulin is present in excess. Usually when a competing antibody is present in excess, it will inhibit specific binding of a reference antibody to a common antigen by at least about 50-55%, 55-60%, 60-65%, 65-70%, 70-75% or more.
Other techniques include, for example, epitope mapping methods, such as, x-ray analyses of crystals of antigen: antibody complexes which provides atomic resolution of the epitope and mass spectrometry combined with hydrogen/deuterium (H/D) exchange which studies the conformation and dynamics of antigen: antibody interactions. Other methods monitor the binding of the antibody to antigen fragments or mutated variations of the antigen where loss of binding due to a modification of an amino acid residue within the antigen sequence is often considered an indication of an epitope component. In addition, computational combinatorial methods for epitope mapping can also be used. These methods rely on the ability of the antibody of interest to affinity isolate specific short peptides from combinatorial phage display peptide libraries. The peptides are then regarded as leads for the definition of the epitope corresponding to the antibody used to screen the peptide library'. For epitope mapping, computational algorithms have also been developed which have been shown to map conformational discontinuous epitopes.
The disclosure also features methods for producing any of the anti-HTRAl antibodies or antigen-binding fragments thereof described herein in some embodiments, methods for preparing an antibody described herein can include immunizing a subject (e.g , a non-human mammal) with an appropriate immunogen (e.g., an HTRA1 polypeptide having the amino acid sequence of SEQ ID NO: 121 or a fragment thereof). Suitable immunogens for generating any of the antibodies described herein are set forth herein. For example, to generate an antibody that binds to HTRA1, a skilled artisan can immunize a suitable subject (e.g., a non- human mammal such as a rat, a mouse, a gerbil, a hamster, a dog, a cat, a pig, a goat, a horse, or a non-human primate) with human HTRA1.
A suitable subject (e.g., a non-human mammal) can be immunized with the appropriate antigen along with subsequent booster immunizations a number of times sufficient to elicit the production of an antibody by the mammal. The immunogen can be administered to a subject (e.g., a non-human mammal) with an adjuvant. Adjuvants useful in producing an antibody in a subject include, but are not limited to, protein adjuvants; bacterial adjuvants. e.g., whole bacteria (BCG, Corynebacterium parvum or Salmonella minnesota) and bacterial components including cell wall skeleton, trehalose dimycolate, monophosphoryl lipid A, methanol extractable residue (MER) of tubercle bacillus, complete or incomplete Freund’s adjuvant; viral adjuvants; chemical adjuvants, e.g., aluminum hydroxide, and iodoacetate and cholesteryl hemisuccinate. Other adjuvants that can be used in the methods for inducing an immune response include, e.g., cholera toxin and parapoxvims proteins. See also Bieg et al. (1999) Autoimmunity 31 ( 1 ) : 15 -24. See also, e.g., Lodmell et al. (2000) Vaccine 18: 1059- 1066; Johnson et al . (1999) J Med Chem 42:4640-4649; Baldridge et al (1999) Methods 19: 103-107; and Gupta et al. (1995) Vaccine 13(14): 1263-1276.
In some embodiments, the methods include preparing a hybridoma cell line that secretes a monoclonal antibody that binds to the immunogen. For example, a suitable mammal such as a laboratory mouse is immunized with a HTRAl polypeptide as described above. Antibody- producing cells (e.g., B cells of the spleen) of die immunized mammal can be isolated two to four days after at least one booster immunization of the immunogen and then grown briefly in culture before fusion with cells of a suitable myeloma cell line. The cells can be fused in the presence of a fusion promoter such as, e.g., vaccinia vinis or polyethylene glycol. Tire hybrid cells obtained in die fusion are cloned, and cell clones secreting die desired antibodies are selected. For example, spleen cells of Balb/c mice immunized with a suitable immunogen can be fused with cells of the myeloma cell line PAI or the myeloma cell line Sp2/0-Ag 14. After the fusion, the cells are expanded in suitable culture medium, which is supplemented with a selection medium, for example HAT medium, at regular intervals in order to prevent normal myeloma cells from overgrowing the desired hybridoma cells. The obtained hybrid ceils are then screened for secretion of the desired antibodies, e.g., an antibody that binds to human HTRA1 as described herein.
In some embodiments, a skilled artisan can identify an anti-HTRA 1 antibody from a non- immune biased library as described in, e.g., U.S. patent no. 6,300,064 (to Knappik et al.; Morphosys AG) and Schoonbroodt et al. (2005) Nucleic Acids Res 33(9):e81.
In some embodiments, the methods described herein can involve, or be used in conjunction with, e.g., phage display technologies, bacterial display, yeast surface display, eukaryotic viral display, mammalian cell display, and cell-free (e.g., ribosomal display) antibody screening techniques (see, e.g., Etz et al. (2001) J Bactenol 183:6924-6935; Cornells (2000) Curr Opin Biotechnol 1 1 :450-454; Klemm et al. (2000) Microbiology 146:3025-3032;Kieke et al. (1997) Protein Eng 10: 1303-1310;Yeung et al. (2002) BiotechnolProg 18:212- 220;Boder et al. (2000) Methods Enzymology328:430-444; Grabherr et al. (2001) Comb Chem High Throughput Screen 4: 185-192; Michael et al. (1995) Gene Ther 2:660-668; Pereboev et al. (2001) J Virol 75:7107-7113; Schaffitzel et al. (1999) I Immunol Methods 231 : 1 19-135; and Hanes et al. (2000) Nat Bioteclmol 18: 1287-1292).
Methods for identifying antibodies using various phage display methods are known in the art. In phage display methods, functional antibody domains are displayed on the surface of phage particles which cany- the polynucleotide sequences encoding them. Such phage can be utilized to display antigen-binding domains of antibodies, such as Fab, Fv, or disulfide-bond stabilized Fv antibody fragments, expressed from a repertoire or combinatorial antibody library (e.g., human or murine). Phage used in these methods are typically filamentous phage such as fd and M13. The antigen binding domains are expressed as a recombinantly fused protein to any of the phage coat proteins pill, pVIII, or pIX. See, e.g., Shi et al. (2010) JMB 397:385-396. Examples of phage display methods that can be used to make the
immunoglobulins, or fragments thereof, described herein include those disclosed in Brinkman et al. (1995) J Immunol Methods 182:41-50; Ames et al. (1995) J Immunol Methods 184: 177-186; Kettleborougli et al. (1994) Bur I Immunol 24:952-958; Persic et al. (1997) Gene 187:9-18; Burton et al. (1994) Advances m Immunology 57: 191-280; and PCT publication nos. WO 90/02809, WO 91/10737, WO 92/01047, WO 92/18619, WO 93/1 1236, WO 95/15982, and WO 95/20401. Suitable methods are also described in, e.g., U.S. patent nos.5, 698, 426; 5,223,409; 5,403,484; 5,580,717; 5,427,908; 5,750,753; 5,821,047;
5,571,698; 5,427,908; 5,516,637; 5,780,225; 5,658,727; 5,733,743 and 5,969, 108.
In some embodiments, the phage display antibody libraries can be generated using mRNA collected from B cells from the immunized mammals. For example, a splenic cell sample comprising B cells can be isolated from mice immunized with a HTRAl polypeptide as described above. mRNA can be isolated from the cells and converted to cDNA using standard molecular biology techniques. See, e.g., Sambrook et al. (1989)‘"Molecular Cloning: A Laboratory Manual, 2nd Edition,” Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.; Harlow and Lane (1988), supra; Benny K. C. Lo (2004), supra; and Borrebaek (1995), supra. The cDNA coding for the variable regions of the heavy chain and light chain polypeptides of immunoglobulins are used to construct the phage display library. Methods for generating such a library' are described in, e.g., Merz et al. (1995) J Neurosci Methods 62(l-2):213-9; Di Niro et al. (2005) Biochem J 388(Pt 3):889-894; and Engherg et al. (1995) Methods Mol Biol 51 :355-376.
In some embodiments, a combination of selection and screening can be employed to identify an antibody of interest from, e.g., a population of hybridoma-derived antibodies or a phage display antibody library. Suitable methods are known in the art and are described in, e.g., Hoogenboom (1997) Trends in Biotechnology 15:62-70; Brinkman et al. (1995), supra; Ames et ai. (1995), supra; Kettleborough et al. (1994), supra; Persic et ai. (1997), supra; and Burton et al. (1994), supra. For example, a plurality of phage mid vectors, each encoding a fusion protein of a bacteriophage coat protein (e.g., pill, pVIII, or pIX of M13 phage) and a different antigen-combining region are produced using standard molecular biology techniques and then introduced into a population of bacteria (e.g., E. coli). Expression of the bacteriophage in bacteria can, in some embodiments, require use of a helper phage. In some embodiments, no helper phage is required (see, e.g., Chasteen et ah, (2006) Nucleic Acids Res 34(21):el45). Phage produced from tire bacteria are recovered and then contacted to, e.g., a target antigen bound to a solid support (immobilized). Phage may also be contacted to antigen in solution, and the complex is subsequently bound to a solid support.
A subpopulation of antibodies screened using the above methods can be characterized for their specificity and binding affinity for a particular antigen (e.g., human HTRA 1) using any immunological or biochemical based method known in the art. For example, specific binding of an antibody to HTRA1, may be determined for example using immunological or biochemical based methods such as, but not limited to, an ELISA assay, SPR assays, immunoprecipitation assay, affinity' chromatography, and equilibrium dialysis as described above immunoassays which can be used to analyze immunospecific binding and cross- reactivity of the antibodies include, but are not limited to, competitive and non- competitive assay systems using techniques such as Western blots, RIA, ELISA (enzyme linked immunosorbent assay),“sandwich” immunoassays, immunoprecipitation assays, immunodiffusion assays, agglutination assays, complement-fixation assays,
immunoradiometric assays, fluorescent immunoassays, and protein A immunoassays. Such assays are routine and well known in the art.
In embodiments where the selected CDR amino acid sequences are short sequences (e.g., fewer than 10-15 amino acids in length), nucleic acids encoding the CDRs can be chemically synthesized as described in, e.g., Shiraishi et al. (2007) Nucleic Acids Symposium Series 51(1): 129-130 and U.S. Patent No. 6,995,259. For a given nucleic acid sequence encoding an acceptor antibody, the region of the nucleic acid sequence encoding the CDRs can be replaced with the chemically synthesized nucleic acids using standard molecular biology techniques. The 5’ and 3' ends of the chemically synthesized nucleic acids can be synthesized to comprise sticky end restriction enzyme sites for use in cloning tire nucleic acids into the nucleic acid encoding the variable region of the donor antibody.
In some embodiments, the anti-HTRAl antibodies described herein comprise an altered heavy chain constant region that has reduced (or no) effector function relative to its corresponding unaltered constant region. Effector functions involving the constant region of tire anti-HTRAl antibody may be modulated by altering properties of the constant or Fc region. Altered effector functions include, for example, a modulation in one or more of the following activities: antibody-dependent cellular cytotoxicity (ADCC), complement- dependent cytotoxicity (CDC), apoptosis, binding to one or more Fc-receptors, and pro- inflammatory' responses. Modulation refers to an increase, decrease, or elimination of an effector function activity exhibited by a subject antibody containing an altered constant region as compared to the activity of the unaltered fomi of the constant region. In particular embodiments, modulation includes situations in which an activity is abolished or completely absent.
An altered constant region with altered FcR binding affinity and/or ADCC activity and/or altered CDC activity is a polypeptide which has either an enhanced or diminished FcR binding activity and/or ADCC activity and/or CDC activity compared to the unaltered form of the constant region. An altered constant region which displays increased binding to an FcR binds at least one FcR with greater affinity than the unaltered polypeptide. An altered constant region which displays decreased binding to an FcR binds at least one FcR with lower affinity than the unaltered form of the constant region. Such variants which display- decreased binding to an FcR may possess little or no appreciable binding to an FcR, e.g., 0 to 50% (e.g , less than 50, 49, 48, 47, 46, 45, 44, 43, 42, 41 , 40, 39, 38, 37, 36, 35, 34, 33, 32,
31, 30, 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11 , 10, 9, 8, 7, 6,
5, 4, 3, 2, or 1%) of the binding to the FcR as compared to the level of binding of a native sequence immunoglobulin constant or Fc region to the FcR. Similarly, an altered constant region that displays modulated ADCC and/or CDC activity- may exhibit either increased or reduced ADCC and/or CDC activity compared to the unaltered constant region. For example, in some embodiments, the anti-HTRAl antibody comprises an altered constant region can exhibit approximately 0 to 50% (e.g., less than 50, 49, 48, 47, 46, 45, 44, 43, 42, 41, 40, 39, 38, 37, 36, 35, 34, 33, 32, 31, 30, 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1%) of the ADCC and/or CDC activity of the unaltered form of the constant region. An anti-HTRAl antibody described herein comprising an altered constant region displaying reduced ADCC and/or CDC may exhibit reduced or no ADCC and/or CDC activity7.
In some embodiments, an anti-HTRAl antibody or antigen-binding fragment described herein exhibits reduced or no effector function. In some embodiments, an anti-HTRA 1 antibody comprises a hybrid constant region, or a portion thereof, such as a G2/G4 hybrid constant region (see e.g., Burton et al . (1992) Adv Immun 51 : 1-18; Canfield et al. (1991) I Exp Med 173: 1483-1491 ; and Mueller et al . ( 1997) Mol Immunol 34(6):441 -452). See above.
In some embodiments, an anti-HTRAl antibody or antigen-binding fragment may contain an altered constant region exhibiting enhanced or reduced complement dependent cytotoxicity (CDC). Modulated CDC activity may he achieved by introducing one or more amino acid substitutions, insertions, or deletions in an Fc region of the antibody. See, e.g., IJ.S. patent no. 6,194,551. Alternatively or additionally, cysteine residue(s) may be introduced in the Fc region, thereby allowing interchain disulfide bond formation in this region. The homodimeric antibody thus generated may have improved or reduced internalization capability and/or increased or decreased complement-mediated cell killing. See, e.g., Caron et al. (1992) I Exp Med 176: 1191-1195 and Shopes (1992) Immunol 148:2918-2922; PCT publication nos. WO 99/51642 and WO 94/29351; Duncan and Winter (1988) Nature 322:738-40; and U.S. Patent Nos. 5,648,260 and 5,624,821.
The antibodies or antigen-binding fragments thereof described herein can be produced using a variety of techniques known in the art of molecular biology and protein chemistry . For example, a nucleic acid encoding one or both of the heavy and light chain polypeptides of an antibody can be inserted into an expression vector that contains transcriptional and translational regulatory sequences, which include, e.g., promoter sequences, ribosomal binding sites, transcriptional start and stop sequences, translational start and stop sequences, transcription terminator signals, polyadenylation signals, and enhancer or activator sequences. The regulatory sequences include a promoter and transcriptional start and stop sequences. In addition, the expression vector can include more than one replication system such that it can be maintained in two different organisms, for example in mammalian or insect cells for expression and in a prokaryotic host for cloning and amplification.
Several possible vector systems are available for the expression of cloned heavy chain and light chain polypeptides from nucleic acids in mammalian cells. One class of vectors relies upon the integration of the desired gene sequences into the host cell genome. Cells which have stably integrated DNA can be selected by simultaneously introducing drug resistance genes such as E. coli gpt (Mulligan and Berg (1981) Proc Natl Acad Sei USA 78:2072) or Tn5 neo (Southern and Berg (1982) Mol Appl Genet 1 :327). The selectable marker gene can be either linked to the DNA gene sequences to be expressed, or introduced into the same cell by co-transfection (Wigler et al. ( 1979) Cell 16:77). A second class of vectors utilizes DNA elements which confer autonomously replicating capabilities to an extrachromosomal plasmid. These vectors can be derived from animal viruses, such as bovine papillomavirus (Sa er et al. (1982) Proc Natl Acad Sci USA, 79:7147), cytomegalovirus, polyoma virus (Deans et al. (1984) Proc Natl Acad Sci USA 81 : 1292), or SV40 virus (Lusky and Botchan (1981) Nature 293:79).
The expression vectors can be introduced into cells in a manner suitable for subsequent expression of the nucleic acid. The method of introduction is largely dictated by the targeted ceil type, discussed below. Exemplary methods include CaPCri precipitation, liposome fusion, cationic liposomes, electroporation, viral infection, dextran-mediated transfection, polybrene-mediated transfection, protoplast fusion, and direct microinjection.
Appropriate host cells for the expression of antibodies or antigen-binding fragments thereof include yeast, bacteria, insect, plant, and mammalian cells. Of particular interest are bacteria such as E. coli, fungi such as Saccharomyces cerevisiae and Pichia pastons, insect cells such as SF9, mammalian cell lines (e.g., human cell lines), as well as primary cell lines.
In some embodiments, an antibody or fragment thereof can be expressed in, and purified from, transgenic animals (e.g., transgenic mammals). For example, an antibody can be produced in transgenic non-human mammals (e.g., rodents) and isolated from milk as described in, e.g., Houdebine (2002) Curr Opin Biotechnol 13(6):625-629; van Kuik- Romeijn et al . (2000) Transgenic Res 9(2): 155-159; and Pollock et al. (1999) J Immunol Methods 231(1-2): 147-157. The antibodies and fragments thereof can be produced from the cells by culturing a host cell transformed with the expression vector containing nucleic acid encoding the antibodies or fragments, under conditions, and for an amount of time, sufficient to allow expression of the proteins. Such conditions for protein expression will vary with the choice of the expression vector and the host cell, and will be easily ascertained by one skilled in the art through routine experimentation. For example, antibodies expressed in E. coil can be refolded from inclusion bodies (see, e.g., Hou et a!. (1998) Cytokine 10:319-30). Bacterial expression systems and methods for their use are well known in the art (see Current Protocols in Molecular Biology, Wiley & Sons, and Molecular Cloning— A Laboratory Manual—3rd Ed., Cold Spring Harbor Laboratory Press, New York (2001)). The choice of codons, suitable expression vectors and suitable host cells will vary depending on a number of factors, and may be easily optimized as needed. An antibody (or fragment thereof) described herein can be expressed in mammalian cells or in oilier expression systems including but not limited to yeast, baeu!oviras, and in vitro expression systems (see, e.g., Kaszubska et al. (2000) Protein Expression and Purification 18:213 -220)
Following expression, the antibodies and fragments thereof can be isolated. An antibody or fragment thereof can be isolated or purified in a variety' of ways known to those skilled in the art depending on what other components are present in the sample. Standard purification methods include electrophoretic, molecular, immunological, and chromatographic techniques, including ion exchange, hydrophobic, affinity, and reverse- phase HPLC chromatography. For example, an antibody can be purified using a standard anti-antibody column (e.g., a protein -A or protein-G column). Ultrafiltration and diafiltration techniques, in conjunction with protein concentration, are also useful. See, e.g.. Scopes (1994)“Protein Purification, 3rd edition,,, Springer-Verlag, New York City, New York. The degree of purification necessary will vary depending on the desired use. In some instances, no purification of the expressed antibody or fragments thereof will be necessary.
Methods for determining the yield or purity of a purified antibody or fragment thereof are known in the art and include, e.g., Bradford assay, UV spectroscopy, Biuret protein assay, Lowry protein assay, amido black protein assay, high pressure liquid chromatography (HPLC), mass spectrometry (MS), and gel electrophoretic methods (e.g., using a protein stain such as Coomassie Blue or colloidal silver stain). The antibodies or antigen -binding fragments thereof can be modified following their expression and purification. The modifications can be covalent or non- covalent modifications. Such modifications can be introduced into the antibodies or fragments by, e.g , reacting targeted amino acid residues of the polypeptide with an organic derivatizing agent that is capable of reacting with selected side chains or terminal residues. Suitable sites for modification can be chosen using any of a variety of criteria including, e.g., structural analysis or amino acid sequence analysis of the antibodies or fragments. in some embodiments, the antibodies or antigen-binding fragments thereof can be conjugated to a heterologous moiety. Tire heterologous moiety can be, e.g., a heterologous polypeptide, a therapeutic agent (e.g., a drug), or a detectable label such as, but not limited to, a radioactive label, an enzymatic label, a fluorescent label, a heavy metal label, a luminescent label, or an affinity tag such as biotin or streptavidin. Suitable heterologous polypeptides include, e.g., an antigenic tag (e.g., FLAG (DYKDDDDK (SEQ ID NO: 122)), polyhistidine (6-His; HHHHHH (SEQ ID NO: 123), hemagglutinin (HA: YPYDVPDYA (SEQ ID NO: 124)), ghitathione-S-transferase (GST), or maltose-binding protein (MBP)) for use in purifying the antibodies or fragments. Heterologous polypeptides also include polypeptides (e.g., enzymes) that are useful as diagnostic or detectable markers, for example, luciferase, a fluorescent protein (e.g., green fluorescent protein (GFP)), or chloramphenicol acetyl transferase (CAT). Suitable radioactive labels include, e.g., 32P, 33P, 14C, 1251, 1311, 35S, and 3H. Suitable fluorescent labels include, without limitation, fluorescein, fluorescein isothiocyanate (FITC), green fluorescent protein (GFP), DyLight™ 488, phycoerythrin (PE), propidium iodide (PI), PerCP, PE-Alexa Fluor® 700, Cy5, allophycocyanin, and Cy7 Luminescent labels include, e.g., any of a variety of luminescent lanthanide (e.g., europium or terbium) chelates. For example, suitable europium chelates include the europium chelate of diethyienetiiaminepentaaceticacid (DTPA)or tetraazacyclododecane- i ,4,7, 10-tetraacetic acid (DOTA). Enzymatic labels include, e.g., alkaline phosphatase, CAT, luciferase, and horseradish peroxidase.
Two proteins (e.g., an antibody and a heterologous moiety) can be cross-linked using any of a number of known chemical cross linkers. Examples of such cross linkers are those which link two amino acid residues via a linkage that includes a“hindered’ disulfide bond. In these linkages, a disulfide bond within the cross-linking unit is protected (by hindering groups on ei ther side of the disulfide bond) from reduction by the action, for example, of reduced glutathione or die enzyme disulfide reductase. One suitable reagent, 4-succinimidyl oxycarbonyl-a-methyl- a(2-pyridyldithio) toluene (SMPT), forms such a linkage between two proteins utilizing a terminal lysine on one of the proteins and a terminal cysteine on the other. Heterobifunctional reagents that cross-link by a different coupling moiety on each protein can also be used. Other useful cross-linkers include, without limitation, reagents which link two am o groups (e.g., N-5-azido-2-nitrobenzoyloxysuccinimide), two sulfliydryi groups (e.g., 1,4-bis-maleimidobutane), an amino group and a sulfliydryi group (e.g., m- maleimido benzoyl-N-hydroxy succinimide ester), an amino group and a carboxyl group (e.g., 4-jp-azidosalicylamido] butylamine), and an amino group and a guanidinium group that is present in the side chain of arginine (e.g., p-azidophenyl glyoxal monohydrate).
In some embodiments, a radioactive label can he directly conjugated to the amino acid backbone of the antibody. Alternatively, the radioactive label can be included as
partofalargermolecule(e .g . , 125 Iinmeta- [ 1251] iodophenyl-N -hy droxysuccinimide
([1251]mIPNHS) which binds to free amino groups to form meta-iodophenyl (mlP) derivatives of relevant proteins (see, e.g., Rogers et al (1997) J Nuci Med 38: 1221 -1229) or chelate (e.g., to DOTA or DTP A) which is in turn bound to the protein backbone. Methods of conjugating the radioactive labels or larger molecules/chelates containing them to the antibodies or antigen-binding fragments described herein are known in the art. Such methods involve incubating the proteins with the radioactive label under conditions (e.g., pH, salt concentration, and/or temperature) that facilitate binding of the radioactive label or chelate to the protein (see, e.g., U.S. Patent No. 6,001,329).
Methods for conjugating a fluorescent label (sometimes referred to as a“fluorophore”) to a protein (e.g., an antibody) are known in the art of protein chemistry. For example, fluorophores can be conjugated to free ammo groups (e.g., of lysines) or sulfhydryl groups (e.g., cysteines) of proteins using succinimidyl (NHS) ester or tetrafluorophenyl (TFP) ester moieties attached to the fluorophores. In some embodiments, the fluorophores can be conjugated to a heterobifunctional cross-linker moiety such as suifo-SMCC Suitable conjugation methods involve incubating an antibody protein, or fragment thereof, with the fluorophore under conditions that facilitate binding of the fluorophore to the protein. See, e.g., Welch and Redvaniy (2003)‘Handbook of Radiopharmaceuticals: Radiochemistry and
Applications,” John Wiley and Sons (ISBN 0471495603). In some embodiments, the antibodies or fragments can be modified, e.g., with a moiety that improves the stabilization and/or retention of the antibodies in circulation, e.g., in blood, serum, or other tissues. For example, the antibody or fragment can be PEGylated as described in, e.g., Lee et al. (1999) Bioconjug Chem 10(6): 973-8; Kinstler et al. (2002) Advanced Drug Deliveries Reviews 54:477-485; and Roberts et al. (2002) Advanced Drug Delivery Reviews 54:459-476 or HESylated (Fresenius Kabi, Germany; see, e.g., Pavisic et al. (2010) Int J Pfaarm 387(1-2): 110-119). The stabilization moiety can improve the stability, or retention of, the antibody (or fragment) by at least about 1.5 (e.g., at least about 2, 5, 10,
15, 20, 25, 30, 40, or 50 or more) fold.
In some embodiments, the antibodies or antigen -binding fragments thereof described herein can be glycosylated. In some embodiments, an antibody or antigen-binding fragment thereof described herein can be subjected to enzymatic or chemical treatment, or produced from a ceil, such that the antibody or fragment has reduced or absent glycosylation. Methods for producing antibodies with reduced glycosylation are known in the art and described in, e.g., U.S. patent no 6,933,368; Wright et al (1991) EMBO .1 10( 10) :2717-2723 ; and Co et al. (1993) Mol Immunol 30: 1361.
The disclosure also provides for one or more polynucleotides that encode any of the anti- HTRAl antibodies or antigen-binding fragments thereof. In some embodiments, the disclosure provides for one or more vectors encoding any one or more of the polynucleotides disclosed herein. In some embodiments, the one or more vectors further comprise one or more promoters that may facilitate the expression of the one or more polynucleotides. In some embodiments, the disclosure provides for a host cell capable of expressing any one or more of the polynucleotides disclosed herein.
In some embodiments, the disclosure provides for vector comprising any of die
polynucleotides disclosed herein encoding any of the antibodies or antigen-binding fragments disclosed herein. In some embodiments, the disclosure provides for a method of administering any of the vectors disclosed herein to a subject in need thereof. In some embodiments, the vector is a viral vector. In some embodiments, the viral vector is a retrovirus, lentivirus, or baculovims vector. In some embodiments, the viral vector is an adenoviral vector. In particular embodiments, the viral vector is an AAV vector. A variety of rAAV vectors may be used to deliver the polynucleotides to the eye and to direct its expression. More than 30 naturally occurring serotypes of AAV from humans and non human primates are known. Many natural variants of the AAV capsid exist, and an rAAV vector of the disclosure may be designed based on an AAV with properties specifically suited for ocular cells.
Recombinant AAV vectors of the present disclosure may be generated from a variety of adeno-associated viruses. For example, ITRs from any AAV serotype are expected to have similar structures and functions with regard to replication, integration, excision and transcriptional mechanisms. Examples of AAV serotypes include AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV 11 and AAV12. In some embodiments, the rAAV vector is generated from serotype AAV1, AAV2, AAV4, AAV5, or AAV8. These serotypes are known to target photoreceptor cells or the retinal pigment epithelium. In particular embodiments, the rAAV vector is generated from serotype AAV2 In certain embodiments, the AAV serotypes include AAVrhS, AAVrhBR or AAVrhl O. It will also be understood that the rAAV vectors may be chimeras of two or more serotypes selected from serotypes AAV1 through AAV12. The tropism of the vector may be altered by packaging the recombinant genome of one serotype into capsids derived from another AAV serotype. In some embodiments, the ITRs of the rAAV vims may be based on the ITRs of any one of AAV1-12 and may be combined with an AAV capsid selected from any one of AAV1-12, AAV-DJ, AAV-DJ8, AAV-DJ9 or other modified serotypes. In certain embodiments, any AAV capsid serotype may be used with the vectors of the disclosure. Examples of AAV serotypes include AAV1, AAV2, AAV 3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV 10, AAVl !, AAV 12, AAV-DJ, AAV-DJ8, AAV-DJ9, AAVrhS, AAVrhSR or AAVrh lO. In certain embodiments, the AAV capsid serotype is AAV2.
Desirable AAV fragments for assembly into vectors may include the cap proteins, including the vpl, vp2, vp3 and hypervariable regions, the rep proteins, including rep 78, rep 68, rep 52, and rep 40, and the sequences encoding these proteins. These fragments may be readily utilized in a variety of vector systems and host cells. Such fragments maybe used, alone, in combination with other AAV serotype sequences or fragments, or in combination with elements from other AAV or non-AAV viral sequences. As used herein, artificial AAV serotypes include, without limitation, AAV with a non-naturally occurring capsid protein. Such an artificial capsid may be generated by any suitable technique using a selected AAV sequence (e.g , a fragment of a vpl capsid protein) in combination with heterologous sequences which may be obtained from a different selected AAV serotype, non-contiguous portions of the same AAV serotype, from a non-AAV viral source, or from a non-viral source. An artificial AAV serotype may be, without limitation, a pseudotyped AAV, a chimeric AAV capsid, a recombinant AAV capsid, or a "humanized" AAV capsid
Pseudotyped vectors, wherein the capsid of one AAV is replaced with a heterologous capsid protein, are useful in the disclosure. In some embodiments, the AAV is AAV2/5. In another embodiment, the AAV is AAV2/8. When pseudotyping an AAV vector, the sequences encoding each of the essential rep proteins may be supplied by different AAV sources (e.g., AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8). For example, the rep78/68 sequences may be from AA V2, whereas the rep52/40 sequences may he from AAV8.
In one embodiment, the vectors of the disclosure contain, at a minimum, sequences encoding a selected AAV serotype capsid, e.g., an AAV2 capsid or a fragment thereof. In another embodiment, the vectors of the disclosure contain, at a minimum, sequences encoding a selected AAV serotype rep protein, e.g., AAV2 rep protein, or a fragment thereof.
Optionally, such vectors may contain both AAV cap and rep proteins. In vectors in which both AAV rep and cap are provided, the AAV rep and AAV cap sequences can both be of one serotype origin, e.g., all AAV2 origin. In certain embodiments, the vectors may comprise rep sequences from an AAV serotype which differs from that which is providing the cap sequences. In some embodiments, the rep and cap sequences are expressed from separate sources (e.g., separate vectors, or a host cell and a vector). In some embodiments, these rep sequences are fused in frame to cap sequences of a different AAV serotype to form a chimeric AAV vector, such as AAV2/8 described in US Patent No. 7,282, 199, which is incorporated by reference herein. Examples of AAV serotypes include AAV1 , AAV2, AAV3, AAV4, AAV 5, AAV6, AAV7, AAV8, AAV9, AAVIO, AAV11, AAV 12, AAV-DJ, AAV-DJ 8, AAV-DJ9, AAVrh8, AAVrh8R or AAVrhlO. In some embodiments, the cap is derived from AAV2.
In some embodiments, any of the vectors disclosed herein includes a spacer, i.e., a DNA sequence interposed between the promoter and the rep gene ATG start site. In some embodiments, the spacer may be a random sequence of nucleotides, or alternatively, it may encode a gene product, such as a marker gene. In some embodiments, the spacer may contain genes which typically incorporate start/ stop and poly A sites. In some embodiments, the spacer may be a non-coding DNA sequence from a prokaryote or eukaryote, a repetitive non- coding sequence, a coding sequence without transcriptional controls or a coding sequence with transcriptional controls. In some embodiments, the spacer is a phage ladder sequences or a yeast ladder sequence. In some embodiments, the spacer is of a size sufficient to reduce expression of the rep78 and rep68 gene products, leaving the rep52, rep40 and cap gene products expressed at normal levels. In some embodiments, the length of the spacer may therefore range from about 10 bp to about 10.0 kbp, preferably in the range of about 100 bp to about 8.0 kbp. In some embodiments, the spacer is less than 2 kbp in length.
In certain embodiments, the capsid is modified to improve therapy. The capsid may be modified using conventional molecular biology techniques. In certain embodiments, the capsid is modified for minimized immunogenicity, better stability' and particle lifetime, efficient degradation, and/or accurate deli very of any of the polynucleotides disclosed herein encoding any of the antibodies or antigen-binding fragments disclosed herein. In some embodiments, the modification or mutation is an amino acid deletion, insertion, substitution, or any combination thereof in a capsid protein. A modified polypeptide may comprise 1, 2, 3, 4, 5, up to 10, or more amino acid substitutions and/or deletions and/or insertions. A
"deletion" may comprise the deletion of individual amino acids, deletion of small groups of amino acids such as 2, 3, 4 or 5 amino acids, or deletion of larger amino acid regions, such as the deletion of specific ammo acid domains or other features. An“insertion” may comprise the insertion of individual amino acids, insertion of small groups of amino acids such as 2, 3, 4 or 5 amino acids, or insertion of larger a mo acid regions, such as the insertion of specific amino acid domains or other features. A“substitution” comprises replacing a wild type amino acid with another (e.g., a non-wild type amino acid). In some embodiments, the another (e.g., non-wild type) or inserted amino acid is Ala (A), His (H), Lys (K), Phe (F), Met (M), Thr (T), Gin (Q), Asp (D), or Giu (E). In some embodiments, the another (e.g., non-wild type) or inserted amino acid is A. In some embodiments, the another (e.g., non-wild type) amino acid is Arg (R), Asn (N), Cys (C), Giy (G), He (I), Leu (L), Pro (P), Ser (S), Trp (W), Tyr (Y), or Val (V). Conventional or naturally occurring amino acids are divided into the following basic groups based on common side-chain properties: (1 ) non-polar: Norleucine, Met, Ala, Val, Leu, He; (2) polar without charge: Cys, Ser, Thr, Asn, Gin; (3) acidic (negatively charged): Asp, Glu; (4) basic (positively charged): Lys, Arg; and (5) residues that influence chain orientation: Gly, Pro; and (6) aromatic: Tip, Tyr, Phe, His. Conventional amino acids include L or D stereochemistry. In some embodiments, the another (e.g., non-wild type) amino acid is a member of a different group (e.g., an aromatic amino acid is substituted for a non-polar amino acid). Substantial modifications in the biological properties of the polypeptide are accomplished by selecting substitutions that differ significantly in their effect on maintaining (a) the structure of the polypeptide backbone in the area of the substitution, for example, as a b-sheet or helical conformation, (b) the charge or hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain . Naturally occurring residues are divided into groups based on common side-chain properties: (1 ) Non-polar: Norleucine, Met, Ala, Val, Leu, Ile;(2) Polar without charge: Cys, Ser, Thr, Asn, Gln;(3) Acidic (negatively charged): Asp, G3u;(4) Basic (positively charged): Lys, Arg;(5) Residues that influence chain orientation: Gly, Pro; and(6) Aromatic: Trp, Tyr, Phe, His. In some embodiments, the another (e.g., non wild type) ammo acid is a member of a different group (e.g., a hydrophobic amino acid for a hydrophilic amino acid, a charged amino acid for a neutral amino acid, an acidic amino acid for a basic amino acid, etc.). In some embodiments, the another (e.g., non-wild type) amino acid is a member of the same group (e.g., another basic amino acid, another acidic amino acid, another neutral ammo acid, another charged amino acid, another hydrophilic amino acid, another hydrophobic amino acid, another polar amino acid, another aromatic amino acid or another aliphatic amino acid). In some embodiments, the another (e.g., non-wild type) amino acid is an unconventional amino acid. Unconventional amino acids are non-naturally occurring amino acids. Examples of an unconventional amino acid include, but are not limited to, aminoadipic acid, beta-alanine, beta-aminopropionic acid, aminobutyric acid, piperidinic acid, aminocaprioic acid, aminoheptanoic acid, aminoisobutyric acid, aminopimelic acid, citrulline, diaminobutyric acid, desmosine, diaminopimelic acid, diaminopropionic acid, N-ethylglycine, N-ethylaspargine, hyroxylysine, allo-hydroxylysine, hydroxyproline, isodesmosine, allo-isoleucine, N-methylglycine, sarcosine, N- methylisoieucine, N-methyl valine, non aline, norleucine, orithine, 4-hydroxyproline, g- carboxyglutamate, e-N,N,N-trimethyllysine, e-N-acetyllysine, O-phosphoserine, N- acetylserine, N -form y 1 m eth i on i n e 3-methylhistidine, 5-hydroxylysine, s-N-methylarginine, and other similar amino acids and amino acids (e.g., 4-hydroxyproline). In some
embodiments, one or more amino acid substitutions are introduced into one or more of VP1, VP2 and VP3. In one aspect, a modified capsid protein comprises 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14, or 15 conservative or non-conservative substitutions relative to the wild-type polypeptide. In another aspect, the modified capsid polypeptide of the disclosure comprises modified sequences, wherein such modifications can include both conservative and non conservative substitutions, deletions, and/or additions, and typically include peptides that share at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 87%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% sequence identity to the corresponding wild-type capsid protein.
In some embodiments, the recombinant AAV vector, rep sequences, cap sequences, and helper functions required for producing the rAA V of the disclosure may be delivered to the packaging host cell using any appropriate genetic element (vector). In some embodiments, a single nucleic acid encoding all three capsid proteins (e.g., VP1, VP2 and VP3) is delivered into the packaging host cell in a single vector. In some embodiments, nucleic acids encoding the capsid proteins are delivered into the packaging host cell by two vectors; a first vector comprising a first nucleic acid encoding two capsid proteins (e.g., VP I and VP2) and a second vector comprising a second nucleic acid encoding a single capsid protein (e.g., VPS). In some embodiments, three vectors, each comprising a nucleic acid encoding a different capsid protein, are delivered to the packaging host cell. The selected genetic element may be delivered by any suitable method, including those described herein. The methods used to construct any embodiment of this disclosure are known to those with skill in nucleic acid manipulation and include genetic engineering, recombinant engineering, and synthetic techniques. See, e.g., Sambrook et al, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press, Cold Spring Harbor, N.Y. Similarly, methods of generating rAAV virions are well known and the selection of a suitable method is not a limi tation on the present disclosure. See, e.g., K. Fisher et al, J. Virol., 70:520-532 (1993) and U.S. Pat. No. 5,478,745.
Two polynucleotide or polypeptide sequences are said to be "identical" if the sequence of nucleotides or ammo acids in the two sequences is the same when aligned for maximum correspondence as described below . Comparisons between two sequences are typically performed by comparing the sequences over a comparison window to identify and compare local regions of sequence similarity. A "comparison window" as used herein, refers to a segment of at least about 20 contiguous positions, usually 30 to about 75, or 40 to about 50, in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned.
Optimal alignment of sequences for comparison may be conducted using the MegAlign® program in the Lasergene® suite of bioinformatics software (DNASTAR®, Inc , Madison, WI), using default parameters. This program embodies several alignment schemes described in the following references: Dayhoff, M.Q., 1978, A model of evolutionary change in proteins - Matrices for detecting distant relationships. In Dayhoff, M.O. (ed.) Atlas of Protein Sequence and Structure, National Biomedical Research Foundation, Washington DC Vol. 5, Suppl. 3, pp. 345-358; Hein J., 1990, Unified Approach to Alignment and Phylogenes pp. 626-645 Methods in Enzymology vol. 183, Academic Press, Inc., San Diego, CA; Higgins, D.G. and Sharp, P.M., 1989, CABIOS 5: 151-153; Myers, E.W. and Muller W, 1988, CABIOS 4: 1 1 -17; Robinson, E.D., 1971 , Comb. Theor. 1 1 : 105; Santou, N., Nes, M., 1987, Mol. Biol. Evol. 4:406-425; Sneath, P.H.A. and Sokal, R.R., 1973, Numerical Taxonomy the Principles and Practice of Numerical Taxonomy, Freeman Press, San Francisco, CA; Wilbur, WJ. and Lipman, D.J., 1983, Proc. Natl . Acad Sci USA 80:726-730.
Preferably, with regard to any of the polynucleotides disclosed herein, the "percentage of sequence identity" is determined by comparing two optimally aligned sequences over a window of comparison of at least 20 positions, wherein the portion of the polynucleotide or polypeptide sequence in the comparison window may comprise additions or deletions (i.e., gaps) of 20 percent or less, usually 5 to 15 percent, or 10 to 12 percent, as compared to the reference sequences (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 bases 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 reference sequence (i.e., the window size) and multiplying the results by 100 to yield the percentage of sequence identity. Suitable“moderately stringent conditions” include prewashing in a solution of 5 X SSC, 0.5% SDS, 1 .0 mM EDTA (pH 8.0); hybridizing at 50°C-65°C, 5 X SSC, overnight; followed by washing twice at 65°C for 20 minutes with each of 2X, 0.5X and 0.2X SSC containing 0.1 % SDS. As used herein,
"highly stringent conditions" or "high stringency conditions" are those that: (1) employ low ionic strength and high temperature for washing, for example 0.015 M sodium
chloride/0.0015 M sodium citrate/0.1% sodium dodecyl sulfate at 50°C; (2) employ during hybridization a denaturing agent, such as formamide, for example, 50% (v/v) formamide with 0.1 % bovine serum albumin/0.1 % Ficoil/0.1% polyvinylpyrrolidone/50 mM sodium phosphate buffer at pH 6.5 with 750 mM sodium chloride, 75 mM sodium citrate at 42°C; or (3) employ 50% formamide, 5 x SSC (0.75 M NaCl, 0.075 M sodium citrate), 50 mM sodium phosphate (pH 6.8), 0.1% sodium pyrophosphate, 5 x Denhardt’s solution, sonicated salmon sperm DNA (50 pg/ml), 0.1% SDS, and 10% dextran sulfate at 42°C, with washes at 42°C in 0.2 x SSC (sodium chloride/sodium citrate) and 50% formamide at 55°C, followed by a high- stringency wash consisting of 0.1 x SSC containing EDTA at 55°C. The skilled artisan will recognize how to adjust the temperature, ionic strength, etc. as necessary to accommodate factors such as probe length and the like.
It will be appreciated by those of ordinary skill in the art that, as a result of the degeneracy of the genetic code, there are many nucleotide sequences that encode a polypeptide as described herein. Some of these polynucleotides bear minimal homology to the nucleotide sequence of any native gene. Nonetheless, polynucleotides that vary due to differences in codon usage are specifically contemplated by the present disclosure. Further, alleles of the genes comprising the polynucleotide sequences provided herein are within the scope of the present disclosure. Alleles are endogenous genes that are altered as a result of one or more mutations, such as deletions, additions and/or substitutions of nucleotides. The resulting mRNA and protein may, but need not, have an altered structure or function. Alleles may be identified using standard techniques (such as hybridization, amplification and/or database sequence comparison).
The nucleic acids/polynucleotides of this disclosure can be obtained using chemical synthesis, recombinant methods, or PCR. Methods of chemical polynucleotide synthesis are well known in the art and need not be described in detail herein. One of skill in the art. can use the sequences provided herein and a commercial DNA synthesizer to produce a desired DNA sequence. In some embodiments, the disclosure provides for a polynucleotide that hybridizes under highly stringent conditions with any of the specific nucleotide sequences disclosed herein. One of ordinary' skill in the art will readily understand that appropriate stringency conditions which promote DNA hybridization can be varied. For example, one could perform the hybridization at 6.0 x sodium chloride/sodium citrate (SSC) at about 45 °C, followed by a wash of 2.0 x SSC at 50 °C. For example, the salt concentration in the wash step can be selected from a low stringency of about 2.0 x SSC at 50 °C to a high stringency of about 0.2 x SSC at 50 °C. In addition, the temperature in the wash step can be increased from low stringency conditions at room temperature, about 22 °C, to high stringency conditions at about 65 °C. Both temperature and salt may be varied, or temperature or salt concentration may he held constant while the other variable is changed. In one embodiment, the disclosure provides nucleic acids which hybridize under low stringency conditions of 6 x SSC at room temperature followed by a wash at 2 x SSC at room temperature.
Isolated nucleic acids which differ due to degeneracy in the genetic code are also within the scope of the disclosure. For example, a number of amino acids are designated by more than one triplet. Codons that specify the same amino acid, or synonyms (for example, CAU and CAC are synonyms for histidine) may result in‘silent” mutations which do not affect the amino acid sequence of the protein. One skilled in the art wall appreciate that these variations m one or more nucleotides (up to about 3-5% of the nucleotides) of the nucleic acids encoding a particular protein may exist among members of a given species due to natural allelic variation. Any and all such nucleotide variations and resulting amino acid polymorphisms are within the scope of this disclosure.
Pharmaceutical Compositions
Also provided herein are pharmaceutical compositions comprising any of the antibodies or antigen-binding fragments disclosed herein, and a pharmaceutically acceptable earner. The pharmaceutical compositions may be suitable for any mode of administration described herein; for example, by intravhreai or intravenous administration.
In some embodiments, use of any of the antibodies or antigen-binding fragments disclosed herein for treating retinal diseases, such as LCA, retinitis pigmentosa, and age-related macular degeneration require the localized delivery of the anti-HTRAI antibody or antigen binding fragment to the cells in the retina. In some embodiments, the cells that will be the treatment target in these diseases are either the photoreceptor cells in the retina or the cells of the RPE underlying the neurosensory retina.
In some embodiments, the pharmaceutical compositions comprising any of the antibodies or antigen-binding fragment described herein and a pharmaceutically acceptable carrier are suitable for administration to a human subject. Such carriers are well known in the art (see, e.g., Remington's Pharmaceutical Sciences, 15th Edition, pp. 1035-1038 and 1570-1580). In some embodiments, the pharmaceu tical compositions comprising any of the an tibodies or antigen-binding fragment described herein and a pharmaceutically acceptable carrier is suitable for ocular injection. In some embodiments, the pharmaceutical composition is suitable for intravitreal injection. In some embodiments, the pharmaceutical composition is suitable for subretinal delivery. Such pharmaceutically acceptable carriers can be sterile liquids, such as water and oil, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, and the like. Saline solutions and aqueous dextrose, polyethylene glycol (PEG) and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. The pharmaceutical composition may further comprise additional ingredients, for example preservatives, buffers, tonicity agents, antioxidants and stabilizers, nonionic wetting or clarifying agents, viscosity-increasing agents, and the like. The pharmaceutical compositions described herein can be packaged in single unit dosages or in multidosage forms. The compositions are generally formulated as sterile and substantially isotonic solution.
In one embodiment, any of the antibodies or antigen-binding fragments disclosed herein is formulated into a pharmaceutical composition intended for subretinal or intravitreal injection. Such formulation involves the use of a pharmaceutically and/or physiologically acceptable vehicle or carrier, particularly one suitable for administration to the eye, e.g., by subretinal injection, such as buffered saline or other buffers, e.g., HEPES, to maintain pH at appropriate physiological levels, and, optionally, other medicinal agents, pharmaceutical agents, stabilizing agents, buffers, carriers, adjuvants, diluents, etc. For injection, the carrier will typically be a liquid. Exemplary physiologically acceptable carriers include sterile, pyrogen- free water and sterile, pyrogen-free, phosphate buffered saline. A variety of such known carriers are provided in US Patent Publication No. 7,629,322, incorporated herein by reference. In one embodiment, the carrier is an isotonic sodium chloride solution. In another embodiment, the carrier is balanced salt solution. In one embodiment, the carrier includes tween. If the antibody or antigen-binding fragment is to be stored long-term, it may be frozen in the presence of glycerol or Tween20. In another embodiment, the pharmaceutically acceptable carrier comprises a surfactant, such as perfluorooctane (Perfluoron liquid).
In certain embodiments of the methods described herein, the pharmaceutical composition described above is administered to the subject by subretinal injection. I n other embodiments, the pharmaceutical composition is administered by intravitreal injection. Other forms of administration that may be useful in the methods described herein include, but are not limited to, direct delivery to a desired organ (e.g., the eye), oral, inhalation, intranasal, intratracheal intravenous, intramuscular, subcutaneous, intradermal, and other parental routes of administration. Routes of administration may be combined, if desired.
In some embodiments, any of tire antibodies or antigen-binding fraginents/pharmaceutical compositions disclosed herein are administered to a patient such that they target cells of any one or more layers or regions of the retina or macula. For example, the compositions disclosed herein target cells of any one or more layers of the retina, including the inner limiting membrane, the nerve fiber layer, the ganglion cell layer (GCL), the inner plexiform layer, the inner nuclear layer, the outer plexiform layer, the outer nuclear layer, the external limiting membrane, the layer of rods and cones, or the retinal pigment epi thelium (RPE) In some embodiments, the compositions disclosed herein target glial cells of the GCL, Muller cells, and/or retinal pigment epithelial cells. In particular embodiments, the compositions disclosed herein target vascular endothelial cells. In some embodiments, the compositions disclosed herein targets cells of any one or more regions of the macula including, for example, the umbo, the foveolar, the fo veal avascular zone, the fovea, the parafovea, or the perifovea. In some embodiments, the route of administration does not specifi cally target neurons. In some embodiments, the route of administration is chosen such that it reduces the risk of retinal detachment in the patient (e.g., intravitreal rather than subretinal
administration). In some embodiments, intravitreal administration is chosen if the antibody or antigen-binding fragment/composition is to be administered to an elderly adult (e.g., at least 60 years of age). In particular embodiments, any of the antibodies/antigen-binding fragments/compositions disclosed herein are administered to a subject intravitreally.
Procedures for intravitreal injection are known in the art (see, e.g., Peyman, G.A., et al.
(2009) Retina 29(7):875-912 and Fagan, X.J. and Al-Qureshi, S. (2013) Clin. Experiment. Ophthalmol. 41(5):500-7). Briefly, a subject for intravitreal injection rnay be prepared for the procedure by pupillary dilation, sterilization of the eye, and administration of anesthetic. Any suitable mydriatic agent known in the art may be used for pupillary dilation. Adequate pupillary dilation may be confirmed before treatment. Sterilization may be achieved by applying a sterilizing eye treatment, e.g., an iodide -containing solution such as Povidone- Iodine (BETADINE©). A similar solution may also be used to clean the eyelid, eyelashes, and any other nearby tissues {e.g., skin). Any suitable anesthetic may be used, such as lidocaine or proparacaine, at any suitable concentration. Anesthetic may be administered by any method known in the art, including without limitation topical drops, gels or jellies, and subconjuctival application of anesthetic. Prior to injection, a sterilized eyelid speculum may be used to clear the eyelashes from die area. The site of the injection may be marked with a syringe. The site of the injection may be chosen based on the lens of the patient. For example, the injection site may be 3-3.5 m from the limns in pseudophakic or aphakic patients, and 3.5-4 mm from the limbus in phakic patients. The patient may look in a direction opposite the injection site. During injection, the needle may be inserted perpendicular to the sclera and pointed to the center of the eye. The needle may be inserted such that the tip ends m the vitreous, rather than the subretinal space. Any suitable volume known in the art for injection may be used. After injection, the eye may be treated with a sterilizing agent such as an antiobiotic. The eye may also be rinsed to remove excess sterilizing agent.
Furthermore, in certain embodiments it is desirable to perform non-invasive retinal imaging and functional studies to identify areas of specific ocular ceils to be targeted for therapy. In these embodiments, clinical diagnostic tests are employed to determine the precise location(s) for one or more subretinal injection(s). These tests may include ophthalmoscopy, electroretinography (ERG) (particularly the b-wave measurement), perimetry, topographical mapping of the layers of the retina and measurement of the thickness of its layers by means of confocal scanning laser ophthalmoscopy (cSLO) and optical coherence tomography (OCT), topographical mapping of cone density via adaptive optics (AO), functional eye exam, etc.
The composition may be delivered in a volume of from about 0.1 pL to about 1 mL, including all numbers within the range, depending on the size of the area to be treated, the amount of antibody oar antigen -binding fragment used, the route of administration, and the desired effect of the method. In one embodiment, the volume is about 50 pL. in another embodiment, the volume is about 70 pL. In a preferred embodiment, the volume is about 100 pL. In another embodiment, the volume is about 125 pL. In another embodiment, the volume is about 150 pL. In another embodiment, the volume is about 175 pL. In yet another embodiment, the volume is about 200 pL. In another embodiment, the volume is about 250 pL. In another embodiment, the volume is about 300 pL. In another embodiment, the volume is about 450 pL. In another embodiment, the volume is about 500 pL. In another
embodiment, the volume is about 600 pL. In another embodiment, the volume is about 750 pL. In another embodiment, the volume is about 850 pL, In another embodiment, the volume is about 1000 pL,
Methods of treatment/prophylaxis Described herein are various methods of preventing, treating, arresting progression of or ameliorating the ocular disorders and retinal changes associated therewith. Generally, the methods include administering to a mammalian subject in need thereof, an effective amount of a composition comprising any of the anti-HTRAl antibodies or antigen-binding fragments disclosed herein. Any of the antibodies or antigen-binding fragments described herein, or any of the vectors disclosed herein encoding any of the antibodies or antigen-binding fragments disclosed herein, are useful in the methods described below. in some embodiments, any of the antibodies or antigen-binding fragments disclosed herein, or any of the vectors disclosed herein encoding any of the antibodies or antigen -binding fragments disclosed herein, are for use in treating retinal diseases, such as LCA, retinitis pigmentosa, and age-related macular degeneration may require the localized delivery of the antibody or antigen-binding fragment to the cells in the retina. The cells that will be the treatment target in these diseases are either tire photoreceptor cells in the retina or the cells of the RPE underlying the neurosensory retina. In some embodiments, deli v ering any of the antibodies or antigen-binding fragments disclosed herein to these cells requires injection into the subretinal space between the retina and the RPE. In some embodiments, any of the antibodies or antigen-binding fragments disclosed herein are administered intravitreally or intravenously.
In a certain aspect, the disclosure provides a method of treating a subject having age-related macular degeneration (AMD), comprising the step of administering to the subject any of the antibodies or antigen-binding fragments of the disclosure. In some embodiments, the AMD is any one of Early AMD; Intermediate AMD; Advanced non-neovascular (“Dr”) AMD; or Advanced neovascuiar (“Wet”) AMD. In some embodiments, the disclosure provides for methods of treating a subject with Wet AMD. In some embodiments, the disclosure provides for methods of treating a subject with Dry AMD. In some embodiments, the disclosure provides for methods of treating a subject with polyploidal choroidal vasculopathy (PCV). In some embodiments, the subject has geographic atrophy.
In certain embodiments, the pharmaceutical compositions of the disclosure comprise a pharmaceutically acceptable carrier. In certain embodiments, the pharmaceutical compositions of the disclosure comprise PBS. In certain embodiments, the pharmaceutical compositions of the disclosure comprise piuronic. In certain embodiments, the
pharmaceutical compositions of the disclosure comprise PBS, NaCl and piuronic. In some embodiments, any of the antibodies or antigen-binding fragments disclosed herein is capable of inhibiting proteolytic activity of an HTRA1 protein in a subject in need thereof. In some embodiments, the antibody or antigen-binding fragment thereof is capable of inhibiting HTRA ! proteolytic activity by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the proteolytic activity of a wildtype HTRAl protein m the absence of the antibody or antigen-binding fragment thereof. In some embodiments, the antibody or antigen-binding fragment thereof is capable of inhibiting HTRA 1 proteolytic activity in a cell by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the proteolytic activity of a wildtype HTRAl protein in the same cell type in the absence of the antibody or antigen-binding fragment thereof. In some embodimen ts, the antibody or antigen-binding fragment thereof is capable of inhibiting HTRA 1 proteolytic activity in an eye by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the proteolytic activity of a wildtype HTRAl protein in an eye in the absence of the antibody or antigen-binding fragment thereof.
In some embodiments, the antibody or antigen-binding fragment thereof is capable of inhibiting HTRAl’s ability to cleave any one or more HTRAl substrate is reduced in a subject. In some embodiments, the HTRAl substrate is selected from the group consisting of: fibromodulin, clusterin, ADAM9, eiastin, vitronectin, a2 -macroglobulin, taiin-1, fasein, LTBP-1, EFEMP1, fibulin 5, tau, RseA, and chloride intracellular channel protein. In some embodiments, the antibody or antigen-binding fragment thereof is capable of inhibiting HTRA l’s ability to cleave a regulator of the complement cascade (e.g., vitronectin, fibromodulin or clusterin). In some embodiments, tire antibody or antigen-binding fragment thereof is capable of inhibiting HTRAl such that HTRAl’s ability to cleave an HTRAl substrate and/or regulator of the complement cascade is reduced by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the ability of the HTRAl to cleave the HTRAl substrate and/or regulator of the complement cascade in the absence of the anti body or antigen-binding fragment thereof. In some embodiments, the antibody or antigen -binding fragment thereof is capable of inhibiting HTRAl such that HTRAl’s ability to trimerize is reduced by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the ability of the HTRA1 to trimerize in the absence of the antibody or antigen-binding fragment thereof.
In some embodiments, any of tire antibodies or antigen binding fragments disclosed herein is administered to celi(s) or tissue(s) in a test subject. In some embodiments, the edits) or tissue(s) in the test subject express a higher level of HTRA1 than expressed in the same cell type or tissue type in a reference control subject or population of reference control subjects.
In some embodiments, the reference control subject is of the same age and/or sex as the test subject. In some embodiments, the reference control subject is a healthy subject, e.g., the subject does not have a disease or disorder of the eye. In some embodiments, the reference control subject does not have a disease or disorder of the eye associated with activation of the complement cascade. In some embodiments, the reference control subject does not have macular degeneration. In some embodiments, the eye or a specific cell type of the eye (e.g, ceils in the foveal region) in the test subject express at least 300%, 250%, 200%, 150%,
100%, 95%, 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 10%, 5%, or 1% more HTRA1 as compared to the levels in the reference control subject or population of reference control subjects. In some embodiments, the eye or a specific cell type of the eye (e.g., cells in the foveal region) in the test subject express an HTRA1 gene having any of the mutations disclosed herein. In some embodiments, the eye or a specific cell type of the eye (e.g., cells in the foveal region) in the reference control subject do not express a H'TRA 1 gene having any of the HTRAl mutations disclosed herein. In some embodiments, administration of any of the antibodies or antigen-binding fragments disclosed herein in the ceil(s) or tissue(s) of the test subject results in an decrease in levels of HTRAl protein or functional HTRAl protein. In some embodiments, administration of any of the antibodies or antigen -binding fragments disclosed herein in the cell(s) or tissue(s) of the test subject is capable of decreasing levels of HTRAl protein or functional HTRAl protein such that the decreased levels are within 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 10%, 5%, or 1% of, or are the same as, the levels of HTRAl protein or functional HTRA l protein expressed by the same cell type or tissue type in the reference control subject or population of reference control subjects. In some embodiments, administration of any of the antibodies or antigen-binding fragments disclosed herein in the cell(s) or tissue(s) of the test subject results in a decrease in levels of HTRAl protein or functional HTRAl protein, but the decreased levels of HTRAl protein or functional HTRAl protein are not below the levels of HTRAl protein or functional HTRAl protein expressed by the same cell type or tissue type in the reference control subject or population of reference control subjects. In some embodiments, administration of any of the antibodies or antigen -binding fragments disclosed herein in the cell(s) or tissue(s) of the test subject is capable of decreasing levels of H'TRAl protein or functional HTRA1 protein, but the decreased levels of HTRA l protein or functional HTRA1 protein are below the levels ofHTRAl protein or functional HTRAl protein by no more than 1%, 5%, 10%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or 100% of the levels expressed by the same cell type or tissue type in the reference control subject or population of reference control subjects.
In some embodiments, any of the treatment and/or prophylactic methods disclosed herein are applied to a subject. In some embodiments, the subject is a mammal. In some embodiments, the subject is a human. In some embodiments, the human is an adult. In some embodiments, the human is an elderly adult. In some embodiments, the human is at least 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, or 95 years of age. In particular embodiments, the human is at least 60 or 65 years of age.
In some embodiments, any of the treatment and/or prophylactic methods disclosed herein is for use in treatment of a patient having one or more mutations that causes macular degeneration (AMD) or that increases the likelihood that a patient develops AMD. In some embodiments, the one or more mutations are in the patient’s HTRAl gene.
In some embodiments, any of the treatment and/or prophylactic methods di sclosed herein is for use in treatment of a subject having one or more mutations in the patient’s HTRAl gene. As used herein,“mutations” encompasses polymorphisms that are associated with increased HTRAl expression. In some embodiments, the mutation is associated with AMD or PC V. In some embodiments, the one or more mutations result in overexpression of the HTRAl gene. In some embodiments, HTRAl is expressed at a level at least 25%, 50%, 75%, 100%, 150%, 200%, 250%, 300%, 350%, 400%, 450%, or 500% greater in the subject having the disease or disorder as compared to the level in a control subject not having the disease or disorder. In some embodiments, the control subject is a subject of the same sex and/or of similar age as the subject having the disease or disorder. In some embodiments, the one or more mutations are not in the coding sequence for the HTRAl gene. In some embodiments, the one or more mutations are in 10q26 in a human patient. In some embodiments, the one or more mutations correspond to any one or more of the following human polymorphisms: rs61871744;
rs59616332; rsl 1200630; rs61871745; rsl 1200632; rsl 1200633; rs61871746; rs61871747; rs370974631; rs200227426; rs201396317; rs!99637836; rsl !20Q634; rs75431719; rs 10490924; rs!44224550; rs36212731; rs36212732; rs36212733; rs3750848; rs3750847; rs3750846; rs566108895; rs3793917; rs3763764; rsl 1200638; rsl049331: rs2293870:
rs2284665; rs60401382; rsl 1200643; rs58077526; rs932275 and/or rs2142308.
The retinal diseases described above are associated with various retinal changes. These may include a loss of photoreceptor structure or function; thinning or thickening of the ou ter nuclear layer (ONL); thinning or thickening of the outer plexiform layer (OPL);
disorganization followed by loss of rod and cone outer segments; shortening of the rod and cone inner segments; retraction of bipolar cell dendrites; thinning or thickening of the inner retinal layers including inner nuclear layer, inner plexiform layer, ganglion cell layer and nerve fiber layer; opsin mislocalization; overexpression of neurofilaments; thinning of specific portions of the retina (such as the fovea or macula); loss of ERG function; loss of visual acuity and contrast sensitivity; loss of optokinetic reflexes; loss of the pupillary light reflex; and loss of visually guided behavior. In one embodiment, a method of preventing, arresting progression of or ameliorating any of the retinal changes associated with these retinal diseases is provided. As a result, the subject's vision is improved, or vision loss is arrested and/or ameliorated.
In a particular embodiment, a method of preventing, arresting progression of or ameliorating vision loss associated with an ocular disorder in the subject is provided. Vision loss associated with an ocular disorder refers to any decrease in peripheral vision, central (reading) vision, night vision, day vision, loss of color perception, loss of contrast sensitivity, or reduction in visual acuity.
In another embodiment, a method of targeting one or more type(s) of ocular cells for gene augmentation therapy in a subject in need thereof is provided. In another embodiment, a method of targeting one or more type of ocular cells for gene suppression therapy in a subject in need thereof is provided. In yet another embodiment, a method of targeting one or more type of ocular cells for gene knockdown/augmentation therapy in a subject in need thereof is provided. In another embodiment, a method of targeting one or more type of ocular cells for gene correction therapy in a subject in need thereof is provided. In still another embodiment, a method of targeting one or more type of ocular cells for neurotropic factor gene therapy in a subject in need thereof is provided. In any of the methods described herein, the targeted ceil may be an ocular cell. In one embodiment, the targeted cell is a glial cell. In one embodiment, the targeted cell is an RPE cell. In another embodiment, the targeted cell is a photoreceptor. In another embodiment, the photoreceptor is a cone cell. In another embodiment, the targeted cell is a Muller cell. In another embodiment, tire targeted cell is a bipolar cell. In yet another embodiment, the targeted cell is a horizontal cell. In another embodiment, the targeted cell is an amacrme cell. In still another embodiment, the targeted cell is a ganglion cell. In still another embodiment, the gene may be expressed and delivered to an intracellular organelle, such as a
mitochondrion or a lysosome.
In some embodiments, any of the methods disclosed herein increase photoreceptor function. As used herein "photoreceptor function loss" means a decrease in photoreceptor function as compared to a normal, non-diseased eye or the same eye at an earlier time point. As used herein, "increase photoreceptor function" means to improve the function of the
photoreceptors or increase the number or percentage of functional photoreceptors as compared to a diseased eye (having the same ocular disease), the same eye at an earlier time point, a non-treated portion of the same eye, or the contralateral eye of the same patient. Photoreceptor function may be assessed using the functional studies described above and in the examples below, e.g., ERG or perimetry, winch are conventional in the art.
For each of the described methods, the treatment may be used to pre vent the occurrence of retinal damage or to rescue eyes having mild or advanced disease. As used herein, the term "rescue" means to prevent progression of the disease to total blindness, prevent spread of damage to uninjured ocular cells, improve damage in injured ocular cells, or to provide enhanced vision. In one embodiment, the composition is administered before the disease becomes symptomatic or prior to photoreceptor loss. By symptomatic is meant onset of any of the various retinal changes described above or vision loss. In another embodiment, the composition is administered after disease becomes symptomatic. In yet another embodiment, the composition is administered after initiation of photoreceptor loss. In another embodiment, the composition is administered after outer nuclear layer (ONE) degeneration begins. In some embodiments, it is desirable that the composition is administered while bipolar cell circuitry to ganglion cells and optic nerve remains intact.
In another embodiment, the composition is administered after initiation of photoreceptor loss. In yet another embodiment, the composition is administered when less than 90% of the photoreceptors are functioning or remaining, as compared to a non-diseased eye. In another embodiment, the composition is administered when less than 80% of the photoreceptors are functioning or remaining. In another embodiment, the composition is administered when less than 70% of the photoreceptors are functioning or remaining. In another embodiment, the composition is administered when less than 60% of the photoreceptors are functioning or remaining. In another embodiment, the composition is administered when less than 50% of the photoreceptors are functioning or remaining. In another embodiment, the composition is administered when less than 40% of the photoreceptors are functioning or remaining. In another embodiment, the composition is administered when less than 30% of the
photoreceptors are functioning or remaining. In another embodiment, the composition is administered when less than 20% of the photoreceptors are functioning or remaining. In another embodiment, tire composition is administered when less than 10% of the
photoreceptors are functioning or remaining. In one embodiment, the composition is administered only to one or more regions of the eye. in another embodiment, the composition is administered to the entire eye.
In another embodiment, the method includes performing functional and imaging studies to determine the efficacy of the treatment. These studies include ERG and in vivo retinal imaging, as described in the examples below. In addition visual field studies, perimetry and microperimetry, pupillometry, mobility testing, visual acuity, contrast sensitivity, color vision testing may be performed.
In yet another embodiment, any of the above described methods is perfonned in combination with another, or secondary, therapy. The therapy may be any now known, or as yet unknown, therapy which helps prevent, arrest or ameliorate any of the described retinal changes and/or vision loss. In one embodiment, the secondary therapy is encapsulated cell therapy (such as that delivering Ciliary Neurotrophic Factor (CNTF)). See, Sieving, P.A. et al, 2006. Proc Natl Acad Sci USA, 103 (10): 3896-3901 , which is hereby incorporated by reference. In another embodiment, the secondary therapy is a neurotrophic factor therapy (such as pigment epithelium-derived factor, PEDF; ciliary' neurotrophic factor 3; rod-derived cone viability factor (RdCVF) or glial -derived neurotrophic factor). In another embodiment, the secondary therapy is anti-apoptosis therapy (such as that deli vering X-iinked inhibitor of apoptosis,
XIAP). In yet another embodiment, the secondary therapy is rod derived cone viability factor 2. The secondary therapy can be administered before, concurrent with, or after administration of the antibodies/antigen-binding fragments described above.
In some embodiments, any of tire antibodies, antigen-binding fragments or compositions disclosed herein is administered to a subject in combination with another therapeutic agent or therapeutic procedure. In some embodiments, the additional therapeutic agent is an anti- VEGF therapeutic agent (e.g., such as an anti-VEGF antibody or fragment thereof such as ranibizumab, bevacizumab or afhbercept), a vitamin or mineral ( e.g ., vitamin C, vitamin E, lutein, zeaxanthin, zinc or copper), omega-3 fatty acids, and/or Visudyne™. In some embodiments, the other therapeutic procedure is a diet having reduced omega-6 fatty acids, laser surgery, laser photocoagulation, submacular surgery, retinal translocation, and/or photodynamic therapy.
Kits
In some embodiments, any of the antibodies or antigen-binding fragments thereof disclosed herein is assembled into a pharmaceutical or diagnostic or research kit to facilitate their use in therapeutic, diagnostic or research applications. A kit may include one or more containers housing any of the antibodies or antigen-binding fragments thereof disclosed herein and instructions for use.
The kit may be designed to facilitate use of the methods described herein by researchers and can take many forms. Each of the compositions of the kit, where applicable, may he provided in liquid fonn (e.g., in solution), or in solid form, (e.g., a dry' powder). In certain cases, some of the compositions may be constitutable or otherwise processable (e.g., to an active form), for example, by the addition of a suitable solvent or other species (for example, water or a cell culture medium), which may or may not be provided with the kit. As used herein, “instructions” can define a component of instruction and/or promotion, and typically involve written instructions on or associated with packaging of the disclosure. Instructions also can include any oral or electronic instructions provided in any manner such that a user will clearly recognize that the instructions are to be associated with the kit, for example, audiovisual (e.g., videotape, DVD, etc.), Internet, and/or web-based communications, etc.
The written instructions may be in a form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which instructions can also reflects approval by the agency of manufacture, use or sale for animal administration. EXAMPLES
The disclosure now being generally described, it will be more readily understood by reference to the following examples, which are included merely for purposes of illustration of certain embodiments and embodiments of the present disclosure, and are not intended to limit the disclosure.
Example 1: Phage Panning for anti-HTRA 1 Binders
Bead-based panning was employed with biotinylated HTRA captured on streptavidin beads. Four rounds of phage panning was performed with varying stringencies A total of 4 conditions (varying antigen concentration and washes) were tested. The different antigens tested were 0.5 mg of biotinylated HTRA1; biotinylated L2 peptide 201 (19-mer); and biotinylated L3 peptide 202 (19-mer). The panning rounds were performed as outlined below:
Figure imgf000063_0001
Phage ELISA on rescued panning outputs showed increased enrichment. A total of 14 plates from rounds 3 and rounds 4 outputs were screened for HTRA 1 binding. Binding was determined by detecting scFv in PPE expression with anti-v5 mAb. Three-hundred and eighty-seven clones were initially identified as ELISA binders and re-arrayed for scFv sequencing. Ninety-one unique sequences and ELISA re-confirmed clones w'ere identified. PPEs of these clones were tested for competition binding with two different control anti- HTRA 1 IgGs (BM1 and BM2) in ELISA format to determine epitope binders. Two control benchmark“BM” anti-HTRA 1 antibodies were used as comparators in these experiments. A streptavidin plate was coated with 1 ug/rnl HTRA1 by passive adsorption. Twenty -five ul scFv PPE and 25 ul 0.08 ug/mi BM IgG in 4% BSA/PBS was added to wells m duplicate plates. Plate 1 was treated w ith HRP anti-human Fc (detects BM IgG), and plate 2 was treated with anti-v5 mAb and HRP anti-mouse IgG (detects scFV) If scFv binds the same epitope as BM IgG, ELISA signal of scFv will be lower than BM IgG. If scFv binds different epitopes than BM IgG, ELISA signal of scFv is not affected by BM IgG. Forty-five clones appear to bind independently of BMl and BM2.
A total of 12 plates from rounds 3 and 4 outputs were screened for Pep2Ql binding. Binding was determined by detecting scFv in PPE expression with anti~v5 mAb. Forty-two clones were initially identified as ELISA binders and re-arrayed for scFv sequencing. Fifteen sequences were unique and ELISA re-confirmed clones were identified. PPEs of these clones were tested for HTRA1 binding via ELISA but none of these clones bound. A total of 12 plates from rounds 3 and 4 outputs were screened for Pep202 binding. Binding was determined by detecting scFv in PPE expression with anti-v5 mAb. Eighty-eight clones were initially identified as ELISA binders and re -arrayed for scFv sequencing. Seventeen sequences were unique and ELIS A re-confirmed clones were identified. PPEs of these clones were tested for HTRA1 binding via ELISA but none of these clones bound.
Binding experiments were performed on Octet HTX at 25°C. Biotimy!ated-HTRAl antigen (0.5 ug/mL) was loaded onto Streptavidin, and loaded sensors were dipped into tested antibodies (50 iiM). Kinetic constants were calculated using a monovalent (1 : 1 ) model. Hie clones showing strong binding were as follows:
Figure imgf000064_0001
Figure imgf000065_0001
Binders were tested in epitope binning experiments. Briefly, binning experiments were performed on Octet HTX at 25°C In tandem, the binning assay format was set up.
Biotinylated antigen bt-HTRAl (10 nM) was loaded onto Streptavidin sensors for 240 seconds. Loaded sensors were dipped into saturating antibody (20 ug/mL) for 900 seconds followed by competing antibody (5 ug/mL for 300 seconds).
Epitope binning experiments were completed by evaluating the degree of binding competition in a full pairwise comparison. In brief, the first antibody was bound to HTRA1 at a saturating concentration. Subsequently the Octet sensor tips were then exposed to a solution of the potential competitor binder and the kinetics and level of binding compared to assess whether the two antibodies bound to a similar epitope. Based on these data six bins were defined along with an additional group where the epitope bin was uncertain. Binders were tested in an AlphaLISA assay format, using AlphaScreen signal inhibition. Briefly, an N-terminal Flag RseA-C-termmal His substrate associates with a donor bead (anti-Flag) and an acceptor bead (Nickel chelator) . When the beads are associated with an uncleaved RseA substrate, they are in close enough proximity to release a signal. HTRA1 is capable of cleaving RseA and reducing the output signal. The optimized assay conditions were as follows. The buffer used included 150m M NaHrPOr pH 8.3, 380mM NaCi, 0.05% Triton X-100, 0.3% BSA Five uL of diluted HTR.41 (400 nM final) and 5 ul of buffer/antibody (400 nM final) were added and incubated for 30 minutes at room temperature. Five ul of diluted RseA (25 nM final) was added and then incubated for 1 or 4 hours at room temperature. Fifteen ul of AlphaLISA Nickel Chelate Acceptor Beads (10 ug/mL final) and 15 uL anti-FLAG Donor beads (20 ug/mL final) were added and incubated for 1 hour at room temperature. The samples were measured with 1 in Spire 2390. The assay indicated that 10 potential inhibitors (purified IgGs) were comparable or beter than the BM antibodies in the assay. A summar ' of the results from the various assays testing the different binders is provided in Figure 1. The CDR sequences for each of the binders listed in Figure 1 are provided in Figure 2.
Example 2: Treatment of Patients with AMD with an Anti-HTRAl Antibody
This study will evaluate the efficacy of the antibodies or antigen-binding fragments of
Example 1 for treating patients with AMD. Patients with AMD will be treated with any of the antibodies or antigen-binding fragments, or a control. The antibodies or antigen-binding fragments will be administered by intravitreal, subretinal or intravenous injection. Patients will be monitored for improvements in AMD symptoms.
It is expected that the antibody or antigen-binding fragment treatments will improve the A AID symptoms.
Example 3: Functional Test inn of anti-HTRAl anti bodies
Recombinant HTRA1 was generated containing a Strepil affinity tag and with a deletion of the HTRA1 N-terminal Mac domain (StrepIl-AMac-HTRAl). Antibody specificity for HTRA! w'as confirmed using western blots. Antibodies SR8047-38, SR8047-40, SR8047-17, SR8047-44, SR8047-16, SR8047-43, SR8047-31, SR8047-06, SR8047-23, and SR8047-42 were ail determined to bind to HTRA1. SR8Q47-38 and SR8047-40 also bound to HTRA4. None of the antibodies showed any detectable binding to HTR A2 or HTRA3.
A. Eiastin Assay
The HTRA1 -mediated proteolysis of eiastin was measured in the presence and absence of anti-HTRAl antibodies in order to determine whether these antibodies inhibit HTRA1 serine protease activity. HTRA1 -mediated proteolysis of eiastin was measured utilizing the Sensolyte® Green Elastase Kit (Anaspec, Inc.). Reaction solutions were prepared and earned out in opaque 96-well pates. Each solution w'as prepared by mixing StrepII-AMac-HTRAl (200 iiM) with of 1.2 mM anti-HTRAl antibody in assay buffer (pH 8.0), followed by subsequent incubation at 37°C for 45 minutes. Following incubation, eiastin labeled with 5-FAM and QXL®520 quencher was added to the reaction solution. HTRA1 -mediated proteolysis of eiastin generated an increase in the fluorescence of the 5-FAM fluorophore with an intensity proportional to the proteolytic activity of SixepIT-AMac -HTRAl The change in fluorescence was measured at 37°C with excitation and emission wavelengths of 490 ran and 520 ran, respectively. Fluorescence intensity' was measured every 60 seconds and plotted as a function of time (Figure 3 A, Figure 3B). Measurements were performed at least in duplicate for each sample. Anti-HTRAl antibodies SR8047-44 and SR8047-43 generated a 70% and 50% (respectively) reduction in HTRA1 -mediated proteolysis of elastin relative to the fully active positive controls (Figure 3C), suggesting that these particular antibodies are inhibitors of HTRA1 proteolytic activity.
B. Fibulin 5 Assay
The HTRA1 -mediated proteolysis of Fibulin 5 was measured in the presence and absence of anti- HTRA1 antibodies in order to test whether such antibodies would exert an inhibitory effect. Reaction solutions containing StrepTI-AMac-HTRAl (100 nM) and an anti-HTRAl antibody (600 nM) were prepared in a buffer containing 25 niM HEPES pH 7.5, and 75 niM NaCl, and allowed to incubate at 37°C for 45 minutes at 300 rpm. The proteolysis reaction was initiated upon addition of Fibulin 5 (1 mM) at 37°C and 300 rpm. Reaction time-points were taken at 0, 1, 3, and 22 hours by quenching a reaction sample with SDS-PAGE gel loading buffer and DTT ( 1 M). Reaction progress was visualized for each time-point using SDS-PAGE (Figure 4A and Figure 4B). HTRA1 -mediated proteolysis of Fibulin 5 generated a decrease in Fibulin 5 band intensities over time. The Fibulin 5 band intensities for the reactions containing antibodies SR8047-17, SR8047-44, SR8Q47-43, and SR8047-42 remained constant over time relative to the control reactions (BM1 and BM2; Figure 4B), suggesting that the these antibodies are capable of inhibiting HTRA1 -mediated proteolysis of fibulin 5.
C. Tau Digest Assay
The antibodies were further tested for their capacity' to inhibit HTRA1 -mediated proteolysis of Tau using substantially the same SDS-PAGE method as described in the Fibulin 5 assay. Reaction solutions containing StreplT-AMac-HTRAl (100 nM) and an anti-HTRAl antibody (600 nM) were prepared in a buffer containing 50 niM Tris pH 8 0, and allowed to incubate at 37°C for 45 minutes. The reaction was initiated upon addition of Tau (800 nM) at 37°C and 300 rpm. Reaction time-points were taken at 0, 7, and 22 hours by quenching a reaction sample with SDS-PAGE gel loading buffer and DTT (1 M). Reaction progress was visualized for each time-point using SDS-PAGE (Figure 5A and Figure 5B). A decrease in Tau band intensities over time represented HTRAl· -mediated proteolysis. HTRAi proteolysis of Tau was not inhibited by SR8047-23, resulting in a large decrease in Tau band intensity after 7 and 22 hours (Figure 5A). Withholding the controls, the Tau band intensities for all other reactions did not decrease, indicating that all antibodies except SR8047-23 inhibited HTRAl -mediated proteolysis of Tau (Figure 5 A and Figure 5B). An above average degree of inhibition was observed for the reactions containing antibodies SR8047-17, SR8047-44, SR8047-43, and SR8047-42 remained constant over time relative to the control reactions (BMi and BM2; Figure 5B) These results suggest that the SR8Q47-17, SR8047-44, SR8047-43, and SR8047-42 antibodies are more effective inhibitors of HTR A 1 -mediated Tan proteolysis.
D. RseA Assay
The antibodies were further screened for their ability to inhibit HTRA 1 -mediated proteolysis of RseA. The SDS-PAGE based screen described in the Fibulin 5 and Tau digest assay s was utilized, and reaction solutions containing StrepII-AMac-HTRAi (100 nM) and an anti-HTRAl antibody (600 nM) were prepared in a buffer containing 150 mM NaTfrPCh (Sodium Phosphate) pH 8.0, and 380 mM Nad. These reaction solutions were allowed to incubate at 37°C for 45 minutes. Following incubation, the reactions were initiated upon addition of RseA (1 mM) at 37°C and 300 rpm. Reaction time-points were taken at 0, 5, and 7 hours by quenching a reaction sample with SDS-PAGE gel loading buffer and DTT ( 1 M). Reaction progress was visualized for each time-point using SDS-PAGE (Figure 6 A and Figure 6B). Similar to the Tau digest assay, RseA band intensities for all reactions, except the BM1 and BM2 controls, decreased over time (Figure 6A). A higher degree of inhibition was observed with the SR8047-44, SR8047-43, SR8047-17, and SR8047-42 antibodies, suggesting that these particular antibodies may be more effective inhibitors of HTRA1 -mediated proteolysis of RseA.
E. 0-Casein digest Assay
The antibodies were further screened for their ability to inhibit HTRA 1 -mediated proteolysis of b-casein using a similar SDS-PAGE screen as utilized above. Reaction solutions containing Strepil-AMae- HTRA1 (100 nM) and an anti-HTRAl antibody (600 nM) were prepared in a buffer containing 50 mM Tris pH 8.0, and 150 mM NaCl. These reaction solutions were allowed to incubate at 37°C for 15 minutes. Following incubation, the reactions were initiated upon addition of b-casein (2 mM) at 37°C and 300 rpm. Reaction time-points were taken at 0, 5, and 30 minutes by quenching a reaction sample with SDS-PAGE gel loading buffer and DTT (1 M). Reaction progress was visualized for each time- point using SDS-PAGE (Figure 7 A and Figure 7B). Aside from the control reactions (BM1 and BM2; Figure 7B), no decreases in [3-casein band intensities were observed for any reaction, suggesting that none of the antibodies were capable of effectively inhibiting HTRA 1 -mediated proteolysis of b-caseia
F. Summary of HTRA1 Anti body and Fragment Screens
The pooled assay data (Figure 8) reveals that SR8047-44 and SR8047-43 were capable of inhibiting HTRA1 in all screens except the b-casein digest assay. These data suggest that the SR8047-44 and SR8047-43 antibodies are two of the most effective inhibitors of HTRA! proteolytic activity. To determine whether the critical fragments of these antibodies would function effectively in isolation, the Fab regions of each antibody were isolated and screened against the elastin assay (Figure 9A). The SR8047-43 and SR8047-44 anti-HTRAl Fab fragments generated nearly equivalent reductions in proteolysis as their anti-HTRAl IgG counterpasts (Figure 9A). The Vm?.x for SR8047-43 and SR8047- 44 Fab fragments and TgGs is approximately equivalent to the Vmax for HTRA 1 in the absence of the antibodies (Figure 9B) Taken together, the data generated in these screening assays highlight the ability for SR8047-43 and SR8047-44, whether as full-length antibodies or as antibody fragments, effectively inhibit the protease activity of HTRA! .
I NCORPORATION BY REFERENCE
All publications and patents mentioned herein are hereby incorporated by reference in their entirety as if each individual publication or patent was specifically and indi vidually indicated to be incorporated by reference.
While specific embodiments of the subject matter have been discussed, the above specification is illustrative and not restrictive. Many variations will become apparent to those skilled in the art upon review of this specification and the claims below. The full scope of the disclosure should be determined by reference to the claims, along with their full scope of equivalents, and the specification, along with such variations.
SEQUENCE LISTING
SEQ ID NO: 1— SR8047-06 HCDR1 Amino Acid Sequence LSFSNYAMS
SEQ ID NO: 2— SR8047-06 HCDR2 Amino Acid Sequence
SGISGSGSNTYYA
SEQ ID NO: 3— SR8047-06 HCDR3 Ammo Acid Sequence CARQPGITM VRGGHYGMDVW
SEQ ID NO: 4— SR8047-06 LCDR1 Ammo Acid Sequence
QASQDISTYLN SEQ ID NO: 5— SR8047-06 LCDR2 Am o Acid Sequence EASALQS
SEQ ID NO: 6— SR8047-06 LCDR3 Amino Acid Sequence
CQQSYTTPLTF
SEQ ID NO: 7— SR8047-08 HCDRi Amino Acid Sequence
YMFTAYYIH
SEQ ID NO: 8— SR8047-08 HCDR2 Ammo Acid Sequence GWMNPN SGDTGYA
SEQ ID NO: 9— SR8047-08 HCDR3 Amino Acid Sequence
CVNPYNWNDRDYW SEQ ID NO: 10— SR8Q47-08 LCDR1 Amino Acid Sequence
RASQA1YSYLA
SEQ ID NO: 1 i— SR8047-08 LCDR2 Amino Acid Sequence DASSLES SEQ ID NO: 12— SR8047-08 LCDR3 Amino Acid Sequence
CLQHNTYPYTF SEQ ID NO: 13— SR8047-12 HCDR1 Ammo Acid Sequence YSFTSQYMH
SEQ ID NO: 14— SR8047-12 HCDR2 Amino Acid Sequence GGIIPIFGTANYA
SEQ ID NO: 15— SR8047-12 HCDR3 Ammo Acid Sequence C AREEY S S S LGD YYYYYMD VW
SEQ ID NO: 16— SR8047-12 LCDR1 Amino Acid Sequence QASQDISNYLN
SEQ ID NO: 17— SR8047-12 LCDR2 Amino Acid Sequence
AASRLQG SEQ ID NO: 18— SR8047-12 LCDR3 Amino Acid Sequence CQQATSFPFTF
SEQ ID NO: 19— SR8Q47-14 HCDR1 Amino Acid Sequence
YPFIGQYLH
SEQ ID NO: 20— SR8047-14 HCDR2 Ammo Acid Sequence GWMNPNSGNTGYA
SEQ ID NO: 21— SR8047-14 HCDR3 Amino Acid Sequence CASWAGVDDYW
SEQ ID NO: 22— SR8047-14 LCDR1 Amino Acid Sequence
RASQGISNYLA SEQ ID NO: 23— SR8Q47-14 LCDR2 Amino Acid Sequence
DASNLET
SEQ ID NO: 24— SR8047-I4 LCDR3 Amino Acid Sequence CLQYNTYPFTF
SEQ ID NO: 25— SR8047-16 HCDR1 Amino Acid Sequence
YTFTRQYMH SEQ ID NO: 26— SR8047-16 HCDR2 Amino Acid Sequence GWINPN SGNTGY A
SEQ ID NO: 27— SR8047-16 HCDR3 Amino Acid Sequence
CARAVLGYFDYW
SEQ ID NO: 28— SR8Q47-16 LCDR1 Amino Acid Sequence
RASQSISRWLA
SEQ ID NO: 29— SR8047-16 LCDR2 Ammo Acid Sequence GASNLET
SEQ ID NO: 30— SR8047-16 LCDR3 Amino Acid Sequence CQQSYSTPLTF SEQ ID NO: 31— SR8047-17 HCDR! Amino Acid Sequence
FTFSDQYMS
SEQ ID NO: 32— SR8047-17 HCDR2 Amino Acid Sequence AVI SYDGSNKYY A
SEQ ID NO: 33— SR8047-17 HCDR3 Ammo Acid Sequence
CTTDEFDDGDYEDYW
SEQ ID NO: 34— SR8047-17 LCDR1 Ammo Acid Sequence RASQDIRSYLA
SEQ ID NO: 35— SR8047-17 LCDR2 Amino Acid Sequence
DASNLEP
SEQ ID NO: 36— SR8047-17 LCDR3 Ammo Acid Sequence
CLQHNTYPFTF
SEQ ID NO: 37— SR8Q47-18 HCDR1 Ammo Acid Sequence FTFTSSAMQ
SEQ ID NO: 38— SR8047-18 HCDR2 Ammo Acid Sequence GGFDPEDGEIIYA SEQ ID NO: 39— SR8047-18 HCDR3 Amino Acid Sequence CALDGYNFW
SEQ ID NO: 40— SR8047-18 LCDR1 Amino Acid Sequence
RASQDIRNYLA
SEQ ID NO: 41— SR8047-18 LCDR2 Ammo Acid Sequence QASSLES
SEQ ID NO: 42— SR8047-18 LCDR3 Amino Acid Sequence CLQHN S YPFTF
SEQ ID NO: 43— SR8047-20 HCDR1 Amino Acid Sequence
FIFYDYAMQ SEQ ID NO: 44— SR8Q47-2G HCDR2 Ammo Acid Sequence
SSISPSSSY1YYA
SEQ ID NO: 45— SR8047-20 HCDR3 Ammo Acid Sequence
CTRGGYDY GDHW SEQ ID NO: 46— SR8047-20 LCDRI Amino Acid Sequence
RASQGISGYLA SEQ ID NO: 47— SR8047-20 LCDR2 Ammo Acid Sequence DASSLET
SEQ ID NO: 48— SR8047-2Q LCDR3 Amino Acid Sequence
CLQHDSYPFTF
SEQ ID NO: 49— SR8Q47-21 HCDR1 Ammo Acid Sequence
GTFRRYLNN
SEQ ID NO: 50— SR8047-21 HCDR2 Amino Acid Sequence GIINPSGGSSSYA
SEQ ID NO: 51— SR8047-21 HCDR3 Ammo Acid Sequence
CARDGGTEGYW SEQ ID NO: 52— SR8047-21 LCDRI Ammo Acid Sequence RASQGISSYLA
SEQ ID NO: 53— SR8Q47-21 LCDR2 Amino Acid Sequence
DGSTLET
SEQ ID NO: 54— SR8047-21 LCDR3 Amino Acid Sequence
CLQHSTYPHTF
SEQ ID NO: 55— SR8047-22 HCDR1 Amino Acid Sequence GTFTTYYMQ
SEQ ID NO: 56— SR8047-22 HCDR2 Amino Acid Sequence GWINPNSGGTNYA SEQ ID NO: 57— SR8047-22 HCDR3 Amino Acid Sequence
CARGQYSSSWYYMDVW
SEQ ID NO: 58— SR8047-22 LCDR1 Amino Acid Sequence RASQSISSYLN
SEQ ID NO: 59— SR8047-22 LCDR2 Amino Acid Sequence AASSLQS SEQ ID NO: 60— SR8047-22 LCDR3 Amino Acid Sequence
CQQSYMTPLTF
SEQ ID NO: 61— SR8047-23 HCDR1 Amino Acid Sequence
YTLTTWYMQ
SEQ ID NO: 62— SR8Q47-23 HCDR2 Amino Acid Sequence
GWMNPN SGDTGYA
SEQ ID NO: 63— SR8047-23 HCDR3 Amino Acid Sequence CARAWNLDHSGYDERDYYYGMDVW
SEQ ID NO: 64— SR8047-23 LCDR1 Amino Acid Sequence
QASQDISNYLN SEQ ID NO: 65— SR8047-23 LCDR2 Amino Acid Sequence
AASSLHS
SEQ ID NO: 66— SR8047-23 LCDR3 Amino Acid Sequence
CQQSYDTPFTF
SEQ ID NO: 67— SR8047-25 HCDR1 Ammo Acid Sequence FTP SNYWMH
SEQ ID NO: 68— SR8047-25 HCDR2 Amino Acid Sequence SALSG SGGSTYY A
SEQ ID NO: 69— SR8047-25 HCDR3 Amino Acid Sequence
CARGGLDYDGVYYY GMDVW
SEQ ID NO: 70— SR8047-25 LCDR1 Ammo Acid Sequence
RSSQSLLHSNGYNYLD
SEQ ID NO: 71— SR8Q47-25 LCDR2 Amino Acid Sequence AASSLQS
SEQ ID NO: 72— SR8047-25 LCDR3 Ammo Acid Sequence
CMQALQ1PLTF SEQ ID NO: 73— SR8047-28 HCDR1 Amino Acid Sequence YTFTNYYMH
SEQ ID NO: 74— SR8047-28 HCDR2 Amino Acid Sequence
CGIIPIFGTANYA
SEQ ID NO: 75— SR8047-28 HCDR3 Amino Acid Sequence
CARPS SGWSNYDYW
SEQ ID NO: 76— SR8047-28 LCDRI Amino Acid Sequence RASQDISSYLV
SEQ ID NO: 77— SR8047-28 LCDR2 Ammo Acid Sequence DASNLET SEQ ID NO: 78— SR8Q47-28 LCDR3 Amino Acid Sequence
CLQHNS YPFTF
SEQ ID NO: 79— SR8047-31 HCDRI Ammo Acid Sequence
YPFIGQYLH SEQ ID NO: 80— SR8047-31 HCDR2 Ammo Acid Sequence GWMNPN SGNTGYA SEQ ID NO: 81— SR8047-31 HCDR3 Ammo Acid Sequence CARGRY GSGSYSYYFDYW
SEQ ID NO: 82— SR8047-3 i LCDR1 Amino Acid Sequence
RASQSISTYLN
SEQ ID NO: 83— SR8047-31 LCDR2 Amino Acid Sequence GASSLQS
SEQ ID NO: 84— SR8047-31 LCDR3 Ammo Acid Sequence CQQGYSTPLTF
SEQ ID NO: 85— SR8047-36 HCDR1 Ammo Acid Sequence
FTFSNSDMN SEQ ID NO: 86— SR8047-36 HCDR2 Amino Acid Sequence
SYISSSSRYIYYA
SEQ ID NO: 87— SR8Q47-36 HCDR3 Ammo Acid Sequence
CAKDGP A V VTPID Y W
SEQ ID NO: 88— SR8047-36 LCDR1 Amino Acid Sequence
KSSQSVLSSSNNKNYLA
SEQ ID NO: 89— SR8047-36 LCDR2 Amino Acid Sequence WASTRES
SEQ ID NO: 90— SR8047-36 LCDR3 Amino Acid Sequence
CHQYYSTPYTF SEQ ID NO: 91— SR8Q47-38 HCDR1 Ammo Acid Sequence
YTFTDYYVH
SEQ ID NO: 92— SR8047-38 HCDR2 Ammo Acid Sequence GWMNPN SGNTGY A
SEQ ID NO: 93— SR8047-38 HCDR3 Amino Acid Sequence
CAKGVWDFSYYLDYW SEQ ID NO: 94— SR8047-38 LCDR1 Amino Acid Sequence
RASQNIGYYLN
SEQ ID NO: 95— SR8047-38 LCDR2 Ammo Acid Sequence
SASTLQR
SEQ ID NO: 96— SR8Q47-38 LCDR3 Amino Acid Sequence CQQSYSTPLTF
SEQ ID NO: 97— SR8047-40 HCDR1 Ammo Acid Sequence YTFTNYGFN
SEQ ID NO: 98— SR8047-4Q HCDR2 Amino Acid Sequence
GLINL SDGNTMY A SEQ ID NO: 99— SR8047-40 HCDR3 Amino Acid Sequence CVK AGY S SGWY A G YF QHW
SEQ ID NO: 100— SR8047-40 LCDR1 Amino Acid Sequence
RSSQSLLHSNGYNYLD
SEQ ID NO: 101— SR8047-40 LCDR2 Ammo Acid Sequence
LGSNRAS
SEQ ID NO: 102— SR8G47-40 LCDR3 Amino Acid Sequence CMQALQTPLTF
SEQ ID NO: 103— SR8047-42 HCDR1 Amino Acid Sequence GTFSSYAIS SEQ ID NO: 104— SR8G47-42 HCDR2 Amino Acid Sequence GVVMN PN SGATG YA
SEQ ID NO: 105— SR8047-42 HCDR3 Amino Acid Sequence
CASGDGWFDYW
SEQ ID NO: 106— SR8047-42 LCDR1 Amino Acid Sequence
RASQYIGSWLA
SEQ ID NO: 107— SR8047-42 LCDR2 Amino Acid Sequence AASSLQS
SEQ ID NO: 108— SR8047-42 LCDR3 Amino Acid Sequence
CQQYYNSPITF SEQ ID NO: 109— SR8G47-43 HCDR1 Amino Acid Sequence
YTFTSQYMH
SEQ ID NO: 110— SR8047-43 HCDR2 Ammo Acid Sequence
GGIIPIFPTPDYA
SEQ ID NO: 111— SR8G47-43 HCDR3 Ammo Acid Sequence CARES S S SGPDGAF D1W
SEQ ID NO: 1 12— SR8047-43 LCDR1 Amino Acid Sequence RAS Q SINHWLA
SEQ ID NO: 113— SR8G47-43 LCDR2 Amino Acid Sequence AASSLQS SEQ ID NO: 114— SR8047-43 LCDR3 Ammo Acid Sequence
CQQSYSIPLTF
SEQ ID NO: 115— SR8047-44 HCDR1 Amino Acid Sequence
YTFTSYYMH
SEQ ID NO: 116— SR8047-44 HCDR2 Amino Acid Sequence
GIINPSGGSTSYA SEQ ID NO: 117— SR8047-44 HCDR3 Amino Acid Sequence
CARDGVDYYMDVW
SEQ ID NO: 118— SR8047-44 LCDR1 Ammo Acid Sequence
QASQDISNYLN
SEQ ID NO: 119— SR8047-44 LCDR2 Ammo Acid Sequence
DASNLET
SEQ ID NO: 120— SR8G47-44 LCDR3 Amino Acid Sequence
CQQSYSTPLTF
SEQ ID NO: 121— Human HTRA 1 Amino Acid Sequence- GenBank Accession No.
NP 002766.1
MQIPRAALLPLLLLLLAAPASAQLSRAGRSAPLAAGCPDRCEPARCPPQPEHCEGGR ARDACGCCEVCGAPEGAACGLQEGPCGEGLQCWPFGVPASATVRRRAQAGLCVC ASSEPVCGSDANTYANLCQLRAASRRSERLHRPPVTVLQRGACGQGQEDPNSLRHKY NFIADVVEKIAPAWHIELFRKLPFSKREVPVASGSGF1VSEDGLIVTNAHVVTNKHR VKVELKNGATYEAKIKDVDEKADIALIKIDHQGKLPVLLLGRSSELRPGEFVVAIGSP FSLQNTVTTGIVSTTQRGGKELGLRNSDMDYIQTDAIINYGNSGGPLVNLDGEVTGTN TLKVTAGISFAIPSDKIKKFLTESHDRQAKGKAITKKKYIGIRMMSLTSSKAKELKDR HRDFPDVISGAYUEVIPDTPAEAGGLKENDVIISINGQSVVSANDVSDVIKRESTLNM VVRRGNEDIMITVIPEEIDP
SEQ ID NO: 122— FLAG DYKDDDDK
SEQ ID NO: 123— polyhistidine
HHHHHH
SEQ ID NO: 124— hemagglutinin
YPYDVPDYA
Figure imgf000081_0001
Figure imgf000082_0001
Figure imgf000082_0002
Figure imgf000083_0001

Claims

We claim:
1. An antibody or antigen-binding fragment thereof that binds to a HTRA 1 protein, or a functional fragment thereof.
2. The antibody or antigen-binding fragment of claim 1, wherein the HTRAI protein is a human HTRAI protein, or functional fragment thereof.
3. The antibody or antigen-binding fragment thereof of claim 1 or 2, wherein the HTRAI protein comprises an amino acid sequence that is at least 80%, 85%, 90%, 92%,
94%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 121 or a functional fragment thereof.
4. The antibody' or antigen-binding fragment thereof of any one of claims 1-3, wherein the antibody or antigen-binding fragment thereof is capable of inhibiting any one or more HTRAI function.
5. The antibody or antigen-binding fragment thereof of any' one of claim s 1-4, wherein the antibody or antigen -binding fragment thereof is capable of inhibiting HTRA 1 proteolytic activity' by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the proteolytic activity of a wildtype HTRAI protein in the absence of the HTRAI antibody or antigen-binding fragment thereof. 6. The antibody or antigen-binding fragment thereof of any one of claim s 1-5, wherein the antibody or antigen -binding fragment thereof is capable of inhibiting HTRA 1 proteolytic activity in a cell by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the proteolytic activity of a wildtype HTRAI protein in the same cell type in the absence of the HTRA 1 antibody or antigen-binding fragment thereof.
7. The antibody or antigen-binding fragment thereof of any one of claim s 1-6, wherein the antibody or antigen -binding fragment thereof is capable of inhibiting HTRA 1 proteolytic activity' in any eye by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the proteolytic activity of a wildtype HTRA1 protein in another eye in the absence of the HTRA1 antibody or antigen-binding fragment thereof. 8. The antibody or antigen-binding fragment thereof of any one of claims 1-6, wherein the antibody or antigen-binding fragment is capable of reducing HTRAl’s ability to cleave any one or more HTRAI substrate.
9. Hie antibody or antigen-binding fragment thereof of claim 8, wherein the HTRAI substrate is selected from the group consisting of: fibromodulin, clast in. clusterin,
ADAM9, vitronectin, a2 -macroglobulin, talin-1, fascin, LTBP-1, EFEMP1, fibulin 5, tau, RseA, and chloride intracellular channel protein.
10. The antibody or antigen-binding fragment thereof of any one of claims 1-9, wherein the antibody or antigen-binding fragment is capable of inhibiting HTRA I’ s ability to cleave a regulator of the complement cascade (e.g., vitronectin, fibromodulin or clusterin).
1 1. The antibody or antigen-binding fragment thereof of any one of claim s 1-9, wherein the antibody or antigen -binding fragment is capable of inhibiting HTRAI such that HTRAI’ s ability to cleave an HTRAI substrate and/or regulator of the complement cascade is reduced by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the ability of the HTRA 1 to cleave the HTRAI substrate and/or regulator of the complement cascade in the absence of the anti- HTRA1 antibodies or antigen-binding fragments thereof.
12. The antibody or antigen-binding fragment of any one of claims 1-11, wherein the antibody or antigen-binding fragment is capable of inhibiting the ability of the HTRAI protein to trimerize. 13. The antibody or antigen-binding fragment of claim 12, wherein the antibody or antigen-binding fragment is capable of inhibiting the ability of the HTRAI protein to trimerize by at least 5%, 10%, 15%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%,
70%, 75%, 80%, 85%, 90%, 95% or 100% as compared to the ability of the HTRAI protein to trimerize in the absence of the antibody or antigen-binding fragment.
14. The antibody or antigen-binding fragment of any one of claims 1-13, wherein the antibody or antigen-binding fragment thereof comprises any one of, or combination of, the CDR sequences indicated in Figure 2
15. The antibody or antigen-binding fragment of any one of claims 1-14, wherein the antibody or antigen-binding fragment thereof compri ses any one of, or any combination of, the CDR amino acid sequences of SEQ ID NOs: 1 -120 16. The antibody or antigen-binding fragment of any one of claims 1-14, wherein the antibody or antigen -binding fragment thereof comprises a set of CDRS as forth as any one of tire following sets of SEQ ID NOs: 1 -6, 7-12, 13-18, 19-24, 25-30, 31-36, 37-42, 43- 48, 49-54, 55-60, 61-66, 67-72, 73-78, 79-84, 85-90, 91-96, 97-102, 103-108, 109-114, or 115-120.
17. Hie antibody or antigen -binding fragment of any one of claims 1-14, wherein the antibody or antigen-binding fragment thereof comprises the CDRs set forth as SEQ ID NOs: 1-6. 1 . The antibody or antigen-binding fragment of any one of claims 1-14, wherein the antibody or antigen-binding fragment thereof compri ses the CDRs set forth as SEQ ID NOs: 7-12.
19. The antibody or antigen-binding fragment of any one of claims 1-14, wherein the antibody or antigen-binding fragment thereof comprises the CDRs set forth as SEQ ID
NOs: 19-24.
20. The antibody or antigen-binding fragment of any one of claim s 1-14, wherein the antibody or antigen-binding fragment thereof comprises the CDRs set forth as SEQ ID NOs: 31-36.
21. The antibody or antigen-binding fragment of any one of claims 1-14, wherein the antibody or antigen -binding fragment thereof comprises the CDRs set forth as SEQ ID NOs: 37-42.
22. The antibody or antigen-binding fragment of any one of claims 1-14, wherein the antibody or antigen-binding fragment thereof comprises the CDRs set forth as SEQ ID NOs: 61 -66.
23. The antibody or antigen-binding fragment of any one of claims 1-14, wherein the antibody or antigen-binding fragment thereof compri ses the CDRs set forth as SEQ ID NOs: 73-78. 24. The antibody or antigen-binding fragment of any one of claims 1-14, wherein the antibody or antigen -binding fragment thereof comprises the CDRs set forth as SEQ ID NOs: 103-108.
25. The antibody or antigen-binding fragment of any one of claims 1-14, wherein the antibody or antigen-binding fragment thereof comprises the CDRs set forth as SEQ ID NOs: 109-114.
26. The antibody or antigen-binding fragment of any one of claims 1-14, wherein the antibody or antigen-binding fragment thereof comprises the CDRs se t forth as SEQ ID NOs: 115-120.
27. The antibody or antigen-binding fragment of any one of claims 1-13, wherein the antibody or antigen-binding fragment thereof comprises any one of or combination of the amino acid sequences of SEQ ID NO: 1-120, but wherein the amino acid sequences comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 , or 12 amino acid substitutions.
28. The antibody or antigen-binding fragment of any one of claims 1-13, wherein the antibody or antigen-binding fragment thereof comprises a set of CDRS as forth as any one of the following sets of SEQ ID NOs: 1-6, 7-12, 13-18, 19-24, 25-30, 31-36, 37-42, 43- 48, 49-54, 55-60, 61 -66, 67-72, 73-78, 79-84, 85-90, 91 -96, 97-102, 103-108, 109-114, or
115-120 but wherein the amino acid sequences comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10,
11, or 12 amino acid substitutions.
29. The antibody or antigen-binding fragment of claim 27 or 28, wherein the substitutions are conservative substitutions.
30. The antibody or antigen-binding fragment of any one of claims 27-29, wherein the substitutions either increase the binding affinity of the antibody or antigen-binding fragment to the HTRA1 epitope, or the substitutions result in a reduction of binding affinity of no more than 5%, 10%, 20%, 30%, 40%, or 50% as compared to the antibody or antigen- binding fragment lacking the substitutions. 31. The antibody or antigen-binding fragment of any one of claims 1-30, wherein the antibody or antigen -binding fragment is a full-length antibody.
32. The antibody or antigen-binding fragment of claim 31, wherein the antibody or antigen-binding fragment is a monoclonal antibody.
33. Hie antibody or antigen-binding fragment of claim 31 or 32, wherein the antibody or antigen-binding fragment is a humanized antibody.
34. The antibody or antigen-binding fragment of any one of claims 1-30, wherein tire antibody or antigen-binding fragment is an antigen-binding fragment thereof.
35. Tire antibody or antigen-binding fragment of claim 34, wherein the antigen binding fragment is an scFv . 36. The antibody or antigen-binding fragment of claim 34, wherein the antigen binding fragment is selected from the group consisting of: a Fab fragment, a F(ab')2 fragment, a Fab' fragment, dAb fragment and/or a dsFv.
37. Tire antibody or antigen-binding fragment of any one of claims 1 -36, wherein the antibody or antigen-binding fragment binds to one or more of the following HTRA1 domains: an insulin-like growth factor binding domain, a kazai domain, a trypsin-like peptidase domain, and/or a PDZ domain.
38. The antibody or antigen -binding fragment of any one of claims 1-37, wherein the antibody or antigen-binding fragment is capable of binding to a non-human species HTRAl .
39. The antibody or antigen-binding fragment of any one of claims 1-38, wherein the non-human species HTRA1 is mouse, rat, rabbit, cow, monkey (e.g., cynomolgus monkey), or ape (e.g. chimpanzee) HTRAl protein.
40. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises 1, 2 or 3 of the CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to any one of SEQ ID NOs: 125-144.
41. The antibody or antigen-binding fragment of any one of claims 1-14 and SO SO, wherein the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 125.
42. The antibody or antigen -binding fragment of any one of claims 1-14 and SO SO, wherein the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 126.
43. The antibody or antigen-binding fragment of any one of claims 1 -14 and 30- 39, wherein the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 127.
44. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 128.
45. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen -binding fragment comprises 1 , 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 129.
46. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises 1 , 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 130.
47. The antibody or antigen-binding fragment of any one of claims 1-14 and SO SO, wherein the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 131
48. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain ammo acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 132
49. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 133.
50. The antibody or antigen-binding fragment of any one of claims 1-14 and SO SO, wherein the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 134.
51. The antibody or antigen -binding fragment of any one of claims 1-14 and SO SO, wherein the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 135.
52. Tire antibody or antigen-binding fragment of any one of claims 1 -14 and 30- 39, wherein the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 136.
53. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises 1 , 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 137.
54. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 138.
55. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain ammo acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 139
56. The antibody or antigen-binding fragment of any one of claims 1-14 and SO SO, wherein the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 140.
57. The antibody or antigen-binding fragment of any one of claims 1-14 and SO SO, wherein the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 141.
58. The antibody or antigen -binding fragment of any one of claims 1-14 and SO SO, wherein the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 142.
59. Tire antibody or antigen-binding fragment of any one of claims 1 -14 and 30- 39, wherein the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 143.
60. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises 1 , 2 or 3 CDRs from a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 144.
61. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises 1, 2 or 3 of the CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to any one of SEQ ID NOs: 145-164.
62. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain ammo acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 145.
63. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 146.
64. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 147.
65. Hie antibody or antigen -binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 148.
66. Tire antibody or antigen-binding fragment of any one of claims 1 -14 and 30- 39, wherein the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 149.
67. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises 1 , 2 or 3 CDRs from a variable light chain ammo acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 150.
68. The antibody or antigen-binding fragment of any one of claims 1-14 and SO SO, wherem the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 151.
69. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain ammo acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 152.
70. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 153.
71. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 154.
72. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 155.
73. Tire antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 156.
74. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises 1 , 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 157.
75. The antibody or antigen-binding fragment of any one of claims 1-14 and SO SO, wherem the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 158.
76. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain ammo acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 159.
77. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 160.
78. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 161.
79. Hie antibody or antigen -binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 162.
80. Tire antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 163.
81. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises 1 , 2 or 3 CDRs from a variable light chain ammo acid sequence that is at least 75%, 80%, 85%, 90%, 95%, or 100% identical to SEQ ID NO: 164.
82. The antibody or antigen-binding fragment of any one of claims 1-14 and SO S , wherem the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from the sequence of SEQ ID NO: 143. 83. The antibody or antigen-binding fragment of any one of claims 1-14 and SO
SO, wherein the antibody or antigen-binding fragment comprises 3 CDRs from the sequence of SEQ ID NO: 143.
84. The antibody or antigen -binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from the sequence of SEQ ID NO: 144.
85. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherem the antibody or antigen-binding fragment comprises 3 CDRs from the sequence of SEQ ID NO: 144.
86. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherem the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from the sequence of SEQ ID NO: 163.
87. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises 3 CDRs from the sequence of SEQ ID NO: 163. 88. Tire antibody or antigen-binding fragment of any one of claims 1-14 and 30-
39, wherein the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from the sequence of SEQ ID NO: 164.
89. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises 3 CDRs from the sequence of SEQ ID NO: 164.
90. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen -binding fragment comprises 1 , 2 or 3 CDRs from the sequence of SEQ ID NO: 143 and 1, 2 or 3 CDRs from the sequence of SEQ ID NO: 163.
91. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises 3 CDRs from the sequence of SEQ ID NO: 143 and 3 CDRs from the sequence of SEQ ID NO: 163. 92. The antibody or antigen-binding fragment of any one of claims 1-14 and 30-
39, wherein the antibody or antigen-binding fragment comprises 1, 2 or 3 CDRs from the sequence of SEQ ID NO: 144 and comprises 1, 2 or 3 CDRs from the sequence of SEQ ID NO: 164.
93. The antibody or antigen-binding fragment of any one of claims 1-14 and SO 39, wherein the antibody or antigen-binding fragment comprises 3 CDRs from the sequence of SEQ ID NO: 144 and comprises 3 CDRs from the sequence of SEQ ID NO: 164.
94. The antibody or antigen-binding fragment of any one of claims 40-93, wherein the CDRs are determined in accordance with Chothia.
95. The antibody or antigen-binding fragment of any one of claims 40-93, wherein the CDRs are determined in accordance with MacCallum.
96. The antibody or antigen-binding fragment of any one of claims 40-93, wherein the CDRs are determined accordance with Rabat.
97. The antibody or antigen-binding fragment of any one of claims 40-93, wherein the CDRs are determined in accordance with AbM. 98. The antibody or antigen-binding fragment of any one of claims 40-93, wherein the CDRs are determined in accordance with TMGT.
99. The antibody or antigen -binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to any of SEQ ID NOs: 125-144.
100. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 125.
101. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen -binding fragment comprises an ammo acid sequence that is at least 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 126.
102. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 127.
103. The antibody or antigen-binding fragment of any one of claims 1-14 and SO SO, wherem the antibody or antigen-binding fragment comprises an ammo acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 128
104. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 129
105. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 130
106. The antibody or antigen -binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 131 .
107. The antibody or antigen-binding fragment of any one of claims 1 -14 and 30- 39, wherein the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 132.
108. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen -binding fragment comprises an ammo acid sequence that is at least 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 133.
109. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 134.
1 10. The antibody or antigen-binding fragment of any one of claims 1-14 and SO SO, wherem the antibody or antigen-binding fragment comprises an ammo acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 135.
1 1 1. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 136
1 12. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 137
113. The antibody or antigen -binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 138.
114. The antibody or antigen-binding fragment of any one of claims 1 -14 and 30- 39, wherein the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 139.
115. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen -binding fragment comprises an ammo acid sequence that is at least 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 140.
116. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 141.
1 17. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherem the antibody or antigen-binding fragment comprises an ammo acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 142
1 18. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 143
1 19. The antibody or antigen-binding fragment of any one of claims 1-14 and SO SO, wherein the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 144
120. The antibody or antigen -binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises a variable light chain am o acid sequence that is at least 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to any of SEQ ID NOs: 145-164.
121 . The antibody or antigen-binding fragment of any one of claims 1 -14 and 30- 39, wherein the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 145.
122. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen -binding fragment comprises an ammo acid sequence that is at least 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 146.
123. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 147.
124. The antibody or antigen-binding fragment of any one of claims 1-14 and SO SO, wherem the antibody or antigen-binding fragment comprises an ammo acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 148
125. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 149
126. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 150
127. The antibody or antigen -binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 151 .
128. The antibody or antigen-binding fragment of any one of claims 1 -14 and 30- 39, wherein the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 152.
129. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen -binding fragment comprises an ammo acid sequence that is at least 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 153.
130. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 154.
131. The antibody or antigen-binding fragment of any one of claims 1-14 and SO SO, wherem the antibody or antigen-binding fragment comprises an ammo acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 155
132. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 156
133. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 157
134. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 158.
135. The antibody or antigen-binding fragment of any one of claims 1 -14 and 30- 39, wherein the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 159.
136. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen -binding fragment comprises an ammo acid sequence that is at least 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 160.
137. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 161.
138. The antibody or antigen-binding fragment of any one of claims 1-14 and SO SO, wherem the antibody or antigen-binding fragment comprises an ammo acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 162
139. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 163
140. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises an amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 164
141. The antibody or antigen -binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 143, and further comprises a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 163.
142. The antibody or antigen-binding fragment of any one of claims 1-14 and 30- 39, wherein the antibody or antigen-binding fragment comprises a variable heavy chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 144, and further comprises a variable light chain amino acid sequence that is at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO: 164
143. A polynucleotide encoding the antibody or antigen-binding fragment of any one of the antibodies or antigen-binding fragments of claims 1-142.
144. A vector comprising the polynucleotide of claim 143.
145. A host cell comprising the vector of claim 144 and capable of expressing the polynucleotide.
146. A host cell expressing one or more polynucleotides encoding the antibody or antigen-binding fragments of claims 1-142.
147. A method of treating a disease or disorder in a subject in need thereof, wherein the disease or disorder is associated with aberrantly expressed HTRA1, wherein the method comprises administering to the subject the antibody or antigen-binding fragment of any one of claims 1-142.
148. A method of treating a disease or disorder in a subject in need thereof, wherein HTRA1 is expressed at a level at least 5%, 10%, 25%, 50%, 75%, 100%, 150%, 200%,
250%, 300%, 350%, 400%, 450%, or 500% greater in the subject having the disease or disorder as compared to the level in a control subject not having the disease or disorder. wherein the method comprises administering to the subject the antibody or antigen-binding fragment of any one of claims 1-142.
149. A method of treating age-related macular degeneration or polyploidal choroidal vasculopathy in a subject in need thereof, wherein the disease or disorder is associated with aberrantly expressed HTRA1, wherein the method comprises administering to the subject the antibody or antigen-binding fragment of any one of claims 1 -142.
150. The method of claim 148, wherein the control subject is a subject of the same sex and/or of similar age as the subject having the disease or disorder.
151. The method of any one of claims 147-150, wherein the subject has one or more mutations in the HTRA1 gene. 152. The method of claim 151, wherein the one or more mutations are not in the coding sequence for the HTRA1 gene.
153. The method of claim 151 , wherein the one or more mutations are in 10q26 in a human subject.
154. The method of any one of claims 151-153, wherein the one or more mutations correspond to any one or more of the following polymorphisms in a human subject:
rs61871744; rs59616332; rsl 1200630; rs61871745; rsl 1200632; rsl 1200633; rs61871746; rs61871747; rs370974631; rs200227426; rs2013963 T7; rsl99637836; rsl 1200634;
rs75431719; rsl0490924; rsl44224550; rs3621273 i ; rs36212732; rs36212733; rs3750848; rs3750847; rs3750846; rs566108895; rs3793917; rs3763764; rsl 1200638; rsl049331;
rs2293870; rs2284665; rs60401382; rsl 1200643; rs58077526; rs932275 and/or rs2142308.
155. Tire method of any one of claims 147-154, wherein the subject has age-related macular degeneration.
156. The method of any one of claims 147-155, wherein the subject is a human.
157. The method of claim 156, wherein the human is at least 40 years of age.
158. The method of claim 156, wherein the human is at least 50 years of age.
159. The method of claim 157, wherein the human is at least 65 years of age
160. The method of any one of claims 147-159, wherein the antibody or antigen binding fragment is administered locally.
161. The method of any one of claims 147-159, wherein the antibody or antigen- binding fragment is administered intravitreally.
162. The method of any one of claims 147-159, wherein the antibody or antigen binding fragment is administered subretinaiiy. 163. The method of any one of claims 147-159, wherein the antibody or antigen binding fragment is administered systemicaJly.
164. The method of any one of claims 147-163, wherein the subject has polypi oidal choroidal vasculopathy.
165. The method of any one of claims 147-164, wherein the subject has Wet AMD.
166. Hie method of any one of claims 147-165, wherein the subject has Dry AMD. 167. A composition comprising the antibody or antigen-binding fragment of any one of claims 1 -142 and a pharmaceutically acceptable carrier.
168. The composition of claim 165, wherein the composition is substantially pyrogen free.
PCT/US2019/061740 2018-11-16 2019-11-15 Antibody-based therapeutics for targeting htra1 and methods of use WO2020102680A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201862768557P 2018-11-16 2018-11-16
US62/768,557 2018-11-16

Publications (1)

Publication Number Publication Date
WO2020102680A1 true WO2020102680A1 (en) 2020-05-22

Family

ID=70730907

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2019/061740 WO2020102680A1 (en) 2018-11-16 2019-11-15 Antibody-based therapeutics for targeting htra1 and methods of use

Country Status (2)

Country Link
TW (1) TW202039585A (en)
WO (1) WO2020102680A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008094370A2 (en) * 2006-12-22 2008-08-07 University Of Utah Research Foundation Method of detecting ocular diseases and pathologic conditions and treatment of same
WO2013055998A1 (en) * 2011-10-14 2013-04-18 Genentech, Inc. ANTI-HtrA1 ANTIBODIES AND METHODS OF USE
WO2017075212A1 (en) * 2015-10-30 2017-05-04 Genentech, Inc. Anti-htra1 antibodies and methods of use thereof

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008094370A2 (en) * 2006-12-22 2008-08-07 University Of Utah Research Foundation Method of detecting ocular diseases and pathologic conditions and treatment of same
WO2013055998A1 (en) * 2011-10-14 2013-04-18 Genentech, Inc. ANTI-HtrA1 ANTIBODIES AND METHODS OF USE
WO2017075212A1 (en) * 2015-10-30 2017-05-04 Genentech, Inc. Anti-htra1 antibodies and methods of use thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
LIN, MICHAEL K. ET AL.: "HTRA1, an age-related macular degeneration protease, processes extracellular matrix proteins EFEMP1 and TSP1", AGING CELL, vol. 17, no. 4, 5 May 2018 (2018-05-05), pages 1 - 9, XP055707708, DOI: 10.1111/acel.12710 *
LU, ZHIGANG ET AL.: "HTRA1 synergizes with oxidized phospholipids in promoting inflammation and macrophage infiltration essential for ocular VEGF expression", PLOS ONE, vol. 14, no. 5, 2019, pages e0216808, XP055707711, DOI: 10.1371/journal.pone.0216808 *

Also Published As

Publication number Publication date
TW202039585A (en) 2020-11-01

Similar Documents

Publication Publication Date Title
ES2881642T3 (en) Anti-pro-myostatin / latent myostatin antibodies and uses thereof
JP7028923B2 (en) Anti-transthyretin antibody
US20160297854A1 (en) Compositions and methods for long acting molecules
US20180265586A1 (en) Anti-sortilin antibodies and methods of use thereof
ES2940311T3 (en) Anti-sortilin antibodies and methods of use thereof
ES2925716T3 (en) Anti-transthyretin antibodies
TWI718121B (en) Anti-transthyretin antibodies
JP7017013B2 (en) Anti-transthyretin antibody
JP2019501132A (en) Anti-complement factor C1q Fab fragment and use thereof
JP7016470B2 (en) Anti-transthyretin antibody
US20170290876A1 (en) Compositions and methods for long acting proteins
JP7076711B2 (en) Anti-transthyretin antibody
EP3013860B1 (en) Cb1 receptor antigen-binding proteins and uses thereof
US20200392229A1 (en) Methods of use of anti-sortilin antibodies
JP2021508498A (en) Anti-TMEM106B antibody and how to use it
WO2013012855A1 (en) Apelin antigen-binding proteins and uses thereof
KR20200131335A (en) Adeno-associated virus (AAV) delivery of anti-FAM19A5 antibodies
JP2020510416A (en) Multispecific binding molecules with specificity for dystroglycan and laminin-2
AU2017230091A1 (en) Activin type 2 receptor binding proteins and uses thereof
US20240101681A1 (en) Methods of use of anti-sortilin antibodies
WO2020102680A1 (en) Antibody-based therapeutics for targeting htra1 and methods of use
US20240132597A1 (en) Methods of use of anti-sortilin antibodies
WO2023108102A1 (en) Anti-lcn-2 antibodies as a treatment for eye disorders
WO2022261648A2 (en) Methods of use of anti-sortilin antibodies
EA043015B1 (en) ANTIBODIES AGAINST SORTILIN AND METHODS OF THEIR APPLICATION

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: 19884062

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 19884062

Country of ref document: EP

Kind code of ref document: A1