Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
PEPTIDES AND METHODS OF USE THEREOF IN TREATING OCULAR DISORDERS
Document Type and Number:
WIPO Patent Application WO/2023/150791
Kind Code:
A1
Abstract:
The present disclosure includes methods of treating an ocular disease or condition at the surface of the eye, including ocular diseases that involve the cornea. More specifically the present disclosure relates to treating an ocular disease or condition such as dry eye syndrome by administering a peptide fragment of the MARCKS protein. Peptide fragments and variants thereof as disclosed in the present disclosure are useful in such methods.

Inventors:
SHAH AMIT (US)
LUMSDEN GRAHAM (US)
Application Number:
PCT/US2023/062129
Publication Date:
August 10, 2023
Filing Date:
February 07, 2023
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
BIOMARCK PHARMACEUTICALS LTD (US)
SHAH AMIT (US)
LUMSDEN GRAHAM (US)
International Classes:
A61K38/08; A61P27/02; A61K38/10; A61K38/16; C07K7/00
Domestic Patent References:
WO2020257162A12020-12-24
Foreign References:
US20150274778A12015-10-01
US20080020031A12008-01-24
US20090220581A12009-09-03
Attorney, Agent or Firm:
MCLEOD, Bonnie Weiss et al. (US)
Download PDF:
Claims:
CLAIMS

1 . A method of treating dry eye syndrome in a subject comprising: administering to said subject a therapeutically effective amount of a composition comprising at least one peptide having an amino acid sequence consisting of from 4 to 24 contiguous amino acids of a reference sequence, GAQFSKTAAKGEAAAERPGEAAVA (SEQ ID NO. 1).

2. The method of claim 1, wherein said peptide had an amino acid sequence consisting of four contiguous amino acid residues of SEQ ID NO: 1.

3. The method of claim 1, wherein said peptide comprises an amino acid sequence of SEQ ID NO: 106 or 219.

4. The method of any one of claims 1-3, wherein said peptide comprises an amino acid sequence of SEQ ID NO: 219.

5. The method of any one of claims 1-4, wherein said peptide consists of an amino acid sequence of SEQ ID NO: 219.

6. The method of any of claims 1-5, wherein the peptide is myristoylated or acetylated at the N-terminal amino acid.

7. The method of any one of claims 1-6, wherein the peptide is acetylated at the N-terminal amino acid, and consists of an amino acid sequence of SEQ ID NO: 219.

8. The method according to any one of claims 1-7, wherein the composition comprises a pharmaceutically acceptable carrier.

9. The method according to claim 1, wherein said subject is a mammal.

10. The method according to claim 9, wherein said mammal is selected from the group consisting of humans, canines, equines and felines.

11. The method according to any one of claims 1-10, wherein the composition is administered by topical administration, intravitreal injection (IVT), subconjuctival injection, subtenon injection (SBT), retrobulbar injection, periocular injection, subretinal injection, intrascleral, transscleral, intrastromal, intravenous injection, intra-ocular administration, or any combination thereof.

12. The method according to any one of claims 1-11, wherein the composition is administered by topical administration to the eye.

13. The method of claim 11 or 12, wherein the topical administration comprises one, two, three, four, five, or six topical administrations daily.

14. The method according to claim 1, wherein the composition comprises a topical formulation, intra-ocular formulation, an eye implant, an eye drop, eye gel, ointment, microspheres, microemulsion, liposomal formulation or any combination thereof.

15. The method according to claim 1, further comprising administration to said subject a second molecule, wherein the second molecule is an antibiotic, an antiviral compound, an antiparasitic compound, an anti-inflammatory compound, an immunomodulatory compound, or any combination thereof.

16. The method of any one of claims 1-15, wherein the composition is administered at a concentration from about 1 pM to about 1 mM.

17. The method of any one of claims 1-16, wherein the composition is administered in an amount of about 100 pM.

18. The method of any one of claims 1-17, wherein the composition is administered in a volume of about 0.01 mL to about 1 mL.

Description:
PEPTIDES AND METHODS OF USE THEREOF IN TREATING OCULAR

DISORDERS

CROSS REFERENCE TO RELATED APPLICATIONS

[0001] This application claims priority from U.S. Provisional Application No. 63/307,364 filed February 7, 2022, which is incorporated by reference herein in its entirety for all purposes.

TECHNICAL FIELD

[0002] The present disclosure relates to peptides or peptide compositions or peptide formulations and methods of their use to treat ocular disorders such as dry eye syndrome. The present disclosure also relates to topical formulations of such peptides.

REFERENCE TO AN ELECTRONIC SEQUENCE LISTING

[0003] The contents of the electronic sequence listing (BMRK_008_01WO_SeqList_ST26.xml; Size: 418,796 bytes; and Date of Creation: February 3, 2023) are herein incorporated by reference in its entirety.

BACKGROUND

[0004] Dry eye syndrome (DES), also known as dry eye disease, keratoconjunctivitis sicca (KCS), keratitis sicca, and others, is a disease of the ocular surface. Dry eye syndrome is associated with tear film instability, dysfunction, and/or loss of homeostasis. Dry eye syndrome is a common condition and can be caused by underlying diseases and disorders including autoimmune disorders, or environmental causes. Currently available treatment options for dry eye syndrome are not adequate to treat the disease effectively and/or require repeated administration many times per day. There is a need in the art for improved methods for treating ocular diseases at the surface of the eye such as dry eye syndrome.

SUMMARY

[0005] The disclosure provides a method of treating dry eye syndrome in a subject comprising, administering to said subject a therapeutically effective amount of a composition comprising at least one peptide having an amino acid sequence selected from the group consisting of: (a) an amino acid sequence having from 4 to 24 contiguous amino acids of a reference sequence, GAQFSKTAAKGEAAAERPGEAAVA (SEQ ID NO. 1); (b) an amino acid sequence having the sequence, GAQFSKTAAKGEAAAERPGEAAVA (SEQ ID NO. 1); and (c) an amino acid sequence with at least about 75% identity to the amino acid sequence defined in (a) or (b).

[0006] In some aspects, the peptide comprises at least nine contiguous amino acid residues of SEQ ID NO: 1. In some aspects, the peptide comprises an amino acid sequence of SEQ ID NO: 106. In some aspects, the peptide comprises an amino acid sequence of SEQ ID NO: 219. In some aspects, the peptide is myristoylated or acetylated at the N-terminal amino acid. In some aspects, the peptide is acetylated at the N-terminal amino acid, and consists of an amino acid sequence of SEQ ID NO: 106 or 219. In some aspects, the peptide is acetylated at the N- terminal amino acid, and consists of an amino acid sequence of SEQ ID NO: 219. In some aspects, the peptide consists of four contiguous amino acid residues of SEQ ID NO: 1.

[0007] In some aspects, the composition comprises a pharmaceutically acceptable carrier. In some aspects, the subject is a mammal. In some aspects, the mammal is selected from the group consisting of humans, canines, equines and felines.

[0008] In some aspects, the administering is done by topical administration. In some aspects, the present disclosure provides topical formulations comprising one or more of the peptides disclosed herein. In some aspects, the composition comprises a topical formulation, intra-ocular formulation, an eye implant, an eye drop, eye gel, ointment, microspheres, microemulsion, liposomal formulation or any combination thereof. In some embodiments, the composition is administered by daily topical administrations. In some embodiments, each daily topical administration comprises one, two, three, four, or five topical administrations on each day, for example, approximately one administration every 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 hours. In some embodiments, on each day that the composition is administered topically, the composition is administered once, twice, or three times.

[0009] In some aspects, the method further comprises administration to the subject a second molecule, wherein the second molecule is an antibiotic, an antiviral compound, an antiparasitic compound, an anti-inflammatory compound, an immunomodulatory compound, or any combination thereof. BRIEF DESCRIPTION OF THE DRAWINGS

[00010] FIG. 1 is a line graph showing corneal opacity scores at baseline, 14 days pre-dose, and at days 1, 3, 7, 14, 21, and 24 after treatment, in each of the following groups: sham control (untreated), PBS control, BIO11006 treated (“MI 006”), and BIO91201 treated (“MI 201”).

[00011] FIG. 2 is a line graph showing corneal fluorescein retention scores at baseline, 14 days pre-dose, and at days 1, 3, 7, 14, 21, and 24 after treatment, in each of the following groups: sham control (untreated), PBS control, BIO11006 treated (“MI 006”), and BIO91201 treated (“MI 201”).

[00012] FIG. 3 is a line graph showing tear production (mm/20s) at baseline, 14 days pre- dose, and at days 7, 14, 21, and 24 after treatment, in each of the following groups: sham control (untreated), PBS control, BIO11006 treated (“MI 006”), and BIO91201 treated (“MI 201”).

DETAILED DESCRIPTION

DEFINITIONS

[00013] It is to be understood that the terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting.

[00014] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood to one of ordinary skill in the art to which the present application belongs. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present application, representative methods and materials are herein described.

[00015] Following long-standing patent law convention, the terms "a", "an", and "the" refer to "one or more" when used in this application, including the claims. Thus, for example, reference to "a carrier" includes mixtures of one or more carriers, two or more carriers, and the like and reference to “the method” includes reference to equivalent steps and/or methods known to those skilled in the art, and so forth.

[00016] Unless otherwise indicated, all numbers expressing quantities of ingredients, reaction conditions, and so forth used in the specification and claims are to be understood as being modified in all instances by the term "about". Accordingly, unless indicated to the contrary, the numerical parameters set forth in the present specification and attached claims are approximations that can vary depending upon the desired properties sought to be obtained by the present application. Generally the term "about", as used herein in references to a measurable value such as an amount of weight, time, dose, etc. is meant to encompass values within an acceptable degree of variability in the art. In some embodiments, degree of variability is based on FDA guidelines.

[00017] As used herein, “treating” or “treatment” and the like is an approach for obtaining beneficial or desired clinical results. For purposes of this disclosure, beneficial or desired clinical results include, but are not limited to, one or more of the following: alleviation of one or more symptoms of an ocular condition at the surface of the eye such as dry eye syndrome. Symptoms of such an ocular condition include, but are not limited to, pain or discomfort in the eye, dryness in the eye, itchiness in the eye, a burning, stinging, or irritating feeling in the eye or a feeling that a foreign object is in the eye, and sensitivity to light. Accordingly, the terms “treating” or “treatment” and the like include lessening the severity of such symptoms in the eye, including reducing the incidence of, managing, ameliorating, preventing, and/or delaying the development or progression of such symptoms in the eye. Treating or treatment herein can also include improving vision or preventing, stopping, or slowing the progression of vision loss.

[00018] The term “effective amount” or “therapeutically effective amount” refers to the amount of an agent that is sufficient to achieve an outcome, for example, to affect beneficial or desired results. The therapeutically effective amount may vary depending upon one or more of: the subject and disease condition being treated, the weight and age of the subject, the severity of the disease condition, the manner of administration and the like.

MARCKS protein

[00019] MARCKS protein is an actin-binding protein and contributes to cytoskeleton orientation and function, and cell migration. Several exogenous stimuli can provoke degranulation of leukocytes via a pathway that involves activation of protein kinase C and subsequent phosphorylation and dephosphorylation events. MARCKS protein (where MARCKS as used herein means “Myristoylated Alanine-Rich C Kinase Substrate”), is a ubiquitous phosphorylation target of protein kinase C (PKC), and is highly expressed in leukocytes. MARCKS protein is mechanistically involved in a process of exocytotic secretion of mucin by goblet cells that line respiratory airways. MARCKS, a protein of approximately 82 kD, has three evolutionarily-conserved regions, an N-terminus, a phosphorylation site domain (or PSD), and a multiple homology 2 (MH2) domain. MARCKS is myristoylated via an amide bond at the N-terminal amino acid in the MARCKS protein’s amino acid sequence at the alpha-amine position of the glycine which resides at the N-terminus (i.e., at position 1) of amino acid sequence via a reaction catalyzed by myristoyl CoA:protein N-myristoyl transferase (NMT). The mechanism appears to involve binding of MARCKS, a myristoylated protein, to membranes of intracellular granules.

[00020] The myristoylated N-terminal region of MARCKS appears to be integral to the secretory process because it has been shown to block both mucin secretion and binding of MARCKS to mucin granule membranes in goblet cells. This peptide contains 24 L-amino acids of the MARCKS protein beginning with the N-terminal glycine of the MARCKS protein which is myristoylated via an amide bond and is known as myristoylated alpha-N-terminal sequence (or “MANS”, also interchangeably referred to as the “MARCKS N-terminus”); i.e., Myristoyl-GAQFSKTAAKGEAAAERPGEAAVA (SEQ ID NO: 1). The peptide fragments of the MANS peptide disclosed herein, also preferably are composed of L-amino acids. As MARCKS is an actin-binding protein, it is critical for cytoskeleton orientation and function and cell migration. In some embodiments, the N-terminal MARCKS peptides disclosed herein inhibit directed migration of human neutrophils, fibroblasts, and airway epithelial cells.

Peptides derived from N-Terminus of MARCKS

[00021] The disclosure provides peptides fragments (interchangeably referred to as just “fragments” or just “peptides”) derived from the MARCKS N-terminus. Exemplary MARCKS -related peptide fragments are discussed in U.S. Publication Nos. 2009-0203620 and 2014-0302057, and in International Patent Publication No. WO 2020/257162, the entire contents of each of which are incorporated herein by reference. In some aspects, these peptide fragments play a role in the reducing the rate and/or amount of release of inflammatory mediators granules or vesicles in inflammatory leukocytes.

[00022] In some aspects, the peptides disclosed herein are derived from the MARCKS N- terminus, i.e., contiguous peptide fragments derived from within the N-terminal l-to-24 amino acid sequence of MARCKS. In some aspects, the peptides are N-terminal amides of such fragments, such as N-terminal acetic acid amides of such fragments, and/or as well as C- terminal amides of such fragments, such as C-terminal amides of ammonia. In some aspects, the peptides have from about 4 to about 23 contiguous amino acid residues of the MANS peptide amino acid sequence. In some aspects, the fragments may be N-terminal-myristoylated if they do not begin with the N-terminal glycine at position 1 in SEQ ID NO: 1, or may be N- terminal-acylated with C2 to C12 acyl groups, including N-terminal-acetylated, and/or C- terminal amidated with an NH2 group.

[00023] Table 1 contains a list of amino acid sequences in single letter abbreviation format together with a respectively corresponding peptide number and SEQ ID NO. The reference peptide amino acid sequence (MANS peptide) is listed as peptide 1. Amino acid sequences of peptides of the disclosure having an amino acid sequence of from 4 to 23 contiguous amino acids of the reference amino acid sequence are listed as peptides 2 to 231, together with the amino acid sequence of a random N-terminal sequence (RNS) comprising amino acids of the MANS peptide as peptide 232. Amino acid sequences of representative variants of amino acid sequences of peptides of the disclosure as described herein and are also listed as peptides 233 to 245 and 247 to 251. The variant peptides listed are not intended to be a limiting group of peptides, but are presented only to serve as representative examples of variant peptides of the disclosure. Also presented is a representative reverse amino acid sequence (peptide 246) and a representative random amino acid sequence of peptide (peptide 232) of the disclosure.

[00024] In some aspects, the peptide comprises an amino acid sequence with at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99%, or at least about 99.5% identity to any one of the amino acid sequences listed in Table 1. In some aspects, the peptide comprises any one of the amino acid sequences listed in Table 1. In some aspects, the peptides consist of any one of the amino acid sequences listed in Table 1.

Table 1: Peptides and Amino Acid Sequences

[00025] In some aspects, the peptide is any one of the peptides listed in Table 1 A.

Table 1A

Ma = Myristoyl; Ac = Acetyl

[00026] The disclosure provides peptides having amino acid sequences comprising less than 24 amino acids with amino acid sequences related to the amino acid sequence of MANS peptide. The peptides of the current disclosure consist of amino acid sequences containing less than 24 amino acids, and may consist of from 4 to 14, from 10 to 12, from 9 to 14, from 9 to 13, from 10 to 13, from 10 to 14, at least 4, at least 9, at least 10, or the like amino acids. The peptides are typically straight chains, but may be cyclic peptides as well. Cyclic peptides are peptides that contain a circular or cyclic ring structure. The circular ring structure can be formed, for example, through connection between the amino and carboxyl ends of the peptide, or between the carboxyl or amino end and a side chain, or between a peptide backbone and the carboxyl or amino end or a side chain, or between two positions on the peptide backbone, or between two side chains. The connections may be formed via an amide bond, or other chemically stable bonds. In some embodiments, the peptide is a head-to-tail cyclic peptide. In some embodiments, the peptides are pegylated (PEGylated). PEGylating is the process of covalently attaching polyethylene glycol (PEG) chains to peptides. In some embodiments, PEGylating enhances solubility and/or half life of peptides, and/or reduces immunogenicity. Thus, in some embodiments, peptide PEGylation therapeutic efficacy and/or tolerability of peptide drugs. In some embodiments, the peptides are synthetic peptides. In some embodiments, the peptides are isolated peptides.

[00027] In some aspects, the peptide has an amino acid sequence selected from the group consisting of (a) an amino acid sequence having from 4 to 23 contiguous amino acids of the reference sequence, peptide 1; (b) a sequence with at least about 75%, at least about 80% identity, at least about 85%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, or at least about 95% identity to the amino acid sequence defined in (a); or (c) a variant of the amino acid sequence defined in (a), which variant is selected from the group consisting of a substitution variant, a deletion variant, an addition variant, and combinations thereof.

[00028] In other embodiments, the amino acid sequence of the peptide does not begin at the N-terminal amino acid of the reference sequence, peptide 1, (SEQ ID NO: 1) but rather begins at the amino acid at position 2 through the amino acid at position 21 of the reference sequence peptide 1. For example, the peptides may have an amino acid sequence selected from the group consisting of (a) an amino acid sequence having from 4 to 23 contiguous amino acids of the reference sequence peptide 1, wherein the amino acid sequence begins at any amino acid between position 2 through position 21 of the reference sequence. These peptides may be between 4 and 23 contiguous amino acids long and may represent peptides in the middle of the reference sequence, peptide 1; (b) a sequence with at least about 75%, at least 80% about identity, at least about 85%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, or at least about 95% identity to the amino acid sequence defined in (a); or (c) a variant of the amino acid sequence defined in (a). These peptides may contain no covalently bound chemical moiety or a chemical moiety on the N-terminal amino acid which is not the N-terminal glycine from or equivalent to the N-terminal glycine of the amino acid sequence SEQ ID NO: 1. Preferably, the chemical moiety is an acyl group, such as an acetyl group or a myristoyl group, in the form of an amide bond, or an alkyl group.

[00029] In yet other embodiments, the amino acid sequence of the peptide includes the contiguous residues A, K, G, and E as in peptide 219 of the reference sequence peptide 1. For example, the peptides may have an amino acid sequence selected from the group consisting of (a) an amino acid sequence having from 4 to 23 contiguous amino acids of the reference sequence peptide 1, wherein the amino acid sequence of the peptide includes the contiguous residues A,K,G, and E as in peptide 219 of the reference peptide 1 (e.g., peptide 219, peptide 45, peptide 79, peptide 67, peptide 80, etc.); (b) a sequence with at least about 75%, at least about 80% identity, at least about 85%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, or at least about 95% identity to the amino acid sequence defined in (a); or (c) a variant of the amino acid sequence defined in (a).

[00030] In further embodiments, the amino acid sequence of the peptide begins from the N- terminal amino acid of the reference sequence peptide 1 and includes the contiguous residues A, K, G, and E as in peptide 219 of the reference sequence peptide 1, while in other embodiments the amino acid sequence of the peptide ends at the C-terminal amino acid of the reference sequence peptide 1 and includes the contiguous residues A,K,G, and E as in peptide 219 of the reference sequence peptide 1. In some embodiments, the amino acid sequence of the peptide consists of SEQ ID NO: 219.

[00031] In exemplary aspects, the peptide is acetylated at the N-terminal amino acid. In exemplary aspects, the peptide comprises or consists of the amino acid sequence of SEQ ID NO: 106, and is acetylated at the N-terminal amino acid. In some aspects, the peptide comprises or consists of the amino acid sequence of SEQ ID NO: 219, and is acetylated at the N-terminal amino acid.

[00032] In some embodiments, the present disclosure provides composition comprising the peptides provided herein and salts thereof. For example, in some embodiments, the disclosure encompasses the peptides provided herein and pharmaceutically acceptable salts thereof. Pharmaceutically acceptable salts of the peptides of this disclosure include, for example, peptides modified by making acid or base salts thereof. Examples of acid addition salts include acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, lactate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, oxalate, palmoate, pectinate, persulfate, 3 -phenylpropionate, picrate, pivalate, propionate, succinate, tartrate, thiocyanate, tosylate, and undecanoate. Base salts include ammonium salts, alkali metal salts such as sodium and potassium salts, alkaline earth metal salts such as calcium and magnesium salts, salts with organic bases such as dicyclohexylamine salts, N-methyl-D-glutamine, and salts with amino acids such as arginine, lysine, and so forth. Also, the basic nitrogen-containing groups may be quaternized with such agents as lower alkyl halides, such as methyl, ethyl, propyl, and butyl chloride, bromides and iodides; dialkyl sulfates like dimethyl, diethyl, dibutyl; and diamyl sulfates, long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides, aralkyl halides like benzyl and phenethyl bromides and others.

Pharmaceutical compositions

[00033] In some aspects, any one of the peptides disclosed herein is contained in a pharmaceutical composition which is useful to prevent, treat, and/or block progression of dry eye syndrome.

[00034] The disclosure also encompasses a composition comprising a peptide as described in the paragraphs above and described herein and an excipient. The disclosure also encompasses a pharmaceutical composition comprising a peptide as described in the paragraphs above and described herein and a pharmaceutically acceptable carrier. The pharmaceutical composition can further preferably be sterile, sterilizable or sterilized. These peptides can be contained in a kit with reagents useful for administration.

[00035] In one aspect, the disclosure relates to a method of administering a pharmaceutical composition. The pharmaceutical composition comprises a therapeutically effective amount of a known compound and a pharmaceutically acceptable carrier. Pharmaceutically acceptable carriers are preferably liquid dosage forms. Liquid preparations may be used and may be prepared in the form of solutions or suspensions, e.g., solutions containing an active ingredient, and a mixture of water, glycerol, and propylene glycol. If desired, such liquid preparations may include one or more of following: thickening agents such as carboxymethylcellulose also may be used as well as other acceptable carriers, the selection of which is known in the art.

[00036] In certain embodiments, the drug product is present in a solid pharmaceutical composition. A solid composition of matter according to the present disclosure may be formed and may be mixed with and/or diluted by an excipient. The solid composition of matter also may be enclosed within a carrier, which may be, for example, in the form of a capsule, sachet, tablet, paper, or other container. When the excipient serves as a diluent, it may be a solid, semi- solid, a gel, or liquid material that acts as a vehicle, carrier, or medium for the composition of matter. For ophthalmic administration, the pharmaceutical formulation with any one of the peptides disclosed herein can be prepared in the form of an eye drop, eye gel, ointment, ointment, implant, microspheres, or liposomal formulation, or microemulsion.

[00037] Various suitable excipients will be understood by those skilled in the art and may be found in the National Formulary, 19: 2404-2406 (2000), the disclosure of pages 2404 to 2406 being incorporated herein in their entirety. Examples of suitable excipients include, but are not limited to, starches, gum arabic, calcium silicate, microcrystalline cellulose, methacrylates, shellac, polyvinylpyrrolidone, cellulose, water, and methylcellulose. The drug product formulations additionally can include lubricating agents such as, for example, talc, magnesium stearate and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl- and propyl hydroxybenzoates. Polyols, buffers, and inert fillers also may be used. Examples of polyols include, but are not limited to, mannitol, sorbitol, xylitol, sucrose, maltose, glucose, lactose, dextrose, and the like. Suitable buffers include, but are not limited to, phosphate, citrate, tartrate, succinate, and the like. Other inert fillers that may be used include those that are known in the art and are useful in the manufacture of various dosage forms. If desired, the solid formulations may include other components such as bulking agents and/or granulating agents, and the like. The drug products of the disclosure may be formulated so as to provide quick, sustained, or delayed release of the active ingredient after administration to the patient by employing procedures well known in the art.

[00038] In the event that the above pharmaceuticals are to be used for parenteral or intra- ocular administration, such a formulation may comprise sterile aqueous injection solutions, non-aqueous injection solutions, or both, comprising the composition of matter of the present disclosure. When aqueous injection solutions are prepared, the composition of matter may be present as a water soluble pharmaceutically acceptable salt. Parenteral or intra-ocular preparations may contain anti-oxidants, buffers, bacteriostats, and solutes which render the formulation isotonic with the blood of the intended recipient. Aqueous and non-aqueous sterile suspensions may comprise suspending agents and thickening agents. The formulations may be presented in unit-dose or multi-dose containers, for example sealed ampules and vials. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described. The parenteral or intra-ocular formulation can also be as liposomal composition.

[00039] The composition of matter also may be formulated such that it may be suitable for topical administration (e.g., ophthalmic drop or gel, or cream). These formulations may contain various excipients known to those skilled in the art. Suitable excipients may include, but are not limited to, cetyl esters wax, cetyl alcohol, white wax, glyceryl monostearate, propylene glycol, monostearate, methyl stearate, benzyl alcohol, sodium lauryl sulfate, glycerin, mineral oil, water, carbomer, ethyl alcohol, acrylate adhesives, polyisobutylene adhesives, and silicone adhesives.

Methods of treating dry eye syndrome and/or other corneal conditions

[00040] Dry eye syndrome, dry eye disease, and keratoconjucintivitis sicca are used interchangeably herein. Dry eye syndrome can be any syndrome associated with dysfunction and/or instability of tear film, including reduced tear secretion and increased tear evaporation. Indications encompassed by the term dry eye syndrome an the like include age-related dry eye, Stevens- Johnson syndrome, Sjogren's syndrome, ocular cicatrical pemphigoid, corneal injury, ocular surface infection, Riley-Day syndrome, congenital alacrima, nutritional disorders or deficiencies (including vitamin deficiencies), pharmacologic side effects, glandular and tissue destruction, autoimmune and other immunodeficient disorders, and inability to blink in comatose patients. Also included are dry eye symptoms caused by environmental exposure to airborne particulates, smoke, smog, and excessively dry air; as well as contact lens intolerance and eye stress caused by computer work or computer gaming.

[00041] Allergic conjunctivitis (seasonal and chronic), blepharitis and Meibomian gland dysfunction are examples of conditions that affect the quality and stability of the tear film, which results in dry eye signs and symptoms. Laser assisted vision correction procedures such as photorefractive keratectomy (PRK), laser-assisted sub-epithelial keratectomy (LASEK) and laser-assisted in situ keratomileusis (LASIK) can also negatively influence tear film functionality and cause dry eye disease.

[00042] The disclosure provides methods of treating dry eye syndrome in a subject by administering to the subject any one of the peptides disclosed herein. In some aspects, the method comprising administering to the subject any one of the peptides listed in Table 1 or Table 1A. In some embodiments, the peptide is present in a topical formulation for administration to the surface of the eye. The disclosure further provides methods of treating dry eye syndrome in a subject by administering to the subject a composition comprising any one of the peptides disclosed herein. In embodiments, the peptide comprises or consists of the sequence of SEQ ID NO: 106 or 219.

[00043] In some aspects, the subject is a mammal, such as humans, canines, equines and felines.

[00044] The method of administration of the peptides and compositions disclosed herein may be by topical administration. The ophthalmic administration generally includes an eye drop, eye gel, ointment, ointment, implant, microspheres, or liposomal formulation. In some embodiments, the method of administration of the peptides and compositions disclosed herein is by IVT injection. In some embodiments, the method of administration of the peptides and compositions disclosed herein is by a combination of IVT injection and topical administration. For example, in some embodiments, the compositions are administered by IVT injection followed by topical administration; or by topical administration followed by IVT injection. Topical administration may be, for example, once daily, twice daily, three times daily, four times daily, or more.

[00045] Additionally, the administration to the subj ect can further include the administration of a second molecule selected from the group consisting of an antibiotic, an antiviral compound, an antiparasitic compound, an anti-inflammatory compound, and an immunomodulator. As used herein, an immunomodulator or immunomodulatory compound is an agent that can affect the functioning of the immune system. In some aspects, the immunomodulatory compound helps normalize or regulate the immune system. Non limiting examples of immunomodulators include azathioprine, methotrexate, cyclosporine, tacrolimus, sirolimus, and everolimus. [00046] In some aspects, the peptide is administered at a concentration from about 1 pM to about 10 mM, such as, for example, about 10 pM, about 20 pM, about 30 pM, about 40 pM, about 50 pM, about 60 pM, about 70 pM, about 80 pM, about 90 pM, about 100 pM, about 150 pM, about 200 pM, about 250 pM, about 300 pM, about 350 pM, about 400 pM, about

450 pM, about 500 pM, about 550 pM, about 600 pM, about 650 pM, about 700 pM, about

750 pM, about 800 pM, about 850 pM, about 900 pM, about 950 pM, about 1 mM, about 2 mM, about 3 mM, about 4 mM, about 5 mM, about 6 mM, about 7 mM, about 8 mM, about 9 mM, or about 10 mM, including all subranges and values that lie therebetween.

[00047] In some aspects, the peptide is administered in an amount of about Ipg to about 5 mg, such as for example, about 10 pg, about 20 pg, about 30 pg, about 40 pg, about 50 pg, about 60 pg, about 70 pg, about 80 pg, about 90 pg, about 100 pg, about 150 pg, about 200 pg, about 250 pg, about 300 pg, about 350 pg, about 400 pg, about 450 pg, about 500 pg, about 550 pg, about 600 pg, about 650 pg, about 700 pg, about 750 pg, about 800 pg, about 850 pg, about 900 pg, about 950 pg, about 1 mg, about 2 mg, about 3 mg, about 4 mg, or about 5 mg, including all subranges and values that lie therebetween.

[00048] In some embodiments, the peptide may be administered in a volume of about 0.01 mL to about 1 mL, such as for example, about O.OlmL, about 0.05 mL, about 0.1 mL, about 0.5 mL, about 0.75 mL, or about 1 mL, including all subranges and values that lie therebetween.

[00049] The disclosure also provides a topical composition or topical formulation comprising at least one peptide having an amino acid sequence selected from the group consisting of: (a) an amino acid sequence having from 4 to 24 contiguous amino acids of a reference sequence, GAQFSKTAAKGEAAAERPGEAAVA (SEQ ID NO. 1); (b) an amino acid sequence having the sequence, GAQFSKTAAKGEAAAERPGEAAVA (SEQ ID NO. 1); and (c) an amino acid sequence with at least about 75% identity to the amino acid sequence defined in (a) or (b), for use in a method of treating dry eye syndrome in a subject, the method comprising administering the composition to the subject. In embodiments, the peptide has an amino acid sequence according to SEQ ID NO: 106 or SEQ ID NO: 219. In embodiments, the peptide is BIO-91201 (Ac-AKGE-OH; SEQ ID NO: 219). In embodiments, the topical composition or topical formulation is suitable for administration to the surface of the eye. [00050] Having now described the disclosure, the same will be illustrated with reference to certain examples, which are included herein for illustration purposes only, and which are not intended to be limiting of the disclosure.

EXAMPLES

Example 1. Topical Peptide inhibitors of MARCKS reduce signs of dry eye in the alacrimal rat model

[00051] Dry eye was induced in the left eye of Sprague-Dawley rats by removal of the extraorbital lacrimal gland. One group of rats had a skin incision, but no lacrimal gland removal (sham group). Topical administration of test compounds was initiated 2 weeks after surgery, given 4 times a day and continued for 24 days. Sham rats (n=6) were untreated. Alacrimal rats (n=6 each) were dosed bilaterally with lOuL of phosphate buffered saline (PBS), or 100 pM of either of two MARCKS inhibitors: BIO-11006 [high molecular weight] or BIO-91201 [lower molecular weight]. The efficacy of these MARCKS inhibitor peptides was assessed via clinical examination using biomicroscopy and phenol red test (PRT) tear volume with the observers masked to the treatments. Examinations were at baseline, 2 weeks after surgery, and on days 1, 3, 7, 14, 21, and 24 after initiating dosing. Clinical scores for corneal opacity, vascularization, and corneal fluorescein (FL) retention were recorded at each examination. The experimental design of the study is provided in the table below.

[00052] Clinical signs of dry eye were observed 2 weeks after surgery in groups with lacrimal gland removal, but not in rats in the sham group. No clinical abnormalities were observed in the right eyes (non- surgical) of the rats throughout the study. Left eyes of rats in the sham group had significantly lower clinical scores and higher PRT values than the PBS- dosed group at each examination day after surgery.

[00053] Corneal scores were assessed as in the table below.

[00054] BIO-91201 treated groups exhibited reduced corneal opacity scores relative to the PBS and BIO-11006 dosed groups. For example, at days 3 and 7, Corneal scores were significantly elevated in the PBS and BIO- 11006 dosed groups compared to sham, but no difference between sham and BIO-91201 was observed. The clinical opacity scores and statistical analyses are provided in FIG 1.

[00055] Similarly, BIO-91201 dosed groups had significantly lower FL retention than PBS or BIO-11006 dosed eyes (p<0.02) and through day 14, BIO- 91201 FL retention scores were not significantly different than sham eyes. PRT was higher in MARCKs inhibitor-dosed groups compared to PBS groups on days 14, 21, and 24 after surgery (p<0.02; t test). Fluorescein retention score was measured as shown in the table below, and the scores and statistical analyses are provided in FIG. 2.

[00056] Fluorescein retention scores.

[00057] FIG. 3 provides the results of the phenol red test (PRT), showing tear measurement in mm/20s averaged for each group.

[00058] Taken together, the results of the study showed that topical MARCKS inhibitor peptides appear well tolerated and may reduce inflammation, corneal opacity, corneal FL retention, and improve tear production in dry eye. In particular, the smaller peptide, BIO- 91201, exhibited a significant positive effect on indicators of dry eye disease including corneal scores and corneal fluorescein retention, as well as improvement in tear production. Both peptides were well tolerated without signs of inflammation. Further development of topical MARCKs inhibitors, especially the smaller peptide, BIO-91201, for treatment of dry eye appears warranted.

[00059] The foregoing examples are illustrative of the present disclosure and are not to be construed as limiting thereof. The disclosure is defined by the following claims, with equivalents of the claims to be included therein.

[00060] All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., "such as") provided herein, is intended merely to better illuminate the invention and does not pose a limitation on the scope of the invention unless otherwise claimed. No language in the specification should be construed as indicating any non- claimed element as essential to the practice of the invention.

[00061] All references, including publications, patent applications, and patents, cited herein are hereby incorporated by reference to the same extent as if each reference were individually and specifically indicated to be incorporated by reference and were set forth in its entirety herein.