NZ729142B2 - Ocular formulations for drug-delivery and protection of the anterior segment of the eye - Google Patents

Ocular formulations for drug-delivery and protection of the anterior segment of the eye Download PDF

Info

Publication number
NZ729142B2
NZ729142B2 NZ729142A NZ72914215A NZ729142B2 NZ 729142 B2 NZ729142 B2 NZ 729142B2 NZ 729142 A NZ729142 A NZ 729142A NZ 72914215 A NZ72914215 A NZ 72914215A NZ 729142 B2 NZ729142 B2 NZ 729142B2
Authority
NZ
New Zealand
Prior art keywords
active agent
months
compound
formulation
days
Prior art date
Application number
NZ729142A
Other versions
NZ729142A (en
Inventor
David P Bingaman
Paul G Chaney
Martin B Wax
Original Assignee
Panoptica Inc
Filing date
Publication date
Application filed by Panoptica Inc filed Critical Panoptica Inc
Priority to NZ767710A priority Critical patent/NZ767710A/en
Priority claimed from PCT/US2015/050818 external-priority patent/WO2016044671A1/en
Publication of NZ729142A publication Critical patent/NZ729142A/en
Publication of NZ729142B2 publication Critical patent/NZ729142B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/12Ketones
    • A61K31/122Ketones having the oxygen directly attached to a ring, e.g. quinones, vitamin K1, anthralin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/427Thiazoles not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/455Nicotinic acids, e.g. niacin; Derivatives thereof, e.g. esters, amides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/02Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/10Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/32Macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. carbomers, poly(meth)acrylates, or polyvinyl pyrrolidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/36Polysaccharides; Derivatives thereof, e.g. gums, starch, alginate, dextrin, hyaluronic acid, chitosan, inulin, agar or pectin
    • A61K47/38Cellulose; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0048Eye, e.g. artificial tears
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/08Solutions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics

Abstract

The present application relates to topical formulations comprising Compound-I, or its free base, and a second active agent selected from nicotinic acid, nicotinamide, and vitamin K, and a combination thereof, for treating ocular neovascularization. The present application also relates to pharmaceutical compositions comprising particles of Compound-I or its free base, and suspension formulations compring the particle compositions of Compound-I or its free base.

Description

OCULAR FORMULATIONS FOR DRUG-DELIVERY AND PROTECTION OF THE ANTERIOR SEGMENT OF THE EYE RELATED APPLICATION This application claims priority to, and the benefit of, U.S.S.N. 62/051,794, filed on September 17, 2014, the contents of which are incorporated by reference in their entirety.
FIELD OF THE APPLICATION Embodiments sed herein are generally directed to topical administration of a combination of a first active agent, e.g., a pharmaceutical compound or a salt thereof, and a second active agent, to treat ocular diseases or conditions. The embodiments disclosed include ocular formulations comprising a first active agent or a salt thereof and a second active agent, where the ation is a solution or a suspension. The on or suspension may further comprise a solubilizing agent, and is suitable for ring the first active agent or a salt thereof, to the ior t of the eye while protecting the anterior segment of the eye of a subject.
BACKGROUND Treatment of diseases or disorders of the posterior segment of the eye with topically applied active agents has not been ive because of inefficient delivery of the active agent to the target site. The vast majority of topical drugs penetrate via the cornea.
However, the cornea is not equally permeable to all topically d active agents, since the basic structure of the cornea dictates the relative penetration of active agent. Effectively, the greatest barrier to active agent penetration is the corneal epithelium which is rich in cellular membranes and is therefore more susceptible to penetration by lipophilic agents. In contrast, since the corneal stroma is largely constituted of water, active agents pass through more readily if they are hydrophilic. The elium represents a monolayer that, once more, is lipophilic. Active agents which are lipophilic or amphiphilic, in that they can behave as either charged or arged, penetrate the cornea best. Similar to the cornea, the conjunctiva! epithelium and blood vessels within or under the conjunctiva! epithelium may be ated by the same type of lipophilic or biphasic agents. However, e of the nature of the lipophilic membranes in the conjunctive and its nt vasculature, most active agents typically do not penetrate through the conjunctiva and into the eye. Agents with limited penetration into the vascular tissues in the conjunctival and subconjunctival regions are drained into the systemic circulation.
Because of the limited permeability of many topical drops to the corneal and conjunctival barriers, one major disadvantage of topical drops may be the need for high concentration of active agents in the topical formulation in order to achieve meaningful therapeutic drug levels in internal ocular tissues. Depending on the active agent, the molecule itself or high concentrations thereof, a topical ation may be toxic to the anterior segment of the eye, including the conjunctiva, cornea and/or lens, causing various injuries to the ocular surface, such as corneal epithelial defects and erosions.
Ocular side—effects observed following treatment with anti-EGFR drug therapies, e. g., anti—EGFR cancer therapies, have illuminated the essential role the EGFR ing pathway plays in maintaining and restoring the health of the human corneal epithelium.
Patients treated with anti—EGFR drug therapies can develop corneal changes, such as epithelial degeneration and defects, tion, corneal epithelial thinning, erosions and/or l edema, keratitis as well as perforation while undergoing therapy or even after discontinuation of the anti-EGFR therapy. The important role of EGFR ing in tasis and pathophysiology of the corneal epithelium is well established. EGFR activation is both necessary and suf?cient for corneal epithelial migration, proliferation, and differentiation. Moreover, EGFR is the primary mediator of wound healing during in vitro experiments with immortalized human corneal epithelial cells. Therefore, treating ocular diseases or ers (e.g., diseases or disorders of the posterior segment of the eye) would benefit from administration of an agent that maintains EGFR activity in addition to the administration of a formulation comprising an active agent (e.g., an active agent having toxic or anti-EGFR activity).
The t application provides novel ations which circumvent the problems encountered in ocular delivery of existing topical therapeutic agents. The t application accomplishes the combined effects of decreasing corneal and anterior t drug exposure and protecting corneal and or segment tissues, while increasing ior segment bioavailability. By ng l exposure, protecting corneal tissues, and increasing posterior segment bioavailability, the formulation of the present application improves ocular tolerability and ses the therapeutic index of the active agent.
SUMMARY The present application s to pharmaceutical formulations in the form of a solution and/or a suspension, which lower the exposure to a ?rst active agent in the or segment of the eye, for example the ocular surface comprised of the cornea and ctiva, and protect the eye, for example, the anterior segment of the eye, through maintenance of EGFR activity. The pharmaceutical formulations of the ation increase the bioavailability of the ?rst active agent at the posterior segment of the eye, for e at the central choroid and/or the central retina, and protect and/or e the health of the anterior segment of the eye.
The present application provides a formulation comprising a ?rst active agent, and optionally a second active agent, in the form of a solution or a suspension, with superior characteristics compared to a composition formed as a gel. The present application provides that the ?rst active agent and/or the second active agent can be formulated together as a on and/or a suspension. Increased levels of the ?rst active agent in the anterior segment of the eye limit ocular tolerability of topical drops containing the ?rst active agent and may cause corneal lial defects and erosions. The presence of the second active agent can prevent damage that may be caused by exposure to the ?rst active agent, treat any damage that may be caused by exposure to the ?rst active agent, and/or improve the overall health of the ocular surface, speci?cally the corneal epithelium. The second active agent can be formulated together with the ?rst active agent, or formulated as a separate formulation that is administered in combination with the ation comprising the ?rst active agent.
The formulations of the present application reduce re of the ?rst active agent at the anterior segment of the eye, such as corneal or conjunctival surface, protect from and/or repair damages to the anterior segment of the eye, such as the corneal or conjunctival surface, and maintain te concentrations of a ?rst active agent necessary to bind the relevant receptors at the target tissues and confer a therapeutic effect in the posterior segment of the eye, such as the choroid and the retina.
The present application relates to formulations and methods useful for ng pathological states that arise from or are exacerbated by ocular angiogenesis, neovascularization, and/or vascular leakage, for example, in diabetic retinopathy (including background diabetic retinopathy, proliferative diabetic retinopathy, and diabetic macular ; age—related macular degeneration (AMD) (including neovascular (wet/exudative) AMD, dry AMD, and phic Atrophy); pathologic choroidal cularization (CNV) and vascular leakage from any mechanism (e.g., high myopia, trauma, sickle cell e; ocular histoplasmosis, angioid streaks, traumatic choroidal rupture, drusen of the optic nerve, or some retinal dystrophies); pathologic retinal neovascularization and vascular leakage from any mechanism (e.g., sickle cell retinopathy, Eales disease, ocular ischemic syndrome, carotid cavernous ?stula, al exudative vitreoretinopathy, hyperviscosity syndrome, idiopathic occlusive arteriolitis, birdshot retinochoroidopathy, l vasculitis, sarcoidosis, or toxoplasmosis); uveitis; retinal vein occlusion (central or branch); ocular trauma; y induced edema; surgery induced neovascularization; cystoid macular edema; ocular ischemia; retinopathy of prematurity; Coat's disease; sickle cell retinopathy; and/or cular glaucoma. In one ment, the pathological state is AMD. In one embodiment, the pathological states arise from or are exacerbated by ocular angiogenesis and/or neovascularization.
The present application also relates to formulations and methods useful for preventing and/or treating corneal epithelium disruptions associated with a disease, including a systemic disease (e.g., cancer, diabetes, etc.) and an eye e, or with a side effect from a locally or systemically stered drug (e.g., anti—EGFR agents or EGFR inhibitors). The formulation of the present application has, at least, one anti-angiogenic agent, anti- atory agent, or anti-vascular permeability agent for use in treating angiogenic ocular disorders and an EGFR tor (e.g., an agent that maintains the activity of or activates EGFR) for use in preventing and/or treating corneal epithelium disruptions.
According to embodiments of the application, the ?rst active agent is an anti- angiogenic kinase inhibitor and the second active agent is a kinase modulator (e.g., activator).
In one embodiment, the ?rst active agent ts a kinase that is ent from the kinase that is modulated (e. g., activity maintained or activated) by the second active agent.
Examples of some kinase inhibitors that can be used to bring about bene?cial therapeutic results include inhibitors of receptor ne kinases, for example, without being limiting, VEGFR, FGFR, Tie-2, and Ephrin kinase receptors. Examples of kinase tors that can be used to bring about ial therapeutic results include activators of ErbB receptor tyrosine kinase, for example, without being limiting, ErbBl/HERl , ErbB2/HER2/Neu, ErbB3/HER3, and ErbB4/HER. In one embodiment, the ErbB receptor tyrosine kinase is EGFRl. In one embodiment, the ?rst active agent is a VEGFR inhibitor.
In one embodiment, the second active agent is an EGFR modulator (e.g., activator). In a further embodiment, the second active agent is nicotinic acid, nicotinamide, or vitamin K, or a combination thereof. In a further embodiment, the second active agent is nicotinic acid or nicotinamide. In r embodiment, the second active agent vitamin K.
In some ments, a second active agent of the present application reduces or alleviates the tion of EGFR at the anterior segment of the eye caused by high concentrations of a ?rst active agent of the present application (e.g., transient high concentrations of the ?rst active agent after administration of the ?rst active agent to the anterior segment of the eye).
The embodiments of the present application provide an ophthalmic formulation for treating ocular neovascularization comprising a ?rst active agent of Formula I: 0 NH; NMS 0 or a pharmaceutically acceptable salt thereof; a second active agent n the second active agent is an EGFR modulator (e.g., activator), such as nicotinic acid, nicotinamide, or n K, or a combination thereof; and ceutically acceptable excipients; the ?rst active agent or the pharmaceutically acceptable salt is t in about 0.02% to about 1.2% w/v such that the formulation forms a solution or suspension, and wherein: X O or S; R1 is H, C1 -C10 alkyl, C2 -C10 alkenyl, C2 -C10 alkynyl, C(O)(C1 -C10 alkyl), (CH2)t(C6 — C10 aryl), (CH2)t(4— 10 membered heterocyclic), C(O)(CH2)t(C6 —C10 aryl), or C(O)(CH2)t (5—10 membered heterocyclic), wherein: t is an integer from 0 to 5; the alkyl group ally includes 1 or 2 hetero moieties selected from O, S and N(R6) with the proviso that two 0 atoms, two S atoms, or an O and an S atoms are not attached directly to each other; the aryl and heterocyclic groups are optionally fused with a C5 -C10 aryl group, a C5 -C8 saturated cyclic group, or a 5-10 membered heterocyclic group; 1 or 2 carbon atoms in the foregoing heterocyclic moieties are optionally substituted with an oxo (=0) moiety or an anion of oxygen; the (CH2) moieties ally include a carbon—carbon double or triple bond when t is an integer from 2 to 5; and the foregoing R1 groups, except H, are optionally substituted with l to 3 R4 R2 is H; R3 is (CH2)t(C6 -C10 aryl), wherein: t is an integer from 0 to 5; the aryl group is optionally fused with a C6 —C10 aryl group, a C5 -Cg saturated cyclic group, or a 5-10 membered heterocyclic group; the (CH2) moieties optionally include a carbon—carbon double or triple bond when t is an integer from 2 to 5; and the foregoing R3 groups are optionally substituted with l to 5 R4 groups; each R4 is ndently selected from C1—C10 alkyl, C2 -C10 alkenyl, C2 -C10 alkynyl, halo, cyano, nitro, trifluoromethyl, tri?uoromethoxy, azido, 0R5, C(O)R5, C(O)OR5, NR6C(O)R5, NR6C(O)OR5, OC(O)R5, NR6S02R5, SOZNR5R6, C(O)NR5R6, NR5R6, S(O)jR7 where j is an integer from 0 to 2, SO3H, NR5(CR6R7)rOR6, (CH2)t(C6 —C10 aryl), 2)t(C6 —C10 aryl), S(CH2)t(C6 —C10 aryl), O(CH2)r(C6 —C10 aryl), (CH2)t(5—10 membered heterocyclic), and (CR6 R7)mOR6, wherein: m is an integer from 1 to 5; t is an integer from 0 to 5; the alkyl group optionally includes 1 or 2 hetero moieties selected from O, S and N(R6) with the proviso that two 0 atoms, two S atoms, or an O and an S atoms are not attached ly to each other; the aryl and heterocyclic groups are optionally fused with a C6 -C10 aryl group, a C5 —C8 saturated cyclic group, or a 5—10 membered heterocyclic group; 1 or 2 carbon atoms in the foregoing heterocyclic moieties are optionally substituted with an oxo (=0) moiety or an anion of oxygen; and the alkyl, aryl and cyclic moieties of the foregoing R4 groups are ally tuted with 1 to 3 substituents independently selected from halo, cyano, nitro, romethyl, tri?uoromethoxy, azido, NR6SOZR5, SOZNR5 R6, C(O)R5, C(O)OR5, OC(O)R5, NR6C(O)R5, C(O)NR5R6, NR5R6, (CR6R7)mOR6 where m is an integer from 1 to 5, 0R5, and the substituents listed in the de?nition of R5 ; R5, R6, and R7 are each independently H or C1 —C6 alkyl.
In one embodiment, R3 is (CH2)t(C6 —C10aryl), wherein t is an integer from 1 to 3 and R3 is optionally substituted with 1 to 4 R4 groups.
In a further embodiment, R3 is benzyl, ally substituted with 1 to 4 substituents independently selected from halo and C1-C4 alkyl. In a further embodiment, R3 is benzyl substituted With l to 4 substituents independently selected from methyl, ?uoro, chloro and bromo.
In one embodiment, R1 is (CH2)t(5—10 membered heterocyclic), wherein t is an integer from 0 to 5, optionally substituted with 1 or 2 substituents independently selected from C1 —C4 alkyl, hydroxy and hydroxymethyl.
The present application provides heterocyclic moiety of the R1 group in Formula I selected from lino, pyrrolidinyl, imidazolyl, piperazinyl, piperidinyl, and 2,5-diaza— bicyclo[2.2.1]heptyl, the t le of the R1 group ranges from 2 to 5, and the R1 group is optionally substituted with one or more hydroxy groups.
For example, the heterocyclic moiety of the R1 group in Formula I of the present application is pyrrolidine.
In a further embodiment of theNpresent application, the ?rst active agent is: (11).
In a further embodiment of the present application, the ?rst active agent is a hydrochloride salt of compound of Formula II, namely Compound-I: is (Compound—I).
In one embodiment, the second active agent is an EGFR modulator (e.g., activator).
In a further embodiment, the second active agent is nic acid, nicotinamide, or vitamin K, or a combination thereof. In a further embodiment, the second active agent is vitamin K.
The embodiments of the present ation provide formulations comprising about 0.005% to about 5.0% w/v of the ?rst active agent of Formula I or II, or a pharmaceutically acceptable salt thereof, for example, Compound-I. In some ments, the tration of nd—I or its free base (Formula II) in the formulations is about 0.005% — about 0.01%, about 0.01% — about 0.05%, about 0.05% - about 0.1%, about 0.1% — about 0.2%, about 0.2% — about 0.3%, about 0.3% — about 0.4%, about 0.4% — about 0.5%, about 0.5% — about 0.6%, about 0.6% — about 0.7%, about 0.7% — about 0.8%, about 0.8% — about 0.9%, about 0.9% — about 1.0%, about 1.1 — about 2.0%, about 2.1 — about 3.0%, about 3.1 — about 4.0%, or about 4.1— about 5.0% W/v for topical stration. In some embodiments, the concentration of nd—I or its free base (Formula II) in the formulations is about 0.1% — about 1.2%, about 0.2% — about 1.2%, about 0.3% — about 1.2%, about 0.4% — about 1.2%, 0.1% - about 1.1%, about 0.2% — about 1.1%, about 0.3% — about 1.1%, about 0.4% — about 1.1%, 0.1% — about 1.0%, about 0.2% — about 1.0%, about 0.3% - about 1.0%, about 0.4% - about 1.0%, 0.1% — about 0.8%, about 0.2% — about 0.8%, about 0.3% - about 08%, about 0.4% - about 0.8%, 0.1% - about 0.6%, about 0.2% - about 0.6%, about 0.3% - about 0.6%, about 0.4% - about 0.6%, 0.1% - about 0.5%, about 0.2% - about 0.5%, about 0.3% — about 0.5%, about 0.4% — about 0.5%, 0.1% - about 0.4%, about 0.2% — about 0.4%, about 0.3% — about 0.4% w/v for topical stration. In some embodiments the formulations include about 0.005%, about 0.05%, about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1.0%, about 2.0%, about 3.0%, about 4.0%, or about 5.0% w/v of Compound—I or its free base (Formula In some embodiments, the present application provides a on of a ?rst active agent (e.g., Compound—I) and a second active agent (e.g., nicotinic acid, nicotinamide, or vitamin K, or a combination thereof), which includes one or more solubilizing agents.
In some embodiments, the ation comprises about 0.005%, about 0.05%, about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1.0%, about 2.0%, about 3.0%, about 4.0%, or about 5.0% W/v of Formula I or II, or a pharmaceutically acceptable salt thereof, for example, Compound-I, and a second active agent and a solubilizing agent.
In some embodiments, the solubilizing agent in the formulation may be cyclodextrin, for example, 2-hydroxypropy1—B—cyclodextrin, methyl-B-cyclodextrin, randomly ated—B—cyclodextrin, ted—B—cyclodextrin, triacetyl—B—cyclodextrin, peracetylated— B—cyclodextrin, carboxymethyl-B-cyclodextrin, hydroxyethyl —B—cyclodextrin, 2—hydroxy—3— (trimethylammonio)propyl-B-cyclodextrin, glucosyl -|3-cyclodextrin, maltosyl-B-cyclodextrin, utyl ether-B-cyclodextrin, branched-B-cyclodextrin, hydroxypropyl-y-cyclodextrin, randomly methylated-y-cyclodextrin, trimethyl-y-cyclodextrin, or a combination thereof.
In one embodiment, the solubilizing agent in the formulation is 2-hydroxypropy1— B—cyclodextrin or B—cyclodextrin sulfobutyl ether.
In one embodiment, the formulation may further comprise one or more of benzalkonium chloride (BAK), sodium chloride, and a pH adjusting agent.
In additional embodiments, the formulation ses about 0.005%, about 0.05%, about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1.0%, about 2.0%, about 3.0%, about 4.0%, or about .0% W/v of a ?rst active agent or a pharmaceutically acceptable salt f, and a buffer, for example, tromethamine. In one embodiment, the formulation ses about 0.005%, about 0.05%, about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1.0%, about 2.0%, about 3.0%, about 4.0%, or about 5.0% W/v of the first active agent or a pharmaceutically acceptable salt thereof, and about 0.3% - about 1.0% w/v tromethamine, and optionally further comprises about 0.005% w/v benzalkonium chloride (BAK).
In some embodiments, the second active agent is an EGFR modulator (e.g., activator). For example, the second active agent includes, but is not d to, nicotinic acid (or niacin/vitamin B3), nicotinamide, and vitamin K, or a combination thereof. "Vitamin K," as de?ned herein, includes one or more s of the vitamin K family and prodrugs thereof, lly occurring or synthetic. The vitamin K family is comprised of vitamin K1 (also known as phylloquinone, phytomenadione or phytonadione), vitamin K2, and any vitamin K2 homologues. The vitamin K2 homologues are called menoquinones and are characterized by the number of isoprenoid residues in their ains. Synthetic vitamin K includes, but is not limited to vitamin K3 (i.e., menadione), vitamin K4, vitamin K5. In one embodiment, the second active agent is nic acid and/or and nicotinamide. In another embodiment, the second active agent is nicotinic acid. In another embodiment, the second active agent is nicotinamide. In yet another embodiment, the second active agent is vitamin K. In another embodiment, the second active agent is vitamin K1. In yet another embodiment, the second active agent is vitamin K2. In yet another embodiment, the second active agent is a vitamin K2 homologue. In yet another embodiment, the second active agent is a synthetic vitamin K (e.g., vitamin K3, Vitamin K4, or vitamin K5). In one embodiment, the second active agent is vitamin K3 (226., menadione).
In some embodiments, the concentration of the second active agent in the formulations is about 0.00001% - about 5.0% w/v for topical administration. In some embodiments, the concentration of the second active agent in the formulations is about 0.00001% — about 1.0%, about 0.00001% — about 0.1%, about 0.00001% - about 0.01%, about 0.00001% — about 0.001%, about 0.00001% — about %, or about 0.00001% — about % W/v for topical administration. In some embodiments, the concentration of the second active agent in the formulations is about 0.00001% — about 0.0001%, 0.000012% — about 0.0001%, 0.000014% - about 0.0001%, 16% - about 0.0001%, 0.000018% - about 0.0001%, 0.00002% - about 0.0001%, 0.00003% - about 0.0001%, 0.00004% - about 0.0001%, 0.00005% - about 0.0001%, 0.00006% — about 0.0001%, 0.00007% - about 0.0001%, 0.00008% - about 0.0001%, 0.00009% — about 0.0001%, 16% - about 0.00009%, 0.000018% - about 0.00009%, 0.00002% — about 0.00009%, 0.00003% - about 0.00009%, 0.00004% - about 0.00009%, 0.00005% — about 0.00009%, 0.00006% - about 0.00009%, 0.00007% - about 0.00009%, or 0.00008% — about 0.00009% w/v for topical administration. In some ments, the formulations include about 0.00001%, 0.00002%, 0.00003%, 0.00004%, 0.00005%, 0.00006%, 0.0000?%, 0.00008%, 0.000081%, 0.000082%, 0.000083%, 0.000084%, 0.000085%, 86%, 0.00008?%, 0.000088%, or 0.000089% w/v of the second active agent, or a pharmaceutically acceptable salt f.
In some embodiments, the concentration of the second active agent in the formulations is about 0.5 uM, about 0.6 pM, about 0.? uM, about 0.8 uM, about 0.9 uM, about 1 uM, about 2 uM, about 3 uM, about 4 uM, about 5 uM, about 6 uM, about 7 uM, about 8 uM, or about 9 uM. In some embodiments, the concentration of the second active agent is about 1 HM.
The present application provides a formulation having a pH value of about 4.5 to about 7.5 at or under about 40 0C. In some embodiments, the pH value of the formulation is between about pH 5.0 to about 7.0. In one embodiment, the pH value of the formulation is about 6.0 at or under about 40 0C.
In another embodiment, the present application provides use of a formulation comprising a ?rst active agent (e.g., Formula I or Compound-I or its free base la II)), and a second active agent (e.g., nicotinic acid, nicotinamide, or vitamin K, or a combination f), for the manufacture of a medicament for ing posterior segment of the eye and/or for treating and/or ameliorating a posterior segment disease or a vasculopathic or in?ammatory disease of the eye, as described herein, e.g., diabetic retinopathy ding background diabetic retinopathy, proliferative diabetic retinopathy and diabetic r edema); age-related macular degeneration (AMD) (including neovascular (wet/exudative) AMD, dry AMD, and Geographic Atrophy); pathologic dal neovascularization (CNV) from any mechanism (e.g., high myopia, trauma, sickle cell disease; ocular lasmosis, angioid s, tic choroidal rupture, drusen of the optic nerve, and some retinal dystrophies); pathologic retinal neovascularization from any mechanism (e.g., sickle cell retinopathy, Eales disease, ocular ischemic syndrome, d cavernous ?stula, familial exudative vitreoretinopathy, hyperviscosity syndrome, idiopathic ive arteriolitis; birdshot retinochoroidopathy, retinal vasculitis, sarcoidosis and toxoplasmosis); uveitis; retinal vein occlusion (central or branch); ocular trauma; surgery induced edema; surgery induced neovascularization; cystoid macular edema; ocular ischemia; retinopathy of prematurity; Coat's disease; sickle cell retinopathy and/or neovascular glaucoma. In one embodiment, the disease of the eye is AMD. In one embodiment, the diseases of the eye arise from or are exacerbated by ocular angiogenesis and/or neovascularization.
In another embodiment, the present application relates to a formulation for use in the manufacture of a medicament suitable for accessing the posterior segment of the eye and/or for treating and/or ameliorating a posterior segment disease or a vasculopathic or in?ammatory disease of the eye. In one embodiment, the formulation ses a first active agent and a second active agent. In one embodiment, the ?rst active agent is a VEGFR inhibitor. In one embodiment, the ?rst active agent is a nd of Formula I or a pharmaceutically acceptable salt thereof. In a further embodiment, the first active agent is a compound of Formula II or a ceutically acceptable salt f. In a further embodiment, the ?rst active agent is Compound—I. In one embodiment, the second active agent is an EGFR tor (e.g., tor). In a further embodiment, the second active agent is selected from nic acid, nicotinamide, and vitamin K, and a combination thereof.
The formulation may further comprise one or more pharmaceutically acceptable excipients.
In another embodiment, the present application relates to a formulation for use in the manufacture of a medicament for treating and/or ameliorating a symptom of an ocular disease or disorder (e.g., a posterior segment or vasculopathic or in?ammatory e of the eye). In one embodiment, the formulation comprises a ?rst active agent and a second active agent. In one embodiment, the ?rst active agent is a VEGFR inhibitor. In one ment, the ?rst active agent is a compound of Formula I or a pharmaceutically acceptable salt thereof. In a further embodiment, the ?rst active agent is a compound of Formula II or a pharmaceutically acceptable salt thereof In a further embodiment, the ?rst active agent is Compound-I. In one ment, the second active agent is an EGFR modulator (e.g., activator). In a further ment, the second active agent is selected from nicotinic acid, nicotinamide, and vitamin K, and a ation thereof. The ation may further comprise one or more pharmaceutically acceptable excipients.
In yet another embodiment, the present ation relates to a formulation for use in a method for accessing posterior segment of the eye and/or for treating and/or ameliorating a posterior segment disease of the eye. In one embodiment, the formulation comprises a ?rst active agent and a second active agent. In one ment, the ?rst active agent is a VEGFR inhibitor. In one embodiment, the ?rst active agent is a compound of Formula I or a pharmaceutically acceptable salt thereof. In a further embodiment, the ?rst active agent is a compound of Formula II or a pharmaceutically acceptable salt thereof. In a further embodiment, the ?rst active agent is Compound—I. In one embodiment, the second active agent is an EGFR modulator (e.g., tor). In one embodiment, the second active agent is selected from nic acid, namide, and n K, and a ation thereof. The formulation may further comprise one or more pharmaceutically acceptable excipients.
In yet another embodiment, the present application relates to a formulation for use in a method for ng and/or ameliorating a symptom of an ocular disease or disorder (e. g., a posterior segment vasculopathic or in?ammatory disease of the eye). In one embodiment, the formulation comprises a ?rst active agent and a second active agent. In one embodiment, the ?rst active agent is a VEGFR inhibitor. In one embodiment, the ?rst active agent is a compound of Formula I or a ceutically acceptable salt thereof. In a further embodiment, the ?rst active agent is a compound of Formula II or a pharmaceutically acceptable salt thereof. In a further embodiment, the ?rst active agent is Compound-I. In one embodiment, the second active agent is an EGFR modulator (e.g., activator). In a further embodiment, the second active agent is selected from nicotinic acid, nicotinamide, and vitamin K, and a combination thereof. The formulation may further comprise one or more pharmaceutically able excipients.
In another embodiment, the present application relates to a combinational therapy for accessing the posterior segment of the eye and/or for treating and/or ameliorating a posterior segment disease of the eye, wherein the therapy comprises stering a ?rst active agent and a second active agent. In one embodiment, the ?rst active agent is a VEGFR inhibitor. In one embodiment, the ?rst active agent is a compound of a I or a ceutically acceptable salt thereof. In a further embodiment, the ?rst active agent is a compound of Formula II or a pharmaceutically acceptable salt thereof. In a further embodiment, the ?rst active agent is Compound-I. In one embodiment, the second active agent is an EGFR modulator (e.g., activator). In a further embodiment, the second active agent is selected from nicotinic acid, nicotinamide, and vitamin K, and a combination thereof.
In one embodiment, the ?rst active agent is administered simultaneously with the second active agent. In another embodiment, the ?rst active agent is administered prior to the stration of the second active agent. In another embodiment, the ?rst active agent is stered after the administration of the second active agent.
In another embodiment, the present ation relates to a combinational therapy for treating and/or ameliorating a symptom of an ocular disease or disorder (e.g., a posterior segment vasculopathic or in?ammatory e of the eye), comprising administering a ?rst active agent and a second active agent. In one embodiment, the ?rst active agent is a VEGFR inhibitor. In one embodiment, the ?rst active agent is a compound of Formula I or a pharmaceutically acceptable salt thereof. In a r embodiment, the ?rst active agent is a compound of Formula II or a pharmaceutically acceptable salt thereof. In a further embodiment, the ?rst active agent is Compound-I. In one embodiment, the second active agent is an EGFR modulator (e.g., activator). In a further embodiment, the second active agent is selected from nicotinic acid, nicotinamide, and Vitamin K, and a combination thereof.
In one embodiment, the ?rst active agent is administered simultaneously with the second active agent. In another embodiment, the ?rst active agent is administered prior to the administration of the second active agent. In another ment, the ?rst active agent is administered after the administration of the second active agent.
In some embodiments, the re time of the ?rst active agent (e.g., Compound—I) and the second active agent is between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 , or 12 months). In some ments, the exposure time of the ?rst active agent (e. g., Compound—I) and the second active agent is longer than 90 days (e. g., 4 months, 6 months, 8 months, or 12 months). In some ments, the dosage regimen involves several courses of topical ocular administration of a formulation comprising the ?rst active agent (e. g., Compound—I) and a second active agent to a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 ) or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months) or for longer than 90 days. For example, the dosage regimen involves once daily, twice daily, three times daily or four times daily administration of the formulation for between 1 and 90 days or for longer than 90 days (e. g., 4 months, 6 months, 8 months, or 12 months) or for longer than 90 days (e. g., 4 months, 6 months, 8 months, or 12 months). For e, the dosage regimen involves once, twice, three times, or four times administration of the formulation on alternate days (126., on day 1, 3, 5, 7 etc.) for up to 90 days. For example, the dosage n involves administering once on day 1, once or twice on day 2 to day 90. For example, the dosage regimen involves administering once, twice, three times, or four times on day 1, ed by once daily for 2—90 days. For example, the dosage n involves administering once, twice, three times, four times on day 1, followed by once, twice, three times, or four times on alternate days (i.e., on day 1, 3, 5, 7 etc.) for up to 90 days. For example, one dosage regimen involves stering once or twice per day for 1, 2, 3, 4, or 5 consecutive days. For twice or three daily dosage regimen, subjects e a l ocular dose of a ?rst active agent (e.g., nd—I) and a second active agent ation on days 1 and 4 approximately about 4, 6, or 8 hours apart. In another embodiment, subjects receive topical ocular doses of a ?rst active agent (e.g., nd—I) and a second active agent formulation approximately about 4, 6, or 8 hours apart for four consecutive days. In some embodiments, subjects receive one or two topical ocular doses of a ?rst active agent (e.g., Compound—I) and a second active agent formulation per day for 5 consecutive days. In yet other embodiments, subjects receive one or two topical ocular dose of a ?rst active agent (e.g, Compound-I) and a second active agent formulation for 5—90 consecutive days. In some embodiments, subjects receive one or two topical ocular doses of a ?rst active agent (e.g., Compound-I) and a second active agent formulation for at least 25 consecutive days. In one embodiment, ts receive one or two topical ocular doses for at least 90 consecutive days or more.
For example, a formulation comprising about 1 mg/mL BID of a ?rst active agent (e.g., Compound—I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e. g., 4 months, 6 months, 8 months, or 12 months) or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 1 mg/mL QD of a ?rst active agent (e.g., nd—I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e. g., 4 months, 6 months, 8 months, or 12 ). In some embodiments a formulation comprising about 1 mg/mL TID of a ?rst active agent (e.g., Compound-I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation sing about 1 mg/mL QID of a ?rst active agent (e.g., Compound-I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a ation comprising about 2 mg/mL BID of a ?rst active agent (e.g., Compound-I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 2 mg/mL QD of a ?rst active agent (e. g., Compound—I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e. g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 2 mg/mL TID of a ?rst active agent (e.g., Compound-I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 2 mg/mL QID of a ?rst active agent (e.g., Compound-I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 ). In some embodiments a formulation sing about 3 mg/mL BID of a ?rst active agent (e.g., Compound-I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 , or 12 months). In some embodiments a formulation comprising about 3 mg/mL QD of a ?rst active agent (e. g., Compound—I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e. g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 3 mg/mL TID of a ?rst active agent (e.g., Compound—I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e. g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation sing about 3 mg/mL QID of a ?rst active agent (e.g., nd-I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 ). In some embodiments a formulation comprising about 4 mg/mL BID of a ?rst active agent (e.g., Compound—I) and a second active agent is administered to one eye or both eyes of a subject for n 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 4 mg/mL QD of a ?rst active agent (e.g., Compound—I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e. g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 4 mg/mL TID of a ?rst active agent (e.g., Compound—I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 ). In some embodiments a formulation comprising about 4 mg/mL QID of a ?rst active agent (e.g., Compound-I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 5 mg/mL BID of a ?rst active agent (e.g., nd—I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some ments a formulation comprising about 5 mg/mL QD of a ?rst active agent (e. g., Compound—I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e. g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 5 mg/mL TID of a ?rst active agent (e.g., Compound—I) and a second active agent is stered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 5 mg/mL QID of a ?rst active agent (e.g., Compound-I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 6 mg/mL BID of a ?rst active agent (e.g., Compound—I) and a second active agent is administered to one eye or both eyes of a t for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 6 mg/mL QD of a ?rst active agent (e.g., Compound—I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e. g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 6 mg/mL TID of a ?rst active agent (e.g., Compound—I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e. g., 4 months, 6 months, 8 , or 12 ). In some embodiments a formulation sing about 6 mg/mL QID of a ?rst active agent (e.g., Compound-I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 , or 12 months). In some embodiments a formulation comprising about 7 mg/mL BID of a ?rst active agent (e.g., Compound—I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 7 mg/mL QD of a ?rst active agent (e. g., Compound-I) and a second active agent is stered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a ation comprising about 7 mg/mL TID of a ?rst active agent (e.g, Compound—I) and a second active agent is administered to one eye or both eyes of a t for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 7 mgmL QID of a ?rst active agent (e.g., Compound-I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 8 mg/mL BID of a ?rst active agent (e.g., Compound—I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some ments a formulation comprising about 8 mg/mL QD of a ?rst active agent (e. g., Compound-I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e. g., 4 months, 6 , 8 months, or 12 months). In some embodiments a formulation comprising about 8 mg/mL TID of a ?rst active agent (e.g., Compound—I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e. g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 8 mg/mL QID of a ?rst active agent (e.g., Compound-I) and a second active agent is administered to one eye or both eyes of a t for between 1 and 90 days or for longer than 90 days (e.g., 4 , 6 months, 8 months, or 12 months). In some embodiments a ation sing about 9 mg/mL BID of a ?rst active agent (e.g., Compound—I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 9 mg/mL QD of a ?rst active agent (e.g., Compound—I) and a second active agent is administered to one eye or both eyes of a t for n 1 and 90 days or for longer than 90 days (e. g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 9 mg/mL TID of a ?rst active agent (e.g., Compound-I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 , 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 9 mg/mL QID of a ?rst active agent (e.g., Compound—I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 ). In some embodiments a formulation comprising about 10 mg/mL QD of a ?rst active agent (e.g., Compound—I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 , 8 months, or 12 months). The dosage regimen for between 1 and 90 days or for longer than 90 days (e. g., 4 months, 6 months, 8 months, or 12 months) may be any of the regimens involving consecutive or alternate days described in the paragraph above. In some embodiments, the formulation of the present application is administered QD or BID. In some embodiments, the formulation of the present ation is administered QD, BID, TID, or QID when administered at low doses (e.g., 1 mg/mL, 2 mg/mL, 3 mg/mL, 4 mg/mL, or 5 mg/mL), and QD or BID at high doses (e.g., 6 mg/mL, 7 mg/mL, 8 mg/mL, 9 mg/mL, or 10 .
In some embodiments, the formulation of a ?rst active agent (e.g., Formula II or Compound—I) and a second active agent is administered to one eye or both eyes of a subject.
For example, about 0.2% - about 1.0 % (w/v) of the compound of Formula II or about 0.1% — 1.2% (w/v) of Compound-I and a second active agent comprising formulation of the present application is administered once a day (QD), twice a day (BID), three times a day (TID), or four times a day (QID) to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e. g., 4 months, 6 months, 8 months, or 12 months). In some embodiments, Formula 11 compound or Compound—I is complexed with a complexing agent, e.g., cyclodextrin (e. g., hydroxypropyl—B—cyclodextrin (HP—B—CD, SE® HPB) (%)) in ratio of about 1:8, in which about 2% — l3 % (w/v) cyclodextrin (e. g., KLEPTOSE® HPB (%)) is added to the formulation. The formulation may further comprise about 0.1% — about 0.2% buffer, e. g., 10 mM ate buffer. The desired osmolality of the formulation may be about 200 — about 300 mOsm, achieved by adding quantity suf?cient to achieve the osmolality with a salt, e. g., sodium chloride. The pH of the formulation may be about 6.0.
The t application relates to a pharmaceutical composition comprising particles of an active agent of the t ation (e.g., a ?rst active agent (e.g., Formula II or Compound-I) and/or a second active agent), or a pharmaceutically able salt f, wherein the particles have a mean er of between 100 nm and 100 um.
The present application relates to a suspension formulation comprising a pharmaceutical ition, wherein the pharmaceutical composition comprises particles of an active agent of the present application (e.g., a ?rst active agent (e.g., Formula II or Compound—I) and/or a second active agent), or a pharmaceutically acceptable salt thereof, as described herein.
BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 is a series of immunoblots of EGFR orylation in immortalized corneal epithelial cells (hTCEpi cells) treated with varying concentrations of a ?rst active agent of the present application or a control (AG1478, an EGFR kinase inhibitor) and of EGF (top panels: immunoblotting of total EGFR, middle panels: immunoblotting showing phosphorylation of tyrosine 1068 of EGFR, bottom panels: immunoblotting g phosphorylation of tyrosine 1045 of EGFR).
Figure 2 is a series of immunoblots of EGFR phosphorylation in immortalized corneal epithelial cells (hTCEpi cells) treated with varying concentrations of a ?rst active agent of the present ation either alone, or after treatment with vitamin K3 (50 uM), nicotinic acid (10 uM), or nicotinamide (10 uM) (?rst row: immunoblotting of total EGFR, second row: immunoblotting showing phosphorylation of tyrosine 1068 of EGFR, third row: immunoblotting showing phosphorylation of tyrosine 1045 of EGFR).
Figure 3A is a series of raphs showing the migration/proliferation of immortalized corneal epithelial cells (hTCEpi cells) treated for 30 minutes with the indicated concentrations of a ?rst active agent of the present application or a control 8, an EGFR kinase inhibitor), followed by 16 hour ent with the first active agent or control together with the indicated concentrations of EGF or VEGF. Figure 3B is a series of bar graphs quantifying the migration/proliferation of hTCEpi cells in Figure 3A.
Figure 4A is a series of micrographs showing the migration/proliferation of immortalized corneal epithelial cells (hTCEpi cells) d with the ted concentrations of vitamin K3, menadione, for 4 hours, and then supplemented with varying concentrations of a first active agent of the present application, followed by treatment with n K3, the first active agent together with the EGF. Figure 4B is a series of bar graphs quantifying the migration/proliferation of hTCEpi cells in Figure 4A.
Figure 5A is a series of images of epithelial wounds at the time of the initial wounding (0 hr) and post—wounding of corneas treated with vehicle, the indicated concentrations of a compound of Formula I or II, or AG1478. Figures 5B and 5C are a series of bar graphs quantifying the wound healing at 16 hours (Figure 5B) or 24 hours (Figure 5C) post ng of corneas treated with e, the indicated trations of a compound of Formula I or II, or AG1478. Figure 5D is a graph showing the time—course wound healing of s treated with vehicle, the indicated trations of a compound of a I or II, or AG1478.
Figure 6A is a series of images of epithelial wounds at the time of the initial wounding (0 hr) and post—wounding of corneas treated with vehicle, menadione, a compound of Formula I or II, or menadione in combination with a compound of Formula I or 11. Figure 6B is a series of bar graphs quantifying the wound healing at 16 hours or 24 hours post wounding of corneas treated with vehicle, menadione, a compound of Formula I or II, or menadione in combination with a compound of Formula I or 11. Figure 6C is a graph showing the time—course wound healing of corneas d with vehicle, menadione, a compound of Formula I or II, or menadione in combination with a compound of Formula I or Figure 7A is a series of immunoblots showing the total VEGFRZ, phosphorylated VEGFR2, and d—tubulin (as loading control) in human retinal endothelial cells treated with the ted concentrations of menadione for 4 hours, followed by 30 minutes treatment with varying concentrations of a compound of aI or II (1 nM, 10 nM, 100 nM, or 1 uM), and then 10 ng/ml VEGF. Figure 7B is a graph quantifying growth of human retinal endothelial cells following treatment with the indicated concentrations of menadione for 4 hours, followed by 30 minutes treatment with varying trations of a compound of Formula I or II (0.1 nM, 1 nM, 10 nM, or 100 nM), and then 10 ng/ml VEGF overnight.
Figure 8 is a series of immunoblots showing the total EGFRZ, phosphorylated EGFR, and d-Tubulin (as loading control) in hTCEpi cells treated with the indicated concentrations of menadione for 4 hours, followed by incubation with 8.0 nM EGF for the indicated periods of time.
Figure 9A shows the size distribution of the les comprising 5% of a compound of Formula II and 1% Pluronic F—127 produced using small milling media for a long period of time at a high roller speed. Figure 9B shows the size bution of the les comprising 5% of a compound of Formula II and 1% ic F-127 produced using large milling media for a short period of time at a low roller speed. Figure 9C shows the size distribution of the particles comprising 3% of nd-I, 0.6% Tris HCl, and 2% glycerol produced without milling. Figure 9D shows the size distribution of the particles sing 0.4% of a compound of Formula II, 10 uM menadione, 0.08% Pluronic F—127, and 2.5% glycerol after storage at 40 °C for 7 days.
Figure 10 shows the size distribution of the particles comprising 5% of menadione and 1% HPMC.
Figure 11 shows the size distribution of the les comprising 5% of a compound of Formula II and 1% Pluronic F-l27 produced using large milling media for a short period of time at a low roller speed.
DETAILED DESCRIPTION The materials, compounds, compositions, articles, and methods described herein may be understood more readily by nce to the following detailed description of speci?c aspects of the disclosed subject matter and the Examples included therein. Before the present materials, compounds, compositions, articles, devices, and methods are disclosed and described, it is to be understood that the aspects described below are not limited to speci?c methods or speci?c reagents, as such may vary. It is also to be understood that the ology used herein is for the purpose of describing particular aspects only and is not ed to be ng.
Also, throughout this specification, various publications are nced. The disclosures of these publications in their entireties are hereby incorporated by reference into this application in order to more fully describe the state of the art to which the disclosed matter pertains. The references disclosed are also individually and speci?cally incorporated by reference herein for the material contained in them that is discussed in the sentence in which the reference is relied upon.
The t application es compositions or formulations that contain a ?rst active agent and/or a second active agent for use in the treatment of ocular disorders caused by endothelial cell proliferation, enhanced vascular permeability, in?ammation, enesis, or neovascularization, and a second active agent for use in the tion and/or treatment of damage caused to the anterior of the eye by the ?rst active agent, a systemic disease and/or an eye disease.
The present ation also relates to a combination of a ?rst active agent, e. g., a compound of Formula I or II, and a second active agent, e.g., nicotinic acid, nicotinamide, n K, or a combination thereof. In one embodiment, a compound of Formula I or II, or a pharmaceutically acceptable salt thereof, and a second active agent or a pharmaceutically acceptable salt thereof, are administered simultaneously. atively, a compound of Formula I or II, or a pharmaceutically acceptable salt thereof, is administered prior to administration of a second active agent, or a pharmaceutically acceptable salt thereof. In another embodiment, a compound of Formula I or II, or a pharmaceutically acceptable salt thereof, is stered after administration of a second active agent, or a pharmaceutically acceptable salt thereof.
The formulations of the application are useful in preventing or inhibiting neovascularization and vascular leakage ated with ocular disorders while preventing or inhibiting corneal diseases. In some cases, the formulations of the ation cause regression of neovascularization. Brie?y, within the context of the present application, the first active agents should be understood to be any molecule, either synthetic or naturally occurring, which acts to inhibit vascular growth, reduce vascular permeability, and/or decrease ation.
The formulations of the application are also useful in preventing and/or treating corneal epithelium tions caused by systemic diseases (e.g., cancer, diabetes, eta), eye es, or a side effect from a locally or systemically administered drug (e.g., anti—EGFR agents, or compounds of Formula I or II having anti—EGFR activity). Brie?y, within the t of the present application, the second active agents should be understood to be any molecule, either synthetic or naturally ing, which acts to protect from and/or repair corneal edema, tion or any other corneal abnormality. In particular, the t ation provides formulations comprising a ?rst active agent and a second active agent each in a therapeutically ive amount.
General De?nitions In this specification and in the claims that follow, nce is made to a number of terms, which shall be defined to have the following meanings. All percentages, ratios and proportions herein are by weight, unless otherwise ed. All temperatures are in degrees Celsius (C) unless otherwise speci?ed.
By "pharmaceutically able" is meant a material that is not biologically or otherwise undesirable, i.e., the material can be administered to an individual along with the relevant active compound without causing clinically unacceptable biological effects or interacting in a deleterious manner with any of the other components of the pharmaceutical composition or formulation in which it is contained.
A weight percent of a component, unless speci?cally stated to the contrary, is based on the total weight of the formulation or composition in which the component is included.
By "effective amount" as used herein means "an amount of one or more of the disclosed compounds, ive at dosages and for periods of time necessary to e the desired or therapeutic result." An effective amount may vary according to factors known in the art, such as the disease state, age, sex, and weight of the human or animal being treated.
Although particular dosage regimes may be described in examples herein, a person skilled in the art would appreciate that the dosage regime may be altered to provide optimum therapeutic response. For example, several divided doses may be administered daily or the dose may be proportionally reduced as indicated by the exigencies of the therapeutic situation. In addition, the formulations of this disclosure can be administered as frequently as necessary to achieve a therapeutic amount.
The effective amount or effective dose in a human can be determined from that in an animal (e.g., an experimental animal). For example, the effective dose in a human may be calculated based on the conversion shown in the table below.
Conversion of animal doses to human—equivalent doses (HEDs) by using the exponent 0.67 for body surface area animal divide animal dose by animal animal dose by Mouse 312 IO—I.081lm—Iultlply Hamster I—0l35 Ferret I—O.189 Guinea pig I—0.216 Dog I—0541 IMonkey | I—0324 | Squirrel monkey I—0189 Baboon I—0541 Mini-pig I—0.946 "Excipient" is used herein to include any other compound that may be ned in or ed with one or more of the sed inhibitors that is not a therapeutically or biologically active compound. As such, an excipient should be pharmaceutically or biologically acceptable or relevant (for example, an ent should generally be non—toxic to the subject). "Excipient" es a single such compound and is also intended to include a plurality of ents. For the purposes of the present application the term "excipient" and "carrier" are used interchangeably throughout the ption of the present application and said terms are de?ned herein as, "ingredients which are used in the practice of formulating a safe and effective pharmaceutical composition." As used herein, by a "subject" is meant an individual. Thus, the "subject" can include domesticated animals (e.g., cats, dogs, eta), ock (e. g., cattle, horses, pigs, sheep, goats, etc), laboratory animals (e.g., mouse, , rat, guinea pig, etc), and birds.
"Subject" can also include a primate or a human.
By "reduce" or other forms of the word, such as "reducing" or "reduction," is meant lowering of an event or characteristic (e.g., vascular leakage, or tissue swelling). It is understood that this is typically in relation to some standard or expected value, in other words it is relative, but that it is not always necessary for the standard or ve value to be referred The term "treat" or other forms of the word such as "treated" or "treatment" is used herein to mean that stration of a compound or formulation of the present application mitigates a disease or a disorder in a host and/or s, inhibits, or eliminates a particular characteristic or event associated with a disorder (e. g., ar e or corneal ulceration).
Insofar as the methods of the present application are directed to preventing disorders, it is understood that the term "prevent" does not require that the disease state be completely thwarted. Rather, as used herein, the term preventing refers to the ability of the skilled artisan to identify a population that is susceptible to disorders, such that administration of the compounds of the present ation may occur prior to onset of a disease. The term does not imply that the e state be completely avoided.
The term "ameliorating" or other forms of the word such as "ameliorate" is used herein to mean that administration of a therapeutic agent of the present application mitigates one or more symptoms of a disease or a disorder in a host and/or reduces, inhibits, or eliminates a particular symptom associated with the disease or disorder prior to and/or post administration of the therapeutic agent.
The disclosed ?rst active agent compounds affect vascular leakage or pathological neovascularization by ting a or tyrosine kinase. The disclosed second active agent compounds affect corneal epithelium disruptions by modulating (e.g., activating) a receptor tyrosine .
Throughout the description and claims of this specification the word ise" and other forms of the word, such as "comprising" and "comprises," means including but not limited to, and is not intended to exclude, for example, other additives, or components.
As used in the description and the appended claims, the singular forms LL 3’ LL 3’ a an and "the" include plural referents unless the context clearly dictates otherwise.
"Optional" or "optionally" means that the subsequently bed event or circumstance can or cannot occur, and that the description includes instances where the event or circumstance occurs and instances where it does not.
The term "about" refers to any minimal alteration in the concentration or amount of a therapeutic agent (e.g., a ?rst active agent or a second active agent) that does not change the ef?cacy of the agent in ation of a formulation and in treatment of a e or disorder. For example, without being limiting, the concentration of a therapeutic agent would be effective if the concentration is varied between 0.005% to 5.0% (e. g., i0.0005%). The term "about" with t to concentration range of the therapeutic/active agents of the present ation, e. g., first active agent or second active agent, also refers to any variation of a stated amount or range which would be an effective amount or range.
Ranges can be expressed herein as from "about" one particular value, and/or to "about" another particular value. When such a range is expressed, another aspect includes from the one particular value and/or to the other particular value. Similarly, when values are expressed as approximations, by use of the antecedent "about," it is understood that the ular value forms another aspect. It is further understood that the endpoints of each of the ranges are significant both in relation to the other endpoint, and independently of the other nt. It is also understood that there are a number of values disclosed , and that each value is also herein disclosed as "about" that particular value in addition to the value itself. For example, if the value "10" is disclosed, then "about 10" is also disclosed. It is also tood that when a value is disclosed, then "less than or equal to" the value, "greater than or equal to the value," and possible ranges n values are also disclosed, as appropriately understood by the skilled artisan. For example, if the value "10" is disclosed, then "less than or equal to 10" as well as "greater than or equal to 10" is also disclosed. It is also understood that throughout the application data are provided in a number of different formats and that this data represent endpoints and starting points and ranges for any combination of the data points. For example, if a particular data point "10" and a particular data point "15" are sed, it is understood that greater than, greater than or equal to, less than, less than or equal to, and equal to 10 and 15 are considered disclosed as well as between and 15. It is also tood that each unit between two particular units are also disclosed.
For example, if 10 and 15 are disclosed, then 11, 12, 13, and 14 are also disclosed.
The term "halo", as used herein, unless otherwise indicated, includes ?uoro, chloro, bromo or iodo. Preferred halo groups are ?uoro, chloro and bromo.
The term "alkyl," as used herein, unless otherwise indicated, includes both branched and straight-chain saturated aliphatic hydrocarbon groups having the speci?ed number of carbon atoms. For example, C1_6 alkyl is intended to include C1, C2, C3, C4, C5, and C6 alkyl groups. Examples of alkyl include, but are not d to, methyl, ethyl, n- propyl, i-propyl, n-butyl, s-butyl, t-butyl, n—pentyl, s—pentyl, and n—hexyl. In certain embodiments, a straight chain or branched chain alkyl has six or fewer carbon atoms in its backbone (e.g., C1-C6 for straight chain, C3—C6 for branched chain), and in another embodiment, a straight chain or branched chain alkyl has four or fewer carbon atoms.
Likewise, cycloalkyls have from three to eight carbon atoms in their ring structure, and in other embodiments, lkyls have ?ve or six carbons in the ring structure.
Alkyl can be tuted by replacing hydrogen on one or more carbons of the hydrocarbon backbone. Such substituents can include, for example, halogen, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylthiocarbonyl, l, phosphate, phosphonato, phosphinato, cyano, amino (including alkylamino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, io, thiocarboxylate, sulfates, alkylsul?nyl, sulfonato, sulfamoyl, sulfonamido, nitro, tri?uoromethyl, cyano, azido, cyclyl, alkylaryl, or an ic or heteroaromatic moiety. Cycloalkyls can be further tuted, e.g., with the substituents described above and onal substituents such as alkyl, alkenyl, and l. An "alkylaryl" or an "aralkyl" moiety is an alkyl tuted with an aryl (e.g., phenylmethyl (benzyl)).
The term "alkenyl," as used herein, unless otherwise indicated, includes unsaturated aliphatic groups analogous in length and possible tution to the alkyls described above, but that contain at least one double bond. For example, the term "alkenyl" includes straight-chain alkenyl groups (e.g., ethenyl, propenyl, butenyl, pentenyl, hexenyl, heptenyl, octenyl, nonenyl, decenyl), branched-chain alkenyl groups, cycloalkenyl (e.g., alicyclic) groups (e. g., ropenyl, cyclopentenyl, cyclohexenyl, cycloheptenyl, cyclooctenyl), alkyl or alkenyl substituted cycloalkenyl groups, and cycloalkyl or cycloalkenyl substituted alkenyl groups. In certain embodiments, a straight chain or ed chain alkenyl group has six or fewer carbon atoms in its backbone (e. g., C2—C6 for straight chain, C3-C6 for branched chain). se, lkenyl groups may have from three to eight carbon atoms in their ring structure, and in some embodiments, cycloalkenyl groups have ?ve or six carbons in the ring structure. The term "C2—C6" includes alkenyl groups containing two to six carbon atoms. The term "C3-C6" includes alkenyl groups containing three to six carbon atoms. Alkenyl can be substituted by replacing hydrogen on one or more arbon backbone carbon atoms. Such substituents can include, for e, alkynyl groups, ns, hydroxyl, arbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, arbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, cyano, amino (including alkylamino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate, sulfates, alkylsul?nyl, sulfonato, sulfamoyl, sulfonamido, nitro, tri?uoromethyl, cyano, azido, heterocyclyl, alkylaryl, or an aromatic or heteroaromatic moiety.
The term "alkynyl", as used herein, unless otherwise indicated, includes unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but which n at least one triple bond. For example, "alkynyl" includes straight-chain alkynyl groups (e. g., ethynyl, propynyl, butynyl, pentynyl, hexynyl, heptynyl, octynyl, nonynyl, decynyl), branched-chain alkynyl groups, and cycloalkyl or cycloalkenyl substituted alkynyl groups. In certain ments, a straight chain or branched chain alkynyl group has six or fewer carbon atoms in its backbone (e. g., C2-C6 for straight chain, C3-C6 for branched chain). The term " includes alkynyl groups containing two to six carbon atoms. The term " includes alkynyl groups ning three to six carbon atoms. Alkynyl can be substituted by replacing hydrogen on one or more hydrocarbon backbone carbon atoms. Such substituents can e, for example, halogens, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy, carbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, minocarbonyl, dialkylaminocarbonyl, alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, cyano, amino (including alkylamino, lamino, arylamino, amino, and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate, sulfates, alkylsulfinyl, sulfonato, sulfamoyl, sulfonamido, nitro, romethyl, cyano, azido, heterocyclyl, alkylaryl, or an aromatic or heteroaromatic moiety.
The term "alkoxy," as used herein, unless otherwise indicated, includes l groups wherein "alkyl" is as de?ned above.
The term "aryl," as used herein, unless otherwise indicated, includes 5— and 6— membered "unconjugated", or single—ring, aromatic groups, as well as "conjugated", or multicyclic, systems with at least one aromatic ring. Examples of aryl groups e benzene, , and the like. Furthermore, the term "aryl" includes yclic aryl groups, e. g., tricyclic, bicyclic, e.g., naphthalene.
Aryl groups having heteroatoms in the ring structure may be referred to as "aromatic heterocycles", "aryl heterocycles", "heterocycles," "heteroaryls" or oaromatics".
The aryl or heteroaryl can be substituted at one or more ring positions with such substituents as described above, as for example, halogen, hydroxyl, alkoxy, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, ycarbonyloxy, carboxylate, alkylcarbonyl, alkylaminocarbonyl, aralkylaminocarbonyl, alkenylaminocarbonyl, alkylcarbonyl, rbonyl, aralkylcarbonyl, alkenylcarbonyl, alkoxycarbonyl, arbonyl, alkylthiocarbonyl, phosphate, onato, phosphinato, cyano, amino (including alkylamino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino (including arbonylamino, rbonylamino, carbamoyl and ureido), amidino, imino, dryl, alkylthio, arylthio, thiocarboxylate, sulfates, alkylsul?nyl, sulfonato, sulfamoyl, sulfonamido, nitro, tri?uoromethyl, cyano, azido, heterocyclyl, alkylaryl, or an aromatic or heteroaromatic moiety. Aryl groups can also be fused or bridged with alicyclic or heterocyclic rings, which are not aromatic so as to form a yclic system (e. g. , tetralin, methylenedioxyphenyl).
The term "4—10 membered heterocyclic," as used herein, unless otherwise indicated, includes aromatic and non-aromatic heterocyclic groups containing one or more heteroatoms each selected from O, S and N, wherein each heterocyclic group has from 4—10 atoms in its ring system. Non-aromatic heterocyclic groups include groups having only 4 atoms in their ring , but aromatic heterocyclic groups must have at least 5 atoms in their ring system. An example of a 4 membered heterocyclic group is azetidinyl (derived from azetidine). An example of a 5 membered heterocyclic group is thiazolyl and an example of a 10 membered heterocyclic group is quinolinyl.
Examples of non-aromatic heterocyclic groups are pyrrolidinyl, tetrahydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, tetrahydrothiopyranyl, piperidino, lino, thiomorpholino, thioxanyl, piperazinyl, azetidinyl, oxetanyl, thietanyl, homopiperidinyl, oxepanyl, nyl, oxazepinyl, diazepinyl, thiazepinyl, l,2,3,6-tetrahydropyridinyl, 2- pyrrolinyl, 3-pyrrolinyl, indolinyl, 2H—pyranyl, anyl, dioxanyl, 1,3-dioxolanyl, pyrazolinyl, nyl, dithiolanyl, dihydropyranyl, dihydrothienyl, dihydrofuranyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, 3—azabicyclo[3.1.0]hexanyl, 3— azabicyclo[4.1.0]heptanyl, 3H-indolyl and quinolizinyl.
Examples of aromatic heterocyclic groups are pyridinyl, imidazolyl, pyrimidinyl, pyrazolyl, triazolyl, pyrazinyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxazolyl, isothiazolyl, pyrrolyl, quinolinyl, isoquinolinyl, indolyl, idazolyl, benzofuranyl, cinnolinyl, indazolyl, indolizinyl, phthalazinyl, pyridazinyl, triazinyl, isoindolyl, pteridinyl, purinyl, zolyl, thiadiazolyl, furazanyl, benzofurazanyl, benzothiophenyl, benzothiazolyl, benzoxazolyl, quinazolinyl, alinyl, naphthyridinyl, and furopyridinyl.
The foregoing groups, as derived from the compounds listed above, may be C— attached or N—attached where such is possible. For instance, a group d from pyrrole may be pyrrol-l-yl (N—attached) or pyrrol—3 —yl (C—attached). The "4— 10 membered cyclic" moiety can be tuted.
The phrase "pharmaceutically acceptable salt(s)," as used herein, unless otherwise indicated, includes salts of acidic or basic groups which may be present in the compounds of ?rst active agents (e.g., FormulaI or II) or second active agents. The compounds of Formula I or II and the second active agents that are basic in nature are capable of forming a wide y of salts with s inorganic and organic acids. The acids that may be used to prepare pharmaceutically acceptable acid addition salts of such basic compounds of Formula I or II and second active agents are those that form xic acid addition salts, i.e., salts containing pharmacologically acceptable anions, such as the hydrochloride, hydrobromide, hydroiodide, nitrate, sulfate, bisulfate, phosphate, acid phosphate, isonicotinate, acetate, lactate, salicylate, citrate, acid citrate, tartrate, pantothenate, bitartrate, ate, succinate, e, gentisinate, te, gluconate, glucaronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate and pamoate [i.e., 1,1'—methylene—bis-(2—hydroxy—3—naphthoate)] salts.
Those nds of ?rst active agents (e.g., FormulaI or II) and second active agents that are acidic in nature are capable of forming base salts with s pharmacologically acceptable cations. Examples of such salts include the alkali metal or alkaline earth metal salts and particularly, the sodium and potassium salts.
In some ments, the salt is an acid addition salt, e. g. HCl salt.
Certain compounds of Formula I or II and certain second active agents may have asymmetric centers and therefore exist in different enantiomeric forms. This application relates to the use of all optical isomers and stereoisomers of the compounds of Formula I or II and mixtures f and of the second active agents and mixtures thereof. The compounds of Formula I or II and the second active agents may also exist as E/Z geometric isomers or tautomers. This application relates to the use of all such ric isomers and tautomers and es thereof.
The subject application also includes isotopically—labeled compounds, and the pharmaceutically acceptable salts thereof, which are identical to those recited in Formula I or II, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. es of isotopes that can be incorporated into nds of the application include isotopes of hydrogen, , nitrogen, oxygen, phosphorous, ?uorine and chlorine, such as 2H, 3 13 H, C, 14C, 15N, 18O, 17O, 358, 18F, and 36C1, tively. Compounds of the FormulaI or II, conjugates thereof, and pharmaceutically acceptable salts of said compounds or of said conjugates which contain the aforementioned isotopes and/or other isotopes of other atoms are within the scope of this application. Certain isotopically—labeled compounds of the present application (e. g., compounds of Formula I or II), for example those into which radioactive isotopes such as 3H and 14C are incorporated, are useful in drug and/or substrate tissue distribution assays. Tritiated, 3 i.e., H, and carbon—14, i.e., 14C isotopes are particularly preferred for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium, i.e., 2H, can afford certain eutic advantages resulting from greater metabolic stability, for example increased in vivo half—life or reduced dosage requirements and, hence, may be red in some circumstances. Isotopically d compounds of Formula I or II of this application and esters or lipid conjugates thereof can generally be prepared by carrying out the procedures disclosed in the Schemes and/or in the Examples and Preparations below, by substituting a y available isotopically labeled reagent for a non-isotopically labeled reagent.
This application also encompasses pharmaceutical formulations containing derivatives of compounds of the Formula I or II, or pharmaceutically able salts thereof and of the second active agents, or pharmaceutically acceptable salts thereof. Compounds of Formula I or II, or pharmaceutically acceptable salts thereof, and second active agents, or pharmaceutically acceptable salts f, having free amino, or amido groups can be converted into conjugated derivatives, wherein an amino acid residue, or a polypeptide chain of two or more (e.g., two, three or four) amino acid residues is covalently joined through an amide or ester bond to a free amino group of compounds of a I or II, or ceutically acceptable salts thereof, or of second active agents or pharmaceutically acceptable salts thereof. The amino acid residues include, but are not limited to, the 20 naturally occurring amino acids commonly ated by three letter symbols and also includes 4-hydroxyproline, hydroxylysine, demosine, osine, 3-methylhistidine, norvalin, beta-alanine, gamma-aminobutyric acid, citrulline homocysteine, homoserine, ornithine and methionine e.
Additional types of derivatives are also encompassed. Amide and ester moieties may incorporate groups including but not limited to ether, amine and carboxylic acid functionalities. Free hydroxy groups may be derivatized using groups including but not limited to hemisuccinates, phosphate esters, dimethylaminoacetates, and phosphoryloxymethyloxycarbonyls, as outlined in D. Fleisher, et al., ADVANCED DRUG DELIVERY REVIEWS (1996) 19, 115. Carbamate conjugates of hydroxy and amino groups are also included, as are carbonate conjugates and sulfate esters of hydroxy groups. tization of hydroxy groups as (acyloxy)methyl and (acyloxy)ethyl ethers n the acyl group may be an alkyl ester, optionally substituted with groups including but not limited to ether, amine and carboxylic acid functionalities, or where the acyl group is an amino acid ester as described above, are also encompassed. Derivatives of this type are bed in R.
P. Robinson et al., J. MEDICINAL CHEMISTRY (1996) 39, 10.
] The term "kinase" refers to any enzyme that catalyzes the addition of phosphate groups to a protein residue; for example, serine and threonine kinases catalyze the addition of phosphate groups to serine and threonine residues.
The terms "VEGFR kinase," and "VEGFR," refer to any of the vascular endothelial growth factor ors.
The terms "VEGF signaling," and "VEGF cascade" refer to both the am and downstream components of the VEGF ing cascade.
The terms "ErbB kinase," and "ErbB receptor," refer to any member of the ErbB family of receptor ne kinases including EGFR (ErbBl or HERI), HERZ/c—neu (ErbBZ), HER3 (ErbB3) and HER4 (ErbB4).
The terms "EGF signaling," and "EGF cascade" refer to both the upstream and downstream components of the EGF signaling cascade.
The term aceutically acceptable" refers to the fact that the carrier, diluent or excipient must be compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
] The terms "administration of a nd" or "administering a compound" refer to the act of providing a compound of the application or pharmaceutical formulation to the subject in need of treatment.
The term "vasculostasis" refers to the maintenance of the homeostatic vascular functioning leading to the normal physiologic functioning.
The term "vasculostatic agents" refers to agents that seek to address ions in which vasculostasis is compromised by preventing the loss of or restoring or maintaining vasculostasis.
In the t application "composition" and "formulation" are used interchangeably and refer to the conventional understanding, as known in the art, of a composition or formulation.
The present application relates to an ophthalmic formulation. In some embodiments, the ophthalmic formulation of the present application is a gel formulation or a semi-gel formulation, or both.
"Gel" according to the present application is a semi-solid dosage form of the t application, ning suspended particles. A semisolid is not pourable; it does not flow or conform to its ner at room temperature. A semisolid does not ?ow at low shear stress and generally exhibits plastic ?ow behavior. A dal dispersion is a system in which particles of colloidal dimension (i.e., typically between 1 nm and l um) are distributed uniformly throughout a liquid.
In some embodiments, "gel" is a semisolid system consisting either of suspensions of small inorganic particles or of organic molecules interpenetrated by a liquid. "Gels" are classed either as single—phase or two—phase systems. "Gels" also consist of a ase, or state of matter intermediate between a liquid and a solid that represents a partially ordered structure, which is the state for the active agents in the "Gel Drop" of the present embodiments. A two—phase gel consists of a network of small te particles. In a two— phase , the gel mass sometimes is referred to as magma (e.g., Bentonite Magma) if the particle size of the suspended material is large. Both gels and magmas are thixotropic, forming lids on standing and becoming liquid on agitation. The semisolid ations should be shaken before administration to ensure homogeneity and should be so labeled (see Suspensions). Single—phase gels consist of organic macromolecules uniformly distributed throughout a liquid in such a manner that no apparent boundaries exist between the dispersed macromolecules and the . Single phase gels may also t of organic low molecular weight (LMW) molecules where the component responsible for gelation is the actual active ingredient. These so called "LMW hydrogels" are different from ional gelators of water such as high molecular weight synthetic polymers, polysaccharides, and proteins. High molecular weight gelators are highly ordered and uni-directional due to hydrogen bonding whereas the forces governing LMW hydrogels are largely non-directional van der Waals forces (hydrophobic) interactions. In practice LMW hydrogels are observed as highly anisotropic (typically ?brillar) structures that propagate throughout the liquid yielding a physically branched or led network. The gels can thus be non—ordered to slightly ordered showing some birefringence, liquid l character. Gels are administered topically or, after shaking, in the form of a el as an eye drop.
The lid "gel" according to the present application is a semisolid per USP definitions and literature referenced therein. The semisolid formulation apparent viscosity increases with tration. The al dosage strength of the t ation ranges from a low strength of 51 mg/mL (0.1%) to a high strength of :6 mg/mL (0.6%). Low th doses are least viscous and fall under the category of a "solution," whereas higher strengths are more viscous and fit the de?nition of a gel.
"Jelly" according to the present application is a class of gels, which are semisolid systems that consist of suspensions made up either small inorganic particles or large organic molecules interpenetrated by a liquid, in which the structural coherent matrix contains a high portion of liquid, y water.
"Solution" according to the present application is a clear, neous liquid dosage form that contains one or more chemical substances dissolved in a solvent or mixture of ly miscible ts. A solution is a liquid preparation that ns one or more dissolved chemical substances in a suitable solvent or mixture of mutually le solvents.
Because molecules of a drug substance in solution are uniformly dispersed, the use of solutions as dosage forms generally provides assurance of uniform dosage upon administration and good accuracy when the solution is diluted or otherwise mixed.
"Liquid" according to the present application is a dosage form consisting of a pure chemical in its liquid state. A liquid is pourable; it ?ows and conforms to its container at room temperature. Liquids display Newtonian or pseudoplastic ?ow behavior.
"Suspension" according to the present application is a liquid dosage form that ns solid particles dispersed in a liquid vehicle.
The compounds of first active agents (e.g, Formulal or H) and the second active agents are formulated into therapeutic formulations as natural or salt forms.
Pharmaceutically acceptable non-toxic salts include the base addition salts (formed with free carboxyl or other anionic groups) which are derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, 2—ethylamino—ethanol, histidine, procaine, and the like. Such salts are formed as acid on salts with any free cationic groups and generally are formed with inorganic acids such as, for e, hydrochloric, sulfuric, or phosphoric acids, or organic acids such as acetic, citric, p—toluenesulfonic, esulfonic acid, oxalic, tartaric, mandelic, and the like. Salts of the application include amine salts formed by the protonation of an amino group with inorganic acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, phosphoric acid, and the like. Salts of the application also include amine salts formed by the protonation of an amino group with suitable organic acids, such as p—toluenesulfonic acid, acetic acid, and the like. onal excipients which are contemplated for use in the practice of the present application are those available to those of ordinary skill in the art, for e, those found in the United States Pharmacopoeia Vol.
XXII and National Formulary Vol. XVII, U.S. Pharmacopoeia Convention, Inc., RockVille, Md. (1989), the relevant contents h is incorporated herein by nce. In addition, rphs of the application compounds are included in the present application.
The embodiments of the present application provide an ophthalmic composition or formulation for treating ocular neovascularization with a first active agent of Formula I: o :ssII, NWS 0 or a ceutically acceptable salt thereof; a second active agent or a pharmaceutically acceptable salt thereof, wherein the second active agent is nicotinic acid, nicotinamide, or vitamin K, or a combination thereof; and pharmaceutically acceptable excipients; the ?rst active agent or the pharmaceutically able salt is present in about 0.02% to about 1.2% w/v, wherein: X O or S; R1 is H, C1-C10 alkyl, C2 -C10alkenyl, C2 -C10alkynyl, C(O)(C1 -C10 , (CH2)I(C6 - C10 aryl), (4— 10 membered heterocyclic), C(O)(CH2)1(C6 -C10 aryl), or C(O)(CH2)t (5—10 membered heterocyclic), wherein: t is an integer from 0 to 5; the alkyl group optionally includes 1 or 2 hetero es selected from O, S and N(R6) with the proviso that two 0 atoms, two S atoms, or an O and an S atoms are not attached directly to each other; the aryl and heterocyclic groups are optionally fused with a C6 -C10 aryl group, a C5 —C8 saturated cyclic group, or a 5—10 membered heterocyclic group; 1 or 2 carbon atoms in the foregoing heterocyclic moieties are optionally substituted with an oxo (=0) moiety or an anion of oxygen; the (CH2) moieties optionally include a carbon—carbon double or triple bond when t is an integer from 2 to 5; and the foregoing R1 groups, except H, are optionally substituted with 1 to 3 R4 groups; R2 is H; R3 is (CH2)t(C6 -C10 aryl), wherein: t is an integer from 0 to 5; the aryl group is optionally fused with a C6 -C10 aryl group, a C5 —Cg saturated cyclic group, or a 5—10 membered cyclic group; the (CH2), moieties optionally include a carbon—carbon double or triple bond when t is an integer from 2 to 5; and each R4 is independently selected from C1—C10 alkyl, C2 -C10 l, C2 -C10 alkynyl, halo, cyano, nitro, tri?uoromethyl, tri?uoromethoxy, azido, 0R5, C(O)R5, C(O)OR5, NR6C(O)R5, NR6C(O)OR5, OC(O)R5, NR6S02R5, SOZNR5R6, C(O)NR5R6, NR5R6, S(O)jR7 where j is an integer from o to 2, SO3H, NR5(CR6R7)tOR6, (CH2)t(C6 -C10 aryl), SOz(CH2)t(C6 —C10 aryl), S(CH2)t(C6 —C10 aryl), O(CH2)t(C6 —C10 aryl), (CH2)t(5—10 ed heterocyclic), and (CR6 R7)mOR6, wherein: m is an integer from 1 to 5; t is an r from 0 to 5; the alkyl group optionally includes 1 or 2 hetero moieties ed from O, S and N(R6) with the proviso that two 0 atoms, two S atoms, or an O and an S atoms are not attached directly to each other; the aryl and heterocyclic groups are optionally fused with a C6 -C10 aryl group, a C5 —C8 saturated cyclic group, or a 5—10 membered heterocyclic group; 1 or 2 carbon atoms in the foregoing heterocyclic moieties are ally substituted with an oxo (=0) moiety or an anion of oxygen; and the alkyl, aryl and heterocyclic moieties of the foregoing R4 groups are optionally substituted with l to 3 substituents independently selected from halo, cyano, nitro, tri?uoromethyl, tri?uoromethoxy, azido, R5, SOZNR5 R6, C(O)R5, C(O)OR5, OC(O)R5, NR6C(O)R5, C(O)NR5R6, NR5R6, (0116117)",0116 where m is an integer from 1 to 5, 0R5, and the substituents listed in the de?nition of R5 ; R5, R6, and R7 are each ndently H or C1 —C6 alkyl.
In one embodiment, R3 is (CH2)t(C6 -C10ary1), n t is an integer from 1 to 3 and R3 is optionally substituted with l to 4 R4 groups.
In a further embodiment, R3 is benzyl, ally substituted with 1 to 4 substituents independently selected from halo and C1 —C4 alkyl. In a further embodiment, R3 is benzyl substituted with l to 4 substituents independently selected from methyl, ?uoro, chloro and bromo.
In one embodiment, R1 is (5—10 membered heterocyclic), wherein t is an integer from 0 to 5, optionally substituted with 1 or 2 substituents independently ed from C1 -C4 alkyl, y and ymethyl.
The present application provides heterocyclic moiety of the R1 group in Formula I selected from morpholino, pyrrolidinyl, imidazolyl, zinyl, piperidinyl, and 2,5-diaza— bicyclo[2.2. l]hept—2-yl, the t variable of the R1 group ranges from 2 to 5, and the R1 group is optionally substituted with one or more hydroxy groups.
For example, the heterocyclic moiety of the R1 group in Formula I of the present application is pyrrolidine.
In further embodiments of the present application, the ?rst active agent is: Br or r 0 N’s O F (11).
A compound of the present application is 3-[(4-bromo-2,6- di?uorophenyl)methoxy] -5 -[[[[4-(l-pyrrolidinyl)buty1]amino]carbony1]amino] isothiazolecarboxamide hydrochloride, of molecular formula: C20H24BrF2N503S - HCl, molecular weight: 568.86 g/mol, and with the property that the molecule does not contain an asymmetric center and is not chiral. A compound of the t application is represented by Compound—I: (Compound—I).
The Compound-I of the present application is an inhibitor of the ne kinase activity of VEGFR—2, which blocks VEGF—stimulated auto—phosphorylation of this receptor as well as elial cell proliferation. It is selective (>500x) relative to the concentration required to inhibit the epidermal growth factor receptor (EGFR) and the n receptor (IR) tyrosine kinases. Compound-I is bed in US Patent No 6,235,764. In some embodiments, the compounds of Formula I or II are VEGFR—2 inhibitors.
The second active agents of the t application are EGFR modulators (e.g., activators) of the tyrosine kinase activity of EGFR.
The ?rst active agent and the second active agent can be administered together as part of the same formulation comprising the ?rst active agent, the second active agent, and a pharmaceutical excipient. The ?rst active agent and the second active agent can also be administered separately. In one embodiment, the ?rst active agent and the second active agent are administered separately as two formulations, wherein one formulation comprises the first active agent and a ceutical excipient, and the second formulation ses the second active agent and a pharmaceutical ent.
] In one embodiment, a compound of a I or II, or a ceutically acceptable salt thereof, and the second active agent or a pharmaceutically acceptable salt thereof, are administered simultaneously. Alternatively, a compound of Formula I or II, or a pharmaceutically acceptable salt thereof, is administered prior to administration of the second active agent, or a pharmaceutically acceptable salt thereof. In another embodiment, a compound of Formula I or II, or a pharmaceutically acceptable salt thereof, is administered after administration of the second active agent, or a pharmaceutically acceptable salt thereof.
General Properties Compound-I of the present application has the characteristics as shown in Table 1.
The embodiments provide three formulations of Compound-I or its free base — the Formula II compound.
Table 1A: General Properties of Compound—I Drug Substance Chemical Name [CAS No] 3—[(4—bromo—2,6-di?uorophenyl)methoxy][[[[4-( 1 - Codes: Compound-I pyrrolidinyl)butyl]amino]carbonyl]amino] isothiazolecarboxamide h drochloride 2520037 A. oearance color, oh sical form White 0 stalline solid ; rane 222.2 —224.8 °C .Ka water Methanol: 4.3 Ethanol: 0.7 itrile: 0.04 Tetrahydrofuran: 0.02 Hexanes: < 0.01 Solubility (mg/mL) 0.1 N NaOH: 0.05 pH 9.0 (0.05 m NazHPO4): 0.7 pH 7.5 (0.05 M 4): 1.0 0.1 N HCl: 0.04 Deionized water 0.5-1.2b The composition of Compound—I formulations are listed in Table 1B. The formulation materials are listed in Table 1C.
Table 18: Compound—I formulations: Gel Drop, suspension, and solution.
Formulation Form Ophthalmic gel drops 0.05% Sodium Phosphate, Monobasic, Monohydrate 1.0-2.0% Glycerin with or without 0.005% Benzalkonium Chloride, NF (BAK) pH ~6.0-7.0 Tris-based suspensions 0.6% Tromethamine, USP (Tris) 1.0-2.0% Glycerin, USP with or without 0.005% Benzalkonium Chloride, NF (BAK) pH ~6.0-7.0 Cyclodextrin-based 1% to 20% ypropyl-B-cyclodextrin (HP-B-CD, ons KLEPTOSE® HPB) 0.1% to 0.9% sodium de pH ~6.0—7‘.0 1% to 20% sulfobutylether-B-cyclodextrin (SBE-B-CD, CAPTISOL®) with or without 0.122% Tromethamine (Tris) 0.1-0.2% sodium phosphate, dibasic, anhydrous 0%—0.6% sodium chloride pH ~6.0—7.0 Formulation Forms 1% to 20% HP—B—CD OSE® HPB or KLEPTOSE® 0.1- 0.2% sodium phosphate, dibasic, anhydrous 0.50%—0.6% sodium chloride pH ~6.0—7.0 Table 1C: Formulation materials Function Compound-I Active Substance Sodium Chloride Tonicity Modi?er Sulfobutyl ether-B-cyclodextrin Solubilizing SOL®, S?ECD) agents oxypropyl-B-cyclodextrin (KLEPTOSE® HPB Parenteral Grade, Trometamol (Tris) Buffer Dibasic ohos ohate buffer 2.0 N NaOH Adjust pH 0.1 N HCl The composition of the Compound—I formulations and a second active are listed in Table 1D. The formulation als are listed in Table 1E.
Table 1D: Compound—l formulations: Gel Drop, suspension, and solution.
Formulation Forms lmic gel drops 0.05% Sodium Phosphate, Monobasrc, Monohydrate 1.0-2.0% Glycerin with or without 0.005% Benzalkonium Chloride, NF (BAK) pH ~6.0-7.0 Tris-based suspensions 0.6% Tromethamine, USP (Tris) 1.0-2.0% Glycerin, USP with or without 0.005% Benzalkonium Chloride, NF (BAK) pH ~6.0—7.0 Cyclodextrin-based 1% to 20% hydroxypropyl-[3-cyclodextrin (HP-B-CD, KLEPTOSE® solutions HPB) 0.1% to 0.9% sodium chloride pH ~6.0-7.0 1% to 20% sulfobutylether-[3-cyclodextrin (SBE-B-CD, OL®) with or without 0.122% Tromethamine (Tris) 0.1—0.2% sodium phosphate, dibasic, anhydrous 0%-0.6% sodium chloride pH ~6.0-7.0 1% to 20% HP—E—CD {KLEPTOSE® HPB 0r KLEPTOSE® HP! Formulation Forms 0.1- 0.2% sodium phosphate, dibasic, anhydrous 0.6% sodium chloride pH ~6.0-7.0 Table 1E: Formulation materials Material Function Como ound-I Active Dru - Substance Second active aent Active Dru_ nce Sodium Chloride Tonicit r Sulfobutyl ether-B-cyclodextrin Solubilizing agents CAPTISOL®, S BECD 2-hydr0xypropyl-B-cyclodextrin (KLEPTOSE® HPB Parenteral Grade, HP 3 CD Dibasic ohos ohate buffer Ophthalmic Solutions The present application provides formulations of a ?rst active agent (e. g., Compound—l and/or its free base (Formula 11 compound» and/or a second active agent, formed as a solution with viscosity similar to water. The solution includes pharmaceutically able agents/excipients, for example, without being limiting, cyclodextrin. The solution thus formed is clear and colorless on, suitable for topical administration to the eye.
The solutions of the t application reduce or segment exposure of the ?rst active agent; thereby they allow increased concentration of the ?rst active agent, e. g., a compound of Formula I or II, in the solution and sed frequency of delivery, thus, promoting maintained high concentration of the ?rst active agent in the posterior segment of the eye.
The solutions of the application comprise about 0.005% to about 5.0% w/v of the ?rst active agent of Formula I or II, or a pharmaceutically acceptable salt thereof, for example, Compound-I. In some embodiments, the concentration of Compound-I or its free base (Formula II) in the solutions is about 0.005% - about 0.01%, about 0.01% - about 0.05%, about 0.05% — about 0.1%, about 0.1% — about 0.2%, about 0.2% - about 0.3%, about 0.3% — about 0.4%, about 0.4% — about 0.5%, about 0.5% — about 0.6%, about 0.6% — about 0.7%, about 0.7% — about 0.8%, about 0.8% — about 0.9%, about 0.9% — about 1.0%, about 1.0 — about 2.0%, about 2.0 — about 3.0%, about 3.0 — about 4.0%, or about 4.0 — about 5.0% w/v for l administration. In some embodiments, the concentration of Compound-I or its free base (Formula II) in the formulations is about 0.1% — about 1.2%, about 0.2% — about 1.2%, about 0.3% — about 1.2%, about 0.4% — about 1.2%, 0.1% — about 1.1%, about 0.2% — about 1.1%, about 0.3% — about 1.1%, about 0.4% — about 1.1%, 0.1% — about 1.0%, about 0.2% — about 1.0%, about 0.3% - about 1.0%, about 0.4% — about 1.0%, 0.1% — about 0.8%, about 0.2% — about 0.8%, about 0.3% — about 0.8%, about 0.4% — about 0.8%, 0.1% — about 0.6%, about 0.2% - about 0.6%, about 0.3% — about 0.6%, about 0.4% — about 0.6%, 0.1% - about 0.5%, about 0.2% - about 0.5%, about 0.3% - about 0.5%, about 0.4% - about 0.5%, 0.1% - about 0.4%, about 0.2% - about 0.4%, about 0.3% - about 0.4% W/v for topical stration. In some embodiments, the solutions include about 0.005%, about 0.05%, about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1.0%, about 2.0%, about 3.0%, about 4.0%, or about 5.0% w/v of Compound—I or its free base (Formula II).
The solutions of the ation may further comprise about 1% — about .0% W/v of a second active agent, or a ceutically acceptable salt thereof, for example, nic acid, nicotinamide, or vitamin K, or a combination thereof. The solutions of the application may further comprise is about 0.00001% — about 1.0%, about 0.00001% — about 0.1%, about 0.00001% - about 0.01%, about 0.00001% - about 0.001%, about 0.00001% - about 0.0002%, or about 1% — about 0.0001% w/v of a second active agent, or a ceutically acceptable salt thereof, for example, nicotinic acid, nicotinamide, or vitamin K, or a combination thereof. In some embodiments, the concentration of the second active agent in the solutions is about 0.00001% — about 0.0001%, 0.000012% — about 0.0001%, 0.000014% - about %, 0.000016% — about 0.0001%, 0.000018% - about 0.0001%, 0.00002% - about 0.0001%, 0.00003% — about 0.0001%, 0.00004% - about 0.0001%, 0.00005% - about 0.0001%, 6% — about 0.0001%, 0.00007% - about 0.0001%, 0.00008% - about 0.0001%, 0.00009% — about 0.0001%, 0.000016% - about 0.00009%, 0.000018% - about 0.00009%, 0.00002% — about 0.00009%, 0.00003% - about 0.00009%, 0.00004% - about 0.00009%, 0.00005% - about 0.00009%, 0.00006% - about 0.00009%, 7% - about 0.00009%, 0.00008% - about 0.00009% W/v for l administration. In some embodiments, the solutions include about 0.00001%, 0.00002%, 0.00003%, 0.00004%, 0.00005%, 0.00006%, 0.0000?%, 0.00008%, 0.000081%, 0.000082%, 0.000083%, 0.000084%, 0.000085%, 0.000086%, 0.00008?%, 0.000088%, or 0.000089% W/v of the second active agent.
The solutions of the application may further comprise about 0.5 uM, about 0.6 uM, about 0.7 uM, about 0.8 uM, about 0.9 pM, about 1 uM, about 2 uM, about 3 uM, about 4 uM, about 5 uM, about 6 uM, about 7 uM, about 8 "M, or about 9 [1M of a second active agent, or a pharmaceutically acceptable salt thereof, for example, nic acid, nicotinamide, or vitamin K, or a combination thereof. In some ments, the concentration of the second active agent is about 1 "M.
In some embodiments, the formulation comprises cyclodextrin for improving solubility of a first active agent (e. g., Compound-I). Cyclodextrin, an accharide made up of six to eight dextrose units joined through one or four bonds increases solubility of active agents that have poor or low solubility in water or s solutions (e. g., in PBS buffer). Cyclodextrins form hydrophilic complexes with hydrophobic active agents.
One or more cyclodextrins may be used in the solution of the present application.
Non-limiting examples of cyclodextrins for use in formulation of the present application are, for example: 2-hydroxypropyl-B-cyclodextrin, methyl—B—cyclodextrin, randomly ated- B-cyclodextrin, ethylated-B-cyclodextrin, triacetyl—B—cyclodextrin, peracetylated-B- extrin, carboxymethyl-B-cyclodextrin, hydroxyethyl -B-cyclodextrin, 2-hydroxy (trimethylammonio)propyl-B-cyclodextrin, glucosyl —B—cyclodextrin, maltosyl-B-cyclodextrin, sulfobutyl ether—B—cyclodextrin, branched—B—cyclodextrin, hydroxypropyl-y-cyclodextrin, randomly methylated-y-cyclodextrin, trimethyl—y—cyclodextrin, or a combination thereof.
In some ments, the solution of Formula II compound or Compound—I comprising cyclodextrin is a clear and colorless solution and has a viscosity similar to water.
In some ments, the t application provides a solution comprising Compound-I, one or more cyclodextrin, and a second active agent for topical application and is topically applied to the eye.
The ophthalmic solution of the present application comprises cyclodextrin and pharmaceutical excipients chosen at or below concentrations optimal for ophthalmic solution.
The excipients of the present ation are, for e, benzalkonium chloride (BAK) and NaCl. In some embodiments, the ophthalmic solution comprises about 0.001 — about 0.005% w/v Benzalkonium chloride (BAK). The BAK amount varies depending on the need of the application.
] The ophthalmic solution comprises, for example, t being limiting, about 0.005% — 5.0% Compound—I or its free base, about 2 — about 25% cyclodextrin, e.g., t being limiting, Hydroxypropyl-B—cyclodextrin (HPBCD) or methylcyclodextrin (KLEPTOSE® HPB), and/or sulfobutyl ether—B—cyclodextrin (CAPTISOL®), about 0.1 — about 0.7% salt, e. g., without being limiting, NaCl, and/or about 0.005% of an anti-microbial agent, for example, without being limiting, Benzalkonium de (BAK). The formulation comprises Compound—I or its free base to cyclodextrin ratio 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, or between 1:10 and 1:20. In some embodiments, the ophthalmic solution comprising cyclodextrin further comprises hamine (also known as Tris, Tris(Hydroxymethyl)aminomethane, or Tris buffer). In some embodiments, the ophthalmic solution comprises about 0.05% - 1% Tris. In some embodiments, the ophthalmic solution comprises about 0.05% - 0.5% Tris. In some embodiments, the ophthalmic solution comprises about 0.05% - 0.2% Tris. In some embodiments, the lmic solution ses about 0.1% — 0.15% Tris. In some embodiments, the ophthalmic solution comprises about 1% Tris. In some embodiments, the ophthalmic solution further comprises about 0.005% — 5.0% second active agent.
] Ophthalmic solutions of the present embodiments include, for example, without being limiting: about 0.3% — about 5.0% Compound—I (about 3 mg/mL — about 50.0 mg/mL), about 0.05% sodium phosphate monobasic drate, about 2% glycerin; about 0.4% nd—I, about 7% HPBCD, about 0.7% NaCl, about 0.005% BAK; about 0.4% Compound—I, about 4% HPBCD, about 0.7% NaCl, about 0.005% BAK; about 0.4% Compound—I, about 7% HPBCD, about 1% tromethamine, about 0.4% NaCl, about 0.005% BAK; and about 0.6% nd—I, about 7% HPBCD, about 0.7% NaCl, about 0.005% BAK. For Compound—I of between about 0.005% to about 5.0% concentrations, cyclodextrin is present at a corresponding molar ratio. In some embodiments, the ophthalmic on further comprises about 0.005% — 5.0% second active agent.
] Additional ophthalmic solutions include, for example, without being limiting: about 0.4% Formula 11 compound (free base), about 7.15% HPBCD, about 0.7% NaCl; about 0.1% Formula 11 compound (free base), about 1.79% HPBCD, about 0.85% NaCl; about 0.2% Formula 11 compound (free base), about 3.57% HPBCD, about 0.8% NaCl; about 0.6% a 11 compound (free base), about 10.72% HPBCD, about 0.6% NaCl; about 0.4% Formula 11 compound (free base), about 8.41% HPBCD, about 0.65% NaCl; about 0.4% Compound-I, about 10.51 HPBCD, about 0.65% NaCl; about 0.4% Formula 11 compound (free base), about 10.51% HPBCD, about 0.15% NaCl, about 1.0% tromethamine (Tris); and/or about 0.1% Formula 11 compound (free base), about 2.63% HPBCD, about 0.8% NaCl; about 0.6% Compound—I (as free base), about 15.77% HPBCD, about 0.37% NaCl. For Formula II of between about 0.005% to about 5.0% concentrations, cyclodextrin is present at a corresponding molar ratio. In some embodiments, the ophthalmic solution further comprises about 0.005% — 5.0% second active agent.
In some embodiments, the ophthalmic solutions include between about 1.0% — about 25% extrin. For example, without being limiting, the nd-I formulations include about 2.0% — about 3.0% HPBCD, about 3.0% — about 5.0% HPBCD, about 5.0% — about 10% HPBCD, or about 10% - about 25% HPBCD. In some embodiments, the ophthalmic solution further comprises about 0.005% — 5.0% second active agent.
] In additional embodiments, the ophthalmic solutions are formulated as, for example, without being limiting: about 8.41% SE® HPB and about 0.142% phosphate; about 8.9% KLEPTOSE® HPB and about 0.142% phosphate; about 4.88% CAPTISOL® and about 0.142 ate; and/or about 4.88% CAPTISOL® and about 0.122% phosphate.
In some embodiments, the ophthalmic solutions comprising cyclodextrins are clear and colorless, and are extremely viscous, moderately viscous, or have viscosity similar to water.
In some embodiments, the ophthalmic solution of the application has a pH value of about 4.5 to about 7.5 at or under about 40 0C.
In some embodiments, the ophthalmic solution of the application has a pH value of about 5.0 to about 7.0 at or under about 40 °C.
For example, the ophthalmic solution of the application has a pH value of about 6.0 at or under about 40 0C.
] The ophthalmic solutions of the present application may contain s additives incorporated ordinarily, such as buffering agents (e.g., phosphate buffers, borate buffers, citrate buffers, tartrate s, acetate buffers, amino acids, sodium acetate, sodium citrate and the like), tonicity agents (e.g., saccharides such as sorbitol, glucose and mannitol, polyhydric alcohols such as glycerin, concentrated glycerin, PEG and Propylene glycol, salts such as sodium chloride, etc), preservatives or antiseptics (e.g., Benzalkonium chloride, Benzatkonium chloride, enzoates such as Methyl p-oxybenzoate or Ethyl p- oxybenzoate, Benzyl alcohol, Phenethyl alcohol, Sorbic acid or its salt, Thimerosal, Chlorobutanol, etc), solubilizing aids or particle stabilizing agents (e. g., water-soluble polymers such as nyl pyrrolidone, surfactants such as tyloxapol, polysorbates, poloxamer, eta), pH modi?ers (e.g., Hydrochloric acid, Acetic acid, Phosphoric acid, Sodium ide, ium hydroxide, Ammonium hydroxide and the like), thickening agents (e.g., HEC, Hydroxypropyl cellulose, Methyl ose, HPMC, Carboxymethyl cellulose and their salts), ing agents (e.g., sodium edetate, sodium e, condensed sodium phosphate etc), and second active agent stabilizers (e.g., EDTA, propyl gallate, and a combination thereof).
The ophthalmic solutions of the present application comprise cyclodextrin, and may further comprise additional excipients, for example, without being limiting, about 0.5% — about 3% surfactant and emulsi?er, for example, without being limiting, polysorbate 80 or equivalent excipients thereof; about 0.05 — about 0.4% nonionic liquid polymer of the alkyl aryl polyether alcohol type, for example, without being limiting tyloxapol; and/or about 0.05% - about 0.6% hydrophilic non-ionic tant, for example, without being limiting, poloxamer, such as poloxamer 407.
In some ments, the ophthalmic solution comprises about 0.01 — about 0.5%, about 0.02 — about 0.5%, about 0.04 — about 0.5%, about 0.06 - about 0.5%, about 0.08 — about 0.5%, about 0.08 — about 0.4%, about 0.08 — about 0.3%, about 0.08 — about 0.2%, about 0.08 — about 0.18%, about 0.08 — about 0.16%, about 0.08 — about 0.14%, or about 0.08 - about 0.12% EDTA. In some embodiments, the ophthalmic solution comprises about 0.04%, about 0.06%, about 0.08%, about 0.1%, about 0.12, about 0.14%, about 0.16%, about 0.18%, or about 0.2% EDTA. In some embodiments, the lmic solution comprises about 0.1% EDTA.
In some embodiments, the ophthalmic solution comprises about 0.001 — about 0.5%, about 0.002 — about 0.5%, about 0.005 — about 0.5%, about 0.01 — about 0.5%, about 0.02 — about 0.5%, about 0.03 — about 0.5%, about 0.04 - about 0.5%, about 0.01 — about 0.4%, about 0.01 — about 0.3%, about 0.01 — about 0.2%, about 0.01 — about 0.1%, about 0.01 — about 0.08%, about 0.01 — about 0.06% propyl gallate. In some ments, the ophthalmic solution comprises about 0.01%, about 0.02%, about 0.03%, about 0.04%, about 0.05%, about 0.06%, about 0.07%, about 0.08%, about 0.09% propyl gallate. In some embodiments, the ophthalmic solution ses about 0.05% propyl gallate.
Concentration in Various Ocular Tissues—Delivered as an Ophthalmic Solution The ocular solution sing extrin improves bioavailability of the ?rst active agents of the present application at the posterior segment of the eye. Without being bound by theory, in an embodiment, the formulation comprising cyclodextrin forms a clear and ess solution, which lowers corneal exposure of the active agent, for example, exposure of Compound—I, by about 5—15 fold compared to the corneal re to with an equimolar Gel Drop formulation.
Without being bound by theory, in one embodiment, an ophthalmic solution sing cyclodextrin increases the therapeutic index of Compound-I during topical ocular administration. Upon administration, the hydrophilic complex of cyclodextrin-Compound—I is pharmacologically inert at the cornea. Without being bound by theory, in some embodiments, the cyclodextrin-Compound—I complex increases corneal tolerability of Compound—I. Without being bound by theory, in some embodiments, spontaneous dissociation of cyclodextrin from Compound—I at the peripheral vasculature increases ilability at the target tissue, e.g., at the choroid or retina.
Unlike other formulations of Compound-I, which in some embodiments contribute to corneal toxicity, the cyclodextrin—based ophthalmic solution comprising similar concentration of Compound—I lower l exposures and, thereby increase the therapeutic index and ponding benefits to patients. In one embodiment, the use of cyclodextrin- based solution of Compound—I provides approximately 10x reduction in corneal exposure, as compared to equimolar concentrations of Gel Drop. In some embodiments, the cyclodextrin- based solution of nd—I reduces corneal exposure of Compound—I by 5x, 20x, 30x, 40x, or 50x. In one embodiment, 1—90 days or 3—9 months of topical ocular dosing of about 0.005% — about 5.0% Compound—I as a cyclodextrin—based solution does not have any e or toxic effect at the cornea, choroid, and/or the retina. In yet r embodiment, 1—90 days of topical ocular dosing of about 0.6% — about 5.0% Compound—I as a cyclodextrin—based solution does not have any adverse or toxic effect at the cornea, choroid, and/or the retina.
The lowering of the corneal exposure is correlated with increasing ilability and therapeutic index of the active agent at the ior segment, for example, at the retina or choroid, of the eye. For example, no toxic effect attributable to the ?rst active agent or a suitable r is observed to the cornea or other parts of the eye when about 0.1% — about .0% Compound—I formulation comprising cyclodextrin is administered lly stered to the eye for at least 30 days or more than 60 days.
In one embodiment, when a formulation comprising about 0.4% (about 4 mg/mL) of Compound-I or its free base, and cyclodextrin, when administered topically to the eye, the central choroid tration is between about 0.2 [1M — about 0.9 uM, central retina tration of the active agent is between about 0.02 [1M — about 0.4 "M, aqueous humor concentration of the active agent is about 0.003 "M — about 0.009 "M, and corneal concentration of the active agent is between 6 [1M — 40 "M. The cyclodextrin used in the formulation is, for example, t being limiting example, KLEPTOSE® HPB or In some embodiments, a cyclodextrin—based solution of Compound—I or its free base increases the bioavailability of the active agent at the central choroid and the central retina, while ng concentration at the cornea. In some embodiments, topical delivery of Compound—I or its free base formulated in the presence of cyclodextrin reduces the corneal tration by about 5 — about 15 fold over the corneal concentration of equimolar Gel Drop.
Without being bound by theory, in some embodiments, the combined effects of sing corneal drug exposure so as to avoid poor ocular tolerability while increasing posterior segment bioavailability increase the therapeutic index and corresponding benefits to In some embodiments, the exposure time of Compound—I and the second active agent is between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments, the dosage regimen involves several courses of topical ocular administration of a formulation comprising nd-I and a second active agent, wherein the second active agent is administered as a separate formulation or as part of the Compound-I formulation, to a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). For example, the dosage regimen involves once daily, twice daily, three times daily or four times daily stration of the formulation for between 1 and 90 days or for longer than 90 days (e. g., 4 months, 6 months, 8 months, or 12 months). For example, the dosage regimen involves once, twice, three times, or four times administration of a compound of Formula I or II and a second active agent on alternate days (126., on day l, 3, 5, 7 etc.) for up to 90 days. For example, the dosage regimen involves administering once on day 1, once or twice on day 2 — day 90. For example, the dosage regimen involves administering once, twice, three times, or four times on day 1, followed by once daily for 2-90 days. For example, the dosage regimen involves administering once, twice, three times, four times on day 1, ed by once, twice, three times, or four times on ate days (tie, on day 1, 3, 5, 7" etc.) for up to 90 days. For example, one dosage regimen es once per day or twice per day for l, 2, 3, 4, or 5 consecutive days. For twice or three daily dosage regimen, ts receive topical ocular dose of a Compound—I formulation and a second active agent on days 1 and 4 approximately about 4, 6, or 8 hours apart. In another embodiment, subjects receive l ocular doses of a Compound-I formulation and a second active agent approximately about 4, 6, or 8 hours apart for four utive days. In some embodiments, subjects receive one or two doses of topical ocular dose of Compound—I formulation and a second active agent per day for 5 consecutive days. In yet other embodiments, subjects receive one or two doses of l ocular dose of Compound—I and a second active agent formulation for 5—90 consecutive days.
In some embodiments, subjects receive one or two doses of topical ocular dose of Compound—I ation and a second active agent for at least 25 utive days. In one embodiment, subjects receive one or two topical ocular doses for at least 90 consecutive days or more. The second active agent can be administered separately or as part of the Compound-I formulation. When administered separately, the second active agent, or a pharmaceutical salt thereof, can be administered alone or as a formulation.
For example, a formulation comprising about 1 mg/mL BID of a ?rst active agent (e. g., Compound—I) and a second active agent is stered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a ation comprising about 1 mg/mL QD of a ?rst active agent (e. g., nd—I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation sing about 1 mg/mL TID of a ?rst active agent (e.g., Compound—I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 , 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 1 mg/mL QID of a ?rst active agent (e.g., Compound-I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 2 mg/mL BID of a ?rst active agent (e.g., Compound—I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some ments a formulation comprising about 2 mg/mL QD of a ?rst active agent (e. g., Compound-I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 2 mg/mL TID of a ?rst active agent (e.g, Compound—I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e. g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a ation comprising about 2 mg/mL QID of a ?rst active agent (e.g., Compound-I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 3 mg/mL BID of a ?rst active agent (e.g., Compound—I) and/or a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 ). In some embodiments a formulation comprising about 3 mg/mL QD of a ?rst active agent (e.g., Compound—I) and/or a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 , 8 months, or 12 months). In some embodiments a formulation comprising about 3 mg/mL TID of a ?rst active agent (e.g., Compound—I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 3 mg/mL QID of a ?rst active agent (e.g., Compound-I) and a second active agent is administered to one eye or both eyes of a subject for n 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 , or 12 months). In some embodiments a formulation comprising about 4 mg/mL BID of a first active agent (e.g., nd—I) and/or a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e. g., 4 months, 6 months, 8 months, or 12 months). In some ments a formulation comprising about 4 mg/mL QD of a ?rst active agent (e. g., Compound—I) and/or a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 , 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 4 mg/mL TID of a ?rst active agent (e.g., Compound—I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e. g., 4 , 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 4 mg/mL QID of a ?rst active agent (e.g., Compound—I) and a second active agent is administered to one eye or both eyes of a t for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some ments a formulation comprising about 5 mg/mL BID of a ?rst active agent (e.g., Compound-I) and/or a second active agent is administered to one eye or both eyes of a t for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 5 mg/mL QD of a ?rst active agent (e. g., Compound—I) and/or a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 5 mg/mL TID of a ?rst active agent (e.g., nd—I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 5 mg/mL QID of a ?rst active agent (e.g., Compound-I) and a second active agent is stered to one eye or both eyes of a subject for n 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 6 mg/mL BID of a ?rst active agent (e.g., Compound-I) and/or a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e. g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 6 mg/mL QD of a ?rst active agent (e. g., Compound—I) and/or a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a ation comprising about 6 mg/mL TID of a ?rst active agent (e.g., Compound—I) and a second active agent is administered to one eye or both eyes of a t for between 1 and 90 days or for longer than 90 days (e. g., 4 months, 6 months, 8 months, or 12 ). In some embodiments a formulation comprising about 6 mg/mL QID of a ?rst active agent (e.g., Compound-I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a ation comprising about 7 mg/mL BID of a ?rst active agent (e.g., Compound—I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a ation comprising about 7 mg/mL QD of a ?rst active agent (e.g., Compound-I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e. g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 7 mg/mL TID of a ?rst active agent (e.g., Compound-I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 , or 12 months). In some ments a formulation comprising about 7 mg/mL QID of a ?rst active agent (e.g., Compound—I) and a second active agent is stered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 8 mg/mL BID of a ?rst active agent (e.g., Compound—I) and a second active agent is administered to one eye or both eyes of a subject for n 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 , 8 , or 12 ). In some embodiments a formulation comprising about 8 mg/mL QD of a ?rst active agent (e.g., Compound—I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e. g., 4 months, 6 , 8 months, or 12 months). In some embodiments a formulation comprising about 8 mg/mL TID of a ?rst active agent (e.g., Compound—I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 8 mg/mL QID of a ?rst active agent (e.g., Compound-I) and a second active agent is administered to one eye or both eyes of a subject for n 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a ation comprising about 9 mg/mL BID of a ?rst active agent (e.g., Compound—I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 9 mg/mL QD of a ?rst active agent (e.g., Compound—I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e. g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation sing about 9 mg/mL TID of a ?rst active agent (e.g., Compound-I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e. g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 9 mg/mL QID of a ?rst active agent (e.g., Compound-I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 10 mg/mL QD of a ?rst active agent (e.g., Compound—I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 ). The dosage regimen for between 1 and 90 days or for longer than 90 days (e. g., 4 months, 6 , 8 months, or 12 months) may be any of the regimens involving consecutive or alternate days described in the paragraph above. In some embodiments, the formulation of the present application is administered QD, BID, TID, or QID when administered at low doses (e. g., 1 mg/mL, 2 mg/mL, 3 mg/mL, 4 mg/mL, or 5 mg/mL), and QD or BID at high doses (e.g., 6 mg/mL, 7 mg/mL, 8 mg/mL, 9 mg/mL, or 10 mg/mL).
In some embodiments, a 1 mg/mL BID of a ?rst active agent (e.g., Compound-I) formulation and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments, a 1 mg/mL QD of a ?rst active agent (e.g., Compound—I) formulation and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments, a 2 mg/mL BID of a ?rst active agent (e.g., Compound—I) formulation and a second active agent is administered to one eye or both eyes of a subject for n 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some ments, a 2 mg/mL QD of a ?rst active agent (e.g., Compound—I) formulation and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 , 8 , or 12 months). In some embodiments, a 3 mg/mL BID of a ?rst active agent (e.g., Compound—I) ation and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 , 6 months, 8 months, or 12 months). In some embodiments, a 3 mg/mL QD of a ?rst active agent (e.g., Compound—I) formulation and a second active agent is administered to one eye or both eyes of a subject for n 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments, a 4 mg/mL BID of a ?rst active agent (e.g., Compound—I) formulation and a second active agent is stered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 , or 12 months). In some embodiments, a 4 mg/mL QD of a ?rst active agent (e.g., nd—I) formulation and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments, a 5 mg/mL BID of a ?rst active agent (e.g., Compound—I) formulation and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments, a 5 mg/mL QD of a ?rst active agent (e.g., Compound-I) formulation and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments, a 6 mg/mL BID of a ?rst active agent (e.g., Compound—I) formulation and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments, a 6 mg/mL QD of a ?rst active agent (e.g., Compound—I) formulation and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments, a 7 mg/mL BID of a ?rst active agent (e.g., Compound—I) formulation and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments, a 7 mg/mL QD of a ?rst active agent (e.g., Compound—I) ation and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 , 6 months, 8 months, or 12 months). In some embodiments, a 8 mg/mL BID of a ?rst active agent (e.g., Compound-I) formulation and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 , or 12 months). In some embodiments, a 8 mg/mL QD of a ?rst active agent (e.g., Compound—I) formulation and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments, a 9 mg/mL BID of a ?rst active agent (e.g., Compound—I) formulation and a second active agent is administered to one eye or both eyes of a t for n 1 and 90 days or for longer than 90 days (e.g., 4 , 6 months, 8 months, or 12 months). In some ments, a 9 mg/mL QD of a ?rst active agent (e.g., Compound—I) formulation and a second active agent is stered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments, a 10 mg/mL BID of a ?rst active agent (e.g., Compound—I) formulation and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments, a 10 mg/mL QD of a ?rst active agent (e.g., Compound—I) formulation and a second active agent is administered to one eye or both eyes of a subject for n 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). The dosage regimen for between 1 and 90 days or for longer than 90 days (e. g., 4 months, 6 months, 8 months, or 12 months) may be any of the regimens involving utive or alternate days described in the paragraph above.
The present application provides cyclodextrin-based solutions containing hydroxypropyl-beta—cyclodextrin (HP—B—CD, KLEPTOSE® HPB) or CAPTISOL® that are well tolerated when administered topically for 30 — 90 days or for 4 — 6 months. In some ments, once or twice daily administration of at about 0.005% — about 5.0% w/v Compound—I or its free base and a second active agent in a solution containing about 1.0% - about 25% HP—B—CD or OL® is well tolerated by the t.
In some embodiments, the formulation of Formula II or Compound-I and a second active agent is administered to one eye or both eyes of a subject. For example, about 0.2 % — about 1.0% (w/v) of the compound of Formula II or about 0.1% — 1.2 % (w/v) of Compound— I formulation and a second active agent comprising formulation of the present application is administered once a day (QD), twice a day (BID), three times a day (BID), or four times a day (QID) to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e. g., 4 , 6 months, 8 months, or 12 months). In some ments, the Formula II or Compound—I formulation and a second active agent are administered to one eye or both eyes of a subject. For example, about 0.2 % — about 1.0% (w/V) of the nd of Formula II or about 0.1% — 1.2 % (w/v) of nd—I formulation and a second active agent is administered once a day (QD), twice a day (BID), three times a day (BID), or four times a day (QID) to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e. g., 4 months, 6 months, 8 months, or 12 months).
In some embodiments, Formula 11 compound or Compound—I is complexed with a complexing agent, e. g., cyclodextrin (e.g., KLEPTOSE® HPB (%)) in ratio of about 1:8, in which about 2% — 13 % (w/v) cyclodextrin (e.g., KLEPTOSE® HPB (%)) is added to the formulation. The formulation may further comprise about 0.1% — about 0.2% buffer, e.g., 10 mM phosphate buffer. The desired osmolality of the formulation is about 200 — about 300 mOsm, achieved by adding quantity suf?cient to achieve the lity with a salt, e.g., sodium de. The pH of the formulation is about 6.0 at or under about 40 0C. The dosage regimen for between 1 and 90 days or for longer than 90 days (e. g., 4 months, 6 months, 8 months, or 12 months) may be any of the regimens involving consecutive or alternate days described in the paragraph above.
Ophthalmic Suspensions The present application provides suspensions of a ?rst active agent (e.g., Compound-I) comprising the agent and pharmaceutically acceptable excipients. The present application also provides suspensions of a ?rst active agent (e.g., Compound-I) and a second active agent comprising the ?rst active agent, the second active agent and pharmaceutically acceptable excipients. For example, nd—I suspensions and second active agent suspensions may include, without being limiting, buffering , acids & bases, for example, without being limiting, HCl and NaOH. In one embodiment, suspensions of Compound—I or its free base may include a ing agent, for example, without being limiting, tromethamine (Tris). In another embodiment, suspensions of Compound—I or its free base and a second active agent may include a buffering agent, for example, without being limiting, hamine (Tris). The hamine—based suspension of Formula II compound or Compound—I and a second active agent is useful for topical administration to the eye.
The suspensions of the ation comprise about 0.005% to about 5.0% w/v of a ?rst active agent of Formula I or II, or a pharmaceutically acceptable salt f, for example, Compound-I. In some embodiments, the concentration of Compound-I or its free base (Formula II) in the suspensions is about 0.005% - about 0.01%, about 0.01% - about 0.05%, about 0.05% — about 0.1%, about 0.1% — about 0.2%, about 0.2% - about 0.3%, about 0.3% — about 0.4%, about 0.4% — about 0.5%, about 0.5% — about 0.6%, about 0.6% — about 0.7%, about 0.7% — about 0.8%, about 0.8% — about 0.9%, about 0.9% — about 1.0%, about 1.0 — about 2.0%, about 2.0 — about 3.0%, about 3.0 — about 4.0%, or about 4.0— about 5.0% w/v for topical administration. In some embodiments, the concentration of nd-I or its free base (Formula II) in the formulations is about 0.1% — about 1.2%, about 0.2% — about 1.2%, about 0.3% — about 1.2%, about 0.4% — about 1.2%, 0.1% — about 1.1%, about 0.2% — about 1.1%, about 0.3% — about 1.1%, about 0.4% — about 1.1%, 0.1% — about 1.0%, about 0.2% — about 1.0%, about 0.3% — about 1.0%, about 0.4% — about 1.0%, 0.1% — about 0.8%, about 0.2% — about 0.8%, about 0.3% — about 0.8%, about 0.4% — about 0.8%, 0.1% — about 0.6%, about 0.2% — about 0.6%, about 0.3% — about 0.6%, about 0.4% — about 0.6%, 0.1% — about 0.5%, about 0.2% — about 0.5%, about 0.3% - about 0.5%, about 0.4% — about 0.5%, 0.1% — about 0.4%, about 0.2% — about 0.4%, about 0.3% — about 0.4% w/v for l stration. In some embodiments, the suspensions include about 0.005%, about 0.05%, about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1.0%, about 2.0%, about 3.0%, about 4.0%, or about 5.0% w/v of Compound—I or its free base (Formula II).
The suspensions of the application may further se about 0.00001% — about .0% W/V of a second active agent, or a pharmaceutically acceptable salt thereof, for example, nicotinic acid, nicotinamide, or vitamin K, or a combination thereof. The suspensions of the application may further comprise about 0.00001% - about 1.0%, about 0.00001% - about 0.1%, about 0.00001% — about 0.01%, about 0.00001% — about 0.001%, about 0.00001% — about 0.0002%, or about 0.00001% — about 0.0001% w/v of a second active agent, or a pharmaceutically able salt thereof, for example, nicotinic acid, nicotinamide, or vitamin K, or a combination thereof. In some embodiments, the concentration of the second active agent in the suspensions is about 0.00001% - about 0.0001%, 0.000012% — about 0.0001%, 0.000014% - about 0.0001%, 16% — about 0.0001%, 0.000018% - about 0.0001%, 0.00002% - about 0.0001%, 0.00003% — about 0.0001%, 0.00004% - about 0.0001%, 0.00005% - about 0.0001%, 0.00006% — about 0.0001%, 0.00007% - about 0.0001%, 0.00008% - about 0.0001%, 9% — about 0.0001%, 0.000016% - about 0.00009%, 0.000018% - about 0.00009%, 0.00002% — about 0.00009%, 0.00003% - about 0.00009%, 0.00004% - about 0.00009%, 0.00005% — about 0.00009%, 0.00006% — about 0.00009%, 0.00007% - about 0.00009%, or 0.00008% - about 0.00009% W/V for topical administration. In some embodiments, the suspensions include about 0.00001%, 0.00002%, 0.00003%, 0.00004%, 0.00005%, 0.00006%, 0.0000?%, 0.00008%, 81%, 0.000082%, 0.000083%, 0.000084%, 0.000085%, 0.000086%, 0.000087%, 88%, or 89% w/v of the second active agent.
The sions of the ation may further se about 0.5 uM, about 0.6 uM, about 0.7 uM, about 0.8 uM, about 0.9 uM, about 1 uM, about 2 uM, about 3 uM, about 4 uM, about 5 uM, about 6 uM, about 7 uM, about 8 uM, or about 9 uM of a second active agent, or a ceutically acceptable salt thereof, for example, nicotinic acid, nicotinamide, or vitamin K, or a combination thereof. In some embodiments, the concentration of the second active agent is about 1 "M.
The ophthalmic suspensions may contain s additives incorporated ordinarily, such as buffering agents (e.g., ate buffers, borate buffers, e buffers, tartrate buffers, acetate buffers, amino acids, sodium acetate, sodium e and the like), tonicity agents (e.g., saccharides such as sorbitol, glucose and mannitol, polyhydric alcohols such as glycerin, concentrated glycerin, PEG and propylene glycol, salts such as sodium chloride), vatives or antiseptics (e.g., benzalkonium chloride, benzatkonium chloride, P—oxybenzoates such as methyl p-oxybenzoate or ethyl p—oxybenzoate, Benzyl alcohol, phenethyl l, Sorbic acid or its salt, Thimerosal, Chlorobutanol and the like), solubilizing aids or particle stabilizing agents (e.g., cyclodextrins and their derivative, water— soluble polymers such as polyvinyl pyrrolidone, surfactants such as tyloxapol, polysorbates, poloxamer), pH modifiers (e.g., hydrochloric acid, acetic acid, phosphoric acid, sodium hydroxide, potassium hydroxide, ammonium hydroxide and the like), thickening agents (e.g., HEC, hydroxypropyl cellulose, methyl cellulose, HPMC, carboxymethyl cellulose and their salts), chelating agents (e. g., sodium edetate, sodium citrate, condensed sodium ate etc), and second active agent stabilizers (e.g, EDTA, propyl gallate, and a combination thereof).
The ophthalmic suspension of the present application comprises pharmaceutical excipients chosen at or below concentrations optimal for ophthalmic solution. The excipients of the present application include, for example, without being limiting, sodium phosphate monohydrate, glycerin, and benzalkonium chloride (BAK).
] In some embodiments, the lmic sion comprises about 0.01 — about 0.5%, about 0.02 - about 0.5%, about 0.04 — about 0.5%, about 0.06 — about 0.5%, about 0.08 - about 0.5%, about 0.08 - about 0.4%, about 0.08 — about 0.3%, about 0.08 — about 0.2%, about 0.08 - about 0.18%, about 0.08 - about 0.16%, about 0.08 - about 0.14%, or about 0.08 - about 0.12% EDTA. In some embodiments, the lmic suspension comprises about 0.04%, about 0.06%, about 0.08%, about 0.1%, about 0.12, about 0.14%, about 0.16%, about 0.18%, or about 0.2% EDTA. In some embodiments, the ophthalmic suspension comprises about 0.1% EDTA.
In some embodiments, the ophthalmic suspension comprises about 0.001 — about 0.5%, about 0.002 — about 0.5%, about 0.005 — about 0.5%, about 0.01 — about 0.5%, about 0.02 — about 0.5%, about 0.03 — about 0.5%, about 0.04 — about 0.5%, about 0.01 — about 0.4%, about 0.01 — about 0.3%, about 0.01 — about 0.2%, about 0.01 — about 0.1%, about 0.01 — about 0.08%, about 0.01 — about 0.06% propyl gallate. In some embodiments, the ophthalmic sion comprises about 0.01%, about 0.02%, about 0.03%, about 0.04%, about 0.05%, about 0.06%, about 0.07%, about 0.08%, about 0.09% propyl gallate. In some embodiments, the ophthalmic suspension comprises about0.05% propyl gallate.
In some embodiments, the ophthalmic suspension comprises about 0.005%, about 0.05%, about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1.0%, about 2.0%, about 3.0%, about 4.0%, or about .0% w/V of Formula I or II, or a ceutically acceptable salt thereof, for example, Compound—I, and a second active agent, and may further comprise tromethamine (i.e., Tris).
In some embodiments, the ophthalmic suspension comprises about 0.05% - 1% Tris. In some embodiments, the ophthalmic suspension comprises about 0.2% — 1.0% Tris. In some embodiments, the ophthalmic suspension comprises about 0.4% - 0.8% Tris. The tromethamine-based suspension of nd-I or its free base and/or a second active agent may comprise additional buffers and excipients, for example, without being limiting, ate buffer. The suspensions may further comprise one or more surfactant and emulsi?er, for example, t being limiting, polysorbate 80 or equivalent excipients thereof; one or more nonionic liquid polymer of the alkyl aryl polyether alcohol type, for example, without being limiting tyloxapol; and/or one or more hydrophilic non-ionic surfactant, for example, without being limiting, poloxamer, such as poloxamer 407.
] The present application provides suspensions of the agents of the present application formulated in the presence of excipients such as, without being limiting, Povidone, polysorbate 80 (P880), polyethylene glycol (PEG) 400, tyloxapol, poloxamer, glycerin, and BAK in a Tris buffer.
] In one embodiment the suspension of Compound—I or its free base ses about 0.1 - 0.5% phosphate buffer. In another embodiment the suspension of Compound-I or its free base and the second active agent comprises about 0.1 - 0.5% phosphate buffer. In some ments, the pH of the tromethamine—based sion is between pH 4-7, for example, pH 6.0. In some embodiments, the suspensions prepared in Tris further comprise about 0.5% — about 2% polysorbate 80; about 0.05 — about 0.2% tyloxapol, and/or about 0.05% — about 0.4% poloxamer 407.
In some such embodiments, the suspensions of the application r se about 0.01 — about 1%, or about 1 — about 2.0% w/v glycerin. In a specific ment, the sions comprise about 2% w/v glycerin.
In some embodiments, the sions of the application further comprise about 0001— about 0.005% w/v Benzalkonium chloride (BAK). The BAK amount may be varied depending on any observed adverse effects. BAK may be damaging to the cells on the ocular surface, and, therefore, the amount in the formulation may be varied to achieve an optimal level of ocular penetration of Compound—I, without mising the ocular cell layer integrity and increased toxicity.
In some embodiments, the suspension optionally comprises s. Buffers when used, for example, can be sodium monophosphate basic, phosphoric acid and Tris buffer.
Compound—I concentration in suspension is about 0.005% — about 5.0% w/v. The suspension prepared without additional buffer ?irther comprises about 0.005% BAK and about 2% glycerin and with a pH 6.0. In another embodiment the suspension prepared without additional buffer comprises about 1% polysorbate 80, about 0.1% tyloxapol, about 0.2% Poloxamer 407, about 0.005% BAK, about 2.0% glycerin, and with a pH 6.0.
In suspensions prepared in phosphoric acid/Tris, the suspension comprises about 0.14% phosphoric acid, about 0.2% Tris base, about 1.0% polysorbate 80, about 0.005% BAK, about 2.0% glycerin and with a pH 6.0. In one embodiment, the sion further comprises about 0.2% tyloxapol. The pH of the suspension varies between about pH 6.0 and The suspensions ed in tromethamine (Tris) alone comprise about 1% polysorbate 80, about 0.1% tyloxapol, about 0.2% Poloxamer 407, about 0.6% Tris, about 0.005% BAK, and about 2.0% glycerin with pH 6.0. In r embodiment, a suspension prepared in Tris comprises, about 1% Tris, about 0.45% NaCl, about 0.025% EDTA, about 0.2% HPMC, about 0.1% polysorbate 80, about 0.005% BAK, with a pH 6.0. In these suspensions 1 N HCl and/or 1N NaOH are used for titration to appropriate pH.
The suspension of the application comprises about 0.005%, about 0.05%, about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1.0%, about 2.0%, about 3.0%, about 4.0%, or about 5.0% of an active agent of Formula I or II, or a pharmaceutically able salt thereof, for example, nd—I, and about 0.01% — about 0.05%, about 0.05 — about 0.09%, or about 0.09 — about 0.2% w/v sodium phosphate monobasic drate and/or about 0.3% — about 1.0% of Tris. Alternatively, the suspension of the application comprises about 0.005%, about 0.05%, about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1.0%, about 2.0%, about 3.0%, about 4.0%, or about .0% of an active agent of FormulaI or II, or a pharmaceutically acceptable salt thereof, for example, Compound—I, about 0.005%, about 0.05%, about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1.0%, about 2.0%, about 3.0%, about 4.0%, or about 5.0% of a second active agent, and about 0.01% — about 0.05%, about 0.05 — about 0.09%, or about 0.09 — about 0.2% W/V sodium ate monobasic monohydrate and/or about 0.3% — about 1.0% of Tris.
In a specific embodiment, the suspension comprises about 0.14% or about 0.2% w/v Tris—buffer. In additional ments, suspensions are prepared in about 0.6% Tris or about 1.0% Tris. Other equivalent buffer systems well known in the art are also used in the sions of the present application. In one embodiment, the Formula II compound or Compound—I is formulated as about 0.4% active agent, about 5% Cremophor RH40, about 2.0% glycerin, and about 0.005% BAK. In another embodiment, the Formula II compound or Compound-I is formulated as about 0.4% active agent, about 0.005% to about 5.0% of the second active agent, about 5% Cremophor RH40, about 2.0% glycerin, and about 0.005% In some embodiments, the suspension of the application has a pH value of about 4.0 to about 7.5 at or under about 40 0C.
In some embodiments, the suspension of the application has a pH value of about .0 to about 7.0 at or under about 40 0C.
] For example, the suspension of the application has a pH value of about 6.0 at or under about 40 0C.
In some embodiments, the ?rst active agent may be formulated as a solution according to the embodiments bed herein, and the second active agent may be formulated as a suspension according to the embodiments bed herein.
In other embodiments, the ?rst active agent may be formulated as a suspension according to the embodiments described herein, and the second active agent may be formulated as a solution according to the embodiments described herein.
Concentration in Various Ocular Tissues—Delivered as an Ophthalmic Suspension In some embodiments, a suspension of Compound-I or its free base provides similar concentration of the ?rst active agent at the central choroid and the central retina compared to the concentration of the ?rst active agent delivered in Gel Drop form (discussed infra).
In some embodiments, Tris—based sion of Compound—I or its free base with or without a second active agent increases the bioavailability of the first active agent at the central d and the central retina, while reducing tration at the cornea and preventing and/or treating l disruptions and or diseases (e. g., corneal edema, ulceration, abnormalities, etc). In some embodiments, topical ry of Compound—I or its free base formulated in Tris—base reduces corneal concentration of Compound—I by about 5— 10x, 10—20x, 20—30x, 30—40x, or about 50—100x compared to the corneal concentration of lar Compound—I or its free base delivered as a Gel Drop.
The ed effects of decreasing corneal drug exposure so as to avoid poor ocular tolerability while maintaining or increasing posterior segment bioavailability so as to se inhibition of receptor tyrosine kinase (RTK), for example, VEGFR, significantly increases the eutic index and corresponding bene?ts to patients. The additional prevention and/or treatment of disruptions to the anterior surface of the eye with a second active agent, such as an EGFR modulator (e.g., activator), further improves the therapeutic index and corresponding benefits to patients.
Once or twice daily stration of about 0.005% - about 5.0% W/v Compound- I sion of the present application for 30-90 days or 4 — 6 months is well tolerated in the Gel Drop In some embodiments the ophthalmic composition or formulation of the present application is formulated as a Gel Drop. The Gel Drop formulation includes no more than about 0.05% of sodium ate monobasic monohydrate to provide the required buffering capacity and free—?owing, ?lterable formulations at about 0.005% — about 2.0% Compound—1 and/or at about 0.005% — about 5% of a second active agent without the need for surfactant The Gel Drop formulation of the application comprises about 0.005% to about 2.0% w/v of the ?rst active agent of Formula I or II, or a pharmaceutically acceptable salt thereof, for example, Compound-I. The concentration of Compound—I or its free base (Formula II) in the Gel Drops may be about 0.005% - about 0.01%, about 0.01% - about 0.05%, about 0.05% - about 0.1%, about 0.1% - about 0.2%, about 0.2% - about 0.3%, about 0.3% — about 0.4%, about 0.4% — about 0.5%, about 0.5% — about 0.6%, about 0.6% - about 0.7%, about 0.7% — about 0.8%, about 0.8% — about 0.9%, about 0.9% - about 1.0%, or about 1.0% — about 2.0% W/V for topical administration. In some embodiments, the Gel Drops include about 0.005%, about 0.05%, about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1.0%, or about 2% w/v of Compound—1 or its free base (Formula 11).
The Gel Drop formulation of the application may further comprise about about 0.00001% — about 5.0% W/v of the second active agent, or a pharmaceutically acceptable salt thereof, for e, nicotinic acid, namide or n K, or a combination thereof.
The Ge1 Drop formulation of the application may further comprise about 0.00001% — about 1.0%, about 0.00001% — about 0.1%, about 0.00001% — about 0.01%, about 0.00001% — about 0.001%, about 0.00001% — about 0.0002%, or about 0.00001% — about 0.0001% w/v of the second active agent, or a pharmaceutically acceptable salt thereof, for example, nicotinic acid, nicotinamide or vitamin K, or a combination thereof. The concentration of the second active agent in the Gel Drops may be about 0.00001% — about 0.0001%, 0.000012% — about 0.0001%, 0.000014% — about 0.0001%, 0.000016% — about 0.0001%, 0.000018% — about 0.0001%, 2% — about 0.0001%, 0.00003% — about 0.0001%, 0.00004% — about %, 0.00005% - about 0.0001%, 0.00006% — about 0.0001%, 0.00007% — about 0.0001%, 0.00008% - about 0.0001%, 0.00009% - about 0.0001%, 0.000016% - about 0.00009%, 0.000018% - about 0.00009%, 0.00002% - about 0.00009%, 0.00003% - about 0.00009%, 0.00004% - about 0.00009%, 0.00005% - about 0.00009%, 0.00006% - about 0.00009%, 0.00007% — about 0.00009%, or 0.00008% — about 0.00009% w/v for topical administration. In some embodiments, the Gel Drops e about 0.00001%, 2%, 3%, 0.00004%, 5%, 0.00006%, 0.0000?%, 23%, 0.000081%, 0.000082%, 0.000083%, 0.000084%, 0.000085%, 0.000086%, 0.000087%, 0.000088%, or 0.000089% W/v of the second active agent.
The Gel Drop formulation of the application may r comprise about 0.5 uM, about 0.6 uM, about 0.7 uM, about 0.8 uM, about 0.9 uM, about 1 uM, about 2 uM, about 3 uM, about 4 uM, about 5 uM, about 6 uM, about 7 uM, about 8 uM, or about 9 uM of a second active agent, or a pharmaceutically able salt thereof, for example, nicotinic acid, namide, or vitamin K, or a combination thereof. In some embodiments, the concentration of the second active agent is about 1 "M.
] In some embodiments, the Gel Drop ophthalmic compositions of the present application include glycerin as a tonicity agent. Some embodiments of the application provide lmic composition including mannitol. The glycerin or mannitol t at an amount to t any changes in the solubility of Compound-I, and at a level of about 2.0 — about 2.5%, glycerin provides an osmolality of about 225 — about 300 mOsm/kg depending on the phosphate concentration. In additional embodiments, glycerin is about 2% and phosphate is about 0.05% of the gel drop ophthalmic composition. The concentrations of in and phosphate of the present application is in an amount that the tonicity level of the ophthalmic ition is about 240 mOsm/kg.
The Gel Drop ophthalmic composition of the t application may further include Benzalkonium Chloride (BAK). In some embodiments, the BAK content is about 0.005%, suf?cient for preservation of the ophthalmic composition against microbial ination. In some embodiments ofthe present ation, BAK is not required for use of ophthalmic composition in a sterile, single—use product.
In some embodiments, the Ge] Drop ophthalmic formulation of nd—I includes: about 0.005% — about 2.0% Compound—I or its free base, about 0.05% sodium phosphate, about 2% glycerin as the tonicity adjusting agent, about 0.005% BAK as a preservative, water (puri?ed, z‘.e., distilled, or deionized) as a vehicle, and sodium hydroxide to adjust pH to 6.0. In one embodiment, no other excipients are added.
The Gel Drop of the application comprises about 0.005% - about 2.0% of the active agent of Formula I or II, or a pharmaceutically acceptable salt thereof, for example, Compound-I, and about 0.01% - about 0.05%, about 0.05 - about 0.09%, or about 0.09 - about 0.2% W/V sodium phosphate monobasic monohydrate. In a ic embodiment, the Gel Drop comprises about 0.05%, about 0.05—0.2%, or about 0.2% W/v sodium phosphate monobasic monohydrate buffer. Other equivalent buffer systems well known in the art are also used in the Gel Drop of the present application. In one embodiment, Compound-I or its free base is formulated as about 0.4% — about 2.0% ?rst active agent, about 5% Cremophor RH40, about 2.0% glycerin, and about 0.005% BAK.
] In one embodiment, the Gel Drop of Compound—I includes about 0.3% — about 2.0% (3 —20 mg/mL) Compound—I, about 0.05% — about 0.2% Sodium Phosphate, and about 2% glycerin. The pH of the composition is between pH 5.0 — 7.0.
The Gel Drop ophthalmic formulation of nd—I and a second active agent includes: about 0.005% - about 2.0% nd—I or its free base, about 0.00001% — about % (e.g., about 0.00001% - about 0.0002%, or about 0.00001% - about 0.0001%) second active agent, about 0.05% sodium phosphate, about 2% glycerin as the tonicity adjusting agent, about 0.005% BAK as a vative, water (puri?ed, i.e., distilled, or deionized) as a vehicle, and sodium hydroxide to adjust pH to 6.0. In one ment, no other excipients are added.
The Gel Drop of the ation comprises about 0.005% — about 2.0% of the active agent of Formula I or II, or a pharmaceutically acceptable salt thereof, for e, Compound—I, about 0.00001% — about 5% (e.g., about 0.00001% — about 0.0002%, or about 0.00001% — about 0.0001%) second active agent, and about 0.01% — about 0.05%, about 0.05 — about 0.09%, or about 0.09 — about 0.2% w/v sodium phosphate monobasic monohydrate.
In a speci?c embodiment, the Gel Drop comprises about 0.05%, about 0.05—0.2%, or about 0.2% w/v sodium phosphate monobasic monohydrate buffer. Other lent buffer systems well known in the art are also used in the Gel Drop of the present application. In one embodiment, Compound—I or its free base is ated as about 0.4% — about 2.0% ?rst active agent, about 0.005% — about 5.0% second active agent, about 5% Cremophor RH40, about 2.0% glycerin, and about 0.005% BAK.
In one embodiment, the Gel Drop of Compound—I includes about 0.3% — about 2.0% (3 —20 mg/mL) Compound-I, about 0.005% — about 5.0% second active agent, about 0.05% — about 0.2% Sodium Phosphate, and about 2% glycerin. The pH of the composition is between pH 5.0 — 7.0.
The present application provides Gel Drop of the agents (e.g., the ?rst active agent and the second active agent) of the present application formulated in the presence of excipients such as, without being limiting example, Povidone, polysorbate 80 (P880), polyethylene glycol (PEG) 400, tyloxapol, poloxamer, glycerin, and BAK in a phosphate buffer.
Eye Drops ] Disclosed herein is a formulation, comprising a ?rst active agent, e.g., a compound of Formula I or II, and/or a second active agent, e.g., nicotinic acid, nicotinamide, or vitamin K, or a combination thereof, as eye drops, a form of drug delivery that is pharmaceutically-acceptable to patients, convenient, safe, with an onset of action of several minutes. A standard eye drop used in therapy according to US. federal regulatory practice is sterile, have a pH of about 4, and, if to be used more than once, contains a preservative but has a limited shelf life after g, usually one month. If the eye drops are packaged in a sterile, single use only unit-dose ser, the vative can be omitted.
One method of eye drop formulation comprises the purest forms of the disclosed compound of Formula I or II (e.g, greater than 99% purity) and/or of the second active agent, and the compound and/or the second active agent are mixed with buffer and tonicity adjusters, to adjust for logical pH and osmolarity. es of buffering agents to maintain or adjust pH include, but are not limited to, acetate buffers, citrate buffers, phosphate buffers and borate buffers. Examples of tonicity adjustors are sodium chloride, mannitol and glycerin. In some ments, other pharmaceutically acceptable ingredients are also added.
The formulated solution is then aliquoted into either a plurality of discrete, sterile disposable cartridges each of which is suitable for unit closing, or a single cartridge for unit dosing. Such a single disposable cartridge is, for example, a conical or cylindrical speci?c volume dispenser, with a ner having side—walls able in a radial ion to a longitudinal axis in order to se the container contents therefrom at one end of the container.
The present application provides ophthalmic eye—drop solutions/suspensions packaged in multi-dose form or single dose form, for example, as a plastic bottle with an eyedropper.
In multi-dose form formulations, preservatives are required to prevent ial contamination after opening of the container. le preservatives include, but are not limited to: benzalkonium chloride, thimerosal, chlorobutanol, methyl paraben, propyl paraben, phenylethyl alcohol, edetate um, sorbic acid, polyquatemium-l, or other agents known to those skilled in the art, and all of which are contemplated for use in the present application. Such preservatives are typically employed at a level of from 0.001 to about 1.0% weight/volume.
Without wishing to be bound by theory, the formulation of the present application in an eye drop provides a pulse entry of the drug. The route by which Compound—I obtains access to the posterior segment is not by direct diffusion through the cornea with subsequent diffusion through the aqueous humor, vitreous humor, retina and ultimately the choroid.
Rather, the Compound-I compound achieves notable bioavailability posteriorly following l lation using a circumferential route around, rather than through, the globe.
In certain clinical conditions, the eye drop solutions/suspensions can be formulated with other pharmaceutical agents, in order to attenuate the irritancy of the other ingredient and to facilitate clinical response. Such agents include, but are not limited to, a vasoconstrictor such as phenylephrine, oxymetazoline, napthazoline or tetrahydrozoline; a mast-cell stabilizer such as adine; an antihistamine such as azelastine; an antibiotic such as ycline; a steroidal anti-in?ammatory drug such as betamethasone; a non- steroidal anti—in?ammatory drug such as diclofenac; an modulator such as mod or interferons; and antiviral agents such as valaciclovir, cidofovir and tri?uridine. The doses used for the above described purposes vary, but are in an effective amount to suppress discomfort, itch, irritation, or pain in the eye. When the compositions are dosed topically, the "pharmaceutically effective amount" of a compound of Formula I or II can lly be in a concentration range of from 0.05 mg/mL to about 10 mg/mL and the "pharmaceutically effective amount" of the second active agent can generally be in a concentration range of from 0.05 mg/mL to about 10 mg/mL, with l to 4 drops of the composition administered as a unit dose 1 to 4 times per day. The most common method of ocular drug delivery is the instillation of drops into the cornea (i.e., "eye drops").
A key requirement is that the formulation be sterile and produced in a sterile environment. An ideal disclosed compound for use in ophthalmic solutions/suspensions should be soluble and/or miscible in s media at normal ocular pH and tonicity.
Moreover, the disclosed compounds should be stable, non-toxic, long , and sufficiently potent to counteract dilution of drug concentration by blinking and tearing.
Dosage Forms The formulation of the present application may be suitable for ophthalmic use. In one embodiment the formulation is a solution. The solution of the present application may be a clear, ess, e, isotonic, buffered aqueous free-?owing liquid preparation. The drug t (e. g., the ?rst active agent and/or the second active agent) has a pH of imately 6.0 and may be stored at +5 °C. The drug product may be provided in a container e system consisting of a semi—transparent ophthalmic dispenser bottle with a dropper tip and cap.
In some embodiments, the clinical concentration of Compound-I ophthalmic solution or suspension is equal to or less than about 0.1 mg/mL, equal to or less than about 0.2 mg/mL, about 0.2 — about 1.0 mg/mL, about 0.3 — about 1.0 mg/mL, about 0.4 — about 1.0 mg/mL, about 0.5 — about 1.0 mg/mL, about 0.6 — about 1.0 mg/mL, about 0.7 — about 1.0 mg/mL, about 0.8 —about 1.0 mg/mL, about 0.9 — about 1.0 mg/mL, about 1.0 — about 2.0 mg/mL, about 2.0 — about 3.0 mg/mL, about 3.0 — about 4.0 mg/mL, about 4.0 — about 5.0 mg/mL, about 5.0 — about 6.0 mg/mL, about 5.0 — about 10.0 mg/mL, about 10 — about 20 mg/mL, about 20 — about 30 mg/mL, about 30 — about 40 mg/mL, or about 40 — about 50 mg/mL.
In other embodiments, the clinical concentrations of Compound-I and the second active agent ophthalmic solution or sion are independently equal to or less than about 0.1 mg/mL, equal to or less than about 0.2 mg/mL, about 0.2 — about 1.0 mg/mL, about 0.3 — about 1.0 mg/mL, about 0.4 — about 1.0 mg/mL, about 0.5 — about 1.0 mg/mL, about 0.6 — about 1.0 mg/mL, about 0.7 — about 1.0 mg/mL, about 0.8 —about 1.0 mg/mL, about 0.9 — about 1.0 mg/mL, about 1.0 — about 2.0 mg/mL, about 2.0 — about 3.0 mg/mL, about 3.0 — about 4.0 mg/mL, about 4.0 — about 5.0 mg/mL, about 5.0 — about 6.0 mg/mL, about 5.0 — about 10.0 mg/mL, about 10 — about 20 mg/mL, about 20 — about 30 mg/mL, about 30 — about 40 mg/mL, or about 40 — about 50 mg/mL.
In one embodiment of the present application the strength of the compound of FormulaI or II is about 0.005% — about 5.0% (about 0.5 — about 50 mg/mL). A d cologic activity (or concentration) of the formulation of the present ation against pathologic choroidal and retinal neovascularization is achieved following ocular administration of formulations containing about 0.005%, about 0.05%, about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1.0%, about 2.0%, about 3.0%, about 4.0%, or about 5.0% w/v of Compound—1.
The present application provides that, following l ocular administration with an l dose (for example, between about 0.005% and about 5.0%) the pharmacologically active concentration is achieved and ined in the central choroid target tissue. In one embodiment the ?rst active agent (Formula II or Compound—I) is formulated as about 0.005 — about 5.0% w/v concentration, the second active agent (nicotinic acid, nicotinamide, or vitamin K, or a combination thereof) is formulated as about 0.005 — about 5.0% w/V concentration, and the combination is dosed once or twice a day per eye for more than 60 consecutive days. The plasma concentrations observed following topical administration are substantially below the level expected to produce systemic toxicity.
Table 2: In vitro Summary of pharmacodynamic properties for Compound—I ICso=nM n_/mL Inhibition of recombinant VEGFR-2 tyrosine kinase using exogenous ate 10.55 (6) Inhibition of recombinant FGFR-2 tyrosine kinase using exogenous ate 8.79 (5) Inhibition of recombinant PDGFR tyrosine kinase using ous substrate 2636.67 (1500) Inhibition of recombinant EGFR tyrosine kinase using exogenous substrate 0 (3330) Inhibition of recombinant IR tyrosine kinase using exo enous substrate 10283.00 5850 Inhibition of VEGF-stimulated VEGFR-2 _ auto hos oho lation in intact cells 5.27 3 Inhibition of VEGF—stimulated mitogenesis in HUVECs 14.06 (8) In some embodiments, Compound—I exhibits potent inhibition of tyrosine kinase activity for several proangiogenic growth factor receptors, with IC50 of less than about 100nM (see Table 3). Compound—I also blocks the high—af?nity VEGF receptors, e. g., VEGFR-l/Flt-l, but with lower potency (with IC50 of about 122nM (69.41 ng/mL)).
Table 3: In Vitro Inhibition of Tyrosine Kinases using a 10-point Titration Curve (257nM — 5000nM) for Compound—I Although VEGFR inhibition appears to be essential for ng vascular permeability and preventing further neovascular growth, the simultaneous tion of VEGF signaling with inhibition of other growth factor signaling pathways (e.g., PDGF and angiopoietins/Tie2) may be linked to unique eutic outcomes. The eutic outcomes of a broader inhibition of ing pathways may contribute to the regression of newly ished pathologic vessels in the posterior segment of the eye.
In some embodiments, about 300nM (about 170.67 ng/mL) of Compound—I inhibits VEGFR-2 kinase function (see Table 4). Substantial blockade of a similar set of proangiogenic growth factor receptors, including -3, Tie-2, and EphB-4 are also observed. An unexpected ?nding is that about 300 nM concentration of Compound—I inhibits the VEGFR-2 kinase function, which falls within the l range found in the central choroid and retina following ?ve days of topical ocular delivery.
Table 4: In Vitro Inhibition of Tyrosine Kinases by 300nM 7 ng/rnL) Compound—1 FLT4 VEGFR-3 __ KDR VEGFR—Z 104 RET Y791F mutation TEK Tie2 Table 5: In vitro Inhibition of Tyrosine Kinases by 1 uM (568.9 ng/mL) Compound—I ABLl c[\9 ABLl E255K ABLl G250E ABLl T3151 ABLl Y253F \0DJ ACVRlB ALK4 AURKB Aurora B BRAF V599E EPHA-l EPHA-8 00000000 {hp—1mm EPHB-l 00 U) EPHB-4 FGFR-l FGFR-2 FGFR-3 FGFR-3 K650E "11GR 05HO FLT-1 VEGFR-l FLT-4 VEGFR-3 \oU] KDR VEGFR-2 LYN A LYN B MAP4K4 HGK 5000\0Obit—Ian MAP4K5 KHSl L MAPK14 (p38 aluha MlNKl >—| oo PDGFRA T6741 PTK6 Brk m 00 RET Y791F SNF1LK2 SRC N1 TEK Tie2 YES 1 Overview ofDrug Substance and Drug Product Drug Product: Compound-I and/0r second active agent ophthalmic formulations for clinical studies are manufactured in dosage strengths n 0.05% - 1.0% of Compound—I and about 0.00001% — about 5% (e.g., about 0.00001% - about 0.0002%, or about 0.00001% — about 0.0001%) the second active agent. In some embodiments, Compound—I dosage in the formulation is 0.05%, 0.1%, 0.2%, 0.3%, 0.4%, 0.6%, 0.8%, or 1.0%. In some embodiments, second active agent dosage in the formulation is 0.05%, 0.1%, 0.2%, 0.3%, 0.4%, 0.6%, 0.8%, 1.0%, 2.0%, 3.0%, 4.0% or 5.0%. Compound—I and/or second active agent Ophthalmic Formulations (solutions or suspensions) are for daily, single use, topical administration to the eye in a clinical setting. In addition to the ?rst and second active ingredients, in some embodiments the drug product may further contain about 0.005% BAK as a preservative, puri?ed water as vehicle, and is pH—adjusted with sodium hydroxide to pH 6.0.
Sodium Phosphate-based Gel Drop The lmic bene?ts of Compound—I in a sodium phosphate-based formulation (listed in Table 6) results from the self—gelling properties of the API in buffers, such as sodium ate. Spontaneous formation of self—forming, thixotropic gel of Compound-I from a clear solution is formed by increasing ?rst active agent concentration in sodium phosphate. Once the ?rst active agent concentration in the phosphate buffer reaches super— saturated state, insoluble particulates of Compound—I are observed within the gel.
The current state of the art predicts that ation of a gel with sed viscosity to the surface of the eye would se corneal residence time. Increased corneal residence time in turn tates ocular drug tion. As a result, the intraocular drug concentrations of Viscous gels would be increased in comparison to non-viscous formulations, such as water—like solutions. One way to increase ity is to use various viscosity—enhancing excipients, e.g, carboxymethylcellulose, which in effect achieves increased cular absorption of different drug substances ing topical ocular administration. The present application provides a thixotropic gel of nd-I and/or a second active agent formed in the absence of any viscosity—enhancing excipients. For example, when Compound—I or Compound—I and a second active agent are dissolved into a simple buffer, such as sodium phosphate, a ropic gel is formed. The thixotropic gel, which is formed without any viscosity—enhancing ents, is formulated as a Gel Drop.
The present application provides ependent and requency dependent delivery of Compound-I to the posterior segment eye tissues.
] The Gel Drop formulations of the present application d in Table 6) differ among each other in several aspects, such as ?rst active concentration, sodium phosphate concentration, presence or absence of tonicity (glycerin) or preservative (benzalkoniumchloride/BAK) agents, solubilizing surfactants (polysorbate 80, tyloxapol, and/or poloxamer), and pH.
Tromethamine-based suspension The present application provides a suspension of Compound—I and/or a second active agent in a tromethamine—based formulation. In some embodiments, the suspension of nd—I and/or the second active agent in a tromethamine-based formulation has equal to or more than 95% of the first active drug nce in an insoluble form. This characteristic is distinguishable from the soluble or semi—soluble state of Compound-I in the Gel Drop (the Gel Drop (gel), which is not an entirely soluble state as concentration of the first active agent increases) or in a Cyclodextrin-based formulation. Tromethamine-based formulations of Compound—I show increased turbidity with increasing ?rst active agent concentration.
Administering a topical drop of Compound—I and/or second active agent suspension to the eye, which is a ation of soluble and insoluble ?rst active agent components, are bene?cial with respect to both safety/tolerability and efficacy.
The present application provides Compound—l in the tromethamine-based suspension, delivered at concentrations to the target tissues between 10—1000x of the cellular IC50 for the various pro-angiogenic RTKs. See, e.g., Table 7.
The corneal safety and tolerability of topical nd-I is a direct uence of the amount of soluble (as opposed to insoluble) ?rst active agent applied to the corneal surface, and the resultant corneal tissue concentration. In some embodiments, subjects who receive topical ocular administration of the tromethamine—based sion are able to tolerate up to higher level of the first active agent concentration in the formulation, as compared to equimolar formulations of the sodium phosphate—based Gel Drop. The corneal safety and tolerability of topical Compound—I is also a uence of the administration of a second active agent, e.g., nicotinic acid, nicotinamide, or Vitamin K, or a combination f, which is a modulator (e.g., activator) of EGFR that prevents or treats corneal disruptions or diseases caused by inhibition of EGFR. [plasma] nM .6 .5 3 6 6 4 1 7 9 1 7 1 9 2 5 5 1 5 1 2 .4 8 .0 8 4 .4 .8 5 5 .4 4 .4 .7 5 5 .7 3 .9 .8 4 2 .8 .9 5 4 .4 4.94 4 .1 SD [AH] nM .2 4 4 1 .3 9 6 6 8 8 3 .0 .4 5 .8 1 1 1 5 .6 4 .1 .0 1 6 .0 !Mean [AH] nM .9 .4 .l .8 .5 .8 .2 .9 .6 .7 .8 .4 .1 Q 2 2 5 0 5 5 Q l 3 1 Q 5 3 0 Q 5 1 4 3 2 1 8 Q 0 1 3 .2 7 6 Q 7 1 6 1 7 19.7 Drop SD a] nM .2 .6 .4 .5 .7 .5 .8 .8 .1 3 0 2 8 53 9 0 0 Q 6 4 2 5 2 3 7 3 0 Phosphate-based Gel Mean [retina] nM .6 .9 .3 o L .8 .9 .8 .7 1 1 9 7 9 2 6 3 6 8 < L 1 2 3 4 1 1 4 2 9 0 3 2 1 2 1 0 102 SD nM 0 5 5 * .4 .1 .6 1 5 8 6 2 2 1 0 2 3 1 0 5 1 .3 4 4 3 4 1 8 0 0 No.: PANO-003/00lUS [MEAN [choroid] [choroid] ID M 2 .4 6 3 9 8 0 0 2 9 6 9 7 2 8 5 9 6 2 3 2 8 2 9 9 Q 1 1 8 2 7 7 7 6 7 6 6 6 1 6 9 5 5 9 5 8 5 3 5 3 5 2 5 1 5 2 6 2 357 ? ? 6 ? ? 2 3 9 3 6 3 5 5 in Sodium [Dose x per [Day 3 x 3 x 3 x 3 x 3 x 3 x 3 x x 3 x 3 x 3 x 3 x 3 x 3 x 3 3 x x 3 x x 3 3 x x 3 x 3 x 3 x 3 x 3 3x 4 Attorney Docket Days Dosing 5 5 5 5 5 5 5 5 5 5 5 4 5 4 5 4 5 5 5 5 5 5 5 5 3 8 7 9 8 4 9 4 3 1 3 4 of Compound-I Osmo 2 0 Q 7 0 Q 7 8 0 4 Q 5 Q Q 3 8 Q 3 3 3 4 Q3 2 Polox Q Q Q Q81 % 0 .2 .2 0 0 .2 ocular formulation rrylox % .1 .1 .2 0 .1 0 .2 0 0 .2 0 0 % ' 1 Gly [PS80 1 1 1 1 1 1 1 % 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 2 5 with topical BAK 5 5 5 5 5 5 5 5 5 5 5 5 5 5 5 5 5 .0 0 0 0 5 0 5 0 5 .10 .0 0 0 .0 0 .0 0 0 .0 0 2 .0 0 0 .0 0 0 .0 0 0 .0 0 .0 0 0 .0 0 50 .0 0 0 .0 .0 0 5 .0 0 0 .0 0 0 .0 0 % .0 0 0.0 .0 0 5 0 .0 0 0 .0 0 0 0.005 0 .1 0 .0 5 0 2 0 .0 5 .20 0 0 .0 0 .20 .0 5 0 .20 0 0 .0 0 .1 0 .2 .1 0 .20 0 0 .0 5 Phos 0.0 .0 5 0 0 .20 0 .0 5 0.2 % 5 .9 .1 PK results 6 .9 5 .0 6 .1 6 6 6 .0 .0 6 6 6 .1 6 6 .0 6 .0 .0 6 .0 6 .1 6 6 .1 6 .0 6 6 5 .9 6 6 pH 5.9 6 6 6 6 4 2 5 2 4 2 2 1 2 2 2 1 2 2 1 2 1 2 5 1 2 CD* mg/ml 6 Table 6: 121610498 vl [plasma] nM .3 6 6 9 4 5 .2 3 .4 SD [AH] M 1 9 .9 8 6 0 .9 !Mean nM n 8 .2 .5 9 1 Q 9 .0 9 SD nM .6 9 .1 1 5 4 Mean a] [retina] [AH] nM .7 Q o 5 L .8 8 L < 4 6 SD nM .1 4 .7 4 4 8 No.: PANO-003/00lUS [choroid] [choroid] lllM 6 9 9 6 5 3 5 3 4 3 3 3 1 9 Q !Dose x !MEAN [Day x 3 x 3 x 3 3x 3 x 3 x Attorney Docket Days Osmo Dosing per 5 5 5 5 5 5 2 Q 7 3 7 Q Q 5 Q 54 rrylox Polox % 0 .2 % .2 0 0 .1 0 .2 % 1 1 .0 Gly IPS80 1 1 0 % 2 2 2 2 2 % 5 0 5 5 5 .0 0 0 .0 0 50 0 .0 20 Phos BAK .0 1 0 .1 .0 0 0 .0 0 0.05 0.005 pH .1 6 6 .0 6 6 .0 6 6 2 2 1 2 1 1 CD* mg/ml * CD: Compound 121610498 vl [plasma] nM .3 3 1 8 8 .2 7 9 9 1 1 9 .2 .3 8 . l 8 .4 5 5 .6 0 1 7 .8 4 .9 .3 3 .4 5 2 .6 l.91 SD [AH] nM 0 9 3 3 5 1 5 7 2 1 .5 3 9 .4 .7 6 6 .5 9 3 9 .5 0 .5 .1 6 4 .3 0.41 Mean [AH] nM 0 5 4 1 1 9 .2 .4 .4 .3 .9 .3 .5 .1 .9 .9 .7 3 0 6 8 0 4 2 7 3 4 2 2 5 5 5 0 1 4 l2 9 1 1 5 3.07 SD [retina] nM .7 2 .8 8 .2 5 .7 .4 .7 .4 7 1 3 4 2 1 7 5 2 8 4 6 3 5 6 3 36.9 Mean [retina] nM 0 Q 1 1 9 9 9 3 o 2 5 1 1 3 6 1 2 1 1 1 .8 .5 .5 7 9 1 5 L 1 < L 3 18.5 SD [choroid] nM 1 7 3 6 7 .1 .4 1 .6 5 5 9 3 2 0 2 2 1 2 1 9 5 5 1 2 6 4 27.8 nM 0 2 0 0 5 1 9 4 0 8 2 5 0 8 1 0 4 6 6 5 2 1 6 9 9 1 7 7 7 5 7 0 6 8 7 5 4 5 4 1 4 0 3 5 3 2 2 8 202 No.: PANO-003/00lUS MEAN 1 1 1 Dose [choroid] X per Dav 3 x ,.., ? )X x 3 ' ? "IX 3 x 3 x x 3 3 x ,.., ? )X 3 x ' ? "IX x 3 3 x x 3 x 3 ' ? "IX 3x Attorney Docket tromethamine-based sion Days Dosin g 5 5 5 4 5 5 5 4 5 5 5 5 5 5 5 5 5 4 6 0 4 1 6 2 Compound-I in Osmo 4 1 3 6 8 3 1 2 2 2 7 1 3 3 % .2 0 PS80 Tylox Polox % .2 .2 0 0 0 .2 0 .1 0.2 1 1 1 1 1 topical ocular % Gly 2 2 2 2 2 2 % 2 2 2 2 2 2 2 2 2 2 2 BAK 5 5 0 5 5 5 5 5 5 5 5 5 5 5 5 5 5 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 .0 .0 0 % .0 0 .0 0 0 .0 .0 0 0 .0 0 .0 0 .0 0 .0 .0 0 0 .0 0 .0 0 .0 0.005 0 results with 0 Tris l .2 0 0 0 0 0 0 0 0 -0 .2 --0 0 .6 , 0 .2 "0 .2 1 l .2 .2 0 -0 .2 0 .6 .2 .2 0 .6 0 .2 -0 i ~ -0 ; ? ~ --0 , ~ ?0.20 .1 4 4 4 % % 4 '% % '% % 4 '% 0 .1 0 .1 0 4 .1 4 .l .1 0 .1 4 4 .1 .1 4 % 4 .1 .1 0 4 .1 Phos 6 .0 0 0 0 0 0 0 0 0.14%; 6 .0 6 6 5 .0 .0 7 6 6 6 .0 .0 6 pH .0 6 .1 6 6 .0 6 6 6 .0 6.0 0 Table 7: PK * CD: Compound-I 121610498 vl 1 5 4 6 4 4 5 6 4 5 2 2 ') I.., 2 2 1 1 m CD* mg/ L The present application provides ocular bioavailability of Compound-I in the posterior t upon administration of a tromethamine-based suspension. The ocular ilability of Compound-I in the ior segment is directly proportional to the total amount of drug, Compound—I, administered (insoluble plus soluble, see Table 7). Although the insoluble drug particulates are not readily available to anterior segment tissues; the inherent and unique physicochemical properties of Compound—I allow both insoluble and e components to gain entry to posterior segment tissues, such as the choroid and retina.
Consequently, even higher drug concentrations than those achieved with Gel Drop formulations containing equivalent amounts of the ?rst active agent are achieved with the tromethamine—based suspension. Thus, tromethamine—based suspension provide: a) improved corneal tolerability and b) ined or increased bioavailability to the posterior segment, particularly to the choroid, the primary target tissue for treating cular (wet) AMD. In addition, a second active agent, e.g, nicotinic acid, nicotinamide, or vitamin K, or a combination thereof, which is a modulator (e.g., activator) of EGFR that prevents or treats l disruptions or es caused by inhibition of EGFR, prevents and/or treats corneal disruptions potentially associated with the administration of Compound-I, thereby sing the therapeutic index of nd—I. extrin-based solution Cyclodextrins, which are cyclic oligosaccharides made up of six to eight dextrose units (0-, [3—, and y—CDs) joined through one to four bonds, are well-known for their ability to act as a solubilizing agent for relatively insoluble drugs. See Stella & He, Cyclodextrins, Toxicol. Pathol, 36: 30—42 (2008).
In some embodiments, 2-hydroxypropy1—B—cyclodextrin CD, also known as KLEPTOSE® HPB) at equal to or more than 1:6 molar ratio or Sulfobutylether-B— cyclodextrin (SBE-B-CD, also known as CAPTISOL®) at equal to or more than 1:2 ratio in the proposed clinical formulation, Compound—I or its free base and a second active agent Ophthalmic Solution, provide solubility that meets clinical dose strengths of 0. 1—1.2% Compound-I.
In some embodiments, cyclodextrin-based solutions of Compound-I or its free base and/or a second active agent not only have ed solubility of the ?rst active agent into a uniform solution, but, upon topical ocular administration, also have a novel and previously unobserved characteristic of signi?cantly sed therapeutic index of the ?rst active agent at the posterior segment of the eye. The solutions of Compound—I and/or the second active agent of the present application reduce anterior t exposure of Compound-I, thereby increasing the concentration of the first active in the solution and increasing the frequency of its ry in order to maintain high ior t concentrations. Both of these bene?cial characteristics are related to the known ty of cyclodextrin to form hydrophilic complexes with hydrophobic drugs. See Stella & He, Cyclodextrins, Toxicol. Pathol, 36: 30—42 (2008). The administration of the second active agent as a combination with the ?rst active agent prevents corneal disruptions or es caused by inhibition of EGFR by systemic diseases (e.g., cancer, diabetes), eye diseases, or administration of the first active agent, e.g., a nd of Formula I or II, thereby increasing the therapeutic index of the first active agent, e.g., Compounds of Formula I or II.
When formulated with nd—I or its free base and/or a second active agent, cyclodextrin can form a clear, colorless solution which exhibits water-like ity. ing l ocular administration, Compound—I/cyclodextrin complex has the appearance of being pharmacologically inactive and metabolically inert. The Compound- I/cyclodextrin complex confers corneal bility until cyclodextrin spontaneously dissociates from the first active agent, thus making available high concentration of Compound—I at its intended site of action in the posterior segment of the eye, e.g., choroid and retina.
In some embodiments, cyclodextrin—based solutions of Compound—I lower corneal exposures of Compound—I compared to Gel Drop formulations at similar drug concentrations.
The use of cyclodextrin—based solutions of Compound—I provides about 10x reduction in corneal trations, as compared to dosing with equimolar formulations of the Gel Drop.
In some embodiments, after 20 — 30 days of topical ocular dosing of about 0.2 — 2.0%, e.g., about 0.6%, Compound—I as a cyclodextrin—based solution, no untoward findings are attributed to test-article or vehicle. The present application es higher concentrations of Compound-I within the posterior segment target tissues, such as at the central choroid and the central retina, when cyclodextrin-based solution of Compound—I is topically applied. In some embodiments, the combined effects of decreasing corneal drug exposure so as to avoid poor ocular bility, while increasing posterior t bioavailability so as to increase RTK inhibition, significantly increases the therapeutic index and corresponding bene?t(s) to treated subjects. In other embodiments, the combined effects of decreasing corneal drug exposure so as to avoid poor ocular tolerability and preventing or treating of corneal disruptions or disease by the second active agent, while increasing posterior segment bioavailability so as to increase RTK inhibition can significantly increase the therapeutic index and corresponding benefit(s) to treated subjects.
The present application provides expansion of the therapeutic window for both suspension—based formulations (see Example 3) and the cyclodextrin formulations of Compound—I due to signi?cantly reduced exposure (about 10—100X or 1—2 log reduction).
The reduced exposure improves corneal safety/tolerability, which allows higher concentrations or frequency of dosing of nd—I to be administered topically. The higher tration enables nd—I to achieve higher back of the eye target tissue concentration, which es the therapeutic ef?cacy of Compound—I.
In some embodiments, topical ocular dosing of ophthalmic gel drops is associated with high corneal tissue exposure M) and corresponding untoward observations in the anterior segment, such as discomfort, corneal and conjunctival in?ammation, corneal epithelial n and/or thinning and degeneration. In contrast, repeated topical ocular dosing of Compound—I ophthalmic solution produces corneal exposure that are roughly 5 tolO-fold lower than an lar dose of lmic gel drops, and are free of untoward clinical or histopathologic ?ndings. Topical ocular dosing with Compound-l ophthalmic solution also achieves equal or higher target therapeutic exposure in the l choroid in comparison to an equimolar dose of the ophthalmic gel drop. Overall, the combination of decreased corneal exposure and corresponding improved ocular tolerability, while simultaneously ining or promoting drug delivery to the posterior segment target tissues, along with improved physicochemical stability, provides greater bene?t to subjects compared to the ophthalmic Gel Drop formulation. 1- to 5-day PK results with topical ocular Compound-I in cyclodextrin-based solutions The present application provides ocular pharmacokinetics of various formulations and dose ns of Compound—I following topical ocular administration. Three dosage strengths in nine (9) ent topical ocular formulations of Compound—I are used for dosing either once per day (q.d.) or twice per day (b.i.d.) for 1, 2, 3, 4, or 5 consecutive days.
Subjects each receive about 30 uL bilateral topical ocular dose of one of three (3) Compound—I formulations, or vehicle ation, using a positive displacement pipette.
The composition of each Compound-I formulation is bed in Table 8A. All doses were administered within i 1 hour of the scheduled dose time. On day 1, Groups 1, 2, 4-6, 8, 10, 11, 13, 15, and 17 receive one dose (q.d.) for either one (1) or four (4) days. On days 1 through 4, Groups 3, 7, 9, 12, 14, and 16 receive b.i.d. dosing imately 8 hours apart at 7:00 AM and 3:00 PM for four (4) days. Some subjects receive b.i.d dosing of vehicle only formulations for ?ve (5) consecutive days.
In some ments, ocular sampling is performed at about 0.5, about 1, about 2, about 3, about 4, about 5, about 6, about 7, about 8, about 9, about 10, about 11, about 12, about 13, about 14, about 15, about 16, about 17, about 18, about 19, about 20, about 21, about 22, about 23, or about 24 hours post—dose relative to the day 1 dose. Aqueous humor, cornea, central and peripheral retina, and central and peripheral choroid samples are collected to monitor effects of treatment. Aqueous humor, , central retina, and l choroid s are d.
Table 8A-C lists 1- to 5-day PK results with topical ocular Compound-I in Cyclodextrin-based ons.
Table 8A: Ocular Formulations Corn n osition: 0.3% Comnound-I 3 m mL Comnound-I 0.05% Sodium Phosphate, monobasic, monohydrate, 2.0% glycerin, USP Physical Description: Clear and colorless, extremely viscous Comn osition: 0.3% Comnound—I 3 m mL nd-I 0.05% Sodium Phosphate, monobasic, monohydrate, 2.0% glycerin, USP pH 5.5 Ph sical Descri ntion: Clear and colorless Composition: 0.4% Compound—I (4 mg/mL Compound-I) 7% Hydroxypropyl—B—cyclodextrin (HPBCD), 0.7% Sodium Chloride, USP, 0.005% Benzalkonium chloride (BAK), NF pH 7.0 Ph sical Descri ntion: Clear and colorless, viscous Composition: 0.4% Compound—I (4 mg/mL Compound-I) 4% Hydroxypropyl—B—cyclodextrin (HPBCD), 0.7% Sodium chloride, USP, 0.005% Benzalkonium chloride (BAK), NF pH 7.0 Ph sical Descri ntion: Clear and colorless, viscous Comn osition: 0.4% Comnound—I 4 m; mL Comnound—I 4% Hydroxypropyl—B—cyclodextrin (HPBCD), 0.7% Sodium Chloride, USP, 0.005% Benzalkonium de (BAK), NF Lot : BCL532-052 5 ALG-OOl Ph sical Descrintion: Clear and colorless, extreme] viscous Comn osition: 0.4% Comnound-I 4 m mL Comnound-I 7% Hydroxypropyl-B-cyclodextrin (HPBCD), 1% Tromethamine, USP, 0.4% Sodium Chloride, USP, 0.005% Benzalkonium chloride (BAK), NF, pH 7.0 Ph sical Descri ntion: Clear and colorless Composition: 0.6% Compound—I (6 mg/mL Compound-I) 7% ypropyl-B-cyclodextrin (HPBCD), 0.7% Sodium Chloride, USP, 0.005% Benzalkonium chloride (BAK), NF 0H 7.0 Physical Description: Clear and colorless, viscous Como osition: 0.6% d—I 6 111; mL Commund-I 7% Hydroxypropyl—B—cyclodextrin ), 0.7% Sodium Chloride, USP, 0.005% Benzalkonium chloride (BAK), NF, 0 H 6.0 Ph sical Descri otion: Clear and colorless, viscous Comosition: 0.4% Com-ound-I 4 m_ mL Comound-I % Cremophor RH40, 2.0% glycerin, USP, 0.005% Benzalkonium chloride BAK H 6.0 , NF, Ph sical tion: Clear and colorless Table 8B lists average nd—I concentrations in aqueous humor, retina, choroid, and cornea (LLOQ: Lower Limit of Quantitation; the LLOQ is the lowest analyte concentration that can be quanti?ed with acceptable precision and accuracy).
Table 8B Avera_e Concentration of Com 1 ound-I ( M) Group $311111: Aqueous Central Peripheral Central Peripheral Cornea Humor Retina Retina Choroid Choroid 1 hr 6 0.0363 0.0961 *0207 0.737 112 2 8 hr 0.0165 0.0380 0.0508 *0237 0.687 32.7 24 hr 0.00336 0.380 *0.205 1.14 29.2 0.0142 0.0407 0.108 0.255 0.765 151 24hr 0.00774 0.0292 0.0597 0.283 0.892 82.1 0.00996 0.0431 0.0883 0.196 0.629 78.7 1 hr 0.00323 0.0463 0.0634 0.319 0.311 21.8 6 8 hr 0.00742 0.0537 0.0349 24 hr 0.00122 0.0241 0.0533 0.00648 0.0469 0.0819 0.514 0.744 35.3 24hr 0.00260 0.0313 0.0293 0.439 0.653 13.0 Average Concentration of nd-I (uM) Group Aqueous Central eral Central Perlpheral Point Cornea Humor Retina Retina Choroid Choroid 0.00978 0.0490 63.2 24hr 0.00483 0.0193 37.4 0.0633 237 1hr 0.00867 0.0667 93.9 AH LLOQ = 0.000903 uM l Retina LLOQ = 0.0181 LLM Peripheral Retina LLOQ = 0.00873 LLM (Grps 1-8); LLOQ = 0.00898 uM (Grps 12-16) Central Choroid LLOQ = 0.175 LLM Peripheral Choroid LLOQ = 0.0349 LLM (Grps 1-8); LLOQ = 00359 M (Grp 12-16) Cornea LLOQ = 0.0181 uM (Grp 1-5); LLOQ = 0.0453 uM (Grp -13,15,16A17); LLOQ = 00873 "M (Grp 4,9,16B) N/A = Not Applicable; Samples not assayed per study protocol.
*Average based on n=1.
] Table 8C lists the summary of average ocular tissue concentrations of Compound— I in aqueous humor, central and peripheral retina, central and peripheral choroid, and cornea for Groups 1 through 10. Any values When all values are Table 8C Average Concentration of Compound-I (uM) Group Aqueous Central Peripheral Central Peripheral Point Cornea Humor Retina Retina Choroid Choroid 1 hr 0.00284 0.0397 N/A 0.193 N/A 20.7 24 hr *000205 *0.0185 N/A *0.179 N/A 0.487 12 1hr 0.00651 I 00521 | 0.0842 I 0.528 0.560 | 27.4 1hr 0.0102 0.0934 0.372 N/A 123 24hr 0.00518 0.0246 0.319 N/A 39.1 14 1 hr 0.0209 0.0817 0.151 7.19 1.00 236 1hr 0.0114 I 00527 | N/A | 0.319 N/A | 82.9 16 1 hr 0.0179 0.0480 0.169 0.495 0.868 169 17 1hr 0.00445 0.0468 0.297 N/A 32.0 AH LLOQ = 0.000903 uM Central Retina LLOQ = 0.0181 LLM Peripheral Retina LLOQ = 0.00873 LLM (Grps 1—8); LLOQ = 0.00898 11M (Grps 12-16) Central d LLOQ = 0.175 uM Peripheral Choroid LLOQ = 0.0349 11M (Grps 1-8); LLOQ = 0.0359 11M (Grp 12-16) Cornea LLOQ = 0.0181 uM (Grp 1-5); LLOQ = 0.0453 uM (Grp -13,15,16A17); LLOQ = 0.0873 11M (Grp 4,9,16B) N/A = Not Applicable; Samples not assayed per study protocol.
*Average based on n=1. -day PK results with topical ocular Compound-I in cyclodextrin-based ons The present application provides ocular pharmacokinetics of various dose regimens of topical ocular solutions of Compound—I containing hydroxypropyl-B- cyclodextrin ("HDBCD") following ocular dose administration. Different l ocular ons of Compound—l are administered either once per day (q.d.) or twice per day (b.i.d.) for either 4 or 5 consecutive days. Subjects each e a 30 [LL bilateral topical ocular dose of one of four Compound-I dosage strengths.
] All doses were administered with i 1 hour of the led dose time, except some subjects receiving on day l. Ocular sampling after administration of Compound-I is performed one hour following the ?rst daily dose on day 5 for subjects, except in a few, where ocular sampling is performed 24 hours after the ?rst daily dose on day 4.
Aqueous humor, cornea, central and eral retina, and central and peripheral choroid samples are collected. Cornea, central , and central choroid samples are assayed; aqueous humor, peripheral retina, and peripheral choroid samples are not assayed.
Table 9 (A-B) lists 5-day PK s with topical ocular Compound-l in cyclodextrin—based solutions.
Table 9A: Ocular Formulations Formulation 1 (A) Composition: 0.4% Compound-I (as free base) 7.15% Hydroxypropyl-B-cyclodextrin 0.7% Sodium chloride 0 H 6.5 Physical Descri otion: Clear and ess Formulation 2 (B) Composition: 0.1% Compound-I (as free base) 1.79% Hydroxypropyl-B-cyclodextrin 0.85% Sodium chloride pH 6.5 Physical -tion: Clear and colorless Formulation 3 (C) Composition: 0.2% Compound—I (as free base) 3.57% Hydroxypropyl—B—cyclodextrin 0.8% Sodium chloride H 6.5 Physical Descrition: Clear and colorless Formulation 4 (D) Composition: 0.6% Compound-I (as free base) .72% Hydroxypropyl-B-cyclodextrin 0.6% Sodium chloride 0 H 6.5 Physical Description: Clear and colorless Formulation 5 (E) ition: 0.4% Comnound-I as free base 8.41% ypropyl-B-cyclodextrin 0.65% Sodium chloride n H 6.5 Physical Descri ntion: Clear and colorless ation 6 (F) Composition: 0.4% Compound-I (as free base) .5 1% Hydroxypropyl-B-cyclodextrin 0.65% Sodium chloride n H 6.5 Physical Descrintion: Clear and colorless Formulation 7 (G) Comnosition: 0.4% Com ound-I as free base .5 1% Hydroxypropyl-B-cyclodextrin 0.15% Sodium de 1.0% Tromethamine (Tris) H 6.5 Physical Description: Clear and colorless Formulation 8 (H) Comnosition: 0.1% Comnound-I as free base 2.63% Hydroxypropyl-B-cyclodextrin 0.8% Sodium chloride 0 H 6.5 Physical Description: Clear and colorless Formulation 9 (I) Comnosition: 0.6% Comnound-I as free base .77% ypropyl-B-cyclodextrin 0.37% Sodium chloride pH 6.5 Physical Description: Clear and colorless Table 9B lists a summary of average ocular tissue concentrations of Compound—I in central retina, central choroid, and cornea. Any values statistical calculations. When all values were reported as the average.
Table 9B: Average Compound-I concentrations in retina, choroid, and cornea. ' e tration ( M) Time ' Grou . Central Central 0.772 n———10.6 m———0.371 0.0332 7-60 Avera e Concentration (uM) Group Central Central Point Retina Choroid "———— Central Retina LLOQ = 0.0218 uM Central Choroid LLOQ = 0.174 uM Cornea LLOQ = 0.0174 uM *Average based on n=1 Concentrations of nd-I (in MW) in various ocular?uids and tissues In some embodiments, concentration of the ?rst active agent in various tissues and ?uids of the eye is measured upon topical ocular stration of a solution of about 0.4% (about 4 mg/mL) Compound—I and cyclodextrin. Average concentration of Compound-I is measured in the central choroid, central retina, aqueous humor, and cornea. Compound—I is in a solution (0.4% or 4 mg/mL) with 8.41% KLEPTOSE® and 0.142% ate buffer; 8.9% KLEPTOSE® HPB and 0.142% phosphate; 4.88% CAPTISOL® and 0.142% phosphate; or 4.88% CAPTISOL® and 0.122% phosphate. See Table 10A—B.
] In some embodiments, upon topical ocular administration of a solution of about 0.4% (about 4 mg/mL) Compound-I and cyclodextrin, the central choroid concentration of Compound—I is between about 0.2 uM and about 0.8 uM. The central retina concentration of Compound—I is between about 0.05 uM — about 0.15 uM. In some ments, upon topical ocular administration of a solution of about 0.4% (about 4 mg/mL) nd—I and cyclodextrin, the aqueous humor concentration of Compound—I is between about 0.003 uM — about 0.008 uM. And the corneal concentration of Compound—I is about 6.0 uM — about 40 uM. KLEPTOSE® HPB or CAPTISOL® is used in the solution of Compound—I administered topically to the eye.
In some embodiments, mean Compound—I ocular tissue concentrations following twice daily topical dosing with 0.3% Compound—I lmic gel drop formulations with and without benzylalkonium chloride is highest in the cornea with between about 200 uM — about 350 uM in the , between about 2.0 uM — about 5.0 uM in the peripheral choroid, between about 0.2 uM — about 0.7 uM in the central choroid, between about 0.05 uM — about 0.5 uM in the peripheral retina, and between about 0.01 uM — about 0.05 uM in the aqueous humor.
In some embodiments, Tris—based suspension formulations of Compound—I is well tolerated, t any l ?ndings, and only with a few sporadic incidences of mild conjunctivitis. In some embodiments, mean Compound-I ocular tissue concentrations, assessed at 1 hour :: 15 minutes after the ?rst daily topical ocular dose on day 30 for the twice daily topical dosing with 0.3% Compound—I Tris—based suspensions with and without benzylalkonium chloride, are highest in the cornea, for example, between about 2.00 uM — about 4.0uM. The peripheral choroid concentration from the same dose is between about 0.7 uM — about 1.5 uM; the central choroid concentration is n about 0.3 uM — about 0.4 uM; the peripheral retina concentration is between about 0.08 uM — about 0.09 uM); l retina concentration is between about 0.04 "M — about 0.0? 11M; and s humor tration is about 0.001 uM — about 0.002 "M.
The present application provides Cyclodextrin—based solutions (e. g., solutions sing hydroxypropyl—beta—cyclodextrin (HP—B—CD, KLEPTOSE® HPB)) of Compound—I that were well tolerated when administered topically for up to 30 days, twice daily at about 0.1% Compound-I (in a solution with about 2.0% — about 2.5% HP—B—CD), twice daily at about 0.2% Compound—I (in a solution with about 4.0% — about 4.5% HP—B— CD), once or twice daily at about 0.4% Compound—I (in a solution with about 8.0% — about 8.5% HP—B—CD), and once or twice daily at about 0.6% Compound—I (in solution with up to about 14% HP-B—CD) in subjects. Moreover, in additional embodiments, cyclodextrin—based ons of about 0.4% w/v Compound—I in KLEPTOSE® HPB, KLEPTOSE® HP, or CAPTISOL® are well—tolerated when dosed twice daily for up to 24 days.
The t application provides dose—limiting corneal toxicity observed with Compound-I ophthalmic Gel Drop formulations. In some embodiments, ophthalmic Gel Drop renders about ?ve—fold to about ?fteen—fold higher corneal concentrations of Compound—I compared to cyclodextrin based solution, and about ?fty—fold to about hundred- fold higher corneal concentrations of Compound—I compared to ased suspensions.
Compound—I Tris—based suspensions and cyclodextrin—based solutions of the present ation are well tolerated with no evidence of overt ocular toxicity. In some embodiments, once or twice daily administration for at least 30 days of about 0.005% to about 5.0% w/v of a cyclodextrin-based solution or a Tris-based suspension of Compound-I is well tolerated in subjects. The present application es highest central choroid concentrations of Compound-I using Cyclodextrin-based solutions compared to lar doses of the gels and/or Tris—based formulations.
Table 10A: Average concentration of Compound—I in "M in s ocular ?uids and tissues Group Central d Central Retina Aqueous Humor Cornea m 0.769 0.124 0.00656 _———— m 0.259 0.0741 0.00313 0.212 0.0531 4 6.49 0.345 0.101 0.00403 Values Choroid LLOQ = 0.184 uM Retina LLOQ = 0.0229 uM AH LLOQ = 0.000918 uM Cornea LLOQ = 0.0918 M Table 10B: Study Design Group Total Conc.* Dose Number Total Dose Number Daily Dose* (%w{v) Volume of Doses Volume of Male m/da L/dose er Da L/da Animals Group 8. Compound-I 0.48 0.4 30/eye 2 120 2 in 8.41% SE® HPB**, 0.142% phosphate Group 9. Compound-I 0.48 0.4 30/eye 2 120 2 in 8.90% KLEPTOSE® HPB, 0.142% hos hate Group 10. Compound- 0.48 0.4 30/eye 2 120 2 I in 4.88% CAPTISOL®***, 0.142% hos hate Group 1 1. Compound- 0.48 0.4 30/eye 2 120 2 I in 4.88% CAPTISOL®, 0.122% hos hate *Total daily dose and concentration are expressed as free base equivalent of nd-I (Formula II).
** Hydroxypropyl-B-cyclodextrin (HPBCD) from Roquette.
*** CAPTISOL® is a polyanionic B-cyclodextrin derivative with a sodium sulfonate salt separated from the lipophilic cavity by a butyl ether spacer group, or sulfobutylether (SBE).
Table 11 shows the corneal and central choroidal concentrations of Compound-I formulations.
Table 11: Compound-I Formulation Dosing Frequency Cornea l Choroid W/V % Type & und 1] [Compound I] Duration M M 0.3% Ophthalmic Twice Daily 236.00 0.340 Gel Drop 29 days Tris Twice Daily Sus-ension 30 da s Ophthalmic Twice Daily Solution 24 days (CAPTISOL® Ophthalmic Twice Daily Solution 24 days (KLEPTOSE® Ophthalmic Twice Daily on 24 days (KLEPTOSE® Phase I olfor Dose-Escalation Study in Patients with NeovascalarAMD The present application provides a Phase I study involving a twelve—week, open— label, dose-escalating, center trial to evaluate the safety, tolerability, and pharmacokinetics following topical ocular administration of Compound-I in patients with neovascular age-related macular degeneration (AMD). Up to 60 patients total are d one to two times daily with topical ocular dosing of Compound-I ophthalmic on for three months, where three dose-escalating monotherapy arms and one adjunct therapy arm using a single intravitreal injection of LUCENTIS® plus the maximally-tolerated monotherapy dose are d (15 patients per treatment arm). Patients that meet pre-speci?ed vision and CNV lesion criteria confirmed by an independent reading center are allowed to simultaneously tinue topical ocular dosing and receive treatment with standard-of—care.
The present application provides 3 dosage strengths, ranging from 0.1% to 1.0% (w/v) (as Compound—I) ophthalmic solution for clinical s. The strengths are about 0.1%, about 0.3%, about 0.6%, and about 1.0% (w/v) Compound—1 HCl.
Formulation Preparation Non-limiting es of formulations of the present application are outlined in Table 12.
Table 12: Overview of product compositions tested in product screening studies Cyclodextrln Type Cyclodextrin Conc.
Buffer type and level HPBCDb 6310189"0 ~ 7 None and Tris. 1:4 10,1:12 $213ng 1.58 to 15.6% 0.1 and 0.6% None and Tris E2319?) 1.58 to 2 63% None and Tris $313513) 14 0.81 to 19.5% 0.1 and 0.6% - Phosphate 1.6 1.8 Cyclodextrin Type Cyclodextrin Cone.
Compound-I Cone. pH Buffer type and level and Ratio Rangea Range .5, 6.5 Phosphate, Tris SBECDC 0 1 0 4% HPBCD 0‘1’ 0'440 .5, 6.5 . Phosphate, Tris a Molar ratio of Compound-I : cyclodextrin. b KLEPTOSE® ° OL® EGFR ne Phosphorylation Assay in Cells to Determine EGFR Activity of Compounds ofFormula I or II An EGFR tyrosine assay in corneal epithelial cells is run to determine whether higher concentrations of EGF can overcome inhibition of EGFR kinase activity. Cells are serum starved and then are pre—treated with different concentrations of aa compound of Formula I or 11, e. g., Compound—I or a control for followed by treatment with EGF. Cells are then harvested and immunoblotted for determination of phosphorylated EGFR and total EGFR concentration which was used to determine receptor activity .
EGFR Tyrosine orylation Assay in Cells to Determine EGFR Activity ofa Combination ofCompounds ofFormala I or II, and a SecondActive Agent An EGFR tyrosine assay in corneal epithelial cells is run to determine if Vitamin K or nicotinic acid/nicotinamide can overcome inhibition of EGFR and if there is an increase in EGFR activity. Cells are serum starved and then are pre-treated with a different concentration of a compound of Formula I or 11, e.g., Compound-I and saturating concentrations of vitamin K or nicotinic acid/nicotinamide followed by treatment with EGF.
Cells are harvested cells and immunoblotted for ination of phosphorylated EGFR (tyrosine 1068 and tyrosine 1045) and total EGFR concentration which was used to determine or activity (ICso). ination ofEffects of Varying trations ofCompounds ofFormula I or II and EGF on Cell Migration/Proliferation in Cells (In Vitro Wound g) ination of cell migration/proliferation in corneal epithelial cells is run to determine cell migration/proliferation in the ce of varying concentrations of a compound of Formula I or II and EGF. Cells are plated with silicone plugs. The cells are then serum starved and pre—treated with g concentrations of a compound of Formula I or II or a control. The silicone plugs are then removed to create the acellular area the cells are treated with EGF. Cell migration is quanti?ed from micrographs.
Determination Cell Migration/Proliferation in Cells Treated with Compounds ofFormula I or 11 and/or a Second Active Agent ] Determination of cell migration/proliferation in l epithelial cells is run to determine cell migration/proliferation in the presence of varying concentrations of a compound of Formula I or II, vitamin K, or nicotinic acid/nicotinamide. Cells are plated with ne plugs. The cells are then serum starved and pre—treated with varying concentrations of a compound of Formula I or II or a control. The plugs are removed and the cells are treated with 1) EGF, 2) EGF and vitamin K or 3) EGF and nicotinic acid/nicotinamide. Cell migration is quanti?ed from micrographs.
Determination ofEffects ofCompounds ofFormula I or II on Basal andEGF-mediated Corneal Wound Healing In Vivo Determination of the s of a compound of Formula I or II on basal and ligand ated rates of corneal wound healing was determined in mice. Corneas of C57/B1 mice are wounded and then eated with a compound of Formula I or II followed by addition of EGF. Wound size is monitored by ?uorescein staining and ?uorescent photography and quanti?ed. ination ofE?ects ofCompounds ofFormula I or II and a Active Agent on Basal and EGF-mediated Corneal Wound Healing In Vivo Determination of the effects of a nd of Formula I or II and/or a second active agent on corneal wound healing was determined in mice. Corneas of C57/B1 mice are wounded and then pre—treated with a compound of Formula I or 11, followed by addition of EGF, vitamin K, or nicotinic acid/nicotinamide. Wound size and closure are monitored.
Doses of Treatment The formulation of the present application is effective in treating (i.e., lesion stabilization or regression) or preventing choroidal and retinal neovascularization (NV) in the eye of a ian subject. The Compound—I of the present application, at a speci?c dose, inhibits a receptor tyrosine kinase and the second active agent at a speci?c dose, inhibits an ErbB receptor tyrosine kinase. In some embodiments, the Compound-I formulation, at a speci?c dose, inhibits receptor tyrosine kinase including, VEGFR, FGFRs, Tie2, and .
The inhibition of several RTKs by the ation of the present application, at a speci?c dose, simultaneously has a synergistic effect, and is ive in the treatment or regression of NV in the posterior segment of the eye. In some embodiment, the second active agent modulates (e.g., activates) directly or indirectly an ErB receptor tyrosine kinase ing, EGFR, HER2, HER3 and Erb4. The activation of several ErB receptor tyrosine kinases by the formulation of the present application, at a speci?c dose, simultaneously may have a synergistic effect, and is effective in the prevention or treatment of corneal disruptions or diseases.
In one embodiment, the present application provides a method of treating (i.e., lesion stabilization or regression) or preventing choroidal and l neovascularization (NV) in the eye by administering to the subject in need thereof a therapeutically effective amount of a compound of Formula I or II, or a pharmaceutically able salt f, and a second active agent or a ceutically acceptable salt thereof, wherein a compound of Formula I or II, or a pharmaceutically acceptable salt thereof, and the second active agent, or a pharmaceutically acceptable salt thereof, are administered simultaneously. Alternatively, a compound of Formula I or II, or a pharmaceutically acceptable salt thereof, is administered prior to administration of a second active agent, or a ceutically acceptable salt thereof.
In another embodiment, a compound of Formula I or II, or a pharmaceutically able salt thereof, is administered after administration of a second active agent, or a ceutically acceptable salt thereof. In another embodiment, the present application provides a method of treating (i.e., lesion stabilization or regression) or preventing choroidal and retinal neovascularization (NV) in the eye by administering to the subject in need thereof a therapeutically effective amount of a compound of Formula I or II, or a pharmaceutically acceptable salt thereof, prior to administering a therapeutically effective dose of a formulation described herein.
In another embodiment, the present ation provides a method of treating (i.e., lesion stabilization or regression) or ting choroidal and retinal neovascularization (NV) in the eye and preventing or treating a corneal tion or disease by administering to the subject in need thereof a therapeutically effective amount of a compound of Formula I or II, or a pharmaceutically acceptable salt thereof, and a second active agent or a ceutically acceptable salt thereof, wherein a compound of Formula I or II, or a pharmaceutically acceptable salt thereof, and the second active agent, or a pharmaceutically acceptable salt thereof, are administered simultaneously. atively, a compound of Formula I or II, or a pharmaceutically acceptable salt thereof, is administered prior to stration of a second active agent, or a pharmaceutically acceptable salt thereof. In another embodiment, a compound of a I or II, or a pharmaceutically able salt thereof, is administered after administration of a second active agent, or a pharmaceutically acceptable salt thereof.
In another embodiment, the present application provides a method of treating (i.e., lesion stabilization or regression) or preventing choroidal and retinal neovascularization (NV) in the eye and ting or ng a l disruption or disease by administering to the subject in need thereof a therapeutically effective amount of a compound of Formula I or II, or a pharmaceutically acceptable salt thereof, prior to administering a therapeutically effective dose of a formulation described herein.
In a further embodiment, the formulation of the present application is effective in treating NV and treating and/or preventing corneal disruption or disease caused by systemic disease, an eye e or administration of a compound of Formula I or II when stered one, two, three, and four times daily by topical ocular delivery of about 0.005% - about 5.0% (about 0.05 — about 50 mg/mL) of Compound-I and a second active agent. The formulation of Compound—I or its free base (Formula II) and a second active agent, for the treatment or regression ofNV and the treatment and/or regression of corneal disruption or disease, is a solution comprising a second active agent, a compound of Formula I or H, and cyclodextrin or in a sion comprising Tris. The solution or suspension when delivered to a subject exposed to atmospheric oxygen to induce oxygen induced retinopathy (01R) or NV, for example, is able to effectively reduce the mean area of pre—retinal NV per retina with little or no corneal disruption or disease. The prevention or treatment ofNV by nd—l formulation and/or suspension is achieved via inhibition of l receptor tyrosine kinases (RTKs), including VEGFR-2. The prevention or treatment of corneal disruptions or disease by a second active agent is achieved via moculation (e.g., activation) of EGFR.
Any of the disclosed es or conditions described herein can be treated or prevented by achieving target tissue concentration of from about 200 nM — about 2 uM of the disclosed nds or pharmaceutically acceptable salts, formulation and/or sion thereof. One embodiment of this application relates to a method for treating ogic angiogenesis in the posterior t of the eye, achieving target tissue concentration of about of about 200 nM — about 2 uM of the disclosed compounds or pharmaceutically acceptable salts, and/or formulation thereof. Another iteration of this embodiment relates to achieving target tissue concentration of about 300 nM — about 2 [1M of one or more of the disclosed compounds or pharmaceutically acceptable salts, and/or formulation thereof.
In an embodiment of the t application about 0.2 — about 1.0% (about 2 — about 10 mg/mL) of Compound-I formulated as a solution or suspension, upon administration, may effectively inhibit VEGFR—2 kinase function and provide substantial blockade of a set of proangiogenic growth factor ors, including FGFRs 1-3, Tie-2, and EphB-4. The 2 - 10 mg/mL concentration of the Compound-l in the formulation provides effective pharmacologically effective concentrations of drug to the central choroid and retina following 1—5 days of topical ocular delivery.
In some embodiments, the exposure time of Compound—I is between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 ). In some embodiments, the dosage regimen involves several courses of topical ocular administration of a formulation sing Compound—I to a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). For example, the dosage regimen involves once daily, twice daily, three times daily or four times daily administration of the formulation for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 , 8 months, or 12 months). For example, the dosage regimen involves once, twice, three times, or four times administration of the formulation on every other day (i.e., on day 1, 3, 5, 7 etc.) for up to 90 days. For example, the dosage regimen es administering once on day 1, once or twice on day 2 — day 90. For example, the dosage regimen involves administering once, twice, three times, or four times on day 1, followed by once daily for 2— 90 days. For example, the dosage regimen involves administering once, twice, three times, four times on day 1, followed by once, twice, three times, or four times on every other day (i.e., on day 1, 3, 5, 7 etc.) for up to 90 days. For example, one dosage regimen involves once per day or twice per day for 1, 2, 3, 4, or 5 consecutive days. For twice or three daily dosage regimen, subjects receive topical ocular dose of a Compound—I formulation on days 1 and 4 approximately about 4, 6, or 8 hours apart. In another embodiment, subjects receive topical ocular doses of a Compound—I ation approximately about 4, 6, or 8 hours apart for four consecutive days. In some embodiments, subjects receive one or two doses of topical ocular dose of Compound—I formulation per day for 5 consecutive days. In yet other embodiments, subjects receive one or two doses of topical ocular dose of Compound—I formulation for 5—90 consecutive days. In some embodiments, subjects receive one or two doses of topical ocular dose of Compound—I formulation for at least 25 utive days. In one embodiment, subjects e one or two l ocular doses for at least 90 consecutive days or more.
In some ments, the present application provides a formulation of Compound-I or its free base and/or a second active agent administered lly to the anterior segment of the eye of the subject to treat AMD, pathologic CNV, and/or pathologic NV. For example, the formulation is stered to the eye of a subject 1, 2, 3, or 4 times daily. In ic embodiments, the formulation is stered to the eye of a subject 2 or 3 times daily. For example, the formulation is administered to one eye or both eyes of a subject. For example, about 1 mg/ml of a ?rst active agent comprising formulation of the t disclosure is administered twice a day (BID) to one eye or both eyes of a subject. In some embodiments, about 1 mgmL once a day (QD) or BID, about 2 mg/mL QD or BID, about 3 mg/mL QD or BID, about 4 mg/mL QD or BID, about 5 mg/mL QD or BID, about 6 mg/mL QD or BID, about 7 mg/mL QD or BID, about 8 mg/mL QD or BID, about 9 mg/mL QD or BID, or about 10 mg/mL QD or BID of is administered to one eye or both eyes of a subject.
For example, a ation comprising about 1 mg/mL BID of Compound—I and/or a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months).
In some embodiments a formulation sing about 1 mg/mL QD of nd-I and/or a second active agent is stered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 , 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 1 mg/mL TID of Compound—I and/or a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 1 mg/mL QID of Compound—I and/or a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 , 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 2 mg/mL BID of Compound—I and/or a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 ). In some embodiments a formulation comprising about 2 mg/mL QD of Compound—I and/or a second active agent is administered to one eye or both eyes of a subject for n 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 2 mg/mL TID of Compound—I and/or a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 2 mg/mL QID of Compound-I and/or a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 , 8 months, or 12 months). In some embodiments a formulation comprising about 3 mg/mL BID of Compound—I and/or a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 , 8 months, or 12 months). In some embodiments a formulation comprising about 3 mg/mL QD of Compound—I and/or a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 3 mg/mL TID of Compound—I and/or a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 3 mg/mL QID of Compound—I and/or a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 , 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 4 mg/mL BID of nd-I and/or a second active agent is administered to one eye or both eyes of a subject for n 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation sing about 4 mg/mL QD of Compound—I and/or a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 , or 12 months). In some ments a formulation comprising about 4 mg/mL TID of Compound—I and/or a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some ments a formulation comprising about 4 mg/mL QID of Compound—I and/or a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 , or 12 months). In some embodiments a formulation comprising about 5 mg/mL BID of Compound—I and/or a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 , 6 , 8 months, or 12 months). In some embodiments a formulation comprising about 5 mg/mL QD of Compound—I and/or a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation sing about 5 mg/mL TID of Compound—I and/or a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 5 mg/mL QID of Compound-I and/or a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 6 mg/mL BID of Compound—I and/or a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 6 mg/mL QD of Compound—I and/or a second active agent is stered to one eye or both eyes of a subject for n 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a ation comprising about 6 mg/mL TID of Compound—I and/or a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 6 mg/mL QID of Compound-I and/or a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 7 mg/mL BID of a ?rst active agent (e.g., Compound—I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 7 mg/mL QD of a ?rst active agent (e.g., Compound—I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e. g., 4 months, 6 months, 8 months, or 12 months). In some ments a formulation comprising about 7 mg/mL TID of a ?rst active agent (e.g., Compound—I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e. g., 4 months, 6 months, 8 , or 12 months). In some embodiments a formulation comprising about 7 mg/mL QID of a ?rst active agent (e.g., Compound-I) and a second active agent is administered to one eye or both eyes of a subject for n 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation sing about 8 mg/mL BID of a ?rst active agent (e.g., Compound—I) and a second active agent is administered to one eye or both eyes of a subject for n 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 8 mg/mL QD of a ?rst active agent (e. g., Compound-I) and a second active agent is administered to one eye or both eyes of a subject for n 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 8 mg/mL TID of a ?rst active agent (e.g., Compound—I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e. g., 4 months, 6 , 8 months, or 12 ). In some embodiments a formulation comprising about 8 mg/mL QID of a ?rst active agent (e.g., Compound-I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 9 mg/mL BID of a ?rst active agent (e.g., Compound—I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 ). In some embodiments a formulation sing about 9 mg/mL QD of a ?rst active agent (e.g., Compound-I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 ). In some embodiments a formulation comprising about 9 mg/mL TID of a first active agent (e.g, Compound—I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 9 mg/mL QID of a ?rst active agent (e.g., nd-I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments a formulation comprising about 10 mg/mL QD of a first active agent (e.g., Compound—I) and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). The dosage n for between 1 and 90 days or for longer than 90 days (e. g., 4 months, 6 months, 8 months, or 12 months) may be any of the regimens involving consecutive or alternate days described in the paragraph above. In some ments, the formulation of the present application is administered QD, BID, TID, or QID when administered at low doses (e.g., 1 mg/mL, 2 mg/mL, 3 mg/mL, 4 mg/mL, or 5 mg/mL), and QD or BID at high doses (e.g., 6 mg/mL, 7 mg/mL, 8 mg/mL, 9 mg/mL, or 10 .
In other embodiment, a 1 mg/mL BID of Compound—I formulation and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments, a 1 mg/mL QD of Compound-I formulation and a second active agent is administered to one eye or both eyes of a t for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments, a 2 mg/mL BID of Compound—I formulation and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 ). In some embodiments, a 2 mg/mL QD of Compound-I ation and a second active agent is administered to one eye or both eyes of a subject for n 1 and 90 days or for longer than 90 days (e.g., 4 , 6 months, 8 months, or 12 months). In some embodiments, a 3 mg/mL BID of Compound-I formulation and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments, a 3 mg/mL QD of Compound—I ation and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some ments a 4 mg/mL BID of Compound-I formulation and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments, a 4 mg/mL QD of Compound—I formulation and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 ). In some ments, a 5 mg/mL BID of Compound-I formulation and a second active agent is administered to one eye or both eyes of a t for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments, a 5 mg/mL QD of Compound—I formulation and a second active agent is administered to one eye or both eyes of a t for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments, a 6 mg/mL BID of Compound—I formulation and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 , or 12 months). In some embodiments, a 6 mg/mL QD of nd—I formulation and a second active agent is administered to one eye or both eyes of a subject for n 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments, a 7 mg/mL BID of Compound-I formulation and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments, a 7 mg/mL QD of Compound—I formulation and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments, a 8 mg/mL BID of Compound-I formulation and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments, a 8 mg/mL QD of Compound—I formulation and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 , 6 months, 8 months, or 12 months). In some ments, a 9 mg/mL BID of Compound-I formulation and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments, a 9 mg/mL QD of Compound—I formulation and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months). In some embodiments, a 10 mg/mL BID of Compound-I formulation and a second active agent is administered to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e.g., 4 , 6 months, 8 months, or 12 ). In some embodiments, a 10 mg/mL QD of Compound-I formulation and a second active agent is administered to one eye or both eyes of a subject for n 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 , 8 , or 12 months). The dosage regimen for between 1 and 90 days or for longer than 90 days (e. g., 4 months, 6 months, 8 months, or 12 months) may be any of the ns involving consecutive or alternate days described in the paragraph above.
The present application provides formulations as shown in Table 13 for administering to one eye or both eyes of a subject.
Table 13 Dose/ Formula nd-I Formula II / KLEPTOSE 10 mM Sodium pH Day II (%) (%) Compound-I ® HPB (%) Phosphate Chloride : CD % % * 0.40 0.427 128 8.411 0.142 QS to about 6 285 mOsm 0.641 1:8 12.626 0.142 QS to about 6 285 mOsm BID 0.10 : 2.103 0.142 QS to about 6 285 mOsm BID 0.20 : 4.205 0.142 QS to about 6 285 mOsm BID 0.30 0.321 128 6.308 0.142 QS to about 6 285 mOsm BID 0.40 0.427 128 8.411 0.142 QS to about 6 285 mOsm *QS = quantity suf?cient for achieving the osmolality In some embodiments, the formulation of Formula II or Compound-I is administered to one eye or both eyes of a subject. For example, about 0.2 % - about 1.0% (w/v) of the compound of Formula II or about 0.1% — 1.2 % (w/v) of Compound-I comprising formulation of the current disclosure is administered once a day (QD) or twice a day (BID) to one eye or both eyes of a subject for between 1 and 90 days or for longer than 90 days (e. g., 4 months, 6 months, 8 , or 12 months). In some embodiments, Formula II compound or Compound—I is complexed with a complexing agent, e.g., cyclodextrin (e.g., KLEPTOSE® HPB (%)) in ratio of about 1:8, in which about 2% — 13 % (w/v) cyclodextrin (e.g., KLEPTOSE® HPB (%)) is added to the formulation. The ation further comprises about 0.1% — about 0.2% buffer, e.g., 10 mM phosphate buffer. The desired osmolality of the formulation is about 200 — about 300 mOsm, ed by adding quantity suf?cient to achieve the osmolality with a salt, e.g., sodium chloride. The pH of the formulation is about 6.0 at or under about 40 OC. The dosage regimen for between 1 and 90 days or for longer than 90 days (e.g., 4 months, 6 months, 8 months, or 12 months) may be any of the regimens ing consecutive or alternate days described in the paragraph above.
In some embodiments, the Formula II nd or Compound-I ation further comprises a second active agent. In some embodiment, Formula II compound or Compound— I formulation is administered with a second active agent.
The second active agent, e.g, nicotinic acid, nicotinamide, or vitamin K or a combination f, at a speci?c dose modulates (e.g., activates) directly or indirectly, ErbB receptor tyrosine kinase (RTK) including, EGFR, HER2, HER3, and HER4. The modulation (e.g., activation) of one or more ErbB RTKs by the formulation of the present application, at a speci?c dose, simultaneous and has a synergistic effect, and is effective in the prevention or treatment of corneal disruptions or diseases, (e.g., l ulcers, corneal epithelial s, keratitis, etc.) in the anterior segment of the eye.
The methods of the present application are combined with the rd of care, including but not limited to laser ent and treatment with inj ectable anti-neovascular agents.
Particle Compositions and Formulations Comprising the Particle Compositions The present application relates to a pharmaceutical composition sing les of an active agent of the present application (e.g., a ?rst active agent (e.g., Formula II or Compound—I) and/or a second active agent), or a pharmaceutically acceptable salt thereof, wherein the particles have a mean diameter of between 100 nm and 100 pm. In some embodiments, the particles have a mean er of n 20 um and 90 um. In some embodiments, the particles have a mean diameter of between 20 um and 80 um. In some embodiments, the particles have a mean diameter of between 20 um and 70 um. In some embodiments, the particles have a mean diameter of between 30 um and 70 pm. In some embodiments, the particles have a mean diameter of between 30 um and 60 pm. In some embodiments, the particles have a mean diameter of n 30 um and 50 um. In some embodiments, the particles have a mean diameter of between 30 um and 40 um. In some embodiments, the particles have a mean diameter of between 50 um and 70 um. In some embodiments, the particles have a mean diameter of between 50 um and 60 um. In some embodiments, the particles have a mean diameter of at least 30 um. In some embodiments, the particles have a mean diameter of about 30 um, about 35 um, about 40 um, about 45 um, about 50 um, about 55 um, about 60 um, about 65 pm, or about 70 um. In some embodiments, the particles have a mean diameter of about 30 um, about 35 um, about 50 um, or about 60 um. In some embodiments, the particles have a mean diameter of between 100 nm and 8 um. In some embodiments, the particles have a mean diameter of between 100 nm and 200 nm. In some ments, the particles have a mean diameter of at most 150 nm.
In some embodiments, the particles have a mean diameter of about 150 nm, about 140 nm, about 130 nm, about 120 nm, about 110 nm, or about 100 nm. In other embodiments, the particles have a mean er of between 1 pm and 5 pm. In some embodiments, the particles have a mean diameter of between 2 pm and 4 pm. In some embodiments, the particles have a mean diameter of about 1 pm, about 2 pm, about 3 pm, about 4 um, or about um. In some embodiments, the particles have a mean diameter of about 3 um.
In some embodiments, at least 90% of the particles have a diameter of 70 um or less. In some embodiments, at least 90% of the particles have a diameter of 60 um or less. In some embodiments, at least 90% of the particles have a diameter of 10 um or less. In some embodiments, at least 90% of the les have a diameter of 9 um or less. In some embodiments, at least 90% of the particles have a er of 8 um or less. In some embodiments, at least 90% of the particles have a diameter of 7 um or less. In some embodiments, at least 90% of the particles have a diameter of 6 um or less. In some embodiments, at least 90% of the particles have a diameter of 5 um or less. In some embodiments, at least 90% of the particles have a diameter of 4 pm or less. In some embodiments, at least 90% of the les have a diameter of 300 nm or less. In some embodiments, at least 90% of the particles have a diameter of 200 nm or less.
In some embodiments, the pharmaceutical composition comprises particles of a ?rst active agent (e.g., a first active agent (e.g., Formula II or nd-1)) and particles of a second active agent (e. g., nicotinic acid, nicotinamide, n K, or a combination thereof).
In some embodiments, the pharmaceutical composition comprises les of a ?rst active agent (e.g., a ?rst active agent (e.g, Formula II or Compound-1)) and particles of vitamin K (e. g., menadione).
In some embodiments, a ?rst active agent (e.g, Formula II or Compound-I) and a second active agent (e. g., nicotinic acid, namide, n K, or a combination thereof) are roller milled together to form particles comprising the ?rst active agent and the second active agent. In some embodiments, the ?rst active agent and the second active agent are roller milled separately, and the particles comprising the ?rst active agent and the particles comprising the second active agent are then mixed or roller milled together. In some embodiments, the ?rst active agent or the second active agent is roller milled ?rst, and the particles are then added to the other active agent for further roller milling.
] In some embodiments, the ceutical composition comprising particles of an active agent of the present application (e.g., a ?rst active agent or a second active agent) r comprises one or more excipients. The ent can be selected from any suitable ent known in the art, for e, for preparing an ophthalmic formulation. In some ments, the excipient is selected from Polysorbate (Tween) 80, Poloxamer nic) F-127, Hypromellose (Hydroxypropyl Methylcellulose or HPMC), Povidone (PVP K—29/32 or K-30), and Tyloxapol, and a combination thereof. In some embodiments, the excipient is ed from HPMC, Tween 80, Pluronic F—127, and Tyloxapol, and a combination thereof.
In some embodiments, the particles comprising a ?rst active agent (e.g., Formula II or Compound—I) and the particles comprising a second active agent (e.g., nicotinic acid, nicotinamide, n K, or a combination thereof) comprise the same excipient(s).
In some embodiments, the pharmaceutical composition comprising particles of an active agent of the present application (e.g., a ?rst active agent or a second active agent) further comprises a surfactant, such as benzalkonium chloride (BAC).
In some embodiments, the pharmaceutical composition comprising particles of an active agent of the t application (e.g., a ?rst active agent or a second active agent) further comprises an excipient (e.g., for enhancing bioavailability of the active agent). In some embodiments, the excipient is hydroxyethyl cellulose (HEC). In some embodiments, the HEC is present in an amount of about 0.1% — about 1%, about 0.1% — about 0.9%, about 0.1% — about 0.8%, about 0.1% — about 0.7%, about 0.1% — about 0.6%, about 0.1% — about 0.5%, about 0.1% — about 0.4%, or about 0.1% — about 0.3%. In some embodiments, the HEC is present in an amount of about 0.2% or about 0.3%.
In some embodiments, the particles of the present application comprise Formula II (i.e., free base of nd-I). In other embodiments, the particles of the present application comprises Compound—I.
In some embodiments, the particles of the present application are prepared by roller milling. Factors that may affect the size of the particles include, but are not limited to, the use of the free base or a salt of the active agent (e.g., Compound—I vs. Formula II), the addition of an excipient, the speed of the roller during milling, the size of the milling media, and duration of the milling.
In some embodiments, the particles of the present application are prepared without roller milling.
In some embodiments, the particles of an active agent of the present application (e.g., a ?rst active agent (e.g., Formula II or Compound—I) and/or a second active agent) are sterilized. In some embodiments, the sterilization is conducted with gamma irradiation. In some embodiments, the particles of an active agent of the present application (e. g., a ?rst active agent (e. g., a II or nd-I) and/or a second active agent) are stable after the sterilization (e. g., remaining substantially pure of any impurities or degradation products as a result of the sterilization).
The present application s to a suspension formulation comprising a pharmaceutical composition, wherein the pharmaceutical composition comprises particles of an active agent of the present application (e.g., a ?rst active agent (e.g., Formula II or Compound—I) and/or a second active agent), or a pharmaceutically acceptable salt f, as described herein.
In some embodiments, the suspension formulation comprises les of a ?rst active agent (e. g., Formula II or Compound—I), n the ?rst active agent is at a tration of about 0.1 mg/mL — about 10 mg/mL. In some embodiments, the ?rst active agent is at a concentration of about 0.2 mg/mL — about 10 mg/mL, about 0.5 mg/mL — about mg/mL, about 1 mg/mL — about 10 mg/mL, about 2 mg/mL — about 9 mg/mL, about 2 mg/mL — about 8 mg/mL, about 3 mg/mL — about 8 mg/mL, about 3 mg/mL — about 7 mg/mL, about 3 mg/mL — about 6 mg/mL, about 4 mg/mL — about 6 mg/mL, or about 4 mg/mL — about 5 mg/mL of a ?rst active agent (e.g., Formula II or Compound—I). In some embodiments, the ?rst active agent is at a concentration of about 0.1 mg/mL, 0.3 mg/mL, 0.5 mg/mL, about 1 mg/mL, about 2 mg/mL, about 3 mg/mL, about 4 mg/mL, about 6 mg/mL, or about 10 mg/mL. In some embodiments, the ?rst active agent is at a concentration of about 1 mg/mL — about 4 mg/mL or about 2 mg/mL — about 4 mg/mL. In some embodiments, the ?rst active agent is at a concentration of about 2 mg/mL or about 4 mg/mL.
In some embodiments, the suspension formulation comprising les of a ?rst active agent (e. g., Formula II or Compound—I) further ses a second active agent (e. g., nic acid, nicotinamide, vitamin K, or a combination thereof). In some embodiments, the second active agent is vitamin K (e.g., menadione). In some embodiments, the second active agent is present in an amount of less than 10 "M. In some embodiments, the second active agent is present in an amount of about 0.5 uM, about 0.6 uM, about 0.7 uM, about 0.8 uM, about 0.9 uM, about 1 uM, about 2 uM, about 3 uM, about 4 uM, about 5 uM, about 6 uM, about 7 uM, about 8 uM, or about 9 uM. In some embodiments, the second active agent is present in an amount of about 1 uM.
In some embodiments, the suspension formulation comprising les of a ?rst active agent (e.g., Formula II or Compound—I) further comprises an excipient selected from Polysorbate (Tween) 80, Poloxamer (Pluronic) F—127, Hypromellose (Hydroxypropyl Methylcellulose or HPMC), Povidone (PVP K-29/32 or K-30), and Tyloxapol, and a combination thereof. In some embodiments, the excipient is Pluronic F-127, Tween 80, HPMC, or Tyloxapol, or a ation thereof. In some embodiments, the excipient is present in a concentration of about 0.01% — about 0.2%, about 0.01% - about 0.15%, about 0.01% — about 0.12%, about 0.01% — about 0.1%, about 0.01% - about 0.09%, about 0.02% — about 0.09%, about 0.03% — about 0.09%, about 0.04% — about 0.09%, or about 0.04% — about 0.08%. In some embodiments, the suspension formulation comprises about 0.08% or about 0.04% ic F—127. In some embodiments, the suspension formulation comprises about 0.08% HPMC. In other embodiments, the suspension formulation comprises about 0.04% Tyloxapol.
] In some ments, the suspension formulation comprising particles of a first active agent (e. g., Formula II or Compound—I) further comprises a buffering agent. In some embodiments, the buffering agent is ed from phosphate buffers, borate buffers, citrate buffers, tartrate buffers, acetate s, amino acids, sodium acetate, sodium citrate, Tris buffers, and the like. In some ments, the buffering agent is Tris. In some embodiments, the buffering agent is present in a concentration of about 0.1% - about 2%, about 0.2% — about 1.8%, about 0.3% — about 1.6%, about 0.4% — about 1.4%, about 0.4% — about 1.2%, about 0.4% — about 1%, about 0.4% — about 0.8%, about 0.4% — about 0.7%, or about 0.5% — about 0.7%. In some ments, the suspension ation comprises about 0.6% Tris.
In some embodiments, the suspension formulation comprising particles of a ?rst active agent (e. g., Formula II or Compound-I) further comprises an osmolality adjusting reagent. In some embodiments, the osmolality adjusting reagent is glycerol. In some embodiments, the osmolality adjusting reagent is present in an amount of about 1% - about %, about 2% — about 10%, about 3% — about 10%, about 4% - about 10%, about 5% — about 10%, about 2% — about 9%, about 2% — about 8%, about 2% — about 7%, about 2% — about 6%, about 2% — about 5%, about 2% — about 4%, or about 2% — about 3%. In some embodiments, the suspension formulation comprises about 2% or about 2.5% glycerol.
] In some embodiments, the suspension formulation comprising particles of a ?rst active agent (e.g., Formula II or Compound—I) has a pH of less than 7.5. In some embodiments the pH is between about 6.0 — about 7.0. In some embodiments the pH is about 6.0 or about 7.0.
In some embodiments, the suspension formulation comprising particles of a ?rst active agent (e. g., Formula II or Compound-I) further comprises hydroxyethyl cellulose (HEC). In some ments, the HEC is present in an amount of about 0.1% — about 1%, about 0.1% — about 0.9%, about 0.1% — about 0.8%, about 0.1% — about 0.7%, about 0.1% — about 0.6%, about 0.1% — about 0.5%, about 0.1% — about 0.4%, or about 0.1% — about 0.3%. In some embodiments, the HEC is present in an amount of about 0.2% or about 0.3%.
One of a ?rst active agent, a second active agent, and one or more ents described in the present application can be present at any concentration or level described herein in combination with the der of the ?rst active agent, the second active agent, and one or more excipients described in the present application at any concentration or level described herein.
In some embodiments, the formulation of the present application comprises a first active agent at about 0.1% to about 1.2% (or any range in between as described herein), Tris at about 0.4% — about 0.8% (or any range in between as described herein), glycerol at about 2% - about 4% (or any range in n as described herein), HEC at about 0.1% — about 0.3% (or any range in between as described herein), and HPMC at about 0.04% — about 0.09% (or any range in n as described ). In some embodiments, the ?rst active agent is Formula II or Compound-I. In some embodiments, the formulation of the t application comprises Formula II or Compound—I at about 0.4%, Tris at about 0.6%, ol at about 2%, HEC at about 0.2%, and HPMC at about 0.08%. In some embodiments, the formulation comprises particles of the ?rst active agent wherein the particles have a mean diameter of between 30 um and 60 mm. In some embodiments, the formulation comprises particles of the ?rst active agent wherein the particles have a mean diameter of between 50 um and 60 um. In some embodiments, the formulation comprises particles of the ?rst active agent wherein the particles have a mean diameter of about 50 pm or 60 pm. In some ments, the formulation has a pH of less than 7.0. In some embodiments, the formulation has a pH of about 6.
In some embodiments, the formulation of the present application comprises a ?rst active agent at about 0.1% to about 1.2% (or any range in between as described herein), a second active agent at about 0.00001% — about 0.0001% (or any range in n as described ), Tris at about 0.4% — about 0.8% (or any range in between as described herein), glycerol at about 2% - about 4% (or any range in between as described herein), HEC at about 0.1% — about 0.3% (or any range in n as described herein), and HPMC at about 0.04% - about 0.09% (or any range in between as described herein). In some embodiments, the first active agent is Formula II or Compound-I. In some embodiments, the second active agent is vitamin K3 (e.g., menadione). In some ments, the ation of the present application comprises Formula II or nd-I at about 0.4%, ne K3 at about 0.000086%, Tris at about 0.6%, glycerol at about 2%, HEC at about 0.2%, and HPMC at about 0.08%. In some embodiments, the formulation comprises particles of the ?rst active agent wherein the particles have a mean diameter of between 30 um and 60 um. In some embodiments, the formulation ses les of the ?rst active agent n the particles have a mean diameter of between 50 um and 60 pm. In some embodiments, the formulation comprises particles of the ?rst active agent wherein the particles have a mean diameter of about 50 um or 60 um. In some embodiments, the particle also comprises the second active agent. In some embodiments, the formulation has a pH of less than 7.0. In some embodiments, the formulation has a pH of about 6.
Indications and Methods of ent Disclosed are methods for the treatment of diseases or conditions of the eye. The disclosed methods relate to treating, preventing, or lling ocular neovascularization (NV), or treating a disease or ion that is related to the onset ofNV by administering to a subject one or more of the disclosed compounds (e.g., a ?rst active agent (e. g., a compound of Formula I or II), and optionally a second active agent), and formulations thereof.
One aspect of the disclosed method relates to treating or preventing NV by administering to a subject an effective amount of one or more of the disclosed compounds of Formula I or II, or pharmaceutically acceptable salts, and optionally a second active agent (e.g., vitamin K, nicotinic acid, or nicotinamide, or a combination thereof), and/or formulations thereof. One embodiment of this aspect relates to a method for treating NV by administering to a subject a composition of: a) an effective amount of one or more of the sed compounds of Formula I or II, or pharmaceutically acceptable salts, and optionally a second active agent (e. g., vitamin K, nicotinic acid, or nicotinamide, or a combination thereof), and/or formulations thereof, and optionally b) one or more carriers or compatible excipients.
The disclosed methods relate to preventing or controlling pathologic ocular neovascularization (NV), or treating a disease or condition that is related to the onset ofNV by administering to a subject one or more of the sed compounds of Formula I or II, and ally a second active agent (e.g., vitamin K, nicotinic acid, or nicotinamide, or a ation thereof), and formulations f.
The current embodiments provide use of a formulation of Compound-I or its free base (Formula II), and ally a second active agent (e.g., n K, nic acid, or nicotinamide, or a combination thereof), for the manufacture of a medicament for treating a subject with a ior segment disease vasculopathic or in?ammatory e of the eye.
These include for example, diabetic retinopathy (including background diabetic retinopathy, proliferative diabetic retinopathy and diabetic macular edema); lated macular degeneration (AMD) (including neovascular (wet/exudative) AMD, dry AMD, and Geographic Atrophy); pathologic choroidal cularization (CNV) from any mechanism (1'. e. high myopia, trauma, sickle cell disease; ocular histoplasmosis, angioid streaks, traumatic choroidal rupture, drusen of the optic nerve, and some retinal dystrophies); pathologic retinal neovascularization from any mechanism (i.e., sickle cell retinopathy, Eales disease, ocular ischemic syndrome, carotid ous fistula, familial exudative vitreoretinopathy, hyperviscosity syndrome, idiopathic occlusive arteriolitis; birdshot retinochoroidopathy, retinal vasculitis, sarcoidosis, or toxoplasmosis); uveitis; retinal vein occlusion (central or branch); ocular trauma; surgery induced edema; y induced neovascularization; cystoid macular edema; ocular ischemia; retinopathy of prematurity; Coat's disease; sickle cell retinopathy and/or neovascular glaucoma. In one embodiment, the disease of the eye is AMD. In one embodiment, the diseases of the eye arise from or are exacerbated by ocular angiogenesis and/or neovascularization.
In one aspect of the present application the formulation is used in the treatment of lated macular degeneration (AMD) (including neovascular (wet/exudative) AMD, dry AMD, and Geographic Atrophy). The solutions or suspensions are used in the treatment of neovascular tive or wet) AMD. In another embodiment, the solutions or suspensions are used to treat dry AMD. In yet another embodiment, the solutions or sions are used to treat Geographic Atrophy.
The formulation of the present application prevents, delays, or treats the onset of pathologic choroidal cularization (CNV) from any mechanism (1'. e. high , trauma, sickle cell disease; ocular histoplasmosis, angioid streaks, traumatic choroidal rupture, drusen of the optic nerve, and some retinal dystrophies) in subjects.
The formulation of the present application delays onset, ts progression, or treats formation of pathological choroidal neovascularization (CNV) below the neurosensory retina. The formulation of the present application is effective in treating CNV.
One aspect of this method relates to treating or preventing ocular neovascularization by administering to a subject an effective amount of one or more of the disclosed compounds of Formula I or II, or ceutically acceptable salts thereof, and ally a second active agent (e.g, vitamin K, nicotinic acid, or nicotinamide, or a combination thereof). One embodiment of this aspect relates to a method for treating ocular edema and neovascularization by administering to a subject a composition of: a) an effective amount of one or more of the disclosed compounds of Formula I or II, or pharmaceutically acceptable salts, and optionally a second active agent (e.g., vitamin K, nicotinic acid, or nicotinamide, or a combination thereof), and/or formulations thereof, and optionally b) one or more carriers or compatible excipients.
The disclosed methods also relate to ting or controlling ocular edema or treating a disease or condition that is related to the onset of ocular edema by administering to a subject one or more or the disclosed compounds of Formula I or II and optionally a second active agent (e. g., vitamin K, nic acid, or nicotinamide, or a combination f).
One aspect of this method relates to treating or preventing ocular edema by administering to a subject an effective amount of one or more of the disclosed compounds of Formula I or II, or pharmaceutically acceptable salts f, and ally a second active agent (e.g., vitamin K, nicotinic acid, or namide, or a combination thereof). One ment of this aspect relates to a method for treating ocular edema by administering to a subject a composition of: a) an effective amount of one or more of the disclosed nds of FormulaI or II, or pharmaceutically acceptable salts, and optionally a second active agent (e.g., vitamin K, nicotinic acid, or nicotinamide, or a combination thereof), and/or formulations f, and optionally b) one or more carriers or compatible excipients.
] Another disclosed method relates to preventing or controlling retinal edema or l neovascularization or treating a disease or condition that is d to the onset of retinal edema or retinal neovascularization by administering to a subject one or more or the disclosed compounds of Formula I or II and optionally a second active agent (e.g., vitamin K, nicotinic acid, or nicotinamide, or a combination thereof). One aspect of this method relates to treating or preventing retinal edema or retinal cularization by administering to a subject an effective amount of one or more of the disclosed nds of Formula I or II, or pharmaceutically acceptable salts thereof, and ally a second active agent (e. g., vitamin K, nicotinic acid, or nicotinamide, or a combination thereof). One embodiment of this aspect relates to a method for treating retinal edema or retinal cularization by administering to a subject a composition of: a) an ive amount of one or more of the disclosed compounds of Formula I or II, or pharmaceutically acceptable salts, and optionally a second active agent (e. g., Vitamin K, nic acid, or nicotinamide, or a combination thereof), and/or formulations thereof, and optionally b) one or more rs or compatible excipients.
Another embodiment of this aspect relates to a method for delaying or preventing progression of non-proliferative retinopathy to proliferative retinopathy by administering to a subject a composition of: a) an effective amount of one or more of the disclosed compounds of Formula I or II, or pharmaceutically acceptable salts, and optionally a second active agent (e. g., n K, nicotinic acid, or nicotinamide, or a combination thereof), and/or formulations thereof, and optionally b) one or more carriers or compatible excipients.
One aspect of the disclosed methods relates to diseases that are a direct or indirect result of diabetes, inter alia, diabetic macular edema and diabetic retinopathy. The ocular ature of the diabetic becomes unstable over time leading to conditions such as non— proliferative retinopathy, macular edema, and proliferative retinopathy. As ?uid leaks into the center of the macula, the part of the eye where sharp, straight-ahead Vision occurs, the buildup of ?uid and the associated protein begin to deposit on or under the macula. This s in swelling that causes the subject's central Vision to lly become distorted. This condition is referred to as "macular edema." Another condition that may occur is non- proliferative retinopathy in which vascular changes, such as microaneurysms, outside the macular region of the eye may be observed. During proliferative DR, pathologic new blood s grow in and up from the retina into to the us body, where these abnormal vessels may alter retinal morphology in the macula, and/or hemorrhage into the Vitreous and obscure the visual axis.
] A further disclosed method relates to treating, preventing or controlling diabetic retinopathy or treating a disease or ion that is related to the onset of ic retinopathy by administering to a t one or more or the disclosed compounds of Formula I or II, and optionally a second active agent (e.g., vitamin K, nicotinic acid, or nicotinamide, or a combination thereof).
One aspect of the disclosed method relates to treating or preventing diabetic retinopathy by administering to a t an effective amount of one or more of the disclosed compounds of Formula I or II, or pharmaceutically acceptable salts thereof, and ally a second active agent (e. g., vitamin K, nicotinic acid, or nicotinamide, or a combination thereof). One embodiment of this aspect relates to a method for treating diabetic pathy by administering to a subject a composition of: a) an effective amount of one or more of the disclosed compounds of Formula I or II, or pharmaceutically acceptable salts, and optionally a second active agent (e. g., vitamin K, nic acid, or nicotinamide, or a combination thereof), and/or formulations thereof, and ally b) one or more carriers or compatible excipients.
Diabetic proliferative retinopathy is characterized by neovascularization. The new blood vessels are fragile and are susceptible to bleeding. The result is scarring of the retina, as well as occlusion or total blockage of the light pathway through the eye due to the abnormal formation of new blood vessels. Typically subjects having diabetic macular edema are suffering from the non-proliferative stage of diabetic pathy; however, it is not uncommon for subjects to only begin manifesting macular edema at the onset of the proliferative stage.
Yet a further disclosed method relates to preventing or controlling diabetic macular edema or treating a e or condition that is related to the onset of diabetic macular edema by administering to a subject one or more or the disclosed compounds of Formula I or II and optionally a second active agent (e.g., vitamin K, nicotinic acid, or nicotinamide, or a combination thereof).
] One aspect of this method relates to treating or preventing diabetic macular edema by administering to a subject an effective amount of one or more of the disclosed compounds of Formula I or II, or ceutically acceptable salts, and optionally a second active agent (e.g., vitamin K, nic acid, or nicotinamide, or a combination thereof), or ations thereof. One embodiment of this aspect s to a method for treating diabetic macular edema by administering to a subject a composition of: a) an effective amount of one or more of the disclosed compounds of Formula I or II, or pharmaceutically acceptable salts, and optionally a second active agent (e.g., vitamin K, nicotinic acid, or nicotinamide, or a ation thereof), and/or formulations f, and b) one or more carriers or compatible excipients.
Another aspect of the sed method relates to treating or preventing NV and treating and/or preventing l disruptions or diseases by administering to a subject an effective amount of one or more of the sed compounds of Formula I or II, or ceutically acceptable salts, a second active agent, and/or formulations thereof. One embodiment of this aspect relates to a method for ng NV and treating and/or preventing corneal disruptions or diseases by administering to a subject a composition of: a) an effective amount of one or more of the disclosed nds of Formula I or II, or pharmaceutically acceptable salts, and/or formulations thereof, b) a second active agent, and optionally c) one or more carriers or compatible excipients. In one embodiment, the l disruption or disease is caused by a compound of Formula I or II.
Also disclosed are kits of the sed compounds and compositions for drug delivery into a human, mammal, or cell. The kits can comprise one or more packaged unit doses of a composition sing one or more compounds of Formula I or II and one or more packaged unit doses of a second active agent to be delivered into a human, mammal, or cell. The unit dosage ampoules or multi—dose containers, in which the compounds of Formula I or II or the second active agent to be delivered are packaged prior to use, can comprise a hermetically sealed container enclosing an amount of the active agent or pharmaceutically acceptable salt, or formulation thereof, suitable for a pharmaceutically effective dose thereof, or multiples of an ive dose. The compounds can be packaged as a sterile formulation, and the hermetically sealed container is designed to preserve sterility of the formulation until use.
The kit of the present application has a single—use eye drop dispenser bottle for delivery of ophthalmic formulation. In an alternative ment, the kit of the present application has a multi—use eye—drop dispenser bottle. The multi—dose dispenser bottle has appropriate amount of anti-infective and/or preservative agent, for example without being limited to, 0.005% BAK. The ophthalmic dispenser of the present application has a top and a cap. The ner of the present application has a semi—transparent LDPE ophthalmic ser bottle with a LDPE dropper tip and HDPE cap. The container may be of other type and form as needed and/or as used in the art.
The following examples are rative, but not limiting, of the methods and compositions of the t application. Other suitable modi?cations and adaptations of the variety of conditions and parameters normally encountered in therapy and that are obvious to those skilled in the art are within the spirit and scope of the embodiments.
General Methodology Roller Mill The horizontal roller mill consists of multiple motor driven rollers contained within a metal housing. Individual containers placed between the rollers will rotate at an rpm determined by the speed of the rollers and the er of the container. Drug slurry consisting of API, and optionally stabilizers, water, and/or milling media are added to the container before being placed between the rollers. The media used are beads of s sizes, e.g., from 800 microns to 3000 microns, and can be made with Yttria Zirconium. After milling, the dispersion is separated from the media by transferring the contents to a fuge tube insert ?tted with a screen mesh. After centrifugation, e.g., at approximately 300 x G for approximately 5 minutes, the dispersion is collected below the mesh (which retained the media).
Optical Microscopy (01%) The morphology and size distribution of the particle compositions can be assessed by l microscopy, and photomicrographs of particles can be taken, for example, using an Olympus BX51 system ed with an oil immersion 100x objective (1000x magni?cation). A calibration bar (from lum to 100 um) can be set as a comparator on each photomicrograph.
Particle Size Distribution (PSD) Particle size distribution can be analyzed using laser diffraction light scattering, e. g., with a Horiba LA—950 V2. Generic assumptions are made in setting conditions and the tive index value. The distributions are volume based. Sample y is adjusted to a generic range of percent transmission on the blue LED light source. A small sample cell (?lled with water) can be used rather than the ?ow through cell, to minimize sample quantity.
Calculation The following equation was used to determine the volume of diluent (50:50 methanol: water) required in order to e choroid, retina, and cornea samples at a speci?ed tissue concentration. uent = (MassTissue/Concmssue) - Volmssue Equation 1 Where: Conanssue = Desired tissue concentration (mg/mL) MassTissue = Mass of tissue (mg) V01.Di1uent = Volume of diluent (50:50 ol: water) (mL) sue = Volume of tissue (mL), assuming density of 1.0 g/mL To calculate the concentration in ng/g (ng of drug / g of tissue), the following equation was used: Conc.ng/g = Conc.ng/mL x (Volmml / Massnssue) Equation 2 Where: Conc.ng/g = Calculated tration (ng of drug / g of tissue) Conc.ng/mL = Calculated concentration (ng of drug / mL of nate) Volmal = Total volume of tissue homogenate (mL) MassTissue = Mass of tissue (g) To then calculate concentration of drug in tissue* in [1M (umol of drug / volume of tissue), the following equation was used, assuming a tissue density of 1 g/mL: Conc.HM = Concmg;g / MW Equation 3 Where: ConanM = Calculated concentration (pmol of drug / volume of tissue) Concng/g = Calculated concentration (ng of drug / g of tissue) MW = Molecular weight e) *Tissue = choroid, retina, or cornea To calculate concentration of drug in ?uid** in HM (umol of drug / volume of ?uid), the following on was used: ConcmM = Conc.ng/mL I MW Equation 4 Where: ConcuM = Calculated concentration (umol of drug / volume of ?uid) Conc.ng/mL = ated concentration (ng of drug / mL of ?uid) MW = Molecular weight (g/mole) **Fluid = plasma Note: any sample concentrations dropped or excluded for the calculation of the statistical values reported (average, rd deviation, and percent coef?cient of variance).
Draize Eye Irritation Scoring Systemfor cornea, iris, and conjunctiva Cornea A. Opacity — Degree of density (area which is most dense is taken for reading) Scattered or diffuse area — details of iris clearly e Easily discernible ucent areas, details of iris slightly ed Opalescent areas, no s of iris visible, size of pupil barely discernible Oocaue, iris invisible LWNt—t B. Area of cornea involved One - uarter or less but not zero Greater than one - uarter but less than one-half Greater than one-half but less than three quarters Greater than three quarters up to whole area bUJNv—l Score equals A x B x 5 Total maximum = 80 A. Values Folds above normal, congestion, swelling, circumcomeal injection (any one or all of these or combination of any f), iris still reacting to light (sluggish reaction is positive) 1 No reaction to light, hemorrhage; gross destruction (any one or all of these 2 Score e uals A X 5 Total ossible maximum = 10 Con'unctiva A. Redness refers to ooalebral con'unctiva onl s de?nitel ed above normal 1 More diffuse, deeper crimson red, individual vessels not easily discernible 2 Diffuse beef red 3 B. Chemosis Any swelling above normal (includes nictitating membrane 1 Obvious swelling with partial eversion of the lids 2 ng with lids about half closed 3 Swelling with lids about half closed to completely closed 4 C. Dischare Any amount different from normal (does not include small amount observed in inner canthus of normal rabbits) 1 Discharge with moistening of the lids and hairs just adjacent to the lids 2 Discharge with moistening of the lids and considerable area around the eye 3 Score equals (A + B + C) x 2 Total maximum = 20 Draize et al. (1944) Methods for the study of irritation and toxicity of substances applied topically to the skin and mucous membranes Scores of 0 are assigned for each parameter ifthe cornea, iris, or conjunctiva, are normal.
EXAMPLES Compound-I is a potent and ive small molecule inhibitor of 2, along with other proangiogenic RTKs such as the FGF receptors (FGFR—l—3), Tie—2, and the ephrin receptor B4 (EPHB-4). Compound-I was shown to inhibit phosphorylation of c RTKs, elial cell proliferation, and pathologic angiogenesis following systemic administration in murine cornea and rat growth plate models, as well as the growth of human tumor xenographs in athymic mice. Regarding ial ophthalmic indications, the Examples of the present application described below demonstrated that l ocular delivery of Compound-I ed signi?cant inhibition of pathologic retinal and choroidal neovascularization in ally-relevant rodent models. A summary of these data follows.
] The following studies were conducted to measure the effect of the disclosed compounds on vascular leak and neovascularization of retina tissue.
EXAMPLE 1 Primary Pharmacodynamics In Vitro Ef?cacy Pharmacofogy ofCompound—I. Compound—I potently inhibits the tyrosine kinase activity of vascular endothelial growth factor receptor-2 (VEGF-2), as well as a select subset of other proangiogenic RTKs, during various in vitro assays. Speci?cally, Compound—I compound blocked VEGF—stimulated VEGFR—2 phosphorylation in whole cells along with the proliferation of cultured endothelial cells. Compound-I inhibited recombinant ne kinase activity of VEGFR—2 and FGFR—2 with a 50% inhibitory concentration (leo) of 10.55 nM (6 ng/mL) and 8.79nM (5 ng/mL), tively; and inhibited VEGFR-2 autophosphorylation in intact cells with an IC50 = 5.2? nM (3 ng/mL). This inhibition was selective versus many other tyrosine kinases, e.g., the VEGFR—2 IC50 was approximately 500x and 1000x lower than those for epidermal growth factor receptor (EGFR) and the insulin receptor (IR) tyrosine kinases, tively (see Table 2).
When using a 10-point titration curve that ranged from 257 to 5000nM (146 — 2845 ng/mL), nd-I exhibited potent inhibition of tyrosine kinase ty for several iogenic growth factor receptors, as evidenced by an ICso < lOOnM (56.89 ng/mL) (see Table 3). The IC50S for this select group of kinases were as follows: recombinant KDR (human m of VEGFR-2) = 1.27nM (0.72 ng/mL), Tie-2 = 10.10nM (5.75 ng/mL), and FGFRs 1—3 = 8.50nM (4.84 anL), 3.08nM (1.75 ng/mL), and 33.9nM (19.29 ng/mL), respectively. The compound also blocked the other high—af?nity VEGF receptor, VEGFR- 1/Flt-1, but with lower potency: IC50 = 122nM (69.41 ng/mL).
Although VEGFR tion appears to be essential for reducing vascular permeability and preventing further neovascular growth, the simultaneous inhibition of VEGF signaling with tion of other growth factor signaling pathways (e.g., PDGF and angiopoietins/Tie2) may be linked to unique therapeutic outcomes. The therapeutic outcomes of a broader inhibition of ing pathways may contribute to the regression of newly established ogic vessels in the ior segment of the eye. 300nM (170.67 ng/mL) of Compound—I completely inhibited VEGFR-2 kinase function (see Table 4) and provided substantial blockade of a similar set of proangiogenic growth factor receptors, including l—3, Tie—2, and EphB-4. An unexpected finding was that the 300 nM concentration was able to completely inhibit the VEGFR-2 kinase function. This concentration falls within the typical range found in the central choroid and retina following ?ve days of topical ocular delivery in s and dogs.
Overview ofDrug Substance andDrug Product Drug Substance: The active pharmaceutical ient (API), Formula II hloride (Compound-I, CP-547,632—01), is a small molecule of a single polymorph.
The API substance is consistently manufactured in purity exceeding 99.7%. Any impurity in drug substance 2 0.15% is suitably quali?ed in toxicology s and the current speci?cation for new unknown dual ties is set to NMT 0.2%. The ?nal drug substance and drug product are analyzed using standard methods.
] Drug Product: Compound—I ophthalmic formulations for clinical studies was manufactured in dosage ths between 0.05% — 1.0% (as nd—I). The strengths used for the GLP batches are 0% (placebo), 0.05%, 0.1%, 0.2%, 0.3%, 0.4%, 0.6%, 0.8%, and 1.0% nd—I. Compound—I ophthalmic formulations (solutions or suspensions) are used for daily, single use, l administration to the eye in the clinical trial. In addition to the active ingredient, the drug product may further contain 0.005% BAK as a preservative, puri?ed water as vehicle, and is pH—adjusted with sodium hydroxide to pH 6.0.
EXAMPLE 2 Sodium Phosphate-based Gel Drop The ophthalmic bene?ts of Compound—I in a sodium phosphate-based formulation (listed in Table 6) results from the self-gelling properties of the API in buffers, such as sodium phosphate. Spontaneous formation of a self—forming, thixotropic gel of Compound-I from a clear on was formed by increasing API concentration in sodium phosphate. This gel initially appeared clear and then demonstrated increased thickness/viscosity at higher API concentrations, as well as becoming increasingly more opaque, i.e., turbid. Once the API concentration in the phosphate buffer reached super—saturated state, insoluble particulates of nd—I also were observed within the gel.
Application of a gel with increased viscosity to the surface of the eye increases corneal residence time. Increased corneal residence time in turn tates ocular drug absorption. As a result, the intraocular drug concentrations of viscous gels increase in comparison to non—viscous formulations, such as water—like solutions. One way to increase viscosity is to use various ity—enhancing excipients, e.g., carboxymethylcellulose, which in effect achieves increased intraocular absorption of different drug substances following topical ocular administration. In this study, r, a thixotropic gel of nd—I was unexpectedly formed in the absence of any viscosity—enhancing excipients.
For example, when Compound—I was dissolved into a simple buffer, such as sodium phosphate, a thixotropic gel was formed. The thixotropic gel, which was formed without any viscosity-enhancing excipients, was formulated as a Gel Drop in this Example.
The Gel Drop of Compound—I was applied to eyes of Dutch—belted rabbits. Gel Drops of Compound—I were administered to Belted rabbits for 4 or 5 utive days, with three times daily dosing. The trations of Compound—I at the target tissues were measured at 1 hour following the last administered dose. Delivery of Compound-I to the posterior segment tissues was dose-dependent and dose-frequency dependent.
The Gel Drop ations (listed in Table 6) differed in several aspects, such as API tration, sodium phosphate concentration, presence or absence of tonicity (glycerin) or preservative (benzalkoniumchloride/BAK) , solubilizing surfactants (polysorbate 80, tyloxapol, and/or poloxamer), and pH.
EXAMPLE 3 Tromethamine-based sion Compound—I (about 1 mg/mL to about 10 mg/mL) in a tromethamine—based formulation formed a suspension. The suspension of nd-I in a tromethamine-based formulation had >95% of the active drug substance in an insoluble form. This teristic is distinguishable from the soluble or semi—soluble state of Compound-I in the Gel Drop (the Gel Drop (gel) is not an entirely soluble state as concentration of the active agent increases) or in a Cyclodextrin-based formulation. Tromethamine-based formulations of Compound-I showed increased turbidity with increasing active agent concentration. Administering a topical drop of Compound—I suspension to the eye (which is a combination of soluble and insoluble active agent components) was expected to provide unique bene?ts relevant to both safety/tolerability and ef?cacy.
Tromethamine-based suspension of Compound—I was administered to Dutch— Belted rabbits for 4 or 5 consecutive days with three times daily dosing. Ocular tissue and plasma concentrations of Compound-I were measured at 1 hour following the last administered dose. nd-I in the tromethamine-based suspension delivered concentrations to the target tissues n 10-1000x of its cellular IC50 for the various pro- angiogenic RTKs. See Table 7.
The l safety and tolerability of topical Compound-l was a direct consequence of the amount of soluble (as opposed to insoluble) active agent applied to the corneal surface, and the ant l tissue concentration. Animals that received topical ocular administration of the tromethamine—based suspension were able to tolerate up to higher level of the active agent tration in the formulation, as compared to equimolar formulations of the sodium phosphate—based Gel Drop. Results obtained from both Dutch— belted rabbits and beagle dogs suggested that ocular side effects, such as discomfort and in?ammation and in some cases, corneal thinning, were more consistently observed when the cornea concentration of Compound—I exceeded IOOpM.
Administration of the tromethamine—based suspension had the unexpected effect on the ocular ilability of Compound-I in the posterior segment. The ocular bioavailability of Compound-I in the ior segment was observed to be directly proportional to the total amount of drug administered (insoluble plus soluble, see Table 7).
Although the insoluble drug particulates were not readily ble to anterior segment tissues; the nt and unique physicochemical properties of Compound—I allowed both insoluble and soluble ents to gain entry to posterior segment s, such as the choroid and retina. Consequently, even higher drug concentrations than those achieved with Gel Drop formulations containing equivalent amounts of the active agent were achieved with the tromethamine—based suspension. Thus, tromethamine—based suspension provided: a) improved l tolerability and b) increased bioavailability to the posterior segment, particularly to the choroid, the primary target tissue for treating neovascular (wet) AMD.
EXAMPLE 4 Cyclodextrin-based solution Cyclodextrins, which are cyclic oligosaccharides made up of six to eight dextrose units (0-, B—, and y—CDs) joined through one to four bonds, are nown for their ability to act as a solubilizing agent for relatively insoluble drugs. See Stella & He, Cyclodextrins, Toxicol. Pathol, 36: 30—42 (2008).
] A clinical formulation of Compound—I or its free base ophthalmic Solution in 2- hydroxypropyl—B—cyclodextrin (HP—B—CD, KLEPTOSE® HPB) at equal to or more than 1:6 molar ratio or Sulfobutylether-B-cyclodextrin (SBE—B—CD, CAPTISOL®) at B at equal to or more than 1:2 ratio provided solubility with clinical dose strengths of 0. 1-1.0% Compound-I.
Cyclodextrin-based solutions of Compound-I or its free base not only improved solubility of the active agent into a uniform solution, but, upon topical ocular administration, also had a novel and previously unobserved characteristic of signi?cantly increased therapeutic index of the active agent at the posterior segment of the eye. The solution d anterior segment exposure, thereby providing increased tration of the active in the solution and increased delivery frequency, which maintained high ior segment concentrations. Both of these ial characteristics are related to the known property of cyclodextrin to form hydrophilic complexes with hydrophobic drugs. See Stella & He, Cyclodextrins, Toxicol. Pathol., 36: 30—42 (2008). When formulated with nd—I or its free base, extrin formed a clear, colorless solution and ted water—like viscosity.
Following topical ocular administration, the Compound—l/cyclodextrin complex had the appearance of being pharmacologically inactive and metabolically inert. The Compound— I/cyclodextrin complex conferred corneal bility until cyclodextrin spontaneously iated from the active agent, thus making available high concentration of Compound-I at its intended site of action in the posterior segment of the eye, e. g., choroid and retina.
During topical ocular dosing s lasting from 1 to 30 days in Dutch-belted rabbits, cyclodextrin—based solutions of Compound—I demonstrated dramatically lowered l exposures compared to Gel Drop formulations (see Example 2) at similar drug concentrations. The use of cyclodextrin—based ons of Compound—I provided an approximate 10x reduction in corneal concentrations, as compared to dosing with equimolar formulations of the Gel Drop. Consequently, after 30 days of topical ocular dosing of 0.6% Compound—I as a cyclodextrin—based solution, no untoward ?ndings were attributed to test— article or vehicle. The cyclodextrin—based solution of Compound—I also achieved equal or significantly higher concentrations of drug within the posterior segment target tissues, such as at the central choroid and the central retina. The combined effects of decreasing l drug exposure so as to avoid poor ocular tolerability, while increasing ior segment bioavailability so as to increase RTK inhibition, may significantly increase the therapeutic index and corresponding bene?t(s) experienced by patients.
For both suspension—based formulations (see Example 3) and the cyclodextrin formulations, the therapeutic window is expanded due to significantly reduced exposure (10— 100X or 1—2 log reduction). The reduced exposure improves corneal /tolerability, which allows higher concentrations or frequency of dosing of Compound—I to be administered topically. The higher concentrations s the drug to achieve higher back of the eye target tissue concentrations, which improves the therapeutic ef?cacy of Compound-I.
] This study demonstrated that in rabbits and dogs, topical ocular dosing of ophthalmic gel drops, was associated with high l tissue exposure (BlOOuM) and corresponding rd observations in the anterior segment, such as discomfort, corneal and conjunctival in?ammation, l epithelial erosion and/or thinning and degeneration.
In contrast, ed topical ocular dosing of Compound—1 ophthalmic solution produced corneal exposure that are roughly 5 tolO—fold lower than an equimolar dose of ophthalmic gel drops, and are free of untoward clinical or histopathologic ?ndings. Furthermore, topical ocular dosing with Compound-I lmic solution achieved equal or higher target therapeutic exposure in the l choroid in comparison to an equimolar dose of the ophthalmic gel drop. Overall, the combination of decreased corneal exposure and corresponding improved ocular tolerability, while simultaneously ining or promoting drug delivery to the posterior segment target s, along with improved physicochemical stability, will provide greater bene?t to patients compared to the ophthalmic Gel Drop formulation.
EXAMPLE 5 I- to 5-day PK results with topical ocular Compound-I in extrin-based Solutions Following topical ocular dose administration of various formulations and dosage ns of Compound-I in Dutch Belted rabbits, ocular pharmacokinetics was investigated.
Nine (9) ent topical ocular formulations having three doses of Compound—I were administered either once per day (q.d.) or twice per day (b.i.d.) for 1, 4, or 5 consecutive days. The study design (see Tables 10A—B) ted of seventy—two (72) rabbits each receiving a 30 uL bilateral topical ocular dose of one of three (3) Compound-I formulations, or vehicle formulation, using a positive displacement pipette.
] The composition of each Compound—I formulation is described in Table 8A. A11 doses were administered within :: 1 hour of the scheduled dose time. On day 1, Groups 1, 2, 4—6, 8, 10, 11, 13, 15, and 17 received one dose (q.d.) for either one (1) or four (4) days. On days 1 through 4, Groups 3, 7, 9, 12, 14, and 16 received b.i.d. dosing approximately 8 hours apart at 7:00 AM and 3:00 PM for four (4) days. Animals in Groups 18 and 19 received b.i.d dosing of vehicle only formulations for ?ve (5) consecutive days.
Ocular sampling occurred for Group 1 at 0.5, 1, 2, 4, 8, or 24 hours post—dose relative to the day 1 dose. Ocular sampling ed for Groups 2 and 6 at 1, 8, and 24 hours post—dose relative to the day 5 morning dose. Ocular sampling for Groups 3, 7, 8, 11, and 13 occurred at 1 and 24 hours post-dose relative to the day 5 morning dose. Ocular ng for Groups 4, 9, 10, 12, 14, 15, 16, and 17 occurred at 1 hour post-dose relative to the day 5 morning dose. Group 5 ocular sampling occurred at 0.5, 1, 2, 4, 8, and 24 hours post-dose relative to the day 1 dose. Animals in Groups 18 and 19 were followed only by clinical observations for ?ve (5) days. s humor, cornea, central and peripheral retina, and central and peripheral choroid samples were collected. Aqueous humor, cornea, central retina, and central choroid samples were then assayed. Peripheral retina and peripheral choroid s were assayed only for Groups 1—8, 12, 14, and 16 per study protocol.
Rabbit aqueous humor, cornea, l and peripheral retina, and central and peripheral choroid samples were analyzed. Calibration curves were prepared in l matrix to determine the concentration of Compound—I in the various tissues.
EXAMPLE 6 -day PK s with topical ocular Compound-I in Cyclodextrin-based Solutions Following ocular dose administration of various dose regimens of topical ocular solutions of Compound-I containing hydroxypropyl-B-cyclodextrin D") in Dutch- Belted rabbits, the ocular pharmacokinetics was calculated. Nine (9) different topical ocular solutions having four different doses of Compound—I were administered either once per day (q.d.) or twice per day (b.i.d.) for either 4 or 5 consecutive days. The study design consisted of forty (40) rabbits each receiving a 30 [LL bilateral topical ocular dose of one of four (4) Compound—I dosage strengths, using a positive displacement pipette.
All doses were administered with i 1 hour of the scheduled dose time, except on day 1 for Groups 1, 4, and 10. The ?rst dose on day l was administered at 12:00 PM, and the second dose (for Groups 1 and 10) was administered approximately 4 hours after. This was due to delayed arrival of formulations. All other dosing for these groups was as scheduled.
On day 1, Groups 4—8, and 11—12 received one dose (q.d.) for four (4) days. On days 1 through 4, Groups 1—3, and 9—10 received b.i.d. dosing imately 8 hours apart for four (4) days.
Ocular sampling occurred one hour following the ?rst daily dose on day 5 for all groups, except Groups 5b and 6b, where ocular ng occurred 24 hours after the ?rst daily dose on day 4.
Blood samples for plasma tion were obtained just prior to scheduled euthanasia for all animals. Aqueous humor, cornea, central and peripheral retina, and central and peripheral choroid samples were collected. Cornea, central retina, and central d samples from groups 1-12 were assayed. Aqueous humor, peripheral retina, and peripheral choroid samples were not assayed.
EXAMPLE 7 Concentrations of Compound-I (in MW) in Various Ocular Fluids and s The ocular on of Compound—I comprising cyclodextrin was prepared and tested in different groups of animals. Upon l ocular administration of a solution of 0.4% (4 mg/mL) nd-I and cyclodextrin, the concentration of the active agent was measured in various tissues and ?uids of the eye. Average concentration of Compound—I was measured in the central choroid, central retina, aqueous humor, and cornea. Compound—I was in a on (0.4% or 4 mg/mL) with 8.41% KLEPTOSE® and 0.142% phosphate buffer; 8.9% KLEPTOSE® HPB and 0.142% phosphate; 4.88% CAPTISOL® and 0.142% ate; or 4.88% CAPTISOL® and 0.122% ate. See Table 10A-B.
The central choroid concentration of Compound—I was between 0.259 [1M and 0.769 uM. See Table 10A. The central retina concentration of Compound—I was between 0.0531 [1M — 0.124 uM. See Table 10A. The aqueous humor concentration of Compound-I was between 0.00313 uM — 6 uM. See Table 10A. And the corneal concentration of Compound—I was 6.49 uM — 30 uM. See Table 10A. The cyclodextrins used to prepare the solutions were KLEPTOSE® HPB or CAPTISOL®. See Table 10B.
EXAMPLE 8 Ocular Toxicology Studies Dose—limiting ocular toxicity was characterized by l and conjunctival ?ndings in Dutch—Belted rabbits and beagle dogs. These ocular ?ndings from repeat—dose toxicology studies with Compound—I ophthalmic gel drops were based upon clinical ophthalmic and histopathologic evaluations and limited to ctival hyperemia, chemosis, congestion, and discharge, corneal opaci?cation and epithelial erosion, and keratoconjunctivitis. No untoward alterations involving deeper structures of the eye (iris, lens, ciliary body, retina, d, sclera) or the optic nerve were observed. Retinal function was normal in all test e and vehicle treated groups during full-?eld electroretinograms performed in rabbits.
The objectives for the exploratory ocular toxicology studies were to identify: a) a topical ocular formulation that was well tolerated and b) one that could achieve the targeted therapeutic concentrations of Compound—I in the l choroid.
Mean Compound-I ocular tissue concentrations following twice daily topical dosing with 0.3% Compound-I lmic gel drop formulations with and without benzylalkonium chloride were t in the cornea (236-260uM) >>periphera1 choroid . 10 uM), central choroid (0.340-0.496uM), peripheral retina (0.150-0.309uM) and aqueous humor (0.0197-0.0395uM) in Dutch-Belted rabbits.
Tris—based suspension ations were well tolerated, where al ophthalmic examinations revealed a notable absence of l ?ndings with only a few sporadic incidences of mild conjunctivitis in Dutch—Belted rabbits. Moreover, the eyes from animals that had received 0.3% w/v Compound—I Tris—based suspension twice daily for 30 days were considered normal during microscopic evaluations. Mean Compound—I ocular tissue concentrations, assessed at 1 hour i 15 minutes after the first daily topical ocular dose on day for the twice daily l dosing with 0.3% Compound—1 Tris—based suspensions with and t benzylalkonium chloride, were highest in the cornea (2.69-3.10uM) >>peripheral d (0.781—1.21uM), central choroid (0.303—0.319uM), peripheral retina (0.0819— 0.0868uM), central retina (0.0495-0.0592uM), and aqueous humor (0.00127—0.00145uM).
Cyclodextrin-based solutions, using hydroxypropyl—beta—cyclodextrin (HP—B—CD, KLEPTOSE® HPB), were well tolerated when stered topically for 30 days, twice daily at 0.1% Compound-I (2.1% HP-B-CD), twice daily at 0.2% Compound-I (4.21% HP-B- CD), once or twice daily at 0.4% Compound—I (8.41% HP—B—CD), and once or twice daily at 0.6% Compound—I (up to 12.62% HP—B—CD) in Dutch—Belted rabbits. Moreover, in a similar repeat dosing study, cyclodextrin—based solutions of 0.4% w/v Compound—1 in KLEPTOSE® HPB, KLEPTOSE® HP, or CAPTISOL® were well—tolerated when dosed twice daily for 24 days. No overt ocular toxicity related to Compound—I or vehicle treatment was found during al ophthalmic or microscopic examinations in either study.
In the 24—day study, ocular tissue concentrations from eyes treated with cyclodextrin—based solutions of Compound—I were assessed at 1 hour :: 15 minutes after the first daily topical ocular dose on day 24 were in descending order highest in the cornea (6.49— 30uM) >> center—punch choroid (0.212—0.769uM) > center—punch retina (0.053 1-0. 124) > aqueous humor 0007).
In summary, dose—limiting corneal toxicity was observed with nd—I ophthalmic Gel Drop formulations. The ophthalmic Gel Drop renders ?ve-fold to fifteen— fold higher corneal concentrations of Compound—I compared to cyclodextrin based solution, and ?fty—fold to hundred-fold higher corneal concentrations of nd—1 compared to ased sions. Compound—I Tris—based sions and extrin—based solutions were well tolerated with no evidence of overt ocular toxicity. Dose levels that were well tolerated for the cyclodextrin-based solutions or ased suspensions of Compound—I when administered once or twice daily ranged from about 0.005% to about 5.0% w/v for at least 30 days. extrin-based solutions also provided the highest central choroid concentrations of Compound-I when using equimolar doses of the three formulations, and met or exceeded target therapeutic concentrations.
EXAMPLE 9 Phase I Protocolfor Dose-Escalation Study in Patients with NeovascularAMD The Phase I study is a twelve—week, abel, dose-escalating, multi-center trial designed to evaluate the safety, tolerability, and cokinetics following topical ocular administration of Compound-I in patients with neovascular age-related macular degeneration (AMD). Up to 60 patients total are d one to two times daily with topical ocular dosing of nd-I ophthalmic solution for three months, where three dose-escalating monotherapy arms and one adjunct therapy arm using a single intravitreal injection of LUCENTIS® plus the maximally-tolerated monotherapy dose are planned (15 patients per treatment arm). Patients that meet eci?ed vision and CNV lesion criteria con?rmed by an independent reading center are allowed to aneously discontinue topical ocular dosing and receive treatment with standard—of—care.
Compound—I ophthalmic solution for clinical studies is manufactured in at least 3 dosage strengths, ranging from 0.1% to 1.0% (as Compound—l). The strengths for the GLP s are 0% (placebo), 0.1%, 0.3%, 0.6%, and 1.0% Compound—1 HCl. Up to 2— to 3—fold ental doses (approximately 1/2 log unit steps) are administered to succeeding cohorts.
EXAMPLE 10 A "non—gel," "non—Viscous," homogeneous ophthalmic solution topical formulation that is both physically and chemically stable over the drug strengths of 01—10% (1 to 10 mg/ml) was prepared by measuring the Compound-1 concentration, cyclodextrin complexing agent concentration, pH, and tonicity, on Compound-1 solubility and ity. A suitable buffering system prevents pH drift on stability at concentrations less than 1 mg/mL.
Both phosphate and Trometamol (Tris) were ted as buffering agents. Sodium chloride was used to adjust tonicity.
The product quality attributes are shown in Table 14.
Table 14: Product Quality Attributes t Solubill' m_ mL Color Appearance Clear colorless with no visually apparent of formulation H 5.5 — 7 .0 Free ?owin_, water-like and ?lterable Formulation Preparation The formulations outlined in Tables 12 and 13 were prepared using the general procedure listed.
The formulation was made up to volume with water for injection and stirred for 30 minutes at 500 rpm. Final pH was checked and adjusted with either, NaOH or HCl to the target range. Approximately 5ml aliquots is directly ?ltered into semi—transparent 5ml LDPE bottles while continuously stirring at nt speed with the aid of Watson Marlow Pumpsil D tubing, fitted to a Flexicon ?ller and attached to 0.2 micron PVDF capsule ?lter. Samples are stored at 2—8°C until all sample preparation is complete. All samples will then be submitted to analytical for storage and testing.
EXAMPLE 11 EGFR tyrosinephosphorylation assay in corneal lial cells (hTCEpi cells) to determine EGFR activity ofcompounds ofFormula I or II An EGFR tyrosine assay in corneal epithelial cells was med to ine whether higher concentrations of EGF can overcome inhibition of EGFR kinase ty by a compound of Formula I or II. mm dishes of hTCEpi cells were serum starved and then were pre—treated with different concentrations of a nd of Formula I or H, e.g., Compound-I, or a control, AG1478 (an EGFR kinase inhibitor), for 30 s, followed by treatment with EGF (10, 50, 100 ng/ml) for 15 minutes. Six concentrations of a compound of Formula I or II, e.g., 0, 1 uM, 3 uM, 10 uM, 30 uM, 100 uM, or 1 uM AG1478 as a control, were used for each EGF tration (10, 50, 100 ng/ml). Additional ls run were no compound of Formula I or II or no EGF. All experiments were repeated three times. Cells were then harvested and immunoblotted for determination of phosphorylated EGFR (tyrosine 1068 and tyrosine 1045) and total EGFR.
EGFR orylation serves as a readout of receptor activity. As shown in Fig. 1, high concentrations of a compound of Formula I or II were able to inhibit ligand- stimulated EGFR tyrosine phosphorylation, and increasing concentrations of a compound of Formula I or II decreased EGFR tyrosine phosphorylation at both sites. Increasing concentrations of EGF overcome the EGFR inhibition and l ity due to the compound of Formula I or II (Fig. l). The EGFR—speci?c inhibitor, AG1478, has an IC50 = 3 nM, and at 1 uM AG1478 completely blocked EGFR phosphorylation. It is estimated that 100 uM of a compound of Formula I or II inhibits EGFR phosphorylation ~ 75-80%.
EXAMPLE 12 EGFR tyrosinephosphorylation assay in corneal epithelial cells (hTCEpi cells) with compounds ofFormula I or II, and a second active agent An EGFR tyrosine assay in l epithelial cells was performed to determine if vitamin K, nicotinic acid, or nicotinamide can prevent compounds of Formula I or 11 from inhibiting EGFR and if there is an increase in EGFR activity. mm dishes of hTCEpi cells were serum starved and then were pre—treated with vitamin K (50 uM), nicotinic acid (10 uM), or nicotinamide (lOuM) for 4 hours, followed by treatment with ent concentrations of a compound of Formula I or H, e.g., Compound-I, for 30 minutes. The cells were then incubated with EGF (50 ng/ml) for 15 s. Cells were harvested and cell lysates were immunoblotted for determination of phosphorylated EGFR (tyrosine 1068 and tyrosine 1045) and total EGFR concentration. As shown in Fig. 2, vitamin K, nicotinic acid, or nicotinamide overcome Formula 1 or 11 nd—mediated corneal adversity (EGFR inhibition). In addition, Fig. 2 shows that Vitamin K3 (menadione) most effectively attenuated Formula 1 or 11 compound—mediated inhibition of EGFR tyrosine phosphorylation.
EXAMPLE 13 Determination ofcell ion/proliferation (in vitro wound g) in corneal epithelial cells (hTCEpi cells): e?ects ofcompounds ofFormula I or II andEGF Determination of cell migration/proliferation in corneal epithelial cells was performed to determine cell migration/proliferation in the presence of varying concentrations of a compound of Formula I or II and EGF. hTCEpi cells were plated with silicone plugs. The cells were then serum starved and pre-treated with g concentrations of a compound of Formula I or 11 or a control compound, AG1478, for 30 s. The silicone plugs were then d to create the acellular area. The cells were photographed and then treated with EGF (10, 50, or 100 ng/ml) or VEGF (10 ng/ml) together with a compound of a I or II or AG1478 for 16 hours.
The cells were again raphed and cell migration was ?ed from the micrographs.
All experiments were repeated three times.
Four concentrations of a compound of Formula I or II (0, 3 uM, 10 uM, 30 uM) or 3.2 uM AG1478 (an EGFR kinase inhibitor), were used for each EGF concentration or VEGF. Additional controls run ed no compound of Formula 1 or 11 and no EGF.
The data allows determination of whether a compound of Formula 1 or 11 prevents the in vitro measures of corneal epithelial wound healing (cell migration and proliferation) and if higher levels of EGF overcome receptor tyrosine kinase inhibition. As shown in Fig. 3A and 3B, a nd of Formula I or 11 caused a dose-dependent inhibition of EGFR- mediated hTCEpi cell in vitro "wound healing".
Determination ofcell ion/proliferation (in vitro wound healing) in corneal epithelial cells (hTCEpi : effects ofcompounds ula I or II and a second active agent Determination of cell migration/proliferation in corneal epithelial cells was performed to determine cell migration/proliferation in the presence of varying concentrations of a compound of Formula I or II alone or together with vitamin K, nicotinic acid, or namide. ] hTCEpi cells were plated with silicone plugs. The cells were then serum starved and pre-treated with varying concentrations of vitamin K3 analog, one, for 4 hours and supplemented with the varying concentrations of a compound of Formula I or II for 30 minutes. The plugs were removed. The cells were photographed and then treated with various concentrations of vitamin K, EGF, and/or a compound of Formula I or II. The cells were again photographed and cell ion was quanti?ed from the micrographs.
Additional controls run included no compound of Formula I or II and no EGF. All experiments were repeated three times.
As shown in Fig. 4A and 4B, the inhibitory effects of a compound of Formula I or II on in vitro wound healing can be reversed by the addition of 0.3 uM menadione. There was a statistically signi?cant difference in in vitro wound healing when 0.3 uM of a compound of Formula I or II was combined with 0.3 uM menadione versus 0.3 uM of a compound of Formula I or II alone.
EXAMPLE 15 Determination l and EGF-mediated corneal wound healing in vivo: e?ects of compounds ofFormula I or II Determination of the effects of a compound of Formula I or II on basal and ligand stimulated rates of corneal wound healing was ined in C57/B1 mice.
Corneas of 8 week old C57/Bl mice were d (1.5 mm super?cial epithelial wound) and then pre-treated with a compound of FormulaI or II (0, 1.0 uM, 10 uM, 30 uM, or 50 uM), followed by addition of EGF (0, 10, 100 ng/ml). As a control, AG1478 (an EGFR kinase inhibitor) was used in the absence and presence of 100 ng/ml EGF. Wound size was monitored by ?uorescein staining and ?uorescent photography over the course of 40 hours.
Wound closure was quantified using Image J software.
As shown in Fig. 5A, 5B, 5C, and 5D, in viva corneal lial wound healing was reduced in the presence of a compound of Formula I or II in a dose dependent .
Maximal inhibition of wound healing was observed with a compound of Formula I or II concentrations that were ?ve times greater than those of AG1478. Pre-treatment with a nd of Formula I or II for 48 hours before making the wound to the cornea inhibited corneal epithelial wound g at a lower concentration of a compound of Formula I or 11 than without the pre-treatment.
EXAMPLE 16 Determination ofbasal and EGF-mediated corneal wound healing in vivo: effects of compound ofFormula I or II and a second active agent Determination of the effects of a compound of Formula I or II and/or a second active agent on corneal wound healing was determined in C57/Bl mice.
Corneas of 8 week old C57/Bl mice were pre—treated with topical administration of vehicle, 0.3 uM menadione, 10 uM of a compound of Formula I or II, 0.3 uM one and 10 uM of a compound of Formula I or II 48 hours before the corneas were d (1.5 mm super?cial epithelial wound). Wound size was monitored by fluorescein staining and ?uorescent raphy. Wound closure was monitored over the course of 40 hours.
As shown in Fig. 6A, 6B, and 6C, menadione (0.3 uM) reversed the effects of a compound of Formula I or II (10 uM) pre-treatment on corneal epithelial wound healing.
EXAMPLE 17 Effect ofmenadione on inhibition of VEGFR by compounds ofFormula I or II Human retinal endothelial cells were treated with 0, 0.3, or 3 uM menadione for 4 hours, followed by 30 minutes of treatment with varying concentrations of a nd of Formula I or II (1 nM, 10 nM, 100 nM, or 1 pM) in menadione. The cells were then stimulated with 0.5 nM (10 ng/ml) VEGF. The cells were lysed. The lysates were resolved by 7.5% SDS—PAGE, transferred to ellulose, and immunoblotted for phosphorylated VEGFR2, total VEGFR2, or a-tubulin as a loading control. As shown in Fig. 7A, menadione did not change the IC50 of inhibition ofVEGFR2 phosphorylation mediated by a compound of Formula I or II.
EXAMPLE 18 E?ect ofmenadione on theproliferation ofprimary human retinal ascular elial cells Human retinal endothelial cells were pretreated with one for 4 hours, then with the indicated concentration of a compound of Formula I or II for 30 minutes, and supplemented with 0.5 nM (10 ng/ml) VEGF overnight. Viable cells were ?ed by Alamar Blue Assay (Thermo Fisher). Data were plated as the fold growth relative to cells treated with no VEGF or a compound of Formula I or II, and are shown as the e :: S.E.M. in Fig. 7B. As shown in Fig. 7B, menadione did not change the IC50 of inhibition of VEGF-mediated retinal endothelial cell proliferation mediated by a compound of Formula I or II.
EXAMPLE 19 Determination ofmechanism by which menadione enhances EGFR Activity hTCEpi cells were pretreated with menadione (0, 0.3, or 3.0 uM) for 4 hours.
Cells were then incubated with 8.0 nM (50 ng/ml) EGF for 0-3 hours. The cells were lysed and the lysates were resolved by SDS—PAGE, and immunoblotted for phosphorylated EGFR (pY1068), total EGFR, or ulin. As shown in Fig. 8, menadione treatment did not change basal EGFR phosphorylation or slow EGFR dephosphorylation. 0.3 mM menadione slowed the kinetics of EGFR degradation, indicating that menadione may sustain EGFR receptor ing by slowing the kinetics of receptor degradation or increasing the rate of new receptor synthesis.
EXAMPLE 20-1 Ocularpharmacokinetics offormulations ofthe application in Dutch Belted rabbits ] A P study was conducted to assess the ocular pharmacokinetics of a ?rst active agent of the present application (e.g., a II or Compound—I), in suspension or solution, when stered once or twice daily, as a topical instillation, to both eyes of Dutch Belted rabbits for four days. The study design (Table 15—1) consisted of ?ve (45) Dutch Belted rabbits, each receiving a 30 uL bilateral topical ocular dose. The administration was conducted according to the dosing regimen in Table 15—1.
Table 15-1: Study design Dosing No. of . . Frequency Sample Test Article Formulation Rabbits and Time Points Durationa 0.4% Formula II; 0.08% Pluronic F-127; 30 ML/ (1015: ), 2.5% Glycerol; eye/ dose 5 Laroe Particles in sus-ension 0.4% Formula II; 1 hr post 0.08% Pluronic F-127; 30 uL/ dose on Day 2.5% Glycerol; eye/ dose Nano Particles in suspension 0.2% Formula II; 1 hr post uL / 0.04% Pluronic F-127; dose 2n Day eye/ dose 2.5% GI cerol; 0.2% Formula II; 1 hr post 0.04% Pluronic F-127; 30 pL / 4 3 dose on Day 2.5% Glycerol; eye/ dose Nano Particles in susoension 1 hr post Compound-l 30 pL/ QD for 4 4 mg/mL dose on Day Ophthalmic on eye/ dose Days 1 hr post Compound-l 30 pL/ 6 F 3 2mg/mL dose on Day lmic solution eye/dose 1 hr post Compound-l 30 pL/ 7 G 3 1mg/mL dose on Day lmic on eye/dose 1 hr post Compound-l 0.1 30 pL/ QD for 4 H 3 dose on Day Ophthalmic solution mg/mL eye/ dose Days II-0.4% Formula II; 1 hr post 0.08% Pluronic F—127; 30 pL/ 4 m imL9 dose on Day 2.5% Glycerol; eye/ dose Laroe Particles in susoension 0.4% Formula II; 1 hr post 0.08% Pluronic F-127; 30 pL / dose on Day 2.5% Glycerol; eye/ dose Nano Particles in suspension 0.2% Formula II; 1 hr post 0.04% Pluronic F-127; 30 uL / dose on Day 2.5% Glycerol; eye/ dose Large Particles in suspension 0.2% Formula II; 1 hr post 0.04% Pluronic F-127; 30 uL / dose on Day 2.5% Glycerol; eye/ dose Nano Particles in suSoension II_-?5%; an: 14 N 3 gzriwlllalmlic solution 1mg/mL egg/lit); B|[|:))af)(/)sr4 (1332251052), .. o s anagram... .3... 4. 8'35: 4.32%.. aA single dose will be administered on Day 5.
CO — once daily; BID — twice daily Ocular sampling occurred one hour following the first daily dose on Day 5 for all groups. Whole blood samples using KzEDTA as an anticoagulant were collected from all animals on Day 5, 1 hour post dose. The whole blood was placed on ice until samples were spun in a centrifuge to separate the plasma. Aqueous humor, conjunctiva, cornea, central retina, peripheral , central choroid, and eral choroid samples were collected following necropsy. Samples were then frozen at —?0°C or lower. All Groups 1—15 plasma, central retina, central choroid, and cornea samples were assayed.
For all dose groups, ocular tissue concentrations of Formula II or Compound—I assessed on Day 5 were in ding order: highest in the cornea >> central choroid > central retina (Table 15-2). The highest cornea concentrations of Formula II or Compound-I were seen in Group 10. The lowest cornea concentrations of Formula II or Compound—I were seen in Group 8. In general, higher l retina and central choroid concentrations were associated with higher cornea concentrations of Formula II or Compound—I. No advantage of other formulations over ophthalmic solutions of Formula II or nd—I was seen with respect to a reduction in cornea concentrations or an increase in central retina or central choroid concentrations of Formula II or Compound-I.
Table 15-2 Plasma* Central Central Cornea Group (pM) Retina (pM) Choroid (pM) (pM) 1 0.00500 0.0560 0.152 22.1 0.00350 0.155 29.0 0.00391 0.336 62.2 11 6 0.162 16.0 12 0.00308 0.111 28.8 1 3 0.00256 0.149 16.1 14 0.00214 0.0316 0.108 6.63 ge based on n=3 Plasma LLOQ = 0.00188 uM Central Retina LLOQ = 0.00751 pM Central Choroid LLOQ = 0.0563 pM Cornea LLOQ = 0.0376 uM Gross ocular examinations using the Draize scale for scoring ocular irritation were med on all animals prior to the ?rst dose on each day during the study. All formulations were well tolerated. In addition, animals were ed for general health pre- study, during , and at necropsy. All animals assigned to study were normal. Necropsy evaluations consisted of observations in addition to those already noted on Draize Score sheets and Ophthalmologic Exam forms. For all formulations, dosing observations were assessed using the Draize ocular irritation scoring system. No al observations were noted except for the following: prior to necropsy on Day 5, Redness Level 1 in both eyes was noted in one animal in Group I and one animal in Group M.
EXAMPLE 20-2 Ocularpharmacokinetics offormulations ofthe application in Dutch Belted rabbits A non—GLP study was conducted to assess the ocular pharmacokinetics of a ?rst active agent of the present application (e.g, Formula II or Compound—I), in suspension or solution, when administered one, three, or four times daily, as a topical instillation, to both eyes of Dutch Belted rabbits for four days. The study design (Table 16—1) consisted of — nine (39) Dutch Belted rabbits, each receiving a 30 uL bilateral topical ocular dose. The administration was conducted according to the dosing regimen in Table 16-1.
Table 16-1: Study design No of Dosing ' Dose Test Article Formulation Conc. Frequency and Rabbits. Volume Duration. a 1 hr post Compound-l QD for 4 Days dose on Ophthalmic Solution (Control) mg/mL eye/ dose Day 5 1 hr post Compound-l QD for 4 Days dose on Ophthalmic Solution (Control) mg/mL eye/ dose Day 5 1 hr post Compound-l . QD for 4 Days Ophthalmic Solution mg/mL eye/ dose daze 2n 1 hr post Compound-l 0 .1 30 pL/ TID for 4 Days Ophthalmic on m eye/ dose (18:9 2h 0.4% Formula II; 1 hr post 4 30 pL/ Large 3 pm particles in QD for 4 Days dose on mg/mL eye/ dose sus.ension Da 5 0.4% Formula II; 1 hr post 4 30 L/ Large les, 3 pm + HPBCD p QD for 4 Days dose on in suspension g/mL eye/ dose Day 5 0.4% a II; 1 hr post Extra-large 35 pm particles In QD for 4 Days dose on mg/mL eye/ dose suspenSIon Day 5 0.2% Formula II; 1 hr post Extra-large 35 um les in OD for 4 Days dose on mgimL eye/ dose suSoension Da 5 0.4% Formula II; 1 hr post 4 30 uL/ 3 Extra-large 35 um + HPBCD in QD for 4 Days dose on mgimL eye/ dose suSoension Da 5 0.3% Comoound-l; 30 L/ QDfor4Das 1hr ost Tris; mg/mL eye/ dose dose on Particles in susoension Da 5 0.3% Compound-l; 1 hr post 3 30 u L/ 11 Tris + HPBCD; QD for 4 Days dose on . . . mg/mL eye/ dose Particles in susoenSIon Da 5 0.4% Formula II; HPMC; 1 hr post 12 L 3 0.3 uM Vit K3; QD for4 Days dose on m lmLg eye/ dose Extra-large particles in Day 5 suspension 0.4% Formula II; HPMC; 1 hr post 13 M 3 1.0 pM Vit K3; QD for 4 Days dose on mg/mL eye/ dose Extra-large les in Day 5 aA single dose will be administered on Day 5.
OD — Once daily, TID - Three times daily, QID - Four times daily Ocular sampling occurred one hour following the ?rst daily dose on Day 5 for all groups. Whole blood samples using KZEDTA as an anticoagulant were collected from all animals on Day 5, 1 hour post dose. The whole blood was placed on ice until samples were spun in a centrifuge to separate the plasma. Aqueous humor, conjunctiva, cornea, central retina, peripheral retina, central choroid, and peripheral choroid samples were collected following necropsy. s were then frozen at —70°C or lower. All Groups 1—13 plasma, central retina, central choroid, and cornea samples were assayed.
For all dosing groups, ocular tissue concentrations of Formula II or nd—I assessed on Day 5 were in descending order highest: in the cornea >> central choroid > central retina (Table 16-2). The highest cornea concentrations of Formula II or Compound-I were seen in Group 1. The lowest cornea concentrations of Formula II or nd—I were seen in Group 3. In l, higher central retina and central choroid concentrations were ated with higher cornea concentrations of Formula II or Compound-I. No advantage of other formulations over Formula II or nd—I ophthalmic on was seen with respect to a reduction in cornea concentrations or an increase in central retina or central choroid concentrations of Formula II or Compound—I.
Table 16-2 Central Central lJ P M M 3 4 0.00257 0.00451 0.00202 0.0204 0.0582 13 0.00193 0.0248 0.0700 3.08 *Average based on n=3 Plasma LLOQ = 0.00188 yM Central Retina LLOQ = 1 pM Central Choroid LLOQ = 0.0282 pM Cornea LLOQ = 0.0376 "M Gross ocular examinations using the Draize scale for scoring ocular irritation were performed on all animals prior to the ?rst dose on each day during the study. All ations were well tolerated. In on, animals were observed for general health pre- study, during dosing, and at sy. All animals assigned to study were normal. Necropsy evaluations consisted of observations in addition to those already noted on Draize Score sheets and Ophthalmologic Exam forms. For all formulations, dosing observations were assessed using the Draize ocular irritation scoring system. The following al observations were noted throughout the duration of the study: animals in Groups 7, 9, 10, 11, 12, and 13 enced increased blinking following dosing throughout various days of the study; one animal in Group 11 exhibited Discharge Level 1 on Day 3.
EXAMPLE 20-3 Ocularpharmacokinetics offormuiations ofthe application in Dutch Belted rabbits A non-GLP study was conducted to assess the ocular pharmacokinetics of a ?rst active agent of the present application (e.g., Formula II or Compound-I), in suspension, when stered one, two, or three times daily, as a topical instillation, to both eyes of Dutch Belted rabbits for four days. The study design (Table l?—1) consisted of thirty—nine (3 9) Dutch Belted rabbits, each receiving a 30 pL bilateral topical ocular dose. Various suspension formulations of the present application were evaluated and compared against two control formulations: a) a cyclodextrin—based eye drops solution ol Group 13, Table 17-1), and b) a pilot suspension formulation used in us rabbit pharmacokinetic s, (Group 10, Table 17-1). A comparison of the formulation variables for each suspension listed in Table 17—1 is provided in Table 17—2. Formula II or Compound—I was used in the test suspensions, whereas the Eye Drops solution formulation is derived with the Compound— Table 17-1: Study design Dosing Frequency Sample Test Article Formulation and Time Points Durationa Formula II; 1 hr post p L / Pluronic; 0.3 mgimL dose on Day eye/ dose pm particles 5 Formula II; 1 hr post p L / ic; 0.3 mgimL dose on Day eye/ dose m oarticles 5 Formula II; 1 hr post p L / HPMC; 4 mg/mL dose on Day eye/ dose m oarticles 5 a II; 1 hr post pL / Pluronic; 4 mg/mL dose on Day eye/ dose m oarticles Control 5 Formula II; 1 hr post uL / HPMC; 4 mg/mL dose on Day eye/ dose 50-60 pm particles 5 Formula II; 1 hr post uL / Pluronic; 4 mg/mL dose on Day eye/ dose 50-60 m oarticles 5 Formula II; 1 hr post uL / PVP; 4 mg/mL dose on Day eye/ dose 50-60 m oarticles 5 Formula II; 1 hr post HPMC; 30 p L / 4 mg/mL dose on Day 1.0 uM Vit K3; eye/ dose 50-60 m oarticles Formula II; 1 hr post ic; 30 p L / 4 mg/mL dose on Day 1.0 uM Vit K3; eye/ dose 50-60 pm particles Compound-I; 1 hr post p L / Tris; 4 mg/mL dose on Day eye/ dose 50-60 m oarticles Control 5 Compound-l; 1 hr post u L / Tris + Tween80; 4 mg/mL dose on Day eye/ dose 50-60 m carticles 5 Compound-l; 1 hr post p L / Tris + HPMC; 4 mgimL dose on Day eye/ dose 50-60 m carticles 5 1 hr post Compound-l; 30 p L / QD for 4 13M 3 4 mgimL dose on Day Ophthalmic Solution eye/ dose Days aA single dose will be administered on Day 5.
OD — Once daily, BID — Twice daily, TID — Three times daily Table 17-2 Group# 1and2 3 4 5 6 7 8 9 10 11 Dose- . 4mg/ 4mgf 4mg/ 4mg! 4mg/ 4mg/ 4*mg/ 4*mg/ th mL mL mL mL mL mL mL mL Free Free Free Free Free Free HCI API Form Free Base HCI Salt Base Salt jzttlc: 35 35 35 50-60 50-60 50-60 50-60 50-60 50-60 . Tris + P'Uron' "5+. izer Pluronic HPMC Pluronic HPMC Pluronic PVP HPMC Tris Tween 0 HPMC Vitamin ' ' ' 1 1 K3 M 2X volume for BID & QD QD QD QD TID dosing (control) (control) freo uencies *Note: requires 4.3 mg/mL of HCI salt to provide 4mg/mL of Free Base Ocular sampling occurred one hour following the ?rst daily dose on Day 5 for all groups. Whole blood samples using KZEDTA as an anticoagulant were collected from all animals on Day 5, 1 hour post dose. The whole blood was placed on ice until s were spun in a fuge to separate the plasma. Aqueous humor, conjunctiva, cornea, central retina, peripheral retina, central choroid, and peripheral choroid samples were collected following necropsy. Samples were then frozen at —70°C or lower. For all Groups l—l3, plasma, central , l choroid, and cornea samples were assayed.
Ocular tissue concentrations of Formula II or Compound—I assessed on Day 5 were in descending order: highest in the cornea >> l choroid > central retina. Corneal trations of Formula II or Compound—I ranged from 1.5 to 14.7 uM with the highest concentrations being observed in Group 13 and the lowest in Group 1. All suspension formulations, Groups 1—12, provided lower average corneal concentrations as compared to the Eye Drops solution control, Group 13. These observations did not seem to be in?uenced by the ce or absence of Vitamin adione in the suspension formulation. Central choroid concentrations ranged from <0.028 to 0.27 "M with the highest concentrations seen in Groups 11 and the lowest in Group 2. Central retina concentrations ranged from 0.009 to 0.069 uM; Group 11 had the highest central retina concentrations and Group 1 had the lowest central retina concentrations. Plasma concentrations of Formula II or Compound-I were very low being approximately 10—fold and 100—fold lower than those in the central retina and central choroid, respectively. See Table 17—3.
Table 17-3 l Central Plasma* Cornea Retina Choroid (P M) (H M) M M 3 0.00205 0.0228 0.0767 4.38 4 0.00220 0.0305 0.0684 5.08 11 0.00410 0.0691 0.265 8.67 12 0.00470 0.0482 0.116 5.82 0.00416 0.0570 0.112 14.7 *Average based on n=3 Plasma LLOQ = 0.00188 pM Central Retina LLOQ = 0.00751 pM Central d LLOQ = 0.0282 pM Cornea LLOQ = 0.0376 pM Additionally, Vitamin K3 concentrations were determined in plasma, l retina, central choroid, and cornea samples for animals from Groups 8 and 9. A calibration curve prepared in control matrix was used to determine the concentration of n K3. See Table 17—4.
Table 17-4 Plasma* Central af?x; Cornea Grou9 (HM) Retina (PM) (M) - —0.465 *Average based on n=3 Plasma LLOQ = 0.291 pM Central Retina LLOQ = 11.6 pm Central Choroid LLOQ = 43.6 "M Cornea LLOQ = 2.91 pM Gross ocular examinations using the Draize scale for scoring ocular irritation were performed on all animals prior to the ?rst dose on each day during the study. All formulations were well tolerated, with the exception that for one animal in Group 11 on Day 4 of dosing, where conjunctival chemosis was noted, and for one animal in Group 12 on Day 4 of dosing, where conjunctival redness was noted. In addition, s were observed for general health udy, during dosing, and at necropsy. All animals assigned to study were normal. Necropsy tions consisted of observations in addition to those already noted on Draize Score sheets. For all formulations, dosing observations were assessed using the Draize ocular irritation scoring system. The following abnormal observations were noted throughout the duration of the study: animals in Groups 4, 6, 7, 8, 9, and 11 experienced increased blinking following dosing throughout various days of the study; one animal in Group 4 exhibited Redness Level 1 in both eyes and Chemosis Level 1 in the left eye prior to necropsy on Day 5; and one animal in Group 5 exhibited Chemosis Level 1 in the right eye prior to necropsy on Day 5.
EXAMPLE 20-5 Ocularpharmacokinetics offormulations ofthe application in Dutch Belted rabbits A non—GLP study was ted to assess the ocular pharmacokinetics of a first active agent of the present application (e.g., Formula II or Compound—I), in sion, when administered one or two times daily, as a topical lation, to both eyes of Dutch Belted s for four days. The study design (Table 18—1) consisted of forty—nine (49) Dutch Belted rabbits, each receiving a 30 uL ral topical ocular dose. Various suspension formulations of the present application were evaluated and compared against a cyclodextrin- based eye drops solution (Control Groups 2 and 3, Table 18—1). A comparison of the formulation variables for each suspension listed in Table 18—1 is provided in Table 18—2.
Formula II or nd—I was used in the test suspensions, whereas the Eye Drops solution formulation is d with Compound—I.
Table 18-1 Dosing No. of Dose Conc. Frequency Sample Time Test Article Formulation Rabbits Volume and Points Duration:11 Compound--;I 0.3 30 IJL/ BID for 4 1 hr post dose Ophthalmic Solution, pH 6 mo/mL e 9/ dose Da 8 on Da 5 Compound--;I 30 IJL/ QD for 4 1 hr post dose Ophthalmic on, pH 6 (Control) mo/mL e e/ dose Da 8 on Da 5 Compound--;I 30 IJL/ QD for 4 1 hr post dose o) 0 Ophthalmic Solution, pH 6 (Control) eye/ dose Days on Day 5 Formula II; 0.08% HPMC+ 2.5% Glycerol +0.2% HEC; 30/I3L/ QB for 4 1 hr post dose .3; U eye ose ays on Day 5 1.0 uM Vit K3; um particles, pH ~7.5 Formula II; 0.08 A) HIDMC + 2.5 A) Glycerol +0 o NJ QD for 4 1 hr post dose 01 [TI 0.2% HEC, eye/ dose Days on Day 5 1.0 uM Vit K3; um les, pH 6 Compound-I; 0.6% TRIS + 0.08% HPMC + 2.0% PH QD for 4 1 hr post dose Glycerol + 0.2% HEC; 1.0 uM Vit K3; eye/ dose Days on Day 5 um particles, pH ~7.5 Compound-I; 0.6% Tris + 2.0% Glycerol + 0.3% 3° "Ll QD for 4 1 hr post dose HEC + 0.04% Tylox; 1.0 uM Vit K3; eye/ dose Days on Day 5 um les, pH 6 Compound-I; 0.6% Tris + 2.0% Glycerol + 0.2% "U QD for 4 1 hr post dose HEC + 0.04% Tylox; 1.0 uM Vit K3; eye/ dose Days on Day 5 um particles, pH 6 Compound-I; 0.6 A) TrIs + 2.0%; Glycerol + 0.3%;0 ' 0 0 pL/ QD for 4 1 hr post dose HEC + 0.04°/ Tylox; 1.0 uM Vit I23; eye/ dose Days on Day 5 m les, oH 7 Compound-I; 0.6 A; TrIs + 2.0%; Glycerol + 0.2%;0 ' 0 0 "Ll QD for 4 1 hr post dose 10J HEC + 0.04°/ Tylox; 1.0 uM Vit I23; eye/ dose Days on Day 5 m articles, 0H 7 Compound-I; 0.6% Tris + 2.0% Glycerol + 0.3% PL/ QD for 4 1 hr post dose 11K HEC + 0.08% HPMC; 1.0 uM Vit K3; eye/ dose Days on Day 5 um particles, pH 6 Compound-l; 0.6 Ag Tm + 2.0%; Glycerol + 0.2/00 ‘ 0 0 43 30 pL/ 1 hr post dose 12L H53 3028: ?3HPMC,0 QD for 4 mg/mL eye/ dose Days on Day 5 um particles, pH 6 Compound—l; 0.6% TrIs + 2.0% Glycerol + 0.2% 4.3 30 NJ QD for 4 1 hr post dose 13 M 3 HEC + 0.08% HPMC; 1_0 uM Vit K3; mgimL eye/ dose Days on Day 5 Native (~50 um), pH 6 (Control) aA single dose will be administer on Day 5; QD — Once daily, BID — Twice daily Dosing Frequency Sample Time Test Article Formulation and Points on:11 Compound-l; 0.6% Tris + 2.0% Glycerol + 0.2% 1 hr post dose HEC + 0.08% HPMC; on Day 5 1.0 uM Vit K3; mgI’mL eye/ dose 3 pm particles, pH 6 Compound-l; 0.6% Tris + 2.0% Glycerol + 0.3% 1 hr post dose HEC + 0.08% HPMC; 1.0 uM Vit K3; mgI’mL eye/ dose on Day 5 m oarticles, 0H 7 Compound-l; 0.6% Tris + 2.0% Glycerol + 0.2% 4.3 30 pL/ QD for4 1 hr post dose 16 P 3 08% HPMC; 1 0 uM Vit K3- mgimL eye/ dose Days on Day 5 pm particles, pH 7 3A single dose will be administer on Day 5; Q0 — Once daily Table 18-2 Group# 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 strength 0.3 4* 4* 4* 4* 4* 4* 4* 4* 4* 4* Salt Salt TRIS TRIS TRIS TRIS TRIS TRIS HPMC HPMC Stabilizer 0.08% . 0.60% 0.60% 0.60% 0.60% 0.60% % Glycerol % HEC Add’l HPMC Tylox Tylox Tylox Tylox HPMC ent 0.08% 0.04% 0.04% 0.04% 0.04% VitK3 (pM) FrequencyB pensio Ocular sampling occurred one hour following the ?rst daily dose on Day 5 for all groups. Whole blood samples using KZEDTA as an anticoagulant were collected from all animals on Day 5, 1 hour post dose. The whole blood was placed on ice until samples were spun in a centrifuge to separate the plasma. Aqueous humor, conjunctiva, cornea, central retina, peripheral retina, central choroid, and eral choroid samples were ted following necropsy. s were then frozen at —70°C or lower. For all Groups l—l6, plasma, central retina, l choroid, and cornea samples were assayed for PAN—90806.
Ocular tissue concentrations of Formula II or Compound—I assessed on Day 5 were in descending order: highest in the cornea >> central choroid > central retina. Corneal concentrations of Formula II or Compound—I ranged from 3 to 34 "M with the highest concentrations being observed in Group 9 and the lowest in Group 1. Central choroid concentrations ranged from <0.056 to 0.24 "M with the t concentrations seen in Groups 6 and 14 and the lowest in Group 1. Central retina concentrations ranged from 0.02 to 0.14 uM; Group 6 had the highest central retina concentrations and Group 1 had the lowest central retina concentrations. Plasma concentrations of Formula II or Compound—I were very low being approximately 5 to 10—fold and 100—fold lower than those in the central retina and central choroid, respectively. See Table 18—3.
Table 18-3 Central Plasma Central . Cornea Group PM) Retina (PM) ChoerIOId (PM) 2 0.00267 7.06 3 0.00450 15.9 4 0.00327 21.3 0.00391 23.8 11 0.00465 0.0796 0.154 12.8 12 0.00366 0.0490 0.137 7.05 13 0.00524 0.0677 0.234 7.82 14 0.00499 0.0992 0.241 20.7 0.00325 I 0.0532 0.200 18.2 16 0.00523 ‘ 0.0744 0.208 14.7 Plasma LLOQ = 0.00188 pM Central Retina LLOQ = 0.00751 [AM Central Choroid LLOQ = 0.0563 pM Cornea LLOQ = 0.0188 pM Gross ocular examinations using the Draize scale for scoring ocular irritation were performed on all animals prior to the ?rst dose on each day during the study. All formulations were well tolerated. In addition, animals were observed for general health pre- study, during dosing, and at necropsy. All animals assigned to study were normal. Necropsy evaluations consisted of observations in on to those already noted on Draize Score sheets. For all formulations, dosing observations were assessed using the Draize ocular tion scoring system and are summarized below. No abnormal observations were noted.
EXAMPLE 20-4 pharmacokinetics offormulations ofthe application in Dutch Belted rabbits A non—GLP study was conducted to assess the ocular pharmacokinetics of a first active agent of the present application (e.g., a II or Compound—I), in suspension, when administered one or two times daily, as a topical instillation, to both eyes of Dutch Belted rabbits for four days. The study design (Table 19—1) ted of forty—seven (47) Dutch Belted rabbits, each receiving a 30 ML bilateral topical ocular dose. Various suspension formulations of the present ation were evaluated and compared t a extrin—based eye drops on (Control Group 1, Table 19—1). A comparison of the formulation variables for each suspension listed in Table 19-1 is provided in Table 19—2.
Formula II or Compound—I was used in the test suspensions as well as the Eye Drops solution. However, different batches of Compound—I were tested in certain suspensions (Groups 13, 14, and 15, Table 19-1).
Table 19-1 Dosing No. of . . Dose Test Article Formulation Conc. Frequency Sample Time Rabbits Volume and Points Durationa ll—OCompound-l; —30 pL/ QD for4 1hr post dose Oohthalmic Solution 0H6 Control o/mL e e/ dose da 5 on Da 5 Compound- I; 2 B HPMC;1hgnpgs: dsosey pm particles Compound-l; 1 hr post dose HPMC, . on Day 5 3 m carticles Compound-I; 1 hr post dose on Day 5 50 m oarticles Control nd-l; 4 30pL/ QD for4 1 hr post dose HPMC; mg/mL eye/ dose days on Day 5 m oarticles Control* Compound-l; 4 30 pL / BID for4 1 hr post dose HPMC; mg/mL eye/ dose days on Day 5 50 m oarticles Compound-l; 1 hr post dose HPMC; mg/mL eye/ dose on Day 5 50 pm particles Compound-l; 1 hr post dose HPMC; mg/mL eye/ dose on Day 5 50 m carticles Compound-l; 2 30 pL / QDfor4 1 hr post dose HPMC; eye/ dose days on Day 5 50 m carticles Compound-l; 2 30 pL / BlDfor4 1 hr post dose HPMC; m9 mL1’ eye/ dose days on Day 5 50 m carticles Compound-l; 6 30 pL / QDfor4 1 hr post dose HPMC; mgimL eye/ dose days on Day 5 50 m oarticles Compound-l; hr post dose HPMC; on Day 5 50 pm particles Compound-l; hr post dose HPMC; on Day 5 50 m oarticles Compound-l; 4 30 pL / QD for 4 1 hr post dose HPMC; mg/mL eye/ dose days on Day 5 50 m les Compound-l; 4 30pL/ QD for 4 1 hr post dose 3 HPMC; mg/mL eye/ dose days on Day 5 50 m oarticles aA single dose will be administer on Day 5 *Sampled analyzed for Vitamin K3 QD — Once daily, BID — Twice daily Table 19-2 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 3 4 2 4 3 3 3 3 3 3 3 3 3 3 3 strength 4* m . lmL IIIIIIII"4* Approx.
PSD III-lll-NativeNativeNative(=50) (=50) (=50) (=50) (=50) (=50) (=50) (=50 (=50 (=50 (=50) (=50) Stabilize r 0.60 GI cerol 0.2 0.2 Add’l HPMC HPMC HPMC c HEM HEM HEM HEM HEM HEM HEM HCP:M HCP:M HEM Excip. 0.08% 008% 008% 0.08 ' ' 0.08%0.08%0.08%0.08%0.08%0.08%0.08%0.08%0.08%0.08% Vitamin 1 1 1 1 1 1 1 1 1 1 1 K3 (PM) pH 6 6 6 6 6 6 6 6 6 6 6 Dose Suspen Suspen- Freq_ Con .
-S|0n sion -tro| con-trol control = 0.43%'In salt form Ocular sampling occurred one hour following the ?rst daily dose on Day 5 for all groups. Whole blood samples using KzEDTA as an anticoagulant were collected from all animals on Day 5, 1 hour post dose. The whole blood was placed on ice until samples were spun in a centrifuge to separate the plasma. Aqueous humor, conjunctiva, cornea, central retina, peripheral retina, central choroid, and peripheral choroid s were collected following necropsy. Samples were then frozen at —70°C or lower. For all Groups l—15, plasma, central retina, central choroid, and cornea samples were assayed. For Group 5 only, samples were analyzed for Vitamin K3.
Ocular tissue concentrations of Formula II or Compound—l assessed on Day 5 1 hour after the ?rst dose were in descending order: highest in the cornea >> l choroid > central retina. Corneal concentrations of a II or Compound-I ranged from 1 to 25 uM with the highest concentrations being observed in Group 12 and the lowest in Group 7. l choroid concentrations ranged from 0.06 to 0.2 "M with the t concentrations seen in Groups 12 and 14 and the lowest in Group 7. Central retina concentrations ranged from 0.03 to 0.1 uM; Group 12 had the highest central retina concentrations and Group 7 had the lowest central retina concentrations. Plasma concentrations of Formula II or Compound-I were very low being approximately 10—fold and ld lower than those in the central retina and central choroid, tively. See Table 19—3.
Table 19-3 Central Central Plasma Cornea Retina Choroid IUM) (PM) 0.00428 0.0551 0.129 14.3 -0.00464 00506 0135 7.48 0.00704 0.0928 0.159 10.2 4 0.00598 0.0735 0.128 3.93 0.00626 0.0442 0.129 4.47 0.00415 0.00218 0.00211 0.00405 -0.00376 0.0507 0.116 5.00 0.00772 0.0915 0.148 7.07 -000861 0.114 0.199 24.7 0.00709 0.0766 0.151 8.55 14 8 0.0384 0.203 3.88 0.00469 0.0630 0.196 4.23 Plasma LLOQ = 0.00188 pM Central Retina LLOQ = 0.00751 pM Central d LLOQ = 0.0563 pM Cornea LLOQ = 0.0376 pM An ical method for the analysis of Vitamin K3 was ined in , central retina, central choroid, and cornea samples from Group 5 animals. A ation curve prepared in control us humor was used to determine the concentration of Vitamin K3. Internal standard (IS) responses were inconsistent for the central choroid and plasma samples. Thus, positive plasma results were likely due to the reduced IS response. The Vitamin K3 response in plasma was similar to background levels. Central choroid samples had a positive Vitamin K3 response, but the reduced IS response made quantitative results suspect. Central retina and cornea samples had a normal IS response and no quantitative results for Vitamin K3. See Table 19—4.
Table 19-4 Central Central Plasma Cornea Retina Choroid (PM) (PM) M M Plasma LLOQ = 0.0145 pM Central Retina LLOQ = 0.581 [M Central Choroid LLOQ = 2.18 [M Cornea LLOQ = 0.145 pM Gross ocular examinations using the Draize scale for scoring ocular tion were performed on all animals prior to the ?rst dose on each day during the study. In general, formulations were well tolerated, with the ion of two animals in Group 6 on Day 4 of dosing, where conjunctival chemosis was noted, one animal in Group 8 on Day 3, where conjunctival rge was noted, one animal in Group 10 on Day 5 of dosing, where conjunctival chemosis and discharge were noted, one animal in Group 13, and one animal in Group 14 on Day 5 of dosing, where conjuctival chemosis was noted. In addition, animals were observed for general health pre—study, during dosing, and at necropsy. All animals assigned to study were . Necropsy evaluations consisted of ations in addition to those already noted on Draize Score sheets. For all formulations, dosing observations were assessed using the Draize ocular irritation scoring system. The following clinical observations were noted throughout the study: one animal in Group 8 exhibited rge in the left eye during the dose on Day 3. Animals in Group 12 exhibited chemosis in both eyes at the time of necropsy.
EXAMPLE 21 Preparation ofparticles comprising Formula II The suspension formulations comprising particles of Formula II were prepared by roller milling. Large particles were produced using larger milling media (e.g., 3 mm in diameter) for a short period of time at low roller speed (Figure 9B). Nanoparticles were produced using smaller media (e.g., 0.8 mm in diameter) for a longer period of time at a high roller speed (Figure 9A). The two trations of Formula II (i.e., 0.4% and 0.2%) were diluted from a 5% API stock with a glycerol solution to obtain the proper tonicity. Best clean precautions included autoclaving of product contact parts, tion of excipient solutions and transfers using prepackaged e supplies. In addition, all transfers were within a laminar flow hood in a Clean Room area. onal examples of particles comprising Formula II or Compound-I is shown in Tables 21 and 22.
Table 21 II$33251;iEfff5¥¥€3§ai??i?ffZ?ZZZZZZZZZZZZZZZZZZZZZZZZZZZZZZZZZZZZZZZZrrrréii:ii:iiiiiiI:ZIiI:iI:ZIiI:Iiiféé— 0°8/HPMC35QG?sphgzs/HEC10M30m _________ 008% HPMC’éifzgglifirgl?gé% HEC’ I'OUM 6mg/mL Freebase Milled 30um ‘ , 06% TEEg???i?g?féG51??? 02% 6mg/mL HC1 Salt Milled 30um 4.3mg/mL HC1 Salt M1lled 0i6?yidi(:p2(iii‘)/i).gdigiiiiargiivlzgigf 284% 30um """""" """""""""dLei/ihis;i058'oiyéérbi,6.i%ii?"o.b.64% """""""" """" "mg/m HC1SaltM1lled 30m Tyloxapol, 1.0uM Vitamin K3, pH 6.0 0.6% Tris, 20% Glycerol, 0.3% HEC, 0.04% mL HC1 Salt Milled 30um Tyloxapol, l.0uM Vitamin K3, pH 7.0 4.3mg/rnL HC1 Salt Milled 0‘6?;5Epi??g??g?i?i?i 3.34% 30um """aa‘et"f"""lll;l"""""""""0 """""""" "mg/mt 6/;PMg?/0ththgnoi/3?c6g08/ all;"""" 4.3mg/mL HC1 Salt Milled "mg?g???y?g??/g$13630" 30um mL HC1 Salt Mllled 0'6%I{Tff?é%fkhy§iizgg?,I?lcég‘om 30um 4.3mg/mL HC1 Salt Milled 06%HTlif/Ig?t?fhhygign?lggigcigog% 30um 4.3mg/mL HC1 Salt Milled 0'6%HTl§LS/f&%$y€?:ggfgg,Iicig'om 30urn Table 22 Particles under . . . .
Mllhng' ' PSD Partlcle 8126 Distribution.
API Form Excipient Stora_e speed med1a51ze Median (nrn) Mean (nm) D90 (nrn) Freebase Pluronic F-127 ~4700 ~5600 ~9000 Pluronic F-127 ~1000 ~2800 Tween 80 low 3mm ~8400 ~11000 Freebase Tween 80 low 3 mm ~3900 ~4100 ~6800 Freebase Tween 80 low 3 mm ~3700 ~4000 ~7000 se Tween 80 1CW 3 mm ~3500 ~4000 ~7600 Pluronic F-127 low 3 mm ~6200 ~8100 ~12000 Freebase Pluronic F-l27 ~2500 ~3100 ~5800 6days, 40 °C -—-_—-— HCl salt HPMC 3 mm ~l70 ~450 ~1200 21 days, RT Freebase Tween 80 0.8 mm ~l60 ~370 ~1000 28 days, RT Freebase Pluronic F-l27 ~3400 ~3700 ~6600 EXAMPLE 22 Preparation ofCompound-I/Tris Formulation ] A non—milled suspension of the Compound-I was prepared. The formulation consisted of: 3% Compound—I; 0.6% Tris—HCl pH 6; 2% Glycerol. Compound—I was added to the solution ly and mixed without g and characterized (Figure 9C).
EXAMPLE 23 Particles comprising Formula II and vitamin K3 Addition of Vitamin K3 (menadione) to the suspension formulation may have a bene?cial in-vivo effect. Menadione was added to the formulation as a milled suspension.
Menadione was initially milled exactly as the Formula II was milled; a 5% suspension with Pluronic F—127 as the stabilizer. This sion was then added to Formula II before milling (large and nanoparticle formulations). The milled 5% sions ining menadione suspension) were then diluted to 0.4% of Formula II (in glycerol, as previously described).
] Menadione did not appear to affect any physical teristics of the milled particles of Formula 11 e 9D). However, at the highest concentration of menadione added (10 HM), it was detected particles of vitamin K3 that were increasing in size.
Additionally, there was a small population of very large particles outside the range of the free base, which was presumably the vitamin K3. It was concluded that the vitamin K3 could not be neously distributed as well as the other particles in the suspension, as they were growing at a faster rate than the Formula II.
EXAMPLE 24 Preparation ofparticles comprising one Formulation of menadione/Formula 11 may form larger crystals over time. An optimized formulation was required to be able to add the one as a suspension.
Formulations containing 5% menadione were screened using a roller mill in the same way as when screening the Formula II formulations. An HPMC formulation produced a ?ne, homogeneous suspension that demonstrated short term stability at 40 0C (Figure 10).
EXAMPLE 25 Preparation ofparticles comprising Formula II The HPMC formulation for menadione was used with Formula II to provide a formulation in which the one could be added as a suspension. It was found that an X- large (30 — 40 micron) particle size using HPMC could be milled in a shorter time than when using Pluronic F—127 as the stabilizer (Figures 11A and 11B). The suspension could then be diluted with a menadione/HPMC suspension.
EXAMPLE 25 Stability ofmenadione in formulations ofthepresent application ] The menadione concentration was 20 "M in the stability study. The menadione was formulated as follows: 0.43 % w/V Compound—I (unmilled, 50 um), 0.6% Tris/HCl, pH 6, 2% Glycerol, 0.2% HEC, 0.08% HPMC and 20 pM menadione. From this formulation, ten test ations were prepared: 1. minus glycerin (no N2) 2. control formulation;(no N2) \OOO\]O\UIJ>UJ .. control formulation; N2 sparged+ 0.3% Na thiosulfate hydrate), N2 sparged*. + 0.1% Na2 EDTA, N2 sparged. + 0.5% TPGS Vitamin E, N2 d. + 0.05% propyl gallate, N2 sparged. + 0.02% BHT, N2 sparged. -- menadione/HPb—cyclodextrin complex, N2 sparged . -- anti-oxidant complex (#5 — #9), N2 sparged 7 mL of each formulation was added to an amber 40 ml Vial (under amber lighting). Filtered nitrogen gas was slowly bubbled into the bottom of the Vial until the generated foam began to exit the top of the vial, which was then quickly capped.
Menadione/HPb—Cyclodextrin complex (#9) was prepared by mixing approximately equimolar concentrated ons of cyclodextrin and menadione before addition (approximately 2 hours) to the formulation.
EXAMPLE 26 Methodsfor roller g and characterization ofthe particles ofthepresent application Roller Mill The horizontal roller mill (US Stoneware, model 755) consists of four, motor driven 12" rubber rollers contained within a metal housing. Individual bottles placed between the rollers will rotate at an rpm determined by the speed of the rollers and the er of the bottle. Drug slurry consisting of API, stabilizers, water and milling media was added to the bottle and the cap tightly sealed before placing between the rollers. The media used was an ely dense Yttria Zirconium bead that varies in diameter from 800 microns to 3000 microns. After milling, the dispersion was ted from the media by transferring the contents to a centrifuge tube insert ?tted with a screen mesh. The small insert was placed into a fuge tube. The centrifuge was then run at approximately 300 x G for approximately 5 minutes. The dispersion collected below the mesh (which ed the media) into the tube.
Optical Microscopy (Oil/I) Optical microscopy photomicrographs of nanoparticles were taken using an Olympus BX51 system equipped with an oil immersion 100x objective (1000x magni?cation). A calibration bar (from lum to 100 um) was set as a ator on each photomicrograph. (The ation bar effectively serves to size larger particles at lower magni?cations, as well.) Particle Size Distribution (PSD) Particle size distribution was analyzed using laser diffraction light ring with a Horiba LA—950 V2. Generic assumptions were made in g conditions and the refractive index value. The distributions were volume based. Sample density was adjusted to a generic range of percent ission on the blue LED light source. A small sample cell (?lled with water) was used rather than the ?ow through cell, to minimize sample quantity.
INCORPORATION BY REFERENCE The entire disclosure of each of the patent documents and scienti?c articles referred to herein is incorporated by reference for all purposes. In the present application the host document is identi?ed with suf?cient particularity and als that are relevant to the disclosure are construed based on the context of the reference. Citation of publications and patent documents is not intended as an admission that any is pertinent prior art, nor does it constitute any admission as to the contents or date of the same. The application having now been described by way of written description, those of skill in the art will recognize that the application can be practiced in a variety of embodiments and the ing description and examples are for purposes of ration and not limitation of the claims that follow.
EQUIVALENTS The application can be embodies in other speci?c forms without ing from the spirit or essential characteristics thereof. The foregoing embodiments are therefore to be considered in all respects rative rather than limiting on the application described herein.
Scope of the application is thus indicated by the appended claims rather than by the foregoing description, and all changes that come within the meaning and range of equivalency of the claims are intended to be embraced therein.

Claims (27)

DEFINITIONS OF THE SPECIFIC EMBODIMENTS OF THE INVENTION AS CLAIMED HEREIN
1. A topical, ocular, suspension formulation, comprising: a. a first active agent of Formula II: (II), or a pharmaceutically acceptable salt thereof; b. a second active agent, or a pharmaceutically acceptable salt thereof, wherein the second active agent is nicotinic acid, nicotinamide, or vitamin K, or a combination thereof; and c. one or more pharmaceutically acceptable excipients; wherein the first active agent or the pharmaceutically able salt f is present in about 0.1% to about 2.0% (w/v).
2. The formulation of claim 1, wherein the one or more pharmaceutically acceptable excipients are selected from y propyl methyl ose (HPMC), carboxylmethyl cellulose and salts thereof, and hydroxyl ethyl cellulose (HEC).
3. The formulation of claim 1, comprising: a. about 0.00001% to about 0.0001% (w/v) second active agent, n the second active agent is vitamin K3, or a pharmaceutically acceptable salt thereof; b. about 0.6% (w/v) tromethamine; c. about 2.0% (w/v) glycerin; d. about 0.2% (w/v) hydroxy ethyl cellulose (HEC); and e. about 0.08% (w/v) Hypromellose.
4. The formulation of any one of claims 1 to 3, comprising the compound of Formula II: (II).
5. The formulation of any one of claims 1 to 3, comprising Compound-I: (Compound-I).
6. The formulation of any one of claims 1 to 3, sing about 0.1% to about 1.0% (w/v) of the compound of a II or a pharmaceutically acceptable salt thereof.
7. The formulation of any one of claims 1 to 3, comprising about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, or about 1.0% (w/v) of the compound of Formula II or a pharmaceutically acceptable salt thereof.
8. The formulation of claim 7, comprising about 0.2% (w/v) of the compound of Formula II, or a ceutically acceptable salt thereof.
9. The formulation of claim 7, comprising about 1.0% (w/v) of the compound of Formula II, or a pharmaceutically acceptable salt f.
10. The formulation of any one of claims 1 to 9, comprising particles of the first active agent, or a pharmaceutically acceptable salt thereof, wherein the particles have a mean diameter of between 100 nm and 100 µm.
11. The formulation of claim 10, wherein the particles have a mean diameter of between 30 µm and 60 µm.
12. The formulation of claim 10, wherein the particles have a mean diameter of between 1 µm and 5 µm.
13. The formulation of claim 10, wherein the particles have a mean diameter of at most 150
14. The formulation of claim 10, wherein the particles have a mean diameter of about 3 µm, about 30 µm, about 35 µm, or about 50 µm.
15. The formulation of claim 1 or claim 2, wherein the second active agent is vitamin K.
16. The formulation of any one of claims 1 to 3, wherein the second active agent is present in an amount of less than 10 µM.
17. The formulation of any one of claims 1 to 3, wherein the second active agent is present in an amount of about 1 µM, about 2 µM, about 3 µM, about 4 µM, or about 5 µM.
18. The formulation of claim 17, wherein the second active agent is present in an amount of about 1 µM.
19. The formulation of any one of claims 1 to 3, further sing a stabilizer for the second active agent.
20. The formulation of any one of claims 1 to 3, further comprising one or more ents selected from a non-ionic liquid polymer and a hydrophilic nic surfactant.
21. The ation of claim 20, wherein the nic liquid polymer is of the alkyl aryl polyether alcohol type.
22. The formulation of claim 20, wherein the hydrophilic non-ionic surfactant is poloxamer.
23. The formulation of claim 1, further comprising one or more excipients selected from Polysorbate (Tween) 80, Poloxamer (Pluronic) F-127, ellose (hydroxy propyl methyl cellulose or HPMC), Povidone, and Tyloxapol, and a combination thereof.
24. The ation of claim 1, wherein the one or more pharmaceutically acceptable excipients are selected from one or more of hydroxyl propyl methyl cellulose (HPMC) and ylmethyl cellulose and salts thereof.
25. The formulation of claim 1, wherein the one or more ceutically acceptable excipients are selected from one or more of hydroxyl propyl methyl cellulose (HPMC) and hydroxyl ethyl cellulose (HEC).
26. The formulation of any one of claims 1 to 3, wherein the formulation has a pH of about 6.
27. The formulation of any one of claims 1 to 3, further comprising a pH modifier selected from hydrochloric acid and sodium hydroxide. mwamme wwwwwq mw am wh?waq aw 3 21w we ww?aa‘gf wraia m: maram uwm E?fwomwgw \\ @3me w or E aw c m E mmmwgng g m a V=u§® omwmm _ . .cmrmm m.mw?mwm mamgggw mam mmcawc Em aw m: m w ..; em mmmwagm? akw Q uwm?gme am . “ram mamwam . macaw a? SE my m we mammggwe E m a aw a a ;%.m a Q w. a 9a mgguwmAgivvmcnogamm mmom Emua?é £23Agiwwncgomgom wmmw>m Emnm?é m§gvmmm ”Eifécsog?om\ mwm mwov%g E25353 $953332 . 93822 m w x mg: + Egg, gm + E; 3 + 3 Q + Q .. g gd?fémona?mmmémcw E c3332:
NZ729142A 2014-09-17 2015-09-17 Ocular formulations for drug-delivery and protection of the anterior segment of the eye NZ729142B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
NZ767710A NZ767710A (en) 2014-09-17 2015-09-17 Ocular formulations for drug-delivery and protection of the anterior segment of the eye

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201462051794P 2014-09-17 2014-09-17
PCT/US2015/050818 WO2016044671A1 (en) 2014-09-17 2015-09-17 Ocular formulations for drug-delivery and protection of the anterior segment of the eye

Publications (2)

Publication Number Publication Date
NZ729142A NZ729142A (en) 2023-08-25
NZ729142B2 true NZ729142B2 (en) 2023-11-28

Family

ID=

Similar Documents

Publication Publication Date Title
AU2017210543B2 (en) Ocular formulations for drug-delivery to the posterior segment of the eye
AU2021200997B2 (en) Ocular formulations for drug-delivery and protection of the anterior segment of the eye
NZ729142B2 (en) Ocular formulations for drug-delivery and protection of the anterior segment of the eye
BR112017005523B1 (en) TOPIC OCULAR SUSPENSION FORMULATION AND USE THEREOF