WO1997030701A2 - Pharmaceutical compositions for the treatment of glaucoma - Google Patents

Pharmaceutical compositions for the treatment of glaucoma Download PDF

Info

Publication number
WO1997030701A2
WO1997030701A2 PCT/US1997/002709 US9702709W WO9730701A2 WO 1997030701 A2 WO1997030701 A2 WO 1997030701A2 US 9702709 W US9702709 W US 9702709W WO 9730701 A2 WO9730701 A2 WO 9730701A2
Authority
WO
WIPO (PCT)
Prior art keywords
eye
group
administration
latrunculin
glaucoma
Prior art date
Application number
PCT/US1997/002709
Other languages
French (fr)
Other versions
WO1997030701A3 (en
Inventor
Paul L. Kaufman
Benjamin Geiger
Original Assignee
Wisconsin Alumni Research Foundation
Yeda Research And Development Co. Ltd.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Wisconsin Alumni Research Foundation, Yeda Research And Development Co. Ltd. filed Critical Wisconsin Alumni Research Foundation
Priority to AU20535/97A priority Critical patent/AU2053597A/en
Priority to IL12573597A priority patent/IL125735A/en
Publication of WO1997030701A2 publication Critical patent/WO1997030701A2/en
Publication of WO1997030701A3 publication Critical patent/WO1997030701A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/365Lactones
    • 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
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • 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
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/407Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with other heterocyclic ring systems, e.g. ketorolac, physostigmine
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene or sparfloxacin
    • 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
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/551Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
    • 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

Definitions

  • the present invention relates to the treatment of ocular disorders of the eye and more particularly to the treatment of glaucoma. This invention was made with United States Government support awarded by the
  • Glaucoma is an ophthalmologic disorder responsible for visual impairment. It is the fourth most common cause of blindness and the second most common cause of visual loss in the United States, and the most common cause of irreversible visual loss among African- Americans. Generally speaking, the disease is characterized by a progressive neuropathy caused at least in part by deleterious effects resulting from increased intraocular pressure on the optic nerve. In normal individuals, intraocular pressures range from 12 to 20 mm Hg., averaging approximately 16 mm Hg. However, in individuals suffering from glaucoma, intraocular pressures generally rise above 25 to 30 mm Hg. and can sometimes reach 70 mm Hg.
  • glaucomas Several different types exist, each having different pathophysiologies and risk factors. In terms of classification, glaucomas may first be deemed to be either "primary” or “secondary.” Primary glaucomas result directly from anatomical and/or physiological disturbances in the flow of aqueous humor (i.e., intraocular fluid).
  • the major types of primary glaucomas include (i) open-angle glaucoma (also known as chronic or simple glaucoma), (ii) angle-closure glaucoma (also known as closed-angle or narrow-angle . glaucomas occur as a sequel to ocular injury (e.g. , trauma inflicted to the eye) or preexisting disease (e.g., an intraocular tumor or an enlarged cataract).
  • Glaucoma has been associated with both pharmacological and non-pharmacologica] factors.
  • Non-pharmacological factors include age, race, family history, diabetes, and blood pressure.
  • African Americans are four-to-five times as likely to develop open- angle glaucoma as are Caucasians.
  • open-angle glaucoma is much more prevalent in individuals over the age of 40, and especially in those over 60 years of age.
  • particular drugs have been associated with glaucoma.
  • prednisone e.g. , prednisone, dexamethasone, and hydrocortisone
  • glaucoma following both ophthalmic and systemic administration systemic administration, by increasing resistance to aqueous humor outflow through the trabecular meshwork via a mechanism somehow genetically linked to primary open angle glaucoma.
  • Dexamethasone has been associated with the most pronounced increase in intraocular pressure, and ophthalmic administration generally leads to greater increases than systemic administration.
  • Topically applied ophthalmic drugs which dilate the pupil may induce angle-closure glaucoma, while the anticholinergics can increase resistance to aqueous humor outflow in susceptible individuals even without causing angle-closure, apparently related to their cycloplegic (ciliary muscle/accommodation-paralyzing) action.
  • adrenergic agonists such as phenylephrine and epinephrine
  • anticholinergics such as atropine, scopolamine, homatropine.
  • cyclopentolate and tropicamide may induce angle-closure glaucoma, while the anticholinergics can increase resistance to aqueous humor outflow in susceptible individuals even without causing angle-closure, apparently related to their cycloplegic (ciliary muscle/accommodation-paralyzing) action.
  • Adrenergic e.g., central nervous system stimulants, appetite suppressants
  • anticholinergic e.g., bowel relaxants and tricyclic anti depressants
  • agents administered systemically may also induce angle closure glaucoma, via secondarily dilating the pupil.
  • the miotic agents i.e.. agents that causes the pupil to contract
  • indirect-acting e.g. , anti-cholinesterases, such as echothiophate and isoflurophate, which prevent the degradation of the endogenous cholinergic neurotransmitter acetylcholine, thereby permitting strong and continuous contraction of the ciliary muscle.
  • the adverse effects associated with the miotic agents include painful ciliary or accommodative spasm and consequent induced fluctuating myopia and blurred vision (in patients under the age of approximately 45 years), ciliary or conjunctival congestion, ocular pain, headache, and the miosis itself, which can cause a disabling reduction of vision in elderly patients with early cataract formation (the combination of the small pupil and the cloudy lens critically reduce the amount of light reaching the retina).
  • the long-acting anticholinesterase are associated with the most severe and prolonged adverse effects, which may include development of cataracts or retinal detachment.
  • mydriatic agents e.g., the sympathomimetic amines epinephrine and dipivefrin
  • mydriatic agents are also useful in the initial treatment of open-angle glaucoma.
  • the adverse ocular effects of these agents include ocular congestion or hyperemia, ocular pain and headache, and blurred vision.
  • epinephrine occasionally causes untoward systemic effects, such as palpitation, tachycardia, and hypertension.
  • the frequently used non-selective ⁇ ,- and ⁇ 2 -adrenergic blocking agents e.g. , timolol and levobunolol
  • ⁇ ,-selective e.g. , betaxolol
  • adrenergic blocking agents are also used in the treatment of open-angle glaucoma and are also associated with several untoward effects.
  • untreated glaucoma can result in severe consequences, including blindness.
  • new treatment methods and agents are needed and would be welcomed by those plagued by glaucoma who either cannot tolerate available treatment regimens or who are unwilling to undergo invasive surgical procedures.
  • compositions that can be used to treat glaucoma.
  • the present invention contemplates a method of enhancing aqueous humor outflow in the eye of a subject to treat glaucoma, comprising: a) providing a subject with glaucoma: and b) administering to the subject an ophthalmic preparation comprising an effective amount of a non-corneotoxic compound selected from the group consisting of serine-threonine kinase inhibitors, tyrosine kinase inhibitors, protein tyrosine phosphatase inhibitors, and an actin-
  • a non-corneotoxic compound selected from the group consisting of serine-threonine kinase inhibitors, tyrosine kinase inhibitors, protein tyrosine phosphatase inhibitors, and an actin-
  • the serine-threonine kinase inhibitor is selected from the group consisting of l-(5-isoquinolinyl-sulfonyl)-2-methylpiperazine [H-7]; l-(5-iodonaphthalene-l - sulfonyl)-l H-hexahydro-l ,4-diazepine [ML-7]; (8 ⁇ *,95*,1 15*)-(-)9-hydroxy-9- methoxycarbonyl-8-methyl-14- «-propoxy-2,3,9, 10-tetrahydro-8,l 1-epoxy, ⁇ H, H, 1 1H- 2,7b, 1 la-triazadibenzo[ ⁇ ,g]cycloocta[cfifeJ trinden-1 -one [KT-5926J; and staurospor
  • the tyrosine kinase inhibitor is selected from the group consisting of 3,5- diisopropyl-4-hydroxybenzylidene, N-benzyl-3,4-dihydroxybenzylidene thioacetamide, N-(3- phenylpropyl)-3,5-diisopropyl-4-hydroxy benzylidene acetamide, and N-(4-phenylpropyl)-3.4- diisopropyl-4-hydroxy benzylidene acetamide.
  • the protein tyrosine phosphatase inhibitor is selected from the group consisting of vanadate and phenylarsin oxide.
  • the method of administration is topical. In other embodiments, the method of administration is intracameral. In still other embodiments, the method of administration is intracanalicular.
  • the present invention also contemplates an ophthalmic composition, comprising an effective amount of a compound capable of enhancing aqueous humor outflow in the eye of a subject to treat glaucoma and a pharmaceutically acceptable carrier, the compound selected from the group consisting of serine-threonine kinase inhibitors, tyrosine kinase inhibitors, protein tyrosine phosphatase inhibitors, and an actin-disrupting compound selected from latrunculin A, latrunculin B and swinholide A.
  • the serine-threonine kinase inhibitor is selected from the group consisting of l-(5- isoquinolinyl-sulfonyl)-2-methylpiperazine; 1 -(5-iodonaphthalene- 1 -sulfonyl)- 1 H-Hexahydro- 1 ,4-diazepine; (8i?*,95'*,l lS*)-(-)9-hydroxy-9-methoxycarbonyl-8-methyl-14- «-propoxy- 2,3,9,10-tetrahydro-8,l l-epoxy, 1H,8H, HH-2,7b,l l a-triazadibenzo[ ⁇ ,g]cycloocta[c ⁇ fe] trinden-1-one; and staurosporine.
  • the tyrosine kinase inhibitor is selected from the group consisting of 3,5-diisopropyl-4-hydroxybenzylidene, N-benzyl-3,4- dihydroxybenzylidene thioacetamide, N-(3-phenylpropyl)-3,5-diisopropyl-4-hydroxy benzylidene acetamide, and N-(4-phenylpropyl)-3,4-diisopropyl-4-hydroxy benzylidene acetamide.
  • the protein tyrosine phosphatase inhibitor is selected from the group consisting of vanadate and phenylarsin oxide.
  • the present invention contemplates the use of a compound selected from the group consisting of serine-threonine kinase inhibitors, tyrosine kinase inhibitors, protein , - A, latrunculin B and swinholide A, for the preparation of an ophthalmic composition for enhancing aqueous humor outflow in the eye of a subject to treat glaucoma.
  • the serine-threonine kinase inhibitor is selected from the group consisting of l-(5-isoquinolinyl-sulfonyl)-2-methylpiperazine; l-(5-iodonaphthalene- l-sulfonyl)-lH-Hexahydro-l,4-diazepine; (8 ⁇ *,95*,l IS*)-(-)9-hydroxy-9-methoxycarbonyl-8- methyl- 14-r?-propoxy-2,3,9, 10-tetrahydro-8, 1 1 -epoxy, 1 H,8H, 1 1 H-2.7b, 1 1 a- triazadibenzo[c7,g]cycloocta[cfife] trinden-1-one; and staurosporine.
  • the tyrosine kinase inhibitor is selected from the group consisting of 3,5-diisopropyl-4- hydroxybenzylidene, N-benzyl-3,4-dihydroxybenzylidene thioacetamide, N-(3-phenylpropyl)-
  • the protein tyrosine phosphatase inhibitor is selected from the group consisting of vanadate and phenylarsin oxide.
  • the use of the compositions entails either topical, intracameral, or intracanalicular administration.
  • the present invention contemplates methods of enhancing aqueous humor outflow in the eye of a subject to treat glaucoma, comprising: a) providing a subject with glaucoma; and b) administering to the subject an ophthalmic preparation comprising an effective amount of a non-corneotoxic compound selected from the group consisting of angiotensin II, Al-adenosine agonists, acepromazine, promazine, aceclidine, lysophosphatidic acid, genistein; endothelin-, transforming growth factor, interleukin- 1 , platelet activating factor, and 3 ⁇ ,5 ⁇ -tetrahydrocortisol.
  • a non-corneotoxic compound selected from the group consisting of angiotensin II, Al-adenosine agonists, acepromazine, promazine, aceclidine, lysophosphatidic acid, genistein; endothelin-, transforming growth factor
  • the compounds are either administered topically, intracamerally, or intracanilicularly.
  • the present invention contemplates an ophthalmic composition, comprising an effective amount of a compound capable of enhancing aqueous humor outflow in the eye of a subject to treat glaucoma and a pharmaceutically acceptable carrier, the compound selected from the group consisting of angiotensin II, Al-adenosine agonists, acepromazine, promazine, aceclidine, lysophosphatidic acid, genistein; endothelin-, transforming growth factor, interleukin- 1 , platelet activating factor, and 3 ⁇ ,5 ⁇ -tetrahydrocortisol.
  • the present invention also contemplates the use of the aforementioned compounds for the preparation of an ophthalmic composition for enhancing aqueous humor outflow in the eye of a subject to treat glaucoma.
  • the present invention provides effective and non-invasive methods of treating glaucoma without causing untoward and unacceptable adverse effects, such as corneal edema.
  • untoward and unacceptable adverse effects such as corneal edema.
  • ophthalmic preparation refers to a composition containing a compound active so as to forestall the progression of glaucoma as well as other pharmaceutically acceptable ingredients.
  • the characteristics of the composition will depend on a number of factors, including the mode of administration.
  • the composition may be a solution, suspension or the like that can be administered topically as an eyedrop.
  • the composition may be in a form suitable for intracameral administration; for example, microinjection through the cornea into the anterior chamber so that the compound can reach the trabecular meshwork.
  • the drug could be administered intracanalicularly, i.e., by retrograde microinjection into the venous collector channels draining Schlemm's canal or into Schlemm's canal itself.
  • the ophthalmic preparation may contain diluents, adjuvants and excipients, among other things.
  • the term "effective amount” refers to that amount of an ophthalmic preparation that is required to successfully treat glaucoma.
  • the effective amount of an ophthalmic preparation may depend on a number of factors, including the age, race, and sex of the subject and the severity of the glaucoma and other factors responsible for biologic variability. Though the term effective amount is not limited to a particular mechanism of action for a specific compound, an effective amount may be that amount of a compound able to disrupt cell junctions in the trabecular meshwork of the eye to increase aqueous humor outflow.
  • subject includes humans as well as animals.
  • glaucoma refers to an ophthalmologic disorder responsible for visual impairment. The disease is characterized by a progressive neuropathy caused at least in part by deleterious effects resulting from intraocular pressure on the optic nerve.
  • the term glaucoma refers broadly to both primary glaucomas, which include open-angle, angle-closure, and congenital glaucomas, and secondary glaucomas, which occur as a sequel to ocular injury or preexisting disease. Though not limited to any particular type of glaucoma, it is anticipated that the pharmacological agents and compositions of the present invention will be most efficacious in the treatment of open-angle glaucoma.
  • aqueous humor outflow refers to the drainage of aqueous humor from the eye. As described in further detail below, aqueous humor leaves the eye by passive bulk flow via two pathways at the anterior chamber angle, the trabecular or conventional route and the , , . drainage via both pathways.
  • ser-threonine kinase inhibitors and "ser-thr inhibitors” refer to compounds known to be myosin light-chain kinase inhibitors. These compounds primarily inhibit myosin light-chain kinase, thus inhibiting actomyosin-driven contractility; upon long exposure (>30 minutes), they cause deterioration of the entire microfilament system. These changes affect predominantly cell-extracellular matrix adhesions.
  • the ser-thr inhibitors include, but are not limited to, H-7, KT-5926, ML-7, and staurosporine.
  • non-corneotoxic refers generally to the absence of medically-unacceptable side effects on the cornea.
  • One of the primary side effects to be avoided is corneal edema, which is manifested by abnormal fluid accumulation within the intercellular spaces of the cornea.
  • a main etiologies of corneal edema is impairment of the corneal endothelium due to the introduction of drugs and other chemical compounds. Specular microscopy can be used to evaluate drug effects on the corneal endothelium, and the absence of a statistical change in corneal endothelial cell counts is indicative of a non-corneotoxic compound.
  • the presence of corneal toxicity can be evaluated through ocular examination, which may include slit lamp biomicroscopy, gonioscopy, and corneal thickness measures.
  • Figure IA schematically depicts the major anatomical structures of the eye.
  • Figure IB schematically illustrates the basic anatomy of the primate anterior chamber angle.
  • the trabecular (conventional) route of aqueous humor flow is indicated by the uninterrupted lines, while the uveoscleral (unconventional) route of aqueous humor flow is indicated by the dashed lines.
  • Figure 2 depicts the chemical structures of some of the compounds contemplated for use with the present invention.
  • Figure 3 is a schematic drawing of the anterior chamber perfusion set-up used in conjunction with some of the experiments.
  • Figure 4 is a schematic diagram showing the attachment of a ciliary muscle strip to strain gauges in the muscle chamber apparatus.
  • the muscle chamber apparatus is used to measure contractile force simultaneously in the longitudinal and circular vectors.
  • Figure 5B is a bar graph illustrating the effect on facility of several concentrations of H-7 topically administered to living monkeys.
  • Figure 6A graphically illustrates the effect of topical H-7 on pupil size.
  • Figure 6B graphically illustrates the effect of topical H-7 on refraction.
  • Figures 7A-H graphically depict miotic response ( Figures 7A, 7C, 7E, and 7G) and accommodative response ( Figures 7B, 7D, 7F, and 7H) to intramuscular pilocarpine in either the presence or absence of H-7.
  • Figures 8A-C depict chart recorder traces obtained from a single in vitro ciliary muscle strip and were designed to determine the effect of H-7 on the ciliary muscle's contractile response to pilocarpine.
  • Figures 9A-D graphically depict the effect in living monkeys of latrunculin A given as an exchange infusion with total outflow facilitites measured by 2-level constant pressure perfusion before and after drug administration.
  • Fig. 9A 0.2 ⁇ M
  • Fig. 9B 0.5 ⁇ M
  • Fig. 9C 2.0 ⁇ M
  • Fig. 9D 5.0 ⁇ M
  • Figure 9E graphically illustrates the effect of latrunculin A in ciliary muscle- disinserted eyes.
  • Figure 10 graphically depicts the reversibility of the facility-effect of latrunculin A.
  • Figures 1 1A-C graphically depict the effect in living monkeys of staurosporine given as an exchange infusion with total outflow facilitites measured by 2-level constant pressure perfusion before and after drug administration (Fig. 11 A: 0.1 ⁇ M; Fig. 1 IB: 01.0 ⁇ M; Fig. 1 1C: 10.0 ⁇ M).
  • the present invention relates to the treatment of glaucoma. While the present invention does not depend on an understanding of the mechanism by which successful treatment is accomplished, it is believed that the compositions of the present invention cause transient, drug-induced dissociation of cell-cell junctions which facilitates fluid flow around the cells and across the monolayer ("pericelluiar route"), leading to reduction in intraocular pressure. Considerations And Aqueous Humor Dynamics; II) Structural Considerations of the Eye; III) Cell Culture, Animal Model, and Experimental Apparatus; IV) Evaluation of Compounds for Use in the Present Invention; V) Potential Therapeutic Chemical Compounds; and VI) Composition and Administration of Compounds. Each of these parts will be discussed in turn.
  • FIG. IA The major anatomical structures of the eye are shown in Figure IA.
  • the anterior portion of the sclera 1, the cornea 2 is modified to allow light rays to enter the eye.
  • the transparent, biconvex lens 6, one of the refractive media of the eye, is secured by the lens ligament (zonule) 7 to the ciliary body 8.
  • zonule lens ligament
  • the anterior chamber 12 of the eye contains a clear liquid material called the aqueous humor; the aqueous humor is also referred to as intraocular fluid.
  • aqueous humor is produced in the ciliary body/ciliary processes 8.
  • FIG. 1B schematically illustrates the basic anatomy of the primate anterior ocular segment and the normal pathways of aqueous humor flow.
  • Aqueous humor enters the posterior chamber 16 from the ciliary body/ciliary processes 8 as a consequence of hydrostatic and osmotic gradients between the posterior chamber and the ciliary process vasculature and stroma and active ionic transport across the ciliary epithelium.
  • the aqueous humor then flows around the lens 6 and through the pupil into the anterior chamber 12.
  • the aqueous humor then leaves the eye by passive bulk flow via two pathways at the anterior chamber angle: (i) the trabecular or conventional route (hereafter "the trabecular route"), or (ii) the , , .
  • aqueous humor travels through the trabecular meshwork 15, across the inner wall of Schlemm's canal 18 into its lumen, and then into collector channels 20, aqueous veins 21 , and the general venous circulation (not labelled) (the trabecular route is indicated by the uninterrupted lines in Figure IB).
  • aqueous humor travels across the iris root, uveal meshwork, and the anterior face of the ciliary muscle, through the connective tissue between the muscle bundles, and then out through the sclera (the uveoscleral route is indicated by the dashed lines in Figure IB).
  • Aqueous humor is continually being produced and drained, and it is the balance between these two processes that regulates the pressure of the intraocular fluid.
  • open- angle glaucoma results from decreased permeability of the aqueous humor through the trabecular meshwork 15, whereas angle-closure glaucoma results from shifting of the iris 9 forward so that the anterior chamber angle 10 is obstructed.
  • cytoskeleton a complex network of cytoplasmic filaments, known, collectively, as the cytoskeleton.
  • cytoskeletal filaments There are three major classes of cytoskeletal filaments, including actin microfilaments, intermediate filaments and microtubules. Among these, most relevant for cell adhesion are the microfilaments which interact with both cell-matrix and cell-cell junctions through a complex submembrane interlinking "plaque". Recent studies have established that this interaction with the cytoskeleton is essential for the formation of stable junctions. Thus, disruption of the actin network or its dissociation from the membrane-bound plaque perturbs cell adhesion.
  • actin filaments In forming this firm anchorage of actin filaments to the junctional membrane, several additional proteins participate, the perturbation of which might alter cell adhesion and which should thus be considered. These include both “anchor proteins” which are involved in the physical linkage of actin to the membrane, and myosin, an actin motor driven by ATP hydrolysis, which is responsible for muscle and non-muscle contractility.
  • myosin light chain kinase In its active form, myosin light chain kinase (MLCK) catalyzes the phosphorylation of a myosin light chain, permitting myosin to interact with actin; this interaction results in the movement of myosin along the actin microfilament leading to cell contraction.
  • the trabecular meshwork consists of interlacing connective tissue beams, each surrounded or "coated” by a single layer of endothelial-like cells.
  • the beams form a latticework, comprising several layers.
  • the spaces between the beams are filled with extracellular matrix, consisting of glycosaminoglycans, proteoglycans, and other macromolecules.
  • the outer-most portion of the meshwork contains no beams, but rather several layers of endothelial cells imbedded in the "sea" of extracellular matrix.
  • the canal of Schlemm which is connected to the general venous circulation.
  • the pressure in Schlemm's canal is ⁇ 9 mm Hg.
  • the resistance of the trabecular meshwork normally is such that intraocular pressure is ⁇ 16 mm Hg, at which aqueous humor leaves the eye at the same rate at which it is produced (-2.5 ⁇ L/minute).
  • intraocular pressure is ⁇ 16 mm Hg, at which aqueous humor leaves the eye at the same rate at which it is produced (-2.5 ⁇ L/minute).
  • the exact pathways by which aqueous humor flows through the meshwork and across the wall and thus into the lumen of Schlemm's canal, and the exact sites of resistance within the meshwork, are still unclear.
  • the barriers constituted by the cells, the extracellular matrix, and the adhesions between them are major determinants of both the flow pathways and the resistance to flow.
  • Several classes of drugs theoretically could act directly on the trabecular meshwork and Schlemm's canal to alter outflow facility.
  • ciliary muscle relaxation may cause an undesired decrease in facility. Because ciliary muscle contraction and relaxation cause an acute increase or decrease in facility, respectively, one must be able to determine whether a drug-induced facility change is due to a direct action on the conventional outflow channels or an indirect action exerted via altered ciliary muscle tone.
  • cytochalasins The disruption of overall trabecular meshwork architecture and the consequent increase in outflow facility by cytochalasins, calcium chelators, and sulfhydryl reactive agents has been .
  • w o potential cellular and junctional proteins whose alteration might rearrange meshwork geometry to therapeutic advantage. More importantly, these agents exert unwanted effects on other anterior segment structures whose integrity depends on those particular proteins.
  • cytochalasins researchers have found that they interfere with actin filaments and enhance fluid outflow from the living monkey eye; this finding was first published in the mid-1970's. [See, e.g. , P.L. Kaufman and E.H.
  • Cytochalasin B Reversibly Increases Outflow Facility in the Eye of the Cynomolgus Monkey," Invest. Ophth. & Visual Science 16(l):47-53 (1977)].
  • the cytochalasins are not presently being used in therapy for several reasons, most notably corneal intolerance upon acute administration.
  • ethacrynic acid has been tested in regards to increasing aqueous humor outflow in the eye.
  • U.S. Patent Nos. 4,757,089 and 5,306,731 to D.L. Epstein hereby incorporated by reference, describes a method of increasing aqueous humor outflow by administering ethacrynic acid, or an analog thereof, containing a chemical group capable of reacting with sulfhydryl groups in the trabecular meshwork.
  • Recent studies have shown that topically administered ethacrynic acid resulted in progressive corneal toxicity that was clinically unacceptable. [M.A. Croft and P.L.
  • the monkey is a suitable experimental animal because the biological apparati associated with aqueous humor formation and drainage closely resemble those of the human. Also, the monkey has a large accommodative amplitude, allowing technically simple, non- , perturbed and non-perturbed conditions. Indeed, a previous report of the Glaucoma Panel of the National Advisory Eye Council noted the preferability of primates as opposed to sub- primate mammals for studies such as those described in this application. [U.S. DHHS. Vision Research - A National Plan: 1983-87. 1987 Evaluation and Update. Report of the National
  • a number of different genera and species of monkeys have provided useful information regarding ocular structure and function, including the owl monkey (Aotus trivirgatus), the vervet monkey (African green; Cercopithecus ethiops), and several species of macaques [cynomolgus - M. fasicularis (formerly M. irus); rhesus - M. mulatto; pigtail - M. nemestrina; and stumptail - M. arctoides].
  • the cynomolgus has been used most frequently, and a large body of information has been accumulated on the functional and structural characteristics and responses of its accommodation and aqueous humor formation and drainage apparati.
  • the apparatus for determining aqueous humor outflow facility in the living monkey eye employs the two-level constant pressure perfusion technique.
  • the basic methodology is well-described in the literature and is standard in the field. [See, e.g., M. J. Menage et al., " ⁇ -Aminocaproic Acid Does Not Inhibit Outflow Resistance Washout in Monkeys," Invest. Ophthalmol Vis. Sci. 36(9): 1745- 1749 (1995)].
  • tissue culture experiments on monkey ciliary muscle have also been performed.
  • the inventors of the present invention developed a custom apparatus (described in the Experimental section) in which the contractile properties of isolated monkey ciliary muscle strips, freed from their posterior attachments to the choroid/sclera and anterior attachments to the scleral spur/trabecular meshwork/cornea, can be studied in both the longitudinal contractile vector (putatively more outflow specific) and the circular contractile vector (putatively more accommodation specific) simultaneously.
  • the apparatus represents an advance over other systems which permit measurement in only one, often undefined, vector at a time.
  • the screening (evaluation) procedures described hereafter can be used to test compounds that might exhibit beneficial effects by altering the cytoskeleton or the cell-cell or cell-extracellular matrix adherens junctions. Specifically, it can be used to examine n men n egr y or membrane anchorage or inhibition of contractility.
  • endothelial cells cultured on glass coverslips, are exposed to different concentrations of each compound for different periods of time. Thereafter, they are fixed and either examined under the microscope directly, or immunofluorescently-labeled for actin (since actin is a central element in both cellular- cellular and cellular-extracellular matrix adherens-type junctions), vinculin (a central element in both types of junctions), and cadherins and catenins (critical components of cell-cell contact whose presence and ability to interact are essential for junction integrity), as well as other junctional molecules. Those compounds which affect junction integrity or the integrity of the microfilament system are selected for further analysis. The effective concentrations in culture also serve as guidelines for the live animal experiments
  • Table 1 sets forth compounds contemplated for use with the methods and compositions of the present invention.
  • Actin-disrupting latrunculin B also affect junction integrity and consequently increase drugs swinholide A outflow facility
  • Tyrphostms inhibitors to selectively inhibit tyrosine phosphorylation of cell-cell and cell-ECM adhesions Tyrphostms may either stabilize or de ⁇ stabilize cell adhesions
  • the present invention is not limited to the potential classes of compounds set forth in Table 1. Rather, the present invention broadlv contemplates screening for compounds that may beneficially alter cell-cell or cell-extracellular matrix adherens junctions.
  • agents that may be screened include , - , , , acid, genistein; endothelin-, transforming growth factor, interleukin- 1 , platelet activating factor, and 3 ⁇ ,5 ⁇ -tetrahydrocortisol.
  • the screening procedure set forth in Table 2A can be used initially to evaluate compounds such as those presented in Table 1 that may be effective in the treatment of glaucoma.
  • Members of the serine-threonine kinase inhibitor class of compounds are particularly effective at enhancing aqueous humor outflow in the eye, and this screen is especially appropriate for compounds in that class.
  • Step III meshwork in living monkeys with disinserted ciliary muscle.
  • myosin light-chain kinase inhibitors were studied in the cynomolgus monkey eye in vivo. Data from one compound (H-7, described below) indicates that while these compounds primarily inhibit myosin light-chain kinase, thus inhibiting actomyosin-driven contractility, other mechanisms of action may also be involved.
  • H-7 One specific compound tested, the myosin light-chain kinase and cyclic nucleotide-dependent protein kinase inhibitor l-(5-isoquinolinyl-sulfonyl)-2-methylpiperazine [H-7] induced an ipsilateral increase in total outflow facility in normal and ciliary muscle- disinserted monkey eyes when perfused through the anterior chamber.
  • H-7 inhibits ipsilateral pilocarpine-induced miosis at intracameral and intravitreal doses far lower than required for inhibition of accommodation.
  • H-7's mechanism of action may involve loosening or weakening of cell-cell or cell- extracellular matrix adherens junctions of trabecular meshwork cells, or altered cellular contractility, via an effect on actin filaments or acto-myosin interactions. Further evidence strongly suggests that H-7 acts primarily to inhibit myosin light-chain kinase and thus inhibits actomyosin-driven contractility, eventually leading to perturbation of microfilaments.
  • Staurosporine Staurosporine is a microbial alkaloid produced by a Streptomyces species.
  • the compound is known to possess several biological activities, including antifungal and hypotensive effects, inhibition of platelet aggregation, and promotion of cell differentiation. [T. Tamaoki et al, Biochem, Biophys. Res. Commun. 135(2):397-402 ( 1986);
  • Staurosporine is a modulator of ser-thr kinases that probably, like H-7, acts through inhibition of myosin light-chain kinase. Its effect on the trabecular meshwork has not been definitively elucidated, though staurosporine might decrease contractility, leading to a total disruption of the microfilament system.
  • ML-7 [l-(5-iodonaphthalene-l -sulfonyl)-lH-Hexahydro-1.4-diazepine] is a myosin light chain kinase inhibitor commercially available from Sigma. Though not as , effectively inhibit mouse lung carcinoma 3LL cell attachment to the fibronectin substratum. [M. Isemura et al , "Myosin light chain kinase inhibitors ML-7 and ML-9 inhibit mouse lung carcinoma cell attachment to the fibronectin substratum," Cell Bio. Int. Rep. 15(10):965-972 (1991 )].
  • KT5926 [(8R*,9S*, ⁇ 15*)-(-)9-hydroxy-9-methoxycarbonyl-8-methyl- 14-,?- propoxy-2,3,9,10-tetrahydro-8, 1 1 -epoxy, 1H.8H, HH-2,7b,l la- triazadibenzo[ ⁇ ,g]cycloocta[cfife] trinden-1-one; Sigma] is a potent and selective inhibitor of myosin light-chain- kinase. The compound has been shown to inhibit both Ca 2 7calmodulin- dependent and -independent smooth muscle myosin light chain kinases as well. Though
  • KT5926 is deemed to be a selective inhibitor of myosin light-chain kinase, it, too inhibits other protein kinases, e.g., cAMP-dependent protein kinase, but with relatively high K, values.
  • the chemical structure of KT5926 is depicted in Figure 2. [S. Nakanishi et al. "KT5926. a potent and selective inhibitor of myosin light chain kinase," Mol Pharmacol 37:482-488 (1990)].
  • Actin assembly proceeds in all cells within the eye. For example, actin assembly occurs at the ciliary muscle, where it entails formation of ⁇ -smooth muscle actin which interacts with myosin during muscle fiber contraction. In addition, actin assembly most likely occurs in all cells at the trabecular meshwork, where it entails polymerization of G-actin monomers into F-actin filaments.
  • Latrunculin A is a macrolide isolated from the red sea sponge Latrunculia magni ⁇ ca that acts by destabilizing actin filaments and interfering with their formation. [Y. Kashman et al. , "Latrunculin, a new 2-thiazolidinione macrolide from the marine sponge Latrunculia magnifica,” Tetrahedron Lett. 21 :3629-3632 (1980)]. Latrunculin-
  • A is the first marine macrolide found to contain 16-membered rings; its structure is set forth in Figure 2.
  • Latrunculin A binds monomeric G-actin in a 1 : 1 molar ratio and thus prevents the nucleation and elongation of actin filaments.
  • researchers have compared the short- and long- term effects of latrunculin A on cell shape and actin organization to those of cytochalasin D.
  • latrunculin A s efficacy as a therapeutic agent for the treatment of glaucoma
  • the inventors of the present invention found that, when perfused through the anterior chamber of the living cynomolgus monkey eye, latrunculin A increases ipsilateral outflow facility (data presented in the Experimental section).
  • the mechanism by which latrunculin A affects outflow facility may involve inhibition of actin assembly in the trabecular meshwork cells, thus causing the cells to loosen their attachments to one another and/or to their extracellular matrix, and to change their shape, thereby enhancing outflow facility.
  • latrunculin A primarily affects cell-cell junctions, in contrast to H-7 which mainly causes deterioration of cell-extracellular matrix adhesions.
  • Swinholide A is isolated from the marine sponge Theonella swinhoei. and has been described as a 44-membered dimeric dilactone macrolide with a 2-fold axis of symmetry. The compound exhibits antifungal activity and has been shown to be highly cytotoxic to several cancer cell lines. Its chemical structure is set forth in Figure 2. Swinholide A reportedly disrupts the actin cytoskeleton of cells grown in culture, sequesters actin dimers in vitro in both polymerizing and non-polymerizing buffers, and rapidly severs F-actin in vitro with high cooperativity. [M.R.
  • the third class of chemical entities contemplated, based on work in in vitro cell culture systems, for facilitating aqueous humor outflow are the tyrosine kinase inhibitors or stimulators.
  • the tyrphostins represent one family of tyrosine kinase inhibitors. These compounds, synthetic tyrosine kinase inhibitors with a highly diversified specificity, effect growth factor . phosphorylation of junctional (cell-cell and/or cell-extracellular matrix) proteins. Tyrphostins may either stabilize or de-stabilize cell adhesions. [See T. Volberg et al , "The effect of tyrosin protein phosphorylation on the assembly of adherens-type junctions," EMBO J 1 1 :1733-1742 (1992); T.
  • triphostins include, but are not limited to, 3,5-diisopropyl-4-hydroxybenzylidene, N-benzyl-3,4-dihydroxybenzylidene thioacetamide. N-(3-phenylpropyl)-3,5-diisopropyl-4-hydroxy benzylidene acetamide, and N- (4-phenylpropyl)-3,4-diisopropyl-4-hydroxy benzylidene acetamide.
  • the present invention also contemplates the use of protein tyrosine phosphatase inhibitors for the treatment of glaucoma.
  • protein tyrosine phosphatase inhibitors for the treatment of glaucoma.
  • adherens junctional proteins such as catenins, paxillin, tensin, and others, may exert dramatic effects on junction integrity.
  • compositions of agents that mainly disrupt cell junctions through an effect on microfilaments and/or membrane-microfilament binding in the ocular fluid drainage pathways to reduce intraocular pressure. It is not intended that the present invention be limited by the particular nature of the therapeutic preparation.
  • such compositions can be provided together with physiologically tolerable liquid (e.g. , saline), gel or solid carriers or vehicles, diluents, adjuvants and excipients, such as pharmaceutical grades of mannitol. lactose, starch, , u sacc ar n, c u ose, magnes um car ona e, an e e. ese compositions typically contain l%-95% of active ingredient, preferably 2%-70%.
  • therapeutic preparations can be administered to mammals for veterinary use, such as with domestic animals, and clinical use in humans in a manner similar to other therapeutic agents.
  • dosage required for therapeutic efficacy will vary according to the type of use and mode of administration, as well as the particularized requirements of individual hosts.
  • compositions contemplated by the present invention may be mixed with diluents or excipients which are physiological tolerable and compatible. Suitable diluents and excipients are, for example, water, saline, dextrose, glycerol, or the like, and combinations thereof.
  • diluents and excipients are, for example, water, saline, dextrose, glycerol, or the like, and combinations thereof.
  • the compositions may contain minor amounts of auxiliary substances such as wetting or emulsifying agents, stabilizing or pH buffering agents or preservatives.
  • compositions of the present invention are generally dissolved or suspended in a carrier like physiological saline (e.g., 0.9%) or methyl cellulose
  • the preferred mode of administration for these compounds entails a solution or suspension that can be administered as an eyedrop.
  • infrequent intracameral administration could be feasible if done infrequently (e.g. , once-yearly) or for acute post-surgical situations.
  • the therapeutic compounds may be investigated for their efficacy via other routes of administration, including oral, intravenous, intramuscular, and injection into the collector channels of Schlemm's canal itself.
  • the compounds may be effective at a dosing interval greater than that of currently available agents. Less frequent administration may be required if extracellular matrix material takes some time to re- accumulate in the ocular fluid drainage pathway following drug-induced wash-out of extracellular material.
  • the following examples serve to illustrate certain preferred embodiments and aspects of the present invention and are not to be construed as limiting the scope thereof.
  • USP United States Pharmacopeia
  • DMSO dimethyl sulfoxide
  • DMEM Dulbecco's Modified Eagle's Medium
  • MES MES (2-[N-morpholino]ethanesulfonic acid); s.e.m. (standard error of the mean); df (degrees of freedom).
  • post-surgical analgesic medications were not employed for relatively minor surgical procedures (e.g., iridectomy, perfusions, tail cut-downs); the rationale was to allow recovery to be judged in the undrugged animal.
  • Post-surgically the animals were isolated in a recovery cage warmed with a heat lamp, given food and water ad libitum, and observed at least once daily by both a veterinarian or caretaker staff and one of the inventors or a project specialist staff.
  • post-surgical analgesic medication (butorphanol 0.2-0.3 mg/kg SQ BID) was administered and the animals received intramuscular antibiotics (procaine penicillin G + dihydrostreptomycin sulfate; Pfizer.
  • glucocorticosteroids methylprednisolone acetate; UpJohn, Kalamazoo, MI
  • Topical Eye drops were applied to the anesthetized monkeys as previously described.
  • Transcorneal A 30 gauge needle was removed from its hub. connected directly to narrow-bore polyethylene tubing, and threaded through the cornea for several millimeters using a needle driver. The beveled tip of the needle was then pushed into the anterior chamber and drug was administered. The volume of solution delivered did not exceed 20 ⁇ l. The needle was then withdrawn from the anterior chamber without any loss of aqueous humor.
  • Intravitreal A 27-30 gauge needle, prepared as above, was inserted directly into the vitreous through the pars plana at either the 10:00 or 2:00 positions. The total volume injected did not exceed 50 ⁇ l.
  • d) Systemic Drug was administered via IM or IV injection. Alternatively, a venous or arterial line of polyethylene tubing was placed in the monkey's tail following a small incision and isolation of the vessel. [See, e.g., B.T. Gabelt and P.L. Kaufman, "Prostaglandin F 2 ⁇ increases uveoscleral outflow in the cynomolgus monkey," Exp. Eye Res. 49:389-402 (1989)].
  • the cells were plated at half confluency on clean glass coverslips and cultured in complete medium for 2-4 days until they reached confluency.
  • the drug was then added to the culture medium for different periods of time, after which the cells were either fixed with a fixative appropriate for the specific antigen being examined or further incubated in a drug-free medium to examine recovery.
  • Fixed or fixed-permeabilized cells were then either fluorescently labelled for actin using a fluorescein- or rhodamine-conjugated phalloidin or immunofluorescently labelled for the different cytoskeletal or junctional proteins.
  • the apparatus for determining aqueous humor outflow facility in the living monkey eye employs the two-level constant pressure perfusion technique.
  • This technique and variations thereof, are well known in the art. [See E.H. Barany. "Simultaneous measurements of changing intraocular pressure and outflow facility in the vervet monkey by constant pressure infusion," Invest. Ophthalmol. 3(2):135-143 (1964); P.L. Kaufman and E.H.
  • Figure 3 schematically illustrates the present apparatus as used for one eye. Two identical set-ups are used when studying two eyes of an animal simultaneously, as in the data presented hereafter. However, only one computer (Apple Macintosh II with National
  • the major components of the apparatus include customized 26-gauge needles and polyethylene tubing which connect the anterior chamber of the eye to a continuously-weighed fluid reservoir (Harvard Precision Adjustable Screw Stand (modified with motor and microswitches to raise and lower the reservoir). Harvard Apparatus Co., Inc., South Natick), the change in weight with time providing a measurement of flow.
  • Other major components include a pressure transducer (Statham Universal Transducer P23XL; Ording Inc.,
  • the infusion system allows regulation and measurement of intraocular pressure and rate of fluid flow from the reservoir and infusion syringe into the eye and calculation of outflow facility from those measurements.
  • the flow and pressure data are calculated from the electronic signals provided by the pressure transducer and strain gauge, the signal processing and calculations being performed on-line in real-time by the amplifier and computer, according to the inventors' own software.
  • the recorder simply provides a back-up record of the flow and pressure data, from which the facility can be calculated off-line, should the computer fail.
  • the anterior chamber of each eye was cannulated transcorneally with one branched and one unbranched needle, and connected to the apparatus as described above.
  • the exchange needles were inserted with the branched needle superior to the single needle and separated by several mm, and the opposite ends of the needles were attached to perfusand-filled tubing through which drug, vehicle, and perfusion solutions could pass into the eye.
  • Anterior chamber exchange was accomplished by attaching the tubing of . detached from the reservoir of the perfusion apparatus and fixed at an intraocular pressure so that fluid could exit the eye without changing the intraocular pressure.
  • Baseline outflow facility measurements were taken for 35-45 minutes, after which the previously-clamped tubing from the unbranched needle was connected to syringes containing drug for one eye and vehicle for the opposite eye via customized cone-tipped needles, and undamped.
  • the syringes were placed in a Harvard Apparatus (Harvard pump model 944) variable-speed infusion pump.
  • One tubing arm of the branched needle was detached from the reservoir of the perfusion apparatus and fixed at an elevated position so that fluid could exit the eye without changing intraocular pressure.
  • the inflow tubing was disconnected and the pump turned on to inflow at a rate that allowed 2 ml of drug/vehicle solution to pass through the anterior chamber in a 10-15 minute period via the single needle; the solution then exited the eye via the detached arm of the branched needle. During this period, the reservoirs were emptied and filled with the same solution that was infused into that eye. Following the exchange, the inflow line was reconnected to the reservoir of the perfusion apparatus and the tubing from the infusion syringes was clamped.
  • H-7 has been tested in the muscle chamber.
  • the muscle chamber will be used for additional compounds that show effectiveness on facility in vivo, and especially if they affect accommodative responses to pilocarpine in vivo.
  • a description of the set-up and methodology follows hereafter.
  • globes were enucleated from rhesus monkeys (Macaca mulatto) aged 6 to 18 years under deep pentobarbital anesthesia (25 mg/kg IV) just prior to or within 5 minutes of euthanization and placed in a cell culture medium (Medium 199 with added penicillin G/streptomycin (50 units/ml) and 10% fetal bovine serum; all from Sigma) at 4°C.
  • the ciliary muscle was then dissected under a surgical microscope (Zeiss OP-MI 6), yielding a section of muscle approximately 4 cm circular x 4 mm meridional and including the entire anterior-posterior extent of the ciliary muscle from the scleral spur to the ora serrata.
  • a 5 mm circular x 4 mm meridional strip was then cut from the section, secured to four acrylic attachment rods with cyanoacrylate adhesive (Tri-Point Medical and B. Braun Melsoder AG) and mounted in the apparatus.
  • Figure 4 is a schematic diagram showing the attachment of a ciliary muscle strip to strain gauges in the muscle chamber apparatus.
  • the muscle chamber apparatus is used to measure contractile force simultaneously in the longitudinal and circular vectors.
  • the first acrylic attachment rod 30 was positioned at the cut anterior end of the muscle 55 analogous to the scleral spur/trabecular meshwork; the second acrylic attachment rod 31 was positioned at the cut posterior end of the muscle analogous to the ora serrata 36.
  • the first rod was connected to a first strain gauge (not shown) via a drop of paraffin wax, while the second, opposing rod, was clamped in position in the apparatus; the position of the clamp is depicted by the solid arrows 40.
  • the third acrylic attachment rod 32 was positioned at the medial cut edge of the muscle 37 and was connected to a second strain gauge (not shown), while the fourth acrylic attachment rod 33 was clamped in position at the opposite lateral cut muscle edge 38; the position of the clamp is depicted by the solid arrows 41.
  • the second strain gauge thus registered force representing primarily the circular (putatively more accommodation-specific) vector of ciliary muscle contraction.
  • the remainder of the original muscle section is stored in culture medium (Medium 199; Sigma) at 4°C for future use.
  • the muscle chamber was maintained at 34°C and perfused continuously with warmed oxygenated Krebs solution [made by the inventors themselves: ionic composition (mM): Na " . , . , . , . , 2 . , 3 . , . , . , pH 7.4, continuously oxygenated with 95% 0 2 /5% C0 2 ] or Krebs solution containing drug.
  • ionic composition (mM) Na " . , . , . , . , 2 . , 3 . , . , . , pH 7.4, continuously oxygenated with 95% 0 2 /5% C0 2 ] or Krebs solution containing drug.
  • the first and third attachment rods were secured via paraffin to two strain gauges (force transducers); the strain gauges were mounted on micropositioners for precise control of muscle resting tension. Muscle strip dimensions were measured by the use of dividers and Vernier calipers prior to and immediately following mounting in the muscle chamber.
  • a muscle strip was mounted in the muscle chamber beginning 1 hour after enucleation, used for 2 to 3 hours, and replaced by new strips (stored at 4°C until use) successively over the next 8 to 10 hours. Muscle strips were also used on the day following enucleation, having been kept in the cell culture medium at 4°C overnight.
  • the muscarinic agonist carbachol induced reproducible dose-dependent, atropine-inhibitable contractions in both vectors, with essentially super-imposable dose- response curves (data not shown). The responses were reproducible for the 2-3 hours use of fresh, same-day-stored and overnight-stored strips.
  • each ciliary muscle strip was exposed to Krebs solution incorporating pilocarpine (10 ⁇ M) or carbachol (1 ⁇ M), concentrations of these cholinergic agonists known to mediate "just- maximal" contraction of the in vitro ciliary muscle in both contractile vectors. After 15 , just-maximal concentration of cholinergic agent plus increasing concentrations of test compound ( 15 minutes at each concentration) until a reversal of some cholinergic-induced tone was seen. Thereafter, higher concentrations of test compound were utilized until a maximal reversal was seen. For compounds which mediate ciliary muscle relaxation, there was no 15 minute period between delivery of test compound at each concentration.
  • Tissue Culture Facilities Tissue culture room contains two BioFlo laminar hoods, for CO 2 incubators, and a full set of accessory tools.
  • Microscopy Two units are available for light microscopy. The first unit is equipped for live-specimen recording, with one upright and one inverted microscope, with temperature-controlled tables, video recording devices, videotape and video disc recorders and a semiautomatic microinjection system. The second unit consists of a digital microscopic system, with several light microscopes including a confocal microscope, and a computerized image reconstruction facility.
  • the electron microscopic unit contains three transmission electron microscopes (Philips EM400, EM410, CMI 2) and two scanning electron microscopes (Philips 515, Jeol 6100) and is fully equipped for sample preparation.
  • the microscope probe was placed on the cornea along the optical axis of the eye and photographs were taken. Prints were made by enlarging the negative 3.5-times, and the prints were overlaid by a grid with 14 x 14 mm squares. The total number of cells in each of four squares was counted and the average taken and multiplied by 100 to obtain the number of cells per square millimeter of the cornea.
  • Ocular Tolerability/Toxicity Ocular exams, including slit lamp biomicroscopy (Zeiss or Haag-Streit), gonioscopy, and binocular indirect ophthalmoscopy (American Optical or Keeler), were performed by a trained ophthalmologist, using when necessary contact lenses specially fabricated for the small monkey eye. [P.L. Kaufman and I.H. Wallow, "Minified diagnostic contact lenses for biomicroscopic examination and photocoagulation of the anterior and posterior segment in small primates," Exp. Eye Res. 40:883-885 (1985)]. In addition, specular microscopy was used to evaluate drug effects on the corneal endothelium.
  • Barany 's solution Barany 's solution, a specialized physiologic phosphate buffer designed to minimize the facility increase induced by anterior chamber perfusion, is well-known in the art. [E.H. Barany, "Simultaneous measurements of changing intraocular pressure and outflow facility in the vervet monkey by constant pressure infusion," Invest. Ophthalmol. 3(2): 135-143 (1964)].
  • the inventors prepared their own solution, one liter of solution containing: 8 g NaCl; 0.35 g KCl; 0.17 g CaCl,; 64 mg MgCl,; 69 mg Na 2 H 2 PO 4 ; 13.7 mg NaH 2 PO 4 ; and 1 g glucose.
  • Source of Chemical Compounds Both H-7 and staurosporine are commercially available from Sigma. Latrunculin A and swinholide A were provided by Dr. I Ian Spector.
  • latrunculin A is a 2-thiazolidinone macrolide isolated from the marine sponge Latrunculia magnifica.
  • the sponges are prominent in and may be collected from the Red Sea, where they can be found at a depth of 6.0 to 30.0 meters.
  • the sponges may be squeezed manually to collect the toxic fluid.
  • a combination of Sephadex LH-20 and Silica-gel chromatographies allows one to get rid of the glycerides in the fluid, and obtain three pure toxins, one of which is latrunculin A which may be isolated via separation techniques known in the art.
  • the structure of latrunculin A, an oil, is presented in Figure 2. [Y.
  • Swinholide A is a macrolide isolated from the marine sponge Theonella swinhoei; the sponge may be collected, among other places, from the Red Sea. Swinholide A may by obtained from frozen Theonella swinhoei lyophilized and successively extracted with petrol- ether and CH 2 Cl 2 /MeOH(8:2).
  • the latter extract may then be chromatographed on Sephadex LH-20(MeOH/CHCl 3 (l . )) and on silica gel (petrol-ether, ether, ethyl acetate).
  • the ethyl acetate fractions provide swinholide A as a microcrystalline compound (melting point, 102°.
  • KT5926 may also be prepared from a related compound, K-252a, which is isolated by techniques, known in the art, from the culture broth of Nocardiopsis sp. K-252. [H. Kase et al , "K-252a, a potent inhibitor of protein kinase C from microbial origin," J. Antibiotic. (Tokyo) 39:1059-1065 (1986)]. K-252a is acetylated with acetic anhydride and dimethylamineopyridine at the -NH and -OH positions.
  • the acetate that is obtained is then acetylated by a Friedel-Crafts reaction and oxidized by a Baeyer-Villiger reaction at the 14- position and the acetyl groups are removed by treatment with sodium methoxide.
  • the 14- hydroxy group introduced above is then H-propylated with sodium hydride and rc-propyl iodide to yield KT5926.
  • KT5926 a potent and selective inhibitor of myosin light chain kinase
  • mean baseline facility for (i) treated eyes (more accurately, to-be-treated eyes) and (ii) control eyes (contralateral eyes from (i)) were determined.
  • the mean baseline ratio of treated eyes/control eyes was then determined; eyes were deemed “equivalent” if they did not differ statistically from 1.0.
  • mean post-drug administration facility and mean control (i.e., post- vehicle) administration facility were determined, and the mean post- administration ratio was compared.
  • the mean ratio of post-administration/baseline was determined for both the treated eyes and the control eyes; using ratios reduces inter-animal variability. Finally, the mean ratio of the ratios determined in the third step was determined:
  • this strategy accounts for differences in baseline values between opposite eyes of the same animal, the effect of the perfusion itself, and differences in starting values between animals, thereby isolating the drug effect from other confounding variables.
  • H-7 Increasing concentrations of H-7 were tested to determine the approximate dose which yielded a statistically significant increase in outflow facility. Initially, each concentration of H-7 was prepared by dissolving the appropriate amount of H-7 in Barany's solution. For the treated eye, H-7 was administered in Barany's solution without DMSO as a vehicle, while the contralateral eye was administered vehicle alone. In separate experiments, 2 ml of H-7 solution was passed through the anterior chamber of the eye in a 10-15 minute period. Facility measurements were taken 60 minutes thereafter.
  • Figure 5A is a bar graph illustrating the effect of several concentrations of H-7 on facility.
  • the "open" bars represent eyes administered vehicle only, while the "speckled” bars indicate eyes administered H-7.
  • ** p ⁇ 0.001 are the probabilities that the double ratio defined above equals 1.0 by the 2- tailed paired t-test.
  • H-7 Does Not Significantly Effect Corneal Endothelial Cell Counts At A Concentration Effective At Increasing Total Outflow Facility
  • the effect of 300 ⁇ M H-7 on corneal endothelium was determined in cynomolgus monkeys via transcorneal intracameral injection.
  • Corneal endothelial cell counts were measured as the number of cells/mm 2 and are reported in Table 3.
  • BL baseline cell counts
  • Rx(l) cell counts 1 hour after H-7 administration
  • Rx(2) cell counts 3 hours after H-7 administration
  • Rx(3) cell counts 2 weeks after H-7 administration
  • Treated eyes administered H-7
  • T/C Treated/Control.
  • agents used in the treatment of glaucoma are administered topically as an eyedrop, rather than being injected into the eye.
  • This example describes the results on outflow facility following the topical administration (i.e., via an eyedrop) of four different concentrations of H-7.
  • ketamine (10 mg/kg) anesthesia
  • slit lamp examination was performed on the monkeys. Thereafter, the monkeys were anesthetized with IM pentobarbital-sodium (35 mg/kg). Baseline facility was measured (7 periods) by 2-level constant pressure perfusion of the anterior chamber; thereafter, the perfusion system was closed.
  • H-7 solutions of differing concentrations were manufactured: (i) a 90 mM solution (-0.3 mg H7 dissolved in 10 ⁇ l of 10% DMSO with Barany's solution, pH of the resulting solution adjusted to approximately 7); (ii) a 150 mM solution (-1.1 mg H7 dissolved in 20 ⁇ l of 25% DMSO with Barany's solution, pH of the resulting solution adjusted to approximately 7); (iii) a 450 mM H-7 solution (-3.3 mg H7 dissolved in 20 ⁇ l of 25% DMSO with Barany's solution, pH of the resulting solution adjusted to approximately 7), and (iv) a 650 mM H-7 solution ( ⁇ 5.7 mg H-7).
  • each concentration of H-7 solution was administered to the superior cornea of one eye and vehicle (25% DMSO) to the opposite eye (5 ⁇ l x 4 drops), waiting 90 seconds between each drop; in addition, the lower eyelid was lifted immediately after each drop to allow adequate drug/vehicle contact with the cornea.
  • the perfusion system was reopened and facility was measured for 9 periods.
  • the resulting data are presented in Figure 5B.
  • the "open" bars represent eyes administered vehicle only, while the "speckled” bars indicate eyes administered H-7.
  • ** pO.OOl are the probabilities that the double ratio defined above will equal 1.0 by the 2- tailed paired t-test. - , ere was a sugges on o a sma e ec on ou ow ac y, u e effect was not statistically significant.
  • topical administration of 150 mM H-7 had a definite statistical effect on outflow facility.
  • the administration of 450 and 650 mM had a larger effect, demonstrating that H-7 has a defined dose-response relationship when administered topically.
  • This example examines the effect of topical administration of H-7 on pupil size and refraction in cynomolgus monkeys.
  • the experimental protocol was as follows. Following administration of ketamine (10 mg/kg) anesthesia, baseline slit lamp examination was performed on the six monkeys tested. Thereafter, the monkeys were anesthetized with IM pentobarbital-sodium (35 mg/kg). Baseline refraction (2-3 readings) and pupil size were then measured as described above. Subsequently, a 150 mM H-7 solution (1.0929 mg H7 dissolved in 20 ⁇ l of 25% DMSO with
  • Figure 6A graphically illustrates the effect of topical H-7 on pupil size, pupil diameter (mm) being indicated as a function of time (min).
  • Time 0 represents drug administration
  • the open squares represent eyes treated with H-7
  • the open diamonds represent the control eyes.
  • the results indicate that the control pupil gets smaller with prolonged anesthesia, while the pupil subjected to H-7 does not.
  • Figure 6B graphically illustrates the effect of topical H-7 on refraction (diopters) indicated as a function of time (min). Again, time 0 represents drug administration, and the open squares represent eyes treated with H-7. while the open diamonds represent the control
  • This example is directed at determining whether selective inhibition of miosis (dependent on contraction of the iris sphincter muscle) as compared to accommodation (dependent on contraction of the ciliary muscle) can be achieved.
  • FIGS 7A-H graphically depict the results of this experiment, and reference to those figures will also assist in understanding the experimental protocol.
  • the experiments of this example examined the miotic response ( Figures 7A, C, E, and G), determined by pupil diameter (mm), and the accommodative response ( Figures 7B, D, F, and H), measured in diopters, to pilocarpine in the presence or absence of H-7 in normal living monkey eyes.
  • Each of the 4 rows of panels represents a different dose of H-7.
  • the left-hand panel represents the pupillary diameter
  • the right-hand panel represents the accommodative response in the same animals.
  • "n" represents the number of animals for that protocol.
  • baseline readings (BL) of the pupillary diameter were obtained with vernier calipers held just in front of the cornea, after which phenylephrine (2.5%) was topically administered to the eyes, resulting in mydriasis.
  • H-7 was then administered, and pupil diameter and accommodation were measured 25 minutes later (H-7 on the graphs).
  • Figures 7A-H are mean ⁇ s.e.m. for n monkeys. The following significance levels are depicted in Figures 7A-H: *P ⁇ 0.05; **P ⁇ 0.01 ; ***P ⁇ 0.001.
  • the significance of the difference between treated and control pupil diameter ( Figures 7A, 7E, 7G) or final accommodative response ( Figure 7H) was determined by the 2-tailed paired t-test.
  • Figures 8A-C depict chart recorder traces obtained from a single in vitro ciliary muscle strip maintained in Krebs solution.
  • muscle strip contraction is indicated in the downward direction per the scale indicated; more specifically, in the scale shown in Figure 8A, the length of the downward-pointing arrow indicates a contraction (tension) of 25 mg, while the length of the horizontal arrow pointing to .
  • H-7 in vivo dissociation of miosis and accommodation in response to an intracameral or intravitreal infusion of H-7 (300 ⁇ M) may have a pharmacokinetic basis. Based on the data from this one concentration, H-7 appears to have no selectivity for one vector over the other; however, vector-selectivity might be observed at other concentrations.
  • bovine aortic endothelial cells were plated at half confluency on clean glass coverslips and cultured in DMEM, containing 1 g of glucose, 9% bovine calf serum, 100 mg penicillin, 100 mg streptomycin per 550 ml, for 2-4 days at 37°C , 7% CO 2 /93% O,, 90%) humidity.
  • the medium was then changed to one containing Latrunculin A concentrations of 0.02, 0.05. 0.2, 0.5, or 2.0 ⁇ M for 1 , 2. 3, 5, or 24 hours, after which the er, er ze w . r on - n , and fixed with 3% paraformaldehyde.
  • the medium was then exposed to rhodamine phalloidin (for fluorescent labelling of actin), or to rabbit pan-cadherin primary antibody followed by goat anti-rabbit secondary antibody (for immunofluorescent labelling of cadherin, a major protein associated with cell-cell adherens junctions); and examined by fluorescence microscopy.
  • Latrunculin A concentrations greater than 0.5 ⁇ M had unequivocal effects after 1 hour of exposure, with partial recovery evident within 1-5 hours after removal of the drug. Lower concentrations had more subtle slower onset effects. Based on these findings, doses of 0.2, 0.5, 2.0, and 5.0 ⁇ M were studied in living monkeys, with the positive findings as described below. It is quite likely, albeit not yet tested, that lower concentrations for longer exposure times might also be effective.
  • This example involved an examination of the dose-dependent effect of latrunculin A, staurosporine, and swinholide A on total outflow facility.
  • the experiments of this example were performed on adult cynomolgus and rhesus monkeys. Both normal monkeys and monkeys with unilateral ciliary muscle disinsertion from the scleral spur/trabecular meshwork were utilized. Briefly, the animals were anesthetized with ketamine (10 mg/kg) followed by pentobarbital sodium (35 mg/kg). perfusion of the anterior chamber of both eyes simultaneously with Barany's solution, as described above. The anterior chamber of both eyes was cannulated with a single branched 26-gauge needle; one arm of the needle was connected by polypropylene tubing to a continuously-weighed reservoir of Barany's solution, while the other arm was connected to a pressure transducer. Facility measurements were taken for 35-45 minutes.
  • 10 ⁇ l of either 5 or 20 ⁇ M latrunculin A or 100 or 300 nM of swinholide A were injected into the inflow tubing in one eye (0.5 ⁇ M, 2.0 ⁇ M, 10 nM, and 30 nM, respectively, in the 100 ⁇ l anterior chamber), vehicle in the other, via a custom-designed micro-t-piece (positioned 2-3 cm for the eye) and a micrometer syringe. After five minutes to allow the perfusand from an elevated reservoir to wash into the anterior chamber and 3 minutes for convection mixing of the anterior chamber by blowing cold air on the cornea, facility measurements were taken for 90 minutes.
  • the anterior chamber of each eye was cannulated with a branched needle connected to a reservoir and an unbranched 26 gauge needle with the tubing clamped off. Baseline facility measurements were taken for 35-45 minutes. The clamped tubing from the unbranched needle was then connected to syringes containing either drug (0.2, 0.5, 2.0, and 5.0 ⁇ M latrunculin A, 0.1 , 1.0 or 10 ⁇ M staurosporine; and 10 or 30 nM swinholide A) or vehicle.
  • drug 0.2, 0.5, 2.0, and 5.0 ⁇ M latrunculin A, 0.1 , 1.0 or 10 ⁇ M staurosporine; and 10 or 30 nM swinholide A
  • the vehicles used were as follows: i) latrunculin A: 0.01% DMSO for 0.2 ⁇ M and 2.0 ⁇ M, 0.25% DMSO for 0.5 ⁇ M, and 0.025% DMSO for 5.0 ⁇ M; ii) staurosporin:
  • swinholide A 0.1 % DMSO for 10 and 30 nM.
  • the monkeys with unilateral ciliary muscle disinsertion received drug in both eyes.
  • the syringes were placed in a variable-speed infusion pump and the reservoir (inflow) tubing disconnected, allowing infusion of 2.0 ml of solution through the anterior chamber over 10-15 minutes (maintaining intraocular pressure at
  • the reservoirs were emptied and filled with the same solution being perfused through the eye.
  • the reservoir tubing was then reconnected and the syringe tubing clamped.
  • outflow facility measurements were immediately taken for 90 minutes.
  • staurosporine the reservoirs were closed to inflow for 45 minutes, then opened and facility measurements were taken for 45 minutes.
  • latrunculin A reversibility exchanges an exchange was done with 5.0 ⁇ M in one eye and vehicle in the other followed
  • BL Baseline
  • PRx Post drug administration
  • CM Dis ciliary muscle disinsertion eye
  • Cont Control eye.
  • Data are meanlSEM for n animals with unilateral CM disinsertion receiving 5.0 ⁇ M Lat-AOU.
  • latrunculin A The minimal reversibility exhibited by latrunculin A was probably due to the washout of resistance-relevant material when the drug was present which does not reform quickly following drug removal. Indeed, it may not reform by the next perfusion, as indicated by the baseline comparisons.
  • Swinholide A doses of 10 and 30 nM did not increase facility, whether administered by bolus or exchange infusion (data not shown). It should be noted, however, that in vitro cell culture experiments with swinholide A did reveal an effect on the cells' actin filaments and cell-cell adhesion. Indeed, those experiments suggested that the dosage range of swinholide A tested in the in vivo monkey experiments described in this example would be at the low end of the dose-duration-efficacy relationship.
  • the apparent ineffectiveness of swinholide A may be due to inadequate dose or exposure time, not to the compound itself. Conversely, this apparent ineffectiveness may not have a pharmacokinetic basis.
  • latrunculin A and staurosporine increased outflow facility. Though swinholide A did not appear to be effective, these are likely due to the testing conditions employed.
  • the results of the latrunculin A and staurosporine experiments indicate that actin cytoskeleton disorganization in the trabecular meshwork can increase outflow facility in monkeys.
  • the present invention provides for new agents for the potential treatment of glaucoma.
  • These agents which act via pharmacological interference with cell adhesions and the associated actin filaments in the ocular fluid drainage pathways, present a clearly needed alternative to the pharmacological modalities currently employed.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Compositions and methods for the treatment of glaucoma are described. The compounds utilized in the compositions and methods cause a perturbation of cell adhesions in the trabecular meshwork, mainly via disruption of the associated cytoskeletal structures or the modulation of their interactions with the underlying membrane. Perturbation of these adhesions reduces the resistance of the trabecular meshwork to fluid flow and thereby reduces intraocular pressure.

Description

FIELD OF THE INVENTION
The present invention relates to the treatment of ocular disorders of the eye and more particularly to the treatment of glaucoma. This invention was made with United States Government support awarded by the
National Institute of Health (NIH), Grant No. EY02698. The United States Government has certain rights in this invention.
BACKGROUND OF THE INVENTION
I. Description And Characterization Of Glaucoma Glaucoma is an ophthalmologic disorder responsible for visual impairment. It is the fourth most common cause of blindness and the second most common cause of visual loss in the United States, and the most common cause of irreversible visual loss among African- Americans. Generally speaking, the disease is characterized by a progressive neuropathy caused at least in part by deleterious effects resulting from increased intraocular pressure on the optic nerve. In normal individuals, intraocular pressures range from 12 to 20 mm Hg., averaging approximately 16 mm Hg. However, in individuals suffering from glaucoma, intraocular pressures generally rise above 25 to 30 mm Hg. and can sometimes reach 70 mm Hg. Importantly, the loss of vision can result from intraocular pressures only slightly above or even within the statistically normal range, in eyes which are unusually pressure-sensitive, over a period of years. Moreover, extremely high pressures (e.g. , 70 mm Hg.) may cause blindness within only a few days. [See, e.g., P.L. Kaufman and T.W. Mittag, "Medical Therapy Of Glaucoma," Ch. 9, Sec. II (pp. 9.7-9.30) In P.L. Kaufman and T.W. Mittag (eds.): Glaucoma (Vol. 7 of S.M. Podos and M. Yanoff (eds): Textbook of Ophthalmology Series). London, Mosby-Year Book Europe Ltd. (1994); A.C. Guyton, Textbook of Medical Physiology (W. B. Saunders Co., Sixth Ed.), pp. 386-89 (1981)].
Several different types of glaucomas exist, each having different pathophysiologies and risk factors. In terms of classification, glaucomas may first be deemed to be either "primary" or "secondary." Primary glaucomas result directly from anatomical and/or physiological disturbances in the flow of aqueous humor (i.e., intraocular fluid). The major types of primary glaucomas include (i) open-angle glaucoma (also known as chronic or simple glaucoma), (ii) angle-closure glaucoma (also known as closed-angle or narrow-angle . glaucomas occur as a sequel to ocular injury (e.g. , trauma inflicted to the eye) or preexisting disease (e.g., an intraocular tumor or an enlarged cataract).
II. Etiology Of Glaucoma Glaucoma has been associated with both pharmacological and non-pharmacologica] factors. Non-pharmacological factors include age, race, family history, diabetes, and blood pressure. For example, African Americans are four-to-five times as likely to develop open- angle glaucoma as are Caucasians. Similarly, open-angle glaucoma is much more prevalent in individuals over the age of 40, and especially in those over 60 years of age. In addition, particular drugs have been associated with glaucoma. The corticosteroids
(e.g. , prednisone, dexamethasone, and hydrocortisone) are known to induce glaucoma following both ophthalmic and systemic administration systemic administration, by increasing resistance to aqueous humor outflow through the trabecular meshwork via a mechanism somehow genetically linked to primary open angle glaucoma. Dexamethasone has been associated with the most pronounced increase in intraocular pressure, and ophthalmic administration generally leads to greater increases than systemic administration.
Topically applied ophthalmic drugs which dilate the pupil (adrenergic agonists such as phenylephrine and epinephrine; anticholinergics such as atropine, scopolamine, homatropine. cyclopentolate and tropicamide) may induce angle-closure glaucoma, while the anticholinergics can increase resistance to aqueous humor outflow in susceptible individuals even without causing angle-closure, apparently related to their cycloplegic (ciliary muscle/accommodation-paralyzing) action. Adrenergic (e.g., central nervous system stimulants, appetite suppressants) and anticholinergic (e.g., bowel relaxants and tricyclic anti depressants) agents administered systemically may also induce angle closure glaucoma, via secondarily dilating the pupil.
III. Therapeutic Modalities
Current glaucoma therapies are restricted to reducing intraocular pressure. Presently, both surgical and pharmacological treatments are used.
A number of different pharmacological modalities are presently employed. Unfortunately, each is associated with adverse effects that limit its usefulness. The miotic agents (i.e.. agents that causes the pupil to contract) represent one frequently used class of . . . , , which act on the muscarinic receptors of the ciliary muscle to produce contraction) or indirect-acting (e.g. , anti-cholinesterases, such as echothiophate and isoflurophate, which prevent the degradation of the endogenous cholinergic neurotransmitter acetylcholine, thereby permitting strong and continuous contraction of the ciliary muscle). The adverse effects associated with the miotic agents include painful ciliary or accommodative spasm and consequent induced fluctuating myopia and blurred vision (in patients under the age of approximately 45 years), ciliary or conjunctival congestion, ocular pain, headache, and the miosis itself, which can cause a disabling reduction of vision in elderly patients with early cataract formation (the combination of the small pupil and the cloudy lens critically reduce the amount of light reaching the retina). The long-acting anticholinesterase are associated with the most severe and prolonged adverse effects, which may include development of cataracts or retinal detachment.
Several mydriatic agents (e.g., the sympathomimetic amines epinephrine and dipivefrin) are also useful in the initial treatment of open-angle glaucoma. The adverse ocular effects of these agents include ocular congestion or hyperemia, ocular pain and headache, and blurred vision. In addition, epinephrine occasionally causes untoward systemic effects, such as palpitation, tachycardia, and hypertension. Likewise, the frequently used non-selective β,- and β2-adrenergic blocking agents (e.g. , timolol and levobunolol) and β,-selective (e.g. , betaxolol) adrenergic blocking agents are also used in the treatment of open-angle glaucoma and are also associated with several untoward effects.
As set forth above, untreated glaucoma can result in severe consequences, including blindness. Clearly, new treatment methods and agents are needed and would be welcomed by those plagued by glaucoma who either cannot tolerate available treatment regimens or who are unwilling to undergo invasive surgical procedures.
SUMMARY OF THE INVENTION
The present invention describes compositions that can be used to treat glaucoma.
The present invention contemplates a method of enhancing aqueous humor outflow in the eye of a subject to treat glaucoma, comprising: a) providing a subject with glaucoma: and b) administering to the subject an ophthalmic preparation comprising an effective amount of a non-corneotoxic compound selected from the group consisting of serine-threonine kinase inhibitors, tyrosine kinase inhibitors, protein tyrosine phosphatase inhibitors, and an actin-
- J , , enhancing aqueous humor outflow in the eye and treating the glaucoma. In one embodiment of the present invention, the serine-threonine kinase inhibitor is selected from the group consisting of l-(5-isoquinolinyl-sulfonyl)-2-methylpiperazine [H-7]; l-(5-iodonaphthalene-l - sulfonyl)-l H-hexahydro-l ,4-diazepine [ML-7]; (8Λ*,95*,1 15*)-(-)9-hydroxy-9- methoxycarbonyl-8-methyl-14-«-propoxy-2,3,9, 10-tetrahydro-8,l 1-epoxy, \ H, H, 1 1H- 2,7b, 1 la-triazadibenzo[α,g]cycloocta[cfifeJ trinden-1 -one [KT-5926J; and staurosporine. In some embodiments, the tyrosine kinase inhibitor is selected from the group consisting of 3,5- diisopropyl-4-hydroxybenzylidene, N-benzyl-3,4-dihydroxybenzylidene thioacetamide, N-(3- phenylpropyl)-3,5-diisopropyl-4-hydroxy benzylidene acetamide, and N-(4-phenylpropyl)-3.4- diisopropyl-4-hydroxy benzylidene acetamide. In further embodiments, the protein tyrosine phosphatase inhibitor is selected from the group consisting of vanadate and phenylarsin oxide. In particular embodiments, the method of administration is topical. In other embodiments, the method of administration is intracameral. In still other embodiments, the method of administration is intracanalicular.
The present invention also contemplates an ophthalmic composition, comprising an effective amount of a compound capable of enhancing aqueous humor outflow in the eye of a subject to treat glaucoma and a pharmaceutically acceptable carrier, the compound selected from the group consisting of serine-threonine kinase inhibitors, tyrosine kinase inhibitors, protein tyrosine phosphatase inhibitors, and an actin-disrupting compound selected from latrunculin A, latrunculin B and swinholide A. In some embodiments of the present invention, the serine-threonine kinase inhibitor is selected from the group consisting of l-(5- isoquinolinyl-sulfonyl)-2-methylpiperazine; 1 -(5-iodonaphthalene- 1 -sulfonyl)- 1 H-Hexahydro- 1 ,4-diazepine; (8i?*,95'*,l lS*)-(-)9-hydroxy-9-methoxycarbonyl-8-methyl-14-«-propoxy- 2,3,9,10-tetrahydro-8,l l-epoxy, 1H,8H, HH-2,7b,l l a-triazadibenzo[α,g]cycloocta[c<fe] trinden-1-one; and staurosporine. In still other embodiments, the tyrosine kinase inhibitor is selected from the group consisting of 3,5-diisopropyl-4-hydroxybenzylidene, N-benzyl-3,4- dihydroxybenzylidene thioacetamide, N-(3-phenylpropyl)-3,5-diisopropyl-4-hydroxy benzylidene acetamide, and N-(4-phenylpropyl)-3,4-diisopropyl-4-hydroxy benzylidene acetamide. In further embodiments, the protein tyrosine phosphatase inhibitor is selected from the group consisting of vanadate and phenylarsin oxide.
Furthermore, the present invention contemplates the use of a compound selected from the group consisting of serine-threonine kinase inhibitors, tyrosine kinase inhibitors, protein , - A, latrunculin B and swinholide A, for the preparation of an ophthalmic composition for enhancing aqueous humor outflow in the eye of a subject to treat glaucoma. In certain embodiments of the present invention, the serine-threonine kinase inhibitor is selected from the group consisting of l-(5-isoquinolinyl-sulfonyl)-2-methylpiperazine; l-(5-iodonaphthalene- l-sulfonyl)-lH-Hexahydro-l,4-diazepine; (8Λ*,95*,l IS*)-(-)9-hydroxy-9-methoxycarbonyl-8- methyl- 14-r?-propoxy-2,3,9, 10-tetrahydro-8, 1 1 -epoxy, 1 H,8H, 1 1 H-2.7b, 1 1 a- triazadibenzo[c7,g]cycloocta[cfife] trinden-1-one; and staurosporine. In particular embodiments, the tyrosine kinase inhibitor is selected from the group consisting of 3,5-diisopropyl-4- hydroxybenzylidene, N-benzyl-3,4-dihydroxybenzylidene thioacetamide, N-(3-phenylpropyl)-
3,5-diisopropyl-4-hydroxy benzylidene acetamide. and N-(4-phenylpropyl)-3.4-diisopropyl-4- hydroxy benzylidene acetamide. In further embodiments, the protein tyrosine phosphatase inhibitor is selected from the group consisting of vanadate and phenylarsin oxide. In other embodiments, the use of the compositions entails either topical, intracameral, or intracanalicular administration.
Moreover, the present invention contemplates methods of enhancing aqueous humor outflow in the eye of a subject to treat glaucoma, comprising: a) providing a subject with glaucoma; and b) administering to the subject an ophthalmic preparation comprising an effective amount of a non-corneotoxic compound selected from the group consisting of angiotensin II, Al-adenosine agonists, acepromazine, promazine, aceclidine, lysophosphatidic acid, genistein; endothelin-, transforming growth factor, interleukin- 1 , platelet activating factor, and 3α,5β-tetrahydrocortisol. In particular embodiments, the compounds are either administered topically, intracamerally, or intracanilicularly. Similarly, the present invention contemplates an ophthalmic composition, comprising an effective amount of a compound capable of enhancing aqueous humor outflow in the eye of a subject to treat glaucoma and a pharmaceutically acceptable carrier, the compound selected from the group consisting of angiotensin II, Al-adenosine agonists, acepromazine, promazine, aceclidine, lysophosphatidic acid, genistein; endothelin-, transforming growth factor, interleukin- 1 , platelet activating factor, and 3α,5β-tetrahydrocortisol. The present invention also contemplates the use of the aforementioned compounds for the preparation of an ophthalmic composition for enhancing aqueous humor outflow in the eye of a subject to treat glaucoma.
The present invention provides effective and non-invasive methods of treating glaucoma without causing untoward and unacceptable adverse effects, such as corneal edema. To facilitate understanding of the invention and the chemical schemes set forth in the disclosure that follows, a number of terms are defined below.
The term "ophthalmic preparation" refers to a composition containing a compound active so as to forestall the progression of glaucoma as well as other pharmaceutically acceptable ingredients. The characteristics of the composition will depend on a number of factors, including the mode of administration. For example, the composition may be a solution, suspension or the like that can be administered topically as an eyedrop. In addition, the composition may be in a form suitable for intracameral administration; for example, microinjection through the cornea into the anterior chamber so that the compound can reach the trabecular meshwork. Alternatively, the drug could be administered intracanalicularly, i.e., by retrograde microinjection into the venous collector channels draining Schlemm's canal or into Schlemm's canal itself. This eliminates the risk of corneotoxicity. The ophthalmic preparation may contain diluents, adjuvants and excipients, among other things. The term "effective amount" refers to that amount of an ophthalmic preparation that is required to successfully treat glaucoma. The effective amount of an ophthalmic preparation may depend on a number of factors, including the age, race, and sex of the subject and the severity of the glaucoma and other factors responsible for biologic variability. Though the term effective amount is not limited to a particular mechanism of action for a specific compound, an effective amount may be that amount of a compound able to disrupt cell junctions in the trabecular meshwork of the eye to increase aqueous humor outflow. The term "subject" includes humans as well as animals.
The term "glaucoma" refers to an ophthalmologic disorder responsible for visual impairment. The disease is characterized by a progressive neuropathy caused at least in part by deleterious effects resulting from intraocular pressure on the optic nerve. The term glaucoma refers broadly to both primary glaucomas, which include open-angle, angle-closure, and congenital glaucomas, and secondary glaucomas, which occur as a sequel to ocular injury or preexisting disease. Though not limited to any particular type of glaucoma, it is anticipated that the pharmacological agents and compositions of the present invention will be most efficacious in the treatment of open-angle glaucoma.
The term "aqueous humor outflow" refers to the drainage of aqueous humor from the eye. As described in further detail below, aqueous humor leaves the eye by passive bulk flow via two pathways at the anterior chamber angle, the trabecular or conventional route and the , , . drainage via both pathways.
The terms "serine-threonine kinase inhibitors" and "ser-thr inhibitors" refer to compounds known to be myosin light-chain kinase inhibitors. These compounds primarily inhibit myosin light-chain kinase, thus inhibiting actomyosin-driven contractility; upon long exposure (>30 minutes), they cause deterioration of the entire microfilament system. These changes affect predominantly cell-extracellular matrix adhesions. The ser-thr inhibitors include, but are not limited to, H-7, KT-5926, ML-7, and staurosporine.
The term "non-corneotoxic" refers generally to the absence of medically-unacceptable side effects on the cornea. One of the primary side effects to be avoided is corneal edema, which is manifested by abnormal fluid accumulation within the intercellular spaces of the cornea. A main etiologies of corneal edema is impairment of the corneal endothelium due to the introduction of drugs and other chemical compounds. Specular microscopy can be used to evaluate drug effects on the corneal endothelium, and the absence of a statistical change in corneal endothelial cell counts is indicative of a non-corneotoxic compound. In addition, the presence of corneal toxicity can be evaluated through ocular examination, which may include slit lamp biomicroscopy, gonioscopy, and corneal thickness measures.
DESCRIPTION OF THE FIGURES
Figure IA schematically depicts the major anatomical structures of the eye. Figure IB schematically illustrates the basic anatomy of the primate anterior chamber angle. The trabecular (conventional) route of aqueous humor flow is indicated by the uninterrupted lines, while the uveoscleral (unconventional) route of aqueous humor flow is indicated by the dashed lines.
Figure 2 depicts the chemical structures of some of the compounds contemplated for use with the present invention.
Figure 3 is a schematic drawing of the anterior chamber perfusion set-up used in conjunction with some of the experiments.
Figure 4 is a schematic diagram showing the attachment of a ciliary muscle strip to strain gauges in the muscle chamber apparatus. The muscle chamber apparatus is used to measure contractile force simultaneously in the longitudinal and circular vectors. - on facility in living monkey given as an exchange infusion with total outflow facilitites measured by 2-level constant pressure perfusion before and after drug administration.
Figure 5B is a bar graph illustrating the effect on facility of several concentrations of H-7 topically administered to living monkeys.
Figure 6A graphically illustrates the effect of topical H-7 on pupil size.
Figure 6B graphically illustrates the effect of topical H-7 on refraction.
Figures 7A-H graphically depict miotic response (Figures 7A, 7C, 7E, and 7G) and accommodative response (Figures 7B, 7D, 7F, and 7H) to intramuscular pilocarpine in either the presence or absence of H-7.
Figures 8A-C depict chart recorder traces obtained from a single in vitro ciliary muscle strip and were designed to determine the effect of H-7 on the ciliary muscle's contractile response to pilocarpine.
Figures 9A-D graphically depict the effect in living monkeys of latrunculin A given as an exchange infusion with total outflow facilitites measured by 2-level constant pressure perfusion before and after drug administration. (Fig. 9A: 0.2 μM; Fig. 9B: 0.5 μM; Fig. 9C: 2.0 μM; and Fig. 9D: 5.0 μM).
Figure 9E graphically illustrates the effect of latrunculin A in ciliary muscle- disinserted eyes. Figure 10 graphically depicts the reversibility of the facility-effect of latrunculin A.
Figures 1 1A-C graphically depict the effect in living monkeys of staurosporine given as an exchange infusion with total outflow facilitites measured by 2-level constant pressure perfusion before and after drug administration (Fig. 11 A: 0.1 μM; Fig. 1 IB: 01.0 μM; Fig. 1 1C: 10.0 μM).
DESCRIPTION OF THE INVENTION
The present invention relates to the treatment of glaucoma. While the present invention does not depend on an understanding of the mechanism by which successful treatment is accomplished, it is believed that the compositions of the present invention cause transient, drug-induced dissociation of cell-cell junctions which facilitates fluid flow around the cells and across the monolayer ("pericelluiar route"), leading to reduction in intraocular pressure. Considerations And Aqueous Humor Dynamics; II) Structural Considerations of the Eye; III) Cell Culture, Animal Model, and Experimental Apparatus; IV) Evaluation of Compounds for Use in the Present Invention; V) Potential Therapeutic Chemical Compounds; and VI) Composition and Administration of Compounds. Each of these parts will be discussed in turn.
I. Anatomical Considerations And Aqueous Humor Dynamics
The major anatomical structures of the eye are shown in Figure IA. Referring to Figure IA, the anterior portion of the sclera 1, the cornea 2, is modified to allow light rays to enter the eye. The transparent, biconvex lens 6, one of the refractive media of the eye, is secured by the lens ligament (zonule) 7 to the ciliary body 8. [See Review of Medical Physiology, W.F. Ganong, Ch. 8: Vision (pp. 1 17-136); Appleton & Lange (Thirteenth Ed. 1987)].
The anterior chamber 12 of the eye contains a clear liquid material called the aqueous humor; the aqueous humor is also referred to as intraocular fluid. As described in more detail below, aqueous humor is produced in the ciliary body/ciliary processes 8. A region exists between the lens 6 and the iris 9, called the posterior chamber 16, that is also filled with aqueous humor.
One of the requirements of normal visual function is an optically clear pathway from the anterior corneal surface to the outer retina. This requirement precludes direct vascular perfusion of the transparent cornea 2 and lens 6 to satisfy their metabolic needs. The normal circulation of aqueous humor through the anterior chamber 12 and posterior chamber 16 against resistance not only satisfies corneal and lenticular metabolism, it also maintains optical clarity and the shape of the eye. Figure IB schematically illustrates the basic anatomy of the primate anterior ocular segment and the normal pathways of aqueous humor flow. Aqueous humor enters the posterior chamber 16 from the ciliary body/ciliary processes 8 as a consequence of hydrostatic and osmotic gradients between the posterior chamber and the ciliary process vasculature and stroma and active ionic transport across the ciliary epithelium. The aqueous humor then flows around the lens 6 and through the pupil into the anterior chamber 12. The aqueous humor then leaves the eye by passive bulk flow via two pathways at the anterior chamber angle: (i) the trabecular or conventional route (hereafter "the trabecular route"), or (ii) the , , . trabecular route, aqueous humor travels through the trabecular meshwork 15, across the inner wall of Schlemm's canal 18 into its lumen, and then into collector channels 20, aqueous veins 21 , and the general venous circulation (not labelled) (the trabecular route is indicated by the uninterrupted lines in Figure IB). In contrast, with the uveoscleral route, aqueous humor travels across the iris root, uveal meshwork, and the anterior face of the ciliary muscle, through the connective tissue between the muscle bundles, and then out through the sclera (the uveoscleral route is indicated by the dashed lines in Figure IB).
Aqueous humor is continually being produced and drained, and it is the balance between these two processes that regulates the pressure of the intraocular fluid. Briefly, open- angle glaucoma results from decreased permeability of the aqueous humor through the trabecular meshwork 15, whereas angle-closure glaucoma results from shifting of the iris 9 forward so that the anterior chamber angle 10 is obstructed.
II. Structural Considerations Of The Eye The shape, dynamics and adhesiveness of eucaryotic cells depends, to a large extent on a complex network of cytoplasmic filaments, known, collectively, as the cytoskeleton. There are three major classes of cytoskeletal filaments, including actin microfilaments, intermediate filaments and microtubules. Among these, most relevant for cell adhesion are the microfilaments which interact with both cell-matrix and cell-cell junctions through a complex submembrane interlinking "plaque". Recent studies have established that this interaction with the cytoskeleton is essential for the formation of stable junctions. Thus, disruption of the actin network or its dissociation from the membrane-bound plaque perturbs cell adhesion.
In forming this firm anchorage of actin filaments to the junctional membrane, several additional proteins participate, the perturbation of which might alter cell adhesion and which should thus be considered. These include both "anchor proteins" which are involved in the physical linkage of actin to the membrane, and myosin, an actin motor driven by ATP hydrolysis, which is responsible for muscle and non-muscle contractility. In its active form, myosin light chain kinase (MLCK) catalyzes the phosphorylation of a myosin light chain, permitting myosin to interact with actin; this interaction results in the movement of myosin along the actin microfilament leading to cell contraction.
Interference with the formation of the actin microfilaments, promotion of disruption of the filaments, interference with actin assembly into bundles, or disturbance of the relationship the extracellular matrix. It should also be pointed out that compounds that affect junctions, but that do not act through interactions with actin, are also candidates for therapeutic compounds. Ideally, perturbation of cell-cell adhesion might be selective, and achieved without completely breaking adhesions to the underlying extracellular matrix, avoiding a massive loss of cells. If such interference occurs in the fluid drainage pathways of the eye. it would enhance aqueous humor egress from the eye, with a concomitant lowering of intraocular pressure. This concept forms the basis for the use of the therapeutic compositions of the present invention. The trabecular meshwork consists of interlacing connective tissue beams, each surrounded or "coated" by a single layer of endothelial-like cells. The beams form a latticework, comprising several layers. The spaces between the beams are filled with extracellular matrix, consisting of glycosaminoglycans, proteoglycans, and other macromolecules. The outer-most portion of the meshwork contains no beams, but rather several layers of endothelial cells imbedded in the "sea" of extracellular matrix. Just beyond this region is a small venous channel, the canal of Schlemm, which is connected to the general venous circulation. The pressure in Schlemm's canal is ~9 mm Hg. The resistance of the trabecular meshwork normally is such that intraocular pressure is ~16 mm Hg, at which aqueous humor leaves the eye at the same rate at which it is produced (-2.5 μL/minute). The exact pathways by which aqueous humor flows through the meshwork and across the wall and thus into the lumen of Schlemm's canal, and the exact sites of resistance within the meshwork, are still unclear. However, it is clear that the barriers constituted by the cells, the extracellular matrix, and the adhesions between them are major determinants of both the flow pathways and the resistance to flow. Several classes of drugs theoretically could act directly on the trabecular meshwork and Schlemm's canal to alter outflow facility. However, these classes of drugs may also affect smooth muscle, such as the ciliary muscle; ciliary muscle relaxation may cause an undesired decrease in facility. Because ciliary muscle contraction and relaxation cause an acute increase or decrease in facility, respectively, one must be able to determine whether a drug-induced facility change is due to a direct action on the conventional outflow channels or an indirect action exerted via altered ciliary muscle tone.
The disruption of overall trabecular meshwork architecture and the consequent increase in outflow facility by cytochalasins, calcium chelators, and sulfhydryl reactive agents has been . , w o e potential cellular and junctional proteins whose alteration might rearrange meshwork geometry to therapeutic advantage. More importantly, these agents exert unwanted effects on other anterior segment structures whose integrity depends on those particular proteins. Regarding the cytochalasins, researchers have found that they interfere with actin filaments and enhance fluid outflow from the living monkey eye; this finding was first published in the mid-1970's. [See, e.g. , P.L. Kaufman and E.H. Barany, "Cytochalasin B Reversibly Increases Outflow Facility in the Eye of the Cynomolgus Monkey," Invest. Ophth. & Visual Science 16(l):47-53 (1977)]. However, the cytochalasins are not presently being used in therapy for several reasons, most notably corneal intolerance upon acute administration.
In addition, ethacrynic acid has been tested in regards to increasing aqueous humor outflow in the eye. For example, U.S. Patent Nos. 4,757,089 and 5,306,731 to D.L. Epstein, hereby incorporated by reference, describes a method of increasing aqueous humor outflow by administering ethacrynic acid, or an analog thereof, containing a chemical group capable of reacting with sulfhydryl groups in the trabecular meshwork. Recent studies have shown that topically administered ethacrynic acid resulted in progressive corneal toxicity that was clinically unacceptable. [M.A. Croft and P.L. Kaufman, "Effect of daily topical ethacrynic acid on aqueous humor dynamics in monkeys," Curr. Eye Res. 14:777-781 (1995)]. Finally, research has been conducted with the calcium chelators EDTA and EGTA demonstrating breaking of cell-cell junctions in the trabecular meshwork and concomitant increases in outflow facility. [See, e.g., A. Bill et al., "Effects of intracameral Na2EDTA and EGTA on aqueous outflow routes in the monkey eye," Invest. Ophthalmol. Vis. Sci. 19:492- 504 (1980); A. Bill, "Basic physiology of the drainage of aqueous humor," In: L.Z. Bito ct al. eds., The Ocular and Cerebrospinal Fluids. Fogarty International Center Symposium.
London: Academic Press. Exp. Eye Res. 25:291-304 (1977)]. These compounds remove calcium ions that are necessary for Ca2+ -dependent junctions, and thus function by a mechanism of action distinct from the compositions of the present invention.
HI. Animal Model And Experimental Apparatus
The monkey is a suitable experimental animal because the biological apparati associated with aqueous humor formation and drainage closely resemble those of the human. Also, the monkey has a large accommodative amplitude, allowing technically simple, non- , perturbed and non-perturbed conditions. Indeed, a previous report of the Glaucoma Panel of the National Advisory Eye Council noted the preferability of primates as opposed to sub- primate mammals for studies such as those described in this application. [U.S. DHHS. Vision Research - A National Plan: 1983-87. 1987 Evaluation and Update. Report of the National
Advisory Eye Council 1987; NIH Publication No. 87-2755:231-278.]
A number of different genera and species of monkeys have provided useful information regarding ocular structure and function, including the owl monkey (Aotus trivirgatus), the vervet monkey (African green; Cercopithecus ethiops), and several species of macaques [cynomolgus - M. fasicularis (formerly M. irus); rhesus - M. mulatto; pigtail - M. nemestrina; and stumptail - M. arctoides]. The cynomolgus has been used most frequently, and a large body of information has been accumulated on the functional and structural characteristics and responses of its accommodation and aqueous humor formation and drainage apparati. The apparatus for determining aqueous humor outflow facility in the living monkey eye employs the two-level constant pressure perfusion technique. The basic methodology is well-described in the literature and is standard in the field. [See, e.g., M. J. Menage et al., "ε-Aminocaproic Acid Does Not Inhibit Outflow Resistance Washout in Monkeys," Invest. Ophthalmol Vis. Sci. 36(9): 1745- 1749 (1995)]. In addition to performing experiments on live monkeys, tissue culture experiments on monkey ciliary muscle have also been performed. The inventors of the present invention developed a custom apparatus (described in the Experimental section) in which the contractile properties of isolated monkey ciliary muscle strips, freed from their posterior attachments to the choroid/sclera and anterior attachments to the scleral spur/trabecular meshwork/cornea, can be studied in both the longitudinal contractile vector (putatively more outflow specific) and the circular contractile vector (putatively more accommodation specific) simultaneously. The apparatus represents an advance over other systems which permit measurement in only one, often undefined, vector at a time.
IV. Evaluation Of Compounds For Use In The Present Invention The screening (evaluation) procedures described hereafter can be used to test compounds that might exhibit beneficial effects by altering the cytoskeleton or the cell-cell or cell-extracellular matrix adherens junctions. Specifically, it can be used to examine n men n egr y or membrane anchorage or inhibition of contractility.
Screening is first performed in cell culture. To that end, endothelial cells, cultured on glass coverslips, are exposed to different concentrations of each compound for different periods of time. Thereafter, they are fixed and either examined under the microscope directly, or immunofluorescently-labeled for actin (since actin is a central element in both cellular- cellular and cellular-extracellular matrix adherens-type junctions), vinculin (a central element in both types of junctions), and cadherins and catenins (critical components of cell-cell contact whose presence and ability to interact are essential for junction integrity), as well as other junctional molecules. Those compounds which affect junction integrity or the integrity of the microfilament system are selected for further analysis. The effective concentrations in culture also serve as guidelines for the live animal experiments
Table 1 sets forth compounds contemplated for use with the methods and compositions of the present invention.
TABLE 1
Class of
Examples Effect Compound
Cause relaxation of acto-myos -induced tension Upon long
Ser- Thr kinase H-7, K.T-5926, ML-7, exposure (>30 minutes), the entire microfilament system inhibitors staurosporine deteriorates These changes affect predominantly C-EC adhesions latrunculin A, Specific and well-defined effects on actin filaments, might
Actin-disrupting latrunculin B, also affect junction integrity and consequently increase drugs swinholide A outflow facility
These compounds, families of synthetic tyrosine kinase inhibitors with a highly diversified specificity, effect growth
Tyrosine kinase factor stimulation Recent work demonstrated their capacity
Tyrphostms inhibitors to selectively inhibit tyrosine phosphorylation of cell-cell and cell-ECM adhesions Tyrphostms may either stabilize or de¬ stabilize cell adhesions
Treatment of cells with compounds enhances protein tyrosine phosphorylation by preventing dephosphorylation of tyrosyl
Protein tyrosine sodium orthovanadate, residues Recent studies established that the phosphorylation phosphatase pervanadate, various of tyrosyl residues on adherens junctional proteins such as inhibitors* vanadyl salts catenins, paxillin, tensin, and others, may exert dramatic effects on junction integrity
Of course, it is to be understood that the present invention is not limited to the potential classes of compounds set forth in Table 1. Rather, the present invention broadlv contemplates screening for compounds that may beneficially alter cell-cell or cell-extracellular matrix adherens junctions. For example, other agents that may be screened include , - , , , , acid, genistein; endothelin-, transforming growth factor, interleukin- 1 , platelet activating factor, and 3α,5β-tetrahydrocortisol.
The screening procedure set forth in Table 2A can be used initially to evaluate compounds such as those presented in Table 1 that may be effective in the treatment of glaucoma. Members of the serine-threonine kinase inhibitor class of compounds are particularly effective at enhancing aqueous humor outflow in the eye, and this screen is especially appropriate for compounds in that class.
TABLE 2A
Step Determination Conclusion
Examine in vitro the effect of compounds on corneal endothelial
I Proceed To Step II. cells (e.g., monkey, cat, rabbit) and moφhology in culture.
Determine the effect of compounds on overall ocular
II Proceed To Step III. tolerability/toxicity.
Determine the effect of compounds administered topically on the
Consider proceeding to
III intraocular pressure by non-invasive measurement techniques (e.g., Screen in Table 2B. applanation tonometry).
Compounds that have shown promise based on the screen in Table 2A can then be examined in the more comprehensive screening procedure set forth below in Table 2B.
TABLE 2B
Step Determination Conclusion
Determine compounds effectiveness on outflow facility in living
I Proceed To Step II. monkeys.
Document whether compound is acting directly on the trabecular
11 Proceed to Step III. meshwork in living monkeys with disinserted ciliary muscle.
Perform electron microscopy to analyze the effect of the
III compounds on the structure of the trabecular meshwork and other Proceed to Step IV. tissues.
Determine compounds effect on other properties, including
Consider Proceeding to
IV refraction, accommodative and pupillary responses to pilocarpine; Step V. and corneal endothelial cell counts and moφhology.
Consider therapeutic use if
V Consider clinical trials on human subjects. compound found to be safe and effective.
As illustrated by this outline of the sequence of experimental procedures and the description of the procedures themselves (described in the Experimental section below), thoughtful consideration allows any compound to be evaluated for use with the present invention. Indeed, as described in detail in the Experimental section, these screening procedures have been employed in the experiments performed on compound H-7. . c
A. Ser-thr Kinase Inhibitors (Affecting Myosin Light-Chain Kinase)
Several compounds known to be myosin light-chain kinase inhibitors were studied in the cynomolgus monkey eye in vivo. Data from one compound (H-7, described below) indicates that while these compounds primarily inhibit myosin light-chain kinase, thus inhibiting actomyosin-driven contractility, other mechanisms of action may also be involved. H-7: One specific compound tested, the myosin light-chain kinase and cyclic nucleotide-dependent protein kinase inhibitor l-(5-isoquinolinyl-sulfonyl)-2-methylpiperazine [H-7] induced an ipsilateral increase in total outflow facility in normal and ciliary muscle- disinserted monkey eyes when perfused through the anterior chamber. In addition, H-7 inhibits ipsilateral pilocarpine-induced miosis at intracameral and intravitreal doses far lower than required for inhibition of accommodation.
H-7's mechanism of action may involve loosening or weakening of cell-cell or cell- extracellular matrix adherens junctions of trabecular meshwork cells, or altered cellular contractility, via an effect on actin filaments or acto-myosin interactions. Further evidence strongly suggests that H-7 acts primarily to inhibit myosin light-chain kinase and thus inhibits actomyosin-driven contractility, eventually leading to perturbation of microfilaments. The chemical structure of H-7 is set forth in Figure 2. Staurosporine: Staurosporine is a microbial alkaloid produced by a Streptomyces species. The compound is known to possess several biological activities, including antifungal and hypotensive effects, inhibition of platelet aggregation, and promotion of cell differentiation. [T. Tamaoki et al, Biochem, Biophys. Res. Commun. 135(2):397-402 ( 1986);
H. Matsumoto and Y. Sasaki, Biochem, Biophys. Res. Commun. 158(1): 105-09 (1989)]. As described in the Experimental section, staurosporine was found to increase outflow facility when perfused through the anterior chamber. Figure 2 depicts the chemical structure of staurosporine.
Staurosporine is a modulator of ser-thr kinases that probably, like H-7, acts through inhibition of myosin light-chain kinase. Its effect on the trabecular meshwork has not been definitively elucidated, though staurosporine might decrease contractility, leading to a total disruption of the microfilament system.
ML-7: ML-7 [l-(5-iodonaphthalene-l -sulfonyl)-lH-Hexahydro-1.4-diazepine] is a myosin light chain kinase inhibitor commercially available from Sigma. Though not as , effectively inhibit mouse lung carcinoma 3LL cell attachment to the fibronectin substratum. [M. Isemura et al , "Myosin light chain kinase inhibitors ML-7 and ML-9 inhibit mouse lung carcinoma cell attachment to the fibronectin substratum," Cell Bio. Int. Rep. 15(10):965-972 (1991 )].
KT5926: KT5926 [(8R*,9S*,\ 15*)-(-)9-hydroxy-9-methoxycarbonyl-8-methyl- 14-,?- propoxy-2,3,9,10-tetrahydro-8, 1 1 -epoxy, 1H.8H, HH-2,7b,l la- triazadibenzo[α,g]cycloocta[cfife] trinden-1-one; Sigma] is a potent and selective inhibitor of myosin light-chain- kinase. The compound has been shown to inhibit both Ca27calmodulin- dependent and -independent smooth muscle myosin light chain kinases as well. Though
KT5926 is deemed to be a selective inhibitor of myosin light-chain kinase, it, too inhibits other protein kinases, e.g., cAMP-dependent protein kinase, but with relatively high K, values. The chemical structure of KT5926 is depicted in Figure 2. [S. Nakanishi et al. "KT5926. a potent and selective inhibitor of myosin light chain kinase," Mol Pharmacol 37:482-488 (1990)].
B. Actin-Disrupting Compounds Affecting Actin Integrity
Actin assembly proceeds in all cells within the eye. For example, actin assembly occurs at the ciliary muscle, where it entails formation of α -smooth muscle actin which interacts with myosin during muscle fiber contraction. In addition, actin assembly most likely occurs in all cells at the trabecular meshwork, where it entails polymerization of G-actin monomers into F-actin filaments.
Latrunculin A: Latrunculin A, is a macrolide isolated from the red sea sponge Latrunculia magniβca that acts by destabilizing actin filaments and interfering with their formation. [Y. Kashman et al. , "Latrunculin, a new 2-thiazolidinione macrolide from the marine sponge Latrunculia magnifica," Tetrahedron Lett. 21 :3629-3632 (1980)]. Latrunculin-
A is the first marine macrolide found to contain 16-membered rings; its structure is set forth in Figure 2.
Latrunculin A binds monomeric G-actin in a 1 : 1 molar ratio and thus prevents the nucleation and elongation of actin filaments. Researchers have compared the short- and long- term effects of latrunculin A on cell shape and actin organization to those of cytochalasin D.
The experimental results indicated that latrunculin A (and latrunculin B, a related compound) were more potent and had significant differences on cell shape and actin organization . , concen ra on- effect relationship for latrunculin A in vivo reflects the dissociation constant for the interaction of latrunculin A with actin in vitro, in contrast to the situation with cytochalasin D. [I. Spector et al, "Latrunculins-Novel Marine Macrolides That Disrupt Microfilament Organization and Affect Cell Growth: I. Comparison With Cytochalasin D," Cell Motil. &
Cytoskel. 13:127-44 (1989)].
In terms of latrunculin A's efficacy as a therapeutic agent for the treatment of glaucoma, the inventors of the present invention found that, when perfused through the anterior chamber of the living cynomolgus monkey eye, latrunculin A increases ipsilateral outflow facility (data presented in the Experimental section). The mechanism by which latrunculin A affects outflow facility may involve inhibition of actin assembly in the trabecular meshwork cells, thus causing the cells to loosen their attachments to one another and/or to their extracellular matrix, and to change their shape, thereby enhancing outflow facility. It is noteworthy that latrunculin A primarily affects cell-cell junctions, in contrast to H-7 which mainly causes deterioration of cell-extracellular matrix adhesions.
Swinholide A: Swinholide A is isolated from the marine sponge Theonella swinhoei. and has been described as a 44-membered dimeric dilactone macrolide with a 2-fold axis of symmetry. The compound exhibits antifungal activity and has been shown to be highly cytotoxic to several cancer cell lines. Its chemical structure is set forth in Figure 2. Swinholide A reportedly disrupts the actin cytoskeleton of cells grown in culture, sequesters actin dimers in vitro in both polymerizing and non-polymerizing buffers, and rapidly severs F-actin in vitro with high cooperativity. [M.R. Bubb et al , "Swineholide A is a Microfilament Disrupting Marine Toxin that Stabilizes Actin Dimers and Severs Actin Filaments," J. Biol. Chem. 270:3463-3466 (1995)]. In relation to its effect on outflow facility in monkeys, one of swinholide A's primary mechanisms of action is believed to entail induction of the formation of G-actin dimers.
C. Tyrosine Kinase Inhibitors Or Stimulators
The third class of chemical entities contemplated, based on work in in vitro cell culture systems, for facilitating aqueous humor outflow are the tyrosine kinase inhibitors or stimulators.
The tyrphostins represent one family of tyrosine kinase inhibitors. These compounds, synthetic tyrosine kinase inhibitors with a highly diversified specificity, effect growth factor . phosphorylation of junctional (cell-cell and/or cell-extracellular matrix) proteins. Tyrphostins may either stabilize or de-stabilize cell adhesions. [See T. Volberg et al , "The effect of tyrosin protein phosphorylation on the assembly of adherens-type junctions," EMBO J 1 1 :1733-1742 (1992); T. Volberg et al , "Modulation of intercellular adherens-type junctions and tyrosine phosphorylation of their components in RSV-transformed cultured chick lens cells," Cell Regulation 2:105-120 (1991)]. Examples of triphostins include, but are not limited to, 3,5-diisopropyl-4-hydroxybenzylidene, N-benzyl-3,4-dihydroxybenzylidene thioacetamide. N-(3-phenylpropyl)-3,5-diisopropyl-4-hydroxy benzylidene acetamide, and N- (4-phenylpropyl)-3,4-diisopropyl-4-hydroxy benzylidene acetamide.
D. Protein Tyrosine Phosphatase Inhibitors
The present invention also contemplates the use of protein tyrosine phosphatase inhibitors for the treatment of glaucoma. Studies have indicated that the phosphorylation of tyrosyl residues on adherens junctional proteins such as catenins, paxillin, tensin, and others, may exert dramatic effects on junction integrity. [Y.R. Hadari et al, "Hepatic tyrosine- phosphorylated proteins identified and localized following in vivo inhibition of protein tyrosine phosphatases: effects of H2O2 and vanadate administration into rat livers," Mol. Cellular Endocrinol. 97:9-17 (1993); T. Volberg et al, "The effect of tyrosin protein phosphorylation on the assembly of adherens-type junctions," EMBO J 1 1 :1733-1742 (1992)]. Examples include, but are not limited to, phenylarsin oxide, sodium orthovanadate, pervanadate, and various vanadyl salts. Treatment of cells with compositions comprising these compounds enhances protein tyrosine phosphorylation by preventing dephosphorylation of tyrosyl residues.
VI. Composition And Administration Of Compounds As indicated above, the present invention contemplates using therapeutic compositions of agents that mainly disrupt cell junctions through an effect on microfilaments and/or membrane-microfilament binding in the ocular fluid drainage pathways to reduce intraocular pressure. It is not intended that the present invention be limited by the particular nature of the therapeutic preparation. For example, such compositions can be provided together with physiologically tolerable liquid (e.g. , saline), gel or solid carriers or vehicles, diluents, adjuvants and excipients, such as pharmaceutical grades of mannitol. lactose, starch, , u sacc ar n, c u ose, magnes um car ona e, an e e. ese compositions typically contain l%-95% of active ingredient, preferably 2%-70%.
These therapeutic preparations can be administered to mammals for veterinary use, such as with domestic animals, and clinical use in humans in a manner similar to other therapeutic agents. In general, the dosage required for therapeutic efficacy will vary according to the type of use and mode of administration, as well as the particularized requirements of individual hosts.
The therapeutic compositions contemplated by the present invention may be mixed with diluents or excipients which are physiological tolerable and compatible. Suitable diluents and excipients are, for example, water, saline, dextrose, glycerol, or the like, and combinations thereof. In addition, if desired the compositions may contain minor amounts of auxiliary substances such as wetting or emulsifying agents, stabilizing or pH buffering agents or preservatives.
For topical administration, the compositions of the present invention are generally dissolved or suspended in a carrier like physiological saline (e.g., 0.9%) or methyl cellulose
(e.g., 3%). The high viscosity of a compound like methyl cellulose allows for increased contact time between the eye and the corneal surface, thereby allowing increased corneal penetration. Similarly, a compound like benzalkonium chloride (e.g., 0.025%) that disrupts the corneal membrane can also be used to facilitate corneal penetration. As alluded to in the preceding paragraph, the preferred mode of administration for these compounds entails a solution or suspension that can be administered as an eyedrop. In addition, infrequent intracameral administration could be feasible if done infrequently (e.g. , once-yearly) or for acute post-surgical situations. Of course, the therapeutic compounds may be investigated for their efficacy via other routes of administration, including oral, intravenous, intramuscular, and injection into the collector channels of Schlemm's canal itself.
Because some data has indicated that the compounds have a long duration of action, they may be effective at a dosing interval greater than that of currently available agents. Less frequent administration may be required if extracellular matrix material takes some time to re- accumulate in the ocular fluid drainage pathway following drug-induced wash-out of extracellular material. The following examples serve to illustrate certain preferred embodiments and aspects of the present invention and are not to be construed as limiting the scope thereof.
In the experimental disclosure which follows, the following abbreviations apply : mm (millimeters); Hg. (mercury); eq (equivalents); M (Molar); mM (millimolar); μM
(micromolar); mmol (millimoles); μmol (micromoles); nmol (nanomoles); g (grams); mg (milligrams); μg (micrograms); kg (kilograms); L (liters); mL or ml (milliliters); μL or μl (microliters); cm (centimeters); mm (millimeters); μm (micrometers); nm (nanometers); mA (milli-amperes); min. (minutes); °C (degrees Centigrade); Ca2+ (calcium ion); IV (intravenous); IM (intramuscular); SQ (subcutaneous); QD (daily); BID (twice daily); TID
(three times daily); QID (four times daily); AIDS (acquired immune deficiency syndrome); SAIDS (simian acquired immune deficiency syndrome); C-C (cell-cell); C-ECM (cell- extracellular matrix); MLCK (myosin light chain kinase); AVMA (American Veterinary Medical Association); American Optical (Scientific Instrument Div., Buffalo, NY); B. Braun Melsungen AG (Germany); Calbiochem (San Diego, CA); Haag-Streit (Carl Zeiss,
Thornwood, NY); Hamilton Co. (Reno, NV); Harvard Apparatus Co., Inc. (South Natick); Keeler (Keeler Instruments, Inc., Broomall, PA); Kipp & Zonen (Bohemia. NY); Norma Goertz Instruments (Elk Grove Village, IL); Seiler (Seiler Instrument Co., St. Louis, MO); Sigma (Sigma Chemical Company; St. Louis, MO); Statham (Ording Inc., Houston, TX); Tri- Point Medical L.P. (Raleigh, NC); SD (standard deviations); FDA (United States Food and
Drug Administration); USP (United States Pharmacopeia); DMSO (dimethyl sulfoxide); DMEM (Dulbecco's Modified Eagle's Medium); MES (2-[N-morpholino]ethanesulfonic acid); s.e.m. (standard error of the mean); df (degrees of freedom).
Unless otherwise indicated, the examples that follow utilized the procedures and experimental animals described hereafter.
1. The Experimental Animals And The Animal Model
Absent definitive statements to the contrary, the procedures were performed on adolescent or young adult cynomolgus (Macaca fascicularis) or rhesus (Macaca mulatto) monkeys. Though both sexes were tested, female monkeys weighing 2-12 kg were primarily used. Rhesus monkeys were used for selective procedures only. Specifically, rhesus monkeys were used in (i) the ciliary muscle disinsertion protocols (which encompassed both rhesus and cynomolgus monkeys) and in (ii) the in vitro muscle contraction experiments (which , , . , n order to minimize the risk of infection, cynomolgus and rhesus monkeys are rarely physically together in the laboratory (i.e., they are infrequently anesthetized in the same room).
In order to avoid the possibility of corneal decompensation, the number of anterior chamber cannulations the eyes underwent was limited. Rhesus monkey eyes had up to 8 perfusions, most of which were 2-needle. With cynomolgus monkeys, animals with less than five perfusions were chosen for the topical drop and perfusion experiments (though some animals not used in these experiments have had up to 20 perfusions). It should be noted that cynomolgus monkey eyes will usually tolerate 10 or more invasions before experiencing corneal decompensation.
Empirically, it was found that approximately 8-10 experiments are required for any drug dose in order to obtain a reliable quantitative, statistically testable estimate of the response. Formal sample size calculations have corroborated this impression, as hereafter described. Generally speaking, the inventors wished to identify mean physiologic responses that were >25% of the baseline value (adjusted for non-drug or non-stimulus-related baseline drift) and >1.5 SD of the mean response. The following standard equation for sample size calculation was used: N = 2 (Zα + Zβ)2/(Δ/σ)2, where Zα = 1.645 or 1.960 for one-sided and two-sided 5% significance, respectively; Zβ = 0.84 or 1.282 for 80% and 90%) power, respectively; Δ = population standard deviation; σ = the difference (i.e. , response) in the parameter being measured (Δ and σ must have the same units). From that equation, it was determined that 5.5 experiments were required to detect differences of 1.5 standard deviations in a paired test at a one-sided 5% significance level with 80% power, while 9.3 experiments were needed to detect such a difference at a two-sided 5% significance level with 90%> power. Some of the experimental animals were donated by or purchased from Corning Hazelton Laboratories (Madison, WI) or from the Wisconsin Regional Primate Research
Center (Madison, WI). Other experimental animals were obtained from one of the inventor's breeding colony. The animals were used repeatedly until humanitarian considerations or loss of suitability required their sacrifice. The animals were used in the following experiments: a) A group of approximately thirty surgically untouched animals underwent anterior chamber perfusion to work out drug dose-facility response relationships and/or drug interactions. b) A group of approximately twelve monkeys underwent unilateral or bilateral ciliary muscle disinsertion. This group then underwent intracameral drug
11 the drugs' effects on ciliary muscle tone. Non-invasive experiments were conducted up to twice weekly, depending on the type and duration of anesthesia; long-duration deep anesthesia experiments were neither performed on an animal more than once weekly nor for too many consecutive weeks. Invasive experiments were performed at four-to-six week intervals
(whether the animal was ciliary muscle-disinserted or not) and only when the anterior chambers have no cells or flare at slit-lamp examination by a trained ophthalmologist.
In general, post-surgical analgesic medications were not employed for relatively minor surgical procedures (e.g., iridectomy, perfusions, tail cut-downs); the rationale was to allow recovery to be judged in the undrugged animal. Post-surgically, the animals were isolated in a recovery cage warmed with a heat lamp, given food and water ad libitum, and observed at least once daily by both a veterinarian or caretaker staff and one of the inventors or a project specialist staff. Following major procedures (e.g., ciliary muscle disinsertion), post-surgical analgesic medication (butorphanol 0.2-0.3 mg/kg SQ BID) was administered and the animals received intramuscular antibiotics (procaine penicillin G + dihydrostreptomycin sulfate; Pfizer.
NY) and glucocorticosteroids (methylprednisolone acetate; UpJohn, Kalamazoo, MI) daily for varying lengths of time.
2. Administration Of Anesthesia And Euthanasia
Several different anesthesia protocols were performed depending on the procedures being utilized or the length thereof. The protocols may be summarized as follows: a) For experiments or procedures lasting less than 1 hr (e.g. , iridectomy, suture removal, slit lamp examination, and photography), the following agents were administered: IM ketamine 10 mg/kg + IM diazepam 1 mg/kg or acepromazine 1 mg/kg. b) For experiments or procedures lasting more than 1 hr (e.g., ciliary muscle disinsertion, anterior chamber perfusion, refraction with drugs), the following agents were administered: IM ketamine 10 mg/kg or IM methohexital-sodium 15 mg/kg in conjunction with IM pentobarbital-sodium 35 mg/kg. For especially long experiments (i.e., experiments exceeding approximately 3-4 hours), hourly supplemental IM pentobarbital doses of 10 mg/kg were usually required after the first 2-3 hours. c) For euthanasia, the following agents were administered: IM ketamine
10 mg/kg + IM/IV pentobarbital overdose. The absence of a heart beat was confirmed in a manner consistent with recommendations of the AVMA Panel of Euthanasia. . urg ca roce ure
The methods of iris removal (limbal incision) and ciliary muscle disinsertion (goniotomy-like approach) are well known and have been described in detail elsewhere. [See, e.g., P.L. Kaufman and E. Lutjen-Drecoll. "Total Irridectomy In The Primate in vivo: Surgical Technique And Postoperative Anatomy," Invest. Ophth. 14:766-771 ( 1975);
P.L. Kaufman and E.H. Barany, "Loss of acute pilocarpine effect on outlfow facility following surgical disinsertion and retrodisplacement of the ciliary muscle from the scleral spur in the cynomolgus monkey," Invest. Ophth. 15:793-807 (1976)]. Those well-known procedures were followed in the experiments of this invention. Ciliary muscle disinsertion was verified gonioscopically, physiologically (i.e., through loss of accommodative and outflow facility responses to pilocarpine), and, in some eyes, histologically.
4. Drug Delivery a) Topical: Eye drops were applied to the anesthetized monkeys as previously described. b) Transcorneal: A 30 gauge needle was removed from its hub. connected directly to narrow-bore polyethylene tubing, and threaded through the cornea for several millimeters using a needle driver. The beveled tip of the needle was then pushed into the anterior chamber and drug was administered. The volume of solution delivered did not exceed 20 μl. The needle was then withdrawn from the anterior chamber without any loss of aqueous humor. c) Intravitreal: A 27-30 gauge needle, prepared as above, was inserted directly into the vitreous through the pars plana at either the 10:00 or 2:00 positions. The total volume injected did not exceed 50 μl. d) Systemic: Drug was administered via IM or IV injection. Alternatively, a venous or arterial line of polyethylene tubing was placed in the monkey's tail following a small incision and isolation of the vessel. [See, e.g., B.T. Gabelt and P.L. Kaufman, "Prostaglandin F increases uveoscleral outflow in the cynomolgus monkey," Exp. Eye Res. 49:389-402 (1989)].
5. Cell Culture To evaluate the junction- and cytoskeleton-disrupting potential of the various drugs, monolayers of cultured bovine aortic endothelial cells were used. These cells form, in , , adhesion to the underlying matrix and a well-developed network of actin microfilaments. The integrity of the monolayer can be determined by light- and electron-microscopy, and the organization of the actin cytoskeleton by fluorescent labeling with fluophore-conjugated phalloidin.
These cells were selected because they are related to the trabecular meshwork cells which line the outflow system and because they were found to be very sensitive indicators for junction integrity.
The cells were plated at half confluency on clean glass coverslips and cultured in complete medium for 2-4 days until they reached confluency. The drug was then added to the culture medium for different periods of time, after which the cells were either fixed with a fixative appropriate for the specific antigen being examined or further incubated in a drug-free medium to examine recovery. Fixed or fixed-permeabilized cells were then either fluorescently labelled for actin using a fluorescein- or rhodamine-conjugated phalloidin or immunofluorescently labelled for the different cytoskeletal or junctional proteins.
Immunofluorescence microscopy and transmitted light microscopy were conducted, and the integrity of the microfilament system in control- and drug-treated cells was then compared visually. At the resolution of the light microscope used (Axiophot, Zeiss Oberkochen, Germany), intact junctions usually appeared as a single actin, viculin-, catenin-, and cadherin- rich "line". Disruption of the junctions led to disappearance of this line or its apparent division or "splitting" into two lines due to the dissociation of the junction. A digital microscopic system [Z. Kam et al. , "Mapping of adherens junction components using microscopic resonance energy transfer imaging," J. Cell Sci. 108: 1051 -1062 (1995)] was used for quantitative analyses, morphometry and 3-dimensional reconstruction.
6. Anterior Chamber Perfusion/Outflow Facility
As previously indicated, the apparatus for determining aqueous humor outflow facility in the living monkey eye employs the two-level constant pressure perfusion technique. This technique, and variations thereof, are well known in the art. [See E.H. Barany. "Simultaneous measurements of changing intraocular pressure and outflow facility in the vervet monkey by constant pressure infusion," Invest. Ophthalmol. 3(2):135-143 (1964); P.L. Kaufman and E.H.
Barany, "Loss of acute pilocarpine effect on outflow facility following surgical disinsertion an re ro sp acemen o e c ary musc e rom e sc era spur n e cynomo gus mon ey, Invest. Ophthalmol. 15:793-807 (1976)].
Figure 3 schematically illustrates the present apparatus as used for one eye. Two identical set-ups are used when studying two eyes of an animal simultaneously, as in the data presented hereafter. However, only one computer (Apple Macintosh II with National
Instruments NB MIO-16 data acquisition board running perfusion program written by David Schuh using Labview for Macintosh, National Instruments, Austin. TX), amplifier and recorder (Soltec Corp. 4-channel recorder models 3314 and 6400, San Fernando; Linseis 4- channel recorder model 2045, Princeton Junction) are needed; they handle the input from both eyes simultaneously. The major components of the apparatus include customized 26-gauge needles and polyethylene tubing which connect the anterior chamber of the eye to a continuously-weighed fluid reservoir (Harvard Precision Adjustable Screw Stand (modified with motor and microswitches to raise and lower the reservoir). Harvard Apparatus Co., Inc., South Natick), the change in weight with time providing a measurement of flow. Other major components include a pressure transducer (Statham Universal Transducer P23XL; Ording Inc.,
Houston, TX), strain gauges (Statham Universal Transducing Cell GM2/GM3 with Microscale Accessory; Ording Inc.) and an infusion system via a series of two- and three-way stopcocks (HV3-2 and HV3-2; Hamilton Co., Reno). The infusion system allows regulation and measurement of intraocular pressure and rate of fluid flow from the reservoir and infusion syringe into the eye and calculation of outflow facility from those measurements. [See, e.g.,
M. J. Menage et al. "ε-Aminocaproic Acid Does Not Inhibit Outflow Resistance Washout in Monkeys," Invest. Ophthalmol Vis. Sci. 36(9): 1745- 1749 (1995)]. The flow and pressure data are calculated from the electronic signals provided by the pressure transducer and strain gauge, the signal processing and calculations being performed on-line in real-time by the amplifier and computer, according to the inventors' own software. The recorder simply provides a back-up record of the flow and pressure data, from which the facility can be calculated off-line, should the computer fail.
In a typical experiment, the anterior chamber of each eye was cannulated transcorneally with one branched and one unbranched needle, and connected to the apparatus as described above. The exchange needles were inserted with the branched needle superior to the single needle and separated by several mm, and the opposite ends of the needles were attached to perfusand-filled tubing through which drug, vehicle, and perfusion solutions could pass into the eye. Anterior chamber exchange was accomplished by attaching the tubing of . detached from the reservoir of the perfusion apparatus and fixed at an intraocular pressure so that fluid could exit the eye without changing the intraocular pressure.
Baseline outflow facility measurements were taken for 35-45 minutes, after which the previously-clamped tubing from the unbranched needle was connected to syringes containing drug for one eye and vehicle for the opposite eye via customized cone-tipped needles, and undamped. The syringes were placed in a Harvard Apparatus (Harvard pump model 944) variable-speed infusion pump. One tubing arm of the branched needle was detached from the reservoir of the perfusion apparatus and fixed at an elevated position so that fluid could exit the eye without changing intraocular pressure.
The inflow tubing was disconnected and the pump turned on to inflow at a rate that allowed 2 ml of drug/vehicle solution to pass through the anterior chamber in a 10-15 minute period via the single needle; the solution then exited the eye via the detached arm of the branched needle. During this period, the reservoirs were emptied and filled with the same solution that was infused into that eye. Following the exchange, the inflow line was reconnected to the reservoir of the perfusion apparatus and the tubing from the infusion syringes was clamped.
Facility measurements were taken for ninety minutes beginning either immediately, or after a waiting period of up to 90 minutes during which the infusion line from the reservoir was clamped. The content of the solutions in the syringes and reservoirs and the number and temporal spacing of infusions varied with the protocol. For example, (i) in a reversibility experiment, there was a second infusion after the post-first infusion measurement, in which both eyes received vehicle without active drug, and (ii) in monkeys with ciliary muscle disinsertion in one eye, both eyes received active drug after the baseline measurements. In all cases, facility was calculated by the method of successive weighted averaging.
7. The Muscle Chamber
H-7 has been tested in the muscle chamber. The muscle chamber will be used for additional compounds that show effectiveness on facility in vivo, and especially if they affect accommodative responses to pilocarpine in vivo. A description of the set-up and methodology follows hereafter.
The muscle chamber and the protocol for rhesus monkey tissue collection, ciliary muscle dissection and chamber mounting have been described in detail elsewhere. [See, e.g., . . , es e so a e r esus mon ey ciliary muscle to muscarinic agonists," Curr. Eye Res. I2(5):413-422 (1993); J.F. Poyer et al, "Prostaglandin F effects on isolated rhesus monkey ciliary muscle," Invest. Ophth. Vis. Sci. 36:2461-2465 (1995)]. Briefly, globes were enucleated from rhesus monkeys (Macaca mulatto) aged 6 to 18 years under deep pentobarbital anesthesia (25 mg/kg IV) just prior to or within 5 minutes of euthanization and placed in a cell culture medium (Medium 199 with added penicillin G/streptomycin (50 units/ml) and 10% fetal bovine serum; all from Sigma) at 4°C. The ciliary muscle was then dissected under a surgical microscope (Zeiss OP-MI 6), yielding a section of muscle approximately 4 cm circular x 4 mm meridional and including the entire anterior-posterior extent of the ciliary muscle from the scleral spur to the ora serrata. A 5 mm circular x 4 mm meridional strip was then cut from the section, secured to four acrylic attachment rods with cyanoacrylate adhesive (Tri-Point Medical and B. Braun Melsungen AG) and mounted in the apparatus.
Figure 4 is a schematic diagram showing the attachment of a ciliary muscle strip to strain gauges in the muscle chamber apparatus. The muscle chamber apparatus is used to measure contractile force simultaneously in the longitudinal and circular vectors. Referring to Figure 4, the first acrylic attachment rod 30 was positioned at the cut anterior end of the muscle 55 analogous to the scleral spur/trabecular meshwork; the second acrylic attachment rod 31 was positioned at the cut posterior end of the muscle analogous to the ora serrata 36. The first rod was connected to a first strain gauge (not shown) via a drop of paraffin wax, while the second, opposing rod, was clamped in position in the apparatus; the position of the clamp is depicted by the solid arrows 40. Force registered from the first strain gauge was represented primarily in the longitudinal (putatively more outflow-specific) vector of ciliary muscle contraction. The third acrylic attachment rod 32 was positioned at the medial cut edge of the muscle 37 and was connected to a second strain gauge (not shown), while the fourth acrylic attachment rod 33 was clamped in position at the opposite lateral cut muscle edge 38; the position of the clamp is depicted by the solid arrows 41. The second strain gauge thus registered force representing primarily the circular (putatively more accommodation-specific) vector of ciliary muscle contraction. The remainder of the original muscle section is stored in culture medium (Medium 199; Sigma) at 4°C for future use.
The muscle chamber was maintained at 34°C and perfused continuously with warmed oxygenated Krebs solution [made by the inventors themselves: ionic composition (mM): Na" . , . , . , . , . , 2 . , 3 . , . , . , pH 7.4, continuously oxygenated with 95% 02/5% C02] or Krebs solution containing drug. As alluded to above, the first and third attachment rods were secured via paraffin to two strain gauges (force transducers); the strain gauges were mounted on micropositioners for precise control of muscle resting tension. Muscle strip dimensions were measured by the use of dividers and Vernier calipers prior to and immediately following mounting in the muscle chamber. The original strip dimensions, reestablished in the muscle chamber, determined the muscle's resting tension (which ranged from 100 to 200 mg in both muscle vectors). Output from both strain gauges was recorded on a 2-channel flatbed recorder [Kipp & Zonen (model BD1 12) and Norma Goertz Instruments (model Servoger 124)].
A muscle strip was mounted in the muscle chamber beginning 1 hour after enucleation, used for 2 to 3 hours, and replaced by new strips (stored at 4°C until use) successively over the next 8 to 10 hours. Muscle strips were also used on the day following enucleation, having been kept in the cell culture medium at 4°C overnight. In this system, the muscarinic agonist carbachol induced reproducible dose-dependent, atropine-inhibitable contractions in both vectors, with essentially super-imposable dose- response curves (data not shown). The responses were reproducible for the 2-3 hours use of fresh, same-day-stored and overnight-stored strips.
After a ciliary muscle strip has been equilibrated, increasing concentrations of the experimental compound dissolved in perfusand (Krebs solution) were perfused through the ciliary muscle chamber to establish a concentration-response curve.
Different protocols were followed for compounds which mediate ciliary muscle contraction and for compounds which mediate ciliary muscle relaxation. For compounds which mediate ciliary muscle contraction, following equilibration of a ciliary muscle strip with plain Krebs solution, the strip was exposed to each successive concentration of test compound for a period of 15 minutes, followed by a further 15 minute period of exposure to plain Krebs solution once again, followed by the next concentration of test compound for 15 minutes, etc. The 15 minute period was used based on the inventors' experience that it is the minimum time necessary for establishment of a full drug effect or reversal of a drug effect. For compounds which mediate ciliary muscle relaxation, after equilibration, each ciliary muscle strip was exposed to Krebs solution incorporating pilocarpine (10 μM) or carbachol (1 μM), concentrations of these cholinergic agonists known to mediate "just- maximal" contraction of the in vitro ciliary muscle in both contractile vectors. After 15 , just-maximal concentration of cholinergic agent plus increasing concentrations of test compound ( 15 minutes at each concentration) until a reversal of some cholinergic-induced tone was seen. Thereafter, higher concentrations of test compound were utilized until a maximal reversal was seen. For compounds which mediate ciliary muscle relaxation, there was no 15 minute period between delivery of test compound at each concentration. However, at the conclusion of the test compound investigation, the strip was given a further 15 minute perfusion with Krebs solution incorporating the cholinergic agent for the purpose of testing the ability of the strip to recontract. Finally, perfusion with plain Krebs solution was used to establish the ability of the tissue to relax once again.
Starting concentrations of each test compound were determined from reports in the literature regarding the compound's effects on ciliary muscle contractility, accommodation, outflow facility and other biological parameters. In addition, starting concentrations were also influenced by in vivo monkey studies performed in the inventors' laboratories.
8. Other Experimental Procedures, Equipment, And Reagents a) Tissue Culture Facilities: Tissue culture room contains two BioFlo laminar hoods, for CO2 incubators, and a full set of accessory tools. b) Microscopy: Two units are available for light microscopy. The first unit is equipped for live-specimen recording, with one upright and one inverted microscope, with temperature-controlled tables, video recording devices, videotape and video disc recorders and a semiautomatic microinjection system. The second unit consists of a digital microscopic system, with several light microscopes including a confocal microscope, and a computerized image reconstruction facility. The electron microscopic unit contains three transmission electron microscopes (Philips EM400, EM410, CMI 2) and two scanning electron microscopes (Philips 515, Jeol 6100) and is fully equipped for sample preparation. c) Refraction/ Accommodation: Refraction and Accommodation were measured with a Hartinger coincidence refractometer (Jena Hartinger model 602520-01220; Seiler). d) Cell Counts and Morphology: Corneal endothelial cell counts and morphology were determined for H-7 by specular microphotography (Wide Field Specular
Microscope fitted with an Olympus OM1 35 mm camera; U.S. Patent No. 4,170,398. hereby . Animals were anesthetized with ketamine alone or ketamine and acepromazine.
The microscope probe was placed on the cornea along the optical axis of the eye and photographs were taken. Prints were made by enlarging the negative 3.5-times, and the prints were overlaid by a grid with 14 x 14 mm squares. The total number of cells in each of four squares was counted and the average taken and multiplied by 100 to obtain the number of cells per square millimeter of the cornea. e) Ocular Tolerability/Toxicity: Ocular exams, including slit lamp biomicroscopy (Zeiss or Haag-Streit), gonioscopy, and binocular indirect ophthalmoscopy (American Optical or Keeler), were performed by a trained ophthalmologist, using when necessary contact lenses specially fabricated for the small monkey eye. [P.L. Kaufman and I.H. Wallow, "Minified diagnostic contact lenses for biomicroscopic examination and photocoagulation of the anterior and posterior segment in small primates," Exp. Eye Res. 40:883-885 (1985)]. In addition, specular microscopy was used to evaluate drug effects on the corneal endothelium. f) Baranv's Solution: Barany 's solution, a specialized physiologic phosphate buffer designed to minimize the facility increase induced by anterior chamber perfusion, is well-known in the art. [E.H. Barany, "Simultaneous measurements of changing intraocular pressure and outflow facility in the vervet monkey by constant pressure infusion," Invest. Ophthalmol. 3(2): 135-143 (1964)]. The inventors prepared their own solution, one liter of solution containing: 8 g NaCl; 0.35 g KCl; 0.17 g CaCl,; 64 mg MgCl,; 69 mg Na2H2PO4; 13.7 mg NaH2PO4; and 1 g glucose. g) Source of Chemical Compounds: Both H-7 and staurosporine are commercially available from Sigma. Latrunculin A and swinholide A were provided by Dr. I Ian Spector.
As previously alluded to, latrunculin A is a 2-thiazolidinone macrolide isolated from the marine sponge Latrunculia magnifica. The sponges are prominent in and may be collected from the Red Sea, where they can be found at a depth of 6.0 to 30.0 meters. The sponges may be squeezed manually to collect the toxic fluid. A combination of Sephadex LH-20 and Silica-gel chromatographies allows one to get rid of the glycerides in the fluid, and obtain three pure toxins, one of which is latrunculin A which may be isolated via separation techniques known in the art. The structure of latrunculin A, an oil, is presented in Figure 2. [Y. Kashman et al, "Latrunculin, a new 2-thiazolidinone macrolide isolated from , . Spector et al , "Latrunculins-Novel Marine Macrolides That Disrupt Microfilament Organization and Affect Cell Growth: 1. Comparison With Cytochalasin D," Cell Motil. & CytoskeJ. 13: 127-44 (1989)]. Swinholide A is a macrolide isolated from the marine sponge Theonella swinhoei; the sponge may be collected, among other places, from the Red Sea. Swinholide A may by obtained from frozen Theonella swinhoei lyophilized and successively extracted with petrol- ether and CH2Cl2/MeOH(8:2). The latter extract may then be chromatographed on Sephadex LH-20(MeOH/CHCl3(l . )) and on silica gel (petrol-ether, ether, ethyl acetate). The ethyl acetate fractions provide swinholide A as a microcrystalline compound (melting point, 102°.
C39H66O10 H20). [S. Carmely and Y. Kashman, "Structure of swinholide A, a new macrolide from the marine sponge Theonella swinhoei," Tetrahedron Letters 26:51 1-514 (1985); M.R. Bubb et al, "Swineholide A is a Microfilament Disrupting Marine Toxin that Stabilizes Actin Dimers and Severs Actin Filaments," J. Biol. Chem. 270:3463-3466 (1995)]. In addition, ML-7 (Sigma) and KT5926 (Sigma, Calbiochem) are commercially available. KT5926 may also be prepared from a related compound, K-252a, which is isolated by techniques, known in the art, from the culture broth of Nocardiopsis sp. K-252. [H. Kase et al , "K-252a, a potent inhibitor of protein kinase C from microbial origin," J. Antibiotic. (Tokyo) 39:1059-1065 (1986)]. K-252a is acetylated with acetic anhydride and dimethylamineopyridine at the -NH and -OH positions. The acetate that is obtained is then acetylated by a Friedel-Crafts reaction and oxidized by a Baeyer-Villiger reaction at the 14- position and the acetyl groups are removed by treatment with sodium methoxide. The 14- hydroxy group introduced above is then H-propylated with sodium hydride and rc-propyl iodide to yield KT5926. [S. Nakanishi et al, "KT5926, a potent and selective inhibitor of myosin light chain kinase," Mol Pharmacol 37:482-488 (1990)].
9. Data Analysis a) Control Experiments: For each animal tested, one eye received drug and the opposite eye received vehicle without drug (i.e., a simultaneous contralateral, sham- treated paired control). For unilaterally ciliary muscle-disinserted animals, both eyes received a dose of drug known to be effective. In this situation, the control is the contralateral normal eye, as the question being addressed is whether ciliary muscle disinsertion matters (i.e..
- J 2 - . statistical analysis is as described below.
For the cell culture experiments, the control was culture medium without drug. There is no statistical analysis in this context, the interpretation being descriptive, analogous to pathology. The effects are routinely monitored by microphotography. b) Statistical Analysis: Well-known and accepted techniques were utilized to determine the effect of the compounds tested. The techniques are summarized below.
First, mean baseline facility for (i) treated eyes (more accurately, to-be-treated eyes) and (ii) control eyes (contralateral eyes from (i)) were determined. The mean baseline ratio of treated eyes/control eyes was then determined; eyes were deemed "equivalent" if they did not differ statistically from 1.0. Second, mean post-drug administration facility and mean control (i.e., post- vehicle) administration facility were determined, and the mean post- administration ratio was compared. Third, the mean ratio of post-administration/baseline was determined for both the treated eyes and the control eyes; using ratios reduces inter-animal variability. Finally, the mean ratio of the ratios determined in the third step was determined:
[(post-drug/baseline)lrealed/(post-drug/baseline)conIrol]; this reduces inter- and intra-animal variability. A value statistically greater than 1.0 indicates a drug-induced facility increase (determined using a 2-tailed paired t-test).
As alluded to in the preceding paragraph, this strategy accounts for differences in baseline values between opposite eyes of the same animal, the effect of the perfusion itself, and differences in starting values between animals, thereby isolating the drug effect from other confounding variables.
The following examples serve to illustrate certain preferred embodiments and aspects of the present invention and are not to be construed as limiting the scope thereof.
EXAMPLE 1
Dose-Dependent Effect Of H-7 On Total Outflow Facility Occurs Through A Direct Effect On The Trabecular Meshwork
This example examined the dose-dependent effect of H-7 on total outflow facility, as measured by 2-level constant pressure perfusion of the anterior chamber (described above) of both eyes simultaneously with Barany 's solution. , - n some o e other compounds, described below) were as follows: (i) after determination of vehicle-only baselines, one eye received drug and the other eye received vehicle without drug; (ii) analogous to the first protocol, with the exception that after the post-drug determination, both eyes received vehicle without drug to determine reversibility of the drug effect; (iii) in monkeys with unilateral disinsertion of the ciliary muscle, both eyes received drug after determination of vehicle-only baseline; (iv) drug-free baseline facility was again determined in both eyes of animals which had received a facility-effective drug dose unilaterally on the first occasion; this protocol was conducted four-to-six weeks after the first. The second and first baselines were then compared, corrected for any change in the control eye, to see if the drug effect had persisted.
Increasing concentrations of H-7 were tested to determine the approximate dose which yielded a statistically significant increase in outflow facility. Initially, each concentration of H-7 was prepared by dissolving the appropriate amount of H-7 in Barany's solution. For the treated eye, H-7 was administered in Barany's solution without DMSO as a vehicle, while the contralateral eye was administered vehicle alone. In separate experiments, 2 ml of H-7 solution was passed through the anterior chamber of the eye in a 10-15 minute period. Facility measurements were taken 60 minutes thereafter.
Figure 5A is a bar graph illustrating the effect of several concentrations of H-7 on facility. In Figure 5A, the difference between the ratio of Rx BL and 1.0 indicates the increase in facility in eyes administered H-7 (ipsilateral eyes) or vehicle alone (i.e. , control or contralateral eyes) over baseline measurements (BL = pre-treatment (baseline) and Rx = post- treatment with H-7 or vehicle alone). In Figure 5A, the "open" bars represent eyes administered vehicle only, while the "speckled" bars indicate eyes administered H-7. The numbers in parentheses represent the number of experimental animals, while * = p<0.05 and
** = p<0.001 are the probabilities that the double ratio defined above equals 1.0 by the 2- tailed paired t-test.
At a concentration of 10 μM, no increase in outflow facility was found. However, at a concentration of 100 μM, a borderline-significant effect was observed (data not shown). When the initial post-treatment values for both the treated and the control eyes were eliminated (thereby examining a longer exposure time) at a concentration of 100 μM H-7, a stronger and statistically-significant effect was observed. Administration of 300 μM H-7 provided a larger and more strongly significant increase in outflow facility. , effect. For this experiment, 300 μM H-7 was administered to nine monkeys with disinserted ciliary muscle in one eye, the contralateral eyes without disinsertion. As expected, the ciliary muscle-disinserted eyes had lower baseline facilities. However, both the ciliary muscle disinserted and the non-disinserted eyes exhibited essentially identical proportional increase in outflow facility following drug admimstration (Figure 5A). In Figure 5A, the "shaded" bar indicates ciliary muscle-disinserted eyes administered H-7, while the "speckled" bar represents normal eyes administered H-7. These data indicate that H-7 exerts its effect directly on the trabecular meshwork and not via the ciliary muscle. By comparison, pilocarpine exerts its effect via the ciliary muscle.
Finally, six-weeks after treatment, drug-free baseline facility was again determined in both eyes of animals which had received a facility-effective drug dose (100 or 300 μM) unilaterally on the first occasion. The second and first baselines were then compared, corrected for any change in the control eye, to see if the drug effect had persisted. As indicated on the far right in Figure 5 A (labeled 100/300), there was a significant increase in facility for the H-7 treated eyes as compared to the control eyes.
The results presented in Figure 5A indicate that H-7 induced a dramatic dose- dependent facility increase. As set forth in the preceding paragraph, this increase is due to a direct effect on the trabecular meshwork. These in vivo experiments provide important information regarding the dose-dependent and the pharmacologic behavior of H-7, demonstrating its potential usefulness as an anti-glaucoma agent in humans.
EXAMPLE 2
H-7 Does Not Significantly Effect Corneal Endothelial Cell Counts At A Concentration Effective At Increasing Total Outflow Facility
In this example, the effect of 300 μM H-7 on corneal endothelium was determined in cynomolgus monkeys via transcorneal intracameral injection.
The basic experimental protocol was as follows. Following administration of ketamine
( 10 mg/kg) anesthesia, slit lamp examination was performed to provide a baseline evaluation of the cornea, lens, and anterior chamber. Thereafter, the monkeys were anesthetized with IM pentobarbital-sodium (35 mg/kg). A 3 mM H-7 solution (2.1858 mg H-7 dissolved in 2 ml
Barany's solution, pH adjusted to 6-7) was manufactured, and 10 μl of that solution was e needle away from the area to be photographed during the transcorneal intracameral injection). Specular micrographs were taken at 1 and 3 hours after H-7 administration to determine corneal endothelial cell counts and morphology, and slit lamp examination was repeated, observing the cornea, lens, and anterior chamber for any changes from baseline. Finally, specular micrographs were performed again at two-three weeks after H-7 administration, allowing the determination of corneal endothelial cell counts and morphology.
Corneal endothelial cell counts were measured as the number of cells/mm2 and are reported in Table 3. In Table 3, BL= baseline cell counts; Rx(l) = cell counts 1 hour after H-7 administration, Rx(2) = cell counts 3 hours after H-7 administration, and Rx(3) = cell counts 2 weeks after H-7 administration; Treated = eyes administered H-7; Control - eyes administered vehicle only; and T/C = Treated/Control. As indicated by the data presented in Table 3, the specular micrographs did not reveal a statistical change in cell counts (n = 4, p = NS).
TABLE 3
Cell Count (cell/mm2)
T/C
Treated Control
BL 2328±51 2344154 0.99±0.02
Rx( l) 2481 ±98 275611 17 0.91 ±0.06
Rx(l )/BL 1.07±0.06 1.18+0.06 0.91 ±0.07
Rx(2) 25131131 2544133 0.9910.05 Rx(2)/BL 1.0810.07 1.0910.04 0.9910.04
Rx(3) 2569191 2525171 1 .0210.04 Rx(3)/BL 1.10±0.05 1.0810.05 1.0210.03
Similarly, no morphological changes in cells were observed. These results suggest that administration of outflow-facilitating doses of H-7 does not have a detrimental effect on corneal safety, at least in the young healthy monkey. Of course, this may or may not be the same situation in the older human population, which has an age-related decrease in endothelial cells.
It should be noted that the above-mentioned basic protocol for corneal endothelial cell counts will also be followed for other agents that show a positive effect on outflow facility. Moreover, determinations of corneal thickness, one measure of corneal endothelial cell function, can be performed to supplement these results. The Effect Of Topical Administration Of H-7 On Outflow Facility In Cynomolgus Monkeys
Generally speaking, agents used in the treatment of glaucoma are administered topically as an eyedrop, rather than being injected into the eye. This example describes the results on outflow facility following the topical administration (i.e., via an eyedrop) of four different concentrations of H-7.
Following administration of ketamine (10 mg/kg) anesthesia, slit lamp examination was performed on the monkeys. Thereafter, the monkeys were anesthetized with IM pentobarbital-sodium (35 mg/kg). Baseline facility was measured (7 periods) by 2-level constant pressure perfusion of the anterior chamber; thereafter, the perfusion system was closed.
Four H-7 solutions of differing concentrations were manufactured: (i) a 90 mM solution (-0.3 mg H7 dissolved in 10 μl of 10% DMSO with Barany's solution, pH of the resulting solution adjusted to approximately 7); (ii) a 150 mM solution (-1.1 mg H7 dissolved in 20 μl of 25% DMSO with Barany's solution, pH of the resulting solution adjusted to approximately 7); (iii) a 450 mM H-7 solution (-3.3 mg H7 dissolved in 20 μl of 25% DMSO with Barany's solution, pH of the resulting solution adjusted to approximately 7), and (iv) a 650 mM H-7 solution (~ 5.7 mg H-7). Thereafter, each concentration of H-7 solution was administered to the superior cornea of one eye and vehicle (25% DMSO) to the opposite eye (5 μl x 4 drops), waiting 90 seconds between each drop; in addition, the lower eyelid was lifted immediately after each drop to allow adequate drug/vehicle contact with the cornea. Two hours after H-7 administration, the perfusion system was reopened and facility was measured for 9 periods. The resulting data are presented in Figure 5B. In Figure 5B, the ratio of Rx/BL indicates the increase in facility in eyes administered H-7 (ipsilateral eyes) or vehicle alone (i.e., control or contralateral eyes) over baseline measurements (BL = pre-treatment (baseline) and Rx = post-treatment with H-7 or vehicle alone). The "open" bars represent eyes administered vehicle only, while the "speckled" bars indicate eyes administered H-7. The numbers in parentheses represent the number of experimental animals, while * = p<0.02 and
** = pO.OOl are the probabilities that the double ratio defined above will equal 1.0 by the 2- tailed paired t-test. - , ere was a sugges on o a sma e ec on ou ow ac y, u e effect was not statistically significant. By comparison, topical administration of 150 mM H-7 had a definite statistical effect on outflow facility. The administration of 450 and 650 mM had a larger effect, demonstrating that H-7 has a defined dose-response relationship when administered topically.
EXAMPLE 4
The Effect Of Topical Administration Of H-7 On Pupil And Accommodation In Cynomolgus Monkeys
This example examines the effect of topical administration of H-7 on pupil size and refraction in cynomolgus monkeys.
The experimental protocol was as follows. Following administration of ketamine (10 mg/kg) anesthesia, baseline slit lamp examination was performed on the six monkeys tested. Thereafter, the monkeys were anesthetized with IM pentobarbital-sodium (35 mg/kg). Baseline refraction (2-3 readings) and pupil size were then measured as described above. Subsequently, a 150 mM H-7 solution (1.0929 mg H7 dissolved in 20 μl of 25% DMSO with
Barany's solution, pH of the resulting solution adjusted to approximately 7) was administered to the cornea of one eye. and vehicle (25% DMSO) to the opposite eye (5 μl x 4 drops).
Beginning 45 minutes after H-7 administration, pupil size and refraction were evaluated every 15 minutes until 2 hours after the H-7 administration. A slit lamp examination was then performed to check for toxicity. The data are presented in Figures 6A and 6B.
Figure 6A graphically illustrates the effect of topical H-7 on pupil size, pupil diameter (mm) being indicated as a function of time (min). Time 0 represents drug administration, and the open squares represent eyes treated with H-7, while the open diamonds represent the control eyes. The results indicate that the control pupil gets smaller with prolonged anesthesia, while the pupil subjected to H-7 does not. The following statistical levels are depicted in Figure 6A: *= p<0.05, ** = p<0.02, and *** = p<0.005 by the 2-tailed paired t- test for differences between eyes unequal to 0.0.
Figure 6B graphically illustrates the effect of topical H-7 on refraction (diopters) indicated as a function of time (min). Again, time 0 represents drug administration, and the open squares represent eyes treated with H-7. while the open diamonds represent the control
S8 - . between the control eye and the treated eye.
Therefore, while H-7 did have an effect on pupil size, a concomitant effect on refraction was not observed in monkeys.
EXAMPLE 5
H-7 Prevents Pupillo-Constrictive Effect Of Pilocarpine, But Not Accommodative Effect Of Pilocarpine
This example is directed at determining whether selective inhibition of miosis (dependent on contraction of the iris sphincter muscle) as compared to accommodation (dependent on contraction of the ciliary muscle) can be achieved.
Figures 7A-H graphically depict the results of this experiment, and reference to those figures will also assist in understanding the experimental protocol. The experiments of this example examined the miotic response (Figures 7A, C, E, and G), determined by pupil diameter (mm), and the accommodative response (Figures 7B, D, F, and H), measured in diopters, to pilocarpine in the presence or absence of H-7 in normal living monkey eyes.
Each of the 4 rows of panels represents a different dose of H-7. In each row, the left-hand panel represents the pupillary diameter, while the right-hand panel represents the accommodative response in the same animals. Within each panel, "n" represents the number of animals for that protocol. In each experiment, baseline readings (BL) of the pupillary diameter were obtained with vernier calipers held just in front of the cornea, after which phenylephrine (2.5%) was topically administered to the eyes, resulting in mydriasis.
Thereafter, different protocols were used in the administration of H-7 to the animals. Six animals (Figures 7A and 7B) received 10 μL of 3 mM H-7 via transcorneal intracameral injection, resulting in a final anterior chamber concentration = 300 μM. The remaining animals received H-7 by intravitreal injection as follows: (i) four animals (Figures 7C and
7D) received 10 μL of 75 mM H-7; (ii) six animals (Figures 7E and 7F) received 20 μL of 75 mM H-7, and (iii) six animals (Figures 7G and 7H) received 20 μL of 150 mM H-7. all in 50% DMSO. The final intravitreal concentrations of H-7 were 300 μM, 600 μM. and 1.2 M, respectively. Contralateral control eyes received Barany's solution with 50% DMSO (Figures 7C-H) or without 50% DMSO (Figures 7A, B). Pupillary diameter and refraction
(accommodation being defined as refraction at a given time point minus the baseline me ure ap x ma e y - m nu es a er p eny ep r ne on e abscissae of the panels). H-7 was then administered, and pupil diameter and accommodation were measured 25 minutes later (H-7 on the graphs).
Thirty minutes following H-7 administration, pilocarpine (1.5 mg/kg) was infused IM into the thigh of the monkeys over a 10-minute period, insuring slowly rising but always equal pilocarpine concentrations reaching the iris sphincter and ciliary muscles of both eyes simultaneously. Pupillary diameter was then measured 30 minutes after pilocarpine administration (Pilo on the left-hand panels), while accommodation was measured every 5 minutes for -50 minutes beginning at the start of the IM infusion (Pilo on the right-hand panels). In Figures 7A-H, the solid lines connecting the solid squares indicate the eyes which will receive or have received H-7, while the solid lines connecting the open diamonds indicate the contralateral eyes of the same animals which received vehicle without H-7. The data in Figures 7A-H are mean±s.e.m. for n monkeys. The following significance levels are depicted in Figures 7A-H: *P<0.05; **P<0.01 ; ***P<0.001. The significance of the difference between treated and control pupil diameter (Figures 7A, 7E, 7G) or final accommodative response (Figure 7H) was determined by the 2-tailed paired t-test.
As depicted in Figures 7A-H, the data show that H-7 dilates the ipsilateral pupil even beyond the effect of pheny lephrine, and inhibits ipsilateral pilocarpine-induced miosis, at intracameral and intravitreal doses far lower than required for inhibition of accommodation. This is best shown in Figures 7A-B for intracamerally administered H-7 and Figures 7E-F and
7G-H for intravitreally administered H-7.
EXAMPLE 6
The Effect Of H-7 On Pilocarpine-induced Contraction Of The Ciliary Muscle
The experiments of this example were designed to determine the effect of H-7 on the ciliary muscle's contractile response to pilocarpine. The muscle chamber apparatus that has been previously described was used in these experiments.
Figures 8A-C depict chart recorder traces obtained from a single in vitro ciliary muscle strip maintained in Krebs solution. In the traces, which are read from left to right, muscle strip contraction is indicated in the downward direction per the scale indicated; more specifically, in the scale shown in Figure 8A, the length of the downward-pointing arrow indicates a contraction (tension) of 25 mg, while the length of the horizontal arrow pointing to . , contractile vector and C = circular contractile vector; Krebs = administration of Krebs solution without other drugs; pilo = administration of pilocarpine; and Pilo+H-7 = concomitant administration of pilocarpine and H-7 In Figure 8 A, a ciliary muscle strip under baseline tone was first exposed to 10 μM pilocarpine, then to 10 μM pilocarpine + 300 μM H-7. As indicated by the chart recorder trace, the initial addition of pilocarpine resulted in contraction. However, the addition of H-7 (i. e. , pilocarpine + H-7) resulted in inhibition of the initial pilocarpine contraction within 10 minutes of exposure. At the conclusion of the experiment, washout of the muscle strip was achieved by exposure of perfusand without drugs for a 90 minute period (indicated by the
"Krebs" designation in Figure 8A).
Subsequent to the washout period, the same ciliary muscle strip was exposed to 10 μM of pilocarpine. As shown by the chart recorder trace in Figure 8B, such exposure elicited no contraction in either vector. Finally, in the same ciliary muscle strip following a further 60 min of washout (now 2.5 hours after removal of H-7), 10 μM of pilocarpine now elicited sluggish contraction in both vectors; this is shown by the chart recorder trace in Figure 8C. The experiments described above suggest that the effects of H-7 are relatively long- lasting, even after its removal from the perfusand. This finding also suggests that the in vivo dissociation of miosis and accommodation in response to an intracameral or intravitreal infusion of H-7 (300 μM) may have a pharmacokinetic basis. Based on the data from this one concentration, H-7 appears to have no selectivity for one vector over the other; however, vector-selectivity might be observed at other concentrations.
EXAMPLE 7
Latrunculin A Inhibition Of Cell Contractility
The experiments in this example were conducted to establish probable dose and time course for latrunculin A for subsequent in vivo studies on living monkeys.
An established line of bovine aortic endothelial cells were plated at half confluency on clean glass coverslips and cultured in DMEM, containing 1 g of glucose, 9% bovine calf serum, 100 mg penicillin, 100 mg streptomycin per 550 ml, for 2-4 days at 37°C , 7% CO2/93% O,, 90%) humidity. The medium was then changed to one containing Latrunculin A concentrations of 0.02, 0.05. 0.2, 0.5, or 2.0 μM for 1 , 2. 3, 5, or 24 hours, after which the er, er ze w . r on - n , and fixed with 3% paraformaldehyde. The medium was then exposed to rhodamine phalloidin (for fluorescent labelling of actin), or to rabbit pan-cadherin primary antibody followed by goat anti-rabbit secondary antibody (for immunofluorescent labelling of cadherin, a major protein associated with cell-cell adherens junctions); and examined by fluorescence microscopy.
In associated experiments to judge the ability of the cells to recover from the drug, after exposure to drug for various concentrations and times (based on the previous experiments), the medium was changed to an identical one without drug for 15 minutes, 1 hour, 5 hours, 18 hours or 22 hours, and the cells washed, fixed, labelled, and examined as above. Structural alterations were found in both the actin filaments and the cell-cell adhesions (in summary, disruption of the actin filaments within the cells and separation of cells from their neighbors) which were both drug-dose and exposure time-dependent; the higher the dose and the longer the exposure time, the more pronounced the effects. Analogously, recovery was also dose- and time-dependent; the lower the dose, the shorter the exposure time, and the longer the drug-free interval, the more normal the appearance.
Latrunculin A concentrations greater than 0.5 μM had unequivocal effects after 1 hour of exposure, with partial recovery evident within 1-5 hours after removal of the drug. Lower concentrations had more subtle slower onset effects. Based on these findings, doses of 0.2, 0.5, 2.0, and 5.0 μM were studied in living monkeys, with the positive findings as described below. It is quite likely, albeit not yet tested, that lower concentrations for longer exposure times might also be effective.
EXAMPLE 8
Dose-Dependent Effects Of Latrunculin A, Staurosporine, And Swinholide A On Total Outflow Facility
This example involved an examination of the dose-dependent effect of latrunculin A, staurosporine, and swinholide A on total outflow facility.
The experiments of this example were performed on adult cynomolgus and rhesus monkeys. Both normal monkeys and monkeys with unilateral ciliary muscle disinsertion from the scleral spur/trabecular meshwork were utilized. Briefly, the animals were anesthetized with ketamine (10 mg/kg) followed by pentobarbital sodium (35 mg/kg). perfusion of the anterior chamber of both eyes simultaneously with Barany's solution, as described above. The anterior chamber of both eyes was cannulated with a single branched 26-gauge needle; one arm of the needle was connected by polypropylene tubing to a continuously-weighed reservoir of Barany's solution, while the other arm was connected to a pressure transducer. Facility measurements were taken for 35-45 minutes. With reservoirs open, 10 μl of either 5 or 20 μM latrunculin A or 100 or 300 nM of swinholide A were injected into the inflow tubing in one eye (0.5 μM, 2.0 μM, 10 nM, and 30 nM, respectively, in the 100 μl anterior chamber), vehicle in the other, via a custom-designed micro-t-piece (positioned 2-3 cm for the eye) and a micrometer syringe. After five minutes to allow the perfusand from an elevated reservoir to wash into the anterior chamber and 3 minutes for convection mixing of the anterior chamber by blowing cold air on the cornea, facility measurements were taken for 90 minutes.
Exchange Infusion: The anterior chamber of each eye was cannulated with a branched needle connected to a reservoir and an unbranched 26 gauge needle with the tubing clamped off. Baseline facility measurements were taken for 35-45 minutes. The clamped tubing from the unbranched needle was then connected to syringes containing either drug (0.2, 0.5, 2.0, and 5.0 μM latrunculin A, 0.1 , 1.0 or 10 μM staurosporine; and 10 or 30 nM swinholide A) or vehicle. The vehicles used were as follows: i) latrunculin A: 0.01% DMSO for 0.2 μM and 2.0 μM, 0.25% DMSO for 0.5 μM, and 0.025% DMSO for 5.0 μM; ii) staurosporin:
0.001% DMSO for 0.1 μM, 0.1% DMSO for 1.0 μM, and 1% DMSO for 10 μM; and iii) swinholide A: 0.1 % DMSO for 10 and 30 nM. The monkeys with unilateral ciliary muscle disinsertion received drug in both eyes. The syringes were placed in a variable-speed infusion pump and the reservoir (inflow) tubing disconnected, allowing infusion of 2.0 ml of solution through the anterior chamber over 10-15 minutes (maintaining intraocular pressure at
- 15 mm Hg).
The reservoirs were emptied and filled with the same solution being perfused through the eye. The reservoir tubing was then reconnected and the syringe tubing clamped. For latrunculin A and swinholide A, outflow facility measurements were immediately taken for 90 minutes. For staurosporine, the reservoirs were closed to inflow for 45 minutes, then opened and facility measurements were taken for 45 minutes. For the latrunculin A reversibility exchanges, an exchange was done with 5.0 μM in one eye and vehicle in the other followed
- 42 y a secon exc ange w t ve c e g ve to ot eyes. ter eac exc ange, ac ty measurements were taken for 60 minutes.
Latrunculin A
With regards to latrunculin A, only exchange infusion induced a dose-dependent facility increase at 0.2, 0.5, 2.0, and 5.0 μM [8 ± 14% (n = 4, NS); 50±17% (n=8, p<0.025);
141 ± 23% (n = 9, pO.OOl); and 329 + 1 19% (n = 6, p<0.05); respectively] in normal monkeys. These data are graphically depicted in Figures 9A-D (Fig. 9A: 0.2 μM; Fig. 9B: 0.5 μM; Fig. 9C: 2.0 μM; and Fig. 9D: 5.0 μM), which show facility as a function of time, drug being administered at time = 0. The following designations apply in those figures: BL Trt = pre-drug administration in eyes to receive drug (open squares with solid lines); BL Cont
= pre-drug administration in eyes to receive vehicle without drug (open circles with dashed lines); p rx Trt = post-drug administration in eyes which received drug (open squares with dashed lines); and p rx Cont = post-drug administration in eyes which received vehicle without drug (open circles with dashed lines). It should be noted that the bolus infusion of latrunculin A was probably ineffective because an effective drug concentration was not maintained for sufficient time. The data are also presented in Table 4, analyzed by 30-minute post-drug time intervals where the average values for each eye are themselves averaged to give group means for the 30-minute intervals.
TABLE 4
Latrunculin Exchange Facilities By 30 Minute Intervals
Facility (/ιL/min/mmHg)
Dose Time Rx/Cont (Rx BL)/(Cont/BL)
Rx Cont
BL 0.3810.04 0.4910.09 0.8010.09
30 min 0.5010.05 0.59+0.1 1 0.9010.12 1.1310.12
0.2μM (n=4)
60 min 0.6510.1 1 0.7910.12 0.8510.13 1.0610.14
90 min 0.7910.19 0.96+0.16 0.8510.18 1 .0610.20
BL 0.3210.05 0.3610.09 1.0210.13
30 min 0.4510.08 0.4710.13 1.2 )10.19 1.1710.18
0.5μM (n-8)
60 min 0.7610.15 0.5910.16 1.5110.23 1 .5310.18-
90 min 1.0710.26 0.6910.17 I .7110.291 1.7710.26 -
BL 0.3310.03 0.42+0.06 0.8510.09
30 min 0.5810.09 0.4510.05 1.3310.13£ l .6110.15+
2.0μM (n-9)
60 min 1.0910.17 0.58+0.08 1.9110.19 2.3310.22:
90 min 1 .7410.30 0.6910.1 1 2.5610.29 3.12±0.36;
BL 0.2810.06 0.2910.09 1.1310.13
30 min 0.9310.30 0.3910.13 2.6910.51 2.3410.30'
5.0μM (n-6)
60 min 2.0010.61 0.5010.20 6.01+2.10 4.8311.30*
90 min 2.5810.80 0.6610.30 7.8913.15 6.1412.02
BL, 0.3210.03 0.39+0.05 0.95+0.08
Baseline
Comparisons BL2 0.4310.09 0.4210.06 1.1810.26
(n=14)
BLJBL, 1.3810.28 1.2010.13 1.2610.22
BL=Baseline: Cont=Control eye; Rx=Drug treated eye. Data are meanlSEM for n animals, Post-drug data encompasses 30 min time windows ending at the post-drug time indicated, i.e., "30 min"=0- 30 min post-drug administration, etc. £p<0.05; yp<0.01 ; -p<0.25; fp<0.005; J<0.0001 for ratios different from 1.0 by the 2-tailed paired t-test.
In addition, latrunculin A was equally effective in ciliary muscle-disinserted eyes, showing a facility increase of 353±129% (n=9, p<0.05), and the control eyes showing a facility increase of 328+35% (pO.OOl , at the 5.0 μM dose). These data, depicted in Figure 9E and presented in Table 5, indicate that latrunculin A has a direct trabecular meshwork effect rather than a secondary ciliary muscle-mediated action. TABLE 5 Latrunculin A - Ciliary Muscle Disinsertion
Facility (μl/min/mmHg)
Dose Time CM Dis/Cont
CM Dis Cont
BL 0.13+0.01 0.38+0.09 0.4510.08
5.0μM OU (n=9) PRx 0.6510.23 1 .51+0.30 0.4310.15
PRx/BL 4.5311.29 4.2810.35 1.0610.27
BL=Baseline; PRx=Post drug administration; CM Dis=ciliary muscle disinsertion eye; Cont=Control eye. Data are meanlSEM for n animals with unilateral CM disinsertion receiving 5.0μM Lat-AOU.
Reversibility of the facility-effect of latrunculin A (5.0 μM) was minimal for the first hour after removal of the drug, as depicted in Figure 10. Referring to Figure 10, each data point is the mean of the facility readings at that time for n monkeys, drug administration being at time = 0. The following designations apply: baseline treated = pre-drug administration in eyes which will receive drug (open squares with solid lines); baseline control = pre-drug administration in eyes which will receive vehicle without drug (open circles with dashed lines); post drug #l/#2 treated = post-drug administration after the first/second exchange in eyes which received drug (open squares with dashed lines); post drug #l/#2 control = post-drug administration after the first/second exchange in eyes which received vehicle without drug (open circles with dashed lines).
The minimal reversibility exhibited by latrunculin A was probably due to the washout of resistance-relevant material when the drug was present which does not reform quickly following drug removal. Indeed, it may not reform by the next perfusion, as indicated by the baseline comparisons.
Staurosporine
Exchange infusion of staurosporine produced a dose-dependent facility increase at 0.1 μM, 1 M. and 10 μM [-2 ± 3% (n = 4, NS); 68 ± 24% (n = 7, p<0.05); and 168 ± 48% (n = 8, p<0.01 ); respectively]. These data are graphically depicted in Figures 11A-C (Fig. 1 1 A: 0.1 μM; Fig. 1 IB: 1.0 μM; Fig. 1 I C: 10.0 μM), which show facility as a function of time, drug being administered at time = 0. The same designations described above for Figures 9A- D also apply for Figures 1 lA-C. In addition, these data are presented in Table 6, where the average value for each eye are themselves averaged to give group means for the entire time period. TABLE 6
Staurosporin Exchange Facilities
Facility (μl min/mmHg)
Dose Time Rx/Cont (Rx/BL)/(Cont/BL)
Rx Cont
BL 0.30+0.09 0.2910.09 1 .04+0.14
0.1 μM (n-4)
PRx 0.2710.07 0.2810.08 1.0310.17 0.9810.04
BL 0.2810.05 0.30+0.05 0.9610.1 1 l .OμM (n=8)
PRx 0.4910.10 0.3310.05 1.57+0.29 1.6810.24'
BL 0.3210.05 0.4410.08 0.8110.12 lOμM (n-8)
PRx 0.7410.18 0.3910.08 1.9910.43 2.68+0.487
3L=Baseline; PRx=Post drug administration; Rx=Treated drug eye; Cont=Control eye. Post-drug data encompasses 45 min, beginning 45 min after drug administration. Data are meanlSEM for n animals. £p<0.05; ρ<0.01 for ratios different from 1.0 by the 2-tailed paired t-test. Swinholide A
Swinholide A doses of 10 and 30 nM did not increase facility, whether administered by bolus or exchange infusion (data not shown). It should be noted, however, that in vitro cell culture experiments with swinholide A did reveal an effect on the cells' actin filaments and cell-cell adhesion. Indeed, those experiments suggested that the dosage range of swinholide A tested in the in vivo monkey experiments described in this example would be at the low end of the dose-duration-efficacy relationship.
Therefore, as previously indicated, the apparent ineffectiveness of swinholide A may be due to inadequate dose or exposure time, not to the compound itself. Conversely, this apparent ineffectiveness may not have a pharmacokinetic basis. Based on the results of these experiments, latrunculin A and staurosporine increased outflow facility. Though swinholide A did not appear to be effective, these are likely due to the testing conditions employed. In addition to being potentially advantageous agents in the treatment of glaucoma, the results of the latrunculin A and staurosporine experiments indicate that actin cytoskeleton disorganization in the trabecular meshwork can increase outflow facility in monkeys.
From the above, it should be evident that the present invention provides for new agents for the potential treatment of glaucoma. These agents, which act via pharmacological interference with cell adhesions and the associated actin filaments in the ocular fluid drainage pathways, present a clearly needed alternative to the pharmacological modalities currently employed.

Claims

We claim:
1. A method of enhancing aqueous humor outflow in the eye of a subject to treat glaucoma, comprising: a) providing a subject with glaucoma; and b) administering to said subject an ophthalmic preparation comprising an effective amount of a non-corneotoxic compound selected from the group consisting of serine-threonine kinase inhibitors, tyrosine kinase inhibitors, protein tyrosine phosphatase inhibitors, and an actin-disrupting compound selected from latrunculin A, latrunculin B and swinholide A. thereby enhancing aqueous humor outflow in the eye and treating the glaucoma.
2. The method of Claim 1 , wherein said serine-threonine kinase inhibitor is selected from the group consisting of l-(5-isoquinolinyl-sulfonyl)-2-methylpiperazine; l-(5- iodonaphthalene- 1 -sulfonyl)- 1 H-Hexahydro- 1 ,4-diazepine; (8R*,9S*,\ 15*)-(-)9-hydroxy-9- methoxycarbonyl-8-methyl-14-«-propoxy-2,3,9,10-tetrahydro-8,l l -epoxy, \H,8H, WH-
2,7b, 1 la-triazadibenzo[α,g]cycloocta[cfife] trinden-1-one; and staurosporine.
3. The method of Claim 1, wherein said tyrosine kinase inhibitor is selected from the group consisting of 3,5-diisopropyl-4-hydroxybenzylidene, N-benzyl-3,4- dihydroxybenzylidene thioacetamide, N-(3-phenylpropyl)-3,5-diisopropyl-4-hydroxy benzylidene acetamide, and N-(4-phenylpropyl)-3,4-diisopropyl-4-hydroxy benzylidene acetamide.
4. The method of Claim 1 , wherein said protein tyrosine phosphatase inhibitor is selected from the group consisting of vanadate and phenylarsin oxide.
5. The method of Claim 1 or 2 or 3 or 4, wherein said administration is topical.
6. The method of Claim 1 or 2 or 3 or 4, wherein said administration is intracameral. . , intracanalicular.
8. An ophthalmic composition, comprising an effective amount of a compound capable of enhancing aqueous humor outflow in the eye of a subject to treat glaucoma and a pharmaceutically acceptable carrier, said compound selected from the group consisting of serine-threonine kinase inhibitors, tyrosine kinase inhibitors, protein tyrosine phosphatase inhibitors, latrunculin A, latrunculin B, and swinholide A.
9. The composition of Claim 8, wherein said serine-threonine kinase inhibitor is selected from the group consisting of l-(5-isoquinolinyl-sulfonyl)-2-methylpiperazine; l -(5- iodonaphthalene- 1 -sulfonyl)- 1 H-Hexahydro- 1 ,4-diazepine; (8R*,9S*, 1 15*)-(-)9-hydroxy-9- methoxycarbonyl-8-methyl-14-w-propoxy-2,3,9,10-tetrahydro-8,l 1 -epoxy, \H,8H, 1 1H- 2,7b, 1 la-triazadibenzo[α,g]cycloocta[c fe] trinden-1-one; and staurosporine.
10. The composition of Claim 8, wherein said tyrosine kinase inhibitor is selected from the group consisting of 3,5-diisopropyl-4-hydroxybenzylidene, N-benzyl-3,4- dihydroxybenzylidene thioacetamide, N-(3-phenylpropyl)-3,5-diisopropyl-4-hydroxy benzylidene acetamide, and N-(4-phenylpropyl)-3,4-diisopropyl-4-hydroxy benzylidene acetamide.
1 1. The composition of Claim 8, wherein said protein tyrosine phosphatase inhibitor is selected from the group consisting of vanadate and phenylarsin oxide.
12. Use of a compound selected from the group consisting of serine-threonine kinase inhibitors, tyrosine kinase inhibitors, protein tyrosine phosphatase inhibitors, latrunculin A, latrunculin B, and swinholide A, for the preparation of an ophthalmic composition for enhancing aqueous humor outflow in the eye of a subject to treat glaucoma. . , - selected from the group consisting of l-(5-isoquinolinyl-sulfonyl)-2-methylpiperazine; l-(5- iodonaphthalene- 1 -sulfonyl)- 1 H-Hexahydro- 1 ,4-diazepine; (8R*,9S* , 1 15'*)-(-)9-hydroxy-9- methoxycarbonyl-8-methyl-14-«-propoxy-2,3,9,10-tetrahydro-8,l 1-epoxy, 1H,8H, 1 1H- 2,7b, 1 la-triazadibenzo[α,g]cycloocta[ccfe] trinden-1-one; and staurosporine.
14. Use according to Claim 12, wherein said tyrosine kinase inhibitor is selected from the group consisting of 3,5-diisopropyl-4-hydroxybenzylidene, N-benzyl-3,4- dihydroxybenzylidene thioacetamide, N-(3-phenylpropyl)-3,5-diisopropyl-4-hydroxy benzylidene acetamide, and N-(4-phenylpropyl)-3,4-diisopropyl-4-hydroxy benzylidene acetamide.
15. Use according to Claim 12, wherein said protein tyrosine phosphatase inhibitor is selected from the group consisting of vanadate and phenylarsin oxide.
16. Use according to Claim 12 or 13 or 14 or 15, wherein said composition is for topical administration.
17. Use according to Claim 12 or 13 or 14 or 15, wherein said composition is for intracameral administration.
18. Use according to Claim 12 or 13 or 14 or 15, wherein said composition is for intracanalicular administration.
19. An ophthalmic composition, comprising an effective amount of a compound capable of enhancing aqueous humor outflow in the eye of a subject to treat glaucoma and a pharmaceutically acceptable carrier, said compound selected from the group consisting of Al- adenosine agonists, acepromazine, promazine, aceclidine, lysophosphatidic acid, genistein; endothelin-, transforming growth factor, interleukin- 1, platelet activating factor, and 3α,5β- tetrahydrocortisol.
PCT/US1997/002709 1996-02-21 1997-02-21 Pharmaceutical compositions for the treatment of glaucoma WO1997030701A2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
AU20535/97A AU2053597A (en) 1996-02-21 1997-02-21 Pharmaceutical compositions for the treatment of glaucoma
IL12573597A IL125735A (en) 1996-02-21 1997-02-21 Pharmaceutical compositions for the treatment of glaucoma comprising an inhibitor of serine-threonine kinase, tyrosine kinase or protein tyrosine phosphatase or an actin-disrupting compound

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US08/604,568 1996-02-21
US08/604,568 US5798380A (en) 1996-02-21 1996-02-21 Cytoskeletal active agents for glaucoma therapy

Publications (2)

Publication Number Publication Date
WO1997030701A2 true WO1997030701A2 (en) 1997-08-28
WO1997030701A3 WO1997030701A3 (en) 1998-02-26

Family

ID=24420147

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1997/002709 WO1997030701A2 (en) 1996-02-21 1997-02-21 Pharmaceutical compositions for the treatment of glaucoma

Country Status (4)

Country Link
US (2) US5798380A (en)
AU (1) AU2053597A (en)
IL (1) IL125735A (en)
WO (1) WO1997030701A2 (en)

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5919813A (en) * 1998-03-13 1999-07-06 Johns Hopkins University, School Of Medicine Use of a protein tyrosine kinase pathway inhibitor in the treatment of diabetic retinopathy
US5980929A (en) * 1998-03-13 1999-11-09 Johns Hopkins University, School Of Medicine Use of a protein tyrosine kinase pathway inhibitor in the treatment of retinal ischmemia or ocular inflammation
US6028099A (en) * 1998-03-13 2000-02-22 John Hopkins University, School Of Medicine Use of an inhibitor of the protein tyrosine kinase pathway in the treatment of choroidal neovascularization
WO2000021531A1 (en) * 1998-10-13 2000-04-20 Kyowa Hakko Kogyo Co., Ltd. Remedies for ocular diseases
US6399655B1 (en) 1998-12-22 2002-06-04 Johns Hopkins University, School Of Medicine Method for the prophylactic treatment of cataracts
EP1034793A4 (en) * 1998-08-17 2003-07-09 Senju Pharma Co Preventives/remedies for glaucoma
WO2005063252A1 (en) * 2003-12-22 2005-07-14 Alcon, Inc. Cdk2 antagonists as short form c-maf transcription factor antagonists for treatment of glaucoma
WO2006061525A2 (en) * 2004-12-09 2006-06-15 Lionel Bueno Compositions for treating surface ocular pathologies
WO2007002670A2 (en) * 2005-06-28 2007-01-04 Bausch & Lomb Incorporated Method of lowering intraocular pressure
US7320974B2 (en) 2005-03-25 2008-01-22 Inspire Pharmaceuticals, Inc. Cytoskeletal active compounds, compositions and use
WO2008079980A1 (en) * 2006-12-22 2008-07-03 Alcon Research, Ltd. Inhibitors of protein kinase c-delta for the treatment of glaucoma
WO2008078762A1 (en) 2006-12-26 2008-07-03 Santen Pharmaceutical Co., Ltd. Novel n-(2-aminophenyl)benzamide derivative having an urea structure
WO2008123395A1 (en) 2007-03-28 2008-10-16 Santen Pharmaceutical Co., Ltd. Ocular hypotensive agent comprising compound capable of inhibiting histone deacetylase as active ingredient
WO2009142321A1 (en) 2008-05-23 2009-11-26 参天製薬株式会社 Novel thiophenediamine derivative having urea structure
US8012972B2 (en) 2007-03-28 2011-09-06 Santen Pharmaceutical Co., Ltd. Pyridinecarboxylic acid (2-aminophenyl) amide derivative having urea structure
US20120088776A1 (en) * 2009-06-02 2012-04-12 Claudiu Supuran Treatment of mammalian disorders mediated by alpha-anhydrase isoforms
WO2015007896A1 (en) * 2013-07-18 2015-01-22 Neuroptis Biotech Method for producing a 1-(5-halonaphthalene-1-sulfonyl)-1h-hexahydro-1,4-diazepine and composition comprising same
EP3034081A1 (en) * 2014-12-18 2016-06-22 Neuroptis Biotech Treatment of an inflammation of the tarsal gland of the eyelid

Families Citing this family (48)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0868186B1 (en) 1995-12-21 2005-03-02 Alcon Laboratories, Inc. Use of certain isoquinolinesulfonyl compounds for the treatment of glaucoma and ocular ischemia
MXPA02008487A (en) * 2000-02-29 2002-12-13 Alcon Lab Inc Diagnostics and therapeutics for glaucoma.
US7708711B2 (en) 2000-04-14 2010-05-04 Glaukos Corporation Ocular implant with therapeutic agents and methods thereof
US20020143284A1 (en) * 2001-04-03 2002-10-03 Hosheng Tu Drug-releasing trabecular implant for glaucoma treatment
US7867186B2 (en) 2002-04-08 2011-01-11 Glaukos Corporation Devices and methods for treatment of ocular disorders
AU2002258754B2 (en) 2001-04-07 2006-08-17 Glaukos Corporation Glaucoma stent and methods thereof for glaucoma treatment
US7431710B2 (en) 2002-04-08 2008-10-07 Glaukos Corporation Ocular implants with anchors and methods thereof
US7678065B2 (en) 2001-05-02 2010-03-16 Glaukos Corporation Implant with intraocular pressure sensor for glaucoma treatment
AU2002305400A1 (en) 2001-05-03 2002-11-18 Glaukos Corporation Medical device and methods of use for glaucoma treatment
US7331984B2 (en) 2001-08-28 2008-02-19 Glaukos Corporation Glaucoma stent for treating glaucoma and methods of use
US7163543B2 (en) * 2001-11-08 2007-01-16 Glaukos Corporation Combined treatment for cataract and glaucoma treatment
US7951155B2 (en) 2002-03-15 2011-05-31 Glaukos Corporation Combined treatment for cataract and glaucoma treatment
WO2003080659A1 (en) * 2002-03-27 2003-10-02 Theratechnologies Inc. Methods and compounds for prevention and treatment of elevated intraocular pressure and related conditions
US9301875B2 (en) 2002-04-08 2016-04-05 Glaukos Corporation Ocular disorder treatment implants with multiple opening
WO2003092693A1 (en) * 2002-05-06 2003-11-13 Washington University Methods of treatment of glaucoma and other conditions mediated by nos-2 expression via inhibition of the egfr pathway
US20040167076A1 (en) * 2003-02-14 2004-08-26 Stamer W. Daniel Modulation of aqueous humor outflow by targeting vascular-endothelial-cadherin in schlemm's canal cells
JP4110324B2 (en) * 2003-10-15 2008-07-02 宇部興産株式会社 New indazole derivatives
WO2005079836A1 (en) 2004-02-18 2005-09-01 Wisconsin Alumni Research Foundation Method for treating glaucoma
WO2005079829A2 (en) * 2004-02-18 2005-09-01 Wisconsin Alumni Research Foundation Method for treating glaucoma
ZA200704959B (en) * 2004-12-27 2009-04-29 Alcon Inc Aminopyrazine analogs for treating glaucoma and other rho kinase-mediated diseases
ES2580108T3 (en) * 2005-07-11 2016-08-19 Aerie Pharmaceuticals, Inc Isoquinoline compounds
US7867999B1 (en) 2005-12-22 2011-01-11 Alcon Research, Ltd. Hydroxyamino- and amino-substituted pyridine analogs for treating rho kinase-mediated diseases and conditions
RU2008130111A (en) * 2005-12-22 2010-01-27 Алькон Рисерч, Лтд. (Us) (INDAZOL-5-IL) Pyrazines and (1,3-dihydroindole-2-OH) Pyrazines for the Treatment of Mediated RNA KINASE DISEASES AND CONDITIONS
US7785624B2 (en) * 2006-03-02 2010-08-31 Inspire Pharmaceuticals, Inc. Pharmaceutical latrunculin formulations
US7414137B2 (en) * 2006-03-23 2008-08-19 Inspire Pharmaceuticals, Inc. Process for the preparation of 3,4-disubstituted-thiazolidin-2-ones
ES2729424T3 (en) 2006-09-20 2019-11-04 Aerie Pharmaceuticals Inc Rho kinase inhibitors
AU2007319383A1 (en) 2006-11-10 2008-05-22 Glaukos Corporation Uveoscleral shunt and methods for implanting same
US20100022517A1 (en) * 2006-12-18 2010-01-28 Richards Lori A Ophthalmic formulation of rho kinase inhibitor compound
AR064420A1 (en) * 2006-12-21 2009-04-01 Alcon Mfg Ltd OPHTHALMIC PHARMACEUTICAL COMPOSITIONS THAT INCLUDE AN EFFECTIVE AMOUNT OF ANALOGS OF 6-AMINOIMIDAZO [1,2B] PIRIDAZINAS, USEFUL FOR THE TREATMENT OF GLAUCOMA AND / OR CONTROL THE NORMAL OR ELEVATED INTRAOCULAR PRESSURE (IOP).
US8455513B2 (en) 2007-01-10 2013-06-04 Aerie Pharmaceuticals, Inc. 6-aminoisoquinoline compounds
US8455514B2 (en) * 2008-01-17 2013-06-04 Aerie Pharmaceuticals, Inc. 6-and 7-amino isoquinoline compounds and methods for making and using the same
US20100152646A1 (en) * 2008-02-29 2010-06-17 Reshma Girijavallabhan Intravitreal injection device and method
US8450344B2 (en) 2008-07-25 2013-05-28 Aerie Pharmaceuticals, Inc. Beta- and gamma-amino-isoquinoline amide compounds and substituted benzamide compounds
WO2010029350A1 (en) * 2008-09-09 2010-03-18 University Of East Anglia Treatment of fibrotic eye disorders
CA2929545C (en) 2009-05-01 2019-04-09 Aerie Pharmaceuticals, Inc. Dual mechanism inhibitors for the treatment of disease
US10206813B2 (en) 2009-05-18 2019-02-19 Dose Medical Corporation Implants with controlled drug delivery features and methods of using same
US20130184318A1 (en) * 2010-04-30 2013-07-18 The Regents Of The University Of California Modulating compliance of trabecular meshwork
EP4193907A1 (en) 2011-09-13 2023-06-14 Glaukos Corporation Intraocular physiological sensor
US9730638B2 (en) 2013-03-13 2017-08-15 Glaukos Corporation Intraocular physiological sensor
HUE061618T2 (en) 2013-03-15 2023-07-28 Aerie Pharmaceuticals Inc Compound for use in the treatment of ocular disorders
WO2017040853A1 (en) 2015-09-02 2017-03-09 Glaukos Corporation Drug delivery implants with bi-directional delivery capacity
US11564833B2 (en) 2015-09-25 2023-01-31 Glaukos Corporation Punctal implants with controlled drug delivery features and methods of using same
AU2015414743B2 (en) 2015-11-17 2019-07-18 Alcon Inc. Process for the preparation of kinase inhibitors and intermediates thereof
US9643927B1 (en) 2015-11-17 2017-05-09 Aerie Pharmaceuticals, Inc. Process for the preparation of kinase inhibitors and intermediates thereof
EP3442479A1 (en) 2016-04-20 2019-02-20 Harold Alexander Heitzmann Bioresorbable ocular drug delivery device
CA3035566A1 (en) 2016-08-31 2018-03-08 Aerie Pharmaceuticals, Inc. Ophthalmic compositions
JP2020515583A (en) 2017-03-31 2020-05-28 アエリエ ファーマシューティカルズ インコーポレイテッド Aryl cyclopropyl-amino-isoquinolinyl amide compound
AU2019337703B2 (en) 2018-09-14 2023-02-02 Aerie Pharmaceuticals, Inc. Aryl cyclopropyl-amino-isoquinolinyl amide compounds

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4343794A (en) * 1980-05-06 1982-08-10 Mt. Sinai Method of reducing intraocular pressure with salts of vanadic acid
US4863912A (en) * 1986-05-19 1989-09-05 New York Medical College Use of tetrahydrocortisol in glaucoma therapy
US4997826A (en) * 1986-05-19 1991-03-05 New York Medical College Tetrahydrocortisol in glaucoma therapy

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4170398A (en) * 1978-05-03 1979-10-09 Koester Charles J Scanning microscopic apparatus with three synchronously rotating reflecting surfaces
US4757089A (en) * 1985-06-14 1988-07-12 Massachusetts Eye And Ear Infirmary Increasing aqueous humor outflow
US5306731A (en) * 1985-06-14 1994-04-26 Massachusetts Eye And Ear Infirmary Method and products for treating the eye

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4343794A (en) * 1980-05-06 1982-08-10 Mt. Sinai Method of reducing intraocular pressure with salts of vanadic acid
US4863912A (en) * 1986-05-19 1989-09-05 New York Medical College Use of tetrahydrocortisol in glaucoma therapy
US4997826A (en) * 1986-05-19 1991-03-05 New York Medical College Tetrahydrocortisol in glaucoma therapy

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
"GOODMAN and GILMAN's The Pharmacological Basis of Therapeutics, 7th Ed." 1985 , MACMILLAN PUBLISHING COMPANY , NEW YORK XP002040802 see page 107 - page 109 *
CURR.EYE RES., vol. 14, 1995, pages 777-781, XP002040795 M.A.CROFT ET AL.: "Effect of daily topical ethacrynic acid on aqueous humor dynamics in monkeys" *
EXP. EYE RES., vol. 45, no. 1, 1987, pages 45-56, XP002040797 N. YOSHIMURA ET AL.: "Analysis of Protein Kinase Activities in Rabbit Ciliary Processes: Identification and Characterization Using Exogenous Substrates" *
INVEST.OPHTH.VISUAL SCIENCE, vol. 16, no. 1, 1977, pages 47-53, XP002040794 P.L.KAUFMAN ET AL.: "Cytochalasin B reversibly increases outflow facility in the eye of the cynomolgus monkey" *
INVEST.OPHTHALMOL.VIS.SCI., vol. 33, 1992, pages 2631-2640, XP002040796 K. ERICKSON-LAMY ET AL.: "Ethacrynic Acid Induces Reversible Shape and Cytoskeletal Changes in Cultured Cells" *
INVESTIGATIVE OPHTHALMOLOGY & VISUAL SCIENCE, vol. 37, no. 3, 15 February 1996, page S202 XP002040799 B. TIAN ET AL.: "THE PROTEIN KINASE INHIBITOR H-7 INCREASES OUTFLOW FACILITY AND INHIBITS MIOTIC BUT NOT ACCOMODATIVE RESPONSES TO PILOCARPINE IN MONKEYS" *
INVESTIGATIVE OPHTHALMOLOGY & VISUAL SCIENCE, vol. 38, no. 4 Part 1-2, 1997, page S1150 XP002040801 H.I.CALVIN ET AL.: "INHIBITION OF MOUSE BSO CATARACTS BY THE PROTEIN KINASE INHIBITOR, H-7" *
J.CELL SCI., vol. 107, no. 3, 1994, pages 683-692, XP002040798 S. CITI ET AL.: "Cytoskeletal involvement in the modulation of cell-cell junctions by the protein kinase inhibitor H-7" *
JAPANESE JOURNAL OF PHYSIOLOGY, vol. 45, no. Suppl. 1, 1995, page S184 XP002040800 H.TODA ET AL.: "EFFECT OF PROTEIN KINASE INHIBITORS INJECTED LOCALLY INTO THE CAT EXTRA-STRIATE CORTEX ON PROLONGED POTENTIATION OF OCULAR CONVERGENCE" *

Cited By (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5919813A (en) * 1998-03-13 1999-07-06 Johns Hopkins University, School Of Medicine Use of a protein tyrosine kinase pathway inhibitor in the treatment of diabetic retinopathy
US5980929A (en) * 1998-03-13 1999-11-09 Johns Hopkins University, School Of Medicine Use of a protein tyrosine kinase pathway inhibitor in the treatment of retinal ischmemia or ocular inflammation
US6028099A (en) * 1998-03-13 2000-02-22 John Hopkins University, School Of Medicine Use of an inhibitor of the protein tyrosine kinase pathway in the treatment of choroidal neovascularization
EP1459742A3 (en) * 1998-08-17 2007-12-26 Senju Pharmaceutical Co., Ltd. Agent for prophylaxis and treatment of asthenopia and pseudomyopia
EP1034793A4 (en) * 1998-08-17 2003-07-09 Senju Pharma Co Preventives/remedies for glaucoma
US6649625B2 (en) 1998-08-17 2003-11-18 Senju Pharmaceutical Co., Ltd. Agent for prophylaxis and treatment of glaucoma
US6673812B1 (en) 1998-08-17 2004-01-06 Senju Pharmaceutical Co., Ltd. Preventives/remedies for glaucoma
EP1459742A2 (en) 1998-08-17 2004-09-22 Senju Pharmaceutical Co., Ltd. Agent for prophylaxis and treatment of asthenopia and pseudomyopia
EP1121932A1 (en) * 1998-10-13 2001-08-08 Kyowa Hakko Kogyo Co., Ltd. Remedies for ocular diseases
US6451787B1 (en) 1998-10-13 2002-09-17 Cephalon, Inc. Remedies for ocular diseases
WO2000021531A1 (en) * 1998-10-13 2000-04-20 Kyowa Hakko Kogyo Co., Ltd. Remedies for ocular diseases
US6399655B1 (en) 1998-12-22 2002-06-04 Johns Hopkins University, School Of Medicine Method for the prophylactic treatment of cataracts
JP2007515426A (en) * 2003-12-22 2007-06-14 アルコン,インコーポレイテッド Cdk2 antagonist as a short form c-Maf transcription factor antagonist for the treatment of glaucoma
AU2004308938B2 (en) * 2003-12-22 2011-06-23 Alcon, Inc. CDK2 antagonists as short form c-Maf transcription factor antagonists for treatment of glaucoma
WO2005063252A1 (en) * 2003-12-22 2005-07-14 Alcon, Inc. Cdk2 antagonists as short form c-maf transcription factor antagonists for treatment of glaucoma
WO2006061525A2 (en) * 2004-12-09 2006-06-15 Lionel Bueno Compositions for treating surface ocular pathologies
WO2006061525A3 (en) * 2004-12-09 2007-02-22 Lionel Bueno Compositions for treating surface ocular pathologies
FR2879100A1 (en) * 2004-12-09 2006-06-16 Lionel Bueno COMPOSITIONS FOR THE TREATMENT OF OCULAR SURFACE PATHOLOGIES AND RETINA
US8273708B2 (en) 2004-12-09 2012-09-25 Lionel Bueno Methods of treatment of ocular surface pathologies
US7320974B2 (en) 2005-03-25 2008-01-22 Inspire Pharmaceuticals, Inc. Cytoskeletal active compounds, compositions and use
JP2008538113A (en) * 2005-03-25 2008-10-09 インスパイアー ファーマシューティカルズ,インコーポレイティド Cytoskeletal active compounds, compositions and uses
US8039465B2 (en) 2005-03-25 2011-10-18 Inspire Pharmaceuticals, Inc. Cytoskeletal active compounds, composition and use
US7666861B2 (en) 2005-03-25 2010-02-23 Inspire Pharmaceuticals, Inc. Cytoskeletal active compounds, composition and use
WO2007002670A2 (en) * 2005-06-28 2007-01-04 Bausch & Lomb Incorporated Method of lowering intraocular pressure
WO2007002670A3 (en) * 2005-06-28 2007-04-05 Bausch & Lomb Method of lowering intraocular pressure
WO2008079980A1 (en) * 2006-12-22 2008-07-03 Alcon Research, Ltd. Inhibitors of protein kinase c-delta for the treatment of glaucoma
WO2008078762A1 (en) 2006-12-26 2008-07-03 Santen Pharmaceutical Co., Ltd. Novel n-(2-aminophenyl)benzamide derivative having an urea structure
US8076332B2 (en) 2006-12-26 2011-12-13 Santen Pharmaceutical Co., Ltd. N- (2-aminophenyl) benzamide derivative having urea structure
US8012972B2 (en) 2007-03-28 2011-09-06 Santen Pharmaceutical Co., Ltd. Pyridinecarboxylic acid (2-aminophenyl) amide derivative having urea structure
WO2008123395A1 (en) 2007-03-28 2008-10-16 Santen Pharmaceutical Co., Ltd. Ocular hypotensive agent comprising compound capable of inhibiting histone deacetylase as active ingredient
WO2009142321A1 (en) 2008-05-23 2009-11-26 参天製薬株式会社 Novel thiophenediamine derivative having urea structure
US8198271B2 (en) 2008-05-23 2012-06-12 Santen Pharmaceutical Co., Ltd. Thiophenediamine derivative having urea structure
US8288380B2 (en) 2008-05-23 2012-10-16 Santen Pharmaceutical Co., Ltd. Thiophenediamine derivative having urea structure
US20120088776A1 (en) * 2009-06-02 2012-04-12 Claudiu Supuran Treatment of mammalian disorders mediated by alpha-anhydrase isoforms
TWI565466B (en) * 2009-06-02 2017-01-11 芬蘭坦帕雷大學 Treatment of mammalian disorders mediated by alpha-carbonic anhydrase isoforms
US9623025B2 (en) * 2009-06-02 2017-04-18 Claudiu Supuran Treatment of mammalian disorders mediated by alpha-carbonic anhydrase isoforms
WO2015007896A1 (en) * 2013-07-18 2015-01-22 Neuroptis Biotech Method for producing a 1-(5-halonaphthalene-1-sulfonyl)-1h-hexahydro-1,4-diazepine and composition comprising same
EP3034081A1 (en) * 2014-12-18 2016-06-22 Neuroptis Biotech Treatment of an inflammation of the tarsal gland of the eyelid

Also Published As

Publication number Publication date
AU2053597A (en) 1997-09-10
IL125735A0 (en) 1999-04-11
IL125735A (en) 2003-09-17
US5798380A (en) 1998-08-25
WO1997030701A3 (en) 1998-02-26
US6110912A (en) 2000-08-29

Similar Documents

Publication Publication Date Title
US6110912A (en) Cytoskeletal active agents for glaucoma therapy
US6586425B2 (en) Cytoskeletal active agents for glaucoma therapy
US8618088B2 (en) Methods of inhibiting choroidal neovascularization
PETERSON et al. Effect of latrunculin-B on outflow facility in monkeys
B Toris Pharmacotherapies for glaucoma
US20030175849A1 (en) Eye treatments using synthetic thyroid hormone compositions
WO1998007435A9 (en) Eye treatments using synthetic thyroid hormone compositions
US8097640B2 (en) Prophylactic or therapeutic agent for diabetic maculopathy
AU2019360953A1 (en) Methods and compositions for treatment of glaucoma and related conditions
Okka et al. Effects of latrunculin B on outflow facility, intraocular pressure, corneal thickness, and miotic and accommodative responses to pilocarpine in monkeys
Kaufman Inhibition of aceclidine-stimulated outflow facility, accommodation and miosis in rhesus monkeys by muscarinic receptor subtype antagonists
Tian et al. Acute effects of H-7 on ciliary epithelium and corneal endothelium in monkey eyes
CA2659511A1 (en) Anticonvulsive pharmaceutical compositions
KR100854058B1 (en) Remedies for retina and choroid diseases containing steroids as the active ingredient
Toris et al. Aqueous humor dynamics II: clinical studies
Waschke New Perspectives in the Pharmacologic Treatment of Primary Open-Angle Glaucoma: Pathogenesis and Patient Factors
AU717608C (en) Eye treatments using synthetic thyroid hormone compositions
Sharma et al. Studies on Corneal Permeation and Oculo-Hypotensive Effect of Benazepril in Chronic and Acute Models of Glaucoma

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AL AM AT AU AZ BB BG BR BY CA CH CN CZ DE DK EE ES FI GB GE HU IL IS JP KE KG KP KR KZ LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK TJ TM TR TT UA UG US UZ VN AM AZ BY KG KZ MD RU TJ TM

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): KE LS MW SD SZ UG AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA

121 Ep: the epo has been informed by wipo that ep was designated in this application
CFP Corrected version of a pamphlet front page
CR1 Correction of entry in section i

Free format text: PAT.BUL.37/97 UNDER INID (81) "DESIGNATED STATES", ADD "BA,CU,LC,YU"

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
AK Designated states

Kind code of ref document: A3

Designated state(s): AL AM AT AU AZ BB BG BR BY CA CH CN CZ DE DK EE ES FI GB GE HU IL IS JP KE KG KP KR KZ LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK TJ TM TR TT UA UG US UZ VN AM AZ BY KG KZ MD RU TJ TM

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): KE LS MW SD SZ UG AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA

NENP Non-entry into the national phase

Ref country code: JP

Ref document number: 97530335

Format of ref document f/p: F

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase