WO1997030085A1 - Troponin subunits and fragments useful as angiogenesis inhibitors - Google Patents

Troponin subunits and fragments useful as angiogenesis inhibitors Download PDF

Info

Publication number
WO1997030085A1
WO1997030085A1 PCT/US1997/002439 US9702439W WO9730085A1 WO 1997030085 A1 WO1997030085 A1 WO 1997030085A1 US 9702439 W US9702439 W US 9702439W WO 9730085 A1 WO9730085 A1 WO 9730085A1
Authority
WO
WIPO (PCT)
Prior art keywords
troponin
subunit
composition
peptide
twitch
Prior art date
Application number
PCT/US1997/002439
Other languages
French (fr)
Other versions
WO1997030085B1 (en
Inventor
Marsha A. Moses
Robert S. Langer
Dimitri G. Wiederschain
Inmin Wu
Arthur Sytkowski
Original Assignee
Children's Medical Center Corporation
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 Children's Medical Center Corporation filed Critical Children's Medical Center Corporation
Priority to JP52953997A priority Critical patent/JP3990456B2/en
Priority to EP97905992A priority patent/EP1007556B1/en
Priority to CA2247247A priority patent/CA2247247C/en
Priority to DE69729125T priority patent/DE69729125T2/en
Priority to AT97905992T priority patent/ATE266675T1/en
Priority to AU22753/97A priority patent/AU707262B2/en
Publication of WO1997030085A1 publication Critical patent/WO1997030085A1/en
Publication of WO1997030085B1 publication Critical patent/WO1997030085B1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4716Muscle proteins, e.g. myosin, actin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention provides for a novel pharmaceutical composition, and method of use thereof for the treatment of diseases or disorders involving abnormal angiogenesis.
  • the present invention is based, in part, on the discovery that troponin subunits C, I and T inhibit stimulated endothelial cell proliferation.
  • Pharmaceutical compositions containing therapeutically effective amounts of troponin C, I, or T, suounits, fragments, or analogs and methods of therapeutic use thereof are provided.
  • Angiogenesis occurs in response to specific signals and involves a complex process characterized by infiltration of the basal lamina by vascular endothelial cells in response to angiogenic growth signal (s) , migration of the endothelial cells toward the source of the signal (s) , and subsequent proliferation and formation of the capillary tube. Blood flow through the newly formed capillary is initiated after the endothelial cells come into contact and connect w th a preexisting capillary.
  • angiogenesis is stringently regulated and spatially and temporally delimited. Under conditions of pathological angiogenesis such as that characterizing solid tumor growth, these regulatory controls fail . Unregulated angiogenesis becomes pathologic and sustains progression of many neoplastic and non-neoplastic diseases .
  • a number of serious diseases are dominated by abnormal neovascularization including solid tumor growth and metastases, arthritis, some types of eye disorders, and psoriasis. See, e . g. , reviews by Moses et al . , 1991,
  • ocular neovascularization occurs in response to the diseased state.
  • ocular disorders include diabetic retinopathy, neovascular glaucoma, inflammatory diseases and ocular tumors (e.g., retinoblastoma) .
  • An inhibitor of angiogenesis could have an important therapeutic role in limiting the contributions of this process to pathological progression of the underlying disease states as well as providing a valuable means of studying their etiology.
  • agents that inhibit tumor neovascularization could play an important role in inhibiting metastatic tumor growth.
  • angiogenesis relating to vascular endothelial cell proliferation, migration and invasion, have been found to be regulated in part by polypeptide growth factors.
  • endothelial cells exposed to a medium containing suitable growth factors can be induced to evoke some or all of the angiogenic responses.
  • polypeptides with in vi tro endothelial growth promoting activity have been identified. Examples include acidic and basic fibroblast growth factors, transforming growth factors ⁇ and ⁇ , platele -derived endothelial cell growth factor, granulocyte colony- stimulating factor, interleukin-8, hepatocyte growth factor, proliferin, vascular endothelial growth factor and placental growth factor. See, e.g., review by Folkman et al . , 1995, N. Engl . J. Med . , 333:1757-1763.
  • Capillary endothelial cells proliferate in response to an angiogenic stimulus during neovascularization. Ausprunk and Folkman, 1977, J “ . Microvasc . Res . l ⁇ : 153-65.
  • An in vi tro assay assessing endothelial cell proliferation in response to known angiogenesis simulating factors, such as acidic or basic fibroblast growth factor (aFGF and bFGF, respectively) has been developed to mimic the process of neovascularization in vi tro .
  • This type of assay is the assay of choice to demonstrate the stimulation of capillary EC proliferation by various angiogenic factors. Shing et al . , 1984, Science 223 :1296-1298.
  • CAM b oassay fertilized chick embryos are cultured in Pet ⁇ dishes.
  • a disc of a release polymer, such as methyl cellulose, impregnated with the test sample or an appropriate control substance is placed onto the vascular membrane at its advancing edge.
  • the area around the implant is observed and evaluated. Avascular zones surrounding the test implant indicate the presence of an inhibitor of embryonic neovascularization.
  • angiogenesis inhibitors effective as combinations include heparin (50 ⁇ g) and hydrocortisone (60 ⁇ g) , and B-cyclodext ⁇ n tetradecasulfate (14 ⁇ g) and hydrocortisone (60 ⁇ g) , reported by Folkman et al. , 1989, Science 243 :1490.
  • polymer pellets of ethylene vinyl acetate copoly er are impregnated with test substance and surgically implanted in a pocket in the rabbit cornea approximately 1 mm from the limbus. Langer et al . , 1976, Science 193 : 707-72.
  • an angiogenesis inhibitor either a piece of carcinoma or some other angiogenic stimulant is implanted distal to the polymer 2 mm from the limbus.
  • control polymer pellets that are empty are implanted next to an angiogenic stimulant in the same way. In these control corneas, capillary blood vessels start growing towards the tumor implant in 5-6 days, eventually sweeping over the blank polymer.
  • Troponin a complex of three polypeptides is an accessory protein that is closely associated with actin filaments in vertebrate muscle
  • the troponin complex acts in conjunction with the muscle form of tropomyosin to mediate the Ca 2* dependency of myosm ATPase activity and thereoy regulate muscle contraction.
  • the troponin polypeptides T, I, and C are named for their tropomyosin binding, inhibitory, and calcium binding activities, respectively Troponin T binds to tropomyosin and is believed to be responsible for positioning the troponin complex on the muscle thin filament Troponin I binds to actin, and the complex formed by troponins I and T, and tropomyosin, inhibits the interaction of actin and myosm.
  • Troponin C is capable of binding up to four calcium molecules. Studies suggest that when the level of calcium in the muscle is raised, troponin C causes troponin I to loose its hold on the actin molecule, causing the tropomyosin molecule shift, thereby exposing the myosm binding sites on actin and stimulating myosm ATPase activity.
  • troponin subunits Prior to the discovery of the present invention, troponin subunits were not known to inhibit the process of endothelial eel proliferation The citation of a reference herein shall not be construed as an admission that such reference is prior art to the present invention.
  • the present invention relates to pharmaceutical compositions containing troponin subunits C, I, or T, or fragments thereof, in therapeutically effective amounts that are capable of inhibiting endothelial cell proliferation
  • the invention also relates to pharmaceutical compositions containing analogs of troponin subunits C, I, or T ana analogs of their fragments, in therapeutically effective amounts that are capable of inhibiting en ⁇ othelial cell proliferation.
  • the invention further relates to treatment of neovascular disorders by administration of a therapeutic compound of the invention.
  • Such therapeutic compounds include. troponin subunits C, I, and T, and fragments and analogs thereof.
  • a Therapeutic of the invention is administered to treat a cancerous condition, or to prevent progression from the pre-neoplastic or pre-malignant state into a neoplastic or a malignant state
  • a Therapeutic of the invention is administered to treat ocular disorders associated with neovascularization
  • troponin subunit when not preceding the terms C, I or T, means generically any of troponin subunits C, I, or T.
  • BCE Endothelial Cell proliferation by troponin C
  • Percent inhibition of bFGF-stimulated BCE proliferation is shown as a function of troponin C concentration (nM) Percent inhibition was determined by comparing results obtained for cells treated with stimulus alone with those obtained for samples exposed to both stimulus and inhibitor Well volume was 200 ⁇ l .
  • FIG. 1 Inhibition of capillary BCE proliferation by troponin I. Percent inhibition of bFGF-stimulated BCE proliferation is shown as a function of troponin I concentration (nM) . Percent inhibition was determined as described in Figure 1. Well volume was 200 ⁇
  • FIG. 5 Inhibition of capillary BCE proliferation by troponin C, I and T. Percent inhibition of bFGF-stimulated BCE proliferation is shown as a function of troponin C, I, and T concentration (nM) . Percent inhibition was determined as described in Figure 1. Well volume was 200 ⁇ l.
  • the present invention relates to therapeutic methods and compositions based on troponin subunits.
  • the invention provides for treatment of neovascular disorders by administration of a therapeutic compound of the invention.
  • therapeutic compounds include: troponin C, I, and T subunits, fragments and analogs thereof (collectively “peptides of the invention”) .
  • the peptides of the invention are characterized by the property of inhibiting bovine endothelial cell proliferation in culture with an IC 50 of 10 ⁇ M or less.
  • a Therapeutic of the invention is administered to treat a cancerous condition, or to prevent progression from a pre-neoplastic or non-malignant state into a neoplastic or a malignant state. In other specific embodiments, a Therapeutic of the invention is administered to treat an ocular disorder associated with neovascularization.
  • a Therapeutic of the invention is a peptide consisting of at least a fragment of troponin C, troponin I, troponin T, or troponins C and I, which is effective to inhibit endothelial cell proliferation.
  • examples of the troponin subunits that can be utilized in accordance with the invention include the subunits of troponin from human fast twitch skeletal muscle, the sequences of which are given below:
  • the invention encompasses peptides which are homologous to human fast-twitch skeletal troponin C (SEQ ID NO:l) or fragments thereof.
  • the ammo acid sequence of the peptide has at least 80% identity compared to the fragment of human fast twitch skeletal troponin C from which it is derived (the "prototype fragment") . In another embodiment, this identity is greater than 85%. In a more preferred embodiment, this identity is greater than 90% . In a most preferred embodiment, the ammo acid sequence of the peptide has at least 95% identity with the prototype fragment. Fragments can be at least 10 am o acids, and m preferred embodiments at least 50, 75, 100 and 120 ammo acids, respectively
  • the invention encompasses peptides which are homologous to human fast-twitch skeletal troponin I (SEQ ID NO:2) or fragments thereof.
  • the ammo acid sequence of the peptide has at least 80% identity with the prototype human fast-twitch skeletal troponin I fragment. In another embodiment, this identity is greater than 85%. In a more preferred embodiment, this identity is greater than 90%.
  • the amino acid sequence of the peptide has at least 95% identity with the prototype fragment. Fragments can be at least 10 amino acids, and in preferred embodiments at least 50, 75, 100 and 120 amino acids, respectively.
  • the invention encompasses peptides which are homologous to human fast-twitch skeletal troponin T (SEQ ID NO:3) or fragments thereof.
  • the ammo acid sequence of the peptide has at least 80% identity with the prototype human fast-twitch skeletal beta troponin T. In another embodiment, this identity is greater than 85% . In a more preferred embodiment, this identity is greater than 90%.
  • the ammo acid sequence of the peptide has at least 95% identity with the prototype fragment Fragments can be at least 10 ammo acids, and m preferred embodiments at least 50, 75, 100, 120 and 200 amino acids length, respectively.
  • the peptides of the invention are troponin C, troponin I and troponin T suounits of the fast twitch, slow twitch and cardiac isoforms from other mammalian species, e.g., human, rabbit, rat, mouse, bovine, ovine and porcine.
  • a Therapeutic of the invention is combined with a therapeutically effective amount of another molecule which negatively regulates angiogenesis which may be, but is not limited to, platelet factor 4, thrombospondin-l, tissue inhibitors of metalloproteases (TIMP1 and TIMP2) prolact (16-Kd fragment) , angiostat (38-Kd fragment of plasminogen) , bfGf soluble receptor, transforming growth factor ⁇ , mterferon alfa, and placental proliferm-related protein.
  • angiogenesis may be, but is not limited to, platelet factor 4, thrombospondin-l, tissue inhibitors of metalloproteases (TIMP1 and TIMP2) prolact (16-Kd fragment) , angiostat (38-Kd fragment of plasminogen) , bfGf soluble receptor, transforming growth factor ⁇ , mterferon alfa, and placental proliferm-related protein.
  • the invention provides for a pharmaceutical composition of the present invention in combination with a chemotherapeutic agent.
  • a Therapeutic of the invention is combined with chemotherapeutic agents or radioactive isotope exposure.
  • the invention is illustrated by way of examples infra which disclose, inter alia , the inhibition of capillary endothelial cell proliferation by troponin subunits C, I, and T and the means for determining inhibition of capillary endothelial cell migration and inhibition of neovascularization m vivo by troponin subunits.
  • the detailed description of the invention is divided into the subsections set forth below.
  • the invention provides for pharmaceutical compositions comprising troponin subunits, fragments, and analogs thereof.
  • the subunits, fragments, or analogs are of fly, frog, mouse, rat, rabbit, pig, cow, dog, monkey, or human troponin subunits.
  • troponin subunit fragments can be made by altering troponin sequences by substitutions, additions or deletions that provide for functionally equivalent molecules. These include, but are not limited to, troponin subunits, fragments, or analogs containing, as a primary amino acid sequence, all or part of the amino acid sequence of a troponin subunit including altered sequences in which functionally equivalent amino acid residues are substituted for residues within the sequence resulting in a silent change. For example, one or more amino acid residues within the sequence can be substituted by another amino acid of a similar polarity which acts as a functional equivalent, resulting in a silent alteration. Substitutes for an amino acid within the sequence may be selected from other members of the class to which the amino acid belongs.
  • the nonpolar (hydrophobic) amino acids include alanine, leucine, isoleucine, valine, proline, phenylalanine, tryptophan and methionine.
  • the polar neutral amino acids include glycine, serine, threonine, cysteine, tyrosine, asparagine, and glutamine.
  • the positively charged (basic) amino acids include arginine, lysine and histidine.
  • the negatively charged (acidic) amino acids include aspartic acid and glutamic acid.
  • One embodiment of the invention provides for molecules consisting of or comprising a fragment of at least 10 (continuous) amino acids of a troponin subunit which is capable of inhibiting endothelial cell proli eration. In other embodiments, this molecule consists of at least 20 or 50 ammo acids of the troponin subunit. In specific embodiments, such molecules consist of or comprise fragments of a troponin subunit that at least 75, 120 or 200 ammo 5 acids.
  • the protein is a mammalian troponin subunit. In alternative embodiments, it is a mammalian troponin C, I, or T subunit.
  • tissue 10 invention can be derived from tissue (see, for example,
  • troponin subunit encoding nucleic acid sequence can be mutated m vi tro or in vivo, to create
  • Any technique for mutagenesis known m tie art can be used, including, but not limited to, in vi tro site- directed mutagenesis (Hutchinson et al . , 1978, J. Biol . Chem . 5 253 :6551) , use of TAB* linkers (Pharmacia) , etc
  • troponin subunit C, I, or T sequence may also be made at the protein level
  • troponin subunit fragments or other fragments or analogs which are 0 differentially modified during or after translation, e . g . , by acetylation, phosphorylation, carboxylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to an antibody molecule or other cellular ligand, etc.
  • any of numerous chemical 5 modifications may be carried out by known techniques, including, but not limited to, specific chemical cleavage toy cyanogen bromide, trypsm, chymotryps , papam, V8 protease, NaBH 4 , acetylation, formylation, oxidation, reduction, etc
  • fragments and analogs of troponin 0 subunits can be chemically synthesized.
  • a peptide corresponding to a portion of a troponin subunit which comprises the desired domain, or which mediates the desired activity m vi tro can be synthesized by use of a peptide synthesizer.
  • nonclassical 5 amino acids or chemical ammo acid analogs can be introduced as a substitution or addition into the troponin subunit sequence.
  • Non-classical ammo acids include, but are not limited to, the D-isomers of the common ammo acids, ⁇ -ammo isobutyric acid, 4-ammobutyr ⁇ c acid, hydroxyprolme, 0 sarcosme, citrulline, cysteic acid, t-butylglycme, t- butylalanme, phenylglyc e, cyclohexylalamne, /3-alanme, designer ammo acids such as 0-methyl ammo acids, C ⁇ -methyl ammo acids, and N ⁇ -methyl ammo acids.
  • the invention encompasses 5 a chimeric, or fusion, protein comprising a troponin s ⁇ bunit or fragment thereof (consisting of at least a domain cr motif of the troponin subunit that is responsible for endothelial cell proliferation) joined at its amino or carboxy-terminus via a peptide bond to an ammo acid sequence of a different protein.
  • a chimeric product can be made by ligating the appropriate nucleic acid sequences encoding
  • such a chimeric product may be made by protein synthetic techniques, e . g. , by use of a peptide synthesizer.
  • troponin subunits fragments and analogs
  • .lb_ can be assayed in vi tro by various methods. These methods are based on the physiological processes involved in angiogenesis and while they are within the scope of the invention, they are not intended to limit the methods by which troponin subunits, fragments and analogs inhibiting
  • _- angiogenesis are defined and/or a therapeutically effective dosage of the pharmaceutical composition is determined.
  • ._ cells (EC) in vi tro various bioassays known in the art can be used, including, but not limited to, radioactive incorporation into nucleic acids, colorimetric assays and cell counting.
  • angiogenesis stimulating factor such as aFGF.
  • the colorimetric determination of cellular acid phosphatase activity is described by Connolly et al . , 1986, J. Anal . Biochem. 152:136-140 According to this method, described in Example 3, capillary endothelial cells are treated with angiogenesis stimulating factors, such as aFGF, and a range of potential inhibitor concentrations These samples are incubated to allow for growth, and then harvested, washed, lysed in a buffer containing a phosphatase substrate, and then incubated a second time A basic solution is added to stop the reaction and color development is determined at 405 ⁇ According to Connolly et al , a linear relationship is obtained between acid phosphatase activity and endothelial cell number up to 10,000 cells/sample Standard curves for acid phosphatase activity are also generated from known cell numbers in order to confirm that the enzyme levels reflect the actual EC numbers Percent inhibition is determined by comparing
  • Troponin C inhibited bFGF-stimulated endothelial cell proliferation in a dose-dependent manner m all concentrations tested (FIG 1) .
  • Percent inhibition of bovine endothelial cell proliferation (“BCE") was 54%, 86%, 83%, and 100% at concentrations of 280 nM, 1.4 ⁇ m, 2.8 ⁇ M and 5.6 ⁇ M, respectively An inhibition of 100% was observed at a concentration of 20 ⁇ g/well (5.6 ⁇ M)
  • IC 50 represents the concentration at which 50% inhibition of aFGF growth factor- induced stimulation was observed.
  • the IC S0 of troponin C was determined to be 278 nM.
  • Troponin I inhibited bFGF-stimulated BCE proliferation at concentrations of 1 and 5 ug/well, but inhibition was not observed the sample tested at 10 ug/well (FIG 2)
  • the percent inhibition of BCE was 33% and 46% at concentrations of 240 nM and 1.2 ⁇ M, respectively.
  • the ICc 0 of troponin I was determined to be 1.14 ⁇ M.
  • Troponin T inhibited bFGF-stimulated EC proliferation at concentrations of 10 and 20 ug/well, but not at concentrations of 1 and 5 ⁇ g/well (FIG 3) .
  • BCE proliferation was inhibited 23% and 62% at 1.6 ⁇ M and 3.3 ⁇ M, respectively.
  • the IC 50 of troponin T was determined to be 2.14 ⁇ M.
  • the combination of troponin subunits C and I inhibited EC at all concentrations tested (FIG 4) .
  • the percent inhibition of BCE was 52%, 54% 73% and 47% at 130 nM, 645 nM, 1.3 ⁇ M and 2.6 ⁇ M, respectively.
  • the IC 50 of this combination was determined to be 110 nM.
  • the troponins samples tested had no detectable inhibitory effect on the growth of Balb/c 3T3 cells, a non- endothelial cell type.
  • the incorporation of radioactive thymidine by capillary endothelial cells represents another means by which to assay for the inhibition of endothelial cell proliferation by a potential angiogenesis inhibitor.
  • a predetermined number of capillary endothelial cells are grown in the presence of 3 H-Thymidine stock, an angiogenesis stimulator such as for example, bFGF, and a range of concentrations of the angiogenesis inhibitor to be tested. Following incubation, the cells are harvested and the extent of thymidine incorporation is determined. See, Example 2.
  • troponin subunits, fragments or analogs The ability of varying concentrations of troponin subunits, fragments or analogs to interfere with the process of capillary endothelial cell migration in response to an angiogenic stimulus can be assayed using the modified Boyden chamber technique. See, Section 2 and Example 4, infra .
  • Another means by which to assay the functional activity of troponin subunits, fragments and analogs involves examining the ability of the compounds to inhibit the directed migration of capillary endothelial cells which ultimately results in capillary tube formation This ability may be assessed for example, using an assay in which capillary endothelial cells plated on collagen gels are challenged with the inhibitor, and determining whether capillary-like tube structures are formed by the cultured endothelial cells.
  • Assays for the ability to inhibit angiogenesis in vivo include the chick chorioallantoic membrane assay (see Section 2 and Example 5, infra ) and rat or rabbit corneal pocket assays. See, Polverini et al . , 1991, Methods Enzymol . 198 • 440-450 According to the corneal pocket assays, a tumor of choice is implanted into the cornea of the test: animal the form of a corneal pocket. The potential angiogenesis inhibitor is applied to the corneal pocket and the corneal pocket is routinely examined for neovascularization See, Section 2 and Example 6 infra .
  • One embodiment of the invention provides for combination of the troponin subunits, fragments, or analogs of the present invention to inhibit angiogenesis.
  • Another embodiment provides for the combination of troponin subunits, fragments, or analogs with other angiogenesis inhibiting factors.
  • angiogenesis inhibiting factors include, but are not limited to: angiostatic steroids, thrombospondm, platelet factor IV, transforming growth factor ⁇ , mterferons, tumor necrosis factor ⁇ , bovine vitreous extract, protamine, tissue inhibitors of metalloprotemases (TIMP-1 and TIMP-2) , prolactm (16-kd fragment) , angiostatm (38-kd fragment of plasmmogen) , bfGf soluble receptor, and placental proliferin-related protein.
  • the therapeutically effective dosage for inhibition of angiogenesis m vivo may be extrapolated from m vi tro inhibition assays using the compositions of the invention above or m combination with other angiogenesis inhibiting factors.
  • the effective dosage is also dependent on the method and means of delivery.
  • the inhibitor is delivered in a topical-ophthalmic carrier.
  • the inhibitor is delivered by means of a biodegradable, polymeric implant.
  • the protein can also be modified, for example, by polyethyleneglycol treatment.
  • the invention provides for treatment of diseases or disorders associated with neovascularization by administration of a therapeutic compound of the invention.
  • therapeutic compounds include troponin subunits and fragments and analogs thereof (e.g., as described infra) .
  • Malignant and metastatic conditions which can be treated with the Therapeutic compounds of the present invention include, but are not limited to, the solid tumors listed in Table 1 (for a review of such disorders, see Fishman et al . , 1985, Medicine, 2d Ed., J.B. Lippmcott Co., Philadelphia) :
  • OCULAR DISORDERS Ocular disorders associated with neovascularization which can be treated with the Therapeutic compounds of the present invention include, but are not limited to: neovascular glaucoma diabetic retinopathy retinoblastoma retrolental fibroplasia uveitis retinopathy of prematurity macular degeneration corneal graft neovascularization
  • Therapeutic compounds of the present invention include, but are not limited to, hemangioma, arthritis, psoriasis, angiofibroma, atherosclerotic plaques, delayed wound healing, granulations, hemophilic joints, hypertrophic scars, nonunion fractures, Osier-Weber syndrome, pyogenic granuloma, scleroderma, trachoma, and vascular adhesions.
  • the Therapeutics of the invention can be tested m vivo for the desired therapeutic or prophylactic activity as well as for determination of therapeutically effective dosage.
  • such compounds can be tested m suitable animal model systems prior to testing in humans, including, but not limited to, rats, mice, chicken, cows, monkeys, rabbits, etc.
  • suitable animal model systems prior to testing in humans, including, but not limited to, rats, mice, chicken, cows, monkeys, rabbits, etc.
  • any animal model system known in the art may be used.
  • the invention provides methods of treatment and prophylaxis) by administration to a subject an effective amount of a Therapeutic of the invention.
  • the Therapeutic is substantially purified as set forth in Example 1.
  • the subject is preferably an animal, including, but not limited to, animals such as cows, pigs, chickens, etc., and is preferably a mammal, and most preferably human.
  • the invention further provides methods of treatment by administration to a sub ect, an effective amount of a Therapeutic of the invention combined with a chemotherapeutic agent and/or radioactive isotope exposure.
  • the invention also provides for methods of treatment of a Therapeutic of the invention for patients who have entered a remission in order to maintain a dormant state.
  • a Therapeutic of the invention e.g., encapsulation in liposo es, microparticles, microcapsules, receptor-mediated endocytosis (see, e.g., Wu and u, 1987, J.
  • Methods of introduction include, but are not limited to , topical , tradermal , intramuscular , mtraperitoneal , intravenous , subcutaneous , tranasal , epidural, ophthalmic, and oral routes.
  • the compounds may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents. It is preferred that administration is localized, but it may be systemic.
  • mtraventricular injection may be facilitated by an intraventricular catheter, for example, attached to a reservoir, such as an Ommaya reservoir.
  • Pulmonary administration can also be employed, e . g. , by use of an inhaler or nebulizer, and formulation with an aerosolizing agent.
  • compositions of the invention may be desirable to administer the pharmaceutical compositions of the invention locally to the area m need of treatment; this may be achieved by, for example, and not by way of limitation, local infusion during surgery, topical application, e.g., in conjunction with a wound dressing after surgery, by injection, by means of a catheter, by means of a suppository, or by means of an implant, said implant being of a porous, non-porous, or gelatinous material, including membranes, sucn as sialastic membranes, or fibers.
  • administration can be by direct injection at the site (or former site) of a malignant tumor or neoplastic or pre- neoplastic tissue.
  • the purified troponin subunit is combined with a carrier so that an effective dosage is delivered, based on the desired activity (i.e., ranging from an effective dosage, for example, of 1.0 ⁇ M to 1.0 mM to prevent localized angiogenesis, endothelial cell migration, and/or inhibition of capillary endothelial cell proliferation.
  • the carrier ⁇ ay m the form of, for example, and not by way of limitation an ointment, cream, gel, paste, foam, aerosol, suppository, pad or gelled stick.
  • a topical Therapeutic for treatment of some cf the eye disorders discussed infra consists of an effective amount of troponin subunit, fragment, or analog, m a ophthalmologically acceptable excipient such as buffered saline, mineral oil, vegetable oils such as corn or arachis oil, petroleum jelly, Miglyol 182, alcohol solutions, or liposomes or liposome-like products. Any of these compositions may also include preservatives, antioxidant ⁇ , antibiotics, immunosuppressants, and other biologicallv or pharmaceutically effective agents which do not exert a detrimental effect on the troponin subunit.
  • the troponin subunit, fragment, or analog composition may be in the form of tablets or capsules, which can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystallme cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as algmic acid, Primogel, cr corn starch; a lubricant such as magnesium stearate or Sterotes, or a glidant such as colloidal silicon dioxide.
  • a binder such as microcrystallme cellulose, gum tragacanth or gelatin
  • an excipient such as starch or lactose, a disintegrating agent such as algmic acid, Primogel, cr corn starch
  • a lubricant such as magnesium stearate or Sterotes
  • a glidant such as colloidal silicon dioxide.
  • dosage unit form can contain, in addition to material of the above type, a liquid carrier such as a fatty oil
  • Suppositories generally contain active ingredient m the range of 0.5% to 10% by weight; oral formulations preferably contain 10% to 95% active ingredient.
  • the Therapeutic can be delivered in a vesicle, in particular a liposome See, Langer et al . , 1990, Sci ence 249 :1527-1533 , Treat et a. , 1989, in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein and Fidler (eds.) , Liss, New York, pp. 353-365; Lopez-Berestein, ibid. , pp. 317-327.
  • the Therapeutic can be delivered in a controlled release system.
  • an infusion pump may be used to administer troponin subunit, such as for example, that used for delivering insulin or chemotherapy to specific organs or tumors (see Langer, supra ; Sefton, CRC Cri t . Ref . Biomed . , 1987, Eng. 14 . :201; Buchwald et al., 1980, Surgery 88:507; Saudek et al . , 1989, N. Engl . J. Med . 321:574.
  • the troponin subunit, fragment, or analog is administered in combination with a biodegradable, biocompatible polymeric implant which releases the troponin subunit, fragment, or analog over a controlled period of time at a selected site.
  • a biodegradable, biocompatible polymeric implant which releases the troponin subunit, fragment, or analog over a controlled period of time at a selected site.
  • preferred polymeric materials include polyanhydrides, polyorthoesters, polyglycolic acid, polylactic acid, polyethylene vinyl acetate, and copolymers and blends thereof. See, Medical Applications of Controlled Release, Langer and Wise (eds.) , 1974, CRC Pres., Boca Raton, Florida; Controlled Drug Bioavailability, Drug Product Design and Performance, Smolen and Ball (eds.) , 1984, Wiley, New York; Ranger and Peppas, 1983, J.
  • a controlled release system can be placed in proximity of the therapeutic target, i . e . , the brain, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, 1989, supra, vol. 2, pp. 115-138) .
  • compositions comprise a therapeutically effective amount of a Therapeutic, and a pharmaceutically acceptable carrier.
  • the pharmaceutical compositions of the invention can be formulated as neutral or salt forms.
  • Pharmaceutically acceptable salts include those formed with free amino groups such as those derived from hydrochloric, phosphoric, acetic, oxalic, tarta ⁇ c acids, etc., and those formed with free carboxyl groups such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, lsopropylamme, triethylamme, 2-ethylammo ethanol, histidine, procaine, etc.
  • “pharmaceutically acceptable” means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognize ⁇ pnarmacopeia for use in animals, and more particularly in humans
  • carrier refers to a diluent, adjuvant, excipient, or vehicle with which the therapeutic is administered.
  • Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents Water is a preferred carrier when the pharmaceutical composition is administered intravenously.
  • Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for ectable solutions.
  • suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like.
  • composition can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents such as acetates, citrates or phosphates
  • Antibacterial agents such as benzyl alcohol or methyl parabens
  • antioxidants such as ascorbic acid or sodium bisulfite, chelatmg agents such as ethylenediammetetraacetic acid
  • agents for the adjustment of tonicity such as sodium chloride or dextrose are also envisioned.
  • the parental preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
  • compositions will contain a therapeutically effective amount of the Therapeutic, preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the patient.
  • the formulation should suit the mode of administration.
  • the composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous administration to human beings.
  • compositions for intravenous administration are solutions in sterile lsotonic aqueous buffer.
  • the composition may also include a solubilizing agent and a local anesthetic such as lignocaine to ease pain at the site of the injection.
  • the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
  • a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
  • the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline.
  • an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
  • the amount of the Therapeutic of the invention which will be effective in the treatment of a particular disorder or condition will depend on the nature of the disorder or condition, and can be determined by standard clinical techniques.
  • m vi tro assays such as those discussed in section 5.2 may optionally be employed to help identify optimal dosage ranges.
  • the precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness of the disease or disorder, and should be decided according to the judgment of the practitioner and each patient's circumstances
  • suitable dosage ranges for intravenous administration are generally about 20-500 micrograms of active compound per kilogram body weight.
  • Suitable dosage ranges for intranasal administration are generally about 0.01 pg/kg body weight to 1 mg/kg body weight.
  • Effective doses may be extrapolated from dose-response curves derived from in vi tro or animal model test bioassays or systems
  • the invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention.
  • Optionally associated with such container (s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
  • compositions of the present invention and methods for use, will be obvious to those skilled in the art from the foregoing detailed description Such modifications and variations are intended to fall within the scope of the appended claims
  • the following non-limiting examples demonstrate the discovery of troponin subunit inhibition of angiogenic stimulus induced endothelial cell proliferation, and means for determining the effective dosage of troponin subunit, fragment, or analog to inhibit angiogenesis, as well as for identifying troponin subunit fragments and analogs (i.e., tnose fragments or analogs of troponin subunit capable of mnibitmg angiogenesis.
  • the troponin subunit used in tne examples is purified as described infra .
  • 248 2125-2133 for purifying troponin can be used. Rabbit back and leg muscles are removed, cleaned of fat and connective tissue, and ground. The ground muscle (1 kg) is stirred for 5 mm. in 2 liters of a solution containing 20 mM KCl, 1 mM KHC0 3 , 0.1 mM CaCl 2 , and 0.1 mM DTT.” The suspension is filtered through cheesecloth, and the washing of the residue is repeated four times Two liters of 95% ethanol are then added to the washed residue and the solution filtered after 10 mm. The ethanol extraction is repeated twice. The residue is then washed 3 times with 2 liters of diethyl ether for 10 min. Finally the residue is allowed to dry at room temperature for 2 to 3 hours.
  • the dried powder (from 1 kg of muscle) is extracted overnight at 22° with 2 liters of a solution containing 1 M KCl, 25 mM Tris (pH 8.0), 0.1 M CaCl 2 , and 1 mM DTT After
  • the extracts are combined and cooled to 4°C. Solid ammonium sulfate is added to produce approximately 40% saturation (230 g per liter) . After 30 mm. the solution is centrifuged and 125 g of ammonium sulfate is then added per liter of supernatant (60% saturation) . After centrifugation the precipitate is dissolved in 500 ml of a solution containing 5 mM Tris (pH 7.5) , 0.1 mM CaCl 2 , and 0.1 mM DTT and dialyzed against 15 liters of the same solution for 6 hours and against a fresh solution overnight.
  • 5 mM Tris pH 7.5
  • CaCl 2 0.1 mM CaCl 2
  • DTT dialyzed against 15 liters of the same solution for 6 hours and against a fresh solution overnight.
  • Solid KCl is added to a final concentration of 1 M and 1 M KCl solution is added to bring the volume to 1 liter.
  • the pH is then adjusted to .6 by addition of HCl, and the tropomyosin precipitate is removed by centrifugation.
  • the pH of the supernatant is adjusted to 7.0 with KOH, and 450 g of ammonium sulfate were added per liter (70% saturation) .
  • the precipitate is dissolved in a solution containing 5 mM Tris (pH 7.5, 0.1 mM CaCl 2 , and 0.1 mM DTT, and dialyzed overnight against the same solution.
  • Bovine hearts are obtained approximately 30 min. after death and immediately cut open, rinsed of blood, and immersed in ice. The left ventricle is removed, trimmed of excess fat and connective tissue, and ground. All subsequent extraction and preparation steps are performed at 0-3° except where noted.
  • the ground muscle 500 g is homogenized in a Waring Blender for 1 min. in 2.5 liters of solution containing 0.09 M KH 2 P0 4 , 0.06 M K 2 HP0,, 0.3 M KCl, 5 mM 2- mercaptoethanol, pH 6.8. The homogenized muscle suspension is then stirred for 30 min. and centrifuged at 1000 x g for 20 mm. The precipitate is re-extracted for 30 mm. and centrifuged.
  • the residue is then washed with 2.5 liters of 5 mM 2-mercaptoethanol and centrifuged at 1000 x g for 10 mm., followed by two successive washings and centrifugations with 1.5 liters of 50 mM KCl, 5mM Tris-HCl (pH 8.1/5) , mM 2- mercaptoethanol.
  • the residue is then washed and centrifuged twice with 1.5 liters of 50 mM Tris-HCl (pH 8.1) , and 5 mM 2- mercaptoethanol.
  • the volume of the residue is measured, and the residue is mixed with 0.5 volume of 3 M KCl, 50 mM Tris- HC1 (pH 8.1) , and 5 mM 2-mercaptoethanol . After a 16- to 20- hour extraction at 0°, the suspension is centrifuged at 15,000 x g for 10 min. The sediment is discarded, and the supernatant is adjusted to pH 7.6 with 0.05 N HCl . The filamentous precipitate which forms upon pH adjustment is removed by filtering the extract through nylon gauze.
  • the protein that precipitates between 30 and 50% ammonium sulfate saturation is collected, dissolved in a solution containing 1 M KCl, and ImM potassium phosphate (pH 6.8) , and 5 mM 2- mercaptoethanol, and dialyzed against the same solution for 4 hours and against a fresh solution overnight.
  • the protein solution is clarified by centrifugation at 105,000 x g for 30 min.
  • the troponin is then purified by chromatography on a hydroxylapatite column with the protein being eluted between 0.08 and 0.10 M phosphate. Greaser et al . , 1972 Cold Spring Harbor Symp . Quant . Biol . 3_7:235-244.
  • Rabbit cardiac troponin is prepared in a similar manner using a pooled batch of hearts which has been stored at -20°C prior to extraction.
  • the troponin subunits are separated by DEAE- Sephadex chromatography in 6 M urea.
  • Bovine cardiac tropomyosin is prepared from the 50% ammonium sulfate saturation supernatant from the troponin extraction scheme (see above) . Ammonium sulfate is added to 65% saturation, and the precipitate is dissolved in and dialyzed versus 1 M KCl, 1 mM potassium phosphate (pH 7.0) , and 5 mM 2- mercaptoethanol. The protein is then purified by hydroxylapatite chromatography. Protein Determination - Protein concentrations are determined by the biuret method of Gornall et al. using bovine serum albumin as a standard. Gornall et al . , 1949, J. Biol . Chem . , Ill :751-766. Separation of Components - A sequence of SP-
  • DNA encoding various troponin subunits and isoforms are known in the art. See, e.g., Wu et al . , 1994, DNA Cell . Biol . 12:217-233; Schreier et al . , 1990, J. Biol . Chem . 265 :21247-21253 ; and Gahlmann et al . , 1990, J. Biol . Chem. 265:12520-12528.
  • the cells are harvested by centrifugation and suspended in 20 ml of 20 mM Tris, 20% sucrose, 1 mM EDTA, 0.2 mM phenylmethylsulfonyl fluoride, 1 mg/ml lysozy e, pH 7.5.
  • the supernatant is made 50 mM in NaCl, 5 mM in CaCl 2 , 1 mM in MgCl 2 , and 1 mM in DTT and then loaded onto a 1.5 X 25-cm phenyl-Sepharose (Pharmacia LKB Biotechnology Inc.) column.
  • the column is washed first with 50 mM Tris, 50 mM NaCl, 5 mM CaCl 2 , 1 mM MgCl 2 , 1 M DTT, pH 7.5, then with 50 mM Tris, 1 mM NaCl, 0.1 mM CaCl 2 , 1 mM DTT, pH 7.5, until no more protein is eluted.
  • the crude troponin subunit is then eluted with 50 mM Tris, 1 mM EDTA, 1 mM DTT, pH 7.5.
  • Fractions that contained troponin subunit, fragment, or analog are pooled, dialyzed against 25 mM Tris, 6 M urea (United States Biochemical Corp.) , 1 mM MgCl 2 , 1 mM DTT, pH 8.0, and loaded onto a 1.5 X 25-cm DE52 (Whatman) column.
  • the column is eluted with a 0-0.6 M NaCl linear gradient.
  • Troponin subunit, fragment, or analog eluted from the column is dialyzed against 0.1 mM NH 4 HC0 3 , 1 mM 3-mercaptoethanol, lyophilized, and stored. Purity is assessed by SDS- polyacrylamide gel electrophoresis and UV spectrophotometry. Typical yields of 6 mg of purified recombinant troponin subunit, fragment, or analog/liter of bacterial culture are expected. The lyophilized recombinant protein is resuspended in a take up buffer consisting of 6M urea, 20 mM Hepes (pH 7.5), 0.5M NaCl, 2mM EDTA, and 5mM DTT.
  • the mixture is nutated at room temperature for 1 hour.
  • the solution is then dialyzed at 4°C for six hours with 1 exchange against a dialysis buffer consisting of 0.5M NaCl, 20mM Hepes (pH 7.5) , and 0.5mM DTT.
  • the lyophilized recombinant protein is resuspended in a take up buffer consisting of 0.1 M NaCl, 20 mM Hepes (pH 7.5) , 2mM EDTA, and 5mM DTT. This solution is dialyzed for 6 hours at 4°C with one exchange against a dialysis buffer of 0.1 M NaCl, 20 mM Hepes (pH 7.5), and 0.5 mM DTT. Protein concentration is determined by measuring absorbance at 280 ⁇ . The extension coefficient for troponin C is 0.18.
  • Protein concentrations having the same reconstitution molar ratios of troponin subunits C, I, and T are maintained for all various combinations. These concentrations of the respective proteins are combined in a reconstitution buffer consisting of 0.1 M NaCl, 0.1 M CaCl2, 5 mM DTT, 5mM Hepes (pH 7.5) . Dialysis is for 20-24 hours at 4°C with three exchanges over a dialysis buffer consisting of 0.1 M NaCl, 0.1 m CaCl 2 , 0.5 mM DTT, and 5 mM Hepes (pH 7.5) . Protein concentration is approximated by measuring absorption at 278 ⁇ . The troponin trimer has an extension coefficient of 0.45 at 278 ⁇ .
  • the inhibitory effect of troponin subunit, fragment, or analog on the proliferation of bFGF-stimulated EC can be measured according to the following procedure .
  • Bovine capillary endothelial cells in DMEM/10% CS/1% GPS are plated onto each well of a 96-well pregelatinized tissue culture plate.
  • the cell media is changed to DMEM, 2% CS, 1% GPS, 0.5% BSA (complete medium) , supplemented with 10 ⁇ l of lmg/ml "cold" thymidine per 50 ml of medium.
  • test samples complete medium are added in duplicate.
  • bFGF is added in each well except for the appropriate controls, to a final concentration of 0.2 ng/well .
  • BALB/c 3T3 cells are trypsinized and resuspended at a concentration of 5 X 10 4 cells/ml. Aliquots of 200 ⁇ l are plated into 0.3 cm 2 microtiter wells (Microtest II tissue Culture Plates, Falcon) . After reaching confluence, in a period of 2 to 3 days, the cells are further incubated for a minimum of 5 days in order to deplete the media of growth promoting factors. These growth conditions yield confluent monolayers of non-dividing BALB/c 3T3 cells. Test samples are dissolved in 50 ⁇ l of 0.15 M NaCl and added to microtiter wells, along with [ 3 H]TdR.
  • autoradiography may be used to quantitate DNA synthesis by punching out the bottoms of the microtiter wells and mounting them on glass slides with silastic glue.
  • the slides are dipped in a 1 g/ml solution of NTB2 nuclear track emulsion (Kodak) and exposed for 3-4 days.
  • the emulsion is developed with Microdol-X solution (Kodak) for 10 minutes, rinsed with distilled H 2 0, and fixed with Rapid Fixer (Kodak) for three minutes.
  • the autoradiographs are stained with a modified Giemsa stain. At least 1000 nuclei are counted in each well and DNA synthesis, expressed as the percentage of nuclei labeled. Cell division is measured by counting the number of cells in microtiter wells with the aid of a grid after 40-48 hour incubations with test samples.
  • a quick and sensitive screen for inhibition of EC proliferation in response to treatment with a troponin subunit, analog, or derivative of the invention involves incubating the cells in the presence of varying concentrations of the inhibitor and determining the number of endothelial cells in culture based on the colorimetric determination of cellular acid phosphatase activity, described by Connolly, et al . , 1986, J. Anal . Biochem . 152:136-140.
  • bFGF angiogenesis factor
  • Capillary endothelial cells and Balb/c 3T3 cells were separately plated (2 x 10 3 /0.2 ml) onto gelatin-coated 96-well tissue culture dishes on day 1.
  • cells were refed with Dulbecco's modified Eagle's medium (Gibco) with 5% calf serum (Hyclone) (DMEM/5) and bFGF (10 ng/ l) (FGF Co.) and increasing concentrations of the troponin subunit. These substances were added simultaneously in volumes that did not exceed 10% of the final volume.
  • Wells containing phosphate buffered saline (PBS) (Gibco) alone and PBS + bFGF were included as controls.
  • PBS phosphate buffered saline
  • Troponin C inhibited bFGF-stimulated endothelial cell proliferation in a dose-dependent manner in all concentrations tested (FIG 1) .
  • Percent inhibition of bovine endothelial cell proliferation (“BCE”) was 54%, 86%, 83%, and
  • IC 50 represents the concentration at which 50% inhibition of bFGF growth factor- induced stimulation was observed.
  • the IC 50 of troponin C was
  • Troponin I inhibited bFGF-stimulated BCE proliferation at concentrations of 1 and 5 ug/well, but inhibition was not observed in the sample tested at 10 ug/well (FIG 2) .
  • the percent inhibition of BCE was 33% and
  • the IC 50 of troponin I was determined to be 1.14 ⁇ M.
  • Troponin T inhibited bFGF-stimulated EC proliferation at concentrations of 10 and 20 ug/well, but not at concentrations of 1 and 5 ⁇ g/well (FIG 3) .
  • 35 proliferation was inhibited 23% and 62% at 1.6 ⁇ M and 3.3 ⁇ M, respectively.
  • the IC 50 of troponin T was determined to be 2.14 ⁇ M.
  • the combination of troponin subunits C and I inhibited EC at all concentrations tested (FIG 4)
  • the percent inhibition of proliferation of BCE was 52%, 54% 73% and 47% at 130 nM, 645 nM, 1 3 ⁇ M and 2.6 ⁇ M, respectively
  • the IC C0 of this combination was determined to be 110 nM
  • troponin subunits C, I and T were observed to inhibit bFGF-stimulated BCE proliferation by 16% at a concentration of 360 nM (5 ug/well, FIG 5)
  • the troponin samples tested had no detectable inhibitory effect on the growth of Balb/c 3T3 cells, a non- endothelial cell type
  • Determination of the ability of the troponin subunit, derivative, or analog to inhibit the angiogenic process of capillary EC migration in response to an angiogenic stimulus can be determined using a modification of the Boyden chamber technique is used to study the effect of troponin subunit, derivative, or analog on capillary EC migration. Falk et al. , 1980, J. Immunol . 118 :239-247 (1980) .
  • a blind-well Boyden chamber consists of two wells (upper and lower) separated by a porous membrane J Exp . Med.
  • Migration is measured using blind well chamDers (Neuroprobe, no. 025-187) and polycarbonate membranes with £ micron pores (Nucleopore) precoated with fibronect (6 67 ⁇ g/ml in PBS) (human, Cooper) Basic FGF (Takeda Co ) diluted DMEM with 1% calf serum (DMEM/l) is added to the lower well at a concentration of 10 ng/ml .
  • the upper wells receive 5 x 10 5 capillary EC/ml and increasing concentrations of purified troponin subunit, derivative or analog is used within 24 hours of purification. Control wells receive 5 DMEM/l, either with or without bFGF.
  • the migration chambers are incubated at 37°C in 10% C0 2 for 4 hours.
  • the cells on the upper surface of the membrane are then wiped off by drawing the membrane over a wiper blade (Neuroprobe) .
  • Example 5 Inhibition of tumor growth as determined by a SCID mouse model system
  • SCID mice in a treatment and a control group of four mice each.
  • a dorsal subcutaneous implantation of 10 6 PC-3 cells was made and observed until a volume of between 100-400 mm 3 was attained. After the tumor reached the threshold volume, twice daily subcutaneous injections of
  • Figure 6 shows that 28 days of treatment resulted in an approximate 50% reduction in tumor volume in the treatment group compared to the tumor volume of the control
  • Example 6 Inhibition in vivo of Neovascularization by troponin as determined by the murine corneal pocket assay
  • Fibroblast Growth Factor 40 ng/pellet was placed in a corneal micropocket of a mouse.
  • the mouse received subcutaneous injections of recombinant Troponin I, 50 mg/kg every 12 hours beginning 48 hors prior to the implantation.
  • Corneal angiogenesis was evaluated by slit lamp microscopy. By day 6 angiogenesis in control eyes results in vessels that have extended into the pellet by day 6. At this time there was observed a 50% reduction in blood vessel density and a 30% inhibition in blood vessel length in the treated animals.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Veterinary Medicine (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biochemistry (AREA)
  • Toxicology (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention concerns methods of inhibiting atopic angiogenesis using troponin subunits and peptide fragments thereof. The invention further concerns pharmaceutical compositions comprising a troponin subunit or a fragment thereof. The pharmaceutical compositions and the methods of the invention are useful in the treatment of solid tumors, particularly tumors of the central nervous system and of the eye.

Description

TROPONIN SUBUN1TS AND FRAGMENTS USEFUL AS ANGIOGENESIS INHIBITORS
1. INTRODUCTION
The present invention provides for a novel pharmaceutical composition, and method of use thereof for the treatment of diseases or disorders involving abnormal angiogenesis.
More particularly, the present invention is based, in part, on the discovery that troponin subunits C, I and T inhibit stimulated endothelial cell proliferation. Pharmaceutical compositions containing therapeutically effective amounts of troponin C, I, or T, suounits, fragments, or analogs and methods of therapeutic use thereof are provided.
2. BACKGROUND
Angiogenesis, the process of new blood vessel development and formation, plays an important role in numerous physiological events, both normal and pathological.
Angiogenesis occurs in response to specific signals and involves a complex process characterized by infiltration of the basal lamina by vascular endothelial cells in response to angiogenic growth signal (s) , migration of the endothelial cells toward the source of the signal (s) , and subsequent proliferation and formation of the capillary tube. Blood flow through the newly formed capillary is initiated after the endothelial cells come into contact and connect w th a preexisting capillary.
The naturally occurring balance between endogenous stimulators and inhibitors of angiogenesis is one in which inhibitory influences predominate. Rastinejad et al . , 1989, Cell 5_6.:345-355. In those rare instances in which neovascularization occurs under normal physiological conditions, such as wound healing, organ regeneration, embryonic development, and female reproductive processes, angiogenesis is stringently regulated and spatially and temporally delimited. Under conditions of pathological angiogenesis such as that characterizing solid tumor growth, these regulatory controls fail . Unregulated angiogenesis becomes pathologic and sustains progression of many neoplastic and non-neoplastic diseases . A number of serious diseases are dominated by abnormal neovascularization including solid tumor growth and metastases, arthritis, some types of eye disorders, and psoriasis. See, e . g. , reviews by Moses et al . , 1991,
Biotech . 9.:630-634; Folkman et al . , 1995, N. Engl . J. Med . , 333 :1757-1763 ; Auerbach et al . , 1985, J. Microvaεc . Res . 29.:401-411; Folkman, 1985, Advances in Cancer Researcn, eds. Klein and einhouse, Academic Press, New York, pp. 175-203; Patz, 1982, Am. J. Opthalmol . .94:715-743; and Folkman et al . , 1983, Science 221 :719-725. In a number of pathological conditions, the process of angiogenesis contributes to the disease state. For example, significant data have accumulated which suggest that the growth of solid tumors is dependent on angiogenesis. Folkman and Klagsbrun, 1987, Science 235 :442-447.
The maintenance of the avascularity of the cornea, lens, and trabecular meshwork is crucial for vision as well as to ocular physiology. There are several eye diseases, many of which lead to blindness, in which ocular neovascularization occurs in response to the diseased state. These ocular disorders include diabetic retinopathy, neovascular glaucoma, inflammatory diseases and ocular tumors (e.g., retinoblastoma) . There are also a number of other eye diseases which are also associated with neovascularization, including retrolental fibroplasia, uveitis, retinopathy of prematurity, macular degeneration, and approximately twenty eye diseases which are associated with choroidal neovascularization and approximately forty eye diseases associated with iris neovascularization. See, e.g., reviews by altman et al . , 1978, Am . J. Ophthal . 8_5:704-710 and Gartner et al . , 1978, Surv. Ophthal . 22=291-312. Currently, the treatment of these diseases, especially once neovascularization has occurred, is inadequate and blindness often results. Studies have suggested that vaso-inhibitory factors which are present in normal ocular tissue (cornea and vitreous) are lost in the diseased state.
An inhibitor of angiogenesis could have an important therapeutic role in limiting the contributions of this process to pathological progression of the underlying disease states as well as providing a valuable means of studying their etiology. For example, agents that inhibit tumor neovascularization could play an important role in inhibiting metastatic tumor growth.
The components of angiogenesis relating to vascular endothelial cell proliferation, migration and invasion, have been found to be regulated in part by polypeptide growth factors. Experiments in culture, indicate that endothelial cells exposed to a medium containing suitable growth factors can be induced to evoke some or all of the angiogenic responses. Several polypeptides with in vi tro endothelial growth promoting activity have been identified. Examples include acidic and basic fibroblast growth factors, transforming growth factors α and β , platele -derived endothelial cell growth factor, granulocyte colony- stimulating factor, interleukin-8, hepatocyte growth factor, proliferin, vascular endothelial growth factor and placental growth factor. See, e.g., review by Folkman et al . , 1995, N. Engl . J. Med . , 333:1757-1763.
Although extracts from several different tissue sources have been shown to contain an i-angiogenic activity, several molecules such as platelet factor-4 , thrombospondin, protamine, and transforming growth factor B, have been found to negatively regulate different aspects of angiogenesis, such as cell proliferation or cell migration, no single tissue-derived macromolecule capable of inhibiting angiogenesis has been identified in the prior art. See, e. g. , reviews by Folkman, J., 1995, N. Engl . J. Med. 333 : 1757-1763 and D'Amore, 1985, Prog. Clin . Biol . Res . 221:269-283. There is therefore a great need for the further identification and characterization of chemical agents which can prevent the continued deregulated spread of vascularization and which would potentially have broad applicability as a therapy for those diseases in which neovascularization plays a prominent role.
Capillary endothelial cells ("EC") proliferate in response to an angiogenic stimulus during neovascularization. Ausprunk and Folkman, 1977, J". Microvasc . Res . l± : 153-65. An in vi tro assay assessing endothelial cell proliferation in response to known angiogenesis simulating factors, such as acidic or basic fibroblast growth factor (aFGF and bFGF, respectively) , has been developed to mimic the process of neovascularization in vi tro . This type of assay is the assay of choice to demonstrate the stimulation of capillary EC proliferation by various angiogenic factors. Shing et al . , 1984, Science 223 :1296-1298.
The process of capillary EC migration through the extracellular matrix towards an angiogenic stimulus is also a critical event required for angiogenesis. See, e . g . , review by Ausprunk et al . , 1977, J". Microvasc . Res . 14.:53-65. This process provides an additional assay by which to mimic the process of neovascularization in vi tro . A modification of the Boyden chamber technique has been developed to monitor EC migration. Boyden et al . , 1962, J. Exptl . Med . 115 :453-456, Example 4. To date, only a few tissue-derived EC cell migration inhibitors are known. See, e.g., review by Langer et al., 1976, Science 193 :70-72.
In the early 1970's, a number of in vivo angiogenesis model bioassays were widely used. These model systems included rabbit corneal pocket, chick chorioaliantoic membrane ("CAM") , rat dorsal air sac and rabbit air chamber bioassays. For review, see, Blood et al . , 1990, Biochem . et Biophys . Acta 1032 :89-118. The development of controlled release polymers capable of releasing large molecules such as angiogenesis stimulators and inhibitors was critical to the use of these assays. Langer et al . , 1976, Na ture 263 :797- 800.
In the CAM b oassay, fertilized chick embryos are cultured in Petπ dishes. On day 6 of development, a disc of a release polymer, such as methyl cellulose, impregnated with the test sample or an appropriate control substance is placed onto the vascular membrane at its advancing edge. On day 8 of development, the area around the implant is observed and evaluated. Avascular zones surrounding the test implant indicate the presence of an inhibitor of embryonic neovascularization. Moses et al . , 1990, Science, 248:1408- 1410 and Taylor et al. , 1982, Nature, 297:307-312. The reported doses for previously described angiogenesis inhibitors tested alone in the CAM assay are 50 μg of protamine (Taylor et al . (1982)) , 200 μg of bovine vitreous extract (Lutty et al . , 1983, Invest. Opthalmol . Vis . Sci . 24:53-56) , and 10 μg of platelet factor IV (Taylor et al . (1982)) . The lowest reported doses of angiogenesis inhibitors effective as combinations include heparin (50 μg) and hydrocortisone (60 μg) , and B-cyclodextπn tetradecasulfate (14 μg) and hydrocortisone (60 μg) , reported by Folkman et al. , 1989, Science 243 :1490.
According to the rabbit corneal pocket assay, polymer pellets of ethylene vinyl acetate copoly er ("EVAC") are impregnated with test substance and surgically implanted in a pocket in the rabbit cornea approximately 1 mm from the limbus. Langer et al . , 1976, Science 193 : 707-72. To test for an angiogenesis inhibitor, either a piece of carcinoma or some other angiogenic stimulant is implanted distal to the polymer 2 mm from the limbus. In the opposite eye of each rabbit, control polymer pellets that are empty are implanted next to an angiogenic stimulant in the same way. In these control corneas, capillary blood vessels start growing towards the tumor implant in 5-6 days, eventually sweeping over the blank polymer. In test corneas, the directional growth of new capillaries from the limbal blood vessel towards the tumor occurs at a reduced rate and is often inhibited such that an avascular region around the polymer is observed. This assay s quantitated by measurement of the maximum vessel lengths with a stereospecifIC microscope
Troponin, a complex of three polypeptides is an accessory protein that is closely associated with actin filaments in vertebrate muscle The troponin complex, acts in conjunction with the muscle form of tropomyosin to mediate the Ca2* dependency of myosm ATPase activity and thereoy regulate muscle contraction. The troponin polypeptides T, I, and C, are named for their tropomyosin binding, inhibitory, and calcium binding activities, respectively Troponin T binds to tropomyosin and is believed to be responsible for positioning the troponin complex on the muscle thin filament Troponin I binds to actin, and the complex formed by troponins I and T, and tropomyosin, inhibits the interaction of actin and myosm. Troponin C is capable of binding up to four calcium molecules. Studies suggest that when the level of calcium in the muscle is raised, troponin C causes troponin I to loose its hold on the actin molecule, causing the tropomyosin molecule shift, thereby exposing the myosm binding sites on actin and stimulating myosm ATPase activity. Prior to the discovery of the present invention, troponin subunits were not known to inhibit the process of endothelial eel proliferation The citation of a reference herein shall not be construed as an admission that such reference is prior art to the present invention.
3. SUMMARY OF THE INVENTION The present invention relates to pharmaceutical compositions containing troponin subunits C, I, or T, or fragments thereof, in therapeutically effective amounts that are capable of inhibiting endothelial cell proliferation The invention also relates to pharmaceutical compositions containing analogs of troponin subunits C, I, or T ana analogs of their fragments, in therapeutically effective amounts that are capable of inhibiting enαothelial cell proliferation. The invention further relates to treatment of neovascular disorders by administration of a therapeutic compound of the invention. Such therapeutic compounds (termed herein "Therapeutics"), include. troponin subunits C, I, and T, and fragments and analogs thereof. In one embodiment, a Therapeutic of the invention is administered to treat a cancerous condition, or to prevent progression from the pre-neoplastic or pre-malignant state into a neoplastic or a malignant state In other specific embodiments, a Therapeutic of the invention is administered to treat ocular disorders associated with neovascularization
3.1. Definitions
As used herein, The term "troponin subunit", when not preceding the terms C, I or T, means generically any of troponin subunits C, I, or T.
4. BRIEF DESCRIPTION OF THE FIGURES Figure 1. Inhibition of bovine capillary
Endothelial Cell (BCE) proliferation by troponin C Percent inhibition of bFGF-stimulated BCE proliferation is shown as a function of troponin C concentration (nM) Percent inhibition was determined by comparing results obtained for cells treated with stimulus alone with those obtained for samples exposed to both stimulus and inhibitor Well volume was 200 μl .
Figure 2. Inhibition of capillary BCE proliferation by troponin I. Percent inhibition of bFGF-stimulated BCE proliferation is shown as a function of troponin I concentration (nM) . Percent inhibition was determined as described in Figure 1. Well volume was 200 μ±
Figure 3. Inhibition of capillary BCE proliferation by troponin T Percent inhibition of bFGF-stimulated BCE proliferation is shown as a function of troponin T concentration (nM) Percent inhibition was determined as described in Figure 1 Well volume was 200 μ± Figure 4. Inhibition of BCE proliferation by troponins C and I. Percent inhibition of bFGF-stimulated BCE proliferation is shown as a function of troponin I and C concentration (nM) . Percent inhibition was determined as described in Figure 1. Well volume was 200 μl .
Figure 5. Inhibition of capillary BCE proliferation by troponin C, I and T. Percent inhibition of bFGF-stimulated BCE proliferation is shown as a function of troponin C, I, and T concentration (nM) . Percent inhibition was determined as described in Figure 1. Well volume was 200 μl.
Figure 6. Inhibition of tumor growth in SCID mice.
5. DETAILED DESCRIPTION OF THE INVENTION The present invention relates to therapeutic methods and compositions based on troponin subunits. The invention provides for treatment of neovascular disorders by administration of a therapeutic compound of the invention. Such therapeutic compounds (termed herein "Therapeutics") include: troponin C, I, and T subunits, fragments and analogs thereof (collectively "peptides of the invention") . The peptides of the invention are characterized by the property of inhibiting bovine endothelial cell proliferation in culture with an IC50 of 10 μM or less. In a preferred embodiment, a Therapeutic of the invention is administered to treat a cancerous condition, or to prevent progression from a pre-neoplastic or non-malignant state into a neoplastic or a malignant state. In other specific embodiments, a Therapeutic of the invention is administered to treat an ocular disorder associated with neovascularization.
In a preferred aspect, a Therapeutic of the invention is a peptide consisting of at least a fragment of troponin C, troponin I, troponin T, or troponins C and I, which is effective to inhibit endothelial cell proliferation. Examples of the troponin subunits that can be utilized in accordance with the invention, include the subunits of troponin from human fast twitch skeletal muscle, the sequences of which are given below:
Human Fast Twitch Skeletal Muscle Troponin C (SKQ ID NQ:1) l M T D Q Q A E A R S Y L S E E M I A E 21 K A A F D M F D A D G G G D I S V K E
Figure imgf000011_0001
Figure imgf000011_0002
Figure imgf000011_0003
In another embodiment, the invention encompasses peptides which are homologous to human fast-twitch skeletal troponin C (SEQ ID NO:l) or fragments thereof. In one embodiment, the ammo acid sequence of the peptide has at least 80% identity compared to the fragment of human fast twitch skeletal troponin C from which it is derived (the "prototype fragment") . In another embodiment, this identity is greater than 85%. In a more preferred embodiment, this identity is greater than 90% . In a most preferred embodiment, the ammo acid sequence of the peptide has at least 95% identity with the prototype fragment. Fragments can be at least 10 am o acids, and m preferred embodiments at least 50, 75, 100 and 120 ammo acids, respectively
In another embodiment, the invention encompasses peptides which are homologous to human fast-twitch skeletal troponin I (SEQ ID NO:2) or fragments thereof. In one embodiment, the ammo acid sequence of the peptide has at least 80% identity with the prototype human fast-twitch skeletal troponin I fragment. In another embodiment, this identity is greater than 85%. In a more preferred embodiment, this identity is greater than 90%. In a most preferred embodiment, the amino acid sequence of the peptide has at least 95% identity with the prototype fragment. Fragments can be at least 10 amino acids, and in preferred embodiments at least 50, 75, 100 and 120 amino acids, respectively.
In another embodiment, the invention encompasses peptides which are homologous to human fast-twitch skeletal troponin T (SEQ ID NO:3) or fragments thereof. In one embodiment, the ammo acid sequence of the peptide has at least 80% identity with the prototype human fast-twitch skeletal beta troponin T. In another embodiment, this identity is greater than 85% . In a more preferred embodiment, this identity is greater than 90%. In a most preferred embodiment, the ammo acid sequence of the peptide has at least 95% identity with the prototype fragment Fragments can be at least 10 ammo acids, and m preferred embodiments at least 50, 75, 100, 120 and 200 amino acids length, respectively.
In other specific embodiments, the peptides of the invention are troponin C, troponin I and troponin T suounits of the fast twitch, slow twitch and cardiac isoforms from other mammalian species, e.g., human, rabbit, rat, mouse, bovine, ovine and porcine.
In a specific embodiment, a Therapeutic of the invention is combined with a therapeutically effective amount of another molecule which negatively regulates angiogenesis which may be, but is not limited to, platelet factor 4, thrombospondin-l, tissue inhibitors of metalloproteases (TIMP1 and TIMP2) prolact (16-Kd fragment) , angiostat (38-Kd fragment of plasminogen) , bfGf soluble receptor, transforming growth factor β , mterferon alfa, and placental proliferm-related protein.
Paradoxically, neovascularization gradually reduces a tumors accessibility to chemotherapeutic drugs due to increased interstitial pressure within the tumor, which causes vascular compression and central necrosis. In vivo results have demonstrated that rodents receiving angiogenic therapy show increased delivery of chemotherapy to a tumor. Teicher et al . , 1994, Int . J. Cancer 57:920-925. Thus, in one embodiment, the invention provides for a pharmaceutical composition of the present invention in combination with a chemotherapeutic agent.
In another preferred aspect, a Therapeutic of the invention is combined with chemotherapeutic agents or radioactive isotope exposure. The invention is illustrated by way of examples infra which disclose, inter alia , the inhibition of capillary endothelial cell proliferation by troponin subunits C, I, and T and the means for determining inhibition of capillary endothelial cell migration and inhibition of neovascularization m vivo by troponin subunits. For clarity of disclosure, and not by way of limitation, the detailed description of the invention is divided into the subsections set forth below.
5.1. TROPONIN SUBUNITS, Fragments AND ANALOGS
The invention provides for pharmaceutical compositions comprising troponin subunits, fragments, and analogs thereof. In particular aspects, the subunits, fragments, or analogs are of fly, frog, mouse, rat, rabbit, pig, cow, dog, monkey, or human troponin subunits.
It is envisioned that troponin subunit fragments can be made by altering troponin sequences by substitutions, additions or deletions that provide for functionally equivalent molecules. These include, but are not limited to, troponin subunits, fragments, or analogs containing, as a primary amino acid sequence, all or part of the amino acid sequence of a troponin subunit including altered sequences in which functionally equivalent amino acid residues are substituted for residues within the sequence resulting in a silent change. For example, one or more amino acid residues within the sequence can be substituted by another amino acid of a similar polarity which acts as a functional equivalent, resulting in a silent alteration. Substitutes for an amino acid within the sequence may be selected from other members of the class to which the amino acid belongs. For example, the nonpolar (hydrophobic) amino acids include alanine, leucine, isoleucine, valine, proline, phenylalanine, tryptophan and methionine. The polar neutral amino acids include glycine, serine, threonine, cysteine, tyrosine, asparagine, and glutamine. The positively charged (basic) amino acids include arginine, lysine and histidine. The negatively charged (acidic) amino acids include aspartic acid and glutamic acid.
One embodiment of the invention provides for molecules consisting of or comprising a fragment of at least 10 (continuous) amino acids of a troponin subunit which is capable of inhibiting endothelial cell proli eration. In other embodiments, this molecule consists of at least 20 or 50 ammo acids of the troponin subunit. In specific embodiments, such molecules consist of or comprise fragments of a troponin subunit that at least 75, 120 or 200 ammo 5 acids.
In a preferred embodiment, the protein is a mammalian troponin subunit. In alternative embodiments, it is a mammalian troponin C, I, or T subunit.
The troponin subunit fragments and analogs of the
10 invention can be derived from tissue (see, for example,
Example 1; Ebashi et al. , 1968, J. Biochem. £4.-465; Yasui et al . , 1968, J. Biol . Chem . 243 :735; Hartshorne et al . , 1968, Biochem . Biophys . Res . Commun . 3.1:647; Shaub et al . , 1969, Biochem. J. 115:993 ; Greaser et al . , 1971, J. Biol . Chem .
15 211:4226-4733; Brekke et al., 1976, J. Biol . Chem . 251:866- 871; and Yates et al . , 1983, J. Biol . Chem . 258 :5770-5774) or produced by various methods known the art . The manipulations which result m their production can occur at the gene or protein level. For example, a cloned ti opomn
20 gene sequence coding for troponin subunits C, I, or T, can be modified by any of numerous strategies known in the art. Sambrook et al. , 1990, Molecular Cloning, A Laboratory Manual, 2d ed. , Cold Spring Harbor Laboratory, Cold Spring Harbor, New York. The sequence can be cleaved at appropriate
25 sites with restriction endonuclease (s) , followed by further enzymatic modification if desired, isolated, and ligated in vi tro . In the production of the gene encoding a derivative or analog of a troponin subunit, care should be taken to ensure that the modified gene remains within the same
30 translational reading frame as the troponin subunit gene, uninterrupted by translational stop signals, in the gene region where the desired troponin activity is encoded.
Additionally, the troponin subunit encoding nucleic acid sequence can be mutated m vi tro or in vivo, to create
35 and/or destroy translation, initiation, and/or termination sequences, or to create variations coding regions and/or form new restriction endonuclease sites or destroy preexisting ones, to facilitate further m vi tro modification. Any technique for mutagenesis known m tie art can be used, including, but not limited to, in vi tro site- directed mutagenesis (Hutchinson et al . , 1978, J. Biol . Chem . 5 253 :6551) , use of TAB* linkers (Pharmacia) , etc
Manipulations of troponin subunit C, I, or T sequence may also be made at the protein level Included within the scope of the invention are troponin subunit fragments or other fragments or analogs which are 0 differentially modified during or after translation, e . g . , by acetylation, phosphorylation, carboxylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to an antibody molecule or other cellular ligand, etc. Any of numerous chemical 5 modifications may be carried out by known techniques, including, but not limited to, specific chemical cleavage toy cyanogen bromide, trypsm, chymotryps , papam, V8 protease, NaBH4, acetylation, formylation, oxidation, reduction, etc In addition, fragments and analogs of troponin 0 subunits can be chemically synthesized. For example, a peptide corresponding to a portion of a troponin subunit which comprises the desired domain, or which mediates the desired activity m vi tro, can be synthesized by use of a peptide synthesizer. Furthermore, if desired, nonclassical 5 amino acids or chemical ammo acid analogs can be introduced as a substitution or addition into the troponin subunit sequence. Non-classical ammo acids include, but are not limited to, the D-isomers of the common ammo acids, α-ammo isobutyric acid, 4-ammobutyrιc acid, hydroxyprolme, 0 sarcosme, citrulline, cysteic acid, t-butylglycme, t- butylalanme, phenylglyc e, cyclohexylalamne, /3-alanme, designer ammo acids such as 0-methyl ammo acids, Cα-methyl ammo acids, and Nα-methyl ammo acids.
In a specific embodiment, the invention encompasses 5 a chimeric, or fusion, protein comprising a troponin sαbunit or fragment thereof (consisting of at least a domain cr motif of the troponin subunit that is responsible for
Figure imgf000016_0001
endothelial cell proliferation) joined at its amino or carboxy-terminus via a peptide bond to an ammo acid sequence of a different protein. Such a chimeric product can be made by ligating the appropriate nucleic acid sequences encoding
5 the desired amino acid sequences to each other by methods known in the art, in the proper coding frame, and expressing the chimeric product by methods commonly known in the art .
Alternatively, such a chimeric product may be made by protein synthetic techniques, e . g. , by use of a peptide synthesizer.
10
5.2. ASSAYS OF TROPONIN PROTEINS FRAGMENTS AND ANALOGS
The functional activity and/or therapeutically effective dose of troponin subunits, fragments and analogs,
.lb_ can be assayed in vi tro by various methods. These methods are based on the physiological processes involved in angiogenesis and while they are within the scope of the invention, they are not intended to limit the methods by which troponin subunits, fragments and analogs inhibiting
_- angiogenesis are defined and/or a therapeutically effective dosage of the pharmaceutical composition is determined.
For example, where one is assaying for the ability of troponin subunits, fragments, and analogs, to inhibit or interfere with the proliferation of capillary endothelial
._ cells (EC) in vi tro, various bioassays known in the art can be used, including, but not limited to, radioactive incorporation into nucleic acids, colorimetric assays and cell counting.
Inhibition of endothelial cell proliferation may be
_0 measured by colorimetric determination of cellular acid phosphatase activity or electronic cell counting. These methods provide a quick and sensitive screen for determining the number of endothelial cells in culture after treatment with the troponin subunit, derivative, or analog of the
invention, and an angiogenesis stimulating factor such as aFGF. The colorimetric determination of cellular acid phosphatase activity is described by Connolly et al . , 1986, J. Anal . Biochem. 152:136-140 According to this method, described in Example 3, capillary endothelial cells are treated with angiogenesis stimulating factors, such as aFGF, and a range of potential inhibitor concentrations These samples are incubated to allow for growth, and then harvested, washed, lysed in a buffer containing a phosphatase substrate, and then incubated a second time A basic solution is added to stop the reaction and color development is determined at 405 λ According to Connolly et al , a linear relationship is obtained between acid phosphatase activity and endothelial cell number up to 10,000 cells/sample Standard curves for acid phosphatase activity are also generated from known cell numbers in order to confirm that the enzyme levels reflect the actual EC numbers Percent inhibition is determined by comparing the cell number of samples exposed to stimulus with those exposed to both stimulus and inhibitor.
Colorimetric assays to determine the effect of troponin subunits C, I, and T on endothelial cell proliferation demonstrate that all three troponin subunits terefere with bFGF-stimulated endothelial cell proliferation.
Troponin C inhibited bFGF-stimulated endothelial cell proliferation in a dose-dependent manner m all concentrations tested (FIG 1) . Percent inhibition of bovine endothelial cell proliferation ("BCE") was 54%, 86%, 83%, and 100% at concentrations of 280 nM, 1.4 μm, 2.8 μM and 5.6 μM, respectively An inhibition of 100% was observed at a concentration of 20 μg/well (5.6 μM) IC50 represents the concentration at which 50% inhibition of aFGF growth factor- induced stimulation was observed. The ICS0 of troponin C was determined to be 278 nM.
Troponin I inhibited bFGF-stimulated BCE proliferation at concentrations of 1 and 5 ug/well, but inhibition was not observed the sample tested at 10 ug/well (FIG 2) The percent inhibition of BCE was 33% and 46% at concentrations of 240 nM and 1.2 μM, respectively. The ICc0 of troponin I was determined to be 1.14 μM.
Troponin T inhibited bFGF-stimulated EC proliferation at concentrations of 10 and 20 ug/well, but not at concentrations of 1 and 5 μg/well (FIG 3) . BCE proliferation was inhibited 23% and 62% at 1.6 μM and 3.3 μM, respectively. The IC50 of troponin T was determined to be 2.14 μM.
The combination of troponin subunits C and I inhibited EC at all concentrations tested (FIG 4) . The percent inhibition of BCE was 52%, 54% 73% and 47% at 130 nM, 645 nM, 1.3 μM and 2.6 μM, respectively. The IC50 of this combination was determined to be 110 nM.
The combination of troponin subunits C, I and T was observed to inhibit aFGF stimulated BCE proliferation by 16% at a concentration of 360 nM (5 ug/well, FIG 5) .
The troponins samples tested had no detectable inhibitory effect on the growth of Balb/c 3T3 cells, a non- endothelial cell type. The incorporation of radioactive thymidine by capillary endothelial cells represents another means by which to assay for the inhibition of endothelial cell proliferation by a potential angiogenesis inhibitor. According to this method, a predetermined number of capillary endothelial cells are grown in the presence of 3H-Thymidine stock, an angiogenesis stimulator such as for example, bFGF, and a range of concentrations of the angiogenesis inhibitor to be tested. Following incubation, the cells are harvested and the extent of thymidine incorporation is determined. See, Example 2.
The ability of varying concentrations of troponin subunits, fragments or analogs to interfere with the process of capillary endothelial cell migration in response to an angiogenic stimulus can be assayed using the modified Boyden chamber technique. See, Section 2 and Example 4, infra . Another means by which to assay the functional activity of troponin subunits, fragments and analogs, involves examining the ability of the compounds to inhibit the directed migration of capillary endothelial cells which ultimately results in capillary tube formation This ability may be assessed for example, using an assay in which capillary endothelial cells plated on collagen gels are challenged with the inhibitor, and determining whether capillary-like tube structures are formed by the cultured endothelial cells.
Assays for the ability to inhibit angiogenesis in vivo include the chick chorioallantoic membrane assay (see Section 2 and Example 5, infra ) and rat or rabbit corneal pocket assays. See, Polverini et al . , 1991, Methods Enzymol . 198 440-450 According to the corneal pocket assays, a tumor of choice is implanted into the cornea of the test: animal the form of a corneal pocket. The potential angiogenesis inhibitor is applied to the corneal pocket and the corneal pocket is routinely examined for neovascularization See, Section 2 and Example 6 infra .
One embodiment of the invention provides for combination of the troponin subunits, fragments, or analogs of the present invention to inhibit angiogenesis. Another embodiment provides for the combination of troponin subunits, fragments, or analogs with other angiogenesis inhibiting factors. Such angiogenesis inhibiting factors include, but are not limited to: angiostatic steroids, thrombospondm, platelet factor IV, transforming growth factor β , mterferons, tumor necrosis factor α, bovine vitreous extract, protamine, tissue inhibitors of metalloprotemases (TIMP-1 and TIMP-2) , prolactm (16-kd fragment) , angiostatm (38-kd fragment of plasmmogen) , bfGf soluble receptor, and placental proliferin-related protein. See, e.g., reviews by Folkman et al. , 1995, N. Engl . J. Med . 333:1757-1763 and Klagsbrun et al . , 1991, Annu . Rev. Physiol . 53 =217-239.
The therapeutically effective dosage for inhibition of angiogenesis m vivo, defined as inhibition of capillary endothelial cell proliferation, migration, and/or blood vessel ingrowth, may be extrapolated from m vi tro inhibition assays using the compositions of the invention above or m combination with other angiogenesis inhibiting factors. The effective dosage is also dependent on the method and means of delivery. For example, in some applications, as m tne treatment of psoriasis or diabetic retinopathy, the inhibitor is delivered in a topical-ophthalmic carrier. In other applications, as in the treatment of solid tumors, the inhibitor is delivered by means of a biodegradable, polymeric implant. The protein can also be modified, for example, by polyethyleneglycol treatment.
5.3. THERAPEUTIC USES
The invention provides for treatment of diseases or disorders associated with neovascularization by administration of a therapeutic compound of the invention. Such therapeutic compounds (termed herein "Therapeutics") include troponin subunits and fragments and analogs thereof (e.g., as described infra) .
5.3.1. MALIGNANCIES
Malignant and metastatic conditions which can be treated with the Therapeutic compounds of the present invention include, but are not limited to, the solid tumors listed in Table 1 (for a review of such disorders, see Fishman et al . , 1985, Medicine, 2d Ed., J.B. Lippmcott Co., Philadelphia) :
TABLE 1 MALIGNANCIES AND RELATED DISORDERS
Solid tumors 5 sarcomas and carcinomas fibrosarcoma myxosarcoma liposarcoma chondrosarcoma osteogenic sarcoma chordoma - angiosarcoma endotheliosarcoma lymphangiosarcoma lymphangioendotheliosarcoma synovioma mesothelioma
Ewing' s tumor leiomyosarcoma 5 rhabdomyosarcoma colon carcinoma pancreatic cancer breast cancer ovarian cancer prostate cancer squamous cell carcinoma basal cell carcinoma 0 adjenocarcmoma sweat gland carcinoma sebaceous gland carcinoma papillary carcinoma papillary adenocarcinomas cystadenocarcinoma medullary carcinoma 25 bronchogenic carcinoma renal cell carcinoma hepatoma bile duct carcinoma choriocarcinoma semino a embryonal carcinoma ,n Wilms' tumor cervical cancer testicular tumor lung carcinoma small cell lung carcinoma bladder carcinoma epithelial carcinoma glioma 35 astrocytoma medulloblastoma craniopharyngioma ependymoma Kaposi ' s sarcoma pmealoma hemangioblastoma acoustic neuroma oligodendroglioma menangioma melanoma neuroblastoma retinoblastoma
5.3.2. OCULAR DISORDERS Ocular disorders associated with neovascularization which can be treated with the Therapeutic compounds of the present invention include, but are not limited to: neovascular glaucoma diabetic retinopathy retinoblastoma retrolental fibroplasia uveitis retinopathy of prematurity macular degeneration corneal graft neovascularization
as well as other eye inflammatory diseases, ocular tumors and diseases associated with choroidal or iris neovascularization. See, e.g., reviews by Waltman et al . , 1978, Am. J. Ophthal . 5:704-710 and Gartner et al . , 1978, Surv. Ophthal . 22:291-312.
5.3.3. OTHER DISORDERS
Other disorders which can be treated with the Therapeutic compounds of the present invention include, but are not limited to, hemangioma, arthritis, psoriasis, angiofibroma, atherosclerotic plaques, delayed wound healing, granulations, hemophilic joints, hypertrophic scars, nonunion fractures, Osier-Weber syndrome, pyogenic granuloma, scleroderma, trachoma, and vascular adhesions.
- Δ . 5.4. DEMONSTRATION OF THERAPEUTIC OR PROPHYLACTIC UTILITY
The Therapeutics of the invention can be tested m vivo for the desired therapeutic or prophylactic activity as well as for determination of therapeutically effective dosage. For example, such compounds can be tested m suitable animal model systems prior to testing in humans, including, but not limited to, rats, mice, chicken, cows, monkeys, rabbits, etc. For m vivo testing, prior to administration to humans, any animal model system known in the art may be used.
5.5. THERAPEUTIC/PROPHYLACTIC
ADMINISTRATION AND COMPOSITIONS
The invention provides methods of treatment and prophylaxis) by administration to a subject an effective amount of a Therapeutic of the invention. In a preferred aspect, the Therapeutic is substantially purified as set forth in Example 1. The subject is preferably an animal, including, but not limited to, animals such as cows, pigs, chickens, etc., and is preferably a mammal, and most preferably human.
The invention further provides methods of treatment by administration to a sub ect, an effective amount of a Therapeutic of the invention combined with a chemotherapeutic agent and/or radioactive isotope exposure.
The invention also provides for methods of treatment of a Therapeutic of the invention for patients who have entered a remission in order to maintain a dormant state.
Various delivery systems are known and can be used to administer a Therapeutic of the invention, e.g., encapsulation in liposo es, microparticles, microcapsules, receptor-mediated endocytosis (see, e.g., Wu and u, 1987, J.
Biol . Chem. 262 =4429-4432) . Methods of introduction include, but are not limited to , topical , tradermal , intramuscular , mtraperitoneal , intravenous , subcutaneous , tranasal , epidural, ophthalmic, and oral routes. The compounds may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents. It is preferred that administration is localized, but it may be systemic. In addition, it may be desirable to introduce the pharmaceutical compositions of the invention into the central nervous system by any suitable route, including mtraventπcular and intrathecal injection; mtraventricular injection may be facilitated by an intraventricular catheter, for example, attached to a reservoir, such as an Ommaya reservoir. Pulmonary administration can also be employed, e . g. , by use of an inhaler or nebulizer, and formulation with an aerosolizing agent.
In a specific embodiment, it may be desirable to administer the pharmaceutical compositions of the invention locally to the area m need of treatment; this may be achieved by, for example, and not by way of limitation, local infusion during surgery, topical application, e.g., in conjunction with a wound dressing after surgery, by injection, by means of a catheter, by means of a suppository, or by means of an implant, said implant being of a porous, non-porous, or gelatinous material, including membranes, sucn as sialastic membranes, or fibers. In one embodiment, administration can be by direct injection at the site (or former site) of a malignant tumor or neoplastic or pre- neoplastic tissue. For topical application, the purified troponin subunit is combined with a carrier so that an effective dosage is delivered, based on the desired activity (i.e., ranging from an effective dosage, for example, of 1.0 μM to 1.0 mM to prevent localized angiogenesis, endothelial cell migration, and/or inhibition of capillary endothelial cell proliferation. In one embodiment, a topical troponin subunit, fragment or analog s applied to the skin for treatment of diseases such as psoriasis. The carrier ~ay m the form of, for example, and not by way of limitation an ointment, cream, gel, paste, foam, aerosol, suppository, pad or gelled stick. A topical Therapeutic for treatment of some cf the eye disorders discussed infra consists of an effective amount of troponin subunit, fragment, or analog, m a ophthalmologically acceptable excipient such as buffered saline, mineral oil, vegetable oils such as corn or arachis oil, petroleum jelly, Miglyol 182, alcohol solutions, or liposomes or liposome-like products. Any of these compositions may also include preservatives, antioxidantε, antibiotics, immunosuppressants, and other biologicallv or pharmaceutically effective agents which do not exert a detrimental effect on the troponin subunit.
For directed internal topical applications, for example for treatment of ulcers or hemorrhoids, the troponin subunit, fragment, or analog composition may be in the form of tablets or capsules, which can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystallme cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as algmic acid, Primogel, cr corn starch; a lubricant such as magnesium stearate or Sterotes, or a glidant such as colloidal silicon dioxide. When the dosage unit form is a capsule, it can contain, in addition to material of the above type, a liquid carrier such as a fatty oil. In addition, dosage unit forms can contain various other materials which modify the physical form of the dosage unit, for example, coatings of sugar, shellac, or other enteric agents.
Suppositories generally contain active ingredient m the range of 0.5% to 10% by weight; oral formulations preferably contain 10% to 95% active ingredient. In another embodiment, the Therapeutic can be delivered in a vesicle, in particular a liposome See, Langer et al . , 1990, Sci ence 249 :1527-1533 , Treat et a. , 1989, in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein and Fidler (eds.) , Liss, New York, pp. 353-365; Lopez-Berestein, ibid. , pp. 317-327.
In yet another embodiment, the Therapeutic can be delivered in a controlled release system. In one embodiment, an infusion pump may be used to administer troponin subunit, such as for example, that used for delivering insulin or chemotherapy to specific organs or tumors ( see Langer, supra ; Sefton, CRC Cri t . Ref . Biomed . , 1987, Eng. 14.:201; Buchwald et al., 1980, Surgery 88:507; Saudek et al . , 1989, N. Engl . J. Med . 321:574.
In a preferred form, the troponin subunit, fragment, or analog is administered in combination with a biodegradable, biocompatible polymeric implant which releases the troponin subunit, fragment, or analog over a controlled period of time at a selected site. Examples of preferred polymeric materials include polyanhydrides, polyorthoesters, polyglycolic acid, polylactic acid, polyethylene vinyl acetate, and copolymers and blends thereof. See, Medical Applications of Controlled Release, Langer and Wise (eds.) , 1974, CRC Pres., Boca Raton, Florida; Controlled Drug Bioavailability, Drug Product Design and Performance, Smolen and Ball (eds.) , 1984, Wiley, New York; Ranger and Peppas, 1983, J. Macromol . Sci . Rev. Macromol . Chem . 22_ -. ξ> \ ; see also Levy et al . , 1985, Science 228 :190; During et al . , 1989, Ann . Neurol . 25:351; Howard et al. , 1989, J. Neurosurg. 71.-105. In yet another embodiment, a controlled release system can be placed in proximity of the therapeutic target, i . e . , the brain, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, 1989, supra, vol. 2, pp. 115-138) .
Other controlled release systems are discussed in the review by Langer (1990, Science 249 : 1527-1533) .
The present invention also provides pharmaceutical compositions. Such compositions comprise a therapeutically effective amount of a Therapeutic, and a pharmaceutically acceptable carrier. The pharmaceutical compositions of the invention can be formulated as neutral or salt forms. Pharmaceutically acceptable salts include those formed with free amino groups such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaπc acids, etc., and those formed with free carboxyl groups such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, lsopropylamme, triethylamme, 2-ethylammo ethanol, histidine, procaine, etc. In a specific embodiment, the term
"pharmaceutically acceptable" means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognizeα pnarmacopeia for use in animals, and more particularly in humans The term "carrier" refers to a diluent, adjuvant, excipient, or vehicle with which the therapeutic is administered. Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents Water is a preferred carrier when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for ectable solutions. Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like. The composition, if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents such as acetates, citrates or phosphates Antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite, chelatmg agents such as ethylenediammetetraacetic acid; and agents for the adjustment of tonicity such as sodium chloride or dextrose are also envisioned. The parental preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
These compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like. The composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides, microcrystallme cellulose, gum tragacanth or gelatin. Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Examples of suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences" by E.W. Martin. Such compositions will contain a therapeutically effective amount of the Therapeutic, preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the patient. The formulation should suit the mode of administration. In a preferred embodiment, the composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous administration to human beings. Typically, compositions for intravenous administration are solutions in sterile lsotonic aqueous buffer. Where necessary, the composition may also include a solubilizing agent and a local anesthetic such as lignocaine to ease pain at the site of the injection. Generally, the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent. Where the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline. Where the composition is administered by injection, an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
The amount of the Therapeutic of the invention which will be effective in the treatment of a particular disorder or condition will depend on the nature of the disorder or condition, and can be determined by standard clinical techniques. In addition, m vi tro assays such as those discussed in section 5.2 may optionally be employed to help identify optimal dosage ranges. The precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness of the disease or disorder, and should be decided according to the judgment of the practitioner and each patient's circumstances However, suitable dosage ranges for intravenous administration are generally about 20-500 micrograms of active compound per kilogram body weight. Suitable dosage ranges for intranasal administration are generally about 0.01 pg/kg body weight to 1 mg/kg body weight. Effective doses may be extrapolated from dose-response curves derived from in vi tro or animal model test bioassays or systems
The invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention. Optionally associated with such container (s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration. Modifications and variations of the compositions of the present invention, and methods for use, will be obvious to those skilled in the art from the foregoing detailed description Such modifications and variations are intended to fall within the scope of the appended claims The following non-limiting examples demonstrate the discovery of troponin subunit inhibition of angiogenic stimulus induced endothelial cell proliferation, and means for determining the effective dosage of troponin subunit, fragment, or analog to inhibit angiogenesis, as well as for identifying troponin subunit fragments and analogs (i.e., tnose fragments or analogs of troponin subunit capable of mnibitmg angiogenesis. The troponin subunit used in tne examples is purified as described infra .
6. EXAMPLES
Example 1: Purification of Troponin subunit Components
Cardiac Troponin Isolation from Tissue
The procedures of Ebashi et al . , 1968, J. Biochem . 64:465-477; Yasui et al . , 1968, J. Biol . Chem . 243 :735-742; Hartshorne et al. , 1969, Biochim . Biophys . Acta , 175 : 0 ; Schaub et al . , 1969, Biochem . J. 115: 993-1004; Greaser et al., 1971, J. Biol . Chem . 246 :4226-4233 ; and Greaser et al . , 1973, J. Biol . Chem . 248 =2125-2133 for purifying troponin can be used. Rabbit back and leg muscles are removed, cleaned of fat and connective tissue, and ground. The ground muscle (1 kg) is stirred for 5 mm. in 2 liters of a solution containing 20 mM KCl, 1 mM KHC03, 0.1 mM CaCl2, and 0.1 mM DTT." The suspension is filtered through cheesecloth, and the washing of the residue is repeated four times Two liters of 95% ethanol are then added to the washed residue and the solution filtered after 10 mm. The ethanol extraction is repeated twice. The residue is then washed 3 times with 2 liters of diethyl ether for 10 min. Finally the residue is allowed to dry at room temperature for 2 to 3 hours.
The dried powder (from 1 kg of muscle) is extracted overnight at 22° with 2 liters of a solution containing 1 M KCl, 25 mM Tris (pH 8.0), 0.1 M CaCl2, and 1 mM DTT After
The abbreviations used are: DDT, dithiothreitol, EGTA, ethylene glycol bis (β-aminoethyl ether) - N, N' - tetraacetate; SDS, sodium dodecyl sulfate, SE-, sulfoethyl . filtration through cheesecloth, the residue is once more extracted with 1 liter of 1 M KCl.
The extracts are combined and cooled to 4°C. Solid ammonium sulfate is added to produce approximately 40% saturation (230 g per liter) . After 30 mm. the solution is centrifuged and 125 g of ammonium sulfate is then added per liter of supernatant (60% saturation) . After centrifugation the precipitate is dissolved in 500 ml of a solution containing 5 mM Tris (pH 7.5) , 0.1 mM CaCl2, and 0.1 mM DTT and dialyzed against 15 liters of the same solution for 6 hours and against a fresh solution overnight.
Solid KCl is added to a final concentration of 1 M and 1 M KCl solution is added to bring the volume to 1 liter. The pH is then adjusted to .6 by addition of HCl, and the tropomyosin precipitate is removed by centrifugation. The pH of the supernatant is adjusted to 7.0 with KOH, and 450 g of ammonium sulfate were added per liter (70% saturation) . The precipitate is dissolved in a solution containing 5 mM Tris (pH 7.5, 0.1 mM CaCl2, and 0.1 mM DTT, and dialyzed overnight against the same solution. Solid KCl is added to bring its concentration to 1 M, the pH adjusted to 4.6, and the precipitate formed removed by centrifugation. The neutralized supernatant is dialyzed against 2 mM Tris (pH 7.5) until the Nessler reaction is negative. The final yield of troponin is usually 2.5 to 3.0 g per kg of fresh muscle.
Cardiac Troponin Isolation from Tissue
Bovine hearts are obtained approximately 30 min. after death and immediately cut open, rinsed of blood, and immersed in ice. The left ventricle is removed, trimmed of excess fat and connective tissue, and ground. All subsequent extraction and preparation steps are performed at 0-3° except where noted. The ground muscle (500 g) is homogenized in a Waring Blender for 1 min. in 2.5 liters of solution containing 0.09 M KH2P04, 0.06 M K2HP0,, 0.3 M KCl, 5 mM 2- mercaptoethanol, pH 6.8. The homogenized muscle suspension is then stirred for 30 min. and centrifuged at 1000 x g for 20 mm. The precipitate is re-extracted for 30 mm. and centrifuged. The residue is then washed with 2.5 liters of 5 mM 2-mercaptoethanol and centrifuged at 1000 x g for 10 mm., followed by two successive washings and centrifugations with 1.5 liters of 50 mM KCl, 5mM Tris-HCl (pH 8.1/5) , mM 2- mercaptoethanol. The residue is then washed and centrifuged twice with 1.5 liters of 50 mM Tris-HCl (pH 8.1) , and 5 mM 2- mercaptoethanol. The volume of the residue is measured, and the residue is mixed with 0.5 volume of 3 M KCl, 50 mM Tris- HC1 (pH 8.1) , and 5 mM 2-mercaptoethanol . After a 16- to 20- hour extraction at 0°, the suspension is centrifuged at 15,000 x g for 10 min. The sediment is discarded, and the supernatant is adjusted to pH 7.6 with 0.05 N HCl . The filamentous precipitate which forms upon pH adjustment is removed by filtering the extract through nylon gauze. The protein that precipitates between 30 and 50% ammonium sulfate saturation is collected, dissolved in a solution containing 1 M KCl, and ImM potassium phosphate (pH 6.8) , and 5 mM 2- mercaptoethanol, and dialyzed against the same solution for 4 hours and against a fresh solution overnight. The protein solution is clarified by centrifugation at 105,000 x g for 30 min. The troponin is then purified by chromatography on a hydroxylapatite column with the protein being eluted between 0.08 and 0.10 M phosphate. Greaser et al . , 1972 Cold Spring Harbor Symp . Quant . Biol . 3_7:235-244. Rabbit cardiac troponin is prepared in a similar manner using a pooled batch of hearts which has been stored at -20°C prior to extraction.
The troponin subunits are separated by DEAE- Sephadex chromatography in 6 M urea. Bovine cardiac tropomyosin is prepared from the 50% ammonium sulfate saturation supernatant from the troponin extraction scheme (see above) . Ammonium sulfate is added to 65% saturation, and the precipitate is dissolved in and dialyzed versus 1 M KCl, 1 mM potassium phosphate (pH 7.0) , and 5 mM 2- mercaptoethanol. The protein is then purified by hydroxylapatite chromatography. Protein Determination - Protein concentrations are determined by the biuret method of Gornall et al. using bovine serum albumin as a standard. Gornall et al . , 1949, J. Biol . Chem . , Ill :751-766. Separation of Components - A sequence of SP-
Sephadex and DEAE-Sephadex chromatography gives complete separation of the three cardiac troponin components.
Recombinant Troponin Isolation and Reconstitution Protocols Troponin I and T
DNA encoding various troponin subunits and isoforms are known in the art. See, e.g., Wu et al . , 1994, DNA Cell . Biol . 12:217-233; Schreier et al . , 1990, J. Biol . Chem . 265 :21247-21253 ; and Gahlmann et al . , 1990, J. Biol . Chem. 265:12520-12528.
To express a troponin subunit, DNA encoding the subunit is subcloned into a high copy number expression plasmid, such as KP3998, using recombinant techniques known in the art . To express the cloned cDNA, E. coli transformed with the sert-contaming pKP1500 vector is grown overnight at 37°C, then inoculated into 4 liters of Luria-Bertani broth (LB) medium and grown at 42°C until mid-log phase. Isopropyl- l-thio-S-D-galactopyranosιde is then added to 0.5 M, and the culture is allowed to grow at 42°C overnight. Purification of expressed troponin subunit, fragment, or analog may be adapted from published procedures (Reinach et al . , 1988, J. Biol . Chem . 250:4628-4633 and Xu et al . , 1988, J. Biol . Chem . 263 :13962-13969) . The cells are harvested by centrifugation and suspended in 20 ml of 20 mM Tris, 20% sucrose, 1 mM EDTA, 0.2 mM phenylmethylsulfonyl fluoride, 1 mg/ml lysozy e, pH 7.5. After incubation on ice for 30 min., 80 ml of 20 mM Tris, 1 mM EDTA, 0.2 mM phenylmethylsulfonyl fluoride, 0.5 mM DTT is added and the cells broken in a French press (SLM Instruments) . The cell debris is pelleted; the supernatant is made 35% in saturated (NH4)2S04 and stirred on ice for 30 mm. After sedimentation, the supernatant is made 50 mM in NaCl, 5 mM in CaCl2, 1 mM in MgCl2, and 1 mM in DTT and then loaded onto a 1.5 X 25-cm phenyl-Sepharose (Pharmacia LKB Biotechnology Inc.) column. The column is washed first with 50 mM Tris, 50 mM NaCl, 5 mM CaCl2, 1 mM MgCl2, 1 M DTT, pH 7.5, then with 50 mM Tris, 1 mM NaCl, 0.1 mM CaCl2, 1 mM DTT, pH 7.5, until no more protein is eluted. The crude troponin subunit is then eluted with 50 mM Tris, 1 mM EDTA, 1 mM DTT, pH 7.5. Fractions that contained troponin subunit, fragment, or analog are pooled, dialyzed against 25 mM Tris, 6 M urea (United States Biochemical Corp.) , 1 mM MgCl2, 1 mM DTT, pH 8.0, and loaded onto a 1.5 X 25-cm DE52 (Whatman) column. The column is eluted with a 0-0.6 M NaCl linear gradient. Troponin subunit, fragment, or analog eluted from the column is dialyzed against 0.1 mM NH4HC03, 1 mM 3-mercaptoethanol, lyophilized, and stored. Purity is assessed by SDS- polyacrylamide gel electrophoresis and UV spectrophotometry. Typical yields of 6 mg of purified recombinant troponin subunit, fragment, or analog/liter of bacterial culture are expected. The lyophilized recombinant protein is resuspended in a take up buffer consisting of 6M urea, 20 mM Hepes (pH 7.5), 0.5M NaCl, 2mM EDTA, and 5mM DTT. The mixture is nutated at room temperature for 1 hour. The solution is then dialyzed at 4°C for six hours with 1 exchange against a dialysis buffer consisting of 0.5M NaCl, 20mM Hepes (pH 7.5) , and 0.5mM DTT.
Protein concentration is determined for each subunit at 280λ. The extension coefficient of Troponin I is 0.40 and Troponin T is 0.50.
Troponin C
The lyophilized recombinant protein is resuspended in a take up buffer consisting of 0.1 M NaCl, 20 mM Hepes (pH 7.5) , 2mM EDTA, and 5mM DTT. This solution is dialyzed for 6 hours at 4°C with one exchange against a dialysis buffer of 0.1 M NaCl, 20 mM Hepes (pH 7.5), and 0.5 mM DTT. Protein concentration is determined by measuring absorbance at 280λ. The extension coefficient for troponin C is 0.18.
Reconstitution of Combined Subunits:
Protein concentrations having the same reconstitution molar ratios of troponin subunits C, I, and T are maintained for all various combinations. These concentrations of the respective proteins are combined in a reconstitution buffer consisting of 0.1 M NaCl, 0.1 M CaCl2, 5 mM DTT, 5mM Hepes (pH 7.5) . Dialysis is for 20-24 hours at 4°C with three exchanges over a dialysis buffer consisting of 0.1 M NaCl, 0.1 m CaCl2, 0.5 mM DTT, and 5 mM Hepes (pH 7.5) . Protein concentration is approximated by measuring absorption at 278λ. The troponin trimer has an extension coefficient of 0.45 at 278λ.
Example 2 : Inhibition of Endothelial Cell
Proliferation measured by DNA synthesis.
The inhibitory effect of troponin subunit, fragment, or analog on the proliferation of bFGF-stimulated EC can be measured according to the following procedure .
Endothelial cell DNA Synthesis;
On day one, 5,000 Bovine capillary endothelial cells in DMEM/10% CS/1% GPS are plated onto each well of a 96-well pregelatinized tissue culture plate. On day two, the cell media is changed to DMEM, 2% CS, 1% GPS, 0.5% BSA (complete medium) , supplemented with 10 μl of lmg/ml "cold" thymidine per 50 ml of medium. On day three, test samples complete medium are added in duplicate. Additionally, bFGF is added in each well except for the appropriate controls, to a final concentration of 0.2 ng/well . On day four, 5 ul of 1:13 diluted H-Thymιdine stock is added to each well and the plate is incubated for 5-6 hours. Following incubation, the medium is aspirated, and the remainder is rinsed once with PBS, then twice for 5 minutes each with methanol followed by two rinses each for 10 minutes with 5% TCA. The cells well contents are then rinsed with water three times, dried to the plate, and lOOμl of 0.3 N NaOH is added to each well. The contents of the well are then transfered to the scintillation counter vials and 3 mis of Ecolume added to each vial. Samples are then counted on the scintillation counter.
3T3 Cell DNA Synthesis:
DNA synthesis in bFGF-stimulated 3T3 cells provides a control with which to evaluate results obtained for bFGF stimulated endothelial cell proliferation. DNA synthesis in the 3T3 cells can be determined according to the following method.
BALB/c 3T3 cells are trypsinized and resuspended at a concentration of 5 X 104 cells/ml. Aliquots of 200 μl are plated into 0.3 cm2 microtiter wells (Microtest II tissue Culture Plates, Falcon) . After reaching confluence, in a period of 2 to 3 days, the cells are further incubated for a minimum of 5 days in order to deplete the media of growth promoting factors. These growth conditions yield confluent monolayers of non-dividing BALB/c 3T3 cells. Test samples are dissolved in 50 μl of 0.15 M NaCl and added to microtiter wells, along with [3H]TdR. After an incubation of at least 24 hours, the media is removed and the cells are washed in PBS. Fixation of the cells and removal of unincorporated [3H] TdR is accomplished by the following successive steps; addition of methanol twice for periods of 5 minutes, 4 washes with H20, addition of cold 5% TCA twice for periods of 10 minutes, and 4 washes with H20. DNA synthesis is measured either by liquid scintillation counting or by autoradiography using a modification of the method described by Haudenschild et al . , 1976, M. Exp. Cell Res . 9_8:175. For scintillation counting, cells are lysed in 150 μl of 0.3 N NaOH and counted in 5 ml. of Insta-Gel liquid scintillation cocktail (Packard) using a Packard Tri-Carb liquid scintillation counter.
Alternatively, autoradiography may be used to quantitate DNA synthesis by punching out the bottoms of the microtiter wells and mounting them on glass slides with silastic glue. The slides are dipped in a 1 g/ml solution of NTB2 nuclear track emulsion (Kodak) and exposed for 3-4 days. The emulsion is developed with Microdol-X solution (Kodak) for 10 minutes, rinsed with distilled H20, and fixed with Rapid Fixer (Kodak) for three minutes. The autoradiographs are stained with a modified Giemsa stain. At least 1000 nuclei are counted in each well and DNA synthesis, expressed as the percentage of nuclei labeled. Cell division is measured by counting the number of cells in microtiter wells with the aid of a grid after 40-48 hour incubations with test samples.
Example 3 : Inhibition of Endothelial Cell
Proliferation measured by colorimetric determination of cellular acid phosphatase activity and electronic cell counting
A quick and sensitive screen for inhibition of EC proliferation in response to treatment with a troponin subunit, analog, or derivative of the invention involves incubating the cells in the presence of varying concentrations of the inhibitor and determining the number of endothelial cells in culture based on the colorimetric determination of cellular acid phosphatase activity, described by Connolly, et al . , 1986, J. Anal . Biochem . 152:136-140. We measured the effect of troponin on the proliferation of capillary endothelial cells (EC) in an assay which measures the ability of this protein to interfere with stimulation of endothelial cell proliferation by a known angiogenesis factor (bFGF) . Capillary endothelial cells and Balb/c 3T3 cells were separately plated (2 x 103 /0.2 ml) onto gelatin-coated 96-well tissue culture dishes on day 1. On day 2, cells were refed with Dulbecco's modified Eagle's medium (Gibco) with 5% calf serum (Hyclone) (DMEM/5) and bFGF (10 ng/ l) (FGF Co.) and increasing concentrations of the troponin subunit. These substances were added simultaneously in volumes that did not exceed 10% of the final volume. Wells containing phosphate buffered saline (PBS) (Gibco) alone and PBS + bFGF were included as controls. On day 5, media was removed and cells were washed with PBS and lysed in 100 μl of buffer containing 0.1 M sodium acetate (pH 5.5) , 0.1% Triton X-100""1 and 100 mM 5 p-nitrophenyl phosphate (Sigma 104 phosphatase substrate) . After incubation for 2 hours at 37°C, the reaction was stopped with the addition of 10 μl of 1 N NAOH. Color development was determined at 405 nm using a rapid microplate reader (Bio-Tek) . 0 Percent inhibition was determined by comparing the cell number of wells exposed to stimulus with those exposed to stimulus and troponin subunits.
All three troponin subunits were found to inhibit bFGF-stimulated EC proliferation, as measured by the
15 colorimetric assay.
Troponin C inhibited bFGF-stimulated endothelial cell proliferation in a dose-dependent manner in all concentrations tested (FIG 1) . Percent inhibition of bovine endothelial cell proliferation ("BCE") was 54%, 86%, 83%, and
20 100% at concentrations of 280 nM, 1.4 μM, 2.8 μM and 5.6 μM, respectively. An inhibition of 100% was observed at a concentration of 20 ug/well (5.6 μM) . IC50 represents the concentration at which 50% inhibition of bFGF growth factor- induced stimulation was observed. The IC50 of troponin C was
25 determined to be 278 nM.
Troponin I inhibited bFGF-stimulated BCE proliferation at concentrations of 1 and 5 ug/well, but inhibition was not observed in the sample tested at 10 ug/well (FIG 2) . The percent inhibition of BCE was 33% and
30 46% at concentrations of 240 nM and 1.2 μM, respectively. The IC50 of troponin I was determined to be 1.14 μM.
Troponin T inhibited bFGF-stimulated EC proliferation at concentrations of 10 and 20 ug/well, but not at concentrations of 1 and 5 μg/well (FIG 3) . BCE
35 proliferation was inhibited 23% and 62% at 1.6 μM and 3.3 μM, respectively. The IC50 of troponin T was determined to be 2.14 μM. The combination of troponin subunits C and I inhibited EC at all concentrations tested (FIG 4) The percent inhibition of proliferation of BCE was 52%, 54% 73% and 47% at 130 nM, 645 nM, 1 3 μM and 2.6 μM, respectively The ICC0 of this combination was determined to be 110 nM
The combination of troponin subunits C, I and T was observed to inhibit bFGF-stimulated BCE proliferation by 16% at a concentration of 360 nM (5 ug/well, FIG 5)
The troponin samples tested had no detectable inhibitory effect on the growth of Balb/c 3T3 cells, a non- endothelial cell type
Example 4: Inhibition of Capillary Endothelial Cell Migration by troponin
Determination of the ability of the troponin subunit, derivative, or analog to inhibit the angiogenic process of capillary EC migration in response to an angiogenic stimulus, can be determined using a modification of the Boyden chamber technique is used to study the effect of troponin subunit, derivative, or analog on capillary EC migration. Falk et al. , 1980, J. Immunol . 118 :239-247 (1980) . A blind-well Boyden chamber, consists of two wells (upper and lower) separated by a porous membrane J Exp . Med. 115 :453-456 (1962) A known concentration of growth factor is placed n the lower wells and a predetermined number of cells and troponin subunit, derivative, or analog is placed m the upper wells Cells attach to the upper surface of the membrane, migrate through and attach to the lower membrane surface The membrane can then be fixed and stained for counting, using the method of Glaser et al , 1980 , Na ture 288 =483-484.
Migration is measured using blind well chamDers (Neuroprobe, no. 025-187) and polycarbonate membranes with £ micron pores (Nucleopore) precoated with fibronect (6 67 μg/ml in PBS) (human, Cooper) Basic FGF (Takeda Co ) diluted DMEM with 1% calf serum (DMEM/l) is added to the lower well at a concentration of 10 ng/ml . The upper wells receive 5 x 105 capillary EC/ml and increasing concentrations of purified troponin subunit, derivative or analog is used within 24 hours of purification. Control wells receive 5 DMEM/l, either with or without bFGF. The migration chambers are incubated at 37°C in 10% C02 for 4 hours. The cells on the upper surface of the membrane are then wiped off by drawing the membrane over a wiper blade (Neuroprobe) . The cells which have migrated through the membrane onto the lower
10 surface are fixed in 2% glutaraldehyde followed by methanol (4°C) and stained with hematoxylin. Migration is quantified by counting the number of cells on the lower surface in 16 oil immersion fields and comparing this number with that obtained for the control .
15
Example 5 : Inhibition of tumor growth as determined by a SCID mouse model system
The effects of recombinant troponin I on the growth of human PC-3 prostatic carcinoma cells were determined in
_0 immunodeficient (SCID) mice in a treatment and a control group of four mice each. A dorsal subcutaneous implantation of 106 PC-3 cells was made and observed until a volume of between 100-400 mm3 was attained. After the tumor reached the threshold volume, twice daily subcutaneous injections of
_ς 50mg/kg recombinant troponin I was begun in the treatment group.
Figure 6 shows that 28 days of treatment resulted in an approximate 50% reduction in tumor volume in the treatment group compared to the tumor volume of the control
30 group.
Example 6: Inhibition in vivo of Neovascularization by troponin as determined by the murine corneal pocket assay
A pellet of sucrose octasulfate, Hydron™ and basic
35 Fibroblast Growth Factor (40 ng/pellet) was placed in a corneal micropocket of a mouse. The mouse received subcutaneous injections of recombinant Troponin I, 50 mg/kg every 12 hours beginning 48 hors prior to the implantation.
Corneal angiogenesis was evaluated by slit lamp microscopy. By day 6 angiogenesis in control eyes results in vessels that have extended into the pellet by day 6. At this time there was observed a 50% reduction in blood vessel density and a 30% inhibition in blood vessel length in the treated animals.

Claims

WE CLAIM :
1. A pharmaceutical composition comprising an amount of a peptide that is effective to inhibit angiogenesis, in which the peptide is = a. an inhibitor of bFGF-stimulated bovine endothelial cell proliferation having an IC50 of at least 10 μM; b. greater than 75 amino acids in length,- and c. greater than 80% homologous with a subunit selected from the group consisting of human fast-twitch troponin subunit C (SEQ ID NO=l) , human fast- twitch troponin subunit I (SEQ ID NO=2) , and human fast-twitch troponin subunit T (SEQ ID NO:3) ; and a pharmaceutically acceptable carrier.
2. The composition of claim 1, in which the subunit is human fast-twitch troponin C or human fast-twitch troponin I .
3. The composition of claim 1, in which the subunit is human fast-twitch troponin C.
4. The composition of claim 1, in which the subunit is human fast-twitch troponin I.
5. The composition of claim 1 in which the peptide is greater than 95 % homologous with a human troponin subunit.
6. The composition of claim 5, in which the subunit is human fast-twitch troponin C.
7 The composition of claim 5, in which the subunit is human fast-twitch troponin I
8. The composition of claim 1 m which the peptide is a mammalian troponin subunit.
9. The composition of claim 8 in which the peptide is a mammalian troponin C or troponin I subunit
10. The composition of claim 9 in which the peptide is a troponin subunit selected from the group consisting of bovine, rabbit, mouse and rat troponin subunits
11. The composition of claim 8 in which the peptide is a human troponin C or troponin I subunit.
12. The composition of claim 8 in which the peptide is a troponin subunit selected from the group consisting of bovine, rabbit, mouse and rat troponin subunits.
13. The composition of claim 1 in which the peptide is a fragment of a mammalian troponin subunit
14 The composition of claim 13 n which the peptide is a fragment of a human troponin C or troponin I subunit .
15. The composition of claim 13 in which the peptide is a fragment of a troponin I or troponin C sununit selected from the group consisting of bovme, rabbit, mouse and rat troponin C and I subunits
16. The composition of claim 1 wherein the carrier is acceptable for topical application to the eye
17. The composition of claim 1 wherein the carrier is acceptable for topical application to the skin.
18. The composition of claim 1 wherein the angiogenesis inhibitor is in a biodegradable, biocompatible polymeric delivery device.
19. A method of inhibiting atopic angiogenesis in a subject, having a disease or disorder causing atopic angiogenesis requiring such inhibition, which comprising the step of applying to a site of atopic angiogenesis an amount of a peptide that is effective to inhibit angiogenesis, in which the peptide is: a. an inhibitor of bFGF-stimulated bovme endothelial cell proliferation having an
IC50 of at least 10 μM; b. greater than 75 ammo acids in length; and c. greater than 80% homologous with a subunit selected from the group consisting of human fast-twitch troponin I subunit, human fast-twitch troponin C subunit and human fast-twitch troponin T subunit .
20. The method of claim 19, in which the subunit is human fast-twitch troponin C or human fast-twitch troponin I.
21. The method of claim 20, in which the disease or disorder is a solid tumor.
22. The method of claim 20, in which the tumor is a tumor of the central nervous system.
23. The method according to claim 20, in wnich the disease or disorder is an ophthalmologic disease or disorder.
PCT/US1997/002439 1996-02-16 1997-02-14 Troponin subunits and fragments useful as angiogenesis inhibitors WO1997030085A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
JP52953997A JP3990456B2 (en) 1996-02-16 1997-02-14 Troponin subunits and fragments useful as inhibitors of angiogenesis
EP97905992A EP1007556B1 (en) 1996-02-16 1997-02-14 Troponin subunits and fragments useful as angiogenesis inhibitors
CA2247247A CA2247247C (en) 1996-02-16 1997-02-14 Troponin subunits and fragments useful as angiogenesis inhibitors
DE69729125T DE69729125T2 (en) 1996-02-16 1997-02-14 TROPONIN SUB-UNITS AND FRAGMENTS FOR USE AS INHIBITORS OF ANGIOGENESIS
AT97905992T ATE266675T1 (en) 1996-02-16 1997-02-14 TROPONIN SUBUNITS AND FRAGMENTS FOR USE AS INHIBITORS OF ANGIOGENESIS
AU22753/97A AU707262B2 (en) 1996-02-16 1997-02-14 Troponin subunits and fragments useful as angiogenesis inhibitors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US08/602,941 US5837680A (en) 1996-02-16 1996-02-16 Pharmaceutical compositions comprising troponin subunits, fragments and analogs thereof and methods of their use to inhibit angiogenesis
US08/602,941 1996-02-16

Publications (2)

Publication Number Publication Date
WO1997030085A1 true WO1997030085A1 (en) 1997-08-21
WO1997030085B1 WO1997030085B1 (en) 1997-09-25

Family

ID=24413387

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1997/002439 WO1997030085A1 (en) 1996-02-16 1997-02-14 Troponin subunits and fragments useful as angiogenesis inhibitors

Country Status (8)

Country Link
US (4) US5837680A (en)
EP (1) EP1007556B1 (en)
JP (1) JP3990456B2 (en)
AT (1) ATE266675T1 (en)
AU (1) AU707262B2 (en)
CA (1) CA2247247C (en)
DE (1) DE69729125T2 (en)
WO (1) WO1997030085A1 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999016889A1 (en) * 1997-10-01 1999-04-08 G.D. Searle & Co. Fusion proteins comprising an angiostatin moiety and their use in anti-tumor treatment
WO2000004145A2 (en) * 1998-07-17 2000-01-27 Carpen Olli Myotilin, an actin-organizing protein
EP1165059A1 (en) * 1999-03-15 2002-01-02 Boston Life Sciences, Inc. Pharmaceutical compositions comprising troponin subunits, fragments and homologs thereof and methods of their use to inhibit angiogenesis
US6586401B1 (en) 1996-02-16 2003-07-01 Children's Medical Center Corporation Troponin subunit I fragment and homologs thereof
WO2004091650A1 (en) * 2003-04-15 2004-10-28 Fengming Liu Application of cardiac troponin i in preparing antitumour medicaments
US7078385B2 (en) 1996-02-16 2006-07-18 Boston Life Sciences, Inc. Methods of inhibiting angiogenesis with fragments and homologs of troponin subunit I
WO2007067611A2 (en) * 2005-12-07 2007-06-14 The Brigham And Women's Hospital, Inc. Methods of treating cardiac reperfusion disease
US7393829B2 (en) 1996-02-16 2008-07-01 Children's Medical Center Corporation Pharmaceutical compositions comprising fragments and homologs of troponin subunits
US7479278B2 (en) * 1998-10-21 2009-01-20 Spectral Diagnostics, Inc Troponin I polypeptide fragments and uses thereof

Families Citing this family (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5837680A (en) 1996-02-16 1998-11-17 Children's Medical Center Corporation Pharmaceutical compositions comprising troponin subunits, fragments and analogs thereof and methods of their use to inhibit angiogenesis
US7220557B2 (en) 1997-04-24 2007-05-22 Human Genome Sciences, Inc. METH1 polynucleotides
US20030166065A1 (en) * 1997-04-24 2003-09-04 Human Genome Sciences, Inc. Novel integrin ligand ITGL-TSP
US6248869B1 (en) * 1997-05-29 2001-06-19 Medical Analysis Systems, Inc. Troponin I forms and use of the same
US6468519B1 (en) * 1997-09-10 2002-10-22 Rutgers, The State University Of New Jersey Polyanhydrides with biologically active degradation products
KR100335397B1 (en) * 1998-05-25 2002-09-05 주식회사 하이닉스반도체 Apparatus for driving sequentially sense amplifier
US6378526B1 (en) * 1998-08-03 2002-04-30 Insite Vision, Incorporated Methods of ophthalmic administration
EP1187849A1 (en) * 1999-05-25 2002-03-20 Human Genome Sciences, Inc. Meth1 and meth2 polynucleotides and polypeptides
US20020142982A1 (en) * 1999-09-02 2002-10-03 Timothy Hla Method for regulating angiogenesis
ES2323407T3 (en) 2000-07-12 2009-07-15 Agensys, Inc. NEW USEFUL TUMORAL ANTIGEN IN THE DIAGNOSIS AND THERAPY OF CANCER DE VEJIGA, OVARIO, PULMON AND RIÑON.
US20030236188A1 (en) * 2001-05-03 2003-12-25 Spytek Kimberly A. Novel human proteins, polynucleotides encoding them and methods of using the same
AU2003208097A1 (en) * 2002-02-04 2003-09-02 Hideo Yoshida Anticancer agents using vero toxin variants
US7261876B2 (en) 2002-03-01 2007-08-28 Bracco International Bv Multivalent constructs for therapeutic and diagnostic applications
US7794693B2 (en) * 2002-03-01 2010-09-14 Bracco International B.V. Targeting vector-phospholipid conjugates
US8623822B2 (en) * 2002-03-01 2014-01-07 Bracco Suisse Sa KDR and VEGF/KDR binding peptides and their use in diagnosis and therapy
US20050100963A1 (en) * 2002-03-01 2005-05-12 Dyax Corporation KDR and VEGF/KDR binding peptides and their use in diagnosis and therapy
US7211240B2 (en) * 2002-03-01 2007-05-01 Bracco International B.V. Multivalent constructs for therapeutic and diagnostic applications
CA2666005C (en) 2002-03-01 2016-01-19 Dyax Corp. Kdr and vegf/kdr binding peptides and their use in diagnosis and therapy
JP2005528362A (en) 2002-03-21 2005-09-22 ユニヴァーシティ オヴ フロリダ Regulation of angiogenesis
WO2004028559A1 (en) * 2002-09-26 2004-04-08 The Board Of Trustees Of The University Of Illinois Anti-angiogenic fragments of pigment epithelium-derived factor (pedf)
PL1944312T3 (en) 2003-03-03 2012-12-31 Dyax Corp Peptides that specifically bind HGF receptor (CMET) and uses thereof
ES2737835T3 (en) * 2003-04-23 2020-01-16 Valeritas Inc Hydraulically driven pump for long-term medication administration
US9089636B2 (en) 2004-07-02 2015-07-28 Valeritas, Inc. Methods and devices for delivering GLP-1 and uses thereof
US8697139B2 (en) 2004-09-21 2014-04-15 Frank M. Phillips Method of intervertebral disc treatment using articular chondrocyte cells
US7265248B1 (en) 2005-04-29 2007-09-04 Technology Innovations, Llc Compositions and methods for the treatment of malaria
US20070130009A1 (en) * 2005-06-01 2007-06-07 Chad Steelberg System and method for media play pricing
ES2566058T3 (en) 2006-03-30 2016-04-08 Valeritas, Inc. Multi-cartridge fluid supply device
US8468561B2 (en) 2006-08-09 2013-06-18 Google Inc. Preemptible station inventory

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5583200A (en) * 1993-02-23 1996-12-10 Pasteur Sanofi Diagnostics Stabilized composition of troponin for immunoassays and process for stabilizing troponin for immunoassays

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE69122956T2 (en) * 1990-01-25 1997-05-22 Childrens Hosp Medical Center METHOD AND COMPOSITIONS FOR INHIBITING ANGIOGENESIS
US5837680A (en) 1996-02-16 1998-11-17 Children's Medical Center Corporation Pharmaceutical compositions comprising troponin subunits, fragments and analogs thereof and methods of their use to inhibit angiogenesis

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5583200A (en) * 1993-02-23 1996-12-10 Pasteur Sanofi Diagnostics Stabilized composition of troponin for immunoassays and process for stabilizing troponin for immunoassays

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
GRABAREK Z, ET AL.: "PROTEOLYTIC FRAGMENTS OF TROPONIN C. INTERACTIONS WITH THE OTHER TROPONIN SUBUNITS AND BIOLOGICAL ACTIVITY", JOURNAL OF BIOLOGICAL CHEMISTRY.(MICROFILMS), AMERICAN SOCIETY OF BIOLOGICAL CHEMISTS, BALTIMORE, MD., US, vol. 256, no. 04, 25 December 1981 (1981-12-25), US, pages 13121 - 13127, XP008037857 *
TANOKURA M, OHTSUKI I: "INTERACTIONS AMONG CHYMOTRYPTIC TROPONIN T SUBFRAGMENTS, TROPOMYOSIN, TROPONIN I AND TROPONIN C", JOURNAL OF BIOCHEMISTRY, OXFORD UNIVERSITY PRESS, GB, vol. 95, no. 05, 1 May 1984 (1984-05-01), GB, pages 1417 - 1421, XP001108929, ISSN: 0021-924X *

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7393829B2 (en) 1996-02-16 2008-07-01 Children's Medical Center Corporation Pharmaceutical compositions comprising fragments and homologs of troponin subunits
US7452866B2 (en) 1996-02-16 2008-11-18 Children's Medical Center Corporation Methods of inhibiting angiogenesis with fragments and homologs of troponin subunit 1
US6586401B1 (en) 1996-02-16 2003-07-01 Children's Medical Center Corporation Troponin subunit I fragment and homologs thereof
US7078385B2 (en) 1996-02-16 2006-07-18 Boston Life Sciences, Inc. Methods of inhibiting angiogenesis with fragments and homologs of troponin subunit I
EP1705248A1 (en) * 1997-10-01 2006-09-27 G.D. Searle LLC. Fusion proteins comprising an angiostatin moiety and their use in anti-tumour treatment
WO1999016889A1 (en) * 1997-10-01 1999-04-08 G.D. Searle & Co. Fusion proteins comprising an angiostatin moiety and their use in anti-tumor treatment
WO2000004145A2 (en) * 1998-07-17 2000-01-27 Carpen Olli Myotilin, an actin-organizing protein
WO2000004145A3 (en) * 1998-07-17 2000-04-20 Olli Carpen Myotilin, an actin-organizing protein
US6458929B1 (en) 1998-07-17 2002-10-01 Licentia Ltd. Myotilin, a novel actin-organizing protein
US7479278B2 (en) * 1998-10-21 2009-01-20 Spectral Diagnostics, Inc Troponin I polypeptide fragments and uses thereof
EP1165059A1 (en) * 1999-03-15 2002-01-02 Boston Life Sciences, Inc. Pharmaceutical compositions comprising troponin subunits, fragments and homologs thereof and methods of their use to inhibit angiogenesis
EP1165059A4 (en) * 1999-03-15 2003-03-05 Boston Life Sciences Inc Pharmaceutical compositions comprising troponin subunits, fragments and homologs thereof and methods of their use to inhibit angiogenesis
JP2002539161A (en) * 1999-03-15 2002-11-19 ボストン ライフ サイエンシズ,インコーポレーテッド. Pharmaceutical compositions comprising troponin subunits, fragments and homologs thereof and methods of using them for inhibiting angiogenesis
WO2004091650A1 (en) * 2003-04-15 2004-10-28 Fengming Liu Application of cardiac troponin i in preparing antitumour medicaments
WO2007067611A3 (en) * 2005-12-07 2008-03-06 Brigham & Womens Hospital Methods of treating cardiac reperfusion disease
WO2007067611A2 (en) * 2005-12-07 2007-06-14 The Brigham And Women's Hospital, Inc. Methods of treating cardiac reperfusion disease

Also Published As

Publication number Publication date
EP1007556A4 (en) 2000-06-14
US6653283B1 (en) 2003-11-25
CA2247247C (en) 2010-08-03
DE69729125T2 (en) 2005-05-12
JP3990456B2 (en) 2007-10-10
US5837680A (en) 1998-11-17
EP1007556A2 (en) 2000-06-14
DE69729125D1 (en) 2004-06-17
EP1007556B1 (en) 2004-05-12
AU707262B2 (en) 1999-07-08
ATE266675T1 (en) 2004-05-15
US6025331A (en) 2000-02-15
AU2275397A (en) 1997-09-02
CA2247247A1 (en) 1997-08-21
JP2000506523A (en) 2000-05-30
US6403558B1 (en) 2002-06-11

Similar Documents

Publication Publication Date Title
US6403558B1 (en) Pharmaceutical compositions comprising troponin subunits, fragments and analogs thereof and methods of their use to inhibit angiogenesis
US7078385B2 (en) Methods of inhibiting angiogenesis with fragments and homologs of troponin subunit I
AU2006313318B2 (en) Improved variants of pigment epithelium derived factor and uses thereof
AU652744B2 (en) Method and compositions for inhibiting angiogenesis
US7452866B2 (en) Methods of inhibiting angiogenesis with fragments and homologs of troponin subunit 1
JP4560616B2 (en) Compositions and methods for modulating cellular NF-κB activation
US8530416B2 (en) Variants of pigment epithelium derived factor and uses thereof
EP0220241A1 (en) Purified protein having angiogenic activity and methods of preparation.
US20080051345A1 (en) Pharmaceutical compositions comprising fragments and homologs of troponin subunits
AU774596B2 (en) Pharmaceutical compositions comprising troponin subunits, fragments and homologs thereof and methods of their use to inhibit angiogenesis
US20020045564A1 (en) Methods of modulating muscle contraction
ZA200600914B (en) Methods and compositions for treating disorders of the extracellular matrix
DE29724845U1 (en) Use of troponin subunits as angiogenesis inhibitors - used for treating e.g. tumours, ocular neovascularisation, arthritis, psoriasis, atherosclerotic plaques or nonunion fractures

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AL AM AU AZ BA BB BG BR BY CA CN CU CZ EE GE HU IL IS JP KG KP KR KZ LC LK LR LT LV MD MG MK MN MX NO NZ PL RO RU SG SI SK TJ TM TR TT UA UZ VN YU AM AZ BY KG KZ MD RU TJ TM

AL Designated countries for regional patents

Kind code of ref document: A1

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 GN ML MR NE SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
ENP Entry into the national phase

Ref document number: 2247247

Country of ref document: CA

Ref country code: CA

Ref document number: 2247247

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 1997905992

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1997905992

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 1997905992

Country of ref document: EP