WO2017141246A1 - Drug eluting stent - Google Patents

Drug eluting stent Download PDF

Info

Publication number
WO2017141246A1
WO2017141246A1 PCT/IL2017/050201 IL2017050201W WO2017141246A1 WO 2017141246 A1 WO2017141246 A1 WO 2017141246A1 IL 2017050201 W IL2017050201 W IL 2017050201W WO 2017141246 A1 WO2017141246 A1 WO 2017141246A1
Authority
WO
WIPO (PCT)
Prior art keywords
leptin
stent
antagonist
leptin antagonist
drug
Prior art date
Application number
PCT/IL2017/050201
Other languages
French (fr)
Inventor
Jacob Schneiderman
Original Assignee
Jacob Schneiderman
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 Jacob Schneiderman filed Critical Jacob Schneiderman
Priority to US16/078,013 priority Critical patent/US10926006B2/en
Publication of WO2017141246A1 publication Critical patent/WO2017141246A1/en
Priority to IL261182A priority patent/IL261182B/en
Priority to US16/153,890 priority patent/US11123461B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F2/00Filters implantable into blood vessels; Prostheses, i.e. artificial substitutes or replacements for parts of the body; Appliances for connecting them with the body; Devices providing patency to, or preventing collapsing of, tubular structures of the body, e.g. stents
    • A61F2/82Devices providing patency to, or preventing collapsing of, tubular structures of the body, e.g. stents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F2/00Filters implantable into blood vessels; Prostheses, i.e. artificial substitutes or replacements for parts of the body; Appliances for connecting them with the body; Devices providing patency to, or preventing collapsing of, tubular structures of the body, e.g. stents
    • A61F2/02Prostheses implantable into the body
    • A61F2/04Hollow or tubular parts of organs, e.g. bladders, tracheae, bronchi or bile ducts
    • A61F2/06Blood vessels
    • A61F2/07Stent-grafts
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L31/00Materials for other surgical articles, e.g. stents, stent-grafts, shunts, surgical drapes, guide wires, materials for adhesion prevention, occluding devices, surgical gloves, tissue fixation devices
    • A61L31/08Materials for coatings
    • A61L31/10Macromolecular materials
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L31/00Materials for other surgical articles, e.g. stents, stent-grafts, shunts, surgical drapes, guide wires, materials for adhesion prevention, occluding devices, surgical gloves, tissue fixation devices
    • A61L31/14Materials characterised by their function or physical properties, e.g. injectable or lubricating compositions, shape-memory materials, surface modified materials
    • A61L31/148Materials at least partially resorbable by the body
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L31/00Materials for other surgical articles, e.g. stents, stent-grafts, shunts, surgical drapes, guide wires, materials for adhesion prevention, occluding devices, surgical gloves, tissue fixation devices
    • A61L31/14Materials characterised by their function or physical properties, e.g. injectable or lubricating compositions, shape-memory materials, surface modified materials
    • A61L31/16Biologically active materials, e.g. therapeutic substances
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F2230/00Geometry of prostheses classified in groups A61F2/00 - A61F2/26 or A61F2/82 or A61F9/00 or A61F11/00 or subgroups thereof
    • A61F2230/0002Two-dimensional shapes, e.g. cross-sections
    • A61F2230/0017Angular shapes
    • A61F2230/0021Angular shapes square
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F2250/00Special features of prostheses classified in groups A61F2/00 - A61F2/26 or A61F2/82 or A61F9/00 or A61F11/00 or subgroups thereof
    • A61F2250/0058Additional features; Implant or prostheses properties not otherwise provided for
    • A61F2250/0067Means for introducing or releasing pharmaceutical products into the body
    • A61F2250/0068Means for introducing or releasing pharmaceutical products into the body the pharmaceutical product being in a reservoir
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2300/00Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices
    • A61L2300/40Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices characterised by a specific therapeutic activity or mode of action
    • A61L2300/416Anti-neoplastic or anti-proliferative or anti-restenosis or anti-angiogenic agents, e.g. paclitaxel, sirolimus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2300/00Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices
    • A61L2300/40Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices characterised by a specific therapeutic activity or mode of action
    • A61L2300/432Inhibitors, antagonists
    • A61L2300/436Inhibitors, antagonists of receptors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2300/00Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices
    • A61L2300/60Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices characterised by a special physical form
    • A61L2300/602Type of release, e.g. controlled, sustained, slow
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2300/00Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices
    • A61L2300/60Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices characterised by a special physical form
    • A61L2300/602Type of release, e.g. controlled, sustained, slow
    • A61L2300/604Biodegradation

Definitions

  • the present invention relates to the field of medicine, and in some embodiments to devices and compositions comprising a leptin antagonist formulated for localized release of a leptin antagonist at the site of treatment, specifically the treatment of ischemia and reperfusion injury.
  • Cardiovascular disease is a class of diseases that involve the heart and/or the blood vessels.
  • CVD cardiovascular disease
  • CRP C- reactive protein
  • osteoprotegerin which is involved with regulation of NF- KB, has been found to be a risk factor for cardiovascular disease and mortality [Venuraju et al., 2010 J. Am. Coll. Cardiol. 55 (19): 2049-61]
  • cardiovascular disorders can benefit from suppression of inflammation-related processes and cellular proliferation as part of a remodeling response (e.g. use of locally released cytotoxic drugs such as paclitaxel or sirolimus in preventing restenosis or use of doxycyciine in treatment of abdominal aortic aneurysm (AAA)), to date, there is no evidence to suggest that cardiovascular disease can benefit from anti-inflammatory treatment.
  • cytotoxic drugs such as paclitaxel or sirolimus in preventing restenosis or use of doxycyciine in treatment of abdominal aortic aneurysm (AAA)
  • IRI ischemia/reperfusion injury
  • IRI is caused by critical reduction of blood supply to an organ followed by renewal of blood flow and re-ox ygenati on. Tissue damage is driven by activation of inflammatory processes, including synthesis of cytokines, chemokines and reactive oxygen species (ROS).
  • ROS reactive oxygen species
  • Chemokines mediate inflammation and regulate pro- inflammatory cytokine, adhesion molecule expression, leukocyte infiltration and activation.
  • ROS reactive oxygen species
  • Reperfusion and oxygenation produce oxygen free radicals, which promote lipid peroxidation and tissue damage by free radicals.
  • IRI tissue damage As mentioned above apply to all tissues.
  • cellular damage correlates with the magnitude of ischemia, and cell type. For instance, kidney and brain cells are extremely sensitive to ischemic damage, which is further exacerbated by reperfusion injury.
  • Leptin is a naturally occurring pleiotropic molecule that regulates food intake as well as metabolic and endocrine functions. Leptin also plays a regulatory role in immunity, inflammation, and hematopoiesis.
  • the human leptin precursor is a linear polypeptide 167 amino acid residues long represented by NCBI Reference Sequence NP 000221.1 (SEQ ID NO 1) encoded by the mRNA having the nucleotide sequence NCBI Reference Sequence NM_000230. Residues 1-
  • Leptin antagonists are also known, see for example, US 7,307,142 and US 8,969,292.
  • the invention in some embodiments, relates to the field of medicine, and more particularly to methods and devices that use leptin antagonists.
  • the invention relates to compositions comprising a leptin antagonist formulated for localized release of a leptin antagonist to inhibit activity of leptin at the site of treatment as well as methods of using such compositions for treating disorders, including cardiovascular disorders.
  • a method of treatment comprising: exposing in vivo tissue of a subject in need thereof to local administration of a pharmaceutically-effective amount of leptin antagonist, thereby providing a therapeutic effect to the tissue.
  • the tissue is substantially continuously exposed to a pharmaceutically-effective amount of leptin antagonist for a period of not less than three days, not less than 5 days, not less than 8 days and even not less than 14 days.
  • a method of treatment comprising implanting in contact with tissue in the body of a subject in need thereof a composition configured for in vivo local administration of leptin antagonist, thereby providing a therapeutic effect to the tissue.
  • the configuration for the in vivo release is such that when the composition is implanted in vivo, leptin antagonist is released from the composition in a pharmaceutically-effective amount for a period of not less than three days, not less than 5 days, not less than 8 days and even not less than 14 days.
  • the need is that the subject suffers from at least one pathology selected from the group consisting of: cardiovascular disease; remodeling of stable athersclerotic plaque into an unstable lesion; ascending aortic aneurysm-associated hypertension, hypercholesterolemia or diabetes mellitus; bicuspid aortic valve; Takayasu disease; rheumatoid arteritis; Marian's syndrome; ankylosing spondylitis; giant cell arteritis; inflammatory aortic aneurysm; pulmonary artery aneurysm in Marian's syndrome; aortic dissection in an aortic or peripheral large artery; angiogenesis; cancer; local discrete lesion therapy; and arteriovenous malformation.
  • pathology selected from the group consisting of: cardiovascular disease; remodeling of stable athersclerotic plaque into an unstable lesion; ascending aortic aneurysm-associated hypertension, hypercholesterolemia or diabetes mellitus; bicuspid aortic
  • the need is that the subject suffers from a cardiovascular disorder, wherein the therapeutic effect is down-regulation of an expression or activity of leptin in a cardiovascular tissue.
  • the cardiovascular tissue is aortic and/or mitral heart valve leaflet tissue.
  • the local administration is effected by positioning a carrier capable of releasing the leptin antagonist on an outer wall (e.g., tunica externa) or the inner wall (e.g., tunica intima) of an aorta.
  • the cardiovascular tissue is arterial or venous wall tissue.
  • the local administration is effected by positioning a carrier capable of releasing the leptin antagonist on an outer wall (e.g., tunica externa) or the inner wall (e.g., tunica intima) of the arterial or venous wall tissue.
  • the cardiovascular disorder is a vascular aneurysm. In some embodiments, the cardiovascular disorder is an aortic vascular disorder. In some embodiments, the cardiovascular disorder is left ventricular remodeling.
  • the local administration is effected via an intravascular catheter. In some embodiments, the local administration is effected via direct injection.
  • a method for treatment of athersclerotic plaque comprising: administering a pharmaceutical ly-effective amount of a leptin antagonist to atherscl erotic plaque accumulated in the inner walls of an artery, thereby at least one of: (a) reducing the rate and (b) reducing the incidence, of conversion of a stable athersclerotic plaque to an unstable lesion.
  • composition comprising a leptin antagonist and a carrier, for use in treating a disorder selected from the group consisting of: cardiovascular disease; remodeling of stable athersclerotic plaque into an unstable lesion; ascending aortic aneurysm-associated hypertension, hypercholesterolemia or diabetes mellitus; bicuspid aortic valve; Takayasu disease; rheumatoid arteritis; Marfan's syndrome; ankylosing spondylitis; giant cell arteritis; inflammatory aortic aneurysm; pulmonary artery aneurysm in Marfan's syndrome; aortic dissection in an aortic or peripheral large artery; site of arterial anastomosis, angiogenesis; cancer; local neoplastic discrete lesion therapy; and arteriovenous malformation, wherein the carrier is configured for localized administration of the leptin antagonist.
  • the carrier is a biodegradable support.
  • the biodegradable support is composed of a polymer selected from the group consisting of a hydrogel, poly gly colic acid (PGA), poly lactic co-glycolic acid (PLGA), polylactide (PL A), and poly (L-lactide) (PLLA), and combinations thereof.
  • the carrier is a hydrogel. In some embodiments, the carrier is configured as a film. In some embodiments, the carrier is a device selected from the group consisting of a mesh, a balloon and a vascular graft. In some embodiments, the carrier is a depot-forming injectable composition.
  • the disorder is a cardiovascular disorder, wherein the leptin antagonist effects down-regulation of an expression or activity of leptin in a cardiovascular tissue.
  • the cardiovascular disorder is a vascular disorder.
  • the vascular disorder is an aortic vascular disorder.
  • the cardiovascular disorder is left ventricular remodeling.
  • the cardiovascular tissue is aortic and/or mitral heart valve leaflet tissue. In some such embodiments, the cardiovascular tissue is arterial or venous wall tissue.
  • the local administration is effected by positioning a carrier capable of releasing the leptin antagonist on a location selected from the group consisting of: an outer wall of an aorta, an outer wall of an artery, an outer wall of an vein, a luminal surface of an aorta, a luminal surface of an artery and a luminal surface of an vein.
  • the local administration is effected by positioning a carrier capable of releasing the leptin antagonist on an outer wall (e.g., tunica externa) or the inner wall (e.g., tunica intima) of the arterial or venous wall tissue.
  • the localized administration is to be effected via an intravascular catheter. In some embodiments, the localized administration is to be effected via direct injection.
  • a method of treating a condition in a subject in need thereof comprising administering intracavitarily to inner walls of a fluid-filled bodily cavity of the subject a composition comprising a leptin antagonist.
  • composition comprising: a leptin antagonist for use in treating a condition, wherein the composition is configured for intracavitary administration to inner walls of a fluid-filled bodily cavity of a subject.
  • an intracavitarily-implantable medical device comprising: at least one solid functional device part configured for deploying the device in a fluid-filled bodily cavity of a subject; and functionally associated with at least one the device component, a leptin antagonist.
  • a surgical connecting device comprising: a solid device body made of a material; and functionally associated with the device body, a pharmaceutically-effective amount of leptin antagonist.
  • the device body is in the form selected from the group consisting of surgical suture thread and a surgical staple.
  • a method for treating ischemia and reperfusion injury (IRI) by administrating leptin antagonist upon reperfusion and maintaining sustained administration of the leptin antagonist into the vessel undergoing the reperfusion procedure, for delivering the leptin antagonist by the blood flow into the damaged cells of the tissue injured by the IRI via bolus intra-arterial injection, or sustained release from a drug eluting stent.
  • IRI ischemia and reperfusion injury
  • a double function drug eluting stent configured to enable sustained release of anti- proliferative drug into the wall of a blood vessel and to enable sustained release of leptin antagonist (LA) into the lumen, so that the leptin antagonist is carried by the blood stream to be uptaken by tissue cells that sustained ischemia and reperfusion injury.
  • any suitable leptin antagonist may be used for implementing the teachings herein.
  • Various types and specific suitable leptin antagonists are listed in the description herein.
  • the leptin antagonist is capable of binding a leptin receptor.
  • the leptin antagonist is incapable of dimerization.
  • the leptin antagonist comprises a polypeptide portion.
  • the leptin antagonist is selected from the group consisting of a polypeptide, a salt, and/or an ester thereof.
  • the leptin antagonist is a modified leptin polypeptide.
  • FIG. la-c illustrate a gel ( Figure la), film ( Figure lb) and mesh ( Figure l c) for local release of a leptin antagonist.
  • FIG. 2 illustrates a balloon catheter configured for local release of a leptin antagonist(drug release indicated by arrows).
  • FIG. 3 illustrates a slow release leptin eluted from a scaffold.
  • FIG, 4 illustrates the location of leptin film application on the anterior outer surface of the ascending aorta.
  • Human arch angiogram depicts mouse anatomy.
  • FIG. 5 illustrates a time course analysis of serum leptin level in ApoE "7" mice that underwent peri-aortic application of leptin film (20 ⁇ g).
  • FIG. 6 illustrates increased ascending aortic diameter at the location of leptin film application versus controls.
  • FIG. 7 illustrates elastica staining and aSMA IHC analysis of ascending aortic cross sections of mice locally treated with leptin versus controls.
  • FIG. 8 illustrates change in left ventricle (LV) wall thickness in leptin-treated (filled columns) versus control (open columns) mice.
  • FIG. 9 illustrates LV diameter in systole and diastole in leptin-treated (filled columns) and control (open columns) mice.
  • FIG. 10 illustrates LV fractional area change in leptin-treated (filled columns) versus control (open columns) mice.
  • FIG. 1 1 illustrates aortic and mitral valve leaflet thickness in leptin-treated and control mice.
  • FIG. 12 illustrates mean systolic blood pressure in angiotensin II treated mice.
  • FIG. 13 illustrates a time course analysis presenting weight of angiotensin II treated mice (open triangles), and mice receiving both, angiotensin II and leptin antagonist (LA).
  • FIG. 14 illustrates number of mice that succumbed due to ruptured abdominal and thoracic aneurysms in angiotensin II treated mice versus mice receiving angiotensin II and leptin antagonist (LA).
  • FIG. 15 illustrates ascending aortic dilatation in angiotensin II treated mice versus mice receiving angiotensin II and leptin antagonist (LA).
  • FIG. 16 illustrates elastic lamella fragmentation and aSMA depletion in angiotensin II treated mice versus mice receiving angiotensin II and leptin antagonist (LA).
  • FIG. 17 illustrates leptin expression in medial SMCs (arrows) and macrophages of atherosclerotic lesions (filled arrowheads) in angiotensin II treated, angiotensin II and leptin antagonist (LA) treated, and control mice.
  • FIG. 18 illustrates LV hypertrophy in angiotensin II treated mice versus mice receiving angiotensin II and leptin antagonist (LA).
  • FIG. 19 illustrates changes in LV diameter in angiotensin II treated mice versus mice receiving angiotensin II and leptin antagonist (LA).
  • FIG. 20 illustrates LV fractional area change in angiotensin II treated mice (open column) versus mice receiving angiotensin II and leptin antagonist (LA) (filled column).
  • FIG. 21 illustrates peak systolic velocity at the aortic valve in angiotensin II treated mice versus mice receiving angiotensin II and leptin antagonist (LA).
  • FIG. 22 illustrates aortic and mitral valve thickness (graph on left), and staining of valve leaflets with H&E (panels F-I).
  • aSMA and TGFP panels J-M', staining for aortic valves
  • FIG. 23 illustrates expression of leptin (D), and leptin receptor (E) in normal human aortic valve leaflet tissue.
  • FIG. 24 illustrates leptin and leptin receptor antigen prevalent in severe aortic valve stenosis, evident in SMC-iike cells, and infiltrating macrophages.
  • FIG. 25 illustrates leptin and leptin receptor mRNA !evels in leaflets of stenosed aortic valve versus normal aortic valve controls, and fat tissue (as positive control);
  • FIG. 26 illustrates proliferation of valve interstitial cells (VICs) in response to leptin stimulation; and
  • FIG. 27 schematically depicts embodiments of surgical connecting devices according to the teachings herein;
  • FIG. 28 schematically depicts embodiments of the teachings herein suitable for intracavitary administration of leptin antagonist
  • FIG. 29 schematically depicts further embodiments of the teachings herein suitable for intracavitary administration of leptin antagonist
  • FIG. 30 schematically depicts an embodiment of a double-function drug eluting stent in accordance with the teachings herein;
  • FIG. 31a is partial cross sectional perspective view of a prior art stent
  • FIGS. 31b and 31c are partial cross sectional perspective views of further embodiments of a double-function drug eluting stent in accordance with the teachings herein;
  • FIGS. 32a and 32b illustrate a time course of leptin mRNA expression and leptin receptor mRNA expression, respectively, in the heart of mice that underwent myocardial IRI and systemic administration of leptin antagonist to stimulate endogenous leptin synthesis, or saline, upon reperfusion;
  • FIG. 33 illustrates percent of ejection fraction (EF%) of mice at 1 and 30 days after undergoing IRI and systemic administration of leptin antagonist or saline upon reperfusion;
  • FIGS. 34 (a-c) illustrate coronal sections of a mouse brain that underwent IRI (a-c);
  • FIGS. 35 (a-c) illustrate coronal sections of a mouse brain that underwent IRI plus selective administration of leptin antagonist upon reperfusion
  • FIGS. 36 (a-c) illustrate coronal sections of a normal mouse brain.
  • the invention in some embodiments, relates to the field of medicine, and more particularly to methods and devices that use leptin antagonists.
  • the invention relates to compositions comprising a leptin antagonist formulated for localized release of a leptin antagonist at the site of treatment as well as methods of using such compositions for treating disorders, including cardiovascular disorders.
  • the compositions comprising a leptin antagonist can be used for localized suppression of leptin-related conditions, including tissue remodeling processes.
  • leptin might play a role in vascular inflammation, oxidative stress, and vascular smooth muscle hypertrophy that may contribute to coronary heart disease among other pathologies, to date no one has conclusively shown that localized down-regulation of leptin activity can be used to treat cardiovascular disorders characterized by remodeling of cardiovascular tissue such as cardiac, arterial or valve tissue.
  • the present inventor set out to elucidate the role of leptin in disorders, such as cardiovascular disorders by employing a leptin antagonist in a localized manner.
  • Experiments conducted by the present inventor demonstrate that localized release of leptin in cardiovascular tissue can lead to cardiovascular tissue remodeling while localized down-regulation of leptin activity can lead to suppression and even reversal of cardiovascular tissue (arterial wall tissue, heart muscle tissue and valve leaflet tissue) remodeling induced by angiotensin II.
  • a locally administered leptin antagonist can be used to treat cardiovascular disorders characterized by tissue remodeling.
  • the present inventor has discovered an unexpected pharmaceutical efficacy of locally administered leptin antagonists, especially leptin antagonists administered by sustained release.
  • the present inventor has found that in vivo implantation of a composition configured for sustained-release of leptin antagonist can have a desirable pharmaceutical effect on tissue in proximity of the implanted composition with limited or no substantial side- effects, for example, no discernible hormonal or immunological effects.
  • the present inventor has also found that such in vivo administration (e.g., by implantation) inside a fluid-filled cavity of the body (for example of the cardiovascular system such as blood vessels or cardiac chambers) can have a desirable pharmaceutical effect on tissue in proximity of the implanted composition with limited or no substantial side- effects: leptin antagonist from the composition has not been found to be washed away by the fluid and, instead, has been found to interact with the tissue providing a desirable pharmaceutical effect.
  • leptin antagonist released from the composition passes into and through the cavity walls (e.g., tunica intima) in pharmaceutically-effective amounts.
  • cavity walls e.g., tunica intima
  • the success of some embodiments of the teachings herein is at least partially attributable to the serendipitous increased permeability of cardiovascular intima during inflammation. It may be that the intima of healthy cardiovascular intima is relatively impermeable to leptin antagonist released from the composition, so that there is little or no passage of leptin antagonist into and through the endothelium and underlying tissue, thereby avoiding substantial negative side-effects. In contrast, it seems that the permeability of the cardiovascular intima during inflammation increases sufficiently to allow passage of a therapeutically-effective amount of leptin antagonist released from the composition into the tissue. It is currently believed that this effect is self-regulating.
  • a higher degree of inflammation leads to a higher degree of intima permeability allowing passage of more leptin antagonist leading to a relatively high dose of leptin antagonist in the more pathological tissue.
  • inflammation decreases (inter alia, due to the pharmaceutical effect of the administered leptin antagonist)
  • intima permeability decreases thereby decreasing the dose of leptin antagonist actually in the tissue that is still sufficient to exercise a desired pharmaceutical effect but with a reduced incidence of substantial negative side effects.
  • the present invention includes local administration of leptin antagonist to treat and attenuate expansion of ascending aortic aneurysm, and corresponding cardiac sequelae (driven by the aorto-ventricular coupling), including left ventricular hypertrophy, as well as hyperplasia of left heart valve leaflets.
  • the present invention includes treatment of peripheral vascular disorders such as the progression of arterial or venous aneurysms while minimizing systemic exposure to the administered leptin antagonist.
  • a composition comprising a leptin antagonist and a carrier configured for localized administration, for treating disorders such as cardiovascular disorders.
  • cardiovascular disorders refer to disorders of the cardiovascular system, i.e. the heart and central, cranial and peripheral vasculature. Examples of such disorders include, but are not limited to valve stenosis, aneurysms, vessel response to vascular injury, cardiomyopathy and the like.
  • the carrier can be a solid, gel or liquid carrier
  • the leptin antagonist can be any agent capable of down-regulating leptin activity in the target tissue.
  • a leptin antagonist include agents capable of binding and/or degrading leptin or leptin receptors as well as agents capable of down-regulating leptin expression (at the DNA or RNA levels, i.e., agents capable of blocking transcription or translation).
  • the leptin antagonist known as SMLA Shpilman et al., J Biol Chem. 20 ⁇ ; 286:4429-4442
  • leptin receptor-leptin antagonist binds to the leptin receptors in the treated tissue/organ, leaving no leptin receptors available to complex with the endogenous leptin.
  • the complex leptin receptor-leptin antagonist is inactive and can't exert any damage to the tissue, locally.
  • Specific preferred leptin antagonists are listed hereinbelow.
  • a leptin antagonist that is an agent capable of down-regulating leptin is an antibody or antibody fragment capable of specifically binding leptin or a leptin receptor.
  • the antibody specifically binds at least one epitope of leptin, e.g., an epitope defined amino acids 26-59 of mammalian leptin (e.g. rat leptin - SEQ ID NO 48).
  • epitope of leptin e.g., an epitope defined amino acids 26-59 of mammalian leptin (e.g. rat leptin - S
  • antibody refers to a substantially intact antibody molecule.
  • antibody fragment refers to a functional fragment of an antibody that is capable of binding to an antigen.
  • Suitable antibody fragments for practicing the present invention include, inter alia, a complementarity-determining region (CDR) of an immunoglobulin light chain (referred to herein as “light chain”), a CDR of an immunoglobulin heavy chain (referred to herein as “heavy chain”), a variable region of a light chain, a variable region of a heavy chain, a light chain, a heavy chain, an Fd fragment, and antibody fragments comprising essentially whole variable regions of both light and heavy chains such as an Fv, a single-chain Fv, an Fab, an Fab', and an F(ab')2.
  • Fv defined as a genetically engineered fragment consisting of the variable region of the light chain and the variable region of the heavy chain expressed as two chains;
  • scFv single-chain Fv
  • Fab a fragment of an antibody molecule containing a monovalent antigen- binding portion of an antibody molecule, obtained by treating whole antibody with the enzyme papain to yield the intact light chain and the Fd fragment of the heavy chain, which consists of the variable and CHI domains thereof;
  • Fab' a fragment of an antibody molecule containing a monovalent antigen- binding portion of an antibody molecule, obtained by treating whole antibody with the enzyme pepsin, followed by reduction (two Fab' fragments are obtained per antibody molecule);
  • F(ab')2 a fragment of an antibody molecule containing a monovalent antigen- binding portion of an antibody molecule, obtained by treating whole antibody with the enzyme pepsin (i.e., a dimer of Fab' fragments held together by two disulfide bonds).
  • Antibodies may be generated via any one of several known methods, which may employ induction of in vivo production of antibody molecules, screening of immunoglobulin libraries (Orlandi, R. et al. (1989). Cloning immunoglobulin variable domains for expression by the polymerase chain reaction. Proc Natl Acad Sci USA 86, 3833-3837; and Winter, G. and Milstein, C. (1991). Man-made antibodies. Nature 349, 293-299), or generation of monoclonal antibody molecules by continuous cell lines in culture.
  • haptens can be coupled to antigenically neutral carriers such as keyhole limpet hemocyanin (KLH) or serum albumin (e.g., bovine serum albumin (BSA)) carriers (see, for example, US. Pat. Nos. 5,189,178 and 5,239,078).
  • KLH keyhole limpet hemocyanin
  • BSA bovine serum albumin
  • Coupling a hapten to a carrier can be effected using methods well known in the art. For example, direct coupling to amino groups can be effected and optionally followed by reduction of the imino linkage formed.
  • the carrier can be coupled using condensing agents such as dicyclohexyl carbodiimide or other carbodiimide dehydrating agents.
  • Condensing agents such as dicyclohexyl carbodiimide or other carbodiimide dehydrating agents.
  • Linker compounds can also be used to effect the coupling; both homobifunctional and heterobifunctional linkers are available from Pierce Chemical Company, Rockford, Illinois, USA.
  • the resulting immunogenic complex can then be injected into suitable mammalian subjects such as mice, rabbits, and others. Suitable protocols involve repeated injection of the immunogen in the presence of adjuvants according to a schedule designed to boost production of antibodies in the serum.
  • the titers of the immune serum can readily be measured using immunoassay procedures which are well known in the art.
  • the antisera obtained can be used directly or monoclonal antibodies may be obtained, as described hereinabove.
  • Antibody fragments may be obtained using methods well known in the art. (See, for example, Harlow, E. and Lane, D. (1988). Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, New York.)
  • antibody fragments according to the present invention can be prepared by proteolytic hydrolysis of the antibody or by expression in E. coli or mammalian cells (e.g., Chinese hamster ovary (CHO) cell culture or other protein expression systems) of DNA encoding the fragment.
  • E. coli or mammalian cells e.g., Chinese hamster ovary (CHO) cell culture or other protein expression systems
  • antibody fragments can be obtained by pepsin or papain digestion of whole antibodies by conventional methods.
  • an (Fab' )2 antibody fragments can be produced by enzymatic cleavage of antibodies with pepsin to provide a 5S fragment. This fragment can be further cleaved using a thiol reducing agent, and optionally a blocking group for the sulfhydryl groups resulting from cleavage of disulfide linkages, to produce 3.5S Fab' monovalent fragments.
  • a thiol reducing agent optionally a blocking group for the sulfhydryl groups resulting from cleavage of disulfide linkages
  • enzymatic cleavage using pepsin produces two monovalent Fab' fragments and an Fc fragment directly.
  • an Fv is composed of paired heavy chain variable and light chain variable domains. This association may be noncovalent (see, for example, Inbar, D. et al. (1972). Localization of antibody-combining sites within the variable portions of heavy and light chains. Proc Natl Acad Sci USA 69, 2659-2662). Alternatively, as described hereinabove, the variable domains may be linked to generate a single-chain Fv by an intermolecular disulfide bond, or alternately such chains may be cross-linked by chemicals such as glutaraldehyde.
  • the Fv is a single-chain Fv.
  • Single-chain Fvs are prepared by constructing a structural gene comprising DNA sequences encoding the heavy chain variable and light chain variable domains connected by an oligonucleotide encoding a peptide linker. The structural gene is inserted into an expression vector, which is subsequently introduced into a host cell such as E. coli. The recombinant host cells synthesize a single polypeptide chain with a linker peptide bridging the two variable domains.
  • Ample guidance for producing single-chain Fvs is provided in the literature of the art (see, e.g.: Whitlow, M. and Filpula, D. (1991 ). Single-chain Fv proteins and their fusion proteins.
  • Isolated complementarity-determining region peptides can be obtained by constructing genes encoding the CDR of an antibody of interest. Such genes may be prepared, for example, by RT-PCR of the mRNA of an antibody-producing cell. Ample guidance for practicing such methods is provided in the literature of the art (e.g., Larrick, J. W. and Fry, K. E. (1991). PCR Amplification of Antibody Genes. METHODS: A Companion to Methods in Enzymology 2(2), 106-110).
  • humanized antibodies are preferred.
  • Humanized forms of non-human (e.g., murine) antibodies are genetically engineered chimeric antibodies or antibody fragments having (preferably minimal) portions derived from non-human antibodies.
  • Humanized antibodies include antibodies in which the CDRs of a human antibody (recipient antibody) are replaced by residues from a CDR of a non-human species (donor antibody), such as mouse, rat, or rabbit, having the desired functionality.
  • donor antibody such as mouse, rat, or rabbit
  • the Fv framework residues of the human antibody are replaced by corresponding non-human residues.
  • Humanized antibodies may also comprise residues found neither in the recipient antibody nor in the imported CDR or framework sequences.
  • the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDRs correspond to those of a non-human antibody and all or substantially all of the framework regions correspond to those of a relevant human consensus sequence.
  • Humanized antibodies optimally also include at least a portion of an antibody constant region, such as an Fc region, typically derived from a human antibody (see, for example: Jones, P. T. et al. (1986). Replacing the complementarity- determining regions in a human antibody with those from a mouse. Nature 321 , 522-525; Riechmann, L. et al. (1988). Reshaping human antibodies for therapy. Nature 332, 323-327; Presta, L. G. (1992b). Curr Opin Struct Biol 2, 593-596; and Presta, L. G. (1992a). Antibody engineering. Curr Opin Biotechnol 3(4), 394-398).
  • a humanized antibody has one or more amino acid residues introduced into it from a source which is non-human. These non-human amino acid residues are often referred to as imported residues, which are typically taken from an imported variable domain. Humanization can be performed essentially as described (see, for example: Jones et al. (1986); Riechmann et al. (1988); Verhoeyen, M. et al. (1988). Reshaping human antibodies: grafting an antilysozyme activity. Science 239, 1534-1536; and U.S. Pat. No. 4,816,567), by substituting human CDRs with corresponding rodent CDRs.
  • humanized antibodies are chimeric antibodies, wherein substantially less than an intact human variable domain has been substituted by the corresponding sequence from a non-human species.
  • humanized antibodies may be typically human antibodies in which some CDR residues and possibly some framework residues are substituted by residues from analogous sites in rodent antibodies.
  • Human antibodies can also be produced using various additional techniques known in the art, including phage-display libraries (Hoogenboom, H. R. and Winter, G. (1991). Bypassing immunization. Human antibodies from synthetic repertoires of germline VH gene segments rearranged in vitro. J Mol Biol 227, 381-388; Marks, J. D. et al. (1991). By-passing immunization. Human antibodies from V-gene libraries displayed on phage. J Mol Biol 222, 581-597; Cole et al. (1985), Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96; and Boerner, P. et al. (1991).
  • Humanized antibodies can also be created by introducing sequences encoding human immunoglobulin loci into transgenic animals, e.g., into mice in which the endogenous immunoglobulin genes have been partially or completely inactivated. Upon antigenic challenge, human antibody production is observed in such animals which closely resembles that seen in humans in all respects, including gene rearrangement, chain assembly, and antibody repertoire. Ample guidance for practicing such an approach is provided in the literature of the art (for example, refer to: U.S. Pat. Nos.
  • antibodies After antibodies have been obtained, they may be tested for activity, for example via enzyme-linked immunosorbent assay (ELISA).
  • ELISA enzyme-linked immunosorbent assay
  • Anti-leptin antibodies as well as epitope sequences suitable for generating antibodies and antibody fragments are described in US20070104708 (SEQ ID NOs 49-55) which is incorporated herein by reference as if fully set-forth herein.
  • Leptin peptide antagonists can also be used with the present invention.
  • leptin antagonist a modified mammalian leptin polypeptide termed superactive leptin mutein is disclosed in US patent 8,969,292 which is incorporated by reference as if fully set-forth herein.
  • peptide encompass native peptides (either degradation products, synthetically synthesized peptides, or recombinant peptides), peptidomimetics (typically, synthetically synthesized peptides), and the peptide analogues peptoids and semipeptoids, and may have, for example, modifications rendering the peptides more stable while in a body or more capable of penetrating into cells. Such modifications include, but are not limited to: N-terminus modifications; C-terminus modifications; peptide bond
  • Natural aromatic amino acids, Trp, Tyr, and Phe may be substituted for synthetic non-natural acids such as, for instance, tetrahydroisoquinoline-3-carboxylic acid (TIC), naphthylelanine (Nol), ring-methylated derivatives of Phe, halogenated derivatives of Phe, and o-methyl-Tyr.
  • TIC tetrahydroisoquinoline-3-carboxylic acid
  • Nol naphthylelanine
  • ring-methylated derivatives of Phe ring-methylated derivatives of Phe
  • halogenated derivatives of Phe halogenated derivatives of Phe
  • o-methyl-Tyr o-methyl-Tyr
  • the peptides of the present invention may also include one or more modified amino acids or one or more non-amino acid monomers (e.g., fatty acids, complex carbohydrates, etc.).
  • modified amino acids e.g., fatty acids, complex carbohydrates, etc.
  • amino acid or “amino acids” is understood to include the 20 naturally occurring amino acids; those amino acids often modified post-translationally in vivo, including, for example, hydroxyproline, phosphoserine, and phosphothreonine; and other less common amino acids, including but not limited to 2-aminoadipic acid, hydroxylysine, isodesmosine, nor-valine, nor-leucine, and ornithine.
  • amino acid includes both D- and L-amino acids.
  • Amino acids are referred to by the standard three letter code. Amino acids are L amino acids unless otherwise noted, for example, by addition of the prefix "D".
  • the code Trp refers to L-tryptophan, while the codes D-Trp and DTrp refers to D-tryptophan.
  • the code Aib refers to 2-aminoisobutyric acid.
  • the code Orn refers to ornithine.
  • Lys-Ac refers to acetyllysine.
  • HomoLys refers to homolysine.
  • H-Cys refers to homocysteine.
  • peptidic leptin antagonists used to implement the teachings herein are utilized in a linear form, although in some embodiments, cyclic forms thereof are used.
  • peptidic leptin antagonists used to implement the teachings herein are synthesized by any techniques that are known to those skilled in the art of peptide synthesis.
  • solid phase peptide synthesis a summary of the many techniques may be found in: Stewart, J. M. and Young, J. D. (1963), “Solid Phase Peptide Synthesis,” W. H. Freeman Co. (San Francisco); and Meienhofer, J (1973). "Hormonal Proteins and Peptides,” vol. 2, p. 46, Academic Press (New York).
  • peptide synthesis methods comprise the sequential addition of one or more amino acids or suitably protected amino acids to a growing peptide chain.
  • amino acids or suitably protected amino acids Normally, either the amino or the carboxyl group of the first amino acid is protected by a suitable protecting group.
  • the protected or derivatized amino acid can then either be attached to an inert solid support or utilized in solution by adding the next amino acid in the sequence having the complimentary (amino or carboxyl) group suitably protected, under conditions suitable for forming the amide linkage.
  • the protecting group is then removed from this newly added amino acid residue and the next amino acid (suitably protected) is then added, and so forth; traditionally this process is accompanied by wash steps as well.
  • any remaining protecting groups are removed sequentially or concurrently, to afford the final peptide compound.
  • a preferred method of preparing the peptide compounds of the present invention involves solid- phase peptide synthesis, utilizing a solid support.
  • peptidic leptin antagonists used to implement the teachings herein are generated using cell expression approaches by utilizing expression vectors for prokaryotic or eukaryotic expression or alternatively, the peptide can be expressed in-situ by delivering a suitable expression construct to cardiovascular tissue.
  • a polynucleotide sequence encoding the peptide is preferably ligated into a nucleic acid construct suitable for mammalian cell expression.
  • a nucleic acid construct suitable for mammalian cell expression.
  • Such a nucleic acid construct includes a promoter sequence for directing transcription of the polynucleotide sequence in the cell in a constitutive or inducible manner.
  • Constitutive promoters suitable for use with the present invention are promoter sequences that are active under most environmental conditions and most types of cells, such as the cytomegalovirus (CMV) and Rous sarcoma virus (RSV).
  • CMV cytomegalovirus
  • RSV Rous sarcoma virus
  • Polyadenylation sequences can also be added to the expression vector in order to increase the efficiency of mRNA translation.
  • Two distinct sequence elements are required for accurate and efficient polyadenylation: GU- or U-rich sequences located downstream from the polyadenylation site and a highly conserved sequence of six nucleotides, namely AAUAAA, located 1 1-30 nucleotides upstream of the site.
  • Termination and polyadenylation signals suitable for the present invention include those derived from SV40.
  • the expression vector of the present invention may typically contain other specialized elements intended to increase the level of expression of cloned nucleic acids or to facilitate the identification of cells that carry the recombinant DNA.
  • a number of animal viruses contain DNA sequences that promote extra-chromosomal replication of the viral genome in permissive cell types. Plasmids bearing these viral replicons are replicated episomally as long as the appropriate factors are provided by genes either carried on the plasmid or with the genome of the host cell.
  • the expression vector of the present invention may or may not include a eukaryotic replicon. If a eukaryotic replicon is present, the vector is capable of amplification in eukaryotic cells using the appropriate selectable marker. If the vector does not comprise a eukaryotic replicon, no episomal amplification is possible.
  • Expression vectors containing regulatory elements from eukaryotic viruses such as retroviruses can be also used.
  • SV40 vectors include pSVT7 and pMT2 (SEQ ID NO 72), for instance.
  • Vectors derived from bovine papilloma virus include pBV-lMTHA, and vectors derived from Epstein-Barr virus include pHEBO and p205.
  • exemplary vectors include pMSG, pAV009/A + , pMTO10/A + , pMAMneo-5, baculovirus pDSVE, and any other vector allowing expression of proteins under the direction of the SV40 early promoter, SV40 later promoter, metallothionein promoter, murine mammary tumor virus promoter, Rous sarcoma virus promoter, polyhedrin promoter, or other promoters shown effective for expression in eukaryotic cells.
  • Viruses are very specialized infectious agents that have evolved, in many cases, to elude host defense mechanisms. Typically, viruses infect and propagate in specific cell types.
  • the targeting specificity of viral vectors utilizes its natural specificity to specifically target predetermined cell types and thereby introduce a recombinant gene into the infected cell.
  • the type of vector used by the present invention will depend on the cell type transformed. The ability to select suitable vectors according to the cell type transformed is well within the capabilities of the ordinarily skilled artisan and as such, no general description of selection considerations is provided herein.
  • bone marrow cells can be targeted using the human T-cell leukemia virus type I (HTLV-I) and kidney cells may be targeted using the heterologous promoter present in the baculovirus Autographa californica multiple nucleopolyhedro virus (AcMNPV), as described by Liang, C. Y. et al. (2004). High efficiency gene transfer into mammalian kidney cells using baculovirus vectors. Arch Virol 149, 51-60.
  • HTLV-I human T-cell leukemia virus type I
  • AcMNPV baculovirus Autographa californica multiple nucleopolyhedro virus
  • Recombinant viral vectors are useful for in vivo expression of a leptin peptide since they offer advantages such as lateral infection and targeting specificity.
  • Lateral infection is inherent in the life cycle of retrovirus, for example, and is the process by which a single infected cell produces many progeny virions that bud off and infect neighboring cells. The result is the rapid infection of a large area of cells, most of which were not initially infected by the original viral particles. This is in contrast to vertical-type infection in which the infectious agent spreads only through daughter progeny.
  • Viral vectors can also be produced that are unable to spread laterally. This characteristic can be useful if the desired purpose is to introduce a specified gene into only a localized number of targeted cells.
  • compositions also called, compositions-of-matter
  • a carrier for local delivery of the leptin antagonist can be a mesh (Figure lc) an injectable gel (e.g. in-situ forming depot) ( Figures la), a thin (preferably biodegradable) film (Figure lb), a scaffold ( Figure 3).
  • the composition is a coating on a medical device ( Figure 27A).
  • a medical device is impregnated with the composition (Figure 27B).
  • the composition is in the form of a sheet (such as the film of Figure lb or the mesh of Figure 1 c) that constitutes a portion of a medical device such as a stent cover (Figure 27C) or graft of a graft-stent assembly (Figure 27D).
  • a medical device such as a stent cover ( Figure 27C) or graft of a graft-stent assembly ( Figure 27D).
  • the composition constitutes a medical device (Figure 27E).
  • a carrier of a composition is a balloon catheter, or a composition is delivered locally using a drug-eluting balloon catheter ( Figure 2).
  • Figure 2 The manufacture and use of drug-eluting balloons for localized delivery of active-pharmaceutical ingredients are well known in the art (especially to the walls of fluid-filled bodily cavities, such as of the cardiovascular system), for example, the In.Pact Admiral® DCB drug-coated balloon by Medtronic (Dublin, Ireland) and Lutonix® 035 by C. R. Bard, Inc. (Murray Hill, New Jersey, USA).
  • in-situ formed depots examples include semi-solid polymers which can be injected as a melt and form a depot upon cooling to body temperature or two part systems which gel upon mixing ( Figure 3a).
  • such compositions can be injected into or in contact with bodily tissue that is to be treated
  • the requirements for a semi-solid ISFDs include low melting or glass transition temperatures in the range of 25-65°C and an intrinsic viscosity in the range of 0.05-0.8 dl/g [12-14]. Below the viscosity threshold of 0.05 dl/g no delayed diffusion could be observed, whereas above 0.8 dl/g the ISFD was no longer injectable using a needle. At injection temperatures above 37°C but below 65°C these polymers behave like viscous fluids which solidify to highly viscous depots. Drugs are incorporated into the molten polymer by mixing without the application of solvents. In the art, it is known to use thermoplastic pastes (TP) can be used to generate a subcutaneous drug reservoir from which diffusion occurs into the systemic circulation.
  • TP thermoplastic pastes
  • thermoplastic paste is used to generate a composition for the sustained release of leptin antagonist from which diffusion occurs into tissue in contact with the composition, thereby effecting sustained-release local administration of the leptin antagonist.
  • In situ cross-linked polymer systems utilize a cross-linked polymer network to control the diffusion of bioactive agents (e.g., leptin antagonists for implementing the teachings herein) over a prolonged period of time, thereby allowing implementation of sustained release compositions comprising leptin antagonists for use in local administration thereof.
  • bioactive agents e.g., leptin antagonists for implementing the teachings herein
  • Use of in situ cross-linking implants necessitate protection of the bioactive agents during the cross-linking reaction. This could be achieved by encapsulation into fast degrading gelatin micro-particles.
  • An ISFD can also be based on polymer precipitation.
  • a water-insoluble and biodegradable polymer is dissolved in a biocompatible organic solvent to which leptin antagonist is added forming a solution or suspension after mixing that constitutes a composition according to the teachings herein.
  • leptin antagonist is added forming a solution or suspension after mixing that constitutes a composition according to the teachings herein.
  • this composition is injected into the body of a subject in need thereof the water miscible organic solvent dissipates and water penetrates into the organic phase. This leads to phase separation and precipitation of the polymer forming a depot at the site of injection.
  • AtrigeleTM ARTIX Laboratories
  • the thus-formed depot is a composition for the sustained release of leptin antagonist from which diffusion occurs into tissue in contact with the composition, thereby effecting sustained-release local administration of the leptin antagonist.
  • Thermally induced gelling systems can also be used as ISFDs. Numerous polymers show abrupt changes in solubility as a function of environmental temperature.
  • the prototypic thermosensitive polymer is poIy(N-isopropyl acryl amide), poly-NIPAAM, which exhibits a rather sharp lower critical solution temperature.
  • Thermoplastic pastes such as the new generation of poly(ortho-esters) developed by AP Pharma can also be used for depot drug delivery.
  • Such pastes include polymers that are semi-solid at room temperature, hence heating for drug incorporation and injection is no longer necessary. Injection is possible through needles no larger than 22 gauge.
  • the leptin antagonist is mixed into the systems in a dry and, therefore, stabilized state. Shrinkage or swelling upon injection is thought to be marginal and, therefore, the initial drug burst is expected to be lower than in the other types of ISFD. An additional advantage is afforded by the self-catalyzed degradation by surface erosion.
  • IFSD compositions are suitable for effecting sustained-release local administration of the leptin antagonist.
  • an IFSD composition can be formulated for sustained-release (SR), extended- release (ER, XR, or XL), time-release or timed-release, controlled-release (CR), or continuous-release.
  • Examples of thin films suitable for release of a leptin antagonist (or polynucleotide encoding same) include polymeric films (for a review of thin films, see Zelikin ACS Nano, 2010, 4 (5), pp 2494-2509; Venkat et al. 2010, Polymer Thin Films for Biomedical Applications, Wiley VCH Verlag GmbH & Co. KGaA, Weinheim).
  • Such thin film carriers can be biodegradable or dissolvable over time.
  • PLLA can also be used for local delivery of a leptin antagonist.
  • Such microspheres can be produced as described by Kim and Park (J Control Release. 2004 Jul 23;98(1): 1 15-25).
  • a balloon such as an angioplasty balloon can also be used to deliver a leptin antagonist to a vascular wall or an inner wall of a heart chamber.
  • Approach for coating/loading a balloon with a peptide are described in EP2643030; US8617136;
  • drug-eluting balloons for use for localized delivery of active-pharmaceutical ingredients are well known in the art, for example, the In. Pact Admiral® drug-coated balloon by Medtronic (Dublin, Ireland) and Lutonix® 035 by C. R. Bard, Inc. (Murray Hill, New Jersey, USA). It is important to note that such drug-eluting balloons are known to administer extended release compositions of active pharmaceutical ingredients, e.g., In.Pact Admiral delivers a composition that provides extended release of a continuous therapeutic dose of Paclitaxel for over 180 days.
  • leptin or leptin receptor binding agents such as those described above (or expression thereof in cardiovascular cells)
  • downregulation of leptin activity at specific tissues can also be effected at the transcript level using a variety of molecules that interfere with transcription and/or translation (e.g., antisense, siRNA, Ribozyme, or DNAzyme).
  • RNA interference can be used to downregulate endogenous leptin via a small interfering RNA (siRNA) molecule.
  • siRNA small interfering RNA
  • RNAi is a two-step process, in the first, the initiation step, input double- stranded (dsRNA) is digested into 21- to 23-nucleotide (nt) small interfering RNAs (siRNAs), probably by the action of Dicer, a member of the RNase III family of dsRNA-specific ribonuc leases, which processes (cleaves) dsRNA (introduced directly or by means of a transgene or a virus) in an ATP-dependent manner.
  • dsRNA input double- stranded
  • nt small interfering RNAs
  • RNAi Nature abhors a double-strand. Curr Opin Gen Dev 12, 225-232; and Bernstein, E. (2001 ). Role for a bidentate ribonuclease in the initiation step of RNA interference. Nature 409, 363-366).
  • the siRNA duplexes bind to a nuclease complex to form the RNA-induced silencing complex (RISC).
  • RISC RNA-induced silencing complex
  • An ATP-dependent unwinding of the siRNA duplex is required for activation of the RISC.
  • the active RISC targets the homologous transcript by base-pairing interactions and cleaves the mRNA into 12-nucieotide fragments from the 3' terminus of the siRNA (Hutvagner and Zamore (2002); Hammond et al. (2001) Nat. Rev. Gen. 2:1 10-1 19 (2001); and Sharp, P. A. (2001). RNA interference. Genes Dev J5, 485-490). Although the mechanism of cleavage remainsl to be elucidated, research indicates that each RISC contains a single siRNA and an RNase (Hutvagner and Zamore (2002)).
  • RNAi molecules suitable for use with the present invention can be effected as follows. First, the leptin mRNA sequence (SEQ ID NO 2) is scanned downstream of the AUG start codon for AA-dinucIeotide sequences. Occurrence of each A A and the 19 3'-adjacent nucleotides is recorded as a potential siRNA target site.
  • siRNA target sites are selected from the open reading frame (ORF), as untranslated regions (UTRs) are richer in regulatory protein binding sites. UTR-binding proteins and/or translation initiation complexes may interfere with binding of the siRNA endonuclease complex (Tuschl (2001)).
  • siRNAs directed at untranslated regions may also be effective, as demonstrated for GAPDH, wherein siRNA directed at the 5' UTR mediated about a 90% decrease in cellular GAPDH mRNA and completely abolished protein levels (wwwdotambiondotcom/techlib/tn/91/912dothtml).
  • potential target sites are compared to an appropriate genomic database (e.g., human, mouse, rat, etc.) using any sequence alignment software, such as the BlastN software available from the NCBI server (wwwdotncbidotnlmdotnihdotgov/BLAST/). Putative target sites that exhibit significant homology to other coding sequences are filtered out.
  • an appropriate genomic database e.g., human, mouse, rat, etc.
  • sequence alignment software available from the NCBI server (wwwdotncbidotnlmdotnihdotgov/BLAST/).
  • Qualifying target sequences are selected as templates for siRNA synthesis.
  • Preferred sequences are those including low G/C content, as these have proven to be more effective in mediating gene silencing as compared with sequences including G/C content higher than 55%.
  • Several target sites are preferably selected along the length of the target gene for evaluation.
  • a negative control is preferably used in conjunction.
  • Negative-control siRNAs preferably include the same nucleotide composition as the siRNAs but lack significant homology to the genome. Thus, a scrambled nucleotide sequence of the siRNA is preferably used, provided it does not display any significant homology to any other gene.
  • DNAzyme molecule Another agent capable of downregulating leptin is a DNAzyme molecule, which is capable of specifically cleaving an mRNA transcript or a DNA sequence of the leptin.
  • DNAzymes are single-stranded polynucleotides that are capable of cleaving both single- and double- stranded target sequences (Breaker, R. R. and Joyce, G. F. (1995).
  • a general purpose RNA-cleaving DNA enzyme Proc Natl Acad Sci USA 94, 4262-4266).
  • DNAzymes complementary to bcr-abl oncogenes were successful in inhibiting the oncogene's expression in leukemia cells, and in reducing relapse rates in autologous bone marrow transplants in cases of Chronic Myelogenous Leukemia (CML) and Acute Lymphoblastic Leukemia(ALL).
  • CML Chronic Myelogenous Leukemia
  • ALL Acute Lymphoblastic Leukemia
  • Downregulation of leptin can also be effected by using an antisense polynucleotide capable of specifically hybridizing with an mRNA transcript encoding leptin.
  • the first aspect is delivery of the oligonucleotide into the cytoplasm of the appropriate cells, while the second aspect is design of an oligonucleotide that specifically binds the designated mRNA within cells in a manner inhibiting the translation thereof.
  • Ribozyme molecule capable of specifically cleaving an mRNA transcript encoding leptin.
  • Ribozymes increasingly are being used for the sequence-specific inhibition of gene expression by the cleavage of mRNAs encoding proteins of interest (Welch, P. J. et al. (1998). Expression of ribozymes in gene transfer systems to modulate target RNA levels. Curr Opin Biotechnol 9, 486-496).
  • TFOs triplex-forming oligonucleotides
  • the present invention can be used to treat cardiovascular disorders affecting heart or vascular tissue.
  • the following describes several option for local delivery of a leptin antagonist to tissue, for example heart and other cardiovascular tissue, specifically muscle and valve tissue.
  • Arterial catheterization can be used to inject as a bolus, or to deploy a medical device such as a mesh, a thin film, a biodegradable scaffold, a stent cover, a stent, a graft assembly, a coil, a stent, a ring or a prosthetic cardiac valve loaded with a leptin antagonist against a luminal wall of an ascending aorta distal to the orifice of the coronary arteries.
  • a medical device such as a mesh, a thin film, a biodegradable scaffold, a stent cover, a stent, a graft assembly, a coil, a stent, a ring or a prosthetic cardiac valve loaded with a leptin antagonist against a luminal wall of an ascending aorta distal to the orifice of the coronary arteries.
  • a medical device such as a mesh, a thin film, a biodegradable scaffold, a
  • An IFSD (for example, as described above, e.g., a gel) loaded with a leptin antagonist can be delivered via a balloon or needle to the aortic wall.
  • the leptin antagonist extended release film or mesh can be applied via open surgery or minimally invasive laparoscopy.
  • a method of treatment comprising: exposing in vivo tissue of a subject in need thereof to a pharmaceutically-effective amount of leptin antagonist thereby providing a therapeutic effect to the tissue.
  • a composition comprising leptin antagonist is administered (e.g., by injection) locally and selectively via intra-vascular approach, or directly into the tissue.
  • the exposing of the in vivo tissue to the leptin antagonist is substantially continuously for a period of not less than three days. In some embodiments, the period is not less than five days, not less than 8 days and even not less than 14 days.
  • an extended release composition comprising leptin antagonist (e.g., a medical device impregnated with, coated with or made from a leptin antagonist is placed directly in contact with the tissue-
  • a method of treatment comprising implanting in contact with tissue in need thereof in the body of a subject a composition configured for the in vivo release of leptin antagonist, thereby providing a therapeutic effect to the tissue.
  • the composition is configured for sustained release of the leptin antagonist.
  • the implanting is intracavitary implanting within a fluid-filled bodily cavity of the subject.
  • sustained release is meant that, when the composition is implanted in vivo, leptin antagonist is released from the composition in pharmaceutically effective amounts for a period of not less than three days, in some embodiments not less than five days, not less than 8 days and even not less than 14 days.
  • the subject in need thereof is any suitable mammalian subject.
  • the subject is a non-human animal.
  • the subject is a human.
  • the need is any suitable need.
  • the need is at least one need selected from the group consisting of: attenuating a pathology; reducing the chance of developing a pathology; reducing the rate of development a pathology; and mitigating the effect of a pathology.
  • the pathology is any pathology that can be treated by local administration of leptin antagonist, and in some embodiments, substantial continuous local exposure to leptin antagonist, in some embodiments, the pathology is at least one pathology selected from the group consisting of:
  • ascending aortic aneurysm in some embodiments ascending aortic aneurysm associated with at least one member of the group consisting of hypertension, dyslipidemia, hypercholesterolemia, obesity, diabetes mellitus and bicuspid aortic valve (BAV);
  • thoracic aortic aneurysm e.g., to prevent rupture or dissection thereof
  • Takayasu disease e.g., to attenuate cellular proliferative response,and aortic wall remodeling in some embodiments, by administration or implantation of a leptin antagonist composition to an inner or outer vessel wall in the vicinity of a vascular lesion;
  • Rheumatoid arteritis e.g., to attenuate aortic wall remodeling, in some embodiments, by administration or implantation of a leptin antagonist composition to an inner or outer vessel wall in the vicinity of a vascular lesion;
  • Marian's syndrome by mitigation or prevention of ascending aortic aneurysms or pulmonary artery aneurysms, in some embodiments, by perivascular administration or deployment of a leptin antagonist composition to the outer or inner wall of the ascending aorta); giant cell arteritis; ankylosing spondylitis; inflammatory aortic aneurysm; peripheral arterial or venous aneurysms; prevention of arterial dilatation at site of anchorage of bridging stent grafts ("landing zone") applied for EVAR (in the abdominal or thoracic aorta),visceral or peripheral arteries; prevention of myointimal hyperplasia at sites of vascular injury; prevention of restenosis following PTA or PTCA (peripheral or cardiac balloon angioplasty); angiogenesis; cancer; and arteriovenous malformation (e.g., administration of leptin antagonist composition directly into a malformation or into the feeding artery).
  • any pathology may be treated in accordance with the teachings herein by local administration of leptin antagonist.
  • administration is local administration of a dose (optionally repeated) of leptin antagonist, for example by direct intravascular injection into the affected tissue or tissue proximal to the affected tissue or with the use of a drug- eluting balloon.
  • administration is local administration by a sustained release composition (that releases a pharmaceutically-effective amount of leptin antagonist for a period of not less than three days) placed in contact with an outer surface of tissue (e.g., with tunica externa), inside the tissue (e.g., injection of a IFSD as described above into the tissue) or with a composition placed in contact with an inner surface of a tissue (e.g. with tunica intima).
  • a sustained release composition that releases a pharmaceutically-effective amount of leptin antagonist for a period of not less than three days
  • an outer surface of tissue e.g., with tunica externa
  • inside the tissue e.g., injection of a IFSD as described above into the tissue
  • a composition placed in contact with an inner surface of a tissue (e.g. with tunica intima).
  • the tissue is any suitable tissue.
  • the tissue is part of the cardiovascular system.
  • the tissue is selected from the group consisting of arteries, coronary arteries, ascending aorta, abdominal aorta, pulmonary artery mitral valve, aortic vaive and pulmonary valve.
  • the tissue is cardiovascular tissue with accumulated plaque, for example, an artery with accumulated plaque.
  • the tissue is a tumor, especially a cancerous tumor that grows or spreads in a process that includes angiogenesis.
  • the tissue is an arteriovenous malformation.
  • the leptin antagonist composition is locally administered during or post surgery, e.g., following carotid thrombendartrectomy or after ablation of atherosclerotic occlusion from a vessel.
  • the leptin antagonist is locally administered by contact to the outside of tissue to be treated, e.g., in contact with tunica externa.
  • the leptin antagonist is locally administered inside tissue, for example, is injected or implanted inside tissue such as a tumor or the site of arteriovenous malformation.
  • the leptin antagonist is locally administered intraluminally, e.g., a leptin antagonist composition is deployed in contact with a tunica intima, inside a fluid-filled bodily cavity such as inside the lumen of a blood vessel e.g., using an intraluminal catheter, for example, in conjunction with a stent or prosthetic cardiac valve.
  • local administration of a leptin antagonist in accordance with the teachings herein at the ascending aorta may be effective in attenuating ascending aortic aneurysms, as well as moderating left ventricular hypertrophy, and left heart valve thickness (aortic and mitral).
  • Administration of leptin antagonist at arterial aneurysms in other locations is anticipated to achieve a similar outcome, attenuating aneurysm expansion.
  • embodiments of the teachings herein are used to treat cardiovascular disorders such as heart valve stenosis, arterial or venous aneurysms, or left ventricular remodeling by enabling localized release of a leptin antagonist at the site of treatment.
  • a pharmaceutical composition comprising: as an active ingredient a leptin antagonist; and a pharmaceutically acceptable carrier configured for in vivo sustained release of the leptin antagoni st.
  • a method of making a pharmaceutical composition comprising: combining a leptin antagonist; and a pharmaceutically acceptable carrier configured for in vivo sustained release of the leptin antagonist.
  • sustained release is meant that, when the composition is implanted in vivo, leptin antagonist is released from the composition in pharmaceutically effective amounts for a period of not less than three days, in some embodiments not less than five days, not less than 8 days and even not less than 14 days.
  • the in vivo implantation is in a human subject.
  • the in vivo implantation is in any suitable location.
  • the in vivo implantation is contacting an organ through a serous tissue layer or adventitia (tunica externa) layer covering the organ, e.g., is placed contacting a blood vessel such as the aorta from the outside of the blood vessel.
  • the in vivo implantation is outside an organ directly contacting tissue of the organ (for organs covered with serous tissue, the composition is implanted underneath the serous tissue).
  • the in vivo implantation is into an organ.
  • the implantation is from inside a hollow defined by the organ, for example, inside a blood vessel lumen contacting the endothelium thereof.
  • the composition is in the form of a leptin antagonist containing sheet, in some such embodiments configured to be contacted with in vivo tissue, for example, by suturing, with the use of biological adhesive, or pressed against the tissue, for example with the help of a stent or such component, e.g., the sheet is used as a stent cover for a balloon-expandable or self-expanding stent.
  • the composition is configured to coat or be supported by an implantable medical device, e.g., is used as a coating for, is adsorbed or absorbed into or onto a stent (thereby constituting a drug-eluting balloon expandable or self-expanding stent), prosthetic valve (e.g., cardiac valve), implantable spike or rod.
  • an implantable medical device e.g., is used as a coating for, is adsorbed or absorbed into or onto a stent (thereby constituting a drug-eluting balloon expandable or self-expanding stent), prosthetic valve (e.g., cardiac valve), implantable spike or rod.
  • the composition is formed into the shape of an implantable medical device, e.g., a bioresorbable stent, a bioresorbable spike or rod.
  • an implantable medical device e.g., a bioresorbable stent, a bioresorbable spike or rod.
  • the composition is injectable, e.g., is a viscous fluid or a fluid that subsequent to injection solidifies or gels, e.g., a hydrogel.
  • the carrier comprises a biodegradable polymer.
  • the carrier comprises a polymer selected from the group consisting of a hydrogel, poly giycolic acid (PGA), poly lactic co-glycolic acid (PLGA), polylactide (PLA), and poly (L-lactide) (PLLA), and combinations thereof.
  • compositions are in accordance with those known in the art of pharmacology, using any suitable method or combination of methods as known in the art such as described in ''Remington's Pharmaceutical Sciences," Mack Publishing Co., Easton, PA, latest edition, which is incorporated herein by reference. Such methods include conventional mixing, curing, polymerizing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing.
  • Pharmaceutical compositions may be formulated in conventional manner using one or more physiologically acceptable carriers, diluents, excipients or auxiliaries which facilitate processing of the leptin antagonist into a pharmaceutical composition.
  • a use of a leptin antagonist according to the teachings herein for the localized treatment of tissue of a living organism comprising, implanting a composition comprising a leptin antagonist in vivo to contact tissue in need thereof so that the tissue is exposed to a pharmaceutically effective amount of leptin antagonist, thereby providing a therapeutic effect to the tissue.
  • the composition and the implanting is such that the tissue is exposed to a pharmaceutically effective amount of leptin antagonist substantially continuously for a period of at least three days. In some embodiments, the period is not less than five days, not less than 8 days and even not less than 14 days.
  • an above administration is periodically repeated.
  • administration of leptin antagonist is repeated.
  • administration of leptin antagonist is repeated after at least a period, the period selected from the group consisting of 1 week, 2 weeks, 3 weeks, 4 weeks, 1 month, 2 months and 3 months.
  • stable athersclerotic plaque accumulates in the inner walls of mammalian arteries.
  • the stable plaque transforms into an unstable lesion such as vulnerable plaque or rupture-prone plaque. Fragment from the unstable lesion may disintegrate, forming emboli.
  • An aspect of the teachings herein is based on the Inventor's discovery that locally synthesized leptin within the carotid atherosclerotic plaque, which characterize unstable plaques (rupture prone), correlates with brain emboli. Therefore, the inventor believes that a leptin antagonist administered to a stable athersclerotic plaque should reduce the rate and/or incidence of the conversion of a stable athersclerotic plaque to an unstable lesion.
  • a method for treatment of athersclerotic plaque comprising: administering a pharmaceutically-effective amount of a leptin antagonist to athersclerotic plaque accumulated in the inner walls of an artery, thereby at least one of: (a) reducing the rate and (b) reducing the incidence, of conversion of a stable athersclerotic plaque to an unstable lesion.
  • the administration is by sustained-release of the leptin antagonist directly to an inner wall in which plaque is accumulated, from a leptin antagonist containing composition.
  • sustained release is substantial continuous release of a pharmaceutically-effective amount of leptin antagonist for a period of not less than three days, not less than 5 days, not less than 8 days and even not less than 14 days.
  • the composition is in direct contact with the surface of the plaque to be treated.
  • the composition is in contact with the inner walls of a blood vessel with accumulated plaque.
  • MIH myointimal hyperplasia
  • MIH Magnetic resonance
  • a surgical connecting device comprising: a solid device body made of a material; and functionally associated with the device body, a pharmaceutically-effective amount of leptin antagonist.
  • the device body is in the form selected from the group consisting of surgical suture thread and a surgical staple.
  • FIG. 27 An embodiment of a suture thread 10 and of a surgical staple 12 in accordance with the teachings herein are schematically depicted in Figure 27.
  • the device body e.g., of thread 10 or staple 12 is absorbable (i.e., bioresorbable).
  • the device body, e.g., of thread 10 or staple 12 is made of any suitable material.
  • the device body is made of a material comprises a polymer selected from the group consisting of poly glycolic acid (PGA), poly lactic co-glycolic acid (PLGA), polylactide (PLA), and poly (L-lactide) (PLLA), and combinations thereof.
  • the leptin antagonist is functionally associated with the device body, e.g., of thread 10 or staple 12, in any suitable manner.
  • the functional association of the leptin antagonist with the device body is that a composition comprising the leptin antagonist coats the device body.
  • thread 10 or staple 12 are made of PGA, and coated with a coating of PLGA that includes a pharmaceutically effective amount of leptin antagonist.
  • the functional association of the leptin antagonist with the device body is that a composition comprising the leptin antagonist impregantes the device body.
  • thread 10 is made of silk and impregnated with a composition that includes a pharmaceutically effective amount of leptin antagonist.
  • the functional association of the leptin antagonist with the device body is that the material from which the device body is made is a composition comprising the leptin antagonist.
  • thread 10 or staple 12 are made of PGA that includes a pharmaceutically effective amount of leptin antagonist.
  • a fluid-filled cavity of the body for example of the cardiovascular system such as blood vessels or cardiac chambers
  • a desirable pharmaceutical effect on tissue in proximity of the implanted composition with limited or no substantia] side-effects:
  • a method of treating a condition in a subject in need thereof comprising administering intracavitarily to inner walls of a fluid-filled bodily cavity of the subject a composition comprising a leptin antagonist.
  • the subject is human.
  • the subject is a non-human animal.
  • the intracavitary administration exposes in vivo tissue to a pharmaceutically-effective amount of the leptin antagonist and thereby provides a therapeutic effect to the in vivo tissue.
  • the in vivo tissue comprises tissue of the inner walls of the cavity (e.g., tunica intima).
  • the administration is local administration.
  • the in vivo tissue exposed to the pharmaceutically-effective amount of the leptin antagonist is exclusively tissue in physical proximity to the administered composition.
  • the in vivo tissue exposed to the pharmaceutically- effective amount of the leptin antagonist is exclusively tissue in physical contact with the administered composition.
  • exposing of the in vivo tissue to a pharmaceutically-effective amount of the leptin antagonist is substantially continuously for a period of at least three days, at least five days, at least eight days and in some embodiments, even at least fourteen days.
  • the composition is a sustained-release composition, configured for sustained release of a pharmaceutically-effective amount of the leptin antagonist when located inside a bodily cavity.
  • the sustained release comprises release of a pharmaceutically-effective amount of the leptin antagonist over a period of at least three days, at least five days, at least eight days and in some embodiments, even at least fourteen days.
  • the intracavitary administering comprises implantation of the composition within the cavity in contact with the inner walls of the cavity.
  • the intracavitary administering comprises deploying an intracavitarily-implantable medical device in the cavity.
  • the medical device is deployed in contact with the inner walls of the cavity.
  • the leptin antagonist is functionally associated with the deployed intracavitarily-implantable medical device.
  • the leptin antagonist is functionally associated with a portion of the deployed intracavitarily- implantable medical device that contacts bodily tissue when the medical device is deployed.
  • the intracavitarily implantable medical device is selected from the group consisting of: a stent cover, a graft assembly, a coil (e.g., aneurysm coil), a stent (e.g., expandable stent, self-expanding stent, covered stent, partially covered stent, not covered stent), a ring (e.g, a graft anchor), a suture, a staple and a prosthetic cardiac valve.
  • a stent cover e.g., a graft assembly
  • a coil e.g., aneurysm coil
  • a stent e.g., expandable stent, self-expanding stent, covered stent, partially covered stent, not covered stent
  • a ring e.g, a graft anchor
  • teachings herein are applicable to any prosthetic cardiac valve, the teachings are particularly advantageous for implementing with catheter-deployed prosthetic cardiac valves (e.g., TAMVI, TAVI): since these valves are typically held in place without sutures so that myointimal hyperplasia that potentially develops as a result of trauma caused during deployment may lead to leakage.
  • catheter-deployed prosthetic cardiac valves e.g., TAMVI, TAVI
  • compositions comprising: a leptin antagonist for use in treating a condition, wherein the composition is configured for intracavitary administration to inner walls of a fluid-filled bodily cavity of a subject.
  • the subject is human.
  • the subject is a non-human animal.
  • the intracavitary administration of the composition exposes in vivo tissue to a pharmaceutically-effective amount of the leptin antagonist, thereby providing a therapeutic effect to the in vivo tissue.
  • such in vivo tissue comprises tissue of inner walls of a the cavity.
  • the administration is local administration.
  • the in vivo tissue exposed to the pharmaceutically-effective amount of the leptin antagonist is exclusively tissue in physical proximity to the administered composition.
  • the in vivo tissue exposed to the pharmaceutically-effective amount of the leptin antagonist is exclusively tissue in physical contact with the administered composition.
  • exposing of the in vivo tissue to a pharmaceutically-effective amount of the leptin antagonist is substantially continuously for a period of at least three days, at least five days, at least eight days and in some embodiments, even at least fourteen days.
  • the composition is a sustained-release composition, configured for sustained release of a pharmaceutically-effective amount of the leptin antagonist when located inside a bodily cavity.
  • the sustained release comprises release of a pharmaceutically-effective amount of the leptin antagonist over a period of at least three days, at least five days, at least eight days and in some embodiments, even at least fourteen days.
  • the configuration for intracavitary administration comprises configuration for implantation of the composition within the cavity in contact with the inner walls of the cavity.
  • the configuration for intracavitary administration comprises configuration for deploying with an intracavitarily-implantabie medical device in the cavity.
  • the medical device is configured for deployment in contact with the inner walls of the cavity.
  • the leptin antagonist is functionally associated with the intracavitarily-implantabie medical device.
  • the leptin antagonist is functionally associated with a portion of the intracavitarily-implantabie medical device that contacts bodily tissue when the medical device is deployed.
  • the intracavitarily implantable medical device is selected from the group consisting of: a stent cover, a graft assembly, a coil (e.g., aneurysm coil), a stent (e.g., expandable stent, self-expanding stent, covered stent, partially covered stent, not covered stent), a ring (e.g, a graft anchor), a suture, a staple and a prosthetic cardiac valve.
  • a stent cover e.g., a graft assembly
  • a coil e.g., aneurysm coil
  • a stent e.g., expandable stent, self-expanding stent, covered stent, partially covered stent, not covered stent
  • a ring e.g, a graft anchor
  • suture e.g., a staple and a prosthetic cardiac valve.
  • the condition is a pathological cardiovascular condition.
  • the condition is a cardiovascular condition selected from the group consisting of atherosclerosis, valve stenosis, aneurysms, vessel response to vascular injury, and cardiomyopathy.
  • the administration of the leptin antagonist is to athersclerotic plaque accumulated in the inner walls of an artery, thereby at least one of: (a) reducing the rate and (b) reducing the incidence, of conversion of a stable athersclerotic plaque to an unstable lesion.
  • the administration of the leptin antagonist is to bodily tissue in order to prevent or mitigate the development of myointimal hyperplasia, for example, myointimal hyperplasia that potentially develops as a result of trauma caused during deployment of a medical device in a the body of a living subject.
  • the composition constitutes a coating of an intracavitarily implantable medical device.
  • the composition is impregnated in an intracavitarily implantable medical device.
  • the composition comprises, in addition to the leptin antagonist, a polymer selected from the group consisting of a hydrogel, poly glycolic acid (PGA), poly lactic co-glycolic acid (PLGA), polylactide (PLA), and poly (L-lactide) (PLLA), and combinations thereof.
  • the composition is in the form selected from the group consisting of a sheet and a tube constituting a portion of an intracavitarily implantable medical device comprising the leptin antagonist.
  • the composition comprises, in addition to the leptin antagonist, a polymer selected from the group consisting of of a hydrogel, poly glycolic acid (PGA), poly lactic co-glycolic acid (PLGA), polylactide (PLA), and poly (L-lactide) (PLLA), and combinations thereof.
  • the intracavitarily implantable medical device comprises a stent and the composition constitutes a stent cover (e.g., a partial or complete stent cover, a balloon-expandable or a self-expanding stent).
  • the intracavitarily implantable medical device comprises a graft-assembly (e.g., a stent-graft or ring-graft assembly) and the composition constitutes a graft portion thereof.
  • some embodiments are configured to function as a stent-graft assembly for treatment of AAA, like the Endurant® II by Medtronic (Dublin, Ireland).
  • the composition constitutes at least a portion of an intracavitarily implantable medical device, and in some embodiments, the composition constitutes substantially an entire intracavitarily implantable medical device.
  • the intracavitarily implantable medical device is selected from the group consisting of a stent cover, a graft assembly, a coil (e.g., aneurysm coil), a stent (e.g., expandable stent, self-expanding stent, covered stent, partially covered stent, not covered stent), a ring (e.g, a graft anchor), a suture, a staple and a prosthetic cardiac valve.
  • a stent cover e.g., a graft assembly
  • a coil e.g., aneurysm coil
  • a stent e.g., expandable stent, self-expanding stent, covered stent, partially covered stent, not covered stent
  • the composition comprises, in addition to the leptin antagonist, a polymer selected from the group consisting of a hydrogel, poly glycolic acid (PGA), poly lactic co- glycolic acid (PLGA), polylactide (PLA), and poly (L-lactide) (PLLA), and combinations thereof.
  • a polymer selected from the group consisting of a hydrogel, poly glycolic acid (PGA), poly lactic co- glycolic acid (PLGA), polylactide (PLA), and poly (L-lactide) (PLLA), and combinations thereof.
  • an intracavitarily-implantable medical device comprising:
  • At least one solid functional device part configured for deploying the device in a fluid- filled bodily cavity of a subject
  • a leptin antagonist functionally associated with at least one the device component, a leptin antagonist.
  • the leptin antagonist is functionally associated with the at least one the device part as a component of a pharmaceutical composition comprising the leptin antagonist.
  • the pharmaceutical composition is a sustained-release composition, configured for sustained release of a pharmaceutically- effective amount of the leptin antagonist when located inside a bodily cavity.
  • sustained release comprises release of a pharmaceutically-effective amount of the leptin antagonist over a period of at least three days, at least five days, at least eight days and in some embodiments, even at least fourteen days.
  • the pharmaceutically-acceptable carrier further comprises a polymer selected from the group consisting of a hydrogel, poly glycolic acid (PGA), poly lactic co-glycolic acid (PLGA), poiylactide (PLA), and poly (L-lactide) (PLLA), and combinations thereof,
  • the functional association is at least one device component having a coating comprising the pharmaceutical composition. In some embodiments of the medical device, the functional association is at least one device component being impregnated with the pharmaceutical composition. In some embodiments of the medical device, the functional association is at least one device component being fashioned of the pharmaceutical composition.
  • the medical device is selected from the group consisting of a stent cover, a graft assembly, a coil (e.g., aneurysm coil), a stent (e.g., expandable stent, self-expanding stent, covered stent, partially covered stent, not covered stent), a ring (e.g, a graft anchor), a suture, a staple and a prosthetic cardiac valve (as noted above, preferably catheter-deployed prosthetic cardiac valves.
  • a stent cover e.g., a graft assembly
  • a coil e.g., aneurysm coil
  • a stent e.g., expandable stent, self-expanding stent, covered stent, partially covered stent, not covered stent
  • a ring e.g, a graft anchor
  • a suture a staple and a prosthetic cardiac valve (as noted
  • the fluid-filled bodily cavity is a bodily cavity of the cardiovascular system.
  • the cavity is selected from the group consisting of a cardiac chamber, an artery and a vein.
  • the cardiac chamber is selected from the group consisting of left ventricle, right ventricle, left atrium and right atrium,
  • the artery is selected from the group consisting of a systemic artery, a coronary artery and a pulmonary artery.
  • the systemic artery is an aorta, for example selected from the group consisting of an ascending aorta, aortic arch, descending aorta and an abdominal aorta.
  • an aneurysm coil 14 according to the teachings herein is schematically depicted.
  • Aneurysm coil 14 is substantially similar to known aneurysm coils except by being functionally associated with a pharmaceutically-effective amount of leptin antagonist, is made in substantially the same way, and is used in substantially the same way.
  • a given aneurysm coil 14 may include one or more of the additional features detailed hereinabove.
  • coil 14 is made of platinum, and coated with a coating of a hydrogel that includes a pharmaceutically effective amount of leptin antagonist.
  • coil 14 is made of a material such as platinum or a polymer textured with micrometer dimension features such as valleys and pores, where inside the features is held a composition (e.g., a gel such as of Example 1 1) that comprises leptin antagonist.
  • a composition e.g., a gel such as of Example 1 1
  • coil 14 is made of a composition comprising the leptin antagonist, e.g., is made of PGA that includes a pharmaceutically effective amount of leptin antagonist.
  • Prosthetic cardiac valve 16 is substantially similar to known prosthetic cardiac valves except by being functionally associated with a pharmaceutically- effective amount of leptin antagonist, is made in substantially the same way, and is used in substantially the same way.
  • prosthetic cardiac valve 16 has at least one component that is functionally associated with a pharmaceutically-effective amount of leptin antagonist.
  • a given prosthetic cardiac valve 16 according to the teachings herein may include one or more components, each having one or more of the additional features detailed hereinabove.
  • the retainer ring of prosthetic cardiac valve 16 is made of a cobalt chromium ring with a polyester cloth cover, the ring and cloth cover both coated with a coating of a hydrogel that includes a pharmaceutically effective amount of leptin antagonist.
  • the leaflets of prosthetic cardiac valve 16 are made of a material such as porcine or bovine tissue (e.g., cardiac leaflets, pericardium) that has been soaked in and is therefore impregnated with a composition (e.g., a gel such as of Example 11) that comprises leptin antagonist.
  • a graft assembly 18 according to the teachings herein is schematically depicted.
  • Graft assembly 18 is a ring-graft assembly suitable for treatment of abdominal aorta aneurysms and includes a flexible graft 20 that defines a conduit for blood flow and three expandable anchoring rings 22 as graft anchors.
  • Each anchoring ring 22 is a radially expandable device that is substantially a single 360° ring of material.
  • some embodiments of graft assemblies are stent-graft assemblies where one or more of the anchors are radially expandable stents, that are longer in the axial direction and/or describe more than a 360° degree rotation and/or comprise more than a single ring of material.
  • Graft assembly 18 is substantially similar to known graft assemblies except by being functionally associated with a pharmaceutically-effective amount of leptin antagonist, is made in substantially the same way, and is used in substantially the same way. Specifically, graft assembly 18 has at least one component that is functionally associated with a pharmaceutically-effective amount of leptin antagonist.
  • a given graft assembly 18 may include one or more components, each having one or more of the additional features detailed hereinabove.
  • graft 20 is substantially a tube made of a high- density multifilament polyester cloth coated with a coating of a hydrogel that includes a pharmaceutically effective amount of leptin antagonist.
  • the coating is on the entire outer surface of graft 20.
  • the coating is on the outer surface of the termini of the three legs of graft 20 (e.g., a 5 cm length from each terminus.
  • graft 20 is made of a high-density multifilament polyester cloth coat that has been soaked in and is therefore impregnated with a composition (e.g., a gel such as of Example 11) that comprises leptin antagonist.
  • a composition e.g., a gel such as of Example 11
  • the entire graft 20 is impregnated with leptin antagonist composition.
  • only the termini of the three legs of graft 20 e.g., a 5 cm length from each terminus
  • rings 22 are made of nitinol, and coated with a coating of a hydrogel that includes a pharmaceutically effective amount of leptin antagonist.
  • rings 22 are made of a material such as nitinol textured with micrometer dimension features such as valleys and pores, where inside the features is held a composition (e.g., a gel such as of Example 11) that comprises leptin antagonist.
  • stents 24, 26 and 28 according to the teachings herein are schematically depicted. Stents 24, 26 and 28 are all elongated, tubular, outwardly radially-expandable frameworks that are known in the art.
  • Stent 24 is a coverless stent without a cover.
  • Stent 26 is a partially-covered stent with a partial cover 30.
  • Partial cover 30 is a sheet secured to the framework of stent 26 in the usual way, e.g., with sutures.
  • Stent 28 is a covered stent with a full cover 32.
  • Full cover 32 is a tube secured to the framework of stent 28 in the usual way, e.g., with sutures or by tension.
  • Stents 24, 26 and 28 are substantially similar to known stents except by being functionally associated with a pharmaceutically-effective amount of Ieptin antagonist, are made in substantially the same way, and are used in substantially the same way. Specifically, each one of stents 24, 26 and 28 has at least one component that is functionally associated with a pharmaceutically-effective amount of Ieptin antagonist. Depending on the embodiment, a given stent 24, 26 and 28 according to the teachings herein may include one or more components, each having one or more of the additional features detailed hereinabove. Embodiments of any one of stents 24, 26 and 28 are self-expanding stents. Embodiments of any one of stents 24, 26 and 28 are balloon-expandable stents.
  • a cover 30 or a cover 32 is made of a high- density multifilament polyester cloth coated with a coating of a hydrogel that includes a pharmaceutically effective amount of Ieptin antagonist, typically on the outer surface of the cover.
  • a cover 30 or a cover 32 is made of a high- density multifilament polyester cloth coated that has been soaked in and is therefore impregnated with a composition (e.g., a gel such as of Example 11) that comprises Ieptin antagonist.
  • a composition e.g., a gel such as of Example 11
  • a cover 30 or a cover 32 is made of a material that is a composition comprising Ieptin antagonist, e.g., a PLGA sheet of example 4.
  • a framework of any one of stents 24, 26 and 28 is made of cobalt chromium coated with a coating of a hydrogel that includes a pharmaceutically effective amount of Ieptin antagonist.
  • a framework of any one of stents 24, 26 and 28 is made of a material such as nitinol or a polymer textured with micrometer dimension features such as valleys and pores, where inside the features is held a composition (e.g., a gel such as of Example 1 1) that comprises Ieptin antagonist.
  • a framework of any one of stents 24, 26 and 28 is made of a material that is a composition comprising leptin antagonist, e.g., PLA or PLLA comprising a pharmaceutically-effective amount of leptin antagonist.
  • renin-angiotensin aldosterone system RAAS
  • AngH angiotensin II
  • RAAS renin-angiotensin aldosterone system
  • AngH angiotensin II
  • ACEIs angiotensin converting enzyme inhibitors
  • angiotensin receptor blockers have protective effects on IRI in the kidney.
  • the present invention relates to the use of leptin antagonist not only in the treatment of a cardiovascular tissue but also in the treatment of an organ injured by ischemia and reperfusion injury (IRI) for alleviating tissue damage and preserving the organ function.
  • IRI ischemia and reperfusion injury
  • ischemia and reperfusion injury by administrating leptin antagonist upon reperfusion and maintaining sustained administration of the leptin antagonist into the vessel undergoing the reperfusion procedure for delivering the leptin antagonist by the blood flow into the damaged cells of the tissue injured by the IRI.
  • the method is implemented by implanting an intracavitarily- implantable drug delivery device configured for sustained delivering the leptin antagonist.
  • the drug delivery device is a drug eluting stent that is used both to restore the flow of blood in an occluded vessel and to deliver leptin antagonist locally to the specific tissue perfused by the treated vessel.
  • the drug delivering device is a drug eluting stent
  • the stent is also configured to deliver an antiproliferative drug to the wall of the vessel for preventing restenosis.
  • the anti-proliferative drug may be any known anti-proliferative drug such as rapamycin or paclitaxel.
  • the total dose of leptin antagonist in the stent may be released into the lumen over a period of at least 3 days, preferably between 10 to 14 days, and even for a longer period.
  • the release rate is determined, inter alia, by the choice of the sustained release material.
  • PLGA Poly lactic-co-glycolic acid 65:35 MW 45000-75000Da was tested in a mouse model and was found suitable to establish this timeframe.
  • the kinetics of drug release for the antiproliferative drug may be designed based parameters known in the art.
  • the stent may be used as a stand alone device, or as a supplement to support an intra-arterial bolus treatment with leptin antagonist, depending on the specific clinical circumstances.
  • the method of treating IRI is applicable to isolated tissue sections that function as an end organ (i.e., are supplied by a single blood vessel), and that were exposed to a period of non-fatal ischemia and underwent reperfusion via thromboslysis or intravascular angioplasty.
  • the method of treating IRI is applicable for the treatment of a cardiac left ventricular tissue after acute MI at the time of primary revascularization for moderation of cellular damage in cardiac cells (cardiomyocytes) that were subjected to ischemia and reperfusion injury and for preserving cardiac function.
  • the method of treating IRI is applicable for treating IRI in any in other end organs, for example, the kidney, intestine or brain.
  • a double function drug eluting stent to be deployed in an occluded vessel at the site of occlusion following balloon angioplasty.
  • the double function drug eluting stent is configured to enable slow release of anti-proliferative drug into the wall of a vessel to enable slow release of leptin antagonist into the lumen, to be carried by the blood stream and be uptaken by cells that sustained ischemia and reperfusion injury.
  • the double function dug eluting stent comprises an expandable structural framework configured to be deployed in a blood vessel.
  • the stent may be a self-expandable stent or a balloon-expandable stent and may comprise a network of elongated struts.
  • FIG. 30 schematically depicts a drug eluting stent 40 according to the teachings herein.
  • Stent 40 is an elongated, tubular, radially-expandable stent comprising a network of struts 41.
  • Struts 41 comprise outer surface reservoirs 42 and inner surface reservoirs 44, which are located, for example, at the junctions between the struts.
  • Outer surface reservoirs 42 that abut the vessel wall when stent 40 is deployed within a vessel contain a sustained release composition of an anti-proliferative drug.
  • Inner surface reservoirs 44 which are in contact with the fluid flowing through the lumen, contain a sustained release composition of leptin antagonist.
  • Reservoirs 42 and 44 can be configured, for example, as pits embedded in the junctions points between struts 41.
  • Fig. 30 illustrates reservoirs 42 and 44 as circular and square pits, respectively, by way of example only. It will be easily realized that the pits may assume any other shape.
  • Stent 40 may be a se!f-expandable or a self-expanding stent.
  • Struts 41 may be made of metal, alloy or biodegradable polymer, as described above in association with Figure 29.
  • Figures 31 b and 31 c illustrate another embodiment of a double function drug eluting stent, depicting cross sectional views of one of the struts that constitute the stent, abutting vessel wall 50.
  • the outer surface of struts 61 and 71 is partially coated by layer, 62 and 72, respectively, of a polymer containing the antiproliferative drug. Layers 62, 72 cover about 75% of the strut's circumference.
  • the inner surface of the strut, facing the lumen, is provided with a long recessed slit, 64, 74, which serves as a reservoir for containing the sustained release composition of the leptin antagonist for slow release into the lumen.
  • Slits 64 and 74 that extend along the length of the strut have circular and triangular cross sections, respectively, but may assume other cross sectional shapes as well.
  • the slit opening, 65, 75, at the luminal aspect of the struts leads to a wider reservoir. This design prevents early detachment of the sustained release matrix.
  • the size of opening 65, 75 can also be selected as one of the parameters that control the release rate.
  • layers 62, 72 and slits 64,74 extend continuously along the length of the stent.
  • layers 54, 62 and slits 52 and 64 may be discontinuous.
  • FIG. 3 la depicts a strut 51 of a prior art drug eluting stent coated by a layer 52 of antiproliferative drug to prevent in-stent stenosis (restonosis). It is noted that prior art drug eluting stents do not provide for selective release of one active agent into the wall of a vessel and a second different active agent into the lumen to be carried by the flow of blood and to be uptaken by the cells to which the vessel supplies blood. Suitable leptin antagonists
  • leptin antagonist Any suitable leptin antagonist may be used in implementing any specific aspect or embodiment of the teachings herein. In some embodiments, a single leptin antagonist is used. In some embodiments, two or more leptin antagonists are used simultaneously or concurrently.
  • the leptin antagonist comprises a polypeptide portion.
  • the leptin antagonist is a polypeptide.
  • leptin antagonists include all of the leptin antagonists listed and taught in US 7,307,342 (SEQ ID NOs 3-35) and US 8,969,292 (SEQ ID NOs 36-47), which are both included by reference as if fully set-forth herein.
  • the leptin antagonist is selected from the group consisting of:
  • a leptin antagonist consisting of: (a) a mammalian leptin polypeptide in which the LDFI (SEQ ID NO:33 in US 7,307,142 or SEQ ID NO 35 in the present application) hydrophobic binding site at the positions corresponding to positions 39-42 of the wild-type human leptin, is modified such that from two to four amino acid residues of the hydrophobic binding site are substituted with different amino acid residues such that the site becomes less hydrophobic, the modified, mammalian leptin polypeptide being a leptin antagonist; (b) a fragment of the modified mammalian leptin polypeptide of (a) comprising the altered hydrophobic binding site, wherein the fragment is itself a leptin antagonist; (c) a fragment of (b)
  • a synthetic leptin antagonist comprising: (d) a full length modified mammalian leptin polypeptide in which: (i) the LDFI hydrophobic binding site at the position corresponding to positions 39-42 of the wild-type human leptin is modified such that from two to four amino acid residues of the hydrophobic binding site are substituted with different amino acid residues such that the site becomes less hydrophobic; and (ii) the aspartic acid at the position corresponding to position 23 of the wild-type human leptin (D23) is substituted with an amino acid residue selected from the group consisting of glycine, alanine, leucine, lysine, arginine, phenylalanine, tryptophan and histidine, or the threonine at the position corresponding to position 12 of the wild-type human leptin (T12) is substituted with a different amino acid residue that is hydrophobic; (e) a synthetic leptin antagonist consisting of a polypeptide
  • a modified mammalian leptin polypeptide in which: (i) the LDFI hydrophobic binding site at the position corresponding to 5 positions 39-42 of the wild-type human leptin is modified such that from two to four amino acid residues of the hydrophobic binding site are substituted with different amino acid residues such that the site becomes less hydrophobic, the modified mammalian leptin polypeptide being a leptin antagonist; and
  • a synthetic leptin agonist comprising: (h) a modified mammalian leptin polypeptide in which D23 is substituted with a different amino acid residue that is not negatively charged or T12 is substituted with a different amino acid residue that is hydrophobic; (j) a fragment of the modified mammalian leptin polypeptide of (h), in which D23 is substituted with a different amino acid residue that is not negatively charged or T12 is substituted with a different amino acid residue that is hydrophobic, wherein the fragment is itself a leptin agonist;
  • intracavitary relates to within an organ or body cavity.
  • Art that provides enabling support for the teachings herein, and that may also be useful in understanding the background and the inventive aspects of the teachings herein includes US 7,307,342; US 8,969,292; "leptin Locally Synthesized in Carotid Atherosclerotic Plaques Being Associated With Lesion Instability and Cerebral Emboli" by Schneiderman J et al in J Am Heart Assoc.
  • a novel mouse model was used to simulate local leptin synthesis in the ascending aorta in order to assess the effect of leptin on aortic remodeling and heart structure and function.
  • the leptin slow-release film was manufactured by impregnating a poly lactic-co- glycolic acid (PLGA) film with leptin.
  • MgCb Fesher Scientific, Loughborough, UK
  • Sodium chloride (10 mg in 0.2 mL distilled water) and 25 ethylene glycol (Sigma-Aldrich, St. Louis, MO, USA) were added to the polymeric solution and sonicated for 20 seconds.
  • Mouse leptin powder (1 mg; #L3772; Sigma-Aldrich, St.
  • Another option of leptin application for local slow-release has been a gel composed of two liquid materials which gel (solidify) upon mixing at the time of injection. These are a modified carboxymethyl cellulose with adipic dihydrazide (CMC-ADH) and an oxidized dextrane in DDW (DX-COH). Methylene blue dye (0.5%) was also added to the DX-COH solution to make the resulting gel more visible. Leptin (Sigma, L3772, St. Louis, MO, USA) was added to the gel by an emulsion technique.
  • CMC-ADH modified carboxymethyl cellulose with adipic dihydrazide
  • DX-COH oxidized dextrane in DDW
  • Methylene blue dye 0.5%) was also added to the DX-COH solution to make the resulting gel more visible.
  • Leptin Sigma, L3772, St. Louis, MO, USA
  • Serum leptin levels were determined in ApoE "A mice after receiving 20 ⁇ g mouse leptin via peri-aortic application (in another experiment, Tao et al. ATVB 2013). Blood was samples on days 0, 7, 14, and 21 , and leptinanalyzeded by ELISA assay (Quantikine Mouse Lep Kit, R&D Systems, Minneapolis, MN, USA): Day 0 - 3.5 ng/mL; day 7 - leptin 8.0 ng/mL, placebo 9.2 ng/mL; day 14 - leptin 12.0 ng/mL, placebo 14.5 ng/mL; and day 21- leptin 12.25 ng/mL, placebo 12.5 ng/mL ( Figure 5).
  • mice in experiment 1 were fed postoperatively with high fat diet (HFD), and were followed up for 45 days.
  • HFD high fat diet
  • Mouse weight and blood pressure (BP) were assessed weekly. All mice recovered from surgery uneventfully.
  • mice gained weight equally during the follow up period, suggesting no systemic leptin effect.
  • Systolic BP measured weekly in mice of experiment 2 was 100 mmHg throughout the first 4 weeks, and increased to 120 mmHg by week 6 in both leptin treated and control mice. Based on two separate experiments, both HFD and normal chow feeding yielded in general similar results.
  • Histological analysis of the ascending aorta revealed features of medial degeneration at the site of leptin application, including fragmentation of the elastic lamellas, as demonstrated by elastica van Giesen staining, and depletion of aSMA in the media (Figure 7). These structural changes likely underlie local stiffening and dilatation in the proximal ascending aorta.
  • Angiotensin II is the key hormone of the renin-angiotensin system, underlying hypertension and cardiovascular remodeling (Renna et al. Pathophysiology of vascular remodeling in hypertension. Int J Hypertens. 2013;2013: 808353).
  • the phenotypes induced by local leptin application described in Example 1 are reminiscent of Angll induced aortic-ventricular (coupling) remodeling, suggesting that leptin mediates these processes.
  • a leptin antagonist was delivered locally to the ascending aorta in order to assess the effects of leptin down-regulation on Angll induced local aortic remodeling, and aortic- ventricular remodeling in mice.
  • An osmotic mini-pump, delivering Angll at a rate of 1000 ng/kg/min was implanted subcutaneously in the back of the neck of 14 week old ApoE "7" mice.
  • Each mouse also underwent left mini-thoracotomy for application of a slow release miniature PLGA film (1x1.5 mm) containing either 5 ⁇ g leptin antagonist (LA), or PLGA matrix devoid of protein (control).
  • the slow release film was deployed on the surface of the proximal ascending aorta at the position described in Experiment 1. Mice were euthanized 4 weeks following surgery.
  • mice treated with Angll either Ang II alone or Ang II with control film applied on the ascending aorta.
  • Death was related to thoracic (28%) or abdominal (6%) aortic aneurysm rupture.
  • mice treated with LA presented less thickening of the left ventricular wall, particularly in diastole (p ⁇ 0.01, Figure 18).
  • LV diameter which increased in response to Angll treatment was preserved within the baseline (pre- Angll treatment) range in the LA treated mice (P ⁇ 0.05).
  • the PSV parameter is likely reflecting the interaction between proximal aortic hemodynamics, and left ventricular systolic contraction.
  • PSV moderation by LA may represent attenuation of both aortic and LV remodeling.
  • the present findings show that application of a leptin antagonist at the pivotal location on the proximal ascending aortic surface prevents rupture of thoracic aneurysms induced by systemic infusion of Ang 11.
  • Local inhibition of leptin activity reduces the degenerative effects of Ang II on the proximal aorta, which underlie aortic wall destabilization.
  • Human AVS and normal arterial valve (AV) samples were collected for analysis, including autopsy samples, freshly collected AVS specimens from patients undergoing aortic valve replacement surgery, and normal aortic valves from explanted hearts.
  • Formalin fixed valve samples were analyzed by immunohistochemistry for leptin, leptin receptor, CD68 and aSMA.
  • Fresh samples of AVS valves and normal aortic valves underwent total RNA extraction and analyzed by qPCR and Nanostring technique to assess leptin and leptin receptor mRNA levels. Retroperitoneal fat was used as a positive control in both assays, Results
  • VICs human valve interstitial cells
  • Poly iactic-co-glycolic acid (PLGA) films containing a leptin antagonist e.g., a leptin antagonist described in US 8,969,292
  • a leptin antagonist e.g., a leptin antagonist described in US 8,969,292
  • PLGA 6535 polymer D,L-lactide:glycolide: 65:35, Mw - 45,000- 75,000 Da; Lakeshore. Biomaterials, Birmingham, AL, USA
  • MeCI 2 Fesher Scientific, Loughborough, UK
  • Sodium chloride (10 mg in 0.2 ml distilled water) and 25 ⁇ L ethylene glycol (Sigma-Aldrich, St. Louis, MO, USA) are added to the PLGA solution and sonicated for 20 seconds, lmg of the leptin antagonist is suspended in 2 mL of the PLGA solution, followed by casting on the flat surface of a Teflon mold to create a film comprising a leptin antagonist.
  • a 2x2.5-mm patch of PLGA film of Example 4 is maintained in 5 mL sterile PBS at 37°C for 28 days. Every 7 days the PBS medium is sampled before being aspirated and replaced by 5 mL of fresh PBS. The medium samples are analyzed by ELISA (RayBiotech, Norcross, GA, USA) for leptin antagonist levels. The in vitro measurement of leptin discharged from the slow-release PLGA film yields substantial release of leptin antagonist for at least two weeks.
  • a first component of the composition is an aqueous solution of modified carboxymethyl cellulose with adipic dihydrazide (CMC-ADH). Dried and finely ground sustained-release gel with leptin antagonist as described in Example 1 is added to the CMC- ADH solution and vortexed to yield a suspension.
  • CMC-ADH modified carboxymethyl cellulose with adipic dihydrazide
  • a second component of the composition is oxidized dextrane in DDW (DX-COH).
  • DX-COH DDW
  • a dye such as methylene blue (0.5%) is optionally added to the DX-COH solution to make the resulting gel more visible.
  • the two components are mixed together to form a viscous fluid that is immediately administered by injection.
  • the viscous fluid gels.
  • a stent cover is fashioned from the PLGA film of Example 4.
  • the stent cover is used to cover the balloon expandable stents of a MULTI-LINK 8 LL coronary stent system, a MULTI-LINK ULTRA coronary stent system and a MULTI- LINK MINI VISION coronary stent system (all of Abbot Laboratories, Abbot Park, Illinois, USA).
  • the graft (stent cover) portion of an Endurant II AAA Stent Graft System (Medtronic, Dublin, Ireland) is impregnated with a leptin antagonist.
  • impregnation is by immersion in a first component of the composition of Example 3, followed by contact with contact with the second component thereof.
  • the stent cover is used as an external cover for a self-expanding WallFIex Stent
  • the covered stents are deployed in the usual way inside the lumen of a blood vessel of a living subject in need thereof. Once implanted, the leptin antagonist elutes from the stent cover through the blood vessel endothelium into the blood vessel to exert a desired pharmaceutical effect.
  • Drug-eluting stent An X1ENCE Alpine coronary stent system (Abbot Laboratories, Abbot Park, Illinois, USA) is prepared in the usual way, but impregnated with a leptin antagonist as an active pharmaceutical ingredient instead of Everolimus.
  • the resulting drug-eluting stent is deployed in the usual way inside the lumen of a blood vessel of a living subject in need thereof.
  • the leptin antagonist elutes from the stent through the blood vessel endothelium into the blood vessel to exert a desired pharmaceutical effect.
  • a bioresorbable balloon-expandable stent is fashioned of bioresorbable polylactide (PLA) comprising a leptin antagonist, substantially as done to fashion a bioresorbable stent by Arterial Remodeling Technologies (Paris, France).
  • PLA bioresorbable polylactide
  • a bioresorbable balloon-expandable stent is fashioned of bioresorbable poly (L- lactide) (PLLA) comprising a leptin antagonist and also comprises a bioresorbable coating of PLLA including a leptin antagonist, substantially as done to fashion an Absorb GTl vascular scaffold stent.
  • PLLA bioresorbable poly (L- lactide)
  • bioresorbable stents are deployed in the usual way inside the lumen of a blood vessel of a living subject in need thereof.
  • the leptin antagonist elutes from the stents as these resorb, to pass through the blood vessel endothelium into the blood vessel to exert a desired pharmaceutical effect.
  • An implantable spike or rod (bioresorbable or not) is made as known in the art, for example as described above with reference to the stents and includes leptin antagonist integrated into the material of the spike or rod, or adsorbed, absorbed or coated onto the spike or rod.
  • the resulting spike or rod is implanted in the usual way inside an organ, for example by piercing the organ.
  • the leptin antagonist elutes from the spike or rod, to exert a desired pharmaceutical effect on the organ.
  • Injectable Gel The two components of the composition of Example 3 are provided. The components are mixed together and implanted in the body of the subject by injection into or onto an organ to form a gelled mass in or on the organ.
  • the leptin antagonist elutes from the gelled mass, to exert a desired pharmaceutical effect on the organ.
  • a sheet of the film of Example 1 is provided.
  • the sheet is placed against the outer surface of an organ (e.g., aorta, for example, ascending aorta, aortic arch, descending aorta, abdominal aorta) and optionally held in place by sutures and/or biological glue (e.g., Evicel® by Ethicon of Johnson and Johnson).
  • organ e.g., aorta, for example, ascending aorta, aortic arch, descending aorta, abdominal aorta
  • biological glue e.g., Evicel® by Ethicon of Johnson and Johnson.
  • the leptin antagonist elutes from the film, to exert a desired pharmaceutical effect on the organ.
  • a human subject is diagnosed with an abdominal aortic aneurysm (AAA).
  • AAA abdominal aortic aneurysm
  • a composition according to the teachings herein in the form of a long sheet such as described in Example 12 (a ribbon) comprising leptin antagonist is provided.
  • the abdominal aorta of the subject is surgically accessed from the outside (e.g., using keyhole surgery) and the composition administered by winding the long sheet around the abdominal aorta to wrap the entire aneurysm as well as a portion of the aorta above and below the aneurysm, and held in place with a biological glue and/or sutures.
  • a stent graft e.g., an Endurant® II AAA Stent Graft System (Medtronic, Dublin, Ireland)
  • leptin antagonist from the composition passes through the tunica externa to provide a beneficial effect to the subject,
  • an AAA stent graft is provided that is similar to known AAA stent grafts (e.g., similar to an Endurant® II AAA Stent Graft System (Medtronic, Dublin, Ireland)) where at least one of: the graft and/or anchoring stents are a composition of the teachings herein and comprises a leptin antagonist; at least the anchoring portions of the graft and/or anchoring stents are impregnated with a composition of the teachings herein that comprises a leptin antagonist; and at least the anchoring portions of the graft and/or anchoring stents are coated with a composition of the teachings herein that comprises a leptin antagonist.
  • known AAA stent grafts e.g., similar to an Endurant® II AAA Stent Graft System (Medtronic, Dublin, Ireland)
  • the graft and/or anchoring stents are a composition of the teachings herein and comprises a leptin antagonist
  • the stent-graft is deployed in the usual way in the abdominal aneurysm, that is to say, where the anchoring stents are expanded against healthy portions of tunica intima above and below the aneurysm. Subsequently, leptin antagonist from the composition passes through the tunica intima to provide a beneficial effect to the subject, for example, preventing or reducing the extent that the aneurysm spreads to portions of tissue in proximity of the anchoring stents, thereby preventing loosening of the anchoring stents.
  • a drug-eluting balloon similar to the In. Pact Admiral® DCB drug-coated balloon by Medtronic (Dublin, Ireland) that is coated with a composition comprising leptin antagonist according to the teachings herein is introduced through the femoral arteries and advanced to areas of the abdominal and iliac arteries that are just above and just below the aneurysm (for example, "landing zones" where anchoring stents of a stent- graft would be deployed.
  • the balloon is expanded to contact the aorta and iliac walls, thereby administering composition to the healthy tissue and preventing advancement of the aneurysm.
  • the administration of leptin antagonist is repeated periodically, e.g., with a frequency that is less than once a month, less than once every two months and even less than once every 3 months.
  • the administration of the leptin antagonist leads to stabilization of the wall tissue, halting the processes of aneurysm formation at the portions of the blood vessels above and below the aneurysm and at the portions of the blood vessel in contact with the deployed stent graft.
  • a human subject is diagnosed with an aneurysm in the thoracic aorta, including one or more of the ascending aorta, aortic arch and descending aorta. It is known that segmental increased stiffness and aortic dilatation cause local aneurysm formation. These structural changes underlie hemodynamic perturbation, which increases left ventricular afterload. This results in left ventricular remodeling, including left ventricular hypertrophy, and thickening of the aortic and mitral valve leaflets. Left ventricular hypertrophy may lead to heart failure, and aortic valve remodeling may progress to the full clinical presentation of aortic valve stenosis.
  • a composition according to the teachings herein in the form of a patch such as described in Example 12 comprising leptin antagonist is provided.
  • the thoracic aorta of the subject is surgically accessed from the outside (e.g., using thorascopy) and the composition administered by contacting the outer surface of the affected portions of the aorta with the patch, and optionally holding the patch in place with a biological glue and/or sutures.
  • aortic valve annulus diameter a drug eluting stent graft impregnated with leptin antagonist is deployed in the aneurysm in the usual way, without oversizing.
  • Another shape of intra-vascular device that may be deployed within an aortic aneurysm is a tubular self-expandable biodegradable (or non-degradable, like bare metal) leptin antagonist slow release mesh or stent.
  • a tubular self-expandable biodegradable (or non-degradable, like bare metal) leptin antagonist slow release mesh or stent.
  • Such a device may self-expand upon deployment, and gently adhere to the luminal surface of the aortic aneurysm (applying minimal radial force to the luminal surface).
  • Leptin antagonist that is associated with, e.g., incorporated within the biodegradable or non-degradable struts of the mesh or stent or covered or coated onto the mesh or stent, will access the aortic wall at the aneurysm by local diffusion.
  • Yet another shape of a device for proximity and local slow release of leptin antagonist at the luminal surface of aortic or peripheral aneurysm will be a single wire (biodegradable or bare metal) possessing the memory of spiral expansion within the aneurysm cavity.
  • the leptin antagonist incorporated within this wire will diffuse into the arterial wall.
  • leptin antagonist from the composition passes into the tissue to provide a beneficial effect to the subject, in some embodiments one or more of attenuate aneurysm progression, stabilize the vessel wall and prevent rupture or dissection of the aneurysm.
  • the administration of the leptin antagonist also leads to reducing the rate of development, or stopping the development and in some embodiments, reversing the remodeling of parts of the heart.
  • a human subject is diagnosed with arterial stenosis that is treatable by angioplasty.
  • a drug-eluting balloon similar to the In.Pact Admiral® DCB drug-coated balloon by Medtronic (Dublin, Ireland) that is coated with a composition comprising leptin antagonist according to the teachings herein is used in the usual way to perform the angioplasty procedure, for example, at sites of arterial bifurcations and in-stent stenoses. At least some of the composition according to the teachings herein that coats the balloon is administered to the surface of the treated blood vessel, thereby administering a composition according to the teachings herein.
  • the administration of leptin antagonist is repeated periodically, e.g., with a frequency that is less than once a month, less than once every two months and even less than once every 3 months, even when there is no express need for repeated angioplasty. Subsequently, leptin antagonist from the composition passes into and/or through the lesion and/or tunica intima to provide a beneficial effect to the subject.
  • composition according to the teachings herein in the form of a stent
  • leptin antagonist from the administered composition passes into and/or through the lesion and/or tunica intima to provide a beneficial effect to the subject.
  • Myocardial infarction causes left ventricular remodeling, leading to progressive impairment of cardiac function.
  • a human subject is diagnosed with an acute myocardial infarction and is treated in the usual way, for example coronary catheterization for primary revascularization and myocardial salvage.
  • a treating health-care professional identifies that the subject has an elevated risk of developing cardiac dysfunction.
  • a composition comprising leptin antagonist is administered to the ascending aorta as described in the preceding examples using one or more of a drug-eluting balloon, a stent, a covered stent and a stent graft. Subsequently, leptin antagonist from the administered composition passes into and/or through the tunica intima of the aorta to provide a beneficial effect to the subject.
  • the beneficial effect is prophylactic, preventing development of or reducing the rate of development of an thoracic aortic aneurysm, and/or remodeling of the heart (in particular the left ventricle and associated valves) and/or a recurring infarction.
  • MI Post Myocardial Ischemia
  • a bolus of leptin antagonist in aqueous solution may be administered into the involved coronary artery.
  • the following strategy may be exercised for acute MI patients: Patients who sustain acute MI are most frequently admitted through the catheterization lab, to undergo coronary
  • PTCA Percutaneous Transarterial Coronary Angioplasty
  • a bolus of leptin antagonist in an aqueous solution is to be injected into the coronary artery through the catheter, afterwhich the catheter will be withdrawn.
  • This new strategy should achieve local distribution of leptin antagonist within the left ventricular heart muscle cells (cardiomyocytes) that were exposed to the ischemic as well as reperfusion insult. Inhibition of cardiomyocyte leptin activity is anticipated to mitigate left ventricular remodeling, and reduce the damage to left ventricular function.
  • Induction of myocardial ischemia in experimental animals may be achieved by temporary balloon inflation within the proximal left anterior descending (LAD) coronary artery.
  • Leptin antagonist aqueous solution may be injected into the LAD after balloon deflation.
  • Control group may receive intracoronary bolus of saline injection.
  • leptin antagonist may be provided into the treated coronary artery after it has been reopened and blood flow to the ischemic myocardium is restored, in order to prevent post MI left ventricular remodeling.
  • Leptin antagonist may be administered by intravascular injection into a vascular territory that sustained ischemia and reperfusion injury. This injection is a localized injection to a specific section of the left ventricle and does not constitute a systemic treatment.
  • MIH Myointimal Hyperplasia
  • MIH myointimal hyperplasia
  • medial smooth muscle cells undergo uncontrolled proliferation that may lead to stenosis or restenosis of the blood vessel in the area of the trauma.
  • trauma include vascular injury caused by expansion of a blood vessel during angioplasty, stent deployment, stent-graft deployment, as a result of surgical anastomosis and associated suturing, clamping of blood vessels, and as a result of blunt and/or penetrating vascular injury.
  • a health-care professional identifies that a subject has an elevated risk of developing myointimal hyperplasia due to some vascular trauma, administers a composition comprising leptin antagonist according to the teachings herein to the site of the trauma.
  • the administered leptin antagonist reduces the rate or stops the uncontrolled proliferation of cells, reducing the rate of development or preventing MIH.
  • Administration includes the use of any of the compositions according to the teachings herein, including localized administration of a leptin antagonist composition on the outer surface of the blood vessel at the site of injury during surgery (e.g., application of a film of Example 4 as a patch), peri-vascular injection of leptin antagonist in solution or within a slow release gel, Intravascular administration using a drug eluting balloon, or by administration of a composition that impregnates or coats a medical device, or a composition that is in the shape of a medical device by deploying the medical device.
  • localized administration of a leptin antagonist composition on the outer surface of the blood vessel at the site of injury during surgery e.g., application of a film of Example 4 as a patch
  • peri-vascular injection of leptin antagonist in solution or within a slow release gel e.g., Intravascular administration using a drug eluting balloon, or by administration of a composition that impregnates or coats a medical device, or a composition
  • specific suitable medical devices include intracavitary devices such as a stent cover, a stent, a graft assembly, a ring, a suture and a prosthetic cardiac valve as well as extraluminal devices such as sheets, all such comprising leptin antagonist according to the teachings herein,
  • a human subject is diagnosed with a peripheral or venous aneurysm, e.g., a visceral artery aneurysm, a cerebral aneurysm, especially a saccular or pseudo -fusiform aneurysm.
  • a peripheral or venous aneurysm e.g., a visceral artery aneurysm, a cerebral aneurysm, especially a saccular or pseudo -fusiform aneurysm.
  • a composition according to the teachings herein associated with a covered stent is provided, e.g., one or more of coating the stent, coating the stent cover, impregnating the stent, impregnating the stent cover, constituting the stent and constituting the stent cover.
  • the covered stent is deployed in the usual way, where the stent cover covers the mouth of the stent. Subsequently, leptin antagonist from the composition passes into and/or through the tissue in proximity to the aneurysm to provide a beneficial effect to the subject.
  • a composition according to the teachings herein is placed inside the cavity of the aneurysm through the mouth thereof, e.g., as a fluid composition (e.g., Example 6) or as an aneurysm coil that is impregnated with leptin antagonist, coated with leptin antagonist or is made of a composition according to the teachings herein.
  • a fluid composition e.g., Example 6
  • an aneurysm coil that is impregnated with leptin antagonist, coated with leptin antagonist or is made of a composition according to the teachings herein.
  • a person having ordinary skill in the art is able to implement coating an aneurysm coil with an active pharmaceutical ingredient with reference to, for example, Cerecyte® (Codman Neuro, a division of DePuySynthes, part of Johnson & Johnson, New Brunswick, NJ, USA), Nexus® (Micro Therapeutics, Inc., Irvine, CA, USA), and HydroCoil®, HydroSoft® (Terumo Corporation, Tokyo, Japan).
  • Cerecyte® Cosmetic Neuro, a division of DePuySynthes, part of Johnson & Johnson, New Brunswick, NJ, USA
  • Nexus® Micro Therapeutics, Inc., Irvine, CA, USA
  • HydroCoil®, HydroSoft® HydroSoft®
  • a human subject is diagnosed with an aneurysm, e.g., aortic aneurysms, which are typically related to a variety of diseases associated with angiotensin II hormonal activity. Also peripheral arterial aneurysms, which affect visceral, carotid, peripheral, and cerebral arteries, as well as venous aneurysms, including pulmonary artery (which carries venous blood) may be diagnosed.
  • an aneurysm e.g., aortic aneurysms, which are typically related to a variety of diseases associated with angiotensin II hormonal activity.
  • peripheral arterial aneurysms which affect visceral, carotid, peripheral, and cerebral arteries, as well as venous aneurysms, including pulmonary artery (which carries venous blood) may be diagnosed.
  • a leptin antagonist eluting stent or scaffold may be provided, e.g., intravascular stent or scaffold device (which may or may not be biodegradable) covered or coated with leptin antagonist, available for slow release into the vessel wall locally, thereby attenuating aortic aneurysm progression.
  • a stent-graft destined for treatment of aortic aneurysm may be provided, covered or coated with leptin antagonist available for slow release into the vessel wall at the specific sites of stent-graft attachment to non-dilated (normal) proximal and distal vessel (landing zones).
  • a stent-like prosthetic heart valve for intravascular application, covered with leptin antagonist may be provided, to prevent local dilation of the hosting tissue ring.
  • a human subject may be exposed to localized vascular injury which may occur as a result of vascular surgery, local balloon angioplasty. This may cause local arterial narrowing due to smooth muscle cell proliferation, namely myointimal hyperplasia (MIH).
  • MIH myointimal hyperplasia
  • This proliferative response may also occur at arterio-venous anastomosis or graft-arterial anastomosis, and in leptin-induced inflammatory and cellular proliferative arterial disease (e.g., Takayasu disease).
  • an intravascular stent or scaffold device may be provided, (which may or may not be biodegradable) which may be associated with, e.g., covered or coated with leptin antagonist that is available for slow release into the vessel wall. Also, peri-vascular injection of leptin antagonist slow release gel can attenuate the MIH response.
  • a human subject is diagnosed with atherosclerotic plaques. These lesions frequently undergo transformation from a stable plaque into an unstable rupture-prone lesion.
  • An intravascular stent or scaffold device (which may or may not be biodegradable) covered with leptin antagonist that is available for slow release into the vessel wall may be provided, in order to provide vessel stabilization. Deployment of such a stent or scaffold device may apply to any arterial site. Also, peri-vascular injection of leptin antagonist slow release gel can pacify the atherosclerotic plaque and prevent its transformation into an unstable lesion.
  • Patients who are diagnosed as hypertensive and hypercholestrolemic and exhibit initial dilation of the ascending aorta may be treated by positioning an intravascular stent or scaffold device at the ascending aorta.
  • the stent or scaffold device may or may not be biodegradable.
  • the device may be associated with, e.g., covered or coated with leptin antagonist that is available for slow release into the vessel wall. Subsequently, slowing down the progression of the aortic aneurysm by intraluminal, locally applied leptin antagonist will prevent the development of left heart failure, and remodeling of aortic and mitral valve leaflets.
  • the intravascular stent or scaffold device carrying or covered with leptin antagonist may be positioned intra-luminally at the ascending aorta.
  • Such a stent or scaffold device may attenuate local dilation and stiffening thereby prevent local hemodynamic perturbation, which activate the aorto-ventricular coupling. This coupling, when turned on promotes the production and release of angiotensin II from left ventricular cells (cardiomyocytes).
  • Angiotensin II drives intracellular synthesis of leptin in cardiomyocytes and in aortic va!ve interstitial cells (VICs).
  • Leptin synthesis in cardiomyocytes and VICs contributes to left ventricular hypertrophy, and aortic/mitral valve thickening, respectively. Therefore, deactivation of the aorto-ventricular coupling will control both left ventricular hypertrophy (heart failure) and also attenuate the progression of aortic/mitral valve disease.
  • Post-MI heart failure involves complex mechanisms.
  • One of these mechanisms is the over-expression of leptin in cardiomyocytes after ischemia and reperfusion injury (IRI).
  • Leptin activity in cardiomyocytes promotes left ventricular hypertrophy and fibrosis, both of which contribute to cardiac dysfunction.
  • Post-MI therapies in rats aimed at blocking leptin activity via systemic (Purham et al., Am. J Physiol. Heart Circ. Physio. 2008: 295. ⁇ 441 - H446) or local (Moro et al., J Cell Mol. Med. 201 1 ; 15: 1688-1694) approaches mitigated post-MI cardiac damage.
  • renin-angiotensin aldosterone system acts via angiotensin II (Angll) in IRI, contributing to tissue damage through various pathways.
  • Angll angiotensin II
  • IRI Activated renin-angiotensin aldosterone system
  • Angll angiotensin II
  • myocardium Angll induces cardiac cell hypertrophy leading to left ventricular dysfunction through the induction of leptin in cardiomyocytes.
  • Past experimental data have provided evidence that both, cardiomyocyte hypertrophy and the resulting heart failure can be mitigated by inhibiting leptin synthesis and activity.
  • the kidney which is an end organ that is solely supplied in most cases by a single artery, mechanisms related to angiotensin II cause cell injury through constriction of renal vessels, enhancement of vascular sensitivity to sympathetic nerve stimulation, increased oxidative stress and induced apoptosis.
  • ACEIs angiotensin converting enzyme inhibitors
  • angiotensin receptor blockers have protective effects on IRI in the kidney.
  • IRI myocardial ischemia and reperfusion injury
  • Leptin mRNA was increased significantly in different tissues throughout the first 48 hours, including adipose tissue and heart. Leptin receptor mRNA presented a similar pattern. Levels of leptin mRNA and leptin receptor mRNA were measured in the heart over a period of 30 days (Figs. 32). Interestingly, the heart exhibited maximal leptin mRNA upregulation 20 hours post IRI. There was also a long-term increase in leptin transcript by 3-6 fold on post operative day (POD) 30, compared to saline controls.
  • POD post operative day
  • leptin antagonist is made directly and exclusively into the infarcted myocardium.
  • a clinical scenario of major brain ischemia and reperfusion injury has been simulated in mice to examine the effects of selective intra-arterial leptin antagonist (LepA) on global brain ischemia and reperfusion injury in mice.
  • LepA selective intra-arterial leptin antagonist
  • mice Eight weeks old C57BL6 male mice underwent bilateral ligation of the common carotid arteries (CCA). After a period of 10 min ischemia, the right CCA was re-perfused. Leptin antagonist (SMLA), 2C ⁇ g dissolved in 50 ⁇ ] saline, was immediately bolus injected selectively into the right ECA, to be delivered to the brain through the right ICA. Twenty four hours later mice were euthanized, and brains were perfused with 4% paraformaldehyde. The brains were removed and additionally fixed in 10% paraformaldehyde over night at 4° C and embedded in paraffin. The brain was cut into coronal sections, and 5 ⁇ slides were stained with H&E ( Figure 34).
  • brain neurodegenerative diseases like Alzheimer's disease are associated with ischemia and inflammation affecting the brain microcirculation (Kalaria RN; Neurol Res. 2000;21 :321-330).
  • ischemia and inflammation affecting the brain microcirculation
  • df-DES selective intraarterial administration of leptin antagonist into the cerebral circulation, through bolus injection or slow release via df-DES may attenuate pathological changes in the brain and mitigate symptoms related to different neurodegenerative disorders, like Alzheimer's disease.

Abstract

Devices and methods for treating ischemia and reperfusion injury (IRI) are configured for sustained-release of anti-proliferative drug into the wall of a blood vessel (to prevent in-stent stenosis), and for sustained-release of leptin antagonist into the lumen to be carried by the blood and be uptaken by tissue cells that were subjected to IRI.

Description

DRUG ELUTING STENT
FIELD OF THE INVENTION The present invention relates to the field of medicine, and in some embodiments to devices and compositions comprising a leptin antagonist formulated for localized release of a leptin antagonist at the site of treatment, specifically the treatment of ischemia and reperfusion injury. BACKGROUND OF THE INVENTION
Cardiovascular disease (CVD) is a class of diseases that involve the heart and/or the blood vessels. Several studies have related inflammatory markers to cardiovascular disease (CVD) and several assays for inflammatory markers are commercially available. For example, C- reactive protein (CRP), a common inflammatory marker, has been found to be present in increased levels in patients who are at risk for cardiovascular disease [Karakas and Koenig, 2009 Herz 34 (8): 607-13] while osteoprotegerin, which is involved with regulation of NF- KB, has been found to be a risk factor for cardiovascular disease and mortality [Venuraju et al., 2010 J. Am. Coll. Cardiol. 55 (19): 2049-61],
As a result of these findings, the number of inflammatory marker tests ordered by clinicians for CVD risk prediction has grown rapidly. However, to date there is no consensus among professionals as to how these markers of inflammation should be used as a basis for clinical treatment.
Although it has been shown that some cardiovascular disorders can benefit from suppression of inflammation-related processes and cellular proliferation as part of a remodeling response (e.g. use of locally released cytotoxic drugs such as paclitaxel or sirolimus in preventing restenosis or use of doxycyciine in treatment of abdominal aortic aneurysm (AAA)), to date, there is no evidence to suggest that cardiovascular disease can benefit from anti-inflammatory treatment.
Cardiovascular diseases are very often associated with ischemia/reperfusion injury (IRI). IRI is caused by critical reduction of blood supply to an organ followed by renewal of blood flow and re-ox ygenati on. Tissue damage is driven by activation of inflammatory processes, including synthesis of cytokines, chemokines and reactive oxygen species (ROS). Chemokines mediate inflammation and regulate pro- inflammatory cytokine, adhesion molecule expression, leukocyte infiltration and activation. During IRI, excessive production of ROS cause oxidative stress, which impacts mitochondrial oxidative phosphorylation, causing ATP depletion, increase intracellular calcium and activation of membrane phospholipids proteases. Reperfusion and oxygenation produce oxygen free radicals, which promote lipid peroxidation and tissue damage by free radicals. Notably, the mechanisms involved in IRI tissue damage as mentioned above apply to all tissues. However, cellular damage correlates with the magnitude of ischemia, and cell type. For instance, kidney and brain cells are extremely sensitive to ischemic damage, which is further exacerbated by reperfusion injury.
Leptin is a naturally occurring pleiotropic molecule that regulates food intake as well as metabolic and endocrine functions. Leptin also plays a regulatory role in immunity, inflammation, and hematopoiesis.
The human leptin precursor is a linear polypeptide 167 amino acid residues long represented by NCBI Reference Sequence NP 000221.1 (SEQ ID NO 1) encoded by the mRNA having the nucleotide sequence NCBI Reference Sequence NM_000230. Residues 1-
21 of the sequence constitute the signal peptide while residues 22-167 constitute the mature hormone.
Leptin antagonists are also known, see for example, US 7,307,142 and US 8,969,292.
SUMMARY OF THE INVENTION
The invention, in some embodiments, relates to the field of medicine, and more particularly to methods and devices that use leptin antagonists. In some embodiments, the invention relates to compositions comprising a leptin antagonist formulated for localized release of a leptin antagonist to inhibit activity of leptin at the site of treatment as well as methods of using such compositions for treating disorders, including cardiovascular disorders.
According to an aspect of some embodiments of the invention, there is provided a method of treatment comprising: exposing in vivo tissue of a subject in need thereof to local administration of a pharmaceutically-effective amount of leptin antagonist, thereby providing a therapeutic effect to the tissue. In some embodiments, the tissue is substantially continuously exposed to a pharmaceutically-effective amount of leptin antagonist for a period of not less than three days, not less than 5 days, not less than 8 days and even not less than 14 days. According to an aspect of some embodiments of the invention, there is also provided a method of treatment comprising implanting in contact with tissue in the body of a subject in need thereof a composition configured for in vivo local administration of leptin antagonist, thereby providing a therapeutic effect to the tissue. In some embodiments, the configuration for the in vivo release is such that when the composition is implanted in vivo, leptin antagonist is released from the composition in a pharmaceutically-effective amount for a period of not less than three days, not less than 5 days, not less than 8 days and even not less than 14 days.
In some embodiments of the methods, the need is that the subject suffers from at least one pathology selected from the group consisting of: cardiovascular disease; remodeling of stable athersclerotic plaque into an unstable lesion; ascending aortic aneurysm-associated hypertension, hypercholesterolemia or diabetes mellitus; bicuspid aortic valve; Takayasu disease; rheumatoid arteritis; Marian's syndrome; ankylosing spondylitis; giant cell arteritis; inflammatory aortic aneurysm; pulmonary artery aneurysm in Marian's syndrome; aortic dissection in an aortic or peripheral large artery; angiogenesis; cancer; local discrete lesion therapy; and arteriovenous malformation.
In some embodiments, the need is that the subject suffers from a cardiovascular disorder, wherein the therapeutic effect is down-regulation of an expression or activity of leptin in a cardiovascular tissue.
In some embodiments, the cardiovascular tissue is aortic and/or mitral heart valve leaflet tissue. In some embodiments, the local administration is effected by positioning a carrier capable of releasing the leptin antagonist on an outer wall (e.g., tunica externa) or the inner wall (e.g., tunica intima) of an aorta.
In some embodiments, the cardiovascular tissue is arterial or venous wall tissue. In some embodiments, the local administration is effected by positioning a carrier capable of releasing the leptin antagonist on an outer wall (e.g., tunica externa) or the inner wall (e.g., tunica intima) of the arterial or venous wall tissue.
In some embodiments, the cardiovascular disorder is a vascular aneurysm. In some embodiments, the cardiovascular disorder is an aortic vascular disorder. In some embodiments, the cardiovascular disorder is left ventricular remodeling.
In some embodiments, the local administration is effected via an intravascular catheter. In some embodiments, the local administration is effected via direct injection.
According to an aspect of some embodiments of the invention, there is also provided a method for treatment of athersclerotic plaque, comprising: administering a pharmaceutical ly-effective amount of a leptin antagonist to atherscl erotic plaque accumulated in the inner walls of an artery, thereby at least one of: (a) reducing the rate and (b) reducing the incidence, of conversion of a stable athersclerotic plaque to an unstable lesion.
According to an aspect of some embodiments of the invention, there is also provided a composition comprising a leptin antagonist and a carrier, for use in treating a disorder selected from the group consisting of: cardiovascular disease; remodeling of stable athersclerotic plaque into an unstable lesion; ascending aortic aneurysm-associated hypertension, hypercholesterolemia or diabetes mellitus; bicuspid aortic valve; Takayasu disease; rheumatoid arteritis; Marfan's syndrome; ankylosing spondylitis; giant cell arteritis; inflammatory aortic aneurysm; pulmonary artery aneurysm in Marfan's syndrome; aortic dissection in an aortic or peripheral large artery; site of arterial anastomosis, angiogenesis; cancer; local neoplastic discrete lesion therapy; and arteriovenous malformation, wherein the carrier is configured for localized administration of the leptin antagonist.
In some embodiments, the carrier is a biodegradable support. In some embodiments, the biodegradable support is composed of a polymer selected from the group consisting of a hydrogel, poly gly colic acid (PGA), poly lactic co-glycolic acid (PLGA), polylactide (PL A), and poly (L-lactide) (PLLA), and combinations thereof.
In some embodiments, the carrier is a hydrogel. In some embodiments, the carrier is configured as a film. In some embodiments, the carrier is a device selected from the group consisting of a mesh, a balloon and a vascular graft. In some embodiments, the carrier is a depot-forming injectable composition.
In some embodiments, the disorder is a cardiovascular disorder, wherein the leptin antagonist effects down-regulation of an expression or activity of leptin in a cardiovascular tissue. In some embodiments, the cardiovascular disorder is a vascular disorder. In some such embodiments, the vascular disorder is an aortic vascular disorder. In some embodiments, the cardiovascular disorder is left ventricular remodeling.
In some such embodiments, the cardiovascular tissue is aortic and/or mitral heart valve leaflet tissue. In some such embodiments, the cardiovascular tissue is arterial or venous wall tissue.
In some embodiments, the local administration is effected by positioning a carrier capable of releasing the leptin antagonist on a location selected from the group consisting of: an outer wall of an aorta, an outer wall of an artery, an outer wall of an vein, a luminal surface of an aorta, a luminal surface of an artery and a luminal surface of an vein. In some such embodiments, the local administration is effected by positioning a carrier capable of releasing the leptin antagonist on an outer wall (e.g., tunica externa) or the inner wall (e.g., tunica intima) of the arterial or venous wall tissue. In some embodiments, the localized administration is to be effected via an intravascular catheter. In some embodiments, the localized administration is to be effected via direct injection.
According to an aspect of some embodiments of the invention, there is also provided a method of treating a condition in a subject in need thereof, the method comprising administering intracavitarily to inner walls of a fluid-filled bodily cavity of the subject a composition comprising a leptin antagonist.
According to an aspect of some embodiments of the invention, there is also provided a composition comprising: a leptin antagonist for use in treating a condition, wherein the composition is configured for intracavitary administration to inner walls of a fluid-filled bodily cavity of a subject.
According to an aspect of some embodiments of the invention, there is also provided an intracavitarily-implantable medical device, comprising: at least one solid functional device part configured for deploying the device in a fluid-filled bodily cavity of a subject; and functionally associated with at least one the device component, a leptin antagonist.
According to an aspect of some embodiments of the invention, there is also provided a surgical connecting device, comprising: a solid device body made of a material; and functionally associated with the device body, a pharmaceutically-effective amount of leptin antagonist. In some embodiments, the device body is in the form selected from the group consisting of surgical suture thread and a surgical staple.
According to an aspect of the invention, there is provided a method for treating ischemia and reperfusion injury (IRI) by administrating leptin antagonist upon reperfusion and maintaining sustained administration of the leptin antagonist into the vessel undergoing the reperfusion procedure, for delivering the leptin antagonist by the blood flow into the damaged cells of the tissue injured by the IRI via bolus intra-arterial injection, or sustained release from a drug eluting stent.
According to an aspect of some embodiments of the invention, there is provided a double function drug eluting stent (df-DES) configured to enable sustained release of anti- proliferative drug into the wall of a blood vessel and to enable sustained release of leptin antagonist (LA) into the lumen, so that the leptin antagonist is carried by the blood stream to be uptaken by tissue cells that sustained ischemia and reperfusion injury.
Any suitable leptin antagonist may be used for implementing the teachings herein. Various types and specific suitable leptin antagonists are listed in the description herein. In some embodiments, the leptin antagonist is capable of binding a leptin receptor. In some embodiments, the leptin antagonist is incapable of dimerization. In some embodiments, the leptin antagonist comprises a polypeptide portion. In some embdiments, the leptin antagonist is selected from the group consisting of a polypeptide, a salt, and/or an ester thereof. In some embodiments, the leptin antagonist is a modified leptin polypeptide.
BRIEF DESCRIPTION OF THE FIGURES
Some embodiments of the invention are described herein with reference to the accompanying figures. The description, together with the figures, makes apparent to a person having ordinary skill in the art how some embodiments of the invention may be practiced. The figures are for the purpose of illustrative discussion and no attempt is made to show structural details of an embodiment in more detail than is necessary for a fundamental understanding of the invention. For the sake of clarity, some objects depicted in the figures are not to scale.
In the Figures:
FIG. la-c illustrate a gel (Figure la), film (Figure lb) and mesh (Figure l c) for local release of a leptin antagonist.
FIG. 2 illustrates a balloon catheter configured for local release of a leptin antagonist(drug release indicated by arrows).
FIG. 3 illustrates a slow release leptin eluted from a scaffold.
FIG, 4 illustrates the location of leptin film application on the anterior outer surface of the ascending aorta. Human arch angiogram depicts mouse anatomy.
FIG. 5 illustrates a time course analysis of serum leptin level in ApoE"7" mice that underwent peri-aortic application of leptin film (20 μg).
FIG. 6 illustrates increased ascending aortic diameter at the location of leptin film application versus controls.
FIG. 7 illustrates elastica staining and aSMA IHC analysis of ascending aortic cross sections of mice locally treated with leptin versus controls.
FIG. 8 illustrates change in left ventricle (LV) wall thickness in leptin-treated (filled columns) versus control (open columns) mice.
FIG. 9 illustrates LV diameter in systole and diastole in leptin-treated (filled columns) and control (open columns) mice.
FIG. 10 illustrates LV fractional area change in leptin-treated (filled columns) versus control (open columns) mice. FIG. 1 1 illustrates aortic and mitral valve leaflet thickness in leptin-treated and control mice.
FIG. 12 illustrates mean systolic blood pressure in angiotensin II treated mice.
FIG. 13 illustrates a time course analysis presenting weight of angiotensin II treated mice (open triangles), and mice receiving both, angiotensin II and leptin antagonist (LA).
FIG. 14 illustrates number of mice that succumbed due to ruptured abdominal and thoracic aneurysms in angiotensin II treated mice versus mice receiving angiotensin II and leptin antagonist (LA).
FIG. 15 illustrates ascending aortic dilatation in angiotensin II treated mice versus mice receiving angiotensin II and leptin antagonist (LA).
FIG. 16 illustrates elastic lamella fragmentation and aSMA depletion in angiotensin II treated mice versus mice receiving angiotensin II and leptin antagonist (LA).
FIG. 17 illustrates leptin expression in medial SMCs (arrows) and macrophages of atherosclerotic lesions (filled arrowheads) in angiotensin II treated, angiotensin II and leptin antagonist (LA) treated, and control mice.
FIG. 18 illustrates LV hypertrophy in angiotensin II treated mice versus mice receiving angiotensin II and leptin antagonist (LA).
FIG. 19 illustrates changes in LV diameter in angiotensin II treated mice versus mice receiving angiotensin II and leptin antagonist (LA).
FIG. 20 illustrates LV fractional area change in angiotensin II treated mice (open column) versus mice receiving angiotensin II and leptin antagonist (LA) (filled column).
FIG. 21 illustrates peak systolic velocity at the aortic valve in angiotensin II treated mice versus mice receiving angiotensin II and leptin antagonist (LA).
FIG. 22 illustrates aortic and mitral valve thickness (graph on left), and staining of valve leaflets with H&E (panels F-I). aSMA and TGFP (panels J-M', staining for aortic valves) in mice receiving angiotensin II versus mice treated with angiotensin ΪΙ and leptin antagonist (LA).
FIG. 23 illustrates expression of leptin (D), and leptin receptor (E) in normal human aortic valve leaflet tissue.
FIG. 24 illustrates leptin and leptin receptor antigen prevalent in severe aortic valve stenosis, evident in SMC-iike cells, and infiltrating macrophages.
FIG. 25 illustrates leptin and leptin receptor mRNA !evels in leaflets of stenosed aortic valve versus normal aortic valve controls, and fat tissue (as positive control); FIG. 26 illustrates proliferation of valve interstitial cells (VICs) in response to leptin stimulation; and
FIG. 27 schematically depicts embodiments of surgical connecting devices according to the teachings herein;
FIG. 28 schematically depicts embodiments of the teachings herein suitable for intracavitary administration of leptin antagonist;
FIG. 29 schematically depicts further embodiments of the teachings herein suitable for intracavitary administration of leptin antagonist;
FIG. 30 schematically depicts an embodiment of a double-function drug eluting stent in accordance with the teachings herein;
FIG. 31a is partial cross sectional perspective view of a prior art stent;
FIGS. 31b and 31c are partial cross sectional perspective views of further embodiments of a double-function drug eluting stent in accordance with the teachings herein;
FIGS. 32a and 32b illustrate a time course of leptin mRNA expression and leptin receptor mRNA expression, respectively, in the heart of mice that underwent myocardial IRI and systemic administration of leptin antagonist to stimulate endogenous leptin synthesis, or saline, upon reperfusion;
FIG. 33 illustrates percent of ejection fraction (EF%) of mice at 1 and 30 days after undergoing IRI and systemic administration of leptin antagonist or saline upon reperfusion;;
FIGS. 34 (a-c) illustrate coronal sections of a mouse brain that underwent IRI (a-c);
FIGS. 35 (a-c) illustrate coronal sections of a mouse brain that underwent IRI plus selective administration of leptin antagonist upon reperfusion; and
FIGS. 36 (a-c) illustrate coronal sections of a normal mouse brain.
DESCRIPTION OF SOME EMBODIMENTS
The invention, in some embodiments, relates to the field of medicine, and more particularly to methods and devices that use leptin antagonists. In some embodiments, the invention relates to compositions comprising a leptin antagonist formulated for localized release of a leptin antagonist at the site of treatment as well as methods of using such compositions for treating disorders, including cardiovascular disorders. In some embodiments, the compositions comprising a leptin antagonist can be used for localized suppression of leptin-related conditions, including tissue remodeling processes.
Although it has been proposed that leptin might play a role in vascular inflammation, oxidative stress, and vascular smooth muscle hypertrophy that may contribute to coronary heart disease among other pathologies, to date no one has conclusively shown that localized down-regulation of leptin activity can be used to treat cardiovascular disorders characterized by remodeling of cardiovascular tissue such as cardiac, arterial or valve tissue.
The present inventor set out to elucidate the role of leptin in disorders, such as cardiovascular disorders by employing a leptin antagonist in a localized manner. Experiments conducted by the present inventor (see Examples section hereinbelow) demonstrate that localized release of leptin in cardiovascular tissue can lead to cardiovascular tissue remodeling while localized down-regulation of leptin activity can lead to suppression and even reversal of cardiovascular tissue (arterial wall tissue, heart muscle tissue and valve leaflet tissue) remodeling induced by angiotensin II. Thus, the present inventor has shown for the first time that a locally administered leptin antagonist can be used to treat cardiovascular disorders characterized by tissue remodeling.
While reducing the present invention to practice, the present inventor has shown that down-regulation of leptin activity at specific sites in the cardiovascular system can lead to suppression and reversal of pathological tissue remodeling and thereby establishing localized leptin down-regulation as a suitable approach for treating various cardiovascular disorders, such as cardiovascular disorders characterized by pathological tissue remodeling.
The present inventor has discovered an unexpected pharmaceutical efficacy of locally administered leptin antagonists, especially leptin antagonists administered by sustained release.
Particularly, the present inventor has found that in vivo implantation of a composition configured for sustained-release of leptin antagonist can have a desirable pharmaceutical effect on tissue in proximity of the implanted composition with limited or no substantial side- effects, for example, no discernible hormonal or immunological effects. The present inventor has also found that such in vivo administration (e.g., by implantation) inside a fluid-filled cavity of the body (for example of the cardiovascular system such as blood vessels or cardiac chambers) can have a desirable pharmaceutical effect on tissue in proximity of the implanted composition with limited or no substantial side- effects: leptin antagonist from the composition has not been found to be washed away by the fluid and, instead, has been found to interact with the tissue providing a desirable pharmaceutical effect. Presumably, leptin antagonist released from the composition passes into and through the cavity walls (e.g., tunica intima) in pharmaceutically-effective amounts. This is particularly surprising in cavities of the cardiovascular system (e.g., veins, arteries and cardiac chambers) where the large volumes of blood passing through such cavities are expected to wash away released leptin antagonist and where it is expected that the cardiovascular intima is relatively non-permeable to passage of compounds, especially proteins.
Without wishing to be held to any one theory, it is currently believed that the success of some embodiments of the teachings herein is at least partially attributable to the serendipitous increased permeability of cardiovascular intima during inflammation. It may be that the intima of healthy cardiovascular intima is relatively impermeable to leptin antagonist released from the composition, so that there is little or no passage of leptin antagonist into and through the endothelium and underlying tissue, thereby avoiding substantial negative side-effects. In contrast, it seems that the permeability of the cardiovascular intima during inflammation increases sufficiently to allow passage of a therapeutically-effective amount of leptin antagonist released from the composition into the tissue. It is currently believed that this effect is self-regulating. A higher degree of inflammation leads to a higher degree of intima permeability allowing passage of more leptin antagonist leading to a relatively high dose of leptin antagonist in the more pathological tissue. As inflammation decreases (inter alia, due to the pharmaceutical effect of the administered leptin antagonist), intima permeability decreases thereby decreasing the dose of leptin antagonist actually in the tissue that is still sufficient to exercise a desired pharmaceutical effect but with a reduced incidence of substantial negative side effects.
In some embodiments, the present invention includes local administration of leptin antagonist to treat and attenuate expansion of ascending aortic aneurysm, and corresponding cardiac sequelae (driven by the aorto-ventricular coupling), including left ventricular hypertrophy, as well as hyperplasia of left heart valve leaflets. In some embodiments, the present invention includes treatment of peripheral vascular disorders such as the progression of arterial or venous aneurysms while minimizing systemic exposure to the administered leptin antagonist. The principles and operation of the present invention may be better understood with reference to the drawings and accompanying descriptions.
Before explaining at least one embodiment of the invention in detail, it is to be understood that the invention is not limited in its application to the details set forth in the following description or exemplified by the Examples. The invention is capable of other embodiments or of being practiced or carried out in various ways. Also, it is to be understood that the phraseology and terminology employed herein is for the purpose of description and should not be regarded as limiting.
Thus, according to one aspect of the present invention there is provided a composition comprising a leptin antagonist and a carrier configured for localized administration, for treating disorders such as cardiovascular disorders. As used herein, "cardiovascular disorders" refer to disorders of the cardiovascular system, i.e. the heart and central, cranial and peripheral vasculature. Examples of such disorders include, but are not limited to valve stenosis, aneurysms, vessel response to vascular injury, cardiomyopathy and the like.
The carrier can be a solid, gel or liquid carrier, while the leptin antagonist can be any agent capable of down-regulating leptin activity in the target tissue. Examples of a leptin antagonist include agents capable of binding and/or degrading leptin or leptin receptors as well as agents capable of down-regulating leptin expression (at the DNA or RNA levels, i.e., agents capable of blocking transcription or translation). For example, the leptin antagonist known as SMLA (Shpilman et al., J Biol Chem. 20 Π; 286:4429-4442) has a 60 fold higher affinity to the leptin receptor compared to the endogenous leptin. Therefore, it binds to the leptin receptors in the treated tissue/organ, leaving no leptin receptors available to complex with the endogenous leptin. The complex leptin receptor-leptin antagonist is inactive and can't exert any damage to the tissue, locally. Specific preferred leptin antagonists are listed hereinbelow. One example of a leptin antagonist that is an agent capable of down-regulating leptin is an antibody or antibody fragment capable of specifically binding leptin or a leptin receptor. Preferably, the antibody specifically binds at least one epitope of leptin, e.g., an epitope defined amino acids 26-59 of mammalian leptin (e.g. rat leptin - SEQ ID NO 48). As used herein, the term "epitope" refers to any antigenic determinant on an antigen to which the paratope of an antibody binds.
As used herein, the term "antibody" refers to a substantially intact antibody molecule. As used herein, the phrase "antibody fragment" refers to a functional fragment of an antibody that is capable of binding to an antigen.
Suitable antibody fragments for practicing the present invention include, inter alia, a complementarity-determining region (CDR) of an immunoglobulin light chain (referred to herein as "light chain"), a CDR of an immunoglobulin heavy chain (referred to herein as "heavy chain"), a variable region of a light chain, a variable region of a heavy chain, a light chain, a heavy chain, an Fd fragment, and antibody fragments comprising essentially whole variable regions of both light and heavy chains such as an Fv, a single-chain Fv, an Fab, an Fab', and an F(ab')2.
Functional antibody fragments comprising whole or essentially whole variable regions of both light and heavy chains are defined as follows:
(i) Fv, defined as a genetically engineered fragment consisting of the variable region of the light chain and the variable region of the heavy chain expressed as two chains;
(ii) single-chain Fv ("scFv"), a genetically engineered single-chain molecule including the variable region of the light chain and the variable region of the heavy chain, linked by a suitable polypeptide linker.
(iii) Fab, a fragment of an antibody molecule containing a monovalent antigen- binding portion of an antibody molecule, obtained by treating whole antibody with the enzyme papain to yield the intact light chain and the Fd fragment of the heavy chain, which consists of the variable and CHI domains thereof;
(iv) Fab', a fragment of an antibody molecule containing a monovalent antigen- binding portion of an antibody molecule, obtained by treating whole antibody with the enzyme pepsin, followed by reduction (two Fab' fragments are obtained per antibody molecule); and
(v) F(ab')2, a fragment of an antibody molecule containing a monovalent antigen- binding portion of an antibody molecule, obtained by treating whole antibody with the enzyme pepsin (i.e., a dimer of Fab' fragments held together by two disulfide bonds).
Methods of generating monoclonal and polyclonal antibodies are well known in the art. Antibodies may be generated via any one of several known methods, which may employ induction of in vivo production of antibody molecules, screening of immunoglobulin libraries (Orlandi, R. et al. (1989). Cloning immunoglobulin variable domains for expression by the polymerase chain reaction. Proc Natl Acad Sci USA 86, 3833-3837; and Winter, G. and Milstein, C. (1991). Man-made antibodies. Nature 349, 293-299), or generation of monoclonal antibody molecules by continuous cell lines in culture. These include, but are not limited to, the hybridoma technique, the human B-cell hybridoma technique, and the Epstein- Barr virus (EBV)-hybridoma technique (Kohler, G. and Milstein, C. (1975). Continuous cultures of fused cells secreting antibody of predefined specificity. Nature 256, 495-497; Kozbor, D. et al. (1985). Specific immunoglobulin production and enhanced tumorigenicity following ascites growth of human hybridomas. J Immunol Methods 81, 31 -42; Cote RJ. et al. (1983). Generation of human monoclonal antibodies reactive with cellular antigens. Proc Natl Acad Sci USA 80, 2026-2030; and Cole, S. P. et al. (1984). Human monoclonal antibodies. Mol Cell Biol 62, 109-120).
In cases where target antigens are too small to elicit an adequate immunogenic response, such antigens (referred to as "haptens") can be coupled to antigenically neutral carriers such as keyhole limpet hemocyanin (KLH) or serum albumin (e.g., bovine serum albumin (BSA)) carriers (see, for example, US. Pat. Nos. 5,189,178 and 5,239,078). Coupling a hapten to a carrier can be effected using methods well known in the art. For example, direct coupling to amino groups can be effected and optionally followed by reduction of the imino linkage formed. Alternatively, the carrier can be coupled using condensing agents such as dicyclohexyl carbodiimide or other carbodiimide dehydrating agents. Linker compounds can also be used to effect the coupling; both homobifunctional and heterobifunctional linkers are available from Pierce Chemical Company, Rockford, Illinois, USA. The resulting immunogenic complex can then be injected into suitable mammalian subjects such as mice, rabbits, and others. Suitable protocols involve repeated injection of the immunogen in the presence of adjuvants according to a schedule designed to boost production of antibodies in the serum. The titers of the immune serum can readily be measured using immunoassay procedures which are well known in the art.
The antisera obtained can be used directly or monoclonal antibodies may be obtained, as described hereinabove.
Antibody fragments may be obtained using methods well known in the art. (See, for example, Harlow, E. and Lane, D. (1988). Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, New York.) For example, antibody fragments according to the present invention can be prepared by proteolytic hydrolysis of the antibody or by expression in E. coli or mammalian cells (e.g., Chinese hamster ovary (CHO) cell culture or other protein expression systems) of DNA encoding the fragment.
Alternatively, antibody fragments can be obtained by pepsin or papain digestion of whole antibodies by conventional methods. As described hereinabove, an (Fab' )2 antibody fragments can be produced by enzymatic cleavage of antibodies with pepsin to provide a 5S fragment. This fragment can be further cleaved using a thiol reducing agent, and optionally a blocking group for the sulfhydryl groups resulting from cleavage of disulfide linkages, to produce 3.5S Fab' monovalent fragments. Alternatively, enzymatic cleavage using pepsin produces two monovalent Fab' fragments and an Fc fragment directly. Ample guidance for practicing such methods is provided in the literature of the art (for example, refer to: U.S. Pat. Nos. 4,036,945 and 4,331,647; and Porter, R. R. (1959). The hydrolysis of rabbit γ-globulin and antibodies with crystalline papain. Biochem J 73, 1 19-126). Other methods of cleaving antibodies, such as separation of heavy chains to form monovalent light-heavy chain fragments, further cleavage of fragments, or other enzymatic, chemical, or genetic techniques may also be used, so long as the fragments retain the ability to bind to the antigen that is recognized by the intact antibody.
As described hereinabove, an Fv is composed of paired heavy chain variable and light chain variable domains. This association may be noncovalent (see, for example, Inbar, D. et al. (1972). Localization of antibody-combining sites within the variable portions of heavy and light chains. Proc Natl Acad Sci USA 69, 2659-2662). Alternatively, as described hereinabove, the variable domains may be linked to generate a single-chain Fv by an intermolecular disulfide bond, or alternately such chains may be cross-linked by chemicals such as glutaraldehyde.
Preferably, the Fv is a single-chain Fv. Single-chain Fvs are prepared by constructing a structural gene comprising DNA sequences encoding the heavy chain variable and light chain variable domains connected by an oligonucleotide encoding a peptide linker. The structural gene is inserted into an expression vector, which is subsequently introduced into a host cell such as E. coli. The recombinant host cells synthesize a single polypeptide chain with a linker peptide bridging the two variable domains. Ample guidance for producing single-chain Fvs is provided in the literature of the art (see, e.g.: Whitlow, M. and Filpula, D. (1991 ). Single-chain Fv proteins and their fusion proteins. METHODS: A Companion to Methods in Enzymology 2(2), 97-105; Bird, R. E. et al. (1988). Single-chain antigen-binding proteins. Science 242, 423-426; Pack, P. et al. (1993). Improved bivalent miniantibodies, with identical avidity as whole antibodies, produced by high cell density feniientation of Escherichia coli. Biotechnology (N.Y.) 11(11 ), 1271-1277; and U.S. Pat. No. 4,946,778).
Isolated complementarity-determining region peptides can be obtained by constructing genes encoding the CDR of an antibody of interest. Such genes may be prepared, for example, by RT-PCR of the mRNA of an antibody-producing cell. Ample guidance for practicing such methods is provided in the literature of the art (e.g., Larrick, J. W. and Fry, K. E. (1991). PCR Amplification of Antibody Genes. METHODS: A Companion to Methods in Enzymology 2(2), 106-110).
It will be appreciated that for human therapy, humanized antibodies are preferred. Humanized forms of non-human (e.g., murine) antibodies are genetically engineered chimeric antibodies or antibody fragments having (preferably minimal) portions derived from non-human antibodies. Humanized antibodies include antibodies in which the CDRs of a human antibody (recipient antibody) are replaced by residues from a CDR of a non-human species (donor antibody), such as mouse, rat, or rabbit, having the desired functionality. In some instances, the Fv framework residues of the human antibody are replaced by corresponding non-human residues. Humanized antibodies may also comprise residues found neither in the recipient antibody nor in the imported CDR or framework sequences. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDRs correspond to those of a non-human antibody and all or substantially all of the framework regions correspond to those of a relevant human consensus sequence. Humanized antibodies optimally also include at least a portion of an antibody constant region, such as an Fc region, typically derived from a human antibody (see, for example: Jones, P. T. et al. (1986). Replacing the complementarity- determining regions in a human antibody with those from a mouse. Nature 321 , 522-525; Riechmann, L. et al. (1988). Reshaping human antibodies for therapy. Nature 332, 323-327; Presta, L. G. (1992b). Curr Opin Struct Biol 2, 593-596; and Presta, L. G. (1992a). Antibody engineering. Curr Opin Biotechnol 3(4), 394-398).
Methods for humanizing non-human antibodies are well known in the art. Generally, a humanized antibody has one or more amino acid residues introduced into it from a source which is non-human. These non-human amino acid residues are often referred to as imported residues, which are typically taken from an imported variable domain. Humanization can be performed essentially as described (see, for example: Jones et al. (1986); Riechmann et al. (1988); Verhoeyen, M. et al. (1988). Reshaping human antibodies: grafting an antilysozyme activity. Science 239, 1534-1536; and U.S. Pat. No. 4,816,567), by substituting human CDRs with corresponding rodent CDRs. Accordingly, humanized antibodies are chimeric antibodies, wherein substantially less than an intact human variable domain has been substituted by the corresponding sequence from a non-human species. In practice, humanized antibodies may be typically human antibodies in which some CDR residues and possibly some framework residues are substituted by residues from analogous sites in rodent antibodies.
Human antibodies can also be produced using various additional techniques known in the art, including phage-display libraries (Hoogenboom, H. R. and Winter, G. (1991). Bypassing immunization. Human antibodies from synthetic repertoires of germline VH gene segments rearranged in vitro. J Mol Biol 227, 381-388; Marks, J. D. et al. (1991). By-passing immunization. Human antibodies from V-gene libraries displayed on phage. J Mol Biol 222, 581-597; Cole et al. (1985), Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96; and Boerner, P. et al. (1991). Production of antigen-specific human monoclonal antibodies from in vitro-primed human splenocytes. J Immunol 147, 86-95). Humanized antibodies can also be created by introducing sequences encoding human immunoglobulin loci into transgenic animals, e.g., into mice in which the endogenous immunoglobulin genes have been partially or completely inactivated. Upon antigenic challenge, human antibody production is observed in such animals which closely resembles that seen in humans in all respects, including gene rearrangement, chain assembly, and antibody repertoire. Ample guidance for practicing such an approach is provided in the literature of the art (for example, refer to: U.S. Pat. Nos. 5,545,807; 5,545,806; 5,569,825; 5,625,126; 5,633,425; and 5,661 ,016; Marks, J. D. et al. (1992). By-passing immunization: building high affinity human antibodies by chain shuffling. Biotechnology (N.Y.) 10(7), 779-783; Lonberg et al., 1994. Nature 368:856-859; Morrison, S. L. (1994). News and View: Success in Specification. Nature 368, 812-813; Fishwild, D. M. et al. (1996). High-avidity human IgG kappa monoclonal antibodies from a novel strain of minilocus transgenic mice. Nat Biotechnol 14, 845-851 ; Neuberger, M. (1996). Generating high-avidity human Mabs in mice. Nat Biotechnol 14, 826; and Lonberg, N. and Huszar, D. (1995). Human antibodies from transgenic mice. Int Rev Immunol 13, 65-93).
After antibodies have been obtained, they may be tested for activity, for example via enzyme-linked immunosorbent assay (ELISA).
Anti-leptin antibodies as well as epitope sequences suitable for generating antibodies and antibody fragments are described in US20070104708 (SEQ ID NOs 49-55) which is incorporated herein by reference as if fully set-forth herein. Leptin peptide antagonists can also be used with the present invention. One leptin antagonist, a modified mammalian leptin polypeptide termed superactive leptin mutein is disclosed in US patent 8,969,292 which is incorporated by reference as if fully set-forth herein.
The term "peptide" as used herein encompass native peptides (either degradation products, synthetically synthesized peptides, or recombinant peptides), peptidomimetics (typically, synthetically synthesized peptides), and the peptide analogues peptoids and semipeptoids, and may have, for example, modifications rendering the peptides more stable while in a body or more capable of penetrating into cells. Such modifications include, but are not limited to: N-terminus modifications; C-terminus modifications; peptide bond
Figure imgf000018_0001
modifications. Methods for preparing peptidomimetic compounds are well known in the art and are specified, for example, in Ramsden, C. A., ed. (1992), Quantitative Drug Design, Chapter 17.2, F. Choplin Pergamon Press, which is incorporated by reference as if fully set forth herein. Further details in this respect are provided hereinbelow.
Figure imgf000018_0002
presented on the carbon atom. These modifications can occur at any of the bonds along the peptide chain and even at several (2-3) at the same time.
Natural aromatic amino acids, Trp, Tyr, and Phe, may be substituted for synthetic non-natural acids such as, for instance, tetrahydroisoquinoline-3-carboxylic acid (TIC), naphthylelanine (Nol), ring-methylated derivatives of Phe, halogenated derivatives of Phe, and o-methyl-Tyr.
In addition to the above, the peptides of the present invention may also include one or more modified amino acids or one or more non-amino acid monomers (e.g., fatty acids, complex carbohydrates, etc.).
The term "amino acid" or "amino acids" is understood to include the 20 naturally occurring amino acids; those amino acids often modified post-translationally in vivo, including, for example, hydroxyproline, phosphoserine, and phosphothreonine; and other less common amino acids, including but not limited to 2-aminoadipic acid, hydroxylysine, isodesmosine, nor-valine, nor-leucine, and ornithine. Furthermore, the term "amino acid" includes both D- and L-amino acids.
Amino acids are referred to by the standard three letter code. Amino acids are L amino acids unless otherwise noted, for example, by addition of the prefix "D". For example, the code Trp refers to L-tryptophan, while the codes D-Trp and DTrp refers to D-tryptophan. The code Aib refers to 2-aminoisobutyric acid. The code Orn refers to ornithine. The code Lys-Ac refers to acetyllysine. The code HomoLys refers to homolysine. The code H-Cys refers to homocysteine.
In some embodiments, peptidic leptin antagonists used to implement the teachings herein are utilized in a linear form, although in some embodiments, cyclic forms thereof are used.
In some embodiments, peptidic leptin antagonists used to implement the teachings herein are synthesized by any techniques that are known to those skilled in the art of peptide synthesis. For solid phase peptide synthesis, a summary of the many techniques may be found in: Stewart, J. M. and Young, J. D. (1963), "Solid Phase Peptide Synthesis," W. H. Freeman Co. (San Francisco); and Meienhofer, J (1973). "Hormonal Proteins and Peptides," vol. 2, p. 46, Academic Press (New York). For a review of classical solution synthesis, see Schroder, G. and Lupke, K. (1965). The Peptides, vol. 1, Academic Press (New York).
In general, peptide synthesis methods comprise the sequential addition of one or more amino acids or suitably protected amino acids to a growing peptide chain. Normally, either the amino or the carboxyl group of the first amino acid is protected by a suitable protecting group. The protected or derivatized amino acid can then either be attached to an inert solid support or utilized in solution by adding the next amino acid in the sequence having the complimentary (amino or carboxyl) group suitably protected, under conditions suitable for forming the amide linkage. The protecting group is then removed from this newly added amino acid residue and the next amino acid (suitably protected) is then added, and so forth; traditionally this process is accompanied by wash steps as well. After all of the desired amino acids have been linked in the proper sequence, any remaining protecting groups (and any solid support) are removed sequentially or concurrently, to afford the final peptide compound. By simple modification of this general procedure, it is possible to add more than one amino acid at a time to a growing chain, for example, by coupling (under conditions which do not racemize chiral centers) a protected tripeptide with a properly protected dipeptide to form, after deprotection, a pentapeptide, and so forth. Further description of peptide synthesis is disclosed in U.S. Pat. No. 6,472,505. A preferred method of preparing the peptide compounds of the present invention involves solid- phase peptide synthesis, utilizing a solid support.
In some embodiments, peptidic leptin antagonists used to implement the teachings herein are generated using cell expression approaches by utilizing expression vectors for prokaryotic or eukaryotic expression or alternatively, the peptide can be expressed in-situ by delivering a suitable expression construct to cardiovascular tissue.
To express the peptide sequence in cardiovascular cells, a polynucleotide sequence encoding the peptide (see, for example, US20130I33089) is preferably ligated into a nucleic acid construct suitable for mammalian cell expression. Such a nucleic acid construct includes a promoter sequence for directing transcription of the polynucleotide sequence in the cell in a constitutive or inducible manner.
Constitutive promoters suitable for use with the present invention are promoter sequences that are active under most environmental conditions and most types of cells, such as the cytomegalovirus (CMV) and Rous sarcoma virus (RSV).
Polyadenylation sequences can also be added to the expression vector in order to increase the efficiency of mRNA translation. Two distinct sequence elements are required for accurate and efficient polyadenylation: GU- or U-rich sequences located downstream from the polyadenylation site and a highly conserved sequence of six nucleotides, namely AAUAAA, located 1 1-30 nucleotides upstream of the site. Termination and polyadenylation signals suitable for the present invention include those derived from SV40.
In addition to the embodiments already described, the expression vector of the present invention may typically contain other specialized elements intended to increase the level of expression of cloned nucleic acids or to facilitate the identification of cells that carry the recombinant DNA. For example, a number of animal viruses contain DNA sequences that promote extra-chromosomal replication of the viral genome in permissive cell types. Plasmids bearing these viral replicons are replicated episomally as long as the appropriate factors are provided by genes either carried on the plasmid or with the genome of the host cell.
The expression vector of the present invention may or may not include a eukaryotic replicon. If a eukaryotic replicon is present, the vector is capable of amplification in eukaryotic cells using the appropriate selectable marker. If the vector does not comprise a eukaryotic replicon, no episomal amplification is possible.
Figure imgf000021_0001
derivatives.
Expression vectors containing regulatory elements from eukaryotic viruses such as retroviruses can be also used. SV40 vectors include pSVT7 and pMT2 (SEQ ID NO 72), for instance. Vectors derived from bovine papilloma virus include pBV-lMTHA, and vectors derived from Epstein-Barr virus include pHEBO and p205. Other exemplary vectors include pMSG, pAV009/A+, pMTO10/A+, pMAMneo-5, baculovirus pDSVE, and any other vector allowing expression of proteins under the direction of the SV40 early promoter, SV40 later promoter, metallothionein promoter, murine mammary tumor virus promoter, Rous sarcoma virus promoter, polyhedrin promoter, or other promoters shown effective for expression in eukaryotic cells.
Viruses are very specialized infectious agents that have evolved, in many cases, to elude host defense mechanisms. Typically, viruses infect and propagate in specific cell types. The targeting specificity of viral vectors utilizes its natural specificity to specifically target predetermined cell types and thereby introduce a recombinant gene into the infected cell. Thus, the type of vector used by the present invention will depend on the cell type transformed. The ability to select suitable vectors according to the cell type transformed is well within the capabilities of the ordinarily skilled artisan and as such, no general description of selection considerations is provided herein. For example, bone marrow cells can be targeted using the human T-cell leukemia virus type I (HTLV-I) and kidney cells may be targeted using the heterologous promoter present in the baculovirus Autographa californica multiple nucleopolyhedro virus (AcMNPV), as described by Liang, C. Y. et al. (2004). High efficiency gene transfer into mammalian kidney cells using baculovirus vectors. Arch Virol 149, 51-60.
Recombinant viral vectors are useful for in vivo expression of a leptin peptide since they offer advantages such as lateral infection and targeting specificity. Lateral infection is inherent in the life cycle of retrovirus, for example, and is the process by which a single infected cell produces many progeny virions that bud off and infect neighboring cells. The result is the rapid infection of a large area of cells, most of which were not initially infected by the original viral particles. This is in contrast to vertical-type infection in which the infectious agent spreads only through daughter progeny. Viral vectors can also be produced that are unable to spread laterally. This characteristic can be useful if the desired purpose is to introduce a specified gene into only a localized number of targeted cells.
As is mentioned hereinabove, compositions (also called, compositions-of-matter) according to the teachings herein also includes a carrier for local delivery of the leptin antagonist. Such a carrier can be a mesh (Figure lc) an injectable gel (e.g. in-situ forming depot) (Figures la), a thin (preferably biodegradable) film (Figure lb), a scaffold (Figure 3). In some embodiments, the composition is a coating on a medical device (Figure 27A). In some embodiments, a medical device is impregnated with the composition (Figure 27B). In some embodiments, the composition is in the form of a sheet (such as the film of Figure lb or the mesh of Figure 1 c) that constitutes a portion of a medical device such as a stent cover (Figure 27C) or graft of a graft-stent assembly (Figure 27D). In some embodiments, the composition constitutes a medical device (Figure 27E).
In some embodiments a carrier of a composition is a balloon catheter, or a composition is delivered locally using a drug-eluting balloon catheter (Figure 2). The manufacture and use of drug-eluting balloons for localized delivery of active-pharmaceutical ingredients are well known in the art (especially to the walls of fluid-filled bodily cavities, such as of the cardiovascular system), for example, the In.Pact Admiral® DCB drug-coated balloon by Medtronic (Dublin, Ireland) and Lutonix® 035 by C. R. Bard, Inc. (Murray Hill, New Jersey, USA).
Examples of in-situ formed depots (ISFD) include semi-solid polymers which can be injected as a melt and form a depot upon cooling to body temperature or two part systems which gel upon mixing (Figure 3a). Depending on the embodiments, such compositions can be injected into or in contact with bodily tissue that is to be treated
The requirements for a semi-solid ISFDs include low melting or glass transition temperatures in the range of 25-65°C and an intrinsic viscosity in the range of 0.05-0.8 dl/g [12-14]. Below the viscosity threshold of 0.05 dl/g no delayed diffusion could be observed, whereas above 0.8 dl/g the ISFD was no longer injectable using a needle. At injection temperatures above 37°C but below 65°C these polymers behave like viscous fluids which solidify to highly viscous depots. Drugs are incorporated into the molten polymer by mixing without the application of solvents. In the art, it is known to use thermoplastic pastes (TP) can be used to generate a subcutaneous drug reservoir from which diffusion occurs into the systemic circulation. In contrast, in some embodiments of the teachings herein, a thermoplastic paste is used to generate a composition for the sustained release of leptin antagonist from which diffusion occurs into tissue in contact with the composition, thereby effecting sustained-release local administration of the leptin antagonist.
In situ cross-linked polymer systems utilize a cross-linked polymer network to control the diffusion of bioactive agents (e.g., leptin antagonists for implementing the teachings herein) over a prolonged period of time, thereby allowing implementation of sustained release compositions comprising leptin antagonists for use in local administration thereof. Use of in situ cross-linking implants necessitate protection of the bioactive agents during the cross-linking reaction. This could be achieved by encapsulation into fast degrading gelatin micro-particles.
An ISFD can also be based on polymer precipitation. A water-insoluble and biodegradable polymer is dissolved in a biocompatible organic solvent to which leptin antagonist is added forming a solution or suspension after mixing that constitutes a composition according to the teachings herein. When this composition is injected into the body of a subject in need thereof the water miscible organic solvent dissipates and water penetrates into the organic phase. This leads to phase separation and precipitation of the polymer forming a depot at the site of injection. One example of such a system is Atrigele™ (ARTIX Laboratories). The thus-formed depot is a composition for the sustained release of leptin antagonist from which diffusion occurs into tissue in contact with the composition, thereby effecting sustained-release local administration of the leptin antagonist.
Thermally induced gelling systems can also be used as ISFDs. Numerous polymers show abrupt changes in solubility as a function of environmental temperature. The prototypic thermosensitive polymer is poIy(N-isopropyl acryl amide), poly-NIPAAM, which exhibits a rather sharp lower critical solution temperature.
Thermoplastic pastes such as the new generation of poly(ortho-esters) developed by AP Pharma can also be used for depot drug delivery. Such pastes include polymers that are semi-solid at room temperature, hence heating for drug incorporation and injection is no longer necessary. Injection is possible through needles no larger than 22 gauge. The leptin antagonist is mixed into the systems in a dry and, therefore, stabilized state. Shrinkage or swelling upon injection is thought to be marginal and, therefore, the initial drug burst is expected to be lower than in the other types of ISFD. An additional advantage is afforded by the self-catalyzed degradation by surface erosion. As noted above, IFSD compositions are suitable for effecting sustained-release local administration of the leptin antagonist. In some embodiments, an IFSD composition can be formulated for sustained-release (SR), extended- release (ER, XR, or XL), time-release or timed-release, controlled-release (CR), or continuous-release.
Examples of thin films (Figure 3b) suitable for release of a leptin antagonist (or polynucleotide encoding same) include polymeric films (for a review of thin films, see Zelikin ACS Nano, 2010, 4 (5), pp 2494-2509; Venkat et al. 2010, Polymer Thin Films for Biomedical Applications, Wiley VCH Verlag GmbH & Co. KGaA, Weinheim). Such thin film carriers can be biodegradable or dissolvable over time.
Biodegradable microspheres fabricated from, for example, PGA, PLGA, PLA, or
PLLA can also be used for local delivery of a leptin antagonist. Such microspheres can be produced as described by Kim and Park (J Control Release. 2004 Jul 23;98(1): 1 15-25).
A balloon such as an angioplasty balloon (Figure 2) can also be used to deliver a leptin antagonist to a vascular wall or an inner wall of a heart chamber. Approach for coating/loading a balloon with a peptide are described in EP2643030; US8617136;
As noted
Figure imgf000024_0001
above, such drug-eluting balloons for use for localized delivery of active-pharmaceutical ingredients are well known in the art, for example, the In. Pact Admiral® drug-coated balloon by Medtronic (Dublin, Ireland) and Lutonix® 035 by C. R. Bard, Inc. (Murray Hill, New Jersey, USA). It is important to note that such drug-eluting balloons are known to administer extended release compositions of active pharmaceutical ingredients, e.g., In.Pact Admiral delivers a composition that provides extended release of a continuous therapeutic dose of Paclitaxel for over 180 days.
Although delivery of leptin or leptin receptor binding agents such as those described above (or expression thereof in cardiovascular cells), is presently preferred, downregulation of leptin activity at specific tissues can also be effected at the transcript level using a variety of molecules that interfere with transcription and/or translation (e.g., antisense, siRNA, Ribozyme, or DNAzyme).
RNA interference can be used to downregulate endogenous leptin via a small interfering RNA (siRNA) molecule. RNAi is a two-step process, in the first, the initiation step, input double- stranded (dsRNA) is digested into 21- to 23-nucleotide (nt) small interfering RNAs (siRNAs), probably by the action of Dicer, a member of the RNase III family of dsRNA-specific ribonuc leases, which processes (cleaves) dsRNA (introduced directly or by means of a transgene or a virus) in an ATP-dependent manner. Successive cleavage events degrade the RNA to 19- to 21 -bp duplexes (the siRNA), each with 2- nucleotide 3' overhangs (Hutvagner, G. and Zamore. P. D. (2002). RNAi: Nature abhors a double-strand. Curr Opin Gen Dev 12, 225-232; and Bernstein, E. (2001 ). Role for a bidentate ribonuclease in the initiation step of RNA interference. Nature 409, 363-366).
In the second step, termed the effector step, the siRNA duplexes bind to a nuclease complex to form the RNA-induced silencing complex (RISC). An ATP-dependent unwinding of the siRNA duplex is required for activation of the RISC. The active RISC then targets the homologous transcript by base-pairing interactions and cleaves the mRNA into 12-nucieotide fragments from the 3' terminus of the siRNA (Hutvagner and Zamore (2002); Hammond et al. (2001) Nat. Rev. Gen. 2:1 10-1 19 (2001); and Sharp, P. A. (2001). RNA interference. Genes Dev J5, 485-490). Although the mechanism of cleavage remainsl to be elucidated, research indicates that each RISC contains a single siRNA and an RNase (Hutvagner and Zamore (2002)).
Synthesis of RNAi molecules suitable for use with the present invention can be effected as follows. First, the leptin mRNA sequence (SEQ ID NO 2) is scanned downstream of the AUG start codon for AA-dinucIeotide sequences. Occurrence of each A A and the 19 3'-adjacent nucleotides is recorded as a potential siRNA target site. Preferably, siRNA target sites are selected from the open reading frame (ORF), as untranslated regions (UTRs) are richer in regulatory protein binding sites. UTR-binding proteins and/or translation initiation complexes may interfere with binding of the siRNA endonuclease complex (Tuschl (2001)). It will be appreciated, however, that siRNAs directed at untranslated regions may also be effective, as demonstrated for GAPDH, wherein siRNA directed at the 5' UTR mediated about a 90% decrease in cellular GAPDH mRNA and completely abolished protein levels (wwwdotambiondotcom/techlib/tn/91/912dothtml).
Second, potential target sites are compared to an appropriate genomic database (e.g., human, mouse, rat, etc.) using any sequence alignment software, such as the BlastN software available from the NCBI server (wwwdotncbidotnlmdotnihdotgov/BLAST/). Putative target sites that exhibit significant homology to other coding sequences are filtered out.
Qualifying target sequences are selected as templates for siRNA synthesis. Preferred sequences are those including low G/C content, as these have proven to be more effective in mediating gene silencing as compared with sequences including G/C content higher than 55%. Several target sites are preferably selected along the length of the target gene for evaluation. For better evaluation of the selected siRNAs, a negative control is preferably used in conjunction. Negative-control siRNAs preferably include the same nucleotide composition as the siRNAs but lack significant homology to the genome. Thus, a scrambled nucleotide sequence of the siRNA is preferably used, provided it does not display any significant homology to any other gene.
Another agent capable of downregulating leptin is a DNAzyme molecule, which is capable of specifically cleaving an mRNA transcript or a DNA sequence of the leptin. DNAzymes are single-stranded polynucleotides that are capable of cleaving both single- and double- stranded target sequences (Breaker, R. R. and Joyce, G. F. (1995). A DNA enzyme with Mg2+-dependent RNA phosphoest erase activity. Curr Biol 2, 655-660; Santoro, S. W. and Joyce, G. F. (1997). A general purpose RNA-cleaving DNA enzyme. Proc Natl Acad Sci USA 94, 4262-4266). A general model (the "10-23" model) for the DNAzyme has been proposed. "10-23" DNAzymes have a catalytic domain of 15 deoxyribonucleo tides, flanked by two substrate-recognition domains of seven to nine deoxyribonucleotides each. This type of DNAzyme can effectively cleave its substrate RNA at purine :pyrimidine junctions (Santoro and Joyce (1997)); for review of DNAzymes, see: Khachigian, L. M. (2002). DNAzymes: cutting a path to a new class of therapeutics. Curr Opin Mol Ther 4, 1 19-121.
Examples of construction and amplification of synthetic, engineered DNAzymes recognizing single- and double- stranded target cleavage sites are disclosed in U.S. Pat. No. 6,326,174 to Joyce et al. DNAzymes of similar design directed against the human Urokinase receptor were recently observed to inhibit Urokinase receptor expression, and successfully inhibit colon cancer cell metastasis in vivo (Itoh, T. et al., Abstract 409, American Society of Gene Therapy 5th Annual Meeting (wwwdotasgtdotorg), June 5-9, 2002, Boston, Mass. USA.). In another application, DNAzymes complementary to bcr-abl oncogenes were successful in inhibiting the oncogene's expression in leukemia cells, and in reducing relapse rates in autologous bone marrow transplants in cases of Chronic Myelogenous Leukemia (CML) and Acute Lymphoblastic Leukemia(ALL).
Downregulation of leptin can also be effected by using an antisense polynucleotide capable of specifically hybridizing with an mRNA transcript encoding leptin.
Design of antisense molecules that can be used to efficiently downregulate a leptin must be effected while considering two aspects important to the antisense approach. The first aspect is delivery of the oligonucleotide into the cytoplasm of the appropriate cells, while the second aspect is design of an oligonucleotide that specifically binds the designated mRNA within cells in a manner inhibiting the translation thereof.
The prior art teaches of a number of delivery strategies which can be used to efficiently deliver oligonucleotides into a wide variety of cell types (see, for example: Luft, F. C. (1998). Making sense out of antisense oligodeoxynucleotide delivery: getting there is half the fun. J Mol Med 75(2), 75-76 (1998); Kronenwett et al. ( 1998). Oligodeoxyribonucleotide uptake in primary human hematopoietic cells is enhanced by cationic lipids and depends on the hematopoietic cell subset. Blood 91, 852-862; Rajur, S. B. et al. (1997). Covalent protein- oligonucleotide conjugates for efficient delivery of antisense molecules. Bioconjug Chem 8, 935-940; Lavigne et al. Biochem Biophys Res Commun 237: 566-71 (1997); and Aoki, M. et al. (1997). In vivo transfer efficiency of antisense oligonucleotides into the myocardium using HVJ-liposome method. Biochem Biophys Res Commun 231, 540-545).
In addition, also available are algorithms for identifying those sequences with the highest predicted binding affinity for their target mRNA based on a thermodynamic cycle that accounts for the energetics of structural alterations in both the target mRNA and the oligonucleotide (see, for example, Walton, S. P. et al. (1999). Prediction of antisense oligonucleotide binding affinity to a structured RNA target. Biotechnol Bioeng 65, 1 -9).
Such algorithms have been successfully used to implement an antisense approach in cells. For example, the algorithm developed by Walton et al. enabled scientists to successfully design antisense oligonucleotides for rabbit beta-globin (RBG) and mouse tumor necrosis factor-alpha (TNF-alpha) transcripts. The same research group has more recently reported that the antisense activity of rationally selected oligonucleotides against three model target mRNAs (human lactate dehydrogenase A and B and rat gpl30) in cell culture as evaluated by a kinetic PCR technique proved effective in almost all cases, including tests against three different targets in two cell types with phosphodiester and phosphorothioate oligonucleotide chemistries.
In addition, several approaches for designing and predicting efficiencies of specific oligonucleotides using an in vitro system were also published (Matveeva, O. et al. (1998). Prediction of antisense oligonucleotide efficacy by in vitro methods. Nature Biotechnology 16, 1374-1375).
Another agent capable of down-regulating leptin is a ribozyme molecule capable of specifically cleaving an mRNA transcript encoding leptin. Ribozymes increasingly are being used for the sequence-specific inhibition of gene expression by the cleavage of mRNAs encoding proteins of interest (Welch, P. J. et al. (1998). Expression of ribozymes in gene transfer systems to modulate target RNA levels. Curr Opin Biotechnol 9, 486-496).
An additional method of regulating the expression of leptin in cardiovascular cells is via triplex-forming oligonucleotides (TFOs). Recent studies show that TFOs can be designed to recognize and bind to polypurine or polypirimidine regions in double-stranded helical DNA in a sequence-specific manner. These recognition rules are outlined in: Maher III, L. J., et al. (1989). Inhibition of DNA binding proteins by oligonucleotide-directed triple helix formation. Science 245, 725-730; Moser, H. E., et al. (1987). Sequence-specific cleavage of double helical DNA by triple helix formation. Science 238, 645-650; Beal, P. A. and Dervan, P. B. (1991). Second structural motif for recognition of DNA by oligonucleotide-directed triple-helix formation. Science 251, 1360-1363; Cooney, M., et al. (1988). Science 241, 456- 459; and Hogan, M. E., et al., EP Publication 375408. Modifications of the oligonucleotides, such as the introduction of intercalators and backbone substitutions, and optimization of binding conditions (e.g., pH and cation concentration) have aided in overcoming inherent obstacles to TFO activity such as charge repulsion and instability, and it was recently shown that synthetic oligonucleotides can be targeted to specific sequences (for a recent review, see Seidman, M. M. and Glazer, P. M. (2003). The potential for gene repair via triple helix formationj Clin Invest 112, 487-494).
As is described hereinabove, the present invention can be used to treat cardiovascular disorders affecting heart or vascular tissue. The following describes several option for local delivery of a leptin antagonist to tissue, for example heart and other cardiovascular tissue, specifically muscle and valve tissue.
(i) Arterial catheterization can be used to inject as a bolus, or to deploy a medical device such as a mesh, a thin film, a biodegradable scaffold, a stent cover, a stent, a graft assembly, a coil, a stent, a ring or a prosthetic cardiac valve loaded with a leptin antagonist against a luminal wall of an ascending aorta distal to the orifice of the coronary arteries. In case of aneurysm at another location along the aorta, a visceral artery, or small tributary; the same intra-arterial approach can be used for local application.
(ii) An IFSD (for example, as described above, e.g., a gel) loaded with a leptin antagonist can be delivered via a balloon or needle to the aortic wall.
(iii) A composition such as a pliable non-degradable or biodegradable mesh or film loaded with the leptin antagonist that is placed in contact with an outer surface of tissue or an organ to be treated, e.g., by surgical delivery via open surgery or thorascopy, for example surgically delivered to the peri-aortic region (above the aortic root level). In case of small aneurysm in the abdominal aorta the leptin antagonist extended release film or mesh can be applied via open surgery or minimally invasive laparoscopy.
Method of Treatment According to an aspect of some embodiments of the invention, there is also provided a method of treatment comprising: exposing in vivo tissue of a subject in need thereof to a pharmaceutically-effective amount of leptin antagonist thereby providing a therapeutic effect to the tissue. For example, in some such embodiments, a composition comprising leptin antagonist is administered (e.g., by injection) locally and selectively via intra-vascular approach, or directly into the tissue. In some embodiments, the exposing of the in vivo tissue to the leptin antagonist is substantially continuously for a period of not less than three days. In some embodiments, the period is not less than five days, not less than 8 days and even not less than 14 days. For example, in some such embodiments, an extended release composition comprising leptin antagonist (e.g., a medical device impregnated with, coated with or made from a leptin antagonist is placed directly in contact with the tissue- According to an aspect of some embodiments of the invention, there is also provided a method of treatment comprising implanting in contact with tissue in need thereof in the body of a subject a composition configured for the in vivo release of leptin antagonist, thereby providing a therapeutic effect to the tissue. In some embodiments, the composition is configured for sustained release of the leptin antagonist. In some embodiments, the implanting is intracavitary implanting within a fluid-filled bodily cavity of the subject. In some embodiments, by sustained release is meant that, when the composition is implanted in vivo, leptin antagonist is released from the composition in pharmaceutically effective amounts for a period of not less than three days, in some embodiments not less than five days, not less than 8 days and even not less than 14 days.
The subject in need thereof is any suitable mammalian subject. In some embodiments, the subject is a non-human animal. In some embodiments, the subject is a human.
The need is any suitable need. In some embodiments, the need is at least one need selected from the group consisting of: attenuating a pathology; reducing the chance of developing a pathology; reducing the rate of development a pathology; and mitigating the effect of a pathology.
The pathology is any pathology that can be treated by local administration of leptin antagonist, and in some embodiments, substantial continuous local exposure to leptin antagonist, in some embodiments, the pathology is at least one pathology selected from the group consisting of:
cardiovascular disease;
remodeling of stable athersclerotic plaque into an unstable lesion (vulnerable plaque or rupture-prone plaque); ascending aortic aneurysm, in some embodiments ascending aortic aneurysm associated with at least one member of the group consisting of hypertension, dyslipidemia, hypercholesterolemia, obesity, diabetes mellitus and bicuspid aortic valve (BAV);
thoracic aortic aneurysm (e.g., to prevent rupture or dissection thereof);
Takayasu disease (e.g., to attenuate cellular proliferative response,and aortic wall remodeling in some embodiments, by administration or implantation of a leptin antagonist composition to an inner or outer vessel wall in the vicinity of a vascular lesion);
Rheumatoid arteritis (e.g., to attenuate aortic wall remodeling, in some embodiments, by administration or implantation of a leptin antagonist composition to an inner or outer vessel wall in the vicinity of a vascular lesion);
Marian's syndrome (by mitigation or prevention of ascending aortic aneurysms or pulmonary artery aneurysms, in some embodiments, by perivascular administration or deployment of a leptin antagonist composition to the outer or inner wall of the ascending aorta); giant cell arteritis; ankylosing spondylitis; inflammatory aortic aneurysm; peripheral arterial or venous aneurysms; prevention of arterial dilatation at site of anchorage of bridging stent grafts ("landing zone") applied for EVAR (in the abdominal or thoracic aorta),visceral or peripheral arteries; prevention of myointimal hyperplasia at sites of vascular injury; prevention of restenosis following PTA or PTCA (peripheral or cardiac balloon angioplasty); angiogenesis; cancer; and arteriovenous malformation (e.g., administration of leptin antagonist composition directly into a malformation or into the feeding artery).
Depending on the embodiment, any pathology, including any pathology listed above, may be treated in accordance with the teachings herein by local administration of leptin antagonist. For example, in some embodiments, administration is local administration of a dose (optionally repeated) of leptin antagonist, for example by direct intravascular injection into the affected tissue or tissue proximal to the affected tissue or with the use of a drug- eluting balloon. For example, in some embodiments, administration is local administration by a sustained release composition (that releases a pharmaceutically-effective amount of leptin antagonist for a period of not less than three days) placed in contact with an outer surface of tissue (e.g., with tunica externa), inside the tissue (e.g., injection of a IFSD as described above into the tissue) or with a composition placed in contact with an inner surface of a tissue (e.g. with tunica intima).
The tissue is any suitable tissue. In some embodiments the tissue is part of the cardiovascular system. In some such embodiments, the tissue is selected from the group consisting of arteries, coronary arteries, ascending aorta, abdominal aorta, pulmonary artery mitral valve, aortic vaive and pulmonary valve. In some embodiments, the tissue is cardiovascular tissue with accumulated plaque, for example, an artery with accumulated plaque. In some embodiments, the tissue is a tumor, especially a cancerous tumor that grows or spreads in a process that includes angiogenesis. In some embodiments, the tissue is an arteriovenous malformation.
In some embodiments, the leptin antagonist composition is locally administered during or post surgery, e.g., following carotid thrombendartrectomy or after ablation of atherosclerotic occlusion from a vessel.
In some embodiments, the leptin antagonist is locally administered by contact to the outside of tissue to be treated, e.g., in contact with tunica externa.
In some embodiments, the leptin antagonist is locally administered inside tissue, for example, is injected or implanted inside tissue such as a tumor or the site of arteriovenous malformation.
In some embodiments, the leptin antagonist is locally administered intraluminally, e.g., a leptin antagonist composition is deployed in contact with a tunica intima, inside a fluid-filled bodily cavity such as inside the lumen of a blood vessel e.g., using an intraluminal catheter, for example, in conjunction with a stent or prosthetic cardiac valve.
It should be noted that in some embodiments, local administration of a leptin antagonist in accordance with the teachings herein at the ascending aorta may be effective in attenuating ascending aortic aneurysms, as well as moderating left ventricular hypertrophy, and left heart valve thickness (aortic and mitral). Administration of leptin antagonist at arterial aneurysms in other locations is anticipated to achieve a similar outcome, attenuating aneurysm expansion.
Accordingly, embodiments of the teachings herein are used to treat cardiovascular disorders such as heart valve stenosis, arterial or venous aneurysms, or left ventricular remodeling by enabling localized release of a leptin antagonist at the site of treatment.
Pharmaceutical Composition and Method of Making Pharmaceutical Composition
According to an aspect of some embodiments of the teachings herein, there is also provided a pharmaceutical composition, comprising: as an active ingredient a leptin antagonist; and a pharmaceutically acceptable carrier configured for in vivo sustained release of the leptin antagoni st.
According to an aspect of some embodiments of the teachings herein, there is also provided a method of making a pharmaceutical composition, comprising: combining a leptin antagonist; and a pharmaceutically acceptable carrier configured for in vivo sustained release of the leptin antagonist.
In some embodiments, by sustained release is meant that, when the composition is implanted in vivo, leptin antagonist is released from the composition in pharmaceutically effective amounts for a period of not less than three days, in some embodiments not less than five days, not less than 8 days and even not less than 14 days.
In some embodiments, the in vivo implantation is in a human subject. The in vivo implantation is in any suitable location. In some embodiments, the in vivo implantation is contacting an organ through a serous tissue layer or adventitia (tunica externa) layer covering the organ, e.g., is placed contacting a blood vessel such as the aorta from the outside of the blood vessel. In some embodiments, the in vivo implantation is outside an organ directly contacting tissue of the organ (for organs covered with serous tissue, the composition is implanted underneath the serous tissue). In some embodiments, the in vivo implantation is into an organ. In some embodiments, the implantation is from inside a hollow defined by the organ, for example, inside a blood vessel lumen contacting the endothelium thereof.
In some embodiments, the composition is in the form of a leptin antagonist containing sheet, in some such embodiments configured to be contacted with in vivo tissue, for example, by suturing, with the use of biological adhesive, or pressed against the tissue, for example with the help of a stent or such component, e.g., the sheet is used as a stent cover for a balloon-expandable or self-expanding stent.
In some embodiments, the composition is configured to coat or be supported by an implantable medical device, e.g., is used as a coating for, is adsorbed or absorbed into or onto a stent (thereby constituting a drug-eluting balloon expandable or self-expanding stent), prosthetic valve (e.g., cardiac valve), implantable spike or rod.
In some embodiments, the composition is formed into the shape of an implantable medical device, e.g., a bioresorbable stent, a bioresorbable spike or rod.
In some embodiments, the composition is injectable, e.g., is a viscous fluid or a fluid that subsequent to injection solidifies or gels, e.g., a hydrogel.
Any suitable pharmaceutically-acceptable carrier that can be configured for in vivo sustained release of the leptin antagonist can be used. In some embodiments the carrier comprises a biodegradable polymer. In some such embodiments, the carrier comprises a polymer selected from the group consisting of a hydrogel, poly giycolic acid (PGA), poly lactic co-glycolic acid (PLGA), polylactide (PLA), and poly (L-lactide) (PLLA), and combinations thereof. The pharmaceutical composition and methods of making such a composition are in accordance with those known in the art of pharmacology, using any suitable method or combination of methods as known in the art such as described in ''Remington's Pharmaceutical Sciences," Mack Publishing Co., Easton, PA, latest edition, which is incorporated herein by reference. Such methods include conventional mixing, curing, polymerizing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing. Pharmaceutical compositions may be formulated in conventional manner using one or more physiologically acceptable carriers, diluents, excipients or auxiliaries which facilitate processing of the leptin antagonist into a pharmaceutical composition.
Use of leptin antagonist
According to an aspect of some embodiments of the teachings herein, there is also provided a use of a leptin antagonist according to the teachings herein for the localized treatment of tissue of a living organism, comprising, implanting a composition comprising a leptin antagonist in vivo to contact tissue in need thereof so that the tissue is exposed to a pharmaceutically effective amount of leptin antagonist, thereby providing a therapeutic effect to the tissue. In some embodiments, the composition and the implanting is such that the tissue is exposed to a pharmaceutically effective amount of leptin antagonist substantially continuously for a period of at least three days. In some embodiments, the period is not less than five days, not less than 8 days and even not less than 14 days.
In some embodiments, an above administration is periodically repeated. For example, in some embodiments, administration of leptin antagonist is repeated. For example, in some embodiments, administration of leptin antagonist is repeated after at least a period, the period selected from the group consisting of 1 week, 2 weeks, 3 weeks, 4 weeks, 1 month, 2 months and 3 months.
Treatment of athersclerotic plaque
It is known that stable athersclerotic plaque accumulates in the inner walls of mammalian arteries. In some instances, the stable plaque transforms into an unstable lesion such as vulnerable plaque or rupture-prone plaque. Fragment from the unstable lesion may disintegrate, forming emboli.
An aspect of the teachings herein is based on the Inventor's discovery that locally synthesized leptin within the carotid atherosclerotic plaque, which characterize unstable plaques (rupture prone), correlates with brain emboli. Therefore, the inventor believes that a leptin antagonist administered to a stable athersclerotic plaque should reduce the rate and/or incidence of the conversion of a stable athersclerotic plaque to an unstable lesion.
According to an aspect of some embodiments of the teachings herein, there is provided a method for treatment of athersclerotic plaque, comprising: administering a pharmaceutically-effective amount of a leptin antagonist to athersclerotic plaque accumulated in the inner walls of an artery, thereby at least one of: (a) reducing the rate and (b) reducing the incidence, of conversion of a stable athersclerotic plaque to an unstable lesion.
Administration of the leptin antagonist is any suitable administration. In some embodiments, the administration is by sustained-release of the leptin antagonist directly to an inner wall in which plaque is accumulated, from a leptin antagonist containing composition. In some embodiments, such sustained release is substantial continuous release of a pharmaceutically-effective amount of leptin antagonist for a period of not less than three days, not less than 5 days, not less than 8 days and even not less than 14 days. In some such embodiments, the composition is in direct contact with the surface of the plaque to be treated. In some embodiments, the composition is in contact with the inner walls of a blood vessel with accumulated plaque.
Surgical connecting devices
It is know that trauma to a blood vessel may lead to myointimal hyperplasia (MIH),
One type of trauma that may cause MIH is caused by surgical connecting devices such as surgical staples and suture threads that are applied to blood vessels, for example, during surgery for example surgical anastomosis. It has been found that in some instances, local administration of leptin antagonist to such wounds may be able to mitigate or prevent MIH.
Thus, according to an aspect of some embodiments of the invention, there is also provided a surgical connecting device, comprising: a solid device body made of a material; and functionally associated with the device body, a pharmaceutically-effective amount of leptin antagonist. In some embodiments, the device body is in the form selected from the group consisting of surgical suture thread and a surgical staple.
An embodiment of a suture thread 10 and of a surgical staple 12 in accordance with the teachings herein are schematically depicted in Figure 27.
In some embodiments, the device body, e.g., of thread 10 or staple 12, is absorbable (i.e., bioresorbable). The device body, e.g., of thread 10 or staple 12, is made of any suitable material. In some embodiments, the device body is made of a material comprises a polymer selected from the group consisting of poly glycolic acid (PGA), poly lactic co-glycolic acid (PLGA), polylactide (PLA), and poly (L-lactide) (PLLA), and combinations thereof.
The leptin antagonist is functionally associated with the device body, e.g., of thread 10 or staple 12, in any suitable manner.
In some embodiments, the functional association of the leptin antagonist with the device body is that a composition comprising the leptin antagonist coats the device body. For example, in some such embodiments thread 10 or staple 12 are made of PGA, and coated with a coating of PLGA that includes a pharmaceutically effective amount of leptin antagonist.
In some embodiments, the functional association of the leptin antagonist with the device body is that a composition comprising the leptin antagonist impregantes the device body. For example, in some such embodiments thread 10 is made of silk and impregnated with a composition that includes a pharmaceutically effective amount of leptin antagonist.
In some embodiments, the functional association of the leptin antagonist with the device body is that the material from which the device body is made is a composition comprising the leptin antagonist. For example, in some such embodiments thread 10 or staple 12 are made of PGA that includes a pharmaceutically effective amount of leptin antagonist.
Administration of leptin antagonist in fluid-filled cavities
As noted above, the present inventor has found that in vivo implantation inside a fluid-filled cavity of the body (for example of the cardiovascular system such as blood vessels or cardiac chambers) can have a desirable pharmaceutical effect on tissue in proximity of the implanted composition with limited or no substantia] side-effects:
Method of treatment in a fluid-filled bodily cavity
Thus, according to an aspect of some embodiments of the invention, there is also provided a method of treating a condition in a subject in need thereof, the method comprising administering intracavitarily to inner walls of a fluid-filled bodily cavity of the subject a composition comprising a leptin antagonist. In some embodiments of the method, the subject is human. In some embodiments of the method, the subject is a non-human animal. In some embodiments of the method, the intracavitary administration exposes in vivo tissue to a pharmaceutically-effective amount of the leptin antagonist and thereby provides a therapeutic effect to the in vivo tissue.
In some embodiments of the method, the in vivo tissue comprises tissue of the inner walls of the cavity (e.g., tunica intima).
In some embodiments of the method, the administration is local administration. In some such embodiments, the in vivo tissue exposed to the pharmaceutically-effective amount of the leptin antagonist is exclusively tissue in physical proximity to the administered composition. In some such embodiments, the in vivo tissue exposed to the pharmaceutically- effective amount of the leptin antagonist is exclusively tissue in physical contact with the administered composition.
In some embodiments of the method, exposing of the in vivo tissue to a pharmaceutically-effective amount of the leptin antagonist is substantially continuously for a period of at least three days, at least five days, at least eight days and in some embodiments, even at least fourteen days.
In some embodiments of the method, the composition is a sustained-release composition, configured for sustained release of a pharmaceutically-effective amount of the leptin antagonist when located inside a bodily cavity. In some embodiments, the sustained release comprises release of a pharmaceutically-effective amount of the leptin antagonist over a period of at least three days, at least five days, at least eight days and in some embodiments, even at least fourteen days.
In some embodiments of the method, the intracavitary administering comprises implantation of the composition within the cavity in contact with the inner walls of the cavity.
In some embodiments of the method, the intracavitary administering comprises deploying an intracavitarily-implantable medical device in the cavity. In some such embodiments, the medical device is deployed in contact with the inner walls of the cavity. In some such embodiments, the leptin antagonist is functionally associated with the deployed intracavitarily-implantable medical device. In some preferred such embodiments, the leptin antagonist is functionally associated with a portion of the deployed intracavitarily- implantable medical device that contacts bodily tissue when the medical device is deployed. In some such embodiments, the intracavitarily implantable medical device is selected from the group consisting of: a stent cover, a graft assembly, a coil (e.g., aneurysm coil), a stent (e.g., expandable stent, self-expanding stent, covered stent, partially covered stent, not covered stent), a ring (e.g, a graft anchor), a suture, a staple and a prosthetic cardiac valve. Although the teachings herein are applicable to any prosthetic cardiac valve, the teachings are particularly advantageous for implementing with catheter-deployed prosthetic cardiac valves (e.g., TAMVI, TAVI): since these valves are typically held in place without sutures so that myointimal hyperplasia that potentially develops as a result of trauma caused during deployment may lead to leakage.
Composition for treatment in a fluid-filled bodily cavity
According to an aspect of some embodiments of the invention, there is also provided a composition comprising: a leptin antagonist for use in treating a condition, wherein the composition is configured for intracavitary administration to inner walls of a fluid-filled bodily cavity of a subject. In some embodiments of the composition, the subject is human. In some embodiments of the composition, the subject is a non-human animal.
In some embodiments, the intracavitary administration of the composition exposes in vivo tissue to a pharmaceutically-effective amount of the leptin antagonist, thereby providing a therapeutic effect to the in vivo tissue. In some embodiments, such in vivo tissue comprises tissue of inner walls of a the cavity.
In some embodiments, the administration is local administration. In some such embodiments, the in vivo tissue exposed to the pharmaceutically-effective amount of the leptin antagonist is exclusively tissue in physical proximity to the administered composition. In some such embodiments, the in vivo tissue exposed to the pharmaceutically-effective amount of the leptin antagonist is exclusively tissue in physical contact with the administered composition.
In some embodiments of the composition, exposing of the in vivo tissue to a pharmaceutically-effective amount of the leptin antagonist is substantially continuously for a period of at least three days, at least five days, at least eight days and in some embodiments, even at least fourteen days.
In some embodiments, the composition is a sustained-release composition, configured for sustained release of a pharmaceutically-effective amount of the leptin antagonist when located inside a bodily cavity. In some embodiments, the sustained release comprises release of a pharmaceutically-effective amount of the leptin antagonist over a period of at least three days, at least five days, at least eight days and in some embodiments, even at least fourteen days. In some embodiments of the composition, the configuration for intracavitary administration comprises configuration for implantation of the composition within the cavity in contact with the inner walls of the cavity.
In some embodiments of the composition, the configuration for intracavitary administration comprises configuration for deploying with an intracavitarily-implantabie medical device in the cavity. In some such embodiments, the medical device is configured for deployment in contact with the inner walls of the cavity. In some such embodiments, the leptin antagonist is functionally associated with the intracavitarily-implantabie medical device. In some preferred such embodiments, the leptin antagonist is functionally associated with a portion of the intracavitarily-implantabie medical device that contacts bodily tissue when the medical device is deployed. In some such embodiments, the intracavitarily implantable medical device is selected from the group consisting of: a stent cover, a graft assembly, a coil (e.g., aneurysm coil), a stent (e.g., expandable stent, self-expanding stent, covered stent, partially covered stent, not covered stent), a ring (e.g, a graft anchor), a suture, a staple and a prosthetic cardiac valve. As noted above, although the teachings herein are applicable to any prosthetic cardiac valve, the teachings are particularly advantageous for implementing with catheter-deployed prosthetic cardiac valves (e.g., TAMVI, TAVI).
In some embodiments of the method or composition, the condition is a pathological cardiovascular condition. In some embodiments of the method or composition, the condition is a cardiovascular condition selected from the group consisting of atherosclerosis, valve stenosis, aneurysms, vessel response to vascular injury, and cardiomyopathy. In some embodiments of the method or composition, the administration of the leptin antagonist is to athersclerotic plaque accumulated in the inner walls of an artery, thereby at least one of: (a) reducing the rate and (b) reducing the incidence, of conversion of a stable athersclerotic plaque to an unstable lesion. In some embodiments of the method or composition, the administration of the leptin antagonist is to bodily tissue in order to prevent or mitigate the development of myointimal hyperplasia, for example, myointimal hyperplasia that potentially develops as a result of trauma caused during deployment of a medical device in a the body of a living subject.
In some embodiments of the method or composition, the composition constitutes a coating of an intracavitarily implantable medical device. In some embodiments of the method or composition, the composition is impregnated in an intracavitarily implantable medical device. In some such embodiments of the method or composition, the composition comprises, in addition to the leptin antagonist, a polymer selected from the group consisting of a hydrogel, poly glycolic acid (PGA), poly lactic co-glycolic acid (PLGA), polylactide (PLA), and poly (L-lactide) (PLLA), and combinations thereof. In some embodiments of the method or composition, the composition is in the form selected from the group consisting of a sheet and a tube constituting a portion of an intracavitarily implantable medical device comprising the leptin antagonist. In some such embodiments of the method or composition, the composition comprises, in addition to the leptin antagonist, a polymer selected from the group consisting of of a hydrogel, poly glycolic acid (PGA), poly lactic co-glycolic acid (PLGA), polylactide (PLA), and poly (L-lactide) (PLLA), and combinations thereof. In some such embodiments of the method or composition, the intracavitarily implantable medical device comprises a stent and the composition constitutes a stent cover (e.g., a partial or complete stent cover, a balloon-expandable or a self-expanding stent). In some such embodiments of the method or composition, the intracavitarily implantable medical device comprises a graft-assembly (e.g., a stent-graft or ring-graft assembly) and the composition constitutes a graft portion thereof. For example, some embodiments are configured to function as a stent-graft assembly for treatment of AAA, like the Endurant® II by Medtronic (Dublin, Ireland). In some embodiments of the method or composition, the composition constitutes at least a portion of an intracavitarily implantable medical device, and in some embodiments, the composition constitutes substantially an entire intracavitarily implantable medical device. As noted above, in some such embodiments of the method or composition, the intracavitarily implantable medical device is selected from the group consisting of a stent cover, a graft assembly, a coil (e.g., aneurysm coil), a stent (e.g., expandable stent, self-expanding stent, covered stent, partially covered stent, not covered stent), a ring (e.g, a graft anchor), a suture, a staple and a prosthetic cardiac valve. In some such embodiments of the method or composition, the composition comprises, in addition to the leptin antagonist, a polymer selected from the group consisting of a hydrogel, poly glycolic acid (PGA), poly lactic co- glycolic acid (PLGA), polylactide (PLA), and poly (L-lactide) (PLLA), and combinations thereof.
Medical device for treatment in a fluid-filled bodily cavity According to an aspect of some embodiments of the invention, there is also provided an intracavitarily-implantable medical device, comprising:
at least one solid functional device part configured for deploying the device in a fluid- filled bodily cavity of a subject; and
functionally associated with at least one the device component, a leptin antagonist.
In some embodiments of the medical device, the leptin antagonist is functionally associated with the at least one the device part as a component of a pharmaceutical composition comprising the leptin antagonist.
In some embodiments of the medical device, the pharmaceutical composition is a sustained-release composition, configured for sustained release of a pharmaceutically- effective amount of the leptin antagonist when located inside a bodily cavity.
In some embodiments of the medical device, sustained release comprises release of a pharmaceutically-effective amount of the leptin antagonist over a period of at least three days, at least five days, at least eight days and in some embodiments, even at least fourteen days.
In some embodiments of the medical device, the pharmaceutically-acceptable carrier further comprises a polymer selected from the group consisting of a hydrogel, poly glycolic acid (PGA), poly lactic co-glycolic acid (PLGA), poiylactide (PLA), and poly (L-lactide) (PLLA), and combinations thereof,
In some embodiments of the medical device, the functional association is at least one device component having a coating comprising the pharmaceutical composition. In some embodiments of the medical device, the functional association is at least one device component being impregnated with the pharmaceutical composition. In some embodiments of the medical device, the functional association is at least one device component being fashioned of the pharmaceutical composition.
In some embodiments of the medical device, the medical device is selected from the group consisting of a stent cover, a graft assembly, a coil (e.g., aneurysm coil), a stent (e.g., expandable stent, self-expanding stent, covered stent, partially covered stent, not covered stent), a ring (e.g, a graft anchor), a suture, a staple and a prosthetic cardiac valve (as noted above, preferably catheter-deployed prosthetic cardiac valves.
In some embodiments of the method, composition and device, the fluid-filled bodily cavity is a bodily cavity of the cardiovascular system. In some such embodiments, the cavity is selected from the group consisting of a cardiac chamber, an artery and a vein. In some such embodiments, the cardiac chamber is selected from the group consisting of left ventricle, right ventricle, left atrium and right atrium, in some such embodiments, the artery is selected from the group consisting of a systemic artery, a coronary artery and a pulmonary artery. In some such embodiments, the systemic artery is an aorta, for example selected from the group consisting of an ascending aorta, aortic arch, descending aorta and an abdominal aorta.
In Figure 28, an aneurysm coil 14 according to the teachings herein is schematically depicted. Aneurysm coil 14 is substantially similar to known aneurysm coils except by being functionally associated with a pharmaceutically-effective amount of leptin antagonist, is made in substantially the same way, and is used in substantially the same way. Depending on the embodiment, a given aneurysm coil 14 may include one or more of the additional features detailed hereinabove. For example, in some embodiments, coil 14 is made of platinum, and coated with a coating of a hydrogel that includes a pharmaceutically effective amount of leptin antagonist. For example, in some embodiments, coil 14 is made of a material such as platinum or a polymer textured with micrometer dimension features such as valleys and pores, where inside the features is held a composition (e.g., a gel such as of Example 1 1) that comprises leptin antagonist. For example, in some embodiments, coil 14 is made of a composition comprising the leptin antagonist, e.g., is made of PGA that includes a pharmaceutically effective amount of leptin antagonist.
In Figure 28, a prosthetic cardiac valve 16 according to the teachings herein is schematically depicted. Prosthetic cardiac valve 16 is substantially similar to known prosthetic cardiac valves except by being functionally associated with a pharmaceutically- effective amount of leptin antagonist, is made in substantially the same way, and is used in substantially the same way. Specifically, prosthetic cardiac valve 16 has at least one component that is functionally associated with a pharmaceutically-effective amount of leptin antagonist. Depending on the embodiment, a given prosthetic cardiac valve 16 according to the teachings herein may include one or more components, each having one or more of the additional features detailed hereinabove. For example, in some embodiments, the retainer ring of prosthetic cardiac valve 16 is made of a cobalt chromium ring with a polyester cloth cover, the ring and cloth cover both coated with a coating of a hydrogel that includes a pharmaceutically effective amount of leptin antagonist. For example, in some embodiments, the leaflets of prosthetic cardiac valve 16 are made of a material such as porcine or bovine tissue (e.g., cardiac leaflets, pericardium) that has been soaked in and is therefore impregnated with a composition (e.g., a gel such as of Example 11) that comprises leptin antagonist. In Figure 28, a graft assembly 18 according to the teachings herein is schematically depicted. Graft assembly 18 is a ring-graft assembly suitable for treatment of abdominal aorta aneurysms and includes a flexible graft 20 that defines a conduit for blood flow and three expandable anchoring rings 22 as graft anchors. Each anchoring ring 22 is a radially expandable device that is substantially a single 360° ring of material. As known in the art, some embodiments of graft assemblies are stent-graft assemblies where one or more of the anchors are radially expandable stents, that are longer in the axial direction and/or describe more than a 360° degree rotation and/or comprise more than a single ring of material. Stents are preferred as anchors as these typically also provide support for the vessel in which deployed and provide greater anchoring of the graft to a vessel in which deployed. Graft assembly 18 is substantially similar to known graft assemblies except by being functionally associated with a pharmaceutically-effective amount of leptin antagonist, is made in substantially the same way, and is used in substantially the same way. Specifically, graft assembly 18 has at least one component that is functionally associated with a pharmaceutically-effective amount of leptin antagonist. Depending on the embodiment, a given graft assembly 18 according to the teachings herein may include one or more components, each having one or more of the additional features detailed hereinabove.
For example, in some embodiments, graft 20 is substantially a tube made of a high- density multifilament polyester cloth coated with a coating of a hydrogel that includes a pharmaceutically effective amount of leptin antagonist. In some embodiments, the coating is on the entire outer surface of graft 20. In some embodiments, the coating is on the outer surface of the termini of the three legs of graft 20 (e.g., a 5 cm length from each terminus.
For example, in some embodiments, graft 20 is made of a high-density multifilament polyester cloth coat that has been soaked in and is therefore impregnated with a composition (e.g., a gel such as of Example 11) that comprises leptin antagonist. In some embodiments, the entire graft 20 is impregnated with leptin antagonist composition. In some embodiments, only the termini of the three legs of graft 20 (e.g., a 5 cm length from each terminus) is impregnated with leptin antagonist composition.
For example, in some embodiments, rings 22 are made of nitinol, and coated with a coating of a hydrogel that includes a pharmaceutically effective amount of leptin antagonist. For example, in some embodiments, rings 22 are made of a material such as nitinol textured with micrometer dimension features such as valleys and pores, where inside the features is held a composition (e.g., a gel such as of Example 11) that comprises leptin antagonist. In Figure 29, stents 24, 26 and 28 according to the teachings herein are schematically depicted. Stents 24, 26 and 28 are all elongated, tubular, outwardly radially-expandable frameworks that are known in the art. Stent 24 is a coverless stent without a cover. Stent 26 is a partially-covered stent with a partial cover 30. Partial cover 30 is a sheet secured to the framework of stent 26 in the usual way, e.g., with sutures. Stent 28 is a covered stent with a full cover 32. Full cover 32 is a tube secured to the framework of stent 28 in the usual way, e.g., with sutures or by tension.
Stents 24, 26 and 28 are substantially similar to known stents except by being functionally associated with a pharmaceutically-effective amount of Ieptin antagonist, are made in substantially the same way, and are used in substantially the same way. Specifically, each one of stents 24, 26 and 28 has at least one component that is functionally associated with a pharmaceutically-effective amount of Ieptin antagonist. Depending on the embodiment, a given stent 24, 26 and 28 according to the teachings herein may include one or more components, each having one or more of the additional features detailed hereinabove. Embodiments of any one of stents 24, 26 and 28 are self-expanding stents. Embodiments of any one of stents 24, 26 and 28 are balloon-expandable stents.
For example, in some embodiments, a cover 30 or a cover 32 is made of a high- density multifilament polyester cloth coated with a coating of a hydrogel that includes a pharmaceutically effective amount of Ieptin antagonist, typically on the outer surface of the cover.
For example, in some embodiments, a cover 30 or a cover 32 is made of a high- density multifilament polyester cloth coated that has been soaked in and is therefore impregnated with a composition (e.g., a gel such as of Example 11) that comprises Ieptin antagonist.
For example, in some embodiments, a cover 30 or a cover 32 is made of a material that is a composition comprising Ieptin antagonist, e.g., a PLGA sheet of example 4.
For example, in some embodiments, a framework of any one of stents 24, 26 and 28 is made of cobalt chromium coated with a coating of a hydrogel that includes a pharmaceutically effective amount of Ieptin antagonist.
For example, in some embodiments, a framework of any one of stents 24, 26 and 28 is made of a material such as nitinol or a polymer textured with micrometer dimension features such as valleys and pores, where inside the features is held a composition (e.g., a gel such as of Example 1 1) that comprises Ieptin antagonist. For example, in some embodiments, a framework of any one of stents 24, 26 and 28 is made of a material that is a composition comprising leptin antagonist, e.g., PLA or PLLA comprising a pharmaceutically-effective amount of leptin antagonist. Treatment of ischemia and reperfusion injury (IRI)
Activated renin-angiotensin aldosterone system (RAAS) and angiotensin II (AngH) in ischemia/reperfusion injury IRI contribute to tissue damage through various pathways. It the myocardium Angll has been shown to induce cardiac cell hypertrophy and left ventricular dysfunction through the induction of leptin in cardiomyocytes. Subsequently, experimental data provide evidence that both, cardiomyocyte hypertrophy and the resulting heart failure can be mitigated by inhibiting leptin synthesis or activity. In the kidney angiotensin II causes cell injury through constriction of renal vessels, enhancement of vascular sensitivity to sympathetic nerve stimulation, increased oxidative stress and induced apoptosis. Also recruitment of macrophages and neutrophil activation in the injured kidney may be driven by angiotensin II and its mediator, leptin. Studies have demonstrated that angiotensin converting enzyme inhibitors (ACEIs) and angiotensin receptor blockers have protective effects on IRI in the kidney.
The present invention relates to the use of leptin antagonist not only in the treatment of a cardiovascular tissue but also in the treatment of an organ injured by ischemia and reperfusion injury (IRI) for alleviating tissue damage and preserving the organ function.
According to an aspect of the invention, there is provided a method for treating ischemia and reperfusion injury (IRI) by administrating leptin antagonist upon reperfusion and maintaining sustained administration of the leptin antagonist into the vessel undergoing the reperfusion procedure for delivering the leptin antagonist by the blood flow into the damaged cells of the tissue injured by the IRI.
According to some embodiments of the invention, the method is implemented by implanting an intracavitarily- implantable drug delivery device configured for sustained delivering the leptin antagonist. In particular the drug delivery device is a drug eluting stent that is used both to restore the flow of blood in an occluded vessel and to deliver leptin antagonist locally to the specific tissue perfused by the treated vessel. When the drug delivering device is a drug eluting stent, the stent is also configured to deliver an antiproliferative drug to the wall of the vessel for preventing restenosis. The anti-proliferative drug may be any known anti-proliferative drug such as rapamycin or paclitaxel. The total dose of leptin antagonist in the stent may be released into the lumen over a period of at least 3 days, preferably between 10 to 14 days, and even for a longer period. The release rate is determined, inter alia, by the choice of the sustained release material. For example, PLGA (Poly lactic-co-glycolic acid) 65:35 MW 45000-75000Da was tested in a mouse model and was found suitable to establish this timeframe. The kinetics of drug release for the antiproliferative drug may be designed based parameters known in the art. The stent may be used as a stand alone device, or as a supplement to support an intra-arterial bolus treatment with leptin antagonist, depending on the specific clinical circumstances.
According to some embodiments of the present invention, the method of treating IRI is applicable to isolated tissue sections that function as an end organ (i.e., are supplied by a single blood vessel), and that were exposed to a period of non-fatal ischemia and underwent reperfusion via thromboslysis or intravascular angioplasty.
According to some embodiments of the present invention, the method of treating IRI is applicable for the treatment of a cardiac left ventricular tissue after acute MI at the time of primary revascularization for moderation of cellular damage in cardiac cells (cardiomyocytes) that were subjected to ischemia and reperfusion injury and for preserving cardiac function.
According to some embodiments of the present invention, the method of treating IRI is applicable for treating IRI in any in other end organs, for example, the kidney, intestine or brain.
Double function drug eluting stent
According to an aspect of the invention, there is provided a double function drug eluting stent (df-DES) to be deployed in an occluded vessel at the site of occlusion following balloon angioplasty. The double function drug eluting stent is configured to enable slow release of anti-proliferative drug into the wall of a vessel to enable slow release of leptin antagonist into the lumen, to be carried by the blood stream and be uptaken by cells that sustained ischemia and reperfusion injury.
The double function dug eluting stent comprises an expandable structural framework configured to be deployed in a blood vessel. The stent may be a self-expandable stent or a balloon-expandable stent and may comprise a network of elongated struts.
Figure 30 schematically depicts a drug eluting stent 40 according to the teachings herein. Stent 40 is an elongated, tubular, radially-expandable stent comprising a network of struts 41. Struts 41 comprise outer surface reservoirs 42 and inner surface reservoirs 44, which are located, for example, at the junctions between the struts. Outer surface reservoirs 42 that abut the vessel wall when stent 40 is deployed within a vessel, contain a sustained release composition of an anti-proliferative drug. Inner surface reservoirs 44, which are in contact with the fluid flowing through the lumen, contain a sustained release composition of leptin antagonist. Reservoirs 42 and 44 can be configured, for example, as pits embedded in the junctions points between struts 41. Fig. 30 illustrates reservoirs 42 and 44 as circular and square pits, respectively, by way of example only. It will be easily realized that the pits may assume any other shape. Stent 40 may be a se!f-expandable or a self-expanding stent. Struts 41 may be made of metal, alloy or biodegradable polymer, as described above in association with Figure 29.
Figures 31 b and 31 c illustrate another embodiment of a double function drug eluting stent, depicting cross sectional views of one of the struts that constitute the stent, abutting vessel wall 50. According to this embodiment, the outer surface of struts 61 and 71 is partially coated by layer, 62 and 72, respectively, of a polymer containing the antiproliferative drug. Layers 62, 72 cover about 75% of the strut's circumference. The inner surface of the strut, facing the lumen, is provided with a long recessed slit, 64, 74, which serves as a reservoir for containing the sustained release composition of the leptin antagonist for slow release into the lumen. Slits 64 and 74 that extend along the length of the strut have circular and triangular cross sections, respectively, but may assume other cross sectional shapes as well. Preferably, the slit opening, 65, 75, at the luminal aspect of the struts leads to a wider reservoir. This design prevents early detachment of the sustained release matrix. The size of opening 65, 75 can also be selected as one of the parameters that control the release rate. According to some embodiments of the invention, layers 62, 72 and slits 64,74 extend continuously along the length of the stent. According to other embodiments, layers 54, 62 and slits 52 and 64 may be discontinuous. Similarly, all or only part of the struts constituting the stent may be loaded with the leptin antagonist and the anti antiproliferative drug. By way of comparison, Figure 3 la depicts a strut 51 of a prior art drug eluting stent coated by a layer 52 of antiproliferative drug to prevent in-stent stenosis (restonosis). It is noted that prior art drug eluting stents do not provide for selective release of one active agent into the wall of a vessel and a second different active agent into the lumen to be carried by the flow of blood and to be uptaken by the cells to which the vessel supplies blood. Suitable leptin antagonists
Any suitable leptin antagonist may be used in implementing any specific aspect or embodiment of the teachings herein. In some embodiments, a single leptin antagonist is used. In some embodiments, two or more leptin antagonists are used simultaneously or concurrently.
Various leptin antagonists suitable for implementing the teachings herein have been described in detail hereinabove.
In some embodiments, the leptin antagonist comprises a polypeptide portion.
In some embodiments, the leptin antagonist is a polypeptide.
Preferred leptin antagonists include all of the leptin antagonists listed and taught in US 7,307,342 (SEQ ID NOs 3-35) and US 8,969,292 (SEQ ID NOs 36-47), which are both included by reference as if fully set-forth herein. In some embodiments, the leptin antagonist is selected from the group consisting of:
a leptin antagonist consisting of: (a) a mammalian leptin polypeptide in which the LDFI (SEQ ID NO:33 in US 7,307,142 or SEQ ID NO 35 in the present application) hydrophobic binding site at the positions corresponding to positions 39-42 of the wild-type human leptin, is modified such that from two to four amino acid residues of the hydrophobic binding site are substituted with different amino acid residues such that the site becomes less hydrophobic, the modified, mammalian leptin polypeptide being a leptin antagonist; (b) a fragment of the modified mammalian leptin polypeptide of (a) comprising the altered hydrophobic binding site, wherein the fragment is itself a leptin antagonist; (c) a fragment of (b)
a synthetic leptin antagonist comprising: (d) a full length modified mammalian leptin polypeptide in which: (i) the LDFI hydrophobic binding site at the position corresponding to positions 39-42 of the wild-type human leptin is modified such that from two to four amino acid residues of the hydrophobic binding site are substituted with different amino acid residues such that the site becomes less hydrophobic; and (ii) the aspartic acid at the position corresponding to position 23 of the wild-type human leptin (D23) is substituted with an amino acid residue selected from the group consisting of glycine, alanine, leucine, lysine, arginine, phenylalanine, tryptophan and histidine, or the threonine at the position corresponding to position 12 of the wild-type human leptin (T12) is substituted with a different amino acid residue that is hydrophobic; (e) a synthetic leptin antagonist consisting of a polypeptide having the amino acid sequence of SEQ ID NO: 1 of US 8,969,292 or SEQ ID NO 36 of the present application;
(f) a modified mammalian leptin polypeptide in which: (i) the LDFI hydrophobic binding site at the position corresponding to 5 positions 39-42 of the wild-type human leptin is modified such that from two to four amino acid residues of the hydrophobic binding site are substituted with different amino acid residues such that the site becomes less hydrophobic, the modified mammalian leptin polypeptide being a leptin antagonist; and
(ii) the aspartic acid at the position corresponding to position 23 of the wild-0 type human leptin (D23) is substituted with a different amino acid residue that is not negatively charged or the threonine at the position corresponding to position 12 of the wild-type human leptin (T12) is substituted with a different amino acid residue that is hydrophobic;
(g) a fragment of the modified mammalian leptin polypeptide of (f), in which D235 is substituted with a different amino acid residue that is not negatively charged or T12 is substituted with a different amino acid residue that is hydrophobic, wherein the fragment is itself a leptin antagonist;
a synthetic leptin agonist comprising: (h) a modified mammalian leptin polypeptide in which D23 is substituted with a different amino acid residue that is not negatively charged or T12 is substituted with a different amino acid residue that is hydrophobic; (j) a fragment of the modified mammalian leptin polypeptide of (h), in which D23 is substituted with a different amino acid residue that is not negatively charged or T12 is substituted with a different amino acid residue that is hydrophobic, wherein the fragment is itself a leptin agonist;
(k) any one of the above wherein the mammalian leptin is selected from the group consisting of human, ovine and murine;
(1) any one of (a), (b), (c), (d), (e), (f), (g), (h), (j) and (k) in pegylated form; and a pharmaceutically acceptable salt of any one of (a)-(l).
Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the invention pertains. In case of conflict, the specification, including definitions, will take precedence.
As used herein, intracavitary relates to within an organ or body cavity. Art that provides enabling support for the teachings herein, and that may also be useful in understanding the background and the inventive aspects of the teachings herein includes US 7,307,342; US 8,969,292; "leptin Locally Synthesized in Carotid Atherosclerotic Plaques Could Be Associated With Lesion Instability and Cerebral Emboli" by Schneiderman J et al in J Am Heart Assoc. 2012, l :e001727 doi: 10.1 161/JAHA.l 12.001727 and "Locally Applied leptin Induces Regional Aortic Wall Degeneration Preceding Aneurysm Formation in Apolipoprotein E -Deficient Mice" by Tao M et al in Arterioscler Thromb Vase Biol. 2013;33:31 1 -320, all four which are included by reference (together with any published supplemental materials) as if fully set-forth herein.
As used herein, the terms "comprising", "including", "having" and grammatical variants thereof are to be taken as specifying the stated features, integers, steps or components but do not preclude the addition of one or more additional features, integers, steps, components or groups thereof. These terms encompass the terms "consisting of and "consisting essentially of.
As used herein, the indefinite articles "a" and "an" mean "at least one" or "one or more" unless the context clearly dictates otherwise.
As used herein, when a numerical value is preceded by the term "about", the term "about" is intended to indicate +/- 10%.
Additional objects, advantages, and novel features of the present invention will become apparent to one ordinarily skilled in the art upon examination of the following examples, which are not intended to be limiting.
EXAMPLES
Reference is now made to the following examples, which together with the above descriptions, illustrate the invention in a non limiting fashion.
EXAMPLE 1
Localized Leptin Synthesis in a Mouse Mode]
A novel mouse model was used to simulate local leptin synthesis in the ascending aorta in order to assess the effect of leptin on aortic remodeling and heart structure and function.
Materials and Methods A slow release leptin film (Figure lb) made of polylactic co-glycolic acid (PLGA) matrix (1 X 1.5 mm), and containing either 2 μg leptin or no protein (control) was applied to the anterior surface of the proximal ascending aorta (Figure 4).
The leptin slow-release film was manufactured by impregnating a poly lactic-co- glycolic acid (PLGA) film with leptin. One gram of PLGA 6535 polymer (D,L- lactide:glycolide: 65:35, Mw = 45,000-75,000 Da; Lakeshore. Biomaterials, Birmingham, AL, USA) was dissolved in 10 mL MgCb (Fisher Scientific, Loughborough, UK). Sodium chloride (10 mg in 0.2 mL distilled water) and 25 ethylene glycol (Sigma-Aldrich, St. Louis, MO, USA) were added to the polymeric solution and sonicated for 20 seconds. Mouse leptin powder (1 mg; #L3772; Sigma-Aldrich, St. Louis, MO, USA) was suspended in 2 mL of the polymeric solution, followed by casting on a flat surface of Teflon molds to create a fiat film. Films were dried in a hood for 48 hours, and then subjected to high vacuum for 12 hours to extract any residual solvent. Control (placebo) films were fabricated in the same way without the addition of leptin. The calculated amount of leptin per 3 X 1.5-mm film used currently for implantation in each mouse was 2 μg.
Another option of leptin application for local slow-release has been a gel composed of two liquid materials which gel (solidify) upon mixing at the time of injection. These are a modified carboxymethyl cellulose with adipic dihydrazide (CMC-ADH) and an oxidized dextrane in DDW (DX-COH). Methylene blue dye (0.5%) was also added to the DX-COH solution to make the resulting gel more visible. Leptin (Sigma, L3772, St. Louis, MO, USA) was added to the gel by an emulsion technique.
Serum leptin levels were determined in ApoE"A mice after receiving 20μg mouse leptin via peri-aortic application (in another experiment, Tao et al. ATVB 2013). Blood was samples on days 0, 7, 14, and 21 , and leptinanalyzeded by ELISA assay (Quantikine Mouse Lep Kit, R&D Systems, Minneapolis, MN, USA): Day 0 - 3.5 ng/mL; day 7 - leptin 8.0 ng/mL, placebo 9.2 ng/mL; day 14 - leptin 12.0 ng/mL, placebo 14.5 ng/mL; and day 21- leptin 12.25 ng/mL, placebo 12.5 ng/mL (Figure 5). Notably, these values fell within the normal range of plasma leptin in ApoE-/- mice receiving Western diet (mean 5.1 ± 1.4 to 17 ± 3.4 ng/mL). It should also be emphasized that circulating leptin levels are known to increase with age, as also observed in our series.
This unique mouse model was utilized to perform two experiments: Mice in experiment 1 were fed postoperatively with high fat diet (HFD), and were followed up for 45 days. In experiment 2 mice received normal chow for 30 or 60 days. Mouse weight and blood pressure (BP) were assessed weekly. All mice recovered from surgery uneventfully. Results
In both mouse model experiments, leptin or control treated mice gained weight equally during the follow up period, suggesting no systemic leptin effect. Systolic BP measured weekly in mice of experiment 2 was 100 mmHg throughout the first 4 weeks, and increased to 120 mmHg by week 6 in both leptin treated and control mice. Based on two separate experiments, both HFD and normal chow feeding yielded in general similar results.
The following data report the results of experiment 2 (normal chow feeding).
Echocardiography of the ascending aorta at 2 mm distal to the aortic valve level revealed an increase in aortic diameter at peak systole in leptin treated mice vs controls (p=0.08, Figure 6; Exp. 1 using HFD yielded P<0.003). That same aortic location exhibited decreased elasticity, which was defined as the percent increase in aortic diameter in systole vs. diastole, in leptin compared to control treated mice. There was no significant difference in diameter further distally on the ascending aorta. Notably, the aortic valve annulus did not dilate in response to local leptin application. Histological analysis of the ascending aorta revealed features of medial degeneration at the site of leptin application, including fragmentation of the elastic lamellas, as demonstrated by elastica van Giesen staining, and depletion of aSMA in the media (Figure 7). These structural changes likely underlie local stiffening and dilatation in the proximal ascending aorta.
Echocardiography (final vs. pre-operative) revealed a concentric remodeling of the left ventricle, with hypertrophy of all LV walls (pO.001). Wall thickening was most pronounced in diastole (p=0.002, Figure 8). Left ventricular diameter was increased in both systole and diastole (p=0.08, p=0.02, respectively, Figure 9), leading to a reduction in the LV fractional area change (FAC, p=0.07, Figure 10).
Local leptin application at the proximal ascending aorta promoted thickening of the mitral and aortic valve leaflets (p=0.01, p<0.001 accordingly, Figure 1 1). Mitral leaflets were diffusely thickened, while aortic valve leaflets displayed thickening in their free edge, composed mostly of ECM and stromal cells. These proliferating cells are assumed analogous to human valvular interstitial cells (VICs). A few stromal cells within these lesions were positive for aSMA and TGFp as shown by IHC staining (in analyzed aortic valve leaflets), suggesting VICs activation (Figure 1 1 ). A trend was observed for increased VIC proliferation through Ki67 IHC in leptin treated mice. However, the lack of statistical significance implies that most leaflet hyperplasia took place at an earlier time. Increased peak systolic velocity (PSV), as measured at the aortic valve in leptin treated vs control mice was short of statistical significance. However, PSV was significantly augmented in postoperative HFD fed animals.
These experiments reveal that available leptin in the proximal ascending aorta induces local aortic stiffening and dilatation. The resulting changes in local hemodynamics likely drive remodeling of the left ventricle, including LV wall hypertrophy and valve thickening through the aorto-ventricular coupling axis.
EXAMPLE 2
Local Leptin Antagonism in an Ang II Mouse Model
Angiotensin II (Angll) is the key hormone of the renin-angiotensin system, underlying hypertension and cardiovascular remodeling (Renna et al. Pathophysiology of vascular remodeling in hypertension. Int J Hypertens. 2013;2013: 808353). The phenotypes induced by local leptin application described in Example 1 are reminiscent of Angll induced aortic-ventricular (coupling) remodeling, suggesting that leptin mediates these processes. As such, a leptin antagonist was delivered locally to the ascending aorta in order to assess the effects of leptin down-regulation on Angll induced local aortic remodeling, and aortic- ventricular remodeling in mice. Materials and Methods
An osmotic mini-pump, delivering Angll at a rate of 1000 ng/kg/min was implanted subcutaneously in the back of the neck of 14 week old ApoE"7" mice. Each mouse also underwent left mini-thoracotomy for application of a slow release miniature PLGA film (1x1.5 mm) containing either 5 μg leptin antagonist (LA), or PLGA matrix devoid of protein (control). The slow release film was deployed on the surface of the proximal ascending aorta at the position described in Experiment 1. Mice were euthanized 4 weeks following surgery. As expected, blood pressure assessed in both Ang II treated groups after one week was increased by approximately 20% (125 mniHg mean systolic), and was sustained throughout the follow up (Figure 12). Weight gain pattern was similar in both groups, indicating no systemic effects related to the leptin antagonist (Figure 13).
Results
To assess the impact of Angll alone versus Angll plus leptin antagonist on mouse longevity, mortality data from the present experiment were combined with data from a previous experiment, which included a similar cohort of ApoE_/~ mice exposed to Angll, in same dose and duration (Tao M, et al. ATVB 2013). Collectively, a 34% mortality (referred to premature death, prior to the completion of the experiment) was observed in mice treated with Angll (either Ang II alone or Ang II with control film applied on the ascending aorta). Death was related to thoracic (28%) or abdominal (6%) aortic aneurysm rupture. Notably, mice treated with Angll that received also LA were protected from thoracic aneurysm rupture (p=0.04, Figure 14). Death rate in mice receiving LA was only 13%, related exclusively to rupture of abdominal aortic aneurysms.
Echocardiograpic imaging of the ascending aorta demonstrated that local LA application in Angll treated mice significantly attenuated dilatation of the ascending aorta compared to Angll alone when measured 2 mm from the valve, at both diastole and systole (p=0.03, p~0.005, respectively, Figure 15). However, these data did not suggest moderation of increased aortic stiffness by LA application.
Histological analysis revealed medial degeneration in both groups that were treated with Angll. Nevertheless, additional LA application resulted in less fragmentation of the elastic lamellas and fewer sites of aSMA depletion in the aortic media (Figure 16). Notably, amongst mice receiving Angll, medial degeneration was rather diffused throughout the aorta. This was in sharp contrast to the effects of local leptin application, which exhibited medial degeneration within the segment in contact with the leptin film alone.
Immunohistochemical analysis for leptin antigen revealed a weak expression in medial SMCs, and a strong signal within foam cells of aortic luminal atherosclerotic plaques (Figure 17).
Mice treated with LA presented less thickening of the left ventricular wall, particularly in diastole (p<0.01, Figure 18). Left ventricular diameter increased similarly in both groups in diastole however, LA treatment attenuated the increase in LV diameter during systole (p=0.05, Figure 19). As anticipated, and corresponding to these results, a decrease in FAC in mice co-treated with Angll and LA, was observed, while mice treated by Angll alone exhibited a decrease in fractional area change by over 15% (p=0.03, Figure 20). Moreover, LV diameter which increased in response to Angll treatment, was preserved within the baseline (pre- Angll treatment) range in the LA treated mice (P<0.05).
Peak systolic velocity was decreased in Angll treated mice that also received LA application, vs. Angll alone (p=0.03, Figure 21). Notably, since no aortic valve obstruction or changes in its annulus diameter were detected, the PSV parameter is likely reflecting the interaction between proximal aortic hemodynamics, and left ventricular systolic contraction. Thus, PSV moderation by LA may represent attenuation of both aortic and LV remodeling.
LA also attenuated remodeling of the LV valve. Angll-induced thickening of aortic and mitral valve leaflets was reduced by LA application in both valves (p=0.06 in both valves, Figure 22 left panels F-I).
The aSMA and TGFP antigens were observed in aortic valve leaflet stromal cells in all Angll treated mice (Figure 22 panels J-M'); decreased proliferation of stromal cells in LA treated mice was demonstrated through Ki67 staining (p=0.26).
Thus, the present findings show that application of a leptin antagonist at the pivotal location on the proximal ascending aortic surface prevents rupture of thoracic aneurysms induced by systemic infusion of Ang 11. Local inhibition of leptin activity reduces the degenerative effects of Ang II on the proximal aorta, which underlie aortic wall destabilization. Thus, moderation of Ang II induced aortic dilatation and attenuates left heart remodeling, presumably via the aorto-ventricular coupling.
These results highlight the role of leptin as a key mediator of Ang II signaling and indicate that leptin which underlie left ventricular hypertrophy also drives the formation of early aortic valve hyperplastic lesions, which may progress to aortic valve stenosis (AVS).
EXAMPLE 3
The Role of Leptin in A VS
Materials and Methods
Human AVS and normal arterial valve (AV) samples were collected for analysis, including autopsy samples, freshly collected AVS specimens from patients undergoing aortic valve replacement surgery, and normal aortic valves from explanted hearts. Formalin fixed valve samples were analyzed by immunohistochemistry for leptin, leptin receptor, CD68 and aSMA. Fresh samples of AVS valves and normal aortic valves underwent total RNA extraction and analyzed by qPCR and Nanostring technique to assess leptin and leptin receptor mRNA levels. Retroperitoneal fat was used as a positive control in both assays, Results
Normal aortic valve leaflets lack leptin (Ob) antigen, and show very few leptin receptor (ObR) positive cells (Figure 23). Advanced AVS disease was characterized by extensive ossification and infiltration of inflammatory macrophages in the non-calcified rim of cellular tissue (Figure 24). Leptin was demonstrated mostly in two cell types, SMC-like elongated cells, and macrophage-like round cells, and its prevalence was proportional to the severity of AVS disease. In situ hybridization analysis performed on AVS samples demonstrated leptin mRNA expression, suggesting de novo synthesis (not shown) leptin and leptin receptor mRNA levels were assessed by qPCR and Nanostrings hybridization, using total RNA extracted from freshly collected AVS. AVS were compared to normal AV leaflets (Figure 25), revealing increased leptin and leptin receptor mRNA in AVS samples.
To investigate the potential impact of Angli and leptin on human valve cells, in vitro analysis revealed that Angll-mediated proliferation of human valve interstitial cells (VICs) is leptin mediated (leptin-induced proliferation of VICs in Figure 26). This suggests that leptin synthesized in aortic valve leaflets by VICs and inflammatory macrophages may elicit VIC proliferation and subsequent ossification via a paracrine/autocrine pathways.
EXAMPLE 4
Preparation of sustained-release film
Poly iactic-co-glycolic acid (PLGA) films containing a leptin antagonist (e.g., a leptin antagonist described in US 8,969,292) is produced in a manner analogous to the described in Webber WL et al "Characterization of soluble, salt-loaded degradable PLGA films and their release of tetracycline" J Biomed Mater Res 1998, 41 , 18-29.
Specifically, 1 g PLGA 6535 polymer (D,L-lactide:glycolide: 65:35, Mw - 45,000- 75,000 Da; Lakeshore. Biomaterials, Birmingham, AL, USA) is dissolved in 10 mL MeCI2 (Fisher Scientific, Loughborough, UK). Sodium chloride (10 mg in 0.2 ml distilled water) and 25 μL ethylene glycol (Sigma-Aldrich, St. Louis, MO, USA) are added to the PLGA solution and sonicated for 20 seconds, lmg of the leptin antagonist is suspended in 2 mL of the PLGA solution, followed by casting on the flat surface of a Teflon mold to create a film comprising a leptin antagonist.
EXAMPLE 3
Evaluation of sustained-release properties of film
A 2x2.5-mm patch of PLGA film of Example 4 is maintained in 5 mL sterile PBS at 37°C for 28 days. Every 7 days the PBS medium is sampled before being aspirated and replaced by 5 mL of fresh PBS. The medium samples are analyzed by ELISA (RayBiotech, Norcross, GA, USA) for leptin antagonist levels. The in vitro measurement of leptin discharged from the slow-release PLGA film yields substantial release of leptin antagonist for at least two weeks. EXAMPLE 6
Preparation of two-component gelling sustained-release gel composition
A first component of the composition is an aqueous solution of modified carboxymethyl cellulose with adipic dihydrazide (CMC-ADH). Dried and finely ground sustained-release gel with leptin antagonist as described in Example 1 is added to the CMC- ADH solution and vortexed to yield a suspension.
A second component of the composition is oxidized dextrane in DDW (DX-COH). A dye such as methylene blue (0.5%) is optionally added to the DX-COH solution to make the resulting gel more visible.
For use the two components are mixed together to form a viscous fluid that is immediately administered by injection. In a short time, the viscous fluid gels.
EXAMPLE 7
Covered stents
A stent cover is fashioned from the PLGA film of Example 4.
The stent cover is used to cover the balloon expandable stents of a MULTI-LINK 8 LL coronary stent system, a MULTI-LINK ULTRA coronary stent system and a MULTI- LINK MINI VISION coronary stent system (all of Abbot Laboratories, Abbot Park, Illinois, USA).
The graft (stent cover) portion of an Endurant II AAA Stent Graft System (Medtronic, Dublin, Ireland) is impregnated with a leptin antagonist. In one example, impregnation is by immersion in a first component of the composition of Example 3, followed by contact with contact with the second component thereof.
The stent cover is used as an external cover for a self-expanding WallFIex Stent
(Boston Scientific Corporation, Natick, MA, USA).
The covered stents are deployed in the usual way inside the lumen of a blood vessel of a living subject in need thereof. Once implanted, the leptin antagonist elutes from the stent cover through the blood vessel endothelium into the blood vessel to exert a desired pharmaceutical effect.
EXAMPLE 8
Drug-eluting stent An X1ENCE Alpine coronary stent system (Abbot Laboratories, Abbot Park, Illinois, USA) is prepared in the usual way, but impregnated with a leptin antagonist as an active pharmaceutical ingredient instead of Everolimus.
The resulting drug-eluting stent is deployed in the usual way inside the lumen of a blood vessel of a living subject in need thereof. Once implanted, the leptin antagonist elutes from the stent through the blood vessel endothelium into the blood vessel to exert a desired pharmaceutical effect.
EXAMPLE 9
Bioresorbable stent
A bioresorbable balloon-expandable stent is fashioned of bioresorbable polylactide (PLA) comprising a leptin antagonist, substantially as done to fashion a bioresorbable stent by Arterial Remodeling Technologies (Paris, France).
A bioresorbable balloon-expandable stent is fashioned of bioresorbable poly (L- lactide) (PLLA) comprising a leptin antagonist and also comprises a bioresorbable coating of PLLA including a leptin antagonist, substantially as done to fashion an Absorb GTl vascular scaffold stent.
The resulting bioresorbable stents are deployed in the usual way inside the lumen of a blood vessel of a living subject in need thereof. Once implanted, the leptin antagonist elutes from the stents as these resorb, to pass through the blood vessel endothelium into the blood vessel to exert a desired pharmaceutical effect.
EXAMPLE 10
Spike or Rod
An implantable spike or rod (bioresorbable or not) is made as known in the art, for example as described above with reference to the stents and includes leptin antagonist integrated into the material of the spike or rod, or adsorbed, absorbed or coated onto the spike or rod.
The resulting spike or rod is implanted in the usual way inside an organ, for example by piercing the organ. Once implanted, the leptin antagonist elutes from the spike or rod, to exert a desired pharmaceutical effect on the organ.
EXAMPLE 11
Injectable Gel The two components of the composition of Example 3 are provided. The components are mixed together and implanted in the body of the subject by injection into or onto an organ to form a gelled mass in or on the organ.
Once implanted, the leptin antagonist elutes from the gelled mass, to exert a desired pharmaceutical effect on the organ.
EXAMPLE 12
Sheet
A sheet of the film of Example 1 is provided. The sheet is placed against the outer surface of an organ (e.g., aorta, for example, ascending aorta, aortic arch, descending aorta, abdominal aorta) and optionally held in place by sutures and/or biological glue (e.g., Evicel® by Ethicon of Johnson and Johnson).
Once implanted, the leptin antagonist elutes from the film, to exert a desired pharmaceutical effect on the organ.
EXAMPLE 13
Treatment of AAA
A human subject is diagnosed with an abdominal aortic aneurysm (AAA).
A composition according to the teachings herein in the form of a long sheet such as described in Example 12 (a ribbon) comprising leptin antagonist is provided. The abdominal aorta of the subject is surgically accessed from the outside (e.g., using keyhole surgery) and the composition administered by winding the long sheet around the abdominal aorta to wrap the entire aneurysm as well as a portion of the aorta above and below the aneurysm, and held in place with a biological glue and/or sutures. Optionally, a stent graft (e.g., an Endurant® II AAA Stent Graft System (Medtronic, Dublin, Ireland)) is deployed in the aneurysm in the usual way, before or after administration of the composition. Subsequently, leptin antagonist from the composition passes through the tunica externa to provide a beneficial effect to the subject,
Alternatively or additionally, an AAA stent graft is provided that is similar to known AAA stent grafts (e.g., similar to an Endurant® II AAA Stent Graft System (Medtronic, Dublin, Ireland)) where at least one of: the graft and/or anchoring stents are a composition of the teachings herein and comprises a leptin antagonist; at least the anchoring portions of the graft and/or anchoring stents are impregnated with a composition of the teachings herein that comprises a leptin antagonist; and at least the anchoring portions of the graft and/or anchoring stents are coated with a composition of the teachings herein that comprises a leptin antagonist. The stent-graft is deployed in the usual way in the abdominal aneurysm, that is to say, where the anchoring stents are expanded against healthy portions of tunica intima above and below the aneurysm. Subsequently, leptin antagonist from the composition passes through the tunica intima to provide a beneficial effect to the subject, for example, preventing or reducing the extent that the aneurysm spreads to portions of tissue in proximity of the anchoring stents, thereby preventing loosening of the anchoring stents.
Alternatively or additionally, a drug-eluting balloon similar to the In. Pact Admiral® DCB drug-coated balloon by Medtronic (Dublin, Ireland) that is coated with a composition comprising leptin antagonist according to the teachings herein is introduced through the femoral arteries and advanced to areas of the abdominal and iliac arteries that are just above and just below the aneurysm (for example, "landing zones" where anchoring stents of a stent- graft would be deployed. The balloon is expanded to contact the aorta and iliac walls, thereby administering composition to the healthy tissue and preventing advancement of the aneurysm. In some such embodiments, the administration of leptin antagonist is repeated periodically, e.g., with a frequency that is less than once a month, less than once every two months and even less than once every 3 months. In some such embodiments, the administration of the leptin antagonist leads to stabilization of the wall tissue, halting the processes of aneurysm formation at the portions of the blood vessels above and below the aneurysm and at the portions of the blood vessel in contact with the deployed stent graft.
EXAMPLE 14
Treatment of thoracic aortic aneurysm
A human subject is diagnosed with an aneurysm in the thoracic aorta, including one or more of the ascending aorta, aortic arch and descending aorta. It is known that segmental increased stiffness and aortic dilatation cause local aneurysm formation. These structural changes underlie hemodynamic perturbation, which increases left ventricular afterload. This results in left ventricular remodeling, including left ventricular hypertrophy, and thickening of the aortic and mitral valve leaflets. Left ventricular hypertrophy may lead to heart failure, and aortic valve remodeling may progress to the full clinical presentation of aortic valve stenosis.
A composition according to the teachings herein in the form of a patch such as described in Example 12 comprising leptin antagonist is provided. The thoracic aorta of the subject is surgically accessed from the outside (e.g., using thorascopy) and the composition administered by contacting the outer surface of the affected portions of the aorta with the patch, and optionally holding the patch in place with a biological glue and/or sutures. ) aortic valve annulus diameter) a drug eluting stent graft impregnated with leptin antagonist is deployed in the aneurysm in the usual way, without oversizing.
Another shape of intra-vascular device that may be deployed within an aortic aneurysm is a tubular self-expandable biodegradable (or non-degradable, like bare metal) leptin antagonist slow release mesh or stent. Such a device may self-expand upon deployment, and gently adhere to the luminal surface of the aortic aneurysm (applying minimal radial force to the luminal surface). Leptin antagonist that is associated with, e.g., incorporated within the biodegradable or non-degradable struts of the mesh or stent or covered or coated onto the mesh or stent, will access the aortic wall at the aneurysm by local diffusion. Yet another shape of a device for proximity and local slow release of leptin antagonist at the luminal surface of aortic or peripheral aneurysm will be a single wire (biodegradable or bare metal) possessing the memory of spiral expansion within the aneurysm cavity. The leptin antagonist incorporated within this wire will diffuse into the arterial wall. Subsequently, leptin antagonist from the composition passes into the tissue to provide a beneficial effect to the subject, in some embodiments one or more of attenuate aneurysm progression, stabilize the vessel wall and prevent rupture or dissection of the aneurysm. In some embodiments of treatment of the ascending aorta, the administration of the leptin antagonist also leads to reducing the rate of development, or stopping the development and in some embodiments, reversing the remodeling of parts of the heart.
EXAMPLE IS
Angioplasty
A human subject is diagnosed with arterial stenosis that is treatable by angioplasty.
A drug-eluting balloon similar to the In.Pact Admiral® DCB drug-coated balloon by Medtronic (Dublin, Ireland) that is coated with a composition comprising leptin antagonist according to the teachings herein is used in the usual way to perform the angioplasty procedure, for example, at sites of arterial bifurcations and in-stent stenoses. At least some of the composition according to the teachings herein that coats the balloon is administered to the surface of the treated blood vessel, thereby administering a composition according to the teachings herein. In some such embodiments, the administration of leptin antagonist is repeated periodically, e.g., with a frequency that is less than once a month, less than once every two months and even less than once every 3 months, even when there is no express need for repeated angioplasty. Subsequently, leptin antagonist from the composition passes into and/or through the lesion and/or tunica intima to provide a beneficial effect to the subject.
Alternatively or additionally, at least one of:
a composition according to the teachings herein in the form of a stent;
a stent impregnated with a composition according to the teachings herein;
a stent coated with a composition according to the teachings herein;
a composition according to the teachings herein in the form of a stent cover;
a stent cover impregnated with a composition according to the teachings herein; and a stent cover coated with a composition according to the teachings herein;
is deployed in the usual way, e.g., during performance of an angioplasty procedure, thereby administering a composition according to the teachings herein. Subsequently, leptin antagonist from the administered composition passes into and/or through the lesion and/or tunica intima to provide a beneficial effect to the subject.
EXAMPLE 16
Myocardial Infarction
Myocardial infarction causes left ventricular remodeling, leading to progressive impairment of cardiac function. A human subject is diagnosed with an acute myocardial infarction and is treated in the usual way, for example coronary catheterization for primary revascularization and myocardial salvage. A treating health-care professional identifies that the subject has an elevated risk of developing cardiac dysfunction.
A composition comprising leptin antagonist is administered to the ascending aorta as described in the preceding examples using one or more of a drug-eluting balloon, a stent, a covered stent and a stent graft. Subsequently, leptin antagonist from the administered composition passes into and/or through the tunica intima of the aorta to provide a beneficial effect to the subject. In some embodiments, the beneficial effect is prophylactic, preventing development of or reducing the rate of development of an thoracic aortic aneurysm, and/or remodeling of the heart (in particular the left ventricle and associated valves) and/or a recurring infarction. Without wishing to be held to any one theory, it is currently believed that such administration of a leptin antagonist in the ascending aorta reduces angiotensin II synthesis in the left ventricle, thereby moderating the hypertrophy response to the ischemic insult associated with the acute myocardial infarction suffered by the subject. EXAMPLE 16 -A
Post Myocardial Ischemia (MI) therapy
In order to minimize the extent of post MI left ventricular remodeling, a bolus of leptin antagonist in aqueous solution may be administered into the involved coronary artery. The following strategy may be exercised for acute MI patients: Patients who sustain acute MI are most frequently admitted through the catheterization lab, to undergo coronary
catheterization and primary PTCA (Percutaneous Transarterial Coronary Angioplasty) for primary revascularization. Once blood-flow is re-established in the coronary artery involved, a bolus of leptin antagonist in an aqueous solution is to be injected into the coronary artery through the catheter, afterwhich the catheter will be withdrawn. This new strategy should achieve local distribution of leptin antagonist within the left ventricular heart muscle cells (cardiomyocytes) that were exposed to the ischemic as well as reperfusion insult. Inhibition of cardiomyocyte leptin activity is anticipated to mitigate left ventricular remodeling, and reduce the damage to left ventricular function.
Induction of myocardial ischemia in experimental animals may be achieved by temporary balloon inflation within the proximal left anterior descending (LAD) coronary artery. Leptin antagonist aqueous solution may be injected into the LAD after balloon deflation. Control group may receive intracoronary bolus of saline injection.
An intravascular injection of leptin antagonist may be provided into the treated coronary artery after it has been reopened and blood flow to the ischemic myocardium is restored, in order to prevent post MI left ventricular remodeling. Leptin antagonist may be administered by intravascular injection into a vascular territory that sustained ischemia and reperfusion injury. This injection is a localized injection to a specific section of the left ventricle and does not constitute a systemic treatment.
EXAMPLE 17
Myointimal Hyperplasia (MIH)
It is known that trauma to a blood vessel may lead to myointimal hyperplasia (MIH), where medial smooth muscle cells undergo uncontrolled proliferation that may lead to stenosis or restenosis of the blood vessel in the area of the trauma. Such trauma include vascular injury caused by expansion of a blood vessel during angioplasty, stent deployment, stent-graft deployment, as a result of surgical anastomosis and associated suturing, clamping of blood vessels, and as a result of blunt and/or penetrating vascular injury.
A health-care professional identifies that a subject has an elevated risk of developing myointimal hyperplasia due to some vascular trauma, administers a composition comprising leptin antagonist according to the teachings herein to the site of the trauma. The administered leptin antagonist reduces the rate or stops the uncontrolled proliferation of cells, reducing the rate of development or preventing MIH. Administration includes the use of any of the compositions according to the teachings herein, including localized administration of a leptin antagonist composition on the outer surface of the blood vessel at the site of injury during surgery (e.g., application of a film of Example 4 as a patch), peri-vascular injection of leptin antagonist in solution or within a slow release gel, Intravascular administration using a drug eluting balloon, or by administration of a composition that impregnates or coats a medical device, or a composition that is in the shape of a medical device by deploying the medical device. In some embodiments, specific suitable medical devices include intracavitary devices such as a stent cover, a stent, a graft assembly, a ring, a suture and a prosthetic cardiac valve as well as extraluminal devices such as sheets, all such comprising leptin antagonist according to the teachings herein,
EXAMPLE 18
Treatment of aneurysm
A human subject is diagnosed with a peripheral or venous aneurysm, e.g., a visceral artery aneurysm, a cerebral aneurysm, especially a saccular or pseudo -fusiform aneurysm.
A composition according to the teachings herein associated with a covered stent is provided, e.g., one or more of coating the stent, coating the stent cover, impregnating the stent, impregnating the stent cover, constituting the stent and constituting the stent cover. The covered stent is deployed in the usual way, where the stent cover covers the mouth of the stent. Subsequently, leptin antagonist from the composition passes into and/or through the tissue in proximity to the aneurysm to provide a beneficial effect to the subject.
Alternatively or additionally, a composition according to the teachings herein is placed inside the cavity of the aneurysm through the mouth thereof, e.g., as a fluid composition (e.g., Example 6) or as an aneurysm coil that is impregnated with leptin antagonist, coated with leptin antagonist or is made of a composition according to the teachings herein. A person having ordinary skill in the art is able to implement coating an aneurysm coil with an active pharmaceutical ingredient with reference to, for example, Cerecyte® (Codman Neuro, a division of DePuySynthes, part of Johnson & Johnson, New Brunswick, NJ, USA), Nexus® (Micro Therapeutics, Inc., Irvine, CA, USA), and HydroCoil®, HydroSoft® (Terumo Corporation, Tokyo, Japan). Subsequently, leptin antagonist from the composition passes into the cavity of the aneurysm, and subsequently to affected tissue to provide a beneficial effect to the subject.
EXAMPLE 19
Treatment of aneurysm
A human subject is diagnosed with an aneurysm, e.g., aortic aneurysms, which are typically related to a variety of diseases associated with angiotensin II hormonal activity. Also peripheral arterial aneurysms, which affect visceral, carotid, peripheral, and cerebral arteries, as well as venous aneurysms, including pulmonary artery (which carries venous blood) may be diagnosed.
A leptin antagonist eluting stent or scaffold may be provided, e.g., intravascular stent or scaffold device (which may or may not be biodegradable) covered or coated with leptin antagonist, available for slow release into the vessel wall locally, thereby attenuating aortic aneurysm progression. A stent-graft destined for treatment of aortic aneurysm may be provided, covered or coated with leptin antagonist available for slow release into the vessel wall at the specific sites of stent-graft attachment to non-dilated (normal) proximal and distal vessel (landing zones). In some embodiments, a stent-like prosthetic heart valve for intravascular application, covered with leptin antagonist may be provided, to prevent local dilation of the hosting tissue ring.
EXAMPLE 20
Treatment of vascular injury
A human subject may be exposed to localized vascular injury which may occur as a result of vascular surgery, local balloon angioplasty. This may cause local arterial narrowing due to smooth muscle cell proliferation, namely myointimal hyperplasia (MIH). This proliferative response may also occur at arterio-venous anastomosis or graft-arterial anastomosis, and in leptin-induced inflammatory and cellular proliferative arterial disease (e.g., Takayasu disease). In order to prevent local arterial narrowing due to cellular proliferation at the sites of vascular injury, and circumstances as described above, an intravascular stent or scaffold device may be provided, (which may or may not be biodegradable) which may be associated with, e.g., covered or coated with leptin antagonist that is available for slow release into the vessel wall. Also, peri-vascular injection of leptin antagonist slow release gel can attenuate the MIH response.
EXAMPLE 21
Treatment of atherosclerotic plaques
A human subject is diagnosed with atherosclerotic plaques. These lesions frequently undergo transformation from a stable plaque into an unstable rupture-prone lesion. An intravascular stent or scaffold device (which may or may not be biodegradable) covered with leptin antagonist that is available for slow release into the vessel wall may be provided, in order to provide vessel stabilization. Deployment of such a stent or scaffold device may apply to any arterial site. Also, peri-vascular injection of leptin antagonist slow release gel can pacify the atherosclerotic plaque and prevent its transformation into an unstable lesion.
EXAMPLE 22
Treatment of left heart failure and aortic valve disease
Patients who are diagnosed as hypertensive and hypercholestrolemic and exhibit initial dilation of the ascending aorta may be treated by positioning an intravascular stent or scaffold device at the ascending aorta. The stent or scaffold device may or may not be biodegradable. The device, may be associated with, e.g., covered or coated with leptin antagonist that is available for slow release into the vessel wall. Subsequently, slowing down the progression of the aortic aneurysm by intraluminal, locally applied leptin antagonist will prevent the development of left heart failure, and remodeling of aortic and mitral valve leaflets.
The intravascular stent or scaffold device carrying or covered with leptin antagonist, may be positioned intra-luminally at the ascending aorta. Such a stent or scaffold device may attenuate local dilation and stiffening thereby prevent local hemodynamic perturbation, which activate the aorto-ventricular coupling. This coupling, when turned on promotes the production and release of angiotensin II from left ventricular cells (cardiomyocytes).
Angiotensin II drives intracellular synthesis of leptin in cardiomyocytes and in aortic va!ve interstitial cells (VICs). Leptin synthesis in cardiomyocytes and VICs contributes to left ventricular hypertrophy, and aortic/mitral valve thickening, respectively. Therefore, deactivation of the aorto-ventricular coupling will control both left ventricular hypertrophy (heart failure) and also attenuate the progression of aortic/mitral valve disease. EXAMPLE 23
Effect of excessive endogenous leptin expression on post-MI heart failure
Post-MI heart failure involves complex mechanisms. One of these mechanisms is the over-expression of leptin in cardiomyocytes after ischemia and reperfusion injury (IRI). Leptin activity in cardiomyocytes promotes left ventricular hypertrophy and fibrosis, both of which contribute to cardiac dysfunction. Post-MI therapies in rats, aimed at blocking leptin activity via systemic (Purham et al., Am. J Physiol. Heart Circ. Physio. 2008: 295.Ή441 - H446) or local (Moro et al., J Cell Mol. Med. 201 1 ; 15: 1688-1694) approaches mitigated post-MI cardiac damage. Patients that suffer from systemic inflammatory disease and exhibit hyperleptinemia have been found to be at an increased risk of heart failure following acute MI. Therefore, to assess the role of increased endogenous leptin synthesis in the context of IRI, we intentionally induced leptin expression in an ischemia and reperfusion mouse model. A single IP injection of high dose leptin antagonist (LepA) with a short half-life transiently disrupted the availability of leptin receptors, and induced endogenous over expression of the leptin gene in multiple tissues, including the heart.
Activated renin-angiotensin aldosterone system (RAAS) acts via angiotensin II (Angll) in IRI, contributing to tissue damage through various pathways. In the myocardium Angll induces cardiac cell hypertrophy leading to left ventricular dysfunction through the induction of leptin in cardiomyocytes. Past experimental data have provided evidence that both, cardiomyocyte hypertrophy and the resulting heart failure can be mitigated by inhibiting leptin synthesis and activity. In the kidney (which is an end organ that is solely supplied in most cases by a single artery, mechanisms related to angiotensin II cause cell injury through constriction of renal vessels, enhancement of vascular sensitivity to sympathetic nerve stimulation, increased oxidative stress and induced apoptosis. Also recruitment of macrophages and neutrophil activation in the injured kidney may be driven by angiotensin II and its mediator, leptin. Studies have demonstrated that angiotensin converting enzyme inhibitors (ACEIs) and angiotensin receptor blockers have protective effects on IRI in the kidney.
In the current experiment, a clinical scenario of myocardial ischemia and reperfusion injury (IRI) has been simulated in C57BL6 mice in order to examine the effects of excessive endogenous leptin on post-MI heart failure. Myocardial IRI was achieved by ligating the anterior descending coronary artery for 45 minutes. Thereafter, the artery was re-opened to restore perfusion. Excessive endogenous leptin expression was induced by intraperitoneal (IP) injection of 25(^g of superactive mouse leptin antagonist (SMLA) that was administered sequentially following the induced myocardial IRI, upon reperfusion. Control mice received intraperitoneal injection of saline solution upon perfusion. It is emphasized that the intraperitoneal injection of SMLA, namely a systemic administration of leptin antagonist, is used as a trigger to disrupt the systemic leptin pathway, stimulating augmented leptin synthesis.
Leptin mRNA was increased significantly in different tissues throughout the first 48 hours, including adipose tissue and heart. Leptin receptor mRNA presented a similar pattern. Levels of leptin mRNA and leptin receptor mRNA were measured in the heart over a period of 30 days (Figs. 32). Interestingly, the heart exhibited maximal leptin mRNA upregulation 20 hours post IRI. There was also a long-term increase in leptin transcript by 3-6 fold on post operative day (POD) 30, compared to saline controls.
Echocardiography performed 24 hours and 30 day following IRI in LepA versus saline injected mice revealed a significant reduction in cardiac function in LepA receivers, on post-IRI day 30 (Figure 33).
As can be clearly seen, IRI mice injected with leptin antagonist exhibited more extensive post-MI cardiac dysfunction compared to IRI control mice. These results imply that the extent of post-MI cardiac dysfunction con-elates directly with local leptin expression. Notably, the capability to block local leptin activity when using local application of leptin antagonist in angiotensin Il-induced leptin mediated aortic aneurysm mouse model has been previously demonstrated experimentally by the current inventor. Collectively, based on multiple data, it is therefore hypothesized that the local inhibition of leptin activity in LV cardiomycytes would attenuate post-MI cardiac tissue damage. It should be emphasized that the strategy of local administration of leptin antagonist selectively into the infarcted territory, should prevent systemic hormonal perturbation. A previous key experiment demonstrated effective mitigation of post-MI heart failure by disrupting leptin synthesis, via, antisense oligodesoxy nucleotide against leptin mRNA that was injected directly into the infarcted zone in rats undergoing cardiac IRI. It is therefore conceivable that direct intracoronary injection of LepA, aimed exclusively at the infarct-related myocardial territory, could be a viable therapeutic option for cardiomyocytes that sustained sub-critical injury, to diminish the extent of post-MI heart failure (HF).
The mRNA and histological analyses suggest that most left ventricular cellular/tissue damage takes place within the first 24 hours. Therefore, a single bolus injection of leptin antagonist that will be administered into the infarct-related coronary artery may be sufficient to inhibit leptin activity during the critical period of leptin synthesis in cardiomyocytes. However, in order to address cases of extended period of increased leptin expression as synthesized by injured cardiomyocytes, it is suggested to supplement the bolus therapy by deploying a df-DES within the involved coronary artery. This approach will achieve extended period of sustained leptin antagonist release over additional 7-14 days. This may specifically apply to patients that suffer from an inflammatory comorbidity that have intrinsic stimulation for leptin synthesis. It is emphasized that according to the present invention the administration of leptin antagonist is made directly and exclusively into the infarcted myocardium.
EXAMPLE 24
Treatment of brain ischemia and reperfusion injury by local injection of LepA
into the revascularized artery
A clinical scenario of major brain ischemia and reperfusion injury has been simulated in mice to examine the effects of selective intra-arterial leptin antagonist (LepA) on global brain ischemia and reperfusion injury in mice.
Eight weeks old C57BL6 male mice underwent bilateral ligation of the common carotid arteries (CCA). After a period of 10 min ischemia, the right CCA was re-perfused. Leptin antagonist (SMLA), 2C^g dissolved in 50μ] saline, was immediately bolus injected selectively into the right ECA, to be delivered to the brain through the right ICA. Twenty four hours later mice were euthanized, and brains were perfused with 4% paraformaldehyde. The brains were removed and additionally fixed in 10% paraformaldehyde over night at 4° C and embedded in paraffin. The brain was cut into coronal sections, and 5μ slides were stained with H&E (Figure 34). As can be seen, while pyramidal cells in regions C A 1 , C A2 and C A3 of the hypocampus in the brain that underwent IRI are severely damaged, most of the pyramidal cells in the same regions of the brain that underwent IRI plus selective administration of leptin antagonist are intact.
These experimental results demonstrate the neuroprotective effects of leptin antagonist when administered locally into the brain immediately after reperfusion, implying that local inhibition of leptin activity in a brain that sustained IRI will attenuate cellular damage, and most likely will preserve cerebral function. It An extended selective local administration of leptin antagonist into the ischemic and re-perfused region may provide additional benefits in regard to brain cell preservation. It is therefore suggested that both routs of administration, including leptin antagonist bolus injection and/or slow release of leptin antagonist by the double function drug eluting stent (df-DES) of the invention may be used to alleviate brain IRI damage. Furthermore, brain neurodegenerative diseases, like Alzheimer's disease are associated with ischemia and inflammation affecting the brain microcirculation (Kalaria RN; Neurol Res. 2000;21 :321-330). it is therefore suggested that selective intraarterial administration of leptin antagonist into the cerebral circulation, through bolus injection or slow release via df-DES may attenuate pathological changes in the brain and mitigate symptoms related to different neurodegenerative disorders, like Alzheimer's disease.
It is appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention, which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable subcombination.
Although the invention has been described in conjunction with specific embodiments thereof, it is evident that many alternatives, modifications and variations will be apparent to those skilled in the art. Accordingly, it is intended to embrace all such alternatives, modifications and variations that fall within the spirit and broad scope of the appended claims. All publications, patents and patent applications mentioned in this specification are herein incorporated in their entirety by reference into the specification, to the same extent as if each individual publication, patent or patent application was specifically and individually indicated to be incorporated herein by reference. In addition, citation or identification of any reference in this application shall not be construed as an admission that such reference is available as prior art to the present invention. Section headings are used herein to ease understanding of the specification and should not be construed as necessarily limiting.
Figure imgf000071_0001
Figure imgf000072_0001

Claims

WHAT IS CLAIMED IS;
1. A drug emting stent for treating ischemia and repetrusion injury,, comprising
a structural framework configured to be deployed in a blood vessel,, the structural tramework having an. outer surface and an inner surface;
wherein said outer surface is configured to enable sustained-release of antiproliferative drug and said inner surface is configured to enable sustained-release of a ieptin antagonist.
2. The drug elutmg stem according to claim 1 , wherein upon deployment of the stent in said blood vessel, said outer surface abut, the blood vessel wail and said inner surface is in contact with blood flowing through the vessel.
3. The drug elating stent, according to claim 1, wherein said structural framework comprises a plurality of outer surface reservoirs containing a sustained-release composition of said antiproliferative drug and a plurality of inner surface reservoirs containg a sustained- release composition of said Ieptin antagonist;
4. The drug elating stent according to claim 3, wherein said outer surface reservoirs and said inner surface reservoirs are configured as pits located in said outer surface and said inner surface, respectively.
5. The drag eluting stent according to claim 3, wherein said outer surface reservoirs comprise a layer coating the outer surface of said structural framework.
6. The drug eluting steal according to claim 3, wherein said inner surface reservoirs are configured as elongated slits extending along said struts.
7. The drug eluting stent according to claim 1 , wherein said structural framework comprises a network of elongated struts.
8. The drug eluting stent according to claim 1 , wherein said stent is a balloon expandable or a .self-expandable stent.
9. The drug elating stent according to claim .1 , wherein said blood vessel is. art aorta,
10. The drug eluiing stent according' to. claim 1, wherein said sustained-release of said ieptin antagonist is continuous for a period of at least three days.
11. The drug eluiing stent according to claim 1 , wherein said sustained-release of .said leptin antagonist is continuous for a period of 7 to 14 days.
12. The drug eluiing stent according to claim 1 , wherein said sustained-release of said leptin antagonist is continuous for a period of at least fourteen day s.
13. The drug elating stent according to claim L wherein said stent comprises a total dose of leptin antagonist in the range of 5 to 30 pg.
14. A method for treating an. ischemia and reper&sion injury (IR1), the method comprising administering leptin antagonist selectively into a re-opened blood vessel by the drug eluiing stent of any one of cl aims 1 -13.
PCT/IL2017/050201 2015-02-26 2017-02-15 Drug eluting stent WO2017141246A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US16/078,013 US10926006B2 (en) 2015-02-26 2017-02-15 Drug eluting stent
IL261182A IL261182B (en) 2016-02-15 2018-08-15 Leptin antagonist for use in the treatment of ischemia and reperfusion injury
US16/153,890 US11123461B2 (en) 2015-02-26 2018-10-08 Treatment of ischemia and reperfusion using leptin antagonist

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201662295178P 2016-02-15 2016-02-15
US62/295,178 2016-02-15

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US15/000,062 Continuation-In-Part US10143780B2 (en) 2015-02-26 2016-01-19 Methods and compositions relating to leptin antagonists

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US16/078,013 A-371-Of-International US10926006B2 (en) 2015-02-26 2017-02-15 Drug eluting stent
US16/153,890 Continuation-In-Part US11123461B2 (en) 2015-02-26 2018-10-08 Treatment of ischemia and reperfusion using leptin antagonist

Publications (1)

Publication Number Publication Date
WO2017141246A1 true WO2017141246A1 (en) 2017-08-24

Family

ID=59625630

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IL2017/050201 WO2017141246A1 (en) 2015-02-26 2017-02-15 Drug eluting stent

Country Status (2)

Country Link
IL (1) IL261182B (en)
WO (1) WO2017141246A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3890618A4 (en) * 2018-12-04 2023-01-11 The Brain Protection Company PTY Ltd Combinatorial therapies including implantable damping devices and therapeutic agents for treating a condition and associated systems and methods of use

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050187607A1 (en) * 2004-02-20 2005-08-25 Akhtar Adil J. Drug delivery device
US20060229715A1 (en) * 2005-03-29 2006-10-12 Sdgi Holdings, Inc. Implants incorporating nanotubes and methods for producing the same
US20090274739A1 (en) * 2006-04-13 2009-11-05 The Trustees Of Columbia University In The City Of New York Methods and compositions for treating neointimal hyperplasia

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050187607A1 (en) * 2004-02-20 2005-08-25 Akhtar Adil J. Drug delivery device
US20060229715A1 (en) * 2005-03-29 2006-10-12 Sdgi Holdings, Inc. Implants incorporating nanotubes and methods for producing the same
US20090274739A1 (en) * 2006-04-13 2009-11-05 The Trustees Of Columbia University In The City Of New York Methods and compositions for treating neointimal hyperplasia

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3890618A4 (en) * 2018-12-04 2023-01-11 The Brain Protection Company PTY Ltd Combinatorial therapies including implantable damping devices and therapeutic agents for treating a condition and associated systems and methods of use

Also Published As

Publication number Publication date
IL261182A (en) 2018-10-31
IL261182B (en) 2022-03-01

Similar Documents

Publication Publication Date Title
CA2626805C (en) Progenitor endothelial cell capturing with a drug eluting implantable medical device
US8088060B2 (en) Progenitor endothelial cell capturing with a drug eluting implantable medical device
US20160136337A1 (en) Progenitor endothelial cell capturing with a drug eluting implantable medical device
Jeschke et al. Polyurethane vascular prostheses decreases neointimal formation compared with expanded polytetrafluoroethylene
US8460367B2 (en) Progenitor endothelial cell capturing with a drug eluting implantable medical device
EP1235536B1 (en) MEDICAL DEVICE comprising a synthetic surface having nucleic acid for in vivo induction of its endothelialisation
US20070141107A1 (en) Progenitor Endothelial Cell Capturing with a Drug Eluting Implantable Medical Device
US9320829B2 (en) Progenitor endothelial cell capturing with a drug eluting implantable medical device
TW200539841A (en) Progenitor endothelial cell capturing with a drug eluting implantable medical device
JP2005514411A (en) Drug delivery system for the prevention and treatment of vascular diseases comprising rapamycin and its derivatives
JP2005523050A (en) Medical device having a coating that promotes endothelial cell adhesion and differentiation
JP2018198954A (en) Improved medical device
US20190117856A1 (en) Methods and compositions relating to leptin antagonists
WO2012167261A2 (en) Compositions and methods for treating and preventing neointimal stenosis
US10926006B2 (en) Drug eluting stent
IL261182A (en) Drug eluting stent
US11123461B2 (en) Treatment of ischemia and reperfusion using leptin antagonist
US10105469B2 (en) Composition-of-matter and method for treating cardiovascular disorders
AU2002220841B2 (en) Medical device
EP1395199A2 (en) Medical device

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 17752800

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 261182

Country of ref document: IL

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 17752800

Country of ref document: EP

Kind code of ref document: A1